70 results on '"Esti Yeger-Lotem"'
Search Results
2. Machine-learning analysis reveals an important role for negative selection in shaping cancer aneuploidy landscapes
- Author
-
Juman Jubran, Rachel Slutsky, Nir Rozenblum, Lior Rokach, Uri Ben-David, and Esti Yeger-Lotem
- Subjects
Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Background Aneuploidy, an abnormal number of chromosomes within a cell, is a hallmark of cancer. Patterns of aneuploidy differ across cancers, yet are similar in cancers affecting closely related tissues. The selection pressures underlying aneuploidy patterns are not fully understood, hindering our understanding of cancer development and progression. Results Here, we apply interpretable machine learning methods to study tissue-selective aneuploidy patterns. We define 20 types of features corresponding to genomic attributes of chromosome-arms, normal tissues, primary tumors, and cancer cell lines (CCLs), and use them to model gains and losses of chromosome arms in 24 cancer types. To reveal the factors that shape the tissue-specific cancer aneuploidy landscapes, we interpret the machine learning models by estimating the relative contribution of each feature to the models. While confirming known drivers of positive selection, our quantitative analysis highlights the importance of negative selection for shaping aneuploidy landscapes. This is exemplified by tumor suppressor gene density being a better predictor of gain patterns than oncogene density, and vice versa for loss patterns. We also identify the importance of tissue-selective features and demonstrate them experimentally, revealing KLF5 as an important driver for chr13q gain in colon cancer. Further supporting an important role for negative selection in shaping the aneuploidy landscapes, we find compensation by paralogs to be among the top predictors of chromosome arm loss prevalence and demonstrate this relationship for one paralog interaction. Similar factors shape aneuploidy patterns in human CCLs, demonstrating their relevance for aneuploidy research. Conclusions Our quantitative, interpretable machine learning models improve the understanding of the genomic properties that shape cancer aneuploidy landscapes.
- Published
- 2024
- Full Text
- View/download PDF
3. sNucConv: A bulk RNA-seq deconvolution method trained on single-nucleus RNA-seq data to estimate cell-type composition of human adipose tissues
- Author
-
Gil Sorek, Yulia Haim, Vered Chalifa-Caspi, Or Lazarescu, Maya Ziv-Agam, Tobias Hagemann, Pamela Arielle Nono Nankam, Matthias Blüher, Idit F. Liberty, Oleg Dukhno, Ivan Kukeev, Esti Yeger-Lotem, Assaf Rudich, and Liron Levin
- Subjects
Integrative aspects of cell biology ,Biocomputational method ,Classification of bioinformatical subject ,Transcriptomics ,Machine learning ,Science - Abstract
Summary: Deconvolution algorithms mostly rely on single-cell RNA-sequencing (scRNA-seq) data applied onto bulk RNA-sequencing (bulk RNA-seq) to estimate tissues’ cell-type composition, with performance accuracy validated on deposited databases. Adipose tissues’ cellular composition is highly variable, and adipocytes can only be captured by single-nucleus RNA-sequencing (snRNA-seq). Here we report the development of sNucConv, a Scaden deep-learning-based deconvolution tool, trained using 5 hSAT and 7 hVAT snRNA-seq-based data corrected by (i) snRNA-seq/bulk RNA-seq highly correlated genes and (ii) individual cell-type regression models. Applying sNucConv on our bulk RNA-seq data resulted in cell-type proportion estimation of 15 and 13 cell types, with accuracy of R = 0.93 (range: 0.76–0.97) and R = 0.95 (range: 0.92–0.98) for hVAT and hSAT, respectively. This performance level was further validated on an independent set of samples (5 hSAT; 5 hVAT). The resulting model was depot specific, reflecting depot differences in gene expression patterns. Jointly, sNucConv provides proof-of-concept for producing validated deconvolution models for tissues un-amenable to scRNA-seq.
- Published
- 2024
- Full Text
- View/download PDF
4. Affected cell types for hundreds of Mendelian diseases revealed by analysis of human and mouse single-cell data
- Author
-
Idan Hekselman, Assaf Vital, Maya Ziv-Agam, Lior Kerber, Ido Yairi, and Esti Yeger-Lotem
- Subjects
single-cell transcriptomics ,Mendelian diseases ,cell-type selectivity ,data integration ,tissue selectivity ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Mendelian diseases tend to manifest clinically in certain tissues, yet their affected cell types typically remain elusive. Single-cell expression studies showed that overexpression of disease-associated genes may point to the affected cell types. Here, we developed a method that infers disease-affected cell types from the preferential expression of disease-associated genes in cell types (PrEDiCT). We applied PrEDiCT to single-cell expression data of six human tissues, to infer the cell types affected in Mendelian diseases. Overall, we inferred the likely affected cell types for 328 diseases. We corroborated our findings by literature text-mining, expert validation, and recapitulation in mouse corresponding tissues. Based on these findings, we explored characteristics of disease-affected cell types, showed that diseases manifesting in multiple tissues tend to affect similar cell types, and highlighted cases where gene functions could be used to refine inference. Together, these findings expand the molecular understanding of disease mechanisms and cellular vulnerability.
- Published
- 2024
- Full Text
- View/download PDF
5. Predicting molecular mechanisms of hereditary diseases by using their tissue‐selective manifestation
- Author
-
Eyal Simonovsky, Moran Sharon, Maya Ziv, Omry Mauer, Idan Hekselman, Juman Jubran, Ekaterina Vinogradov, Chanan M Argov, Omer Basha, Lior Kerber, Yuval Yogev, Ayellet V Segrè, Hae Kyung Im, GTEx Consortium, Ohad Birk, Lior Rokach, and Esti Yeger‐Lotem
- Subjects
data integration ,genomic medicine ,machine learning ,omics ,tissue selectivity ,Biology (General) ,QH301-705.5 ,Medicine (General) ,R5-920 - Abstract
Abstract How do aberrations in widely expressed genes lead to tissue‐selective hereditary diseases? Previous attempts to answer this question were limited to testing a few candidate mechanisms. To answer this question at a larger scale, we developed “Tissue Risk Assessment of Causality by Expression” (TRACE), a machine learning approach to predict genes that underlie tissue‐selective diseases and selectivity‐related features. TRACE utilized 4,744 biologically interpretable tissue‐specific gene features that were inferred from heterogeneous omics datasets. Application of TRACE to 1,031 disease genes uncovered known and novel selectivity‐related features, the most common of which was previously overlooked. Next, we created a catalog of tissue‐associated risks for 18,927 protein‐coding genes ( https://netbio.bgu.ac.il/trace/ ). As proof‐of‐concept, we prioritized candidate disease genes identified in 48 rare‐disease patients. TRACE ranked the verified disease gene among the patient's candidate genes significantly better than gene prioritization methods that rank by gene constraint or tissue expression. Thus, tissue selectivity combined with machine learning enhances genetic and clinical understanding of hereditary diseases.
- Published
- 2023
- Full Text
- View/download PDF
6. Small heat-shock protein HSPB3 promotes myogenesis by regulating the lamin B receptor
- Author
-
Tatiana Tiago, Barbara Hummel, Federica F. Morelli, Valentina Basile, Jonathan Vinet, Veronica Galli, Laura Mediani, Francesco Antoniani, Silvia Pomella, Matteo Cassandri, Maria Giovanna Garone, Beatrice Silvestri, Marco Cimino, Giovanna Cenacchi, Roberta Costa, Vincent Mouly, Ina Poser, Esti Yeger-Lotem, Alessandro Rosa, Simon Alberti, Rossella Rota, Anat Ben-Zvi, Ritwick Sawarkar, and Serena Carra
- Subjects
Cytology ,QH573-671 - Abstract
Abstract One of the critical events that regulates muscle cell differentiation is the replacement of the lamin B receptor (LBR)-tether with the lamin A/C (LMNA)-tether to remodel transcription and induce differentiation-specific genes. Here, we report that localization and activity of the LBR-tether are crucially dependent on the muscle-specific chaperone HSPB3 and that depletion of HSPB3 prevents muscle cell differentiation. We further show that HSPB3 binds to LBR in the nucleoplasm and maintains it in a dynamic state, thus promoting the transcription of myogenic genes, including the genes to remodel the extracellular matrix. Remarkably, HSPB3 overexpression alone is sufficient to induce the differentiation of two human muscle cell lines, LHCNM2 cells, and rhabdomyosarcoma cells. We also show that mutant R116P-HSPB3 from a myopathy patient with chromatin alterations and muscle fiber disorganization, forms nuclear aggregates that immobilize LBR. We find that R116P-HSPB3 is unable to induce myoblast differentiation and instead activates the unfolded protein response. We propose that HSPB3 is a specialized chaperone engaged in muscle cell differentiation and that dysfunctional HSPB3 causes neuromuscular disease by deregulating LBR.
- Published
- 2021
- Full Text
- View/download PDF
7. The landscape of molecular chaperones across human tissues reveals a layered architecture of core and variable chaperones
- Author
-
Netta Shemesh, Juman Jubran, Shiran Dror, Eyal Simonovsky, Omer Basha, Chanan Argov, Idan Hekselman, Mehtap Abu-Qarn, Ekaterina Vinogradov, Omry Mauer, Tatiana Tiago, Serena Carra, Anat Ben-Zvi, and Esti Yeger-Lotem
- Subjects
Science - Abstract
Tissue-specific differences in protein folding capacities are poorly understood. Here, the authors show that the human chaperone system consists of ubiquitous core chaperones and tissue-specific variable chaperones, perturbation of which leads to tissue-specific phenotypes.
- Published
- 2021
- Full Text
- View/download PDF
8. Dosage-sensitive molecular mechanisms are associated with the tissue-specificity of traits and diseases
- Author
-
Juman Jubran, Idan Hekselman, Lena Novack, and Esti Yeger-Lotem
- Subjects
Hereditary diseases ,Complex traits ,Paralogs ,Data integration ,Linear mixed models ,Biotechnology ,TP248.13-248.65 - Abstract
Hereditary diseases and complex traits often manifest in specific tissues, whereas their causal genes are expressed in many tissues that remain unaffected. Among the mechanisms that have been suggested for this enigmatic phenomenon is dosage-sensitive compensation by paralogs of causal genes. Accordingly, tissue-selectivity stems from dosage imbalance between causal genes and paralogs that occurs particularly in disease-susceptible tissues. Here, we used a large-scale dataset of thousands of tissue transcriptomes and applied a linear mixed model (LMM) framework to assess this and other dosage-sensitive mechanisms. LMM analysis of 382 hereditary diseases consistently showed evidence for dosage-sensitive compensation by paralogs across diseases subsets and susceptible tissues. LMM analysis of 135 candidate genes that are strongly associated with 16 tissue-selective complex traits revealed a similar tendency among half of the trait-associated genes. This suggests that dosage-sensitive compensation by paralogs affects the tissue-selectivity of complex traits, and can be used to illuminate candidate genes' modes of action. Next, we applied LMM to analyze dosage imbalance between causal genes and three classes of genetic modifiers, including regulatory micro-RNAs, pseudogenes, and genetic interactors. Our results propose modifiers as a fundamental axis in tissue-selectivity of diseases and traits, and demonstrates the power of LMM as a statistical framework for discovering treatment avenues.
- Published
- 2020
- Full Text
- View/download PDF
9. Role of duplicate genes in determining the tissue-selectivity of hereditary diseases.
- Author
-
Ruth Barshir, Idan Hekselman, Netta Shemesh, Moran Sharon, Lena Novack, and Esti Yeger-Lotem
- Subjects
Genetics ,QH426-470 - Abstract
A longstanding puzzle in human genetics is what limits the clinical manifestation of hundreds of hereditary diseases to certain tissues, while their causal genes are expressed throughout the human body. A general conception is that tissue-selective disease phenotypes emerge when masking factors operate in unaffected tissues, but are specifically absent or insufficient in disease-manifesting tissues. Although this conception has critical impact on the understanding of disease manifestation, it was never challenged in a systematic manner across a variety of hereditary diseases and affected tissues. Here, we address this gap in our understanding via rigorous analysis of the susceptibility of over 30 tissues to 112 tissue-selective hereditary diseases. We focused on the roles of paralogs of causal genes, which are presumably capable of compensating for their aberration. We show for the first time at large-scale via quantitative analysis of omics datasets that, preferentially in the disease-manifesting tissues, paralogs are under-expressed relative to causal genes in more than half of the diseases. This was observed for several susceptible tissues and for causal genes with varying number of paralogs, suggesting that imbalanced expression of paralogs increases tissue susceptibility. While for many diseases this imbalance stemmed from up-regulation of the causal gene in the disease-manifesting tissue relative to other tissues, it was often combined with down-regulation of its paralog. Notably in roughly 20% of the cases, this imbalance stemmed only from significant down-regulation of the paralog. Thus, dosage relationships between paralogs appear as important, yet currently under-appreciated, modifiers of disease manifestation.
- Published
- 2018
- Full Text
- View/download PDF
10. An Asymmetrically Balanced Organization of Kinases versus Phosphatases across Eukaryotes Determines Their Distinct Impacts.
- Author
-
Ilan Smoly, Netta Shemesh, Michal Ziv-Ukelson, Anat Ben-Zvi, and Esti Yeger-Lotem
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Protein phosphorylation underlies cellular response pathways across eukaryotes and is governed by the opposing actions of phosphorylating kinases and de-phosphorylating phosphatases. While kinases and phosphatases have been extensively studied, their organization and the mechanisms by which they balance each other are not well understood. To address these questions we performed quantitative analyses of large-scale 'omics' datasets from yeast, fly, plant, mouse and human. We uncovered an asymmetric balance of a previously-hidden scale: Each organism contained many different kinase genes, and these were balanced by a small set of highly abundant phosphatase proteins. Kinases were much more responsive to perturbations at the gene and protein levels. In addition, kinases had diverse scales of phenotypic impact when manipulated. Phosphatases, in contrast, were stable, highly robust and flatly organized, with rather uniform impact downstream. We validated aspects of this organization experimentally in nematode, and supported additional aspects by theoretic analysis of the dynamics of protein phosphorylation. Our analyses explain the empirical bias in the protein phosphorylation field toward characterization and therapeutic targeting of kinases at the expense of phosphatases. We show quantitatively and broadly that this is not only a historical bias, but stems from wide-ranging differences in their organization and impact. The asymmetric balance between these opposing regulators of protein phosphorylation is also common to opposing regulators of two other post-translational modification systems, suggesting its fundamental value.
- Published
- 2017
- Full Text
- View/download PDF
11. A Differentiation Transcription Factor Establishes Muscle-Specific Proteostasis in Caenorhabditis elegans.
- Author
-
Yael Bar-Lavan, Netta Shemesh, Shiran Dror, Rivka Ofir, Esti Yeger-Lotem, and Anat Ben-Zvi
- Subjects
Genetics ,QH426-470 - Abstract
Safeguarding the proteome is central to the health of the cell. In multi-cellular organisms, the composition of the proteome, and by extension, protein-folding requirements, varies between cells. In agreement, chaperone network composition differs between tissues. Here, we ask how chaperone expression is regulated in a cell type-specific manner and whether cellular differentiation affects chaperone expression. Our bioinformatics analyses show that the myogenic transcription factor HLH-1 (MyoD) can bind to the promoters of chaperone genes expressed or required for the folding of muscle proteins. To test this experimentally, we employed HLH-1 myogenic potential to genetically modulate cellular differentiation of Caenorhabditis elegans embryonic cells by ectopically expressing HLH-1 in all cells of the embryo and monitoring chaperone expression. We found that HLH-1-dependent myogenic conversion specifically induced the expression of putative HLH-1-regulated chaperones in differentiating muscle cells. Moreover, disrupting the putative HLH-1-binding sites on ubiquitously expressed daf-21(Hsp90) and muscle-enriched hsp-12.2(sHsp) promoters abolished their myogenic-dependent expression. Disrupting HLH-1 function in muscle cells reduced the expression of putative HLH-1-regulated chaperones and compromised muscle proteostasis during and after embryogenesis. In turn, we found that modulating the expression of muscle chaperones disrupted the folding and assembly of muscle proteins and thus, myogenesis. Moreover, muscle-specific over-expression of the DNAJB6 homolog DNJ-24, a limb-girdle muscular dystrophy-associated chaperone, disrupted the muscle chaperone network and exposed synthetic motility defects. We propose that cellular differentiation could establish a proteostasis network dedicated to the folding and maintenance of the muscle proteome. Such cell-specific proteostasis networks can explain the selective vulnerability that many diseases of protein misfolding exhibit even when the misfolded protein is ubiquitously expressed.
- Published
- 2016
- Full Text
- View/download PDF
12. Comparative analysis of human tissue interactomes reveals factors leading to tissue-specific manifestation of hereditary diseases.
- Author
-
Ruth Barshir, Omer Shwartz, Ilan Y Smoly, and Esti Yeger-Lotem
- Subjects
Biology (General) ,QH301-705.5 - Abstract
An open question in human genetics is what underlies the tissue-specific manifestation of hereditary diseases, which are caused by genomic aberrations that are present in cells across the human body. Here we analyzed this phenomenon for over 300 hereditary diseases by using comparative network analysis. We created an extensive resource of protein expression and interactions in 16 main human tissues, by integrating recent data of gene and protein expression across tissues with data of protein-protein interactions (PPIs). The resulting tissue interaction networks (interactomes) shared a large fraction of their proteins and PPIs, and only a small fraction of them were tissue-specific. Applying this resource to hereditary diseases, we first show that most of the disease-causing genes are widely expressed across tissues, yet, enigmatically, cause disease phenotypes in few tissues only. Upon testing for factors that could lead to tissue-specific vulnerability, we find that disease-causing genes tend to have elevated transcript levels and increased number of tissue-specific PPIs in their disease tissues compared to unaffected tissues. We demonstrate through several examples that these tissue-specific PPIs can highlight disease mechanisms, and thus, owing to their small number, provide a powerful filter for interrogating disease etiologies. As two thirds of the hereditary diseases are associated with these factors, comparative tissue analysis offers a meaningful and efficient framework for enhancing the understanding of the molecular basis of hereditary diseases.
- Published
- 2014
- Full Text
- View/download PDF
13. Cancer evolution is associated with pervasive positive selection on globally expressed genes.
- Author
-
Sheli L Ostrow, Ruth Barshir, James DeGregori, Esti Yeger-Lotem, and Ruth Hershberg
- Subjects
Genetics ,QH426-470 - Abstract
Cancer is an evolutionary process in which cells acquire new transformative, proliferative and metastatic capabilities. A full understanding of cancer requires learning the dynamics of the cancer evolutionary process. We present here a large-scale analysis of the dynamics of this evolutionary process within tumors, with a focus on breast cancer. We show that the cancer evolutionary process differs greatly from organismal (germline) evolution. Organismal evolution is dominated by purifying selection (that removes mutations that are harmful to fitness). In contrast, in the cancer evolutionary process the dominance of purifying selection is much reduced, allowing for a much easier detection of the signals of positive selection (adaptation). We further show that, as a group, genes that are globally expressed across human tissues show a very strong signal of positive selection within tumors. Indeed, known cancer genes are enriched for global expression patterns. Yet, positive selection is prevalent even on globally expressed genes that have not yet been associated with cancer, suggesting that globally expressed genes are enriched for yet undiscovered cancer related functions. We find that the increased positive selection on globally expressed genes within tumors is not due to their expression in the tissue relevant to the cancer. Rather, such increased adaptation is likely due to globally expressed genes being enriched in important housekeeping and essential functions. Thus, our results suggest that tumor adaptation is most often mediated through somatic changes to those genes that are important for the most basic cellular functions. Together, our analysis reveals the uniqueness of the cancer evolutionary process and the particular importance of globally expressed genes in driving cancer initiation and progression.
- Published
- 2014
- Full Text
- View/download PDF
14. Algorithms for Regular Tree Grammar Network Search and Their Application to Mining Human-Viral Infection Patterns.
- Author
-
Ilan Y. Smoly, Amir Carmel, Yonat Shemer-Avni, Esti Yeger Lotem, and Michal Ziv-Ukelson
- Published
- 2015
- Full Text
- View/download PDF
15. ProAct: quantifying the differential activity of biological processes in tissues, cells, and user-defined contexts
- Author
-
Moran Sharon, Gil Gruber, Chanan M Argov, Miri Volozhinsky, and Esti Yeger-Lotem
- Subjects
Genetics - Abstract
The distinct functions and phenotypes of human tissues and cells derive from the activity of biological processes that varies in a context-dependent manner. Here, we present the Process Activity (ProAct) webserver that estimates the preferential activity of biological processes in tissues, cells, and other contexts. Users can upload a differential gene expression matrix measured across contexts or cells, or use a built-in matrix of differential gene expression in 34 human tissues. Per context, ProAct associates gene ontology (GO) biological processes with estimated preferential activity scores, which are inferred from the input matrix. ProAct visualizes these scores across processes, contexts, and process-associated genes. ProAct also offers potential cell-type annotations for cell subsets, by inferring them from the preferential activity of 2001 cell-type-specific processes. Thus, ProAct output can highlight the distinct functions of tissues and cell types in various contexts, and can enhance cell-type annotation efforts. The ProAct webserver is available at https://netbio.bgu.ac.il/ProAct/.
- Published
- 2023
16. The differential activity of biological processes in tissues and cell subsets can illuminate disease-related processes and cell-type identities
- Author
-
Moran Sharon, Ekaterina Vinogradov, Chanan M Argov, Or Lazarescu, Yazeed Zoabi, Idan Hekselman, and Esti Yeger-Lotem
- Subjects
Statistics and Probability ,Computational Mathematics ,Computational Theory and Mathematics ,Molecular Biology ,Biochemistry ,Computer Science Applications - Abstract
Motivation The distinct functionalities of human tissues and cell types underlie complex phenotype–genotype relationships, yet often remain elusive. Harnessing the multitude of bulk and single-cell human transcriptomes while focusing on processes can help reveal these distinct functionalities. Results The Tissue-Process Activity (TiPA) method aims to identify processes that are preferentially active or under-expressed in specific contexts, by comparing the expression levels of process genes between contexts. We tested TiPA on 1579 tissue-specific processes and bulk tissue transcriptomes, finding that it performed better than another method. Next, we used TiPA to ask whether the activity of certain processes could underlie the tissue-specific manifestation of 1233 hereditary diseases. We found that 21% of the disease-causing genes indeed participated in such processes, thereby illuminating their genotype–phenotype relationships. Lastly, we applied TiPA to single-cell transcriptomes of 108 human cell types, revealing that process activities often match cell-type identities and can thus aid annotation efforts. Hence, differential activity of processes can highlight the distinct functionality of tissues and cells in a robust and meaningful manner. Availability and implementation TiPA code is available in GitHub (https://github.com/moranshar/TiPA). In addition, all data are available as part of the Supplementary Material. Supplementary information Supplementary data are available at Bioinformatics online.
- Published
- 2022
17. Network Modeling of Tissues and Cell Types
- Author
-
Maya Ziv and Esti Yeger-Lotem
- Published
- 2023
18. Affected cell types for hundreds of Mendelian diseases revealed by analysis of human and mouse single-cell data
- Author
-
Idan Hekselman, Assaf Vital, Maya Ziv-Agam, Lior Kerber, and Esti Yeger-Lotem
- Abstract
Hereditary diseases manifest clinically in certain tissues, however their affected cell types typically remain elusive. Single-cell expression studies showed that overexpression of disease-associated genes may point to the affected cell types. Here, we developed a method that infers disease-affected cell types from the preferential expression of disease-associated genes in cell types (PrEDiCT). We applied PrEDiCT to single-cell expression data of six human tissues, to infer the cell types affected in 1,113 hereditary diseases. Overall, we identified 110 cell types affected by 714 diseases. We corroborated our findings by literature text-mining and recapitulation in mouse corresponding tissues. Based on these findings, we explored features of disease-affected cell types and cell classes, highlighted cell types affected by mitochondrial diseases and heritable cancers, and identified diseases that perturb intercellular communication. This study expands our understanding of disease mechanisms and cellular vulnerability.
- Published
- 2022
19. The AS/400 Cluster Engine: A Case Study.
- Author
-
Gera Goft and Esti Yeger Lotem
- Published
- 1999
- Full Text
- View/download PDF
20. Differential network analysis of multiple human tissue interactomes highlights tissue-selective processes and genetic disorder genes
- Author
-
Idan Hekselman, Chanan M Argov, Liad Alfandari, Raviv Artzy, Yazeed Zoabi, Omer Basha, Vered Chalifa-Caspi, and Esti Yeger-Lotem
- Subjects
Statistics and Probability ,Computer science ,Gene regulatory network ,Computational biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Gene Regulatory Networks ,Protein Interaction Maps ,Molecular Biology ,Gene ,Biological Phenomena ,030304 developmental biology ,0303 health sciences ,Genetic disorder ,medicine.disease ,Computer Science Applications ,Computational Mathematics ,Gene Ontology ,Computational Theory and Mathematics ,030217 neurology & neurosurgery ,Differential (mathematics) ,Biological network ,Network analysis - Abstract
Motivation Differential network analysis, designed to highlight network changes between conditions, is an important paradigm in network biology. However, differential network analysis methods have been typically designed to compare between two conditions and were rarely applied to multiple protein interaction networks (interactomes). Importantly, large-scale benchmarks for their evaluation have been lacking. Results Here, we present a framework for assessing the ability of differential network analysis of multiple human tissue interactomes to highlight tissue-selective processes and disorders. For this, we created a benchmark of 6499 curated tissue-specific Gene Ontology biological processes. We applied five methods, including four differential network analysis methods, to construct weighted interactomes for 34 tissues. Rigorous assessment of this benchmark revealed that differential analysis methods perform well in revealing tissue-selective processes (AUCs of 0.82–0.9). Next, we applied differential network analysis to illuminate the genes underlying tissue-selective hereditary disorders. For this, we curated a dataset of 1305 tissue-specific hereditary disorders and their manifesting tissues. Focusing on subnetworks containing the top 1% differential interactions in disease-relevant tissue interactomes revealed significant enrichment for disorder-causing genes in 18.6% of the cases, with a significantly high success rate for blood, nerve, muscle and heart diseases. Summary Altogether, we offer a framework that includes expansive manually curated datasets of tissue-selective processes and disorders to be used as benchmarks or to illuminate tissue-selective processes and genes. Our results demonstrate that differential analysis of multiple human tissue interactomes is a powerful tool for highlighting processes and genes with tissue-selective functionality and clinical impact. Availability and implementation Datasets are available as part of the Supplementary data. Supplementary information Supplementary data are available at Bioinformatics online.
- Published
- 2020
21. Mechanisms of tissue and cell-type specificity in heritable traits and diseases
- Author
-
Esti Yeger-Lotem and Idan Hekselman
- Subjects
0303 health sciences ,Cell type ,Genotype ,Systems biology ,Infant, Newborn ,Disease ,Biology ,Phenotype ,Germline ,Omics data ,03 medical and health sciences ,0302 clinical medicine ,Evolutionary biology ,Databases, Genetic ,Genetics ,Humans ,Narrow range ,Genetic Predisposition to Disease ,Molecular Biology ,Gene ,030217 neurology & neurosurgery ,Genetics (clinical) ,030304 developmental biology - Abstract
Hundreds of heritable traits and diseases that are caused by germline aberrations in ubiquitously expressed genes manifest in a remarkably limited number of cell types and tissues across the body. Unravelling mechanisms that govern their tissue-specific manifestations is critical for our understanding of disease aetiologies and may direct efforts to develop treatments. Owing to recent advances in high-throughput technologies and open resources, data and tools are now available to approach this enigmatic phenomenon at large scales, both computationally and experimentally. Here, we discuss the large prevalence of tissue-selective traits and diseases, describe common molecular mechanisms underlying their tissue-selective manifestation and present computational strategies and publicly available resources for elucidating the molecular basis of their genotype–phenotype relationships. The pathology of heritable human traits and diseases often affects a narrow range of tissues, even when causal genes are expressed widely across the body. In this Review, Hekselman and Yeger-Lotem discuss the latest understanding of tissue specificity in human traits and disease, including the diverse underlying molecular mechanisms, experimental and bioinformatics resources to leverage omics data, and implications for understanding disease aetiology.
- Published
- 2020
22. Dosage-sensitive molecular mechanisms are associated with the tissue-specificity of traits and diseases
- Author
-
Lena Novack, Juman Jubran, Idan Hekselman, and Esti Yeger-Lotem
- Subjects
Candidate gene ,animal structures ,Linear mixed models ,Pseudogene ,lcsh:Biotechnology ,Biophysics ,Biology ,Biochemistry ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Structural Biology ,lcsh:TP248.13-248.65 ,Genetics ,Gene ,030304 developmental biology ,0303 health sciences ,Hereditary diseases ,Paralogs ,Complex traits ,Computer Science Applications ,Tissue specificity ,030220 oncology & carcinogenesis ,Hereditary Diseases ,Data integration ,Biotechnology ,Research Article - Abstract
Hereditary diseases and complex traits often manifest in specific tissues, whereas their causal genes are expressed in many tissues that remain unaffected. Among the mechanisms that have been suggested for this enigmatic phenomenon is dosage-sensitive compensation by paralogs of causal genes. Accordingly, tissue-selectivity stems from dosage imbalance between causal genes and paralogs that occurs particularly in disease-susceptible tissues. Here, we used a large-scale dataset of thousands of tissue transcriptomes and applied a linear mixed model (LMM) framework to assess this and other dosage-sensitive mechanisms. LMM analysis of 382 hereditary diseases consistently showed evidence for dosage-sensitive compensation by paralogs across diseases subsets and susceptible tissues. LMM analysis of 135 candidate genes that are strongly associated with 16 tissue-selective complex traits revealed a similar tendency among half of the trait-associated genes. This suggests that dosage-sensitive compensation by paralogs affects the tissue-selectivity of complex traits, and can be used to illuminate candidate genes' modes of action. Next, we applied LMM to analyze dosage imbalance between causal genes and three classes of genetic modifiers, including regulatory micro-RNAs, pseudogenes, and genetic interactors. Our results propose modifiers as a fundamental axis in tissue-selectivity of diseases and traits, and demonstrates the power of LMM as a statistical framework for discovering treatment avenues.
- Published
- 2020
23. Dynamic Voting for Consistent Primary Components.
- Author
-
Esti Yeger Lotem, Idit Keidar, and Danny Dolev
- Published
- 1997
- Full Text
- View/download PDF
24. The TissueNet v.3 Database: Protein-protein Interactions in Adult and Embryonic Human Tissue contexts
- Author
-
Maya Ziv, Gil Gruber, Moran Sharon, Ekaterina Vinogradov, and Esti Yeger-Lotem
- Subjects
Adult ,Structural Biology ,Protein Interaction Mapping ,Humans ,Proteins ,Protein Interaction Maps ,Databases, Protein ,Embryo, Mammalian ,Molecular Biology ,Software - Abstract
Tissue contexts are extremely valuable when studying protein functions and their associated phenotypes. Recently, the study of proteins in tissue contexts was greatly facilitated by the availability of thousands of tissue transcriptomes. To provide access to these data we developed the TissueNet integrative database that displays protein-protein interactions (PPIs) in tissue contexts. Through TissueNet, users can create tissue-sensitive network views of the PPI landscape of query proteins. Unlike other tools, TissueNet output networks highlight tissue-specific and broadly expressed proteins, as well as over- and under-expressed proteins per tissue. The TissueNet v.3 upgrade has a much larger dataset of proteins and PPIs, and represents 125 adult tissues and seven embryonic tissues. Thus, TissueNet provides an extensive, quantitative, and user-friendly interface to study the roles of human proteins in adulthood and embryonic stages. TissueNet v3 is freely available at https://netbio.bgu.ac.il/tissuenet3.
- Published
- 2021
25. The Organ-Disease Annotations (ODiseA) Database of Hereditary Diseases and Inflicted Tissues
- Author
-
Idan Hekselman, Lior Kerber, Maya Ziv, Gil Gruber, and Esti Yeger-Lotem
- Subjects
Phenotype ,Structural Biology ,Organ Specificity ,Databases, Genetic ,Genetic Diseases, Inborn ,Computational Biology ,Humans ,Molecular Biology - Abstract
Hereditary diseases tend to manifest clinically in few selected tissues. Knowledge of those tissues is important for better understanding of disease mechanisms, which often remain elusive. However, information on the tissues inflicted by each disease is not easily obtainable. Well-established resources, such as the Online Mendelian Inheritance in Man (OMIM) database and Human Phenotype Ontology (HPO), report on a spectrum of disease manifestations, yet do not highlight the main inflicted tissues. The Organ-Disease Annotations (ODiseA) database contains 4,357 thoroughly-curated annotations for 2,181 hereditary diseases and 45 inflicted tissues. Additionally, ODiseA reports 692 annotations of 635 diseases and the pathogenic tissues where they emerge. ODiseA can be queried by disease, disease gene, or inflicted tissue. Owing to its expansive, high-quality annotations, ODiseA serves as a valuable and unique tool for biomedical and computational researchers studying genotype-phenotype relationships of hereditary diseases. ODiseA is available at https://netbio.bgu.ac.il/odisea.
- Published
- 2021
26. Unraveling the hidden role of a uORF-encoded peptide as a kinase inhibitor of PKCs
- Author
-
Vijayasteltar Belsamma Liju, Debra Toiber, Divya Ram Jayaram, Ilan Smoly, Rose Sinay, Ofra Novoplansky, Esti Yeger-Lotem, Nikhil Ponnoor Anto, Chen Keasar, Chanan M Argov, Amitha Muraleedharan, Moshe Elkabets, Noah Isakov, Assaf Ben-Ari, Etta Livneh, and Sigal A. Frost
- Subjects
Untranslated region ,kinase inhibitor ,Substrate Specificity ,Open Reading Frames ,Cell Line, Tumor ,Humans ,Amino Acid Sequence ,PKC ,Kinase activity ,Protein kinase A ,Protein Kinase Inhibitors ,Protein Kinase C ,Protein kinase C ,Multidisciplinary ,Chemistry ,Kinase ,Cell growth ,pseudosubstrate ,PKCS ,Cell Biology ,Biological Sciences ,Cell biology ,Open reading frame ,Peptides ,uORF - Abstract
Significance Bioinformatic analysis revealed that approximately 40% of human messenger RNAs contain upstream open reading frames (uORFs) in their 5′ untranslated regions. Some of these sequences are translated, but the function of the encoded peptides remains unknown. Our study revealed a uORF encoding for a peptide exhibiting kinase inhibitory activity. This uORF, upstream of a PKC family member, possess the typical pseudosubstrate motif, which autoinhibits the catalytic activity of all PKCs. Using mouse models and human cells, we show that this peptide inhibits cancer cell survival, tumor progression, invasion, and metastasis and synergizes with chemotherapy by interfering with DNA damage response. Together, we point to a previously unrecognized function of a uORF-encoded peptide as a kinase inhibitor, pertinent to cancer therapy., Approximately 40% of human messenger RNAs (mRNAs) contain upstream open reading frames (uORFs) in their 5′ untranslated regions. Some of these uORF sequences, thought to attenuate scanning ribosomes or lead to mRNA degradation, were recently shown to be translated, although the function of the encoded peptides remains unknown. Here, we show a uORF-encoded peptide that exhibits kinase inhibitory functions. This uORF, upstream of the protein kinase C-eta (PKC-η) main ORF, encodes a peptide (uPEP2) containing the typical PKC pseudosubstrate motif present in all PKCs that autoinhibits their kinase activity. We show that uPEP2 directly binds to and selectively inhibits the catalytic activity of novel PKCs but not of classical or atypical PKCs. The endogenous deletion of uORF2 or its overexpression in MCF-7 cells revealed that the endogenously translated uPEP2 reduces the protein levels of PKC-η and other novel PKCs and restricts cell proliferation. Functionally, treatment of breast cancer cells with uPEP2 diminished cell survival and their migration and synergized with chemotherapy by interfering with the response to DNA damage. Furthermore, in a xenograft of MDA-MB-231 breast cancer tumor in mice models, uPEP2 suppressed tumor progression, invasion, and metastasis. Tumor histology showed reduced proliferation, enhanced cell death, and lower protein expression levels of novel PKCs along with diminished phosphorylation of PKC substrates. Hence, our study demonstrates that uORFs may encode biologically active peptides beyond their role as translation regulators of their downstream ORFs. Together, we point to a unique function of a uORF-encoded peptide as a kinase inhibitor, pertinent to cancer therapy.
- Published
- 2021
27. Large-scale analysis of human gene expression variability associates highly variable drug targets with lower drug effectiveness and safety
- Author
-
Ronen Schuster, Esti Yeger-Lotem, and Eyal Simonovsky
- Subjects
Statistics and Probability ,Drug ,Sequence analysis ,media_common.quotation_subject ,Population ,Gene Expression ,Computational biology ,Biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Humans ,Distribution (pharmacology) ,education ,Molecular Biology ,Gene ,030304 developmental biology ,media_common ,Supplementary data ,0303 health sciences ,education.field_of_study ,Sequence Analysis, RNA ,Original Papers ,Computer Science Applications ,Computational Mathematics ,Variable (computer science) ,Computational Theory and Mathematics ,030220 oncology & carcinogenesis - Abstract
Motivation The effectiveness of drugs tends to vary between patients. One of the well-known reasons for this phenomenon is genetic polymorphisms in drug target genes among patients. Here, we propose that differences in expression levels of drug target genes across individuals can also contribute to this phenomenon. Results To explore this hypothesis, we analyzed the expression variability of protein-coding genes, and particularly drug target genes, across individuals. For this, we developed a novel variability measure, termed local coefficient of variation (LCV), which ranks the expression variability of each gene relative to genes with similar expression levels. Unlike commonly used methods, LCV neutralizes expression levels biases without imposing any distribution over the variation and is robust to data incompleteness. Application of LCV to RNA-sequencing profiles of 19 human tissues and to target genes of 1076 approved drugs revealed that drug target genes were significantly more variable than protein-coding genes. Analysis of 113 drugs with available effectiveness scores showed that drugs targeting highly variable genes tended to be less effective in the population. Furthermore, comparison of approved drugs to drugs that were withdrawn from the market showed that withdrawn drugs targeted significantly more variable genes than approved drugs. Last, upon analyzing gender differences we found that the variability of drug target genes was similar between men and women. Altogether, our results suggest that expression variability of drug target genes could contribute to the variable responsiveness and effectiveness of drugs, and is worth considering during drug treatment and development. Availability and implementation LCV is available as a python script in GitHub (https://github.com/eyalsim/LCV). Supplementary information Supplementary data are available at Bioinformatics online.
- Published
- 2019
28. The landscape of molecular chaperones across human tissues reveals a layered architecture of core and variable chaperones
- Author
-
Ekaterina Vinogradov, Eyal Simonovsky, Idan Hekselman, Omry Mauer, Serena Carra, Omer Basha, Anat Ben-Zvi, Juman Jubran, Mehtap Abu-Qarn, Chanan M Argov, Netta Shemesh, Shiran Dror, Tatiana Tiago, and Esti Yeger-Lotem
- Subjects
0301 basic medicine ,Aging ,Proteome ,Evolution ,Science ,Systems analysis ,General Physics and Astronomy ,Computational biology ,Animals ,Caenorhabditis elegans ,Cell Line ,Conserved Sequence ,Evolution, Molecular ,Gene Expression Regulation ,Humans ,Mice ,Molecular Chaperones ,Open Reading Frames ,Organ Specificity ,Article ,General Biochemistry, Genetics and Molecular Biology ,Conserved sequence ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Chaperones ,Regulation of gene expression ,Multidisciplinary ,biology ,Multitier architecture ,Molecular ,General Chemistry ,biology.organism_classification ,Open reading frame ,030104 developmental biology ,Chaperone (protein) ,biology.protein ,Data integration ,030217 neurology & neurosurgery - Abstract
The sensitivity of the protein-folding environment to chaperone disruption can be highly tissue-specific. Yet, the organization of the chaperone system across physiological human tissues has received little attention. Through computational analyses of large-scale tissue transcriptomes, we unveil that the chaperone system is composed of core elements that are uniformly expressed across tissues, and variable elements that are differentially expressed to fit with tissue-specific requirements. We demonstrate via a proteomic analysis that the muscle-specific signature is functional and conserved. Core chaperones are significantly more abundant across tissues and more important for cell survival than variable chaperones. Together with variable chaperones, they form tissue-specific functional networks. Analysis of human organ development and aging brain transcriptomes reveals that these functional networks are established in development and decline with age. In this work, we expand the known functional organization of de novo versus stress-inducible eukaryotic chaperones into a layered core-variable architecture in multi-cellular organisms., Tissue-specific differences in protein folding capacities are poorly understood. Here, the authors show that the human chaperone system consists of ubiquitous core chaperones and tissue-specific variable chaperones, perturbation of which leads to tissue-specific phenotypes.
- Published
- 2021
29. A tissue-aware machine learning framework enhances the mechanistic understanding and genetic diagnosis of Mendelian and rare diseases
- Author
-
Hae Kyung Im, Lior Kerber, Eyal Simonovsky, Maya Ziv, Ekaterina Vinogradov, Ayellet V. Segrè, Juman Jubran, Lior Rokach, Yuval Yogev, Ohad S. Birk, Idan Hekselman, Omry Mauer, Moran Sharon, Omer Basha, Esti Yeger-Lotem, and Chanan M Argov
- Subjects
Candidate gene ,business.industry ,Disease ,Biology ,Machine learning ,computer.software_genre ,Mendelian disease ,symbols.namesake ,Mendelian inheritance ,symbols ,Human genome ,Artificial intelligence ,Genetic diagnosis ,business ,Disease manifestation ,computer ,Gene - Abstract
Genetic studies of Mendelian and rare diseases face the critical challenges of identifying pathogenic gene variants and their modes-of-action. Previous efforts rarely utilized the tissue-selective manifestation of these diseases for their elucidation. Here we introduce an interpretable machine learning (ML) platform that utilizes heterogeneous and large-scale tissue-aware datasets of human genes, and rigorously, concurrently and quantitatively assesses hundreds of candidate mechanisms per disease. The resulting tissue-aware ML platform is applicable in gene-specific, tissue-specific, or patient-specific modes. Application of the platform to selected Mendelian disease genes pinpointed mechanisms that lead to tissue-specific disease manifestation. When applied jointly to diseases that manifest in the same tissue, the models revealed common known and previously underappreciated factors that underlie tissue-selective disease manifestation. Lastly, we harnessed our ML platform toward genetic diagnosis of tissue-selective rare diseases. Patient-specific models of candidate disease-causing genes from 50 patients successfully prioritized the pathogenic gene in 86% of the cases, implying that the tissue-selectivity of rare diseases aids in filtering out unlikely candidate genes. Thus, interpretable tissue-aware ML models can boost mechanistic understanding and genetic diagnosis of tissue-selective heritable diseases. A webserver supporting gene prioritization is available at https://netbio.bgu.ac.il/trace/.
- Published
- 2021
30. Multi-cellular communities are perturbed in the aging human brain and Alzheimer’s disease
- Author
-
Gilad Green, Anael Cain, Orit Rozenblatt-Rosen, Cristin McCabe, Feng Zhang, Idan Hekselman, Philip L. De Jager, Mariko Taga, Esti Yeger-Lotem, Aviv Regev, Vilas Menon, David A. Bennett, Naomi Habib, Hyun-Sik Yang, and Charles C. White
- Subjects
Cell type ,Tau pathology ,Frontal cortex ,medicine.anatomical_structure ,Cellular architecture ,medicine ,Human brain ,Disease ,Biology ,Cognitive decline ,Neuroscience ,Nucleus - Abstract
The role of different cell types and their interactions in Alzheimer’s disease (AD) is an open question. Here we pursued it by assembling a high-resolution cellular map of the aging frontal cortex by single nucleus RNA-seq of 24 individuals with different clinicopathologic characteristics. We used the map to infer the neocortical cellular architecture of 638 individuals profiled by bulk RNA-seq, providing the sample size necessary for identifying statistically robust associations. We uncovered diverse cell populations associated with AD, including inhibitory neuronal subtypes and oligodendroglial states. We further recovered a network of multicellular communities, each composed of coordinated subpopulations of neuronal, glial and endothelial cells, and found that two of these communities are altered in AD. Finally, we used mediation analyses to prioritize cellular changes that might contribute to cognitive decline. Thus, our deconstruction of the aging neocortex provides a roadmap for evaluating the cellular microenvironments underlying AD and dementia.
- Published
- 2020
31. The GTEx Consortium atlas of genetic regulatory effects across human tissues
- Author
-
Deborah C. Mash, Kevin S. Smith, Lappalainen T, Jeffrey A. Thomas, Rajinder Kaul, Paul Flicek, Maghboeba Mosavel, Yuxin Zou, Barbara E. Stranger, Brandon L. Pierce, Yanyu Liang, Andrew R. Hamel, Lihua Jiang, Marcus Hunter, Jimmie B. Vaught, Hae Kyung Im, John M. Rouhana, François Aguet, Ferran Reverter, Jason Bridge, Farzana Jasmine, Scott D. Jewell, William F. Leinweber, Gad Getz, Jonah Einson, Kevin Myer, SE Castel, Barbara E. Engelhardt, Stephen B. Montgomery, Brunilda Balliu, Gary Walters, Helen M. Moore, Daniel Nachun, Zerbino, Lori E. Brigham, Gao Wang, Farhad Hormozdiari, Pejman Mohammadi, Kasper D. Hansen, Nicole A. Teran, Fred A. Wright, Bryan Gillard, Sarah Kim-Hellmuth, CC Powell, Susan E. Koester, Wucher, Aaron Graubert, Duyen T. Nguyen, Shin Lin, Mike Moser, John A. Stamatoyannopoulos, Liqun Qi, Princy Parsana, Peter Hickey, Latarsha J. Carithers, Saboor Shad, Eric R. Gamazon, Jennifer A. Doherty, Stephen J. Trevanion, Kane Hadley, Kate R. Rosenbloom, Anita H. Undale, Robert E. Handsaker, Debra Bradbury, Shankara Anand, Meng Wang, David E. Tabor, Karna Robinson, S. Gabriel, Esti Yeger-Lotem, Kimberly Ramsey, Mary Barcus, Daniel G. MacArthur, Yuan He, Nancy Roche, Alvaro N. Barbeira, Ayellet V. Segrè, Dan Sheppard, Souvik Das, AR Little, Nathan S. Abell, Xiaoquan Wen, Elise D. Flynn, Nicole M. Ferraro, Hua Tang, Jared L. Nedzel, Jessica Wheeler, Abhi Rao, Meier, Thomas Juettemann, Sandra Linder, Bruce A. Roe, Daniel J. Cotter, David A. Davis, Christopher Johns, Lin Chen, Seva Kashin, Muhammad G. Kibriya, Ana Viñuela, Ellen Todres, Ashis Saha, Matthew Stephens, Chiara Sabatti, Manolis Kellis, Laura A. Siminoff, Phillip Branton, Xiao Li, Michael Snyder, Kathryn Demanelis, Gen Li, Barbara A. Foster, Leslie H. Sobin, Simona Volpi, Magali Ruffier, Christopher D. Brown, Ping Guan, Benjamin J. Strober, Alexis Battle, Michael J. Gloudemans, Silva Kasela, Manuel Muñoz-Aguirre, Ellen Karasik, OM deGoede, Roderic Guigó, Michael Washington, Alisa McDonald, Andrew A. Brown, Meritxell Oliva, Kieron Taylor, Nancy J. Cox, Daniel C. Rohrer, Paul J. Hoffman, Gene Kopen, Qin Li, Andrew D Skol, Rodrigo Bonazzola, Tiffany Eulalio, Mark H. Johnson, Laure Fresard, Lindsay F. Rizzardi, Abhiram Rao, T Krubit, W. J. Kent, Alan Kwong, Anna M. Smith, Pedro G. Ferreira, HM Gardiner, Andrew P. Feinberg, Rick Hasz, Lei Hou, Marta Melé, Andrew B. Nobel, Katherine H. Huang, Laura Barker, Maximilian Haeussler, Kristin G. Ardlie, Concepcion R. Nierras, Christopher Lee, Joshua M. Akey, Eskin E, Jeffrey McLean, Donald F. Conrad, Jin Billy Li, YoSon Park, Serghei Mangul, Emmanouil T. Dermitzakis, Brian Jo, D Garrido-Martin, and Barcelona Supercomputing Center
- Subjects
Informàtica::Aplicacions de la informàtica::Bioinformàtica [Àrees temàtiques de la UPC] ,0303 health sciences ,Linkage disequilibrium ,Multidisciplinary ,Genotype-Tissue Expression (GTEx) ,Atlas (topology) ,Human tissues ,Cancer susceptibility ,Diseases ,Genome-wide association study ,RNA-Seq ,Computational biology ,Biology ,Expressió gènica ,Human genetics ,03 medical and health sciences ,Tissues ,0302 clinical medicine ,Allelic heterogeneity ,Gene expression ,Genètica ,030217 neurology & neurosurgery ,030304 developmental biology ,Teixits (Histologia) - Abstract
The Genotype-Tissue Expression (GTEx) project dissects how genetic variation affects gene expression and splicing. Some human genetic variants affect the amount of RNA produced and the splicing of gene transcripts, crucial steps in development and maintaining a healthy individual. However, some of these changes only occur in a small number of tissues within the body. The Genotype-Tissue Expression (GTEx) project has been expanded over time, and, looking at the final data in version 8, Aguet et al. present a deep characterization of genetic associations and gene expression and splicing in 838 individuals over 49 tissues (see the Perspective by Wilson). This large study was able to characterize the details underlying many aspects of gene expression and provides a resource with which to better understand the fundamental molecular mechanisms of how genetic variants affect gene regulation and complex traits in humans. Science, this issue p. 1318; see also p. 1298 The Genotype-Tissue Expression (GTEx) project was established to characterize genetic effects on the transcriptome across human tissues and to link these regulatory mechanisms to trait and disease associations. Here, we present analyses of the version 8 data, examining 15,201 RNA-sequencing samples from 49 tissues of 838 postmortem donors. We comprehensively characterize genetic associations for gene expression and splicing in cis and trans, showing that regulatory associations are found for almost all genes, and describe the underlying molecular mechanisms and their contribution to allelic heterogeneity and pleiotropy of complex traits. Leveraging the large diversity of tissues, we provide insights into the tissue specificity of genetic effects and show that cell type composition is a key factor in understanding gene regulatory mechanisms in human tissues. We thank the donors and their families for their generous gifts of organ donation for transplantation and tissue donations for the GTEx research project; the Genomics Platform at the Broad Institute for data generation; J. Struewing for support and leadership of the GTEx project; M. Khan and C. Stolte for the illustrations in Fig. 1; and R. Do, D. Jordan, and M. Verbanck for providing GWAS pleiotropy scores. Funding: This work was supported by the Common Fund of the Office of the Director, U.S. National Institutes of Health, and by NCI, NHGRI, NHLBI, NIDA, NIMH, NIA, NIAID, and NINDS through NIH contracts HHSN261200800001E (Leidos Prime contract with NCI: A.M.S., D.E.T., N.V.R., J.A.M., L.S., M.E.B., L.Q., T.K., D.B., K.R., and A.U.), 10XS170 (NDRI: W.F.L., J.A.T., G.K., A.M., S.S., R.H., G.Wa., M.J., M.Wa., L.E.B., C.J., J.W., B.R., M.Hu., K.M., L.A.S., H.M.G., M.Mo., and L.K.B.), 10XS171 (Roswell Park Cancer Institute: B.A.F., M.T.M., E.K., B.M.G., K.D.R., and J.B.), 10X172 (Science Care Inc.), 12ST1039 (IDOX), 10ST1035 (Van Andel Institute: S.D.J., D.C.R., and D.R.V.), HHSN268201000029C (Broad Institute: F.A., G.G., K.G.A., A.V.S., X.Li., E.T., S.G., A.G., S.A., K.H.H., D.T.N., K.H., S.R.M., and J.L.N.), 5U41HG009494 (F.A., G.G., and K.G.A.), and through NIH grants R01 DA006227-17 (University of Miami Brain Bank: D.C.M. and D.A.D.), Supplement to University of Miami grant DA006227 (D.C.M. and D.A.D.), R01 MH090941 (University of Geneva), R01 MH090951 and R01 MH090937 (University of Chicago), R01 MH090936 (University of North Carolina–Chapel Hill), R01MH101814 (M.M.-A., V.W., S.B.M., R.G., E.T.D., D.G.-M., and A.V.), U01HG007593 (S.B.M.), R01MH101822 (C.D.B.), U01HG007598 (M.O. and B.E.S.), U01MH104393 (A.P.F.), extension H002371 to 5U41HG002371 (W.J.K.), as well as other funding sources: R01MH106842 (T.L., P.M., E.F., and P.J.H.), R01HL142028 (T.L., Si.Ka., and P.J.H.), R01GM122924 (T.L. and S.E.C.), R01MH107666 (H.K.I.), P30DK020595 (H.K.I.), UM1HG008901 (T.L.), R01GM124486 (T.L.), R01HG010067 (Y.Pa.), R01HG002585 (G.Wa. and M.St.), Gordon and Betty Moore Foundation GBMF 4559 (G.Wa. and M.St.), 1K99HG009916-01 (S.E.C.), R01HG006855 (Se.Ka. and R.E.H.), BIO2015-70777-P, Ministerio de Economia y Competitividad and FEDER funds (M.M.-A., V.W., R.G., and D.G.-M.), la Caixa Foundation ID 100010434 under agreement LCF/BQ/SO15/52260001 (D.G.-M.), NIH CTSA grant UL1TR002550-01 (P.M.), Marie-Skłodowska Curie fellowship H2020 Grant 706636 (S.K.-H.), R35HG010718 (E.R.G.), FPU15/03635, Ministerio de Educación, Cultura y Deporte (M.M.-A.),R01MH109905, 1R01HG010480 (A.Ba.), Searle Scholar Program (A.Ba.), R01HG008150 (S.B.M.), 5T32HG000044-22, NHGRI Institutional Training Grant in Genome Science (N.R.G.), EU IMI program (UE7-DIRECT-115317-1) (E.T.D. and A.V.), FNS funded project RNA1 (31003A_149984) (E.T.D. and A.V.), DK110919 (F.H.), F32HG009987 (F.H.), Massachusetts Lions Eye Research Fund Grant (A.R.H.), Wellcome grant WT108749/Z/15/Z (P.F.), and European Molecular Biology Laboratory (P.F. and D.Z.). Peer Reviewed "Article signat per 1 autors/es del BSC membres del THE GTEX CONSORTIUM: Marta Mele Messeguer"
- Published
- 2020
32. The landscape of molecular chaperones across human tissues reveals a layered architecture of core and variable chaperones
- Author
-
Ekaterina Vinogradov, Anat Ben-Zvi, Omer Basha, Shiran Dror, Juman Jubran, Mehtap Abu-Qarn, Idan Hekselman, Netta Shemesh, Eyal Simonovky, Esti Yeger-Lotem, and Serena Carra
- Subjects
Muscle tissue ,Transcriptome ,medicine.anatomical_structure ,biology ,Downregulation and upregulation ,Chaperone (protein) ,medicine ,biology.protein ,Skeletal muscle ,Myocyte ,Core (group theory) ,Gene ,Cell biology - Abstract
The sensitivity of the protein-folding environment to chaperone disruption can be highly tissue-specific. Yet, the organization of the chaperone system across physiological human tissues has received little attention. Here, we used human tissue RNA-sequencing profiles to analyze the expression and organization of chaperones across 29 main tissues. We found that relative to protein-coding genes, chaperones were significantly more ubiquitously and highly expressed across all tissues. Nevertheless, differential expression analysis revealed that most chaperones were up- or down-regulated in certain tissues, suggesting that they have tissue-specific roles. In agreement, chaperones that were upregulated in skeletal muscle were highly enriched in mouse myoblasts and in nematode’s muscle tissue, and overlapped significantly with chaperones that are causal for muscle diseases. We also identified a distinct subset of chaperones that formed a uniformly-expressed, cross-family core group conducting basic cellular functions that was significantly more essential for cell survival. Altogether, this suggests a layered architecture of chaperones across tissues that is composed of shared core elements that are complemented by variable elements which give rise to tissue-specific functions and sensitivities, thereby contributing to the tissue-specificity of protein misfolding diseases.Significance StatementProtein misfolding diseases, such as neurodegenerative disorders and myopathies, are often manifested in a specific tissue or even a specific cell type. Enigmatically, however, they are typically caused by mutations in widely expressed proteins. Here we focused on chaperones, the main and basic components of the protein-folding machinery of cells. Computational analyses of large scale tissue transcriptomes unveils that the chaperone system is composed of core essential elements that are uniformly expressed across tissues, and of variable elements that are differentially expressed in a tissue-specific manner. This organization allows each tissue to fit the quality control system to its specific requirements and illuminates the mechanisms that underlie a tissue’s susceptibility to protein-misfolding diseases.
- Published
- 2020
- Full Text
- View/download PDF
33. A vast resource of allelic expression data spanning human tissues
- Author
-
Nancy J. Cox, Sayantan Das, Abhi Rao, Pejman Mohammadi, Alan Kwong, Brandon L. Pierce, Yanyu Liang, Yuxin Zou, Anna M. Smith, Matthew Stephens, Chiara Sabatti, Yuan He, Kasper D. Hansen, Lei Hou, Meritxell Oliva, W. James Kent, Stacey Gabriel, Andrew R. Hame, Tanya Krubit, Gary Walters, Lori E. Brigham, Gao Wang, Kevin S. Smith, Michael J. Gloudemans, Barbara E. Engelhardt, Yongjin Park, Nicole A. Teran, David A. Davis, Thomas Juettemann, Kimberley Ramsey, Fred A. Wright, Lin Chen, Valentin Wucher, Benjamin J. Strober, Duyen T. Nguyen, Eleazar Eskin, Kane Hadley, Deborah C. Mash, Michael Snyder, Sarah Kim-Hellmuth, Laura A. Siminoff, Maghboeba Mosavel, Shin Lin, Richard Hasz, Daniel C. Rohrer, Latarsha J. Carithers, Kevin Myer, Rajinder Kaul, Andrew D. Skol, Bryan Gillard, Dana R. Valley, Philip A. Branton, Stephane E. Castel, Robert E. Handsaker, Debra Bradbury, Meng Wang, Mary Barcus, Xiaoquan Wen, Hua Tang, Daniel J. Cotter, Lihua Jiang, Jason Bridge, Ashis Saha, Gen Li, Susan E. Koester, Qin Li, Mark H. Johnson, Barbara E. Stranger, Jimmie B. Vaught, Hae Kyung Im, Paul Flicek, Marcus Hunter, François Aguet, Elise D. Flynn, Sandra Linder, Nancy Roche, Daniel R. Zerbino, Xiao Li, Barbara A. Foster, Stephen B. Montgomery, Daniel Nachun, Serghei Mangul, Emmanouil T. Dermitzakis, Brian Jo, Simona Volpi, Farzana Jasmine, Scott D. Jewell, Jonah Einson, Tuuli Lappalainen, Farhad Hormozdiari, John M. Rouhana, Ana Viñuela, Daniel G. MacArthur, William F. Leinweber, Gad Getz, Peter Hickey, Eric R. Gamazon, Brunilda Balliu, Jennifer A. Doherty, Christopher D. Brown, Roderic Guigó, Gene Kopen, Rodrigo Bonazzola, Pedro G. Ferreira, Andrew P. Feinberg, Shankara Anand, Helen M. Moore, Paul J. Hoffman, Heather M. Gardiner, Ping Guan, Ferran Reverter, Jin Billy Li, Tiffany Eulalio, Joseph Wheeler, Alvaro N. Barbeira, Jared L. Nedzel, Seva Kashin, Laure Fresard, Lindsay F. Rizzardi, Abhiram Rao, Muhammad G. Kibriya, David Tabor, Leslie H. Sobin, A. Roger Little, Stephen J. Trevanion, Nicole M. Ferraro, Kate R. Rosenbloom, John A. Stamatoyannopoulos, Liqun Qi, Princy Parsana, Ayellet V. Segrè, Dan Sheppard, Nathan S. Abell, Kathryn Demanelis, Manolis Kellis, Silva Kasela, Xin Li, Conner C. Powell, YoSon Park, Michael Washington, Magali Ruffier, Saboor Shad, Christopher Johns, Jeffrey A. Thomas, Andrew Brown, Alisa McDonald, Karna Robinson, Esti Yeger-Lotem, Manuel Muñoz-Aguirre, Kieron Taylor, Marta Melé, Diego Garrido-Martín, Brian Roe, Michael T. Moser, Andrew B. Nobel, Alexis Battle, Maximilian Haeussler, Concepcion R. Nierras, Ellen Karasik, Sam Meier, Anita H. Undale, Ellen Todres, Aaron Graubert, Joshua M. Akey, Jeffrey McLean, Donald F. Conrad, Olivia M. De Goede, Katherine H. Huang, Laura Barker, Kristin G. Ardlie, and Christopher Lee
- Subjects
animal structures ,Future studies ,lcsh:QH426-470 ,Short Report ,Gene Expression ,Genomics ,Computational biology ,Biology ,eQTL ,Polymorphism, Single Nucleotide ,ASE ,Regulatory variation ,03 medical and health sciences ,0302 clinical medicine ,Resource (project management) ,Humans ,SNP ,Allele ,lcsh:QH301-705.5 ,Alleles ,Allele specific ,030304 developmental biology ,0303 health sciences ,Allelic expression ,Genome, Human ,Sequence Analysis, RNA ,Haplotype ,Functional genomics ,Human genetics ,lcsh:Genetics ,Haplotypes ,lcsh:Biology (General) ,Expression data ,Expression quantitative trait loci ,GTEx ,030217 neurology & neurosurgery - Abstract
Allele expression (AE) analysis robustly measures cis-regulatory effects. Here, we present and demonstrate the utility of a vast AE resource generated from the GTEx v8 release, containing 15,253 samples spanning 54 human tissues for a total of 431 million measurements of AE at the SNP level and 153 million measurements at the haplotype level. In addition, we develop an extension of our tool phASER that allows effect sizes of cis-regulatory variants to be estimated using haplotype-level AE data. This AE resource is the largest to date, and we are able to make haplotype-level data publicly available. We anticipate that the availability of this resource will enable future studies of regulatory variation across human tissues.
- Published
- 2020
34. A reference map of the human binary protein interactome
- Author
-
Eyal Simonovsky, Joseph C. Mellor, Amélie Dricot, Marc Vidal, Liana Goehring, Miquel Duran-Frigola, Florian Goebels, Dayag Sheykhkarimli, Thomas Rolland, Murat Tasan, John Rasla, Steffi De Rouck, Carles Pons, Sadie Schlabach, Yoseph Kassa, Claudia Colabella, Dong-Sic Choi, Yves Jacob, Joseph N. Paulson, Javier De Las Rivas, Madeleine F. Hardy, Francisco J. Campos-Laborie, Xinping Yang, Soon Gang Choi, Frederick P. Roth, Kerstin Spirohn, Nishka Kishore, Luke Lambourne, Cassandra D’Amata, Dawit Balcha, Adriana San-Miguel, Anupama Yadav, Anjali Gopal, Suet-Feung Chin, Suzanne Gaudet, Yang Wang, István Kovács, Elodie Hatchi, Natascha van Lieshout, Michael A. Calderwood, Yu Xia, Gloria M. Sheynkman, Robert J. Weatheritt, Marinella Gebbia, Atina G. Cote, Bridget E. Begg, Mohamed Helmy, Katja Luck, Bridget Teeking, Quan Zhong, Serena Landini, David E. Hill, Sudharshan Rangarajan, Georges Coppin, Ghazal Haddad, Omer Basha, Carl Pollis, Dylan Markey, Alice Desbuleux, Hanane Ennajdaoui, Dae-Kyum Kim, Vincent Tropepe, Roujia Li, Steven Deimling, Jennifer J. Knapp, Jan Tavernier, Mariana Babor, Benoit Charloteaux, Gary D. Bader, Alexander O. Tejeda, Aaron Richardson, Ruth Brignall, Ashyad Rayhan, Irma Lemmens, Tong Hao, Christian Bowman-Colin, Janusz Rak, David De Ridder, Jochen Weile, Wenting Bian, Jean-Claude Twizere, Patrick Aloy, Esti Yeger-Lotem, Meaghan Daley, Tiziana M. Cafarelli, Andrew MacWilliams, Miles W. Mee, Yun Shen, National Institutes of Health (US), National Human Genome Research Institute (US), Canadian Institutes of Health Research, Natural Sciences and Engineering Research Council of Canada, Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), Fonds de la Recherche Scientifique (Fédération Wallonie-Bruxelles), Epidémiologie et Physiopathologie des Virus Oncogènes (EPVO (UMR_3569 / U-Pasteur_3)), Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Institut Pasteur [Paris], Dana-Farber Cancer Institute [Boston], Harvard Medical School [Boston] (HMS), Génétique Moléculaire des Virus à ARN - Molecular Genetics of RNA Viruses (GMV-ARN (UMR_3569 / U-Pasteur_2)), Institut Pasteur [Paris] (IP)-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), University of Toronto, Mount Sinai Health System, Canadian Institute for Advanced Research (CIFAR), and This work was primarily supported by the National Institutes of Health (NIH) National Human Genome Research Institute (NHGRI) grant U41HG001715 (M.V., F.P.R., D.E.H., M.A.C., G.D.B. and J.T.) with additional support from NIH grants P50HG004233 (M.V. and F.P.R.), U01HL098166 (M.V.), U01HG007690 (M.V.), R01GM109199 (M.A.C.), Canadian Institute for Health Research (CIHR) Foundation Grants (F.P.R. and J. Rak), the Canada Excellence Research Chairs Program (F.P.R.) and an American Heart Association grant 15CVGPS23430000 (M.V.). D.-K.K. was supported by a Banting Postdoctoral Fellowship through the Natural Sciences and Engineering Research Council (NSERC) of Canada and by the Basic Science Research Program through the National Research Foundation (NRF) of Korea funded by the Ministry of Education (2017R1A6A3A03004385). C. Pons was supported by a Ramon Cajal fellowship (RYC-2017-22959). G.M.S. was supported by NIH Training Grant T32CA009361. M.V. is a Chercheur Qualifié Honoraire from the Fonds de la Recherche Scientifique (FRS-FNRS, Wallonia-Brussels Federation, Belgium).
- Subjects
0301 basic medicine ,Multidisciplinary ,Proteome ,[SDV]Life Sciences [q-bio] ,Computational biology ,Biology ,Genome ,Phenotype ,Interactome ,[SDV.BIBS]Life Sciences [q-bio]/Quantitative Methods [q-bio.QM] ,Article ,Protein–protein interaction ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Organ Specificity ,Protein Interaction Mapping ,Reference map ,Humans ,Cellular organization ,Extracellular Space ,030217 neurology & neurosurgery ,Function (biology) - Abstract
et al., Global insights into cellular organization and genome function require comprehensive understanding of the interactome networks that mediate genotype–phenotype relationships1,2. Here we present a human ‘all-by-all’ reference interactome map of human binary protein interactions, or ‘HuRI’. With approximately 53,000 protein–protein interactions, HuRI has approximately four times as many such interactions as there are high-quality curated interactions from small-scale studies. The integration of HuRI with genome3, transcriptome4 and proteome5 data enables cellular function to be studied within most physiological or pathological cellular contexts. We demonstrate the utility of HuRI in identifying the specific subcellular roles of protein–protein interactions. Inferred tissue-specific networks reveal general principles for the formation of cellular context-specific functions and elucidate potential molecular mechanisms that might underlie tissue-specific phenotypes of Mendelian diseases. HuRI is a systematic proteome-wide reference that links genomic variation to phenotypic outcomes., This work was primarily supported by the National Institutes of Health (NIH) National Human Genome Research Institute (NHGRI) grant U41HG001715 (M.V., F.P.R., D.E.H., M.A.C., G.D.B. and J.T.) with additional support from NIH grants P50HG004233 (M.V. and F.P.R.), U01HL098166 (M.V.), U01HG007690 (M.V.), R01GM109199 (M.A.C.), Canadian Institute for Health Research (CIHR) Foundation Grants (F.P.R. and J. Rak), the Canada Excellence Research Chairs Program (F.P.R.) and an American Heart Association grant 15CVGPS23430000 (M.V.). D.-K.K. was supported by a Banting Postdoctoral Fellowship through the Natural Sciences and Engineering Research Council (NSERC) of Canada and by the Basic Science Research Program through the National Research Foundation (NRF) of Korea funded by the Ministry of Education (2017R1A6A3A03004385). C. Pons was supported by a Ramon Cajal fellowship (RYC-2017-22959). G.M.S. was supported by NIH Training Grant T32CA009361. M.V. is a Chercheurv Qualifié Honoraire from the Fonds de la Recherche Scientifique (FRS-FNRS, Wallonia-Brussels Federation, Belgium).
- Published
- 2020
35. Identifying cis-mediators for trans-eQTLs across many human tissues using genomic mediation analysis
- Author
-
Eric Gamazon, Martijn Van de Bunt, Roger Little, Jemma Nelson, Thomas Juettemann, Olivier Delaneau, Fan Wu, Magali Ruffier, Halit Ongen, Daniel MacArthur, Daniel Zerbino, Peter Hickey, Yaping Liu, Esti Yeger-Lotem, Rajinder Kaul, Dan Nicolae, David Davis, Ruth Barshir, Michael Sammeth, Diego Garrido-Martín, Jiebiao Wang, Joshua Akey, and GTEx, Consortium
- Subjects
0301 basic medicine ,Gene regulatory network ,Method ,Genome-wide association study ,Medical and Health Sciences ,2.1 Biological and endogenous factors ,Tissue Distribution ,Gene Regulatory Networks ,ddc:576.5 ,Aetiology ,Genetics (clinical) ,Genetics ,0303 health sciences ,030305 genetics & heredity ,Confounding ,Genomics ,Single Nucleotide ,Biological Sciences ,Biotechnology ,Mediation (statistics) ,Bioinformatics ,1.1 Normal biological development and functioning ,Quantitative Trait Loci ,Computational biology ,Quantitative trait locus ,Biology ,GTEx Consortium ,Databases ,03 medical and health sciences ,Genetic ,Underpinning research ,Genetic variation ,Humans ,SNP ,Genetic Predisposition to Disease ,Polymorphism ,Selection ,Gene ,030304 developmental biology ,Mechanism (biology) ,Gene Expression Profiling ,Human Genome ,Gene expression profiling ,Good Health and Well Being ,030104 developmental biology ,Gene Expression Regulation ,Expression quantitative trait loci ,Generic health relevance ,Genome-Wide Association Study - Abstract
The impact of inherited genetic variation on gene expression in humans is well-established. The majority of known expression quantitative trait loci (eQTLs) impact expression of local genes (cis-eQTLs). More research is needed to identify effects of genetic variation on distant genes (trans-eQTLs) and understand their biological mechanisms. One common trans-eQTLs mechanism is “mediation” by a local (cis) transcript. Thus, mediation analysis can be applied to genome-wide SNP and expression data in order to identify transcripts that are “cis-mediators” of trans-eQTLs, including those “cis-hubs” involved in regulation of many trans-genes. Identifying such mediators helps us understand regulatory networks and suggests biological mechanisms underlying trans-eQTLs, both of which are relevant for understanding susceptibility to complex diseases. The multitissue expression data from the Genotype-Tissue Expression (GTEx) program provides a unique opportunity to study cis-mediation across human tissue types. However, the presence of complex hidden confounding effects in biological systems can make mediation analyses challenging and prone to confounding bias, particularly when conducted among diverse samples. To address this problem, we propose a new method: Genomic Mediation analysis with Adaptive Confounding adjustment (GMAC). It enables the search of a very large pool of variables, and adaptively selects potential confounding variables for each mediation test. Analyses of simulated data and GTEx data demonstrate that the adaptive selection of confounders by GMAC improves the power and precision of mediation analysis. Application of GMAC to GTEx data provides new insights into the observed patterns of cis-hubs and trans-eQTL regulation across tissue types.
- Published
- 2017
36. The DifferentialNet database of differential protein–protein interactions in human tissues
- Author
-
Rotem Shpringer, Esti Yeger-Lotem, Omer Basha, and Chanan M Argov
- Subjects
0301 basic medicine ,Male ,Human Protein Atlas ,Biology ,computer.software_genre ,Kidney ,Interactome ,Bone and Bones ,Protein–protein interaction ,03 medical and health sciences ,0302 clinical medicine ,Atlases as Topic ,Protein Interaction Mapping ,Genetics ,Database Issue ,Humans ,Databases, Protein ,Muscle, Skeletal ,Gene ,Lung ,Internet ,Database ,Ovary ,Prostate ,Kidney metabolism ,Brain ,High-Throughput Nucleotide Sequencing ,Proteins ,Phenotype ,030104 developmental biology ,Organ Specificity ,030220 oncology & carcinogenesis ,DECIPHER ,Protein–protein interaction prediction ,Female ,computer ,Software - Abstract
DifferentialNet is a novel database that provides users with differential interactome analysis of human tissues (http://netbio.bgu.ac.il/diffnet/). Users query DifferentialNet by protein, and retrieve its differential protein–protein interactions (PPIs) per tissue via an interactive graphical interface. To compute differential PPIs, we integrated available data of experimentally detected PPIs with RNA-sequencing profiles of tens of human tissues gathered by the Genotype-Tissue Expression consortium (GTEx) and by the Human Protein Atlas (HPA). We associated each PPI with a score that reflects whether its corresponding genes were expressed similarly across tissues, or were up- or down-regulated in the selected tissue. By this, users can identify tissue-specific interactions, filter out PPIs that are relatively stable across tissues, and highlight PPIs that show relative changes across tissues. The differential PPIs can be used to identify tissue-specific processes and to decipher tissue-specific phenotypes. Moreover, they unravel processes that are tissue-wide yet tailored to the specific demands of each tissue.
- Published
- 2017
37. Genetic effects on gene expression across human tissues
- Author
-
Manuel Muñoz-Aguirre, Pejman Mohammadi, Boxiang Liu, Eric Gamazon, Martijn Van de Bunt, Roger Little, Ferran Reverter, Richard Sandstrom, Jemma Nelson, Omer Basha, Thomas Juettemann, Yi-Hui Zhou, Olivier Delaneau, Robert Handsaker, Gerald Quon, Fan Wu, Panagiotis Papasaikas, Magali Ruffier, Halit Ongen, Daniel MacArthur, Daniel Zerbino, Carlos D. Bustamante, Peter Hickey, Pedro Ferreira, Sarah Kim-Hellmuth, Paul Flicek, Yaping Liu, Tuuli Lappalainen, Barbara Engelhardt, Esti Yeger-Lotem, Christine Peterson, Rajinder Kaul, Dan Nicolae, David Davis, Ruth Barshir, Michael Sammeth, Stephen Montgomery, Diego Garrido-Martín, Kasper Hansen, Andrew Brown, Taru Tukiainen, Tyler Shimko, Laure Frésard, Mark McCarthy, Joshua Akey, Brian Jo, Universitat Politècnica de Catalunya. Departament d'Estadística i Investigació Operativa, Brown, Andrew Anand, Delaneau, Olivier, Dermitzakis, Emmanouil, Howald, Cédric, Ongen, Halit, Panousis, Nikolaos, Other departments, Laboratory, Data Analysis &Coordinating Center (LDACC)-Analysis Working Group, and Statistical Methods groups-Analysis Working Group
- Subjects
0301 basic medicine ,Male ,Informàtica::Aplicacions de la informàtica::Bioinformàtica [Àrees temàtiques de la UPC] ,Genotype ,Bioinformatics ,Quantitative trait locus ,Biology ,Gene Expression Regulation/genetics ,Organ Specificity/genetics ,03 medical and health sciences ,Bioinformàtica ,Genetic variation ,Humans ,ddc:576.5 ,Allele ,Transcriptomics ,Gene ,Alleles ,Regulation of gene expression ,Genetics ,Multidisciplinary ,Gene Expression Profiling ,Genetic Variation ,Chromosomes, Human/genetics ,Expressió gènica ,Human genetics ,Gene regulation ,Gene expression profiling ,030104 developmental biology ,Disease/genetics ,Expression quantitative trait loci ,Genome, Human/genetics ,Female ,Gene expression ,Quantitative Trait Loci/genetics - Abstract
Characterization of the molecular function of the human genome and its variation across individuals is essential for identifying the cellular mechanisms that underlie human genetic traits and diseases. The Genotype-Tissue Expression (GTEx) project aims to characterize variation in gene expression levels across individuals and diverse tissues of the human body, many of which are not easily accessible. Here we describe genetic effects on gene expression levels across 44 human tissues. We find that local genetic variation affects gene expression levels for the majority of genes, and we further identify inter-chromosomal genetic effects for 93 genes and 112 loci. On the basis of the identified genetic effects, we characterize patterns of tissue specificity, compare local and distal effects, and evaluate the functional properties of the genetic effects. We also demonstrate that multi-tissue, multi-individual data can be used to identify genes and pathways affected by human disease-associated variation, enabling a mechanistic interpretation of gene regulation and the genetic basis of disease.
- Published
- 2017
38. Aging promotes reorganization of the CD4 T cell landscape toward extreme regulatory and effector phenotypes
- Author
-
Eyal Simonovsky, Idan Hekselman, Kritika Mittal, Nir Friedman, Ekaterina Eremenko, Vered Chalifa-Caspi, Anna Nemirovsky, Esti Yeger-Lotem, Alon Monsonego, Itai Strominger, Omer Berner, Assaf Vital, Inbal Eizenberg-Magar, Maya Schiller, and Yehezqel Elyahu
- Subjects
CD4-Positive T-Lymphocytes ,Aging ,Immunology ,Cell ,Inflammation ,Biology ,Immunomodulation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Single-cell analysis ,T-Lymphocyte Subsets ,Immunity ,medicine ,Animals ,Cytotoxic T cell ,Research Articles ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Sequence Analysis, RNA ,Effector ,Systems Biology ,High-Throughput Nucleotide Sequencing ,SciAdv r-articles ,RNA ,biochemical phenomena, metabolism, and nutrition ,Phenotype ,Cell biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Single-Cell Analysis ,medicine.symptom ,Research Article - Abstract
Single-cell analysis of CD4 T cells reveals a population structure associated with inflammation and declined immunity in aging., Age-associated changes in CD4 T-cell functionality have been linked to chronic inflammation and decreased immunity. However, a detailed characterization of CD4 T cell phenotypes that could explain these dysregulated functional properties is lacking. We used single-cell RNA sequencing and multidimensional protein analyses to profile thousands of CD4 T cells obtained from young and old mice. We found that the landscape of CD4 T cell subsets differs markedly between young and old mice, such that three cell subsets—exhausted, cytotoxic, and activated regulatory T cells (aTregs)—appear rarely in young mice but gradually accumulate with age. Most unexpected were the extreme pro- and anti-inflammatory phenotypes of cytotoxic CD4 T cells and aTregs, respectively. These findings provide a comprehensive view of the dynamic reorganization of the CD4 T cell milieu with age and illuminate dominant subsets associated with chronic inflammation and immunity decline, suggesting new therapeutic avenues for age-related diseases.
- Published
- 2019
39. ResponseNet v.3: revealing signaling and regulatory pathways connecting your proteins and genes across human tissues
- Author
-
Omry Mauer, Eyal Simonovsky, Omer Basha, Esti Yeger-Lotem, and Rotem Shpringer
- Subjects
Internet ,Genome, Human ,Proteins ,Computational biology ,Biology ,Precision medicine ,Interactome ,Genome ,Human genetics ,Transcriptome ,MicroRNAs ,Interaction network ,microRNA ,Web Server Issue ,Genetics ,Humans ,Gene Regulatory Networks ,Protein Interaction Maps ,Databases, Nucleic Acid ,Databases, Protein ,Gene ,Software - Abstract
ResponseNet v.3 is an enhanced version of ResponseNet, a web server that is designed to highlight signaling and regulatory pathways connecting user-defined proteins and genes by using the ResponseNet network optimization approach (http://netbio.bgu.ac.il/respnet). Users run ResponseNet by defining source and target sets of proteins, genes and/or microRNAs, and by specifying a molecular interaction network (interactome). The output of ResponseNet is a sparse, high-probability interactome subnetwork that connects the two sets, thereby revealing additional molecules and interactions that are involved in the studied condition. In recent years, massive efforts were invested in profiling the transcriptomes of human tissues, enabling the inference of human tissue interactomes. ResponseNet v.3 expands ResponseNet2.0 by harnessing ∼11,600 RNA-sequenced human tissue profiles made available by the Genotype-Tissue Expression consortium, to support context-specific analysis of 44 human tissues. Thus, ResponseNet v.3 allows users to illuminate the signaling and regulatory pathways potentially active in the context of a specific tissue, and to compare them with active pathways in other tissues. In the era of precision medicine, such analyses open the door for tissue- and patient-specific analyses of pathways and diseases.
- Published
- 2019
40. Differential network analysis of human tissue interactomes highlights tissue-selective processes and genetic disorder genes
- Author
-
Liad Alfandari, Omer Basha, Idan Hekselman, Yazeed Zoabi, Esti Yeger-Lotem, Vered Chalifa-Caspi, Raviv Artzy, and Chanan M Argov
- Subjects
Heart tissues ,Benchmark (computing) ,Genetic disorder ,medicine ,Computational biology ,Biology ,medicine.disease ,Subnetwork ,Gene ,Differential (mathematics) ,Biological network ,Network analysis - Abstract
MotivationDifferential network analysis, designed to highlight interaction changes between conditions, is an important paradigm in network biology. However, network analysis methods have been typically designed to compare between few conditions, were rarely applied to protein interaction networks (interactomes). Moreover, large-scale benchmarks for their evaluation have been lacking.ResultsHere, we assess five network analysis methods by applying them to 34 human tissues interactomes. For this, we created a manually-curated benchmark of 6,499 tissue-specific, gene ontology biological processes, and analyzed the ability of each method to expose these tissue-process associations. The four differential network analysis methods outperformed the non-differential, expression-based method (AUCs of 0.82-0.9 versus 0.69, respectively). We then created another benchmark, of 1,527 tissue-specific disease cases, and analyzed the ability of differential network analysis methods to highlight additional disease-related genes. Compared to a non-differential subnetworks surrounding a known disease-causing gene, the extremely-differential subnetwork (top 1%) was significantly enriched for additional disease-causing genes in 18.6% of the cases (p≤10e-3). In 5/10 tissues tested, including Muscle, nerve and heart tissues (p = 2.54E-05, 2.71E-04, 3.63E-19), such enrichments were highly significant.SummaryAltogether, our study demonstrates that differential network analysis of human tissue interactomes is a powerful tool for highlighting processes and genes with tissue-selective functionality and clinical impact. Moreover, it offers expansive manually-curated datasets of tissue-selective processes and diseases that could serve for benchmark and for analyses in many other studies.Contactestiyl@bgu.ac.il
- Published
- 2019
41. A reference map of the human protein interactome
- Author
-
Madeleine F. Hardy, Bridget Teeking, Francisco J. Campos-Laborie, Dayag Sheykhkarimli, Dawit Balcha, Anjali Gopal, Marinella Gebbia, Ashyad Rayhan, Carles Pons, Gloria M. Sheynkman, Yves Jacob, Suzanne Gaudet, Aaron Richardson, Yoseph Kassa, Elodie Hatchi, Kerstin Spirohn, Dong-Sic Choi, Yu Xia, Eyal Simonovsky, David E. Hill, Hanane Ennajdaoui, Steven Deimling, Joseph N. Paulson, Natascha van Lieshout, Vincent Tropepe, Michael A. Calderwood, István Kovács, Gary D. Bader, Luke Lambourne, Sudharshan Rangarajan, Tiziana M. Cafarelli, Carl Pollis, Suet-Feung Chin, Alice Desbuleux, Andrew MacWilliams, Amélie Dricot, Jean-Claude Twizere, Patrick Aloy, Atina G. Cote, Marc Vidal, Jan Tavernier, Javier De Las Rivas, Cassandra D’Amata, Alexander O. Tejeda, Esti Yeger-Lotem, Liana Goehring, Joseph C. Mellor, Meaghan Daley, Irma Lemmens, Soon Gang Choi, Christian Bowman-Colin, Ghazal Haddad, Janusz Rak, Florian Goebels, Robert J. Weatheritt, Mariana Babor, Yang Wang, Thomas Rolland, Steffi De Rouck, Jochen Weile, Serena Landini, John Rasla, Sadie Schlabach, Nishka Kishore, Bridget E. Begg, Ruth Brignall, Quan Zhong, Tong Hao, David De Ridder, Claudia Colabella, Frederick P. Roth, Anupama Yadav, Mohamed Helmy, Katja Luck, Omer Basha, Dae-Kyum Kim, Benoit Charloteaux, Georges Coppin, Dylan Markey, Roujia Li, Miquel Duran-Frigola, Adriana San-Miguel, Wenting Bian, Miles W. Mee, Jennifer J. Knapp, Yun Shen, Murat Tasan, Xinping Yang, Dana-Farber Cancer Institute [Boston], Division of Medical Physics in Radiology [Heidelberg], German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), Barcelona Supercomputing Center - Centro Nacional de Supercomputacion (BSC - CNS), Cancer Research UK Cambridge Institute (CRUK), University of Cambridge [UK] (CAM), Génétique Moléculaire des Virus à ARN - Molecular Genetics of RNA Viruses (GMV-ARN (UMR_3569 / U-Pasteur_2)), Centre National de la Recherche Scientifique (CNRS)-Université Paris Diderot - Paris 7 (UPD7)-Institut Pasteur [Paris], Harvard Medical School [Boston] (HMS), University of Toronto, Universidad de Salamanca, McGill University Health Center [Montreal] (MUHC), Mount Sinai Hospital [Toronto, Canada] (MSH), Université de Liège, Wigner Research Centre for Physics [Budapest], Hungarian Academy of Sciences (MTA), Northeastern University [Boston], Vlaams Instituut voor Biotechnologie [Ghent, Belgique] (VIB), Universiteit Gent = Ghent University [Belgium] (UGENT), Barcelona Institute of Science and Technology (BIST), Ben-Gurion University of the Negev (BGU), Università degli Studi di Perugia (UNIPG), Institut Pasteur [Paris]-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Canadian Institute for Advanced Research (CIFAR), Universiteit Gent = Ghent University (UGENT), Università degli Studi di Perugia = University of Perugia (UNIPG), and Institut Pasteur [Paris] (IP)-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0303 health sciences ,Context (language use) ,Computational biology ,Biology ,[SDV.BIBS]Life Sciences [q-bio]/Quantitative Methods [q-bio.QM] ,Genome ,Interactome ,Human genetics ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Human interactome ,Proteome ,030217 neurology & neurosurgery ,Function (biology) ,030304 developmental biology - Abstract
Global insights into cellular organization and function require comprehensive understanding of interactome networks. Similar to how a reference genome sequence revolutionized human genetics, a reference map of the human interactome network is critical to fully understand genotype-phenotype relationships. Here we present the first human “all-by-all” binary reference interactome map, or “HuRI”. With ~53,000 high-quality protein-protein interactions (PPIs), HuRI is approximately four times larger than the information curated from small-scale studies available in the literature. Integrating HuRI with genome, transcriptome and proteome data enables the study of cellular function within essentially any physiological or pathological cellular context. We demonstrate the use of HuRI in identifying specific subcellular roles of PPIs and protein function modulation via splicing during brain development. Inferred tissue-specific networks reveal general principles for the formation of cellular context-specific functions and elucidate potential molecular mechanisms underlying tissue-specific phenotypes of Mendelian diseases. HuRI thus represents an unprecedented, systematic reference linking genomic variation to phenotypic outcomes.
- Published
- 2019
42. Population-scale tissue transcriptomics maps long non-coding RNAs to complex disease
- Author
-
Olivia M. de Goede, Daniel C. Nachun, Nicole M. Ferraro, Michael J. Gloudemans, Abhiram S. Rao, Craig Smail, Tiffany Y. Eulalio, François Aguet, Bernard Ng, Jishu Xu, Alvaro N. Barbeira, Stephane E. Castel, Sarah Kim-Hellmuth, YoSon Park, Alexandra J. Scott, Benjamin J. Strober, Christopher D. Brown, Xiaoquan Wen, Ira M. Hall, Alexis Battle, Tuuli Lappalainen, Hae Kyung Im, Kristin G. Ardlie, Sara Mostafavi, Thomas Quertermous, Karla Kirkegaard, Stephen B. Montgomery, Shankara Anand, Stacey Gabriel, Gad A. Getz, Aaron Graubert, Kane Hadley, Robert E. Handsaker, Katherine H. Huang, Xiao Li, Daniel G. MacArthur, Samuel R. Meier, Jared L. Nedzel, Duyen T. Nguyen, Ayellet V. Segrè, Ellen Todres, Brunilda Balliu, Rodrigo Bonazzola, Andrew Brown, Donald F. Conrad, Daniel J. Cotter, Nancy Cox, Sayantan Das, Emmanouil T. Dermitzakis, Jonah Einson, Barbara E. Engelhardt, Eleazar Eskin, Elise D. Flynn, Laure Fresard, Eric R. Gamazon, Diego Garrido-Martín, Nicole R. Gay, Roderic Guigó, Andrew R. Hamel, Yuan He, Paul J. Hoffman, Farhad Hormozdiari, Lei Hou, Brian Jo, Silva Kasela, Seva Kashin, Manolis Kellis, Alan Kwong, Xin Li, Yanyu Liang, Serghei Mangul, Pejman Mohammadi, Manuel Muñoz-Aguirre, Andrew B. Nobel, Meritxell Oliva, Yongjin Park, Princy Parsana, Ferran Reverter, John M. Rouhana, Chiara Sabatti, Ashis Saha, Matthew Stephens, Barbara E. Stranger, Nicole A. Teran, Ana Viñuela, Gao Wang, Fred Wright, Valentin Wucher, Yuxin Zou, Pedro G. Ferreira, Gen Li, Marta Melé, Esti Yeger-Lotem, Debra Bradbury, Tanya Krubit, Jeffrey A. McLean, Liqun Qi, Karna Robinson, Nancy V. Roche, Anna M. Smith, David E. Tabor, Anita Undale, Jason Bridge, Lori E. Brigham, Barbara A. Foster, Bryan M. Gillard, Richard Hasz, Marcus Hunter, Christopher Johns, Mark Johnson, Ellen Karasik, Gene Kopen, William F. Leinweber, Alisa McDonald, Michael T. Moser, Kevin Myer, Kimberley D. Ramsey, Brian Roe, Saboor Shad, Jeffrey A. Thomas, Gary Walters, Michael Washington, Joseph Wheeler, Scott D. Jewell, Daniel C. Rohrer, Dana R. Valley, David A. Davis, Deborah C. Mash, Mary E. Barcus, Philip A. Branton, Leslie Sobin, Laura K. Barker, Heather M. Gardiner, Maghboeba Mosavel, Laura A. Siminoff, Paul Flicek, Maximilian Haeussler, Thomas Juettemann, W. James Kent, Christopher M. Lee, Conner C. Powell, Kate R. Rosenbloom, Magali Ruffier, Dan Sheppard, Kieron Taylor, Stephen J. Trevanion, Daniel R. Zerbino, Nathan S. Abell, Joshua Akey, Lin Chen, Kathryn Demanelis, Jennifer A. Doherty, Andrew P. Feinberg, Kasper D. Hansen, Peter F. Hickey, Farzana Jasmine, Lihua Jiang, Rajinder Kaul, Muhammad G. Kibriya, Jin Billy Li, Qin Li, Shin Lin, Sandra E. Linder, Brandon L. Pierce, Lindsay F. Rizzardi, Andrew D. Skol, Kevin S. Smith, Michael Snyder, John Stamatoyannopoulos, Hua Tang, Meng Wang, Latarsha J. Carithers, Ping Guan, Susan E. Koester, A. Roger Little, Helen M. Moore, Concepcion R. Nierras, Abhi K. Rao, Jimmie B. Vaught, and Simona Volpi
- Subjects
Multifactorial Inheritance ,Population ,Quantitative Trait Loci ,Coronary Artery Disease ,Disease ,Computational biology ,Quantitative trait locus ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Humans ,education ,Gene ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,Gene Expression Profiling ,Genetic Variation ,Inflammatory Bowel Diseases ,Long non-coding RNA ,Diabetes Mellitus, Type 1 ,Diabetes Mellitus, Type 2 ,Organ Specificity ,Expression quantitative trait loci ,Trait ,RNA, Long Noncoding ,030217 neurology & neurosurgery - Abstract
Long non-coding RNA (lncRNA) genes have well-established and important impacts on molecular and cellular functions. However, among the thousands of lncRNA genes, it is still a major challenge to identify the subset with disease or trait relevance. To systematically characterize these lncRNA genes, we used Genotype Tissue Expression (GTEx) project v8 genetic and multi-tissue transcriptomic data to profile the expression, genetic regulation, cellular contexts, and trait associations of 14,100 lncRNA genes across 49 tissues for 101 distinct complex genetic traits. Using these approaches, we identified 1,432 lncRNA gene-trait associations, 800 of which were not explained by stronger effects of neighboring protein-coding genes. This included associations between lncRNA quantitative trait loci and inflammatory bowel disease, type 1 and type 2 diabetes, and coronary artery disease, as well as rare variant associations to body mass index.
- Published
- 2021
43. The TissueNet v.2 database: A quantitative view of protein-protein interactions across human tissues
- Author
-
Eugene Lerman, Moran Sharon, Ruth Barshir, Idan Hekselman, Binyamin F. Kirson, Omer Basha, and Esti Yeger-Lotem
- Subjects
0301 basic medicine ,Molecular interactions ,Database ,Computational Biology ,A protein ,Binding (Molecular Function) ,Biology ,computer.software_genre ,Phenotype ,Protein–protein interaction ,03 medical and health sciences ,030104 developmental biology ,Organ Specificity ,Expression data ,Protein Interaction Mapping ,Genetics ,Database Issue ,Humans ,Databases, Protein ,Human proteins ,computer ,Software - Abstract
Knowledge of the molecular interactions of human proteins within tissues is important for identifying their tissue-specific roles and for shedding light on tissue phenotypes. However, many protein-protein interactions (PPIs) have no tissue-contexts. The TissueNet database bridges this gap by associating experimentally-identified PPIs with human tissues that were shown to express both pair-mates. Users can select a protein and a tissue, and obtain a network view of the query protein and its tissue-associated PPIs. TissueNet v.2 is an updated version of the TissueNet database previously featured in NAR. It includes over 40 human tissues profiled via RNA-sequencing or protein-based assays. Users can select their preferred expression data source and interactively set the expression threshold for determining tissue-association. The output of TissueNet v.2 emphasizes qualitative and quantitative features of query proteins and their PPIs. The tissue-specificity view highlights tissue-specific and globally-expressed proteins, and the quantitative view highlights proteins that were differentially expressed in the selected tissue relative to all other tissues. Together, these views allow users to quickly assess the unique versus global functionality of query proteins. Thus, TissueNet v.2 offers an extensive, quantitative and user-friendly interface to study the roles of human proteins across tissues. TissueNet v.2 is available at http://netbio.bgu.ac.il/tissuenet.
- Published
- 2016
44. A Quantitative Proteome Map of the Human Body
- Author
-
Lihua Jiang, Meng Wang, Shin Lin, Ruiqi Jian, Xiao Li, Joanne Chan, Guanlan Dong, Huaying Fang, Aaron E. Robinson, Michael P. Snyder, François Aguet, Shankara Anand, Kristin G. Ardlie, Stacey Gabriel, Gad Getz, Aaron Graubert, Kane Hadley, Robert E. Handsaker, Katherine H. Huang, Seva Kashin, Daniel G. MacArthur, Samuel R. Meier, Jared L. Nedzel, Duyen Y. Nguyen, Ayellet V. Segrè, Ellen Todres, Brunilda Balliu, Alvaro N. Barbeira, Alexis Battle, Rodrigo Bonazzola, Andrew Brown, Christopher D. Brown, Stephane E. Castel, Don Conrad, Daniel J. Cotter, Nancy Cox, Sayantan Das, Olivia M. de Goede, Emmanouil T. Dermitzakis, Barbara E. Engelhardt, Eleazar Eskin, Tiffany Y. Eulalio, Nicole M. Ferraro, Elise Flynn, Laure Fresard, Eric R. Gamazon, Diego Garrido-Martín, Nicole R. Gay, Roderic Guigó, Andrew R. Hamel, Yuan He, Paul J. Hoffman, Farhad Hormozdiari, Lei Hou, Hae Kyung Im, Brian Jo, Silva Kasela, Manolis Kellis, Sarah Kim-Hellmuth, Alan Kwong, Tuuli Lappalainen, Xin Li, Yanyu Liang, Serghei Mangul, Pejman Mohammadi, Stephen B. Montgomery, Manuel Muñoz-Aguirre, Daniel C. Nachun, Andrew B. Nobel, Meritxell Oliva, YoSon Park, Yongjin Park, Princy Parsana, Ferran Reverter, John M. Rouhana, Chiara Sabatti, Ashis Saha, Andrew D. Skol, Matthew Stephens, Barbara E. Stranger, Benjamin J. Strober, Nicole A. Teran, Ana Viñuela, Gao Wang, Xiaoquan Wen, Fred Wright, Valentin Wucher, Yuxin Zou, Pedro G. Ferreira, Gen Li, Marta Melé, Esti Yeger-Lotem, Mary E. Barcus, Debra Bradbury, Tanya Krubit, Jeffrey A. McLean, Liqun Qi, Karna Robinson, Nancy V. Roche, Anna M. Smith, Leslie Sobin, David E. Tabor, Anita Undale, Jason Bridge, Lori E. Brigham, Barbara A. Foster, Bryan M. Gillard, Richard Hasz, Marcus Hunter, Christopher Johns, Mark Johnson, Ellen Karasik, Gene Kopen, William F. Leinweber, Alisa McDonald, Michael T. Moser, Kevin Myer, Kimberley D. Ramsey, Brian Roe, Saboor Shad, Jeffrey A. Thomas, Gary Walters, Michael Washington, Joseph Wheeler, Scott D. Jewell, Daniel C. Rohrer, Dana R. Valley, David A. Davis, Deborah C. Mash, Philip A. Branton, Laura K. Barker, Heather M. Gardiner, Maghboeba Mosavel, Laura A. Siminoff, Paul Flicek, Maximilian Haeussler, Thomas Juettemann, W. James Kent, Christopher M. Lee, Conner C. Powell, Kate R. Rosenbloom, Magali Ruffier, Dan Sheppard, Kieron Taylor, Stephen J. Trevanion, Daniel R. Zerbino, Nathan S. Abell, Joshua Akey, Lin Chen, Kathryn Demanelis, Jennifer A. Doherty, Andrew P. Feinberg, Kasper D. Hansen, Peter F. Hickey, Farzana Jasmine, Rajinder Kaul, Muhammad G. Kibriya, Jin Billy Li, Qin Li, Sandra E. Linder, Brandon L. Pierce, Lindsay F. Rizzardi, Kevin S. Smith, John Stamatoyannopoulos, Hua Tang, Latarsha J. Carithers, Ping Guan, Susan E. Koester, A. Roger Little, Helen M. Moore, Concepcion R. Nierras, Abhi K. Rao, Jimmie B. Vaught, and Simona Volpi
- Subjects
Proteomics ,Cell signaling ,Proteome ,Quantitative proteomics ,Gene Expression ,Disease ,Computational biology ,Biology ,ENCODE ,Article ,General Biochemistry, Genetics and Molecular Biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Secretion ,RNA, Messenger ,Gene ,030304 developmental biology ,0303 health sciences ,Gene Expression Profiling ,RNA ,Metabolism ,Phenotype ,Secretory protein ,030217 neurology & neurosurgery - Abstract
Determining protein levels in each tissue and how they compare with RNA levels is important for understanding human biology and disease as well as regulatory processes that control protein levels. We quantified the relative protein levels from 12,627 genes across 32 normal human tissue types prepared by the GTEx project. Known and new tissue specific or enriched proteins (5,499) were identified and compared to transcriptome data. Many ubiquitous transcripts are found to encode highly tissue specific proteins. Discordance in the sites of RNA expression and protein detection also revealed potential sites of synthesis and action of protein signaling molecules. Overall, these results provide an extraordinary resource, and demonstrate that understanding protein levels can provide insights into metabolism, regulation, secretome, and human diseases.SummaryQuantitative proteome study of 32 human tissues and integrated analysis with transcriptome data revealed that understanding protein levels could provide in-depth knowledge to post transcriptional or translational regulations, human metabolism, secretome, and diseases.
- Published
- 2020
45. MyProteinNet: build up-to-date protein interaction networks for organisms, tissues and user-defined contexts
- Author
-
Omer Basha, Ruth Barshir, Ilan Smoly, Dvir Flom, Shoval Tirman, and Esti Yeger-Lotem
- Subjects
Protein structure database ,Internet ,Web server ,Gene Expression Profiling ,Molecular Sequence Annotation ,Computational biology ,Biology ,Bioinformatics ,computer.software_genre ,Interactome ,Protein–protein interaction ,Identification (information) ,Gene Ontology ,Protein Annotation ,Interaction network ,Protein Interaction Mapping ,Genetics ,Humans ,Web Server issue ,Databases, Protein ,computer ,Software - Abstract
The identification of the molecular pathways active in specific contexts, such as disease states or drug responses, often requires an extensive view of the potential interactions between a subset of proteins. This view is not easily obtained: it requires the integration of context-specific protein list or expression data with up-to-date data of protein interactions that are typically spread across multiple databases. The MyProteinNet web server allows users to easily create such context-sensitive protein interaction networks. Users can automatically gather and consolidate data from up to 11 different databases to create a generic protein interaction network (interactome). They can score the interactions based on reliability and filter them by user-defined contexts including molecular expression and protein annotation. The output of MyProteinNet includes the generic and filtered interactome files, together with a summary of their network attributes. MyProteinNet is particularly geared toward building human tissue interactomes, by maintaining tissue expression profiles from multiple resources. The ability of MyProteinNet to facilitate the construction of up-to-date, context-specific interactomes and its applicability to 11 different organisms and to tens of human tissues, make it a powerful tool in meaningful analysis of protein networks. MyProteinNet is available at http://netbio.bgu.ac.il/myproteinnet.
- Published
- 2015
46. Co-expression networks reveal the tissue-specific regulation of transcription and splicing
- Author
-
Morgan Diegel, Laure Fresard, Lindsay F. Rizzardi, Yuan He, Monkol Lek, Daniel C. Rohrer, Boxiang Liu, Maximilian Haeussler, Heather M. Traino, Concepcion R. Nierras, Joseph Wheeler, Serghei Mangul, Fan Wu, Hualin S. Xi, Andrew D. Skol, Steven Hunter, Yaping Liu, Casandra A. Trowbridge, Brandon L. Pierce, Daniel Bates, Peter Hickey, Susan E. Koester, Bryan Gillard, Eric R. Gamazon, Jennifer A. Doherty, Jared L. Nedzel, Eric Haugen, Lori E. Brigham, Gao Wang, Dana R. Valley, Zachary Zappala, Emmanouil T. Dermitzakis, Seva Kashin, Ira M. Hall, John Vivian, Philip A. Branton, Barbara E. Stranger, Magali Ruffier, Melina Claussnitzer, Nancy Roche, Michael Washington, Halit Ongen, Brian Jo, Rachna Kumar, Jean Monlong, Yi-Hui Zhou, Kristen Lee, Stephane E. Castel, Mark Miklos, Alisa McDonald, Diego Garrido-Martín, Jimmie B. Vaught, Hae Kyung Im, Leslie H. Sobin, John T. Lonsdale, Audra K. Johnson, Rui Zhang, Nancy J. Cox, Christopher D. Brown, Paul Flicek, Ferran Reverter, Roderic Guigó, Tuuli Lappalainen, Sarah E. Gould, Deborah C. Mash, Michael T. Moser, Andrew B. Nobel, Takunda Matose, Jingchun Zhu, Joe R. Davis, Andrey A. Shabalin, Jie Quan, Pedro G. Ferreira, Taru Tukiainen, Ellen Gelfand, Cédric Howald, Buhm Han, Emily K. Tsang, Andrew P. Feinberg, Caroline Linke, Kane Hadley, Richard Sandstrom, Mark D. Johnson, Joshua M. Akey, Ian C. McDowell, Daniel R. Zerbino, Alexis Battle, Brian Roe, Daniel G. MacArthur, Ellen Karasik, Marcus Hunter, Anjené M. Addington, Thomas Juettemann, Konrad J. Karczewski, Duyen T. Nguyen, Lei Hou, Stephen B. Montgomery, YoSon Park, Nicole C. Lockart, Lin Chen, Rajinder Kaul, Ruiqi Jian, Robert G. Montroy, Xiao Li, Michael Snyder, Beryl B. Cummings, Kimberly M. Valentino, Ariel D. H. Gewirtz, François Aguet, Jeffrey McLean, Gary Walters, Farhad Hormozdiari, William F. Leinweber, Gad Getz, Jeffery P. Struewing, Anne Ndungu, Dan L. Nicolae, Benoit Molinie, Lihua Jiang, Michael Sammeth, W. James Kent, John Palowitch, Brian Craft, Donald F. Conrad, Kathryn Demanelis, Jason Bridge, Jin Billy Li, A. Roger Little, Nicholas Van Wittenberghe, Stephen J. Trevanion, Pejman Mohammadi, Michael S. Noble, Kate R. Rosenbloom, Marian S. Fernando, Benjamin J. Strober, Ping Guan, Brunilda Balliu, Yungil Kim, Kevin Myer, Christine B. Peterson, Pushpa Hariharan, Jae Hoon Sul, Abhi Rao, Michael F. Salvatore, Qin Li, Eun Yong Kang, Matthew T. Maurano, Ayellet V. Segrè, Dan Sheppard, Fred A. Wright, Matthew Stephens, Kasper D. Hansen, Chiara Sabatti, Kevin S. Smith, Xin Li, Ruth Barshir, Muhammad G. Kibriya, Farhan N. Damani, Manolis Kellis, Olivier Delaneau, Shin Lin, Richard Hasz, Michael J. Gloudemans, Anita H. Undale, Mary Goldman, Fidencio J. Neri, Katherine H. Huang, David E. Tabor, Manuel Muñoz-Aguirre, Maghboeba Mosavel, Simona Volpi, Latarsha J. Carithers, Anna M. Smith, Genna Gliner, Eleazar Eskin, Nikolaos I Panousis, Benedict Paten, Andrew A. Brown, Jessica Lin, Kieron Taylor, Robert E. Handsaker, Laura Barker, Casey Martin, Meng Wang, Farzana Jasmine, Scott D. Jewell, Nathan S. Abell, Kristin G. Ardlie, Shilpi Singh, Mary Barcus, Anthony Payne, Christopher Lee, Xiaoquan Wen, Nicola J. Rinaldi, Hua Tang, Yongjin Park, Christopher Johns, Saboor Shad, Judith B. Zaugg, Reza Sodaei, Maria M. Tomaszewski, David A. Davis, Joanne Chan, Laura A. Siminoff, Mark I. McCarthy, Ki Sung Um, Karna Robinson, Esti Yeger-Lotem, Martijn van de Bunt, Meritxell Oliva, Jemma Nelson, Negin Vatanian, Colby Chiang, Jeffrey A. Thomas, Alexandra J. Scott, Omer Basha, Jessica Halow, Panagiotis Papasaikas, Barbara A. Foster, Barbara E. Engelhardt, Sarah Kim-Hellmuth, Li Wang, Gireesh K. Bogu, Sandra Linder, Sarah Urbut, Ashis Saha, Gen Li, Bernadette Mestichelli, Chuan Gao, John A. Stamatoyannopoulos, Liqun Qi, Princy Parsana, Helen M. Moore, Gene Kopen, and GTEx, Consortium
- Subjects
Gene isoform ,0301 basic medicine ,Genotyping Techniques ,Bioinformatics ,RNA Splicing ,1.1 Normal biological development and functioning ,Gene regulatory network ,Method ,Genomics ,Computational biology ,Biology ,Medical and Health Sciences ,GTEx Consortium ,Transcriptome ,03 medical and health sciences ,Databases ,0302 clinical medicine ,Genetic ,Transcription (biology) ,Underpinning research ,Genetic variation ,Gene expression ,Genetics ,Humans ,ddc:576.5 ,Gene Regulatory Networks ,Polymorphism ,Gene ,Genetics (clinical) ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Gene Expression Profiling ,Human Genome ,Bayes Theorem ,Single Nucleotide ,Biological Sciences ,Gene expression profiling ,030104 developmental biology ,Gene Expression Regulation ,Organ Specificity ,RNA splicing ,RNA ,Generic health relevance ,Sequence Analysis ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Gene co-expression networks capture biologically important patterns in gene expression data, enabling functional analyses of genes, discovery of biomarkers, and interpretation of regulatory genetic variants. Most network analyses to date have been limited to assessing correlation between total gene expression levels in a single or small sets of tissues. Here, we have reconstructed networks that capture a much more complete set of regulatory relationships, specifically including regulation of relative isoform abundance and splicing, and tissue-specific connections unique to each of a diverse set of tissues. Using the Genotype-Tissue Expression (GTEx) project v6 RNA-sequencing data across 44 tissues in 449 individuals, we evaluated shared and tissue-specific network relationships. First, we developed a framework called Transcriptome Wide Networks (TWNs) for combining total expression and relative isoform levels into a single sparse network, capturing the complex interplay between the regulation of splicing and transcription. We built TWNs for sixteen tissues, and found that hubs with isoform node neighbors in these networks were strongly enriched for splicing and RNA binding genes, demonstrating their utility in unraveling regulation of splicing in the human transcriptome, and providing a set of candidate shared and tissue-specific regulatory hub genes. Next, we used a Bayesian biclustering model that identifies network edges between genes with co-expression in a single tissue to reconstruct tissue-specific networks (TSNs) for 27 distinct GTEx tissues and for four subsets of related tissues. Using both TWNs and TSNs, we characterized gene co-expression patterns shared across tissues. Finally, we found genetic variants associated with multiple neighboring nodes in our networks, supporting the estimated network structures and identifying 33 genetic variants with distant regulatory impact on transcription and splicing. Our networks provide an improved understanding of the complex relationships between genes in the human transcriptome, including tissue-specificity of gene co-expression, regulation of splicing, and the coordinated impact of genetic variation on transcription.
- Published
- 2017
47. RSRC1 mutation affects intellect and behaviour through aberrant splicing and transcription, downregulating IGFBP3
- Author
-
Ohad S. Birk, Gal Meiri, Tatiana Rabinski, Esti Yeger-Lotem, Zamir Shorer, Yonatan Perez, Shay Menascu, Hila Romi, Idan Cohen, Rotem Kadir, Omer Basha, and Rivka Ofir
- Subjects
0301 basic medicine ,Male ,Pluripotent Stem Cells ,Developmental Disabilities ,Down-Regulation ,Rett syndrome ,Biology ,MECP2 ,Small hairpin RNA ,03 medical and health sciences ,Consanguinity ,Mice ,Intellectual Disability ,medicine ,Gene silencing ,Animals ,Humans ,RNA, Small Interfering ,Induced pluripotent stem cell ,Child ,Cell Line, Transformed ,Mice, Knockout ,Alternative splicing ,Infant ,Nuclear Proteins ,Cell Differentiation ,medicine.disease ,Hypotonia ,Cell biology ,Alternative Splicing ,030104 developmental biology ,Gene Ontology ,Insulin-Like Growth Factor Binding Protein 3 ,Child, Preschool ,RNA splicing ,Female ,Neurology (clinical) ,medicine.symptom ,Follow-Up Studies - Abstract
RSRC1, whose polymorphism is associated with altered brain function in schizophrenia, is a member of the serine and arginine rich-related protein family. Through homozygosity mapping and whole exome sequencing we show that RSRC1 mutation causes an autosomal recessive syndrome of intellectual disability, aberrant behaviour, hypotonia and mild facial dysmorphism with normal brain MRI. Further, we show that RSRC1 is ubiquitously expressed, and that the RSRC1 mutation triggers nonsense-mediated mRNA decay of the RSRC1 transcript in patients' fibroblasts. Short hairpin RNA (shRNA)-mediated lentiviral silencing and overexpression of RSRC1 in SH-SY5Y cells demonstrated that RSRC1 has a role in alternative splicing and transcription regulation. Transcriptome profiling of RSRC1-silenced cells unravelled specific differentially expressed genes previously associated with intellectual disability, hypotonia and schizophrenia, relevant to the disease phenotype. Protein-protein interaction network modelling suggested possible intermediate interactions by which RSRC1 affects gene-specific differential expression. Patient-derived induced pluripotent stem cells, differentiated into neural progenitor cells, showed expression dynamics similar to the RSRC1-silenced SH-SY5Y model. Notably, patient neural progenitor cells had 9.6-fold downregulated expression of IGFBP3, whose brain expression is affected by MECP2, aberrant in Rett syndrome. Interestingly, Igfbp3-null mice have behavioural impairment, abnormal synaptic function and monoaminergic neurotransmission, likely correlating with the disease phenotype.
- Published
- 2017
48. Role of duplicate genes in determining the tissue-selectivity of hereditary diseases
- Author
-
Idan Hekselman, Esti Yeger-Lotem, Moran Sharon, Lena Novack, Ruth Barshir, and Netta Shemesh
- Subjects
0301 basic medicine ,Cancer Research ,Gene Dosage ,Gene Expression ,Disease ,Biochemistry ,Germline ,Gene Duplication ,Gene duplication ,Gene expression ,Medicine and Health Sciences ,Tissue Distribution ,Musculoskeletal System ,Genetics (clinical) ,Thyroid ,Genetics ,Brain Diseases ,Muscles ,Liver Diseases ,Germline Mutation ,Phenotype ,Neurology ,Organ Specificity ,Anatomy ,Research Article ,animal structures ,lcsh:QH426-470 ,Systems biology ,Endocrine System ,Context (language use) ,Dermatology ,Gastroenterology and Hepatology ,Biology ,Skin Diseases ,Gene dosage ,03 medical and health sciences ,Germline mutation ,Genes, Duplicate ,Humans ,Genetic Predisposition to Disease ,Pharmacokinetics ,Molecular Biology ,Gene ,Ecology, Evolution, Behavior and Systematics ,Pharmacology ,Gene Expression Profiling ,fungi ,Genetic Diseases, Inborn ,Biology and Life Sciences ,Human genetics ,Gene expression profiling ,lcsh:Genetics ,030104 developmental biology ,Skeletal Muscles ,Mutation ,Hereditary Diseases - Abstract
A longstanding puzzle in human genetics is what limits the clinical manifestation of hundreds of hereditary diseases to certain tissues, while their causal genes are expressed throughout the human body. A general conception is that tissue-selective disease phenotypes emerge when masking factors operate in unaffected tissues, but are specifically absent or insufficient in disease-manifesting tissues. Although this conception has critical impact on the understanding of disease manifestation, it was never challenged in a systematic manner across a variety of hereditary diseases and affected tissues. Here, we address this gap in our understanding via rigorous analysis of the susceptibility of over 30 tissues to 112 tissue-selective hereditary diseases. We focused on the roles of paralogs of causal genes, which are presumably capable of compensating for their aberration. We show for the first time at large-scale via quantitative analysis of omics datasets that, preferentially in the disease-manifesting tissues, paralogs are under-expressed relative to causal genes in more than half of the diseases. This was observed for several susceptible tissues and for causal genes with varying number of paralogs, suggesting that imbalanced expression of paralogs increases tissue susceptibility. While for many diseases this imbalance stemmed from up-regulation of the causal gene in the disease-manifesting tissue relative to other tissues, it was often combined with down-regulation of its paralog. Notably in roughly 20% of the cases, this imbalance stemmed only from significant down-regulation of the paralog. Thus, dosage relationships between paralogs appear as important, yet currently under-appreciated, modifiers of disease manifestation., Author summary A longstanding enigma in human genetics is what limits the clinical manifestation of hundreds of hereditary diseases to certain tissues or cell types, while their causal genes are present and expressed throughout the human body. A general conception was that the tissue-wide robustness to the causal aberration is achieved owing to the presence of a compensatory factor, and that disease phenotypes emerge wherever this factor is limited. Here, we tested this general conception at large-scale for the first time. We focused on paralogs of disease-causing genes, which share their functionality and may compensate for their aberration. Based on quantitative analyses of several types of omics data, we show that paralogs of causal genes are down-regulated relative to the disease-causing gene preferentially in the respective disease-manifesting tissue. This tendency is common across various subsets of causal genes, diseases, and tissues. Thus, paralogs of causal genes appear to contribute to the tissue-wide robustness against causal aberrations, and serve as important, yet currently under-appreciated, modifiers of disease manifestation.
- Published
- 2017
- Full Text
- View/download PDF
49. The impact of rare variation on gene expression across tissues
- Author
-
Manuel Muñoz-Aguirre, Pejman Mohammadi, Boxiang Liu, Eric Gamazon, Martijn Van de Bunt, Roger Little, Richard Sandstrom, Jemma Nelson, Thomas Juettemann, Yi-Hui Zhou, Olivier Delaneau, Alexandra Scott, Fan Wu, Panagiotis Papasaikas, Magali Ruffier, Halit Ongen, Daniel MacArthur, Daniel Zerbino, Peter Hickey, Pedro Ferreira, Xin Li, Yaping Liu, Esti Yeger-Lotem, Gaelen Hess, Rajinder Kaul, Dan Nicolae, David Davis, Ruth Barshir, Michael Sammeth, Stephen Montgomery, Diego Garrido-Martín, Kasper Hansen, Andrea Ganna, Mark McCarthy, Joshua Akey, Brown, Andrew Anand, Delaneau, Olivier, Dermitzakis, Emmanouil, Howald, Cédric, Panousis, Nikolaos, and GTEx, Consortium
- Subjects
Male ,0301 basic medicine ,Genotype ,Genomics ,Biology ,Article ,03 medical and health sciences ,Genetic variation ,Humans ,ddc:576.5 ,DNA sequencing ,Allele ,Gene ,Genetic association ,Genetics ,Multidisciplinary ,Models, Genetic ,Genome, Human ,Sequence Analysis, RNA ,Genetic Variation ,Bayes Theorem ,RNA sequencing ,Gene expression profiling ,Human genetics ,030104 developmental biology ,Organ Specificity ,Female ,Data integration ,Human genome ,Gene expression - Abstract
Rare genetic variants are abundant in humans and are expected to contribute to individual disease risk1,2,3,4. While genetic association studies have successfully identified common genetic variants associated with susceptibility, these studies are not practical for identifying rare variants1,5. Efforts to distinguish pathogenic variants from benign rare variants have leveraged the genetic code to identify deleterious protein-coding alleles1,6,7, but no analogous code exists for non-coding variants. Therefore, ascertaining which rare variants have phenotypic effects remains a major challenge. Rare non-coding variants have been associated with extreme gene expression in studies using single tissues8,9,10,11, but their effects across tissues are unknown. Here we identify gene expression outliers, or individuals showing extreme expression levels for a particular gene, across 44 human tissues by using combined analyses of whole genomes and multi-tissue RNA-sequencing data from the Genotype-Tissue Expression (GTEx) project v6p release12. We find that 58% of underexpression and 28% of overexpression outliers have nearby conserved rare variants compared to 8% of non-outliers. Additionally, we developed RIVER (RNA-informed variant effect on regulation), a Bayesian statistical model that incorporates expression data to predict a regulatory effect for rare variants with higher accuracy than models using genomic annotations alone. Overall, we demonstrate that rare variants contribute to large gene expression changes across tissues and provide an integrative method for interpretation of rare variants in individual genomes.
- Published
- 2017
50. A Differentiation Transcription Factor Establishes Muscle-Specific Proteostasis in Caenorhabditis elegans
- Author
-
Netta Shemesh, Anat Ben-Zvi, Shiran Dror, Rivka Ofir, Yael Bar-Lavan, and Esti Yeger-Lotem
- Subjects
0301 basic medicine ,Cancer Research ,Embryology ,Muscle Physiology ,Muscle Functions ,Physiology ,Cellular differentiation ,Muscle Proteins ,Gene Expression ,MyoD ,Muscle Development ,Biochemistry ,Contractile Proteins ,Animal Cells ,Myosin ,Morphogenesis ,Medicine and Health Sciences ,Myocyte ,Promoter Regions, Genetic ,Genetics (clinical) ,Heat-Shock Proteins ,biology ,Myogenesis ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Cell Differentiation ,Muscle Differentiation ,Cell biology ,DNA-Binding Proteins ,Myogenic Regulatory Factors ,Cellular Types ,Anatomy ,Research Article ,lcsh:QH426-470 ,Motor Proteins ,Muscle Tissue ,Actin Motors ,Embryonic Development ,Nerve Tissue Proteins ,Myosins ,03 medical and health sciences ,Molecular Motors ,Heat shock protein ,Genetics ,Animals ,HSP90 Heat-Shock Proteins ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Muscle Cells ,Binding Sites ,Embryos ,fungi ,Biology and Life Sciences ,Proteins ,Cell Biology ,HSP40 Heat-Shock Proteins ,Cytoskeletal Proteins ,lcsh:Genetics ,030104 developmental biology ,Proteostasis ,Biological Tissue ,Chaperone (protein) ,biology.protein ,Molecular Chaperones ,Transcription Factors ,Developmental Biology - Abstract
Safeguarding the proteome is central to the health of the cell. In multi-cellular organisms, the composition of the proteome, and by extension, protein-folding requirements, varies between cells. In agreement, chaperone network composition differs between tissues. Here, we ask how chaperone expression is regulated in a cell type-specific manner and whether cellular differentiation affects chaperone expression. Our bioinformatics analyses show that the myogenic transcription factor HLH-1 (MyoD) can bind to the promoters of chaperone genes expressed or required for the folding of muscle proteins. To test this experimentally, we employed HLH-1 myogenic potential to genetically modulate cellular differentiation of Caenorhabditis elegans embryonic cells by ectopically expressing HLH-1 in all cells of the embryo and monitoring chaperone expression. We found that HLH-1-dependent myogenic conversion specifically induced the expression of putative HLH-1-regulated chaperones in differentiating muscle cells. Moreover, disrupting the putative HLH-1-binding sites on ubiquitously expressed daf-21(Hsp90) and muscle-enriched hsp-12.2(sHsp) promoters abolished their myogenic-dependent expression. Disrupting HLH-1 function in muscle cells reduced the expression of putative HLH-1-regulated chaperones and compromised muscle proteostasis during and after embryogenesis. In turn, we found that modulating the expression of muscle chaperones disrupted the folding and assembly of muscle proteins and thus, myogenesis. Moreover, muscle-specific over-expression of the DNAJB6 homolog DNJ-24, a limb-girdle muscular dystrophy-associated chaperone, disrupted the muscle chaperone network and exposed synthetic motility defects. We propose that cellular differentiation could establish a proteostasis network dedicated to the folding and maintenance of the muscle proteome. Such cell-specific proteostasis networks can explain the selective vulnerability that many diseases of protein misfolding exhibit even when the misfolded protein is ubiquitously expressed., Author Summary Molecular chaperones protect proteins from misfolding and aggregation. In multi-cellular organisms, the composition and expression levels of chaperones vary between tissues. However, little is known of how such differential expression is regulated. We hypothesized that the cellular differentiation that regulates the cell-type specific expression program may be involved in establishing a cell-type specific chaperone network. To test this possibility, we addressed the myogenic commitment transcription factor HLH-1 (CeMyoD) that converts embryonic cells to muscle cells in Caenorhabditis elegans. We demonstrated that HLH-1 regulates the expression of muscle chaperones during muscle differentiation. Moreover, we showed that HLH-1-dependent expression of chaperones is required for embryonic development and muscle function. We propose that cellular differentiation results in cell-specific differences in the chaperone network that may be detrimental in terms of the susceptibility of neurons and muscle cells to protein misfolding diseases.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.