Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 202
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(9): 1877-1894.e27, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37116470

ABSTRACT

Negative-stranded RNA viruses can establish long-term persistent infection in the form of large intracellular inclusions in the human host and cause chronic diseases. Here, we uncover how cellular stress disrupts the metastable host-virus equilibrium in persistent infection and induces viral replication in a culture model of mumps virus. Using a combination of cell biology, whole-cell proteomics, and cryo-electron tomography, we show that persistent viral replication factories are dynamic condensates and identify the largely disordered viral phosphoprotein as a driver of their assembly. Upon stress, increased phosphorylation of the phosphoprotein at its interaction interface with the viral polymerase coincides with the formation of a stable replication complex. By obtaining atomic models for the authentic mumps virus nucleocapsid, we elucidate a concomitant conformational change that exposes the viral genome to its replication machinery. These events constitute a stress-mediated switch within viral condensates that provide an environment to support upregulation of viral replication.


Subject(s)
Mumps virus , Persistent Infection , Humans , Mumps virus/physiology , Nucleocapsid , Phosphoproteins/metabolism , Virus Replication
2.
Cell ; 181(4): 818-831.e19, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32359423

ABSTRACT

Cells sense elevated temperatures and mount an adaptive heat shock response that involves changes in gene expression, but the underlying mechanisms, particularly on the level of translation, remain unknown. Here we report that, in budding yeast, the essential translation initiation factor Ded1p undergoes heat-induced phase separation into gel-like condensates. Using ribosome profiling and an in vitro translation assay, we reveal that condensate formation inactivates Ded1p and represses translation of housekeeping mRNAs while promoting translation of stress mRNAs. Testing a variant of Ded1p with altered phase behavior as well as Ded1p homologs from diverse species, we demonstrate that Ded1p condensation is adaptive and fine-tuned to the maximum growth temperature of the respective organism. We conclude that Ded1p condensation is an integral part of an extended heat shock response that selectively represses translation of housekeeping mRNAs to promote survival under conditions of severe heat stress.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Fungal/genetics , Protein Biosynthesis/genetics , Saccharomyces cerevisiae Proteins/metabolism , DEAD-box RNA Helicases/physiology , Gene Expression/genetics , Genes, Essential/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Response/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/physiology
3.
Cell ; 181(2): 346-361.e17, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302572

ABSTRACT

Stressed cells shut down translation, release mRNA molecules from polysomes, and form stress granules (SGs) via a network of interactions that involve G3BP. Here we focus on the mechanistic underpinnings of SG assembly. We show that, under non-stress conditions, G3BP adopts a compact auto-inhibited state stabilized by electrostatic intramolecular interactions between the intrinsically disordered acidic tracts and the positively charged arginine-rich region. Upon release from polysomes, unfolded mRNAs outcompete G3BP auto-inhibitory interactions, engendering a conformational transition that facilitates clustering of G3BP through protein-RNA interactions. Subsequent physical crosslinking of G3BP clusters drives RNA molecules into networked RNA/protein condensates. We show that G3BP condensates impede RNA entanglement and recruit additional client proteins that promote SG maturation or induce a liquid-to-solid transition that may underlie disease. We propose that condensation coupled to conformational rearrangements and heterotypic multivalent interactions may be a general principle underlying RNP granule assembly.


Subject(s)
Cytoplasmic Granules/metabolism , DNA Helicases/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Ribonucleoproteins/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , HeLa Cells , Humans , Nucleic Acid Conformation , Organelles/metabolism , Phosphorylation , RNA, Messenger/metabolism , Stress, Physiological/genetics
4.
Nat Rev Mol Cell Biol ; 22(3): 196-213, 2021 03.
Article in English | MEDLINE | ID: mdl-33510441

ABSTRACT

Biomolecular condensates are membraneless intracellular assemblies that often form via liquid-liquid phase separation and have the ability to concentrate biopolymers. Research over the past 10 years has revealed that condensates play fundamental roles in cellular organization and physiology, and our understanding of the molecular principles, components and forces underlying their formation has substantially increased. Condensate assembly is tightly regulated in the intracellular environment, and failure to control condensate properties, formation and dissolution can lead to protein misfolding and aggregation, which are often the cause of ageing-associated diseases. In this Review, we describe the mechanisms and regulation of condensate assembly and dissolution, highlight recent advances in understanding the role of biomolecular condensates in ageing and disease, and discuss how cellular stress, ageing-related loss of homeostasis and a decline in protein quality control may contribute to the formation of aberrant, disease-causing condensates. Our improved understanding of condensate pathology provides a promising path for the treatment of protein aggregation diseases.


Subject(s)
Aging , Macromolecular Substances/chemistry , Multiprotein Complexes/physiology , Protein Aggregation, Pathological/etiology , Stress, Physiological/physiology , Aging/metabolism , Aging/pathology , Animals , Cell Physiological Phenomena , Humans , Macromolecular Substances/metabolism , Protein Aggregates/physiology , Protein Aggregation, Pathological/metabolism
5.
Cell ; 174(3): 688-699.e16, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29961577

ABSTRACT

Proteins such as FUS phase separate to form liquid-like condensates that can harden into less dynamic structures. However, how these properties emerge from the collective interactions of many amino acids remains largely unknown. Here, we use extensive mutagenesis to identify a sequence-encoded molecular grammar underlying the driving forces of phase separation of proteins in the FUS family and test aspects of this grammar in cells. Phase separation is primarily governed by multivalent interactions among tyrosine residues from prion-like domains and arginine residues from RNA-binding domains, which are modulated by negatively charged residues. Glycine residues enhance the fluidity, whereas glutamine and serine residues promote hardening. We develop a model to show that the measured saturation concentrations of phase separation are inversely proportional to the product of the numbers of arginine and tyrosine residues. These results suggest it is possible to predict phase-separation properties based on amino acid sequences.


Subject(s)
RNA-Binding Protein FUS/genetics , RNA-Binding Proteins/physiology , Amino Acid Sequence , Amino Acids/chemistry , Animals , Arginine/chemistry , Computer Simulation , HeLa Cells , Humans , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/physiology , Phase Transition , Prion Proteins/chemistry , Prion Proteins/genetics , Prions/genetics , Prions/physiology , Protein Domains , RNA-Binding Protein FUS/physiology , RNA-Binding Proteins/isolation & purification , Sf9 Cells , Tyrosine/chemistry
6.
Cell ; 169(6): 1066-1077.e10, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575670

ABSTRACT

Centrosomes are non-membrane-bound compartments that nucleate microtubule arrays. They consist of nanometer-scale centrioles surrounded by a micron-scale, dynamic assembly of protein called the pericentriolar material (PCM). To study how PCM forms a spherical compartment that nucleates microtubules, we reconstituted PCM-dependent microtubule nucleation in vitro using recombinant C. elegans proteins. We found that macromolecular crowding drives assembly of the key PCM scaffold protein SPD-5 into spherical condensates that morphologically and dynamically resemble in vivo PCM. These SPD-5 condensates recruited the microtubule polymerase ZYG-9 (XMAP215 homolog) and the microtubule-stabilizing protein TPXL-1 (TPX2 homolog). Together, these three proteins concentrated tubulin ∼4-fold over background, which was sufficient to reconstitute nucleation of microtubule asters in vitro. Our results suggest that in vivo PCM is a selective phase that organizes microtubule arrays through localized concentration of tubulin by microtubule effector proteins.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cell Cycle Proteins/metabolism , Centrosome/chemistry , Microtubules/metabolism , Tubulin/metabolism , Animals , Caenorhabditis elegans/cytology , Carrier Proteins/metabolism , Centrosome/metabolism , Protein Serine-Threonine Kinases/metabolism
7.
Cell ; 166(3): 637-650, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27471966

ABSTRACT

Most vertebrate oocytes contain a Balbiani body, a large, non-membrane-bound compartment packed with RNA, mitochondria, and other organelles. Little is known about this compartment, though it specifies germline identity in many non-mammalian vertebrates. We show Xvelo, a disordered protein with an N-terminal prion-like domain, is an abundant constituent of Xenopus Balbiani bodies. Disruption of the prion-like domain of Xvelo, or substitution with a prion-like domain from an unrelated protein, interferes with its incorporation into Balbiani bodies in vivo. Recombinant Xvelo forms amyloid-like networks in vitro. Amyloid-like assemblies of Xvelo recruit both RNA and mitochondria in binding assays. We propose that Xenopus Balbiani bodies form by amyloid-like assembly of Xvelo, accompanied by co-recruitment of mitochondria and RNA. Prion-like domains are found in germ plasm organizing proteins in other species, suggesting that Balbiani body formation by amyloid-like assembly could be a conserved mechanism that helps oocytes function as long-lived germ cells.


Subject(s)
Amyloid/metabolism , Organelle Biogenesis , T-Box Domain Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Benzothiazoles , Female , Fluorescent Dyes , Mitochondria/metabolism , Oocytes/cytology , Organelles/metabolism , Prions/chemistry , Protein Domains , Protein Transport , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sf9 Cells , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/genetics , Thiazoles , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus laevis , Zebrafish
8.
Cell ; 166(6): 1572-1584.e16, 2016 Sep 08.
Article in English | MEDLINE | ID: mdl-27594427

ABSTRACT

P granules are non-membrane-bound RNA-protein compartments that are involved in germline development in C. elegans. They are liquids that condense at one end of the embryo by localized phase separation, driven by gradients of polarity proteins such as the mRNA-binding protein MEX-5. To probe how polarity proteins regulate phase separation, we combined biochemistry and theoretical modeling. We reconstitute P granule-like droplets in vitro using a single protein PGL-3. By combining in vitro reconstitution with measurements of intracellular concentrations, we show that competition between PGL-3 and MEX-5 for mRNA can regulate the formation of PGL-3 droplets. Using theory, we show that, in a MEX-5 gradient, this mRNA competition mechanism can drive a gradient of P granule assembly with similar spatial and temporal characteristics to P granule assembly in vivo. We conclude that gradients of polarity proteins can position RNP granules during development by using RNA competition to regulate local phase separation.


Subject(s)
Caenorhabditis elegans/metabolism , RNA, Messenger/metabolism , Animals , Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans Proteins/metabolism , Cell Polarity , Embryo, Nonmammalian , Intracellular Space/chemistry , Intracellular Space/metabolism , Models, Theoretical , Protein Binding , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism
9.
Cell ; 163(3): 712-23, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26496610

ABSTRACT

The organization of a cell emerges from the interactions in protein networks. The interactome is critically dependent on the strengths of interactions and the cellular abundances of the connected proteins, both of which span orders of magnitude. However, these aspects have not yet been analyzed globally. Here, we have generated a library of HeLa cell lines expressing 1,125 GFP-tagged proteins under near-endogenous control, which we used as input for a next-generation interaction survey. Using quantitative proteomics, we detect specific interactions, estimate interaction stoichiometries, and measure cellular abundances of interacting proteins. These three quantitative dimensions reveal that the protein network is dominated by weak, substoichiometric interactions that play a pivotal role in defining network topology. The minority of stable complexes can be identified by their unique stoichiometry signature. This study provides a rich interaction dataset connecting thousands of proteins and introduces a framework for quantitative network analysis.


Subject(s)
Protein Interaction Mapping , Proteomics/methods , Cell Line , Chromosomes, Artificial, Bacterial/genetics , Humans
10.
Cell ; 162(5): 1066-77, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26317470

ABSTRACT

Many proteins contain disordered regions of low-sequence complexity, which cause aging-associated diseases because they are prone to aggregate. Here, we study FUS, a prion-like protein containing intrinsically disordered domains associated with the neurodegenerative disease ALS. We show that, in cells, FUS forms liquid compartments at sites of DNA damage and in the cytoplasm upon stress. We confirm this by reconstituting liquid FUS compartments in vitro. Using an in vitro "aging" experiment, we demonstrate that liquid droplets of FUS protein convert with time from a liquid to an aggregated state, and this conversion is accelerated by patient-derived mutations. We conclude that the physiological role of FUS requires forming dynamic liquid-like compartments. We propose that liquid-like compartments carry the trade-off between functionality and risk of aggregation and that aberrant phase transitions within liquid-like compartments lie at the heart of ALS and, presumably, other age-related diseases.


Subject(s)
Aging/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Mutation , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/genetics , Aging/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Cell Nucleus/chemistry , Cytoplasm/chemistry , Humans , Prions/chemistry , Protein Aggregates , Protein Structure, Tertiary , RNA-Binding Protein FUS/metabolism
11.
Mol Cell ; 82(19): 3712-3728.e10, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36150385

ABSTRACT

Recognition of pathogen-derived foreign nucleic acids is central to innate immune defense. This requires discrimination between structurally highly similar self and nonself nucleic acids to avoid aberrant inflammatory responses as in the autoinflammatory disorder Aicardi-Goutières syndrome (AGS). How vast amounts of self RNA are shielded from immune recognition to prevent autoinflammation is not fully understood. Here, we show that human SAM-domain- and HD-domain-containing protein 1 (SAMHD1), one of the AGS-causing genes, functions as a single-stranded RNA (ssRNA) 3'exonuclease, the lack of which causes cellular RNA accumulation. Increased ssRNA in cells leads to dissolution of RNA-protein condensates, which sequester immunogenic double-stranded RNA (dsRNA). Release of sequestered dsRNA from condensates triggers activation of antiviral type I interferon via retinoic-acid-inducible gene I-like receptors. Our results establish SAMHD1 as a key regulator of cellular RNA homeostasis and demonstrate that buffering of immunogenic self RNA by condensates regulates innate immune responses.


Subject(s)
Interferon Type I , RNA, Double-Stranded , Antiviral Agents , Autoimmune Diseases of the Nervous System , Exonucleases/genetics , Humans , Immunity, Innate/genetics , Interferon Type I/genetics , Nervous System Malformations , RNA, Double-Stranded/genetics , SAM Domain and HD Domain-Containing Protein 1/genetics
12.
Nat Rev Mol Cell Biol ; 18(5): 285-298, 2017 05.
Article in English | MEDLINE | ID: mdl-28225081

ABSTRACT

Biomolecular condensates are micron-scale compartments in eukaryotic cells that lack surrounding membranes but function to concentrate proteins and nucleic acids. These condensates are involved in diverse processes, including RNA metabolism, ribosome biogenesis, the DNA damage response and signal transduction. Recent studies have shown that liquid-liquid phase separation driven by multivalent macromolecular interactions is an important organizing principle for biomolecular condensates. With this physical framework, it is now possible to explain how the assembly, composition, physical properties and biochemical and cellular functions of these important structures are regulated.


Subject(s)
Eukaryotic Cells/cytology , Organelles/chemistry , Organelles/physiology , Animals , Biochemical Phenomena , Energy Metabolism , Humans , Kinetics
13.
Cell ; 152(4): 909-22, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23394947

ABSTRACT

Genetic interaction (GI) maps, comprising pairwise measures of how strongly the function of one gene depends on the presence of a second, have enabled the systematic exploration of gene function in microorganisms. Here, we present a two-stage strategy to construct high-density GI maps in mammalian cells. First, we use ultracomplex pooled shRNA libraries (25 shRNAs/gene) to identify high-confidence hit genes for a given phenotype and effective shRNAs. We then construct double-shRNA libraries from these to systematically measure GIs between hits. A GI map focused on ricin susceptibility broadly recapitulates known pathways and provides many unexpected insights. These include a noncanonical role for COPI, a previously uncharacterized protein complex affecting toxin clearance, a specialized role for the ribosomal protein RPS25, and functionally distinct mammalian TRAPP complexes. The ability to rapidly generate mammalian GI maps provides a potentially transformative tool for defining gene function and designing combination therapies based on synergistic pairs.


Subject(s)
Biological Transport , Epistasis, Genetic , Ricin/toxicity , Atorvastatin , Carrier Proteins/metabolism , Cell Line, Tumor , Coat Protein Complex I/metabolism , Endoplasmic Reticulum/metabolism , Heptanoic Acids/pharmacology , Humans , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Pyrroles/pharmacology , RNA, Small Interfering , Ribosomal Proteins/metabolism , Vesicular Transport Proteins/metabolism
14.
Annu Rev Cell Dev Biol ; 30: 39-58, 2014.
Article in English | MEDLINE | ID: mdl-25288112

ABSTRACT

Cells organize many of their biochemical reactions in non-membrane compartments. Recent evidence has shown that many of these compartments are liquids that form by phase separation from the cytoplasm. Here we discuss the basic physical concepts necessary to understand the consequences of liquid-like states for biological functions.


Subject(s)
Cell Compartmentation , Intracellular Fluid/chemistry , Animals , Cell Compartmentation/physiology , Cytoplasm/chemistry , Diffusion , Entropy , Gels , Origin of Life , Phase Transition , Solubility , Terminology as Topic
15.
Cell ; 149(6): 1339-52, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682253

ABSTRACT

We present a genetic interaction map of pairwise measures including ∼40% of nonessential S. pombe genes. By comparing interaction maps for fission and budding yeast, we confirmed widespread conservation of genetic relationships within and between complexes and pathways. However, we identified an important subset of orthologous complexes that have undergone functional "repurposing": the evolution of divergent functions and partnerships. We validated three functional repurposing events in S. pombe and mammalian cells and discovered that (1) two lumenal sensors of misfolded ER proteins, the kinase/nuclease Ire1 and the glucosyltransferase Gpt1, act together to mount an ER stress response; (2) ESCRT factors regulate spindle-pole-body duplication; and (3) a membrane-protein phosphatase and kinase complex, the STRIPAK complex, bridges the cis-Golgi, the centrosome, and the outer nuclear membrane to direct mitotic progression. Each discovery opens new areas of inquiry and-together-have implications for model organism-based research and the evolution of genetic systems.


Subject(s)
Epistasis, Genetic , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/genetics , Biological Evolution , Endosomal Sorting Complexes Required for Transport/metabolism , Membrane Glycoproteins , Mitosis , Multiprotein Complexes/metabolism , Protein Interaction Maps , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins , Schizosaccharomyces/cytology , Schizosaccharomyces/metabolism , Spindle Apparatus , Unfolded Protein Response
16.
Cell ; 150(4): 855-66, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22901814

ABSTRACT

Understanding the in vivo dynamics of protein localization and their physical interactions is important for many problems in biology. To enable systematic protein function interrogation in a multicellular context, we built a genome-scale transgenic platform for in vivo expression of fluorescent- and affinity-tagged proteins in Caenorhabditis elegans under endogenous cis regulatory control. The platform combines computer-assisted transgene design, massively parallel DNA engineering, and next-generation sequencing to generate a resource of 14,637 genomic DNA transgenes, which covers 73% of the proteome. The multipurpose tag used allows any protein of interest to be localized in vivo or affinity purified using standard tag-based assays. We illustrate the utility of the resource by systematic chromatin immunopurification and automated 4D imaging, which produced detailed DNA binding and cell/tissue distribution maps for key transcription factor proteins.


Subject(s)
Animals, Genetically Modified , Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans/genetics , Genetic Engineering/methods , Genome, Helminth , Transcription Factors/analysis , Animals , Caenorhabditis elegans/chemistry , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Transcription Factors/genetics
17.
Proc Natl Acad Sci U S A ; 121(3): e2314699121, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38198527

ABSTRACT

Energy metabolism supports neuronal function. While it is well established that changes in energy metabolism underpin brain plasticity and function, less is known about how individual neurons modulate their metabolic states to meet varying energy demands. This is because most approaches used to examine metabolism in living organisms lack the resolution to visualize energy metabolism within individual circuits, cells, or subcellular regions. Here, we adapted a biosensor for glycolysis, HYlight, for use in Caenorhabditis elegans to image dynamic changes in glycolysis within individual neurons and in vivo. We determined that neurons cell-autonomously perform glycolysis and modulate glycolytic states upon energy stress. By examining glycolysis in specific neurons, we documented a neuronal energy landscape comprising three general observations: 1) glycolytic states in neurons are diverse across individual cell types; 2) for a given condition, glycolytic states within individual neurons are reproducible across animals; and 3) for varying conditions of energy stress, glycolytic states are plastic and adapt to energy demands. Through genetic analyses, we uncovered roles for regulatory enzymes and mitochondrial localization in the cellular and subcellular dynamic regulation of glycolysis. Our study demonstrates the use of a single-cell glycolytic biosensor to examine how energy metabolism is distributed across cells and coupled to dynamic states of neuronal function and uncovers unique relationships between neuronal identities and metabolic landscapes in vivo.


Subject(s)
Glycolysis , Neurons , Animals , Energy Metabolism , Caenorhabditis elegans , Neuronal Plasticity
18.
Nat Methods ; 20(5): 673-676, 2023 05.
Article in English | MEDLINE | ID: mdl-37024650

ABSTRACT

The discovery of biomolecular condensates transformed our understanding of intracellular compartmentalization of molecules. To integrate interdisciplinary scientific knowledge about the function and composition of biomolecular condensates, we developed the crowdsourcing condensate database and encyclopedia ( cd-code.org ). CD-CODE is a community-editable platform, which includes a database of biomolecular condensates based on the literature, an encyclopedia of relevant scientific terms and a crowdsourcing web application. Our platform will accelerate the discovery and validation of biomolecular condensates, and facilitate efforts to understand their role in disease and as therapeutic targets.


Subject(s)
Crowdsourcing , Databases, Factual , Software
19.
Cell ; 147(6): 1224-5, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22153069

ABSTRACT

Although the parts list is nearly complete for many cellular structures, mechanisms that control their size remain poorly understood. Loughlin and colleagues now show that phosphorylation of a single residue of katanin, a microtubule-severing protein, largely accounts for the difference in spindle length between two closely related frogs.

20.
Cell ; 145(3): 470-82, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21529718

ABSTRACT

High-content screening for gene profiling has generally been limited to single cells. Here, we explore an alternative approach-profiling gene function by analyzing effects of gene knockdowns on the architecture of a complex tissue in a multicellular organism. We profile 554 essential C. elegans genes by imaging gonad architecture and scoring 94 phenotypic features. To generate a reference for evaluating methods for network construction, genes were manually partitioned into 102 phenotypic classes, predicting functions for uncharacterized genes across diverse cellular processes. Using this classification as a benchmark, we developed a robust computational method for constructing gene networks from high-content profiles based on a network context-dependent measure that ranks the significance of links between genes. Our analysis reveals that multi-parametric profiling in a complex tissue yields functional maps with a resolution similar to genetic interaction-based profiling in unicellular eukaryotes-pinpointing subunits of macromolecular complexes and components functioning in common cellular processes.


Subject(s)
Caenorhabditis elegans/genetics , Computational Biology/methods , Gene Regulatory Networks , Genetic Techniques , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Embryo, Nonmammalian/metabolism , Gene Knockdown Techniques , Gonads/embryology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL