Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 301
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 156(1-2): 249-60, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24439380

RESUMEN

Sleep is characterized by behavioral quiescence, homeostasis, increased arousal threshold, and rapid reversibility. Understanding how these properties are encoded by a neuronal circuit has been difficult, and no single molecular or neuronal pathway has been shown to be responsible for the regulation of sleep. Taking advantage of the well-mapped neuronal connections of Caenorhabditis elegans and the sleep-like states in this animal, we demonstrate the changed properties of both sensory neurons and downstream interneurons that mediate sleep and arousal. The ASH sensory neuron displays reduced sensitivity to stimuli in the sleep-like state, and the activity of the corresponding interneurons in ASH's motor circuit becomes asynchronous. Restoration of interneuron synchrony is sufficient for arousal. The multilevel circuit depression revealed provides an elegant strategy to promote a robust decrease in arousal while allowing for rapid reversibility of the sleep state.


Asunto(s)
Caenorhabditis elegans/fisiología , Células Receptoras Sensoriales/metabolismo , Animales , Nivel de Alerta , Calcio/metabolismo , Interneuronas/metabolismo , Sueño
2.
Mol Cell ; 79(2): 207-220.e8, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32544389

RESUMEN

RNA polymerase II (RNA Pol II) contains a disordered C-terminal domain (CTD) whose length enigmatically correlates with genome size. The CTD is crucial to eukaryotic transcription, yet the functional and evolutionary relevance of this variation remains unclear. Here, we investigate how CTD length and disorder influence transcription. We find that length modulates the size and frequency of transcriptional bursting. Disorder is highly conserved and facilitates CTD-CTD interactions, an ability we show is separable from protein sequence and necessary for efficient transcription. We build a data-driven quantitative model, simulations of which recapitulate experiments and support that CTD length promotes initial polymerase recruitment to the promoter and slows down its release from it and that CTD-CTD interactions enable recruitment of multiple polymerases. Our results reveal how these parameters provide access to a range of transcriptional activity, offering a new perspective for the mechanistic significance of CTD length and disorder in transcription across eukaryotes.


Asunto(s)
Dominio Catalítico , ARN Polimerasa II/metabolismo , Saccharomycetales/enzimología , Saccharomycetales/genética , Transcripción Genética , Secuencia de Aminoácidos , Modelos Genéticos , ARN Polimerasa II/química , RNA-Seq , Relación Estructura-Actividad , Transcriptoma
3.
Proc Natl Acad Sci U S A ; 121(25): e2322588121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38861598

RESUMEN

The nematode intestine is the primary site for nutrient uptake and storage as well as the synthesis of biomolecules; lysosome-related organelles known as gut granules are important for many of these functions. Aspects of intestine biology are not well understood, including the export of the nutrients it imports and the molecules it synthesizes, as well as the complete functions and protein content of the gut granules. Here, we report a mass spectrometry (MS)-based proteomic analysis of the intestine of the Caenorhabditis elegans and of its gut granules. Overall, we identified approximately 5,000 proteins each in the intestine and the gonad and showed that most of these proteins can be detected in samples extracted from a single worm, suggesting the feasibility of individual-level genetic analysis using proteomes. Comparing proteomes and published transcriptomes of the intestine and the gonad, we identified proteins that appear to be synthesized in the intestine and then transferred to the gonad. To identify gut granule proteins, we compared the proteome of individual intestines deficient in gut granules to the wild type. The identified gut granule proteome includes proteins known to be exclusively localized to the granules and additional putative gut granule proteins. We selected two of these putative gut granule proteins for validation via immunohistochemistry, and our successful confirmation of both suggests that our strategy was effective in identifying the gut granule proteome. Our results demonstrate the practicability of single-tissue MS-based proteomic analysis in small organisms and in its future utility.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Lisosomas , Proteómica , Animales , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteómica/métodos , Lisosomas/metabolismo , Proteoma/metabolismo , Intestinos , Mucosa Intestinal/metabolismo , Gónadas/metabolismo , Espectrometría de Masas/métodos , Orgánulos/metabolismo
4.
Cell ; 145(6): 981-92, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21663799

RESUMEN

Existing theories explain why operons are advantageous in prokaryotes, but their occurrence in metazoans is an enigma. Nematode operon genes, typically consisting of growth genes, are significantly upregulated during recovery from growth-arrested states. This expression pattern is anticorrelated to nonoperon genes, consistent with a competition for transcriptional resources. We find that transcriptional resources are initially limiting during recovery and that recovering animals are highly sensitive to any additional decrease in transcriptional resources. We provide evidence that operons become advantageous because, by clustering growth genes into operons, fewer promoters compete for the limited transcriptional machinery, effectively increasing the concentration of transcriptional resources and accelerating recovery. Mathematical modeling reveals how a moderate increase in transcriptional resources can substantially enhance transcription rate and recovery. This design principle occurs in different nematodes and the chordate C. intestinalis. As transition from arrest to rapid growth is shared by many metazoans, operons could have evolved to facilitate these processes.


Asunto(s)
Regulación de la Expresión Génica , Operón , Animales , Ciona intestinalis/genética , Ciona intestinalis/crecimiento & desarrollo , Ciona intestinalis/metabolismo , Modelos Genéticos , Nematodos/genética , Nematodos/crecimiento & desarrollo , Nematodos/metabolismo , Transcripción Genética
5.
Proc Natl Acad Sci U S A ; 120(19): e2218023120, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37126715

RESUMEN

Many chemosensory cues evoke responses of the same valence under widely varying physiological conditions. It remains unclear whether similar or distinct neural mechanisms are involved in the detection and processing of such chemosensory cues across contexts. We show that in Caenorhabditis elegans, a chemosensory cue is processed by distinct neural mechanisms at two different life stages that share the same valence state. Both starved adults and dauer larvae are attracted to carbon dioxide (CO2), but CO2 evokes different patterns of neural activity and different motor outputs at the two life stages. Moreover, the same interneuron within the CO2 microcircuit plays a different role in driving CO2-evoked motor output at the two life stages. The dauer-specific patterns of CO2-evoked activity in this interneuron require a dauer-specific gap junction complex and insulin signaling. Our results demonstrate that functionally distinct microcircuits are engaged in response to a chemosensory cue that triggers the same valence state at different life stages, revealing an unexpected complexity to chemosensory processing.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Caenorhabditis elegans/fisiología , Señales (Psicología) , Dióxido de Carbono , Interneuronas/fisiología , Transducción de Señal/fisiología , Larva
6.
Proc Natl Acad Sci U S A ; 120(51): e2221680120, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38096407

RESUMEN

Animals integrate sensory information from the environment and display various behaviors in response to external stimuli. In Caenorhabditis elegans hermaphrodites, 33 types of sensory neurons are responsible for chemosensation, olfaction, and mechanosensation. However, the functional roles of all sensory neurons have not been systematically studied due to the lack of facile genetic accessibility. A bipartite cGAL-UAS system has been previously developed to study tissue- or cell-specific functions in C. elegans. Here, we report a toolkit of new cGAL drivers that can facilitate the analysis of a vast majority of the 60 sensory neurons in C. elegans hermaphrodites. We generated 37 sensory neuronal cGAL drivers that drive cGAL expression by cell-specific regulatory sequences or intersection of two distinct regulatory regions with overlapping expression (split cGAL). Most cGAL-drivers exhibit expression in single types of cells. We also constructed 28 UAS effectors that allow expression of proteins to perturb or interrogate sensory neurons of choice. This cGAL-UAS sensory neuron toolkit provides a genetic platform to systematically study the functions of C. elegans sensory neurons.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Células Receptoras Sensoriales/metabolismo
7.
Development ; 149(9)2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35394033

RESUMEN

Diapause arrest in animals such as Caenorhabditis elegans is tightly regulated so that animals make appropriate developmental decisions amidst environmental challenges. Fully understanding diapause requires mechanistic insight of both entry and exit from the arrested state. Although a steroid hormone pathway regulates the entry decision into C. elegans dauer diapause, its role in the exit decision is less clear. A complication to understanding steroid hormonal regulation of dauer has been the peculiar fact that steroid hormone mutants such as daf-9 form partial dauers under normal growth conditions. Here, we corroborate previous findings that daf-9 mutants remain capable of forming full dauers under unfavorable growth conditions and establish that the daf-9 partial dauer state is likely a partially exited dauer that has initiated but cannot complete the dauer exit process. We show that the steroid hormone pathway is both necessary for and promotes complete dauer exit, and that the spatiotemporal dynamics of steroid hormone regulation during dauer exit resembles that of dauer entry. Overall, dauer entry and dauer exit are distinct developmental decisions that are both controlled by steroid hormone signaling.


Asunto(s)
Proteínas de Caenorhabditis elegans , Diapausa , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Hormonas/metabolismo , Larva/metabolismo , Mutación/genética , Esteroides/metabolismo
8.
PLoS Comput Biol ; 20(3): e1011968, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38527066

RESUMEN

Enrichment analysis is frequently used in combination with differential expression data to investigate potential commonalities amongst lists of genes and generate hypotheses for further experiments. However, current enrichment analysis approaches on pathways ignore the functional relationships between genes in a pathway, particularly OR logic that occurs when a set of proteins can each individually perform the same step in a pathway. As a result, these approaches miss pathways with large or multiple sets because of an inflation of pathway size (when measured as the total gene count) relative to the number of steps. We address this problem by enriching on step-enabling entities in pathways. We treat sets of protein-coding genes as single entities, and we also weight sets to account for the number of genes in them using the multivariate Fisher's noncentral hypergeometric distribution. We then show three examples of pathways that are recovered with this method and find that the results have significant proportions of pathways not found in gene list enrichment analysis.


Asunto(s)
Perfilación de la Expresión Génica , Perfilación de la Expresión Génica/métodos
9.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35210367

RESUMEN

Mounting evidence suggests that nematode infection can protect against disorders of immune dysregulation. Administration of live parasites or their excretory/secretory (ES) products has shown therapeutic effects across a wide range of animal models for immune disorders, including asthma. Human clinical trials of live parasite ingestion for the treatment of immune disorders have produced promising results, yet concerns persist regarding the ingestion of pathogenic organisms and the immunogenicity of protein components. Despite extensive efforts to define the active components of ES products, no small molecules with immune regulatory activity have been identified from nematodes. Here we show that an evolutionarily conserved family of nematode pheromones called ascarosides strongly modulates the pulmonary immune response and reduces asthma severity in mice. Screening the inhibitory effects of ascarosides produced by animal-parasitic nematodes on the development of asthma in an ovalbumin (OVA) murine model, we found that administration of nanogram quantities of ascr#7 prevented the development of lung eosinophilia, goblet cell metaplasia, and airway hyperreactivity. Ascr#7 suppressed the production of IL-33 from lung epithelial cells and reduced the number of memory-type pathogenic Th2 cells and ILC2s in the lung, both key drivers of the pathology of asthma. Our findings suggest that the mammalian immune system recognizes ascarosides as an evolutionarily conserved molecular signature of parasitic nematodes. The identification of a nematode-produced small molecule underlying the well-documented immunomodulatory effects of ES products may enable the development of treatment strategies for allergic diseases.


Asunto(s)
Inflamación/prevención & control , Nematodos/química , Tráquea/efectos de los fármacos , Animales , Asma/fisiopatología , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Hipersensibilidad/fisiopatología , Inflamación/inducido químicamente , Ratones , Ratones Endogámicos BALB C , Nematodos/patogenicidad , Ovalbúmina/efectos adversos , Bibliotecas de Moléculas Pequeñas/farmacología , Tráquea/fisiopatología
10.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33361149

RESUMEN

Acetylcholine (ACh) promotes various cell migrations in vitro, but there are few investigations into this nonsynaptic role of ACh signaling in vivo. Here we investigate the function of a muscarinic receptor on an epithelial cell migration in Caenorhabditis elegans We show that the migratory gonad leader cell, the linker cell (LC), uses an M1/M3/M5-like muscarinic ACh receptor GAR-3 to receive extrasynaptic ACh signaling from cholinergic neurons for its migration. Either the loss of the GAR-3 receptor in the LC or the inhibition of ACh release from cholinergic neurons resulted in migratory path defects. The overactivation of the GAR-3 muscarinic receptor caused the LC to reverse its orientation through its downstream effectors Gαq/egl-30, PLCß/egl-8, and TRIO/unc-73 This reversal response only occurred in the fourth larval stage, which corresponds to the developmental time when the GAR-3::yellow fluorescent protein receptor in the membrane relocalizes from a uniform to an asymmetric distribution. These findings suggest a role for the GAR-3 muscarinic receptor in determining the direction of LC migration.


Asunto(s)
Acetilcolina/metabolismo , Movimiento Celular/fisiología , Receptores Muscarínicos/metabolismo , Acetilcolina/fisiología , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/fisiología , Células Epiteliales/metabolismo , Contracción Muscular/fisiología , Terminales Presinápticos/metabolismo , Receptores Muscarínicos/fisiología , Transducción de Señal
11.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33972425

RESUMEN

Proper left-right symmetry breaking is essential for animal development, and in many cases, this process is actomyosin-dependent. In Caenorhabditis elegans embryos active torque generation in the actomyosin layer promotes left-right symmetry breaking by driving chiral counterrotating cortical flows. While both Formins and Myosins have been implicated in left-right symmetry breaking and both can rotate actin filaments in vitro, it remains unclear whether active torques in the actomyosin cortex are generated by Formins, Myosins, or both. We combined the strength of C. elegans genetics with quantitative imaging and thin film, chiral active fluid theory to show that, while Non-Muscle Myosin II activity drives cortical actomyosin flows, it is permissive for chiral counterrotation and dispensable for chiral symmetry breaking of cortical flows. Instead, we find that CYK-1/Formin activation in RhoA foci is instructive for chiral counterrotation and promotes in-plane, active torque generation in the actomyosin cortex. Notably, we observe that artificially generated large active RhoA patches undergo rotations with consistent handedness in a CYK-1/Formin-dependent manner. Altogether, we conclude that CYK-1/Formin-dependent active torque generation facilitates chiral symmetry breaking of actomyosin flows and drives organismal left-right symmetry breaking in the nematode worm.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Corteza Cerebral/metabolismo , Forminas/metabolismo , Transducción de Señal/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Actomiosina/genética , Actomiosina/metabolismo , Animales , Animales Modificados Genéticamente , Blastómeros/citología , Blastómeros/metabolismo , Tipificación del Cuerpo/genética , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Corteza Cerebral/embriología , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Forminas/genética , Lateralidad Funcional/genética , Lateralidad Funcional/fisiología , Transducción de Señal/genética , Torque , Proteína de Unión al GTP rhoA/genética
12.
Mamm Genome ; 34(4): 531-544, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37666946

RESUMEN

Comparing genomic and biological characteristics across multiple species is essential to using model systems to investigate the molecular and cellular mechanisms underlying human biology and disease and to translate mechanistic insights from studies in model organisms for clinical applications. Building a scalable knowledge commons platform that supports cross-species comparison of rich, expertly curated knowledge regarding gene function, phenotype, and disease associations available for model organisms and humans is the primary mission of the Alliance of Genome Resources (the Alliance). The Alliance is a consortium of seven model organism knowledgebases (mouse, rat, yeast, nematode, zebrafish, frog, fruit fly) and the Gene Ontology resource. The Alliance uses a common set of gene ortholog assertions as the basis for comparing biological annotations across the organisms represented in the Alliance. The major types of knowledge associated with genes that are represented in the Alliance database currently include gene function, phenotypic alleles and variants, human disease associations, pathways, gene expression, and both protein-protein and genetic interactions. The Alliance has enhanced the ability of researchers to easily compare biological annotations for common data types across model organisms and human through the implementation of shared programmatic access mechanisms, data-specific web pages with a unified "look and feel", and interactive user interfaces specifically designed to support comparative biology. The modular infrastructure developed by the Alliance allows the resource to serve as an extensible "knowledge commons" capable of expanding to accommodate additional model organisms.


Asunto(s)
Bases de Datos Genéticas , Pez Cebra , Ratas , Ratones , Animales , Humanos , Pez Cebra/genética , Genoma , Genómica , Fenotipo , Internet
13.
Cell ; 134(4): 646-56, 2008 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-18724937

RESUMEN

The orientation of asymmetric cell division contributes to the organization of cells within a tissue or organ. For example, mirror-image symmetry of the C. elegans vulva is achieved by the opposite division orientation of the vulval precursor cells (VPCs) flanking the axis of symmetry. We characterized the molecular mechanisms contributing to this division pattern. Wnts MOM-2 and LIN-44 are expressed at the axis of symmetry and orient the VPCs toward the center. These Wnts act via Fz/LIN-17 and Ryk/LIN-18, which control beta-catenin localization and activate gene transcription. In addition, VPCs on both sides of the axis of symmetry possess a uniform underlying "ground" polarity, established by the instructive activity of Wnt/EGL-20. EGL-20 establishes ground polarity via a novel type of signaling involving the Ror receptor tyrosine kinase CAM-1 and the planar cell polarity component Van Gogh/VANG-1. Thus, tissue polarity is determined by the integration of multiple Wnt pathways.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Polaridad Celular , Organogénesis , Transducción de Señal , Proteínas Wnt/metabolismo , Animales , Femenino , Glicoproteínas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Vulva/embriología , beta Catenina/metabolismo
14.
Nucleic Acids Res ; 48(D1): D762-D767, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31642470

RESUMEN

WormBase (https://wormbase.org/) is a mature Model Organism Information Resource supporting researchers using the nematode Caenorhabditis elegans as a model system for studies across a broad range of basic biological processes. Toward this mission, WormBase efforts are arranged in three primary facets: curation, user interface and architecture. In this update, we describe progress in each of these three areas. In particular, we discuss the status of literature curation and recently added data, detail new features of the web interface and options for users wishing to conduct data mining workflows, and discuss our efforts to build a robust and scalable architecture by leveraging commercial cloud offerings. We conclude with a description of WormBase's role as a founding member of the nascent Alliance of Genome Resources.


Asunto(s)
Caenorhabditis elegans/genética , Bases de Datos Genéticas , Genes de Helminto , Animales , Minería de Datos , Genómica , Internet , Interfaz Usuario-Computador
15.
J Am Chem Soc ; 143(36): 14676-14683, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34460264

RESUMEN

The recently discovered modular glucosides (MOGLs) form a large metabolite library derived from combinatorial assembly of moieties from amino acid, neurotransmitter, and lipid metabolism in the model organism C. elegans. Combining CRISPR-Cas9 genome editing, comparative metabolomics, and synthesis, we show that the carboxylesterase homologue Cel-CEST-1.2 is responsible for specific 2-O-acylation of diverse glucose scaffolds with a wide variety of building blocks, resulting in more than 150 different MOGLs. We further show that this biosynthetic role is conserved for the closest homologue of Cel-CEST-1.2 in the related nematode species C. briggsae, Cbr-CEST-2. Expression of Cel-cest-1.2 and MOGL biosynthesis are strongly induced by starvation conditions in C. elegans, one of the premier model systems for mechanisms connecting nutrition and physiology. Cel-cest-1.2-deletion results in early death of adult animals under starvation conditions, providing first insights into the biological functions of MOGLs.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Glucósidos/biosíntesis , Inanición/metabolismo , Acilación , Animales , Glucósidos/química , Metabolómica , ortoaminobenzoatos/metabolismo
16.
Hum Mol Genet ; 28(13): 2271-2281, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31220273

RESUMEN

Autism spectrum disorder (ASD) involves thousands of alleles in over 850 genes, but the current functional inference tools are not sufficient to predict phenotypic changes. As a result, the causal relationship of most of these genetic variants in the pathogenesis of ASD has not yet been demonstrated and an experimental method prioritizing missense alleles for further intensive analysis is crucial. For this purpose, we have designed a pipeline that uses Caenorhabditis elegans as a genetic model to screen for phenotype-changing missense alleles inferred from human ASD studies. We identified highly conserved human ASD-associated missense variants in their C. elegans orthologs, used a CRISPR/Cas9-mediated homology-directed knock-in strategy to generate missense mutants and analyzed their impact on behaviors and development via several broad-spectrum assays. All tested missense alleles were predicted to perturb protein function, but we found only 70% of them showed detectable phenotypic changes in morphology, locomotion or fecundity. Our findings indicate that certain missense variants in the C. elegans orthologs of human CACNA1D, CHD7, CHD8, CUL3, DLG4, GLRA2, NAA15, PTEN, SYNGAP1 and TPH2 impact neurodevelopment and movement functions, elevating these genes as candidates for future study into ASD. Our approach will help prioritize functionally important missense variants for detailed studies in vertebrate models and human cells.


Asunto(s)
Trastorno del Espectro Autista/genética , Caenorhabditis elegans/genética , Alelos , Animales , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Fertilidad/genética , Estudios de Asociación Genética , Locomoción/genética , Mutación Missense , Trastornos del Neurodesarrollo/genética , Fenotipo
17.
Trends Genet ; 34(4): 253-255, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29395380

RESUMEN

Dejima and colleagues report using CRISPR/Cas9 to generate a new collection of greatly improved balancer chromosomes in the standard laboratory nematode Caenorhabditis elegans, using methods previously reported by the same laboratory, expanding the set of C. elegans balancers to cover nearly 90% of coding genes.


Asunto(s)
Sistemas CRISPR-Cas , Caenorhabditis elegans/genética , Animales , Cromosomas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas
18.
Proc Natl Acad Sci U S A ; 115(13): E2930-E2939, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29531064

RESUMEN

RNA-sequencing (RNA-seq) is commonly used to identify genetic modules that respond to perturbations. In single cells, transcriptomes have been used as phenotypes, but this concept has not been applied to whole-organism RNA-seq. Also, quantifying and interpreting epistatic effects using expression profiles remains a challenge. We developed a single coefficient to quantify transcriptome-wide epistasis that reflects the underlying interactions and which can be interpreted intuitively. To demonstrate our approach, we sequenced four single and two double mutants of Caenorhabditis elegans From these mutants, we reconstructed the known hypoxia pathway. In addition, we uncovered a class of 56 genes with HIF-1-dependent expression that have opposite changes in expression in mutants of two genes that cooperate to negatively regulate HIF-1 abundance; however, the double mutant of these genes exhibits suppression epistasis. This class violates the classical model of HIF-1 regulation but can be explained by postulating a role of hydroxylated HIF-1 in transcriptional control.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Epistasis Genética , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Transcriptoma , Animales , Caenorhabditis elegans/crecimiento & desarrollo
19.
Proc Natl Acad Sci U S A ; 115(15): 3900-3905, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29581308

RESUMEN

Bipartite expression systems, such as the GAL4-UAS system, allow fine manipulation of gene expression and are powerful tools for interrogating gene function. Recently, we established cGAL, a GAL4-based bipartite expression system for transgene control in Caenorhabditis elegans, where a single promoter dictates the expression pattern of a cGAL driver, which then binds target upstream activation sequences to drive expression of a downstream effector gene. Here, we report a split strategy for cGAL using the split intein gp41-1 for intersectional control of transgene expression. Split inteins are protein domains that associate, self-excise, and covalently ligate their flanking peptides together. We split the DNA binding domain and transcriptional activation domain of cGAL and fused them to the N terminal of gp41-1-N-intein and the C terminal of gp41-1-C-intein, respectively. In cells where both halves of cGAL are expressed, a functional cGAL driver is reconstituted via intein-mediated protein splicing. This reconstitution allows expression of the driver to be dictated by two promoters for refined spatial control or spatiotemporal control of transgene expression. We apply the split cGAL system to genetically access the single pair of MC neurons (previously inaccessible with a single promoter), and reveal an important role of protein kinase A in rhythmic pharyngeal pumping in C. elegans Thus, the split cGAL system gives researchers a greater degree of spatiotemporal control over transgene expression, and will be a valuable genetic tool in C. elegans for dissecting gene function with finer cell-specific resolution.


Asunto(s)
Caenorhabditis elegans/genética , Proteínas de Unión al ADN/genética , Proteínas Fúngicas/genética , Regulación de la Expresión Génica , Inteínas , Transgenes , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Fúngicas/metabolismo , Neuronas/metabolismo , Regiones Promotoras Genéticas , Empalme de Proteína , Saccharomyces/genética
20.
Dev Biol ; 454(2): 170-180, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31242447

RESUMEN

Phenotypic plasticity allows animals to survive in changing environments through the alteration of phenotypes or development. One of the best-studied examples of phenotypic plasticity is dauer larval development in the free-living roundworm Caenorhabditis elegans. When faced with hostile environments, C. elegans larvae can exit reproductive development and enter the stress-resistant and spore-like dauer larval stage. However, knowledge about how the dauer entry decision is made, and how the different tissues of the animal coordinate to execute transformation into dauer, is limited. This is because identifying animals that make the entry decision, or that fail to coordinately remodel their tissues during dauer development, is time-consuming and labor-intensive. Utilizing our previously reported RNA-seq of animals going through dauer or reproductive development (Lee et al., 2017), we have identified genetic markers for conveniently tracking and manipulating the dauer entry decision. These include col-183 (which tracks dauer fate in the hypodermis), ets-10 (neurons and intestine), nhr-246 (intestine and hypodermis), and F53F1.4 (reproductive fate in the hypodermis). Using condition shift experiments, we demonstrate that the dauer-specific fluorescent expression of the markers correspond to the commitment event of the dauer entry decision, and therefore label when the decision is made. We show that these markers can be used to manipulate the entry decision by driving the reproduction-promoting gene daf-9 under the control of the dauer-specific marker col-183, through which we could shift animals into non-dauer development. We further demonstrate that the markers can be used to track tissue coordination during the decision. daf-9, daf-15, and daf-18 partial dauers exhibit incomplete expression of the ets-10 marker, with our results indicating that the same gene (e.g. daf-9 or daf-18) can affect dauer development differently in different tissues. Our findings provide molecular tools for studying phenotypic plasticity during a whole animal decision.


Asunto(s)
Adaptación Fisiológica/genética , Caenorhabditis elegans/genética , Diapausa/genética , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Diapausa/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Marcadores Genéticos , Larva/metabolismo , Mutación , Neuronas/metabolismo , Fenotipo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA