Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Development ; 151(3)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38345109

RESUMEN

The field of developmental biology has declined in prominence in recent decades, with off-shoots from the field becoming more fashionable and highly funded. This has created inequity in discovery and opportunity, partly due to the perception that the field is antiquated or not cutting edge. A 'think tank' of scientists from multiple developmental biology-related disciplines came together to define specific challenges in the field that may have inhibited innovation, and to provide tangible solutions to some of the issues facing developmental biology. The community suggestions include a call to the community to help 'rebrand' the field, alongside proposals for additional funding apparatuses, frameworks for interdisciplinary innovative collaborations, pedagogical access, improved science communication, increased diversity and inclusion, and equity of resources to provide maximal impact to the community.


Asunto(s)
Biología Evolutiva
2.
Cell ; 149(1): 75-87, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22464324

RESUMEN

Crossovers (COs) between homologous chromosomes ensure their faithful segregation during meiosis. We identify C. elegans COSA-1, a cyclin-related protein conserved in metazoa, as a key component required to convert meiotic double-strand breaks (DSBs) into COs. During late meiotic prophase, COSA-1 localizes to foci that correspond to the single CO site on each homolog pair and indicate sites of eventual concentration of other conserved CO proteins. Chromosomes gain and lose competence to load CO proteins during meiotic progression, with competence to load COSA-1 requiring prior licensing. Our data further suggest a self-reinforcing mechanism maintaining CO designation. Modeling of a nonlinear dose-response relationship between IR-induced DSBs and COSA-1 foci reveals efficient conversion of DSBs into COs when DSBs are limiting and a robust capacity to limit cytologically differentiated CO sites when DSBs are in excess. COSA-1 foci serve as a unique live cell readout for investigating CO formation and CO interference.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/citología , Intercambio Genético , Ciclinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Meiosis , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Cromosomas/metabolismo , Ciclinas/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , Modelos Moleculares , Mutación
3.
Cell ; 145(3): 470-82, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21529718

RESUMEN

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.


Asunto(s)
Caenorhabditis elegans/genética , Biología Computacional/métodos , Redes Reguladoras de Genes , Técnicas Genéticas , Animales , Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Embrión no Mamífero/metabolismo , Técnicas de Silenciamiento del Gen , Gónadas/embriología , Fenotipo
4.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35022236

RESUMEN

The fidelity of a signaling pathway depends on its tight regulation in space and time. Extracellular signal-regulated kinase (ERK) controls wide-ranging cellular processes to promote organismal development and tissue homeostasis. ERK activation depends on a reversible dual phosphorylation on the TEY motif in its active site by ERK kinase (MEK) and dephosphorylation by DUSPs (dual specificity phosphatases). LIP-1, a DUSP6/7 homolog, was proposed to function as an ERK (MPK-1) DUSP in the Caenorhabditis elegans germline primarily because of its phenotype, which morphologically mimics that of a RAS/let-60 gain-of-function mutant (i.e., small oocyte phenotype). Our investigations, however, reveal that loss of lip-1 does not lead to an increase in MPK-1 activity in vivo. Instead, we show that loss of lip-1 leads to 1) a decrease in MPK-1 phosphorylation, 2) lower MPK-1 substrate phosphorylation, 3) phenocopy of mpk-1 reduction-of-function (rather than gain-of-function) allele, and 4) a failure to rescue mpk-1-dependent germline or fertility defects. Moreover, using diverse genetic mutants, we show that the small oocyte phenotype does not correlate with increased ectopic MPK-1 activity and that ectopic increase in MPK-1 phosphorylation does not necessarily result in a small oocyte phenotype. Together, these data demonstrate that LIP-1 does not function as an MPK-1 DUSP in the C. elegans germline. Our results caution against overinterpretation of the mechanistic underpinnings of orthologous phenotypes, since they may be a result of independent mechanisms, and provide a framework for characterizing the distinct molecular targets through which LIP-1 may mediate its several germline functions.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Proteínas de Ciclo Celular/metabolismo , Células Germinativas/enzimología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Proteínas de Caenorhabditis elegans/genética , Proteínas de Ciclo Celular/genética , Proliferación Celular , Activación Enzimática , Mutación/genética , Oocitos/citología , Oocitos/metabolismo , Fase Paquiteno , Fenotipo , Fosforilación , Proteínas Tirosina Fosfatasas/genética , Especificidad por Sustrato , Complejo Sinaptonémico/metabolismo , Temperatura
5.
Proc Natl Acad Sci U S A ; 119(21): e2117865119, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35576467

RESUMEN

Crossover formation is essential for proper segregation of homologous chromosomes during meiosis. Here, we show that Caenorhabditis elegans cyclin-dependent kinase 2 (CDK-2) partners with cyclin-like protein COSA-1 to promote crossover formation by promoting conversion of meiotic double-strand breaks into crossover­specific recombination intermediates. Further, we identify MutSγ component MSH-5 as a CDK-2 phosphorylation target. MSH-5 has a disordered C-terminal tail that contains 13 potential CDK phosphosites and is required to concentrate crossover­promoting proteins at recombination sites. Phosphorylation of the MSH-5 tail appears dispensable in a wild-type background, but when MutSγ activity is partially compromised, crossover formation and retention of COSA-1 at recombination sites are exquisitely sensitive to phosphosite loss. Our data support a model in which robustness of crossover designation reflects a positive feedback mechanism involving CDK-2­mediated phosphorylation and scaffold-like properties of the MSH5 C-terminal tail, features that combine to promote full recruitment and activity of crossover­promoting complexes.


Asunto(s)
Proteínas de Caenorhabditis elegans , Quinasa 2 Dependiente de la Ciclina , Proteínas de Unión al ADN , Meiosis , Complejo Sinaptonémico , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Segregación Cromosómica , Intercambio Genético , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Proteínas de Unión al ADN/metabolismo , Fosforilación , Complejo Sinaptonémico/genética , Complejo Sinaptonémico/metabolismo
6.
Hum Mol Genet ; 31(17): 2934-2950, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-35405010

RESUMEN

DROSHA encodes a ribonuclease that is a subunit of the Microprocessor complex and is involved in the first step of microRNA (miRNA) biogenesis. To date, DROSHA has not yet been associated with a Mendelian disease. Here, we describe two individuals with profound intellectual disability, epilepsy, white matter atrophy, microcephaly and dysmorphic features, who carry damaging de novo heterozygous variants in DROSHA. DROSHA is constrained for missense variants and moderately intolerant to loss-of-function (o/e = 0.24). The loss of the fruit fly ortholog drosha causes developmental arrest and death in third instar larvae, a severe reduction in brain size and loss of imaginal discs in the larva. Loss of drosha in eye clones causes small and rough eyes in adult flies. One of the identified DROSHA variants (p.Asp1219Gly) behaves as a strong loss-of-function allele in flies, while another variant (p.Arg1342Trp) is less damaging in our assays. In worms, a knock-in that mimics the p.Asp1219Gly variant at a worm equivalent residue causes loss of miRNA expression and heterochronicity, a phenotype characteristic of the loss of miRNA. Together, our data show that the DROSHA variants found in the individuals presented here are damaging based on functional studies in model organisms and likely underlie the severe phenotype involving the nervous system.


Asunto(s)
Epilepsia , Discapacidad Intelectual , MicroARNs , Microcefalia , Malformaciones del Sistema Nervioso , Humanos , Discapacidad Intelectual/genética , MicroARNs/genética , MicroARNs/metabolismo , Microcefalia/genética , Ribonucleasa III/genética , Ribonucleasa III/metabolismo
7.
Mol Reprod Dev ; 89(9): 353-374, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35908193

RESUMEN

During oogenesis, oocytes arrest at meiotic prophase I to acquire competencies for resuming meiosis, fertilization, and early embryonic development. Following this arrested period, oocytes resume meiosis in response to species-specific hormones, a process known as oocyte maturation, that precedes ovulation and fertilization. Involvement of endocrine and autocrine/paracrine factors and signaling events during maintenance of prophase I arrest, and resumption of meiosis is an area of active research. Studies in vertebrate and invertebrate model organisms have delineated the molecular determinants and signaling pathways that regulate oocyte maturation. Cell cycle regulators, such as cyclin-dependent kinase (CDK1), polo-like kinase (PLK1), Wee1/Myt1 kinase, and the phosphatase CDC25 play conserved roles during meiotic resumption. Extracellular signal-regulated kinase (ERK), on the other hand, while activated during oocyte maturation in all species, regulates both species-specific, as well as conserved events among different organisms. In this review, we synthesize the general signaling mechanisms and focus on conserved and distinct functions of ERK signaling pathway during oocyte maturation in mammals, non-mammalian vertebrates, and invertebrates such as Drosophila and Caenorhabditis elegans.


Asunto(s)
Proteínas de Drosophila , Meiosis , Animales , Caenorhabditis elegans/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Drosophila , Proteínas de Drosophila/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Hormonas/metabolismo , Mamíferos , Oocitos/metabolismo , Oogénesis/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal/fisiología
8.
Proc Natl Acad Sci U S A ; 116(3): 960-969, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30593561

RESUMEN

DICER1 gene alterations and decreased expression are associated with developmental disorders and diseases in humans. Oscillation of Dicer1 phosphorylation and dephosphorylation regulates its function during the oocyte-to-embryo transition in Caenorhabditis elegans Dicer1 is also phosphorylated upon FGF stimulation at conserved serines in mouse embryonic fibroblasts and HEK293 cells. However, whether phosphorylation of Dicer1 has a role in mammalian development remains unknown. To investigate the consequence of constitutive phosphorylation, we generated phosphomimetic knock-in mouse models by replacing conserved serines 1712 and 1836 with aspartic acids individually or together. Dicer1S1836D/S1836D mice display highly penetrant postnatal lethality, and the few survivors display accelerated aging and infertility. Homozygous dual-phosphomimetic Dicer1 augments these defects, alters metabolism-associated miRNAs, and causes a hypermetabolic phenotype. Thus, constitutive phosphorylation of Dicer1 results in multiple pathologic processes in mice, indicating that phosphorylation tightly regulates Dicer1 function and activity in mammals.


Asunto(s)
Envejecimiento , ARN Helicasas DEAD-box , Homocigoto , Mutación Missense , Ribonucleasa III , Envejecimiento/genética , Envejecimiento/metabolismo , Sustitución de Aminoácidos , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Femenino , Técnicas de Sustitución del Gen , Células HEK293 , Humanos , Masculino , Ratones , Fosforilación/genética , Ribonucleasa III/genética , Ribonucleasa III/metabolismo
9.
Development ; 145(10)2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29695611

RESUMEN

Adult C. elegans germline stem cells (GSCs) and mouse embryonic stem cells (mESCs) exhibit a non-canonical cell cycle structure with an abbreviated G1 phase and phase-independent expression of Cdk2 and cyclin E. Mechanisms that promote the abbreviated cell cycle remain unknown, as do the consequences of not maintaining an abbreviated cell cycle in these tissues. In GSCs, we discovered that loss of gsk-3 results in reduced GSC proliferation without changes in differentiation or responsiveness to GLP-1/Notch signaling. We find that DPL-1 transcriptional activity inhibits CDK-2 mRNA accumulation in GSCs, which leads to slower S-phase entry and progression. Inhibition of dpl-1 or transgenic expression of CDK-2 via a heterologous germline promoter rescues the S-phase entry and progression defects of the gsk-3 mutants, demonstrating that transcriptional regulation rather than post-translational control of CDK-2 establishes the abbreviated cell cycle structure in GSCs. This highlights an inhibitory cascade wherein GSK-3 inhibits DPL-1 and DPL-1 inhibits cdk-2 transcription. Constitutive GSK-3 activity through this cascade maintains an abbreviated cell cycle structure to permit the efficient proliferation of GSCs necessary for continuous tissue output.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/embriología , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Células Germinativas/citología , Glucógeno Sintasa Quinasa 3/metabolismo , Fase S/fisiología , Células Madre/citología , Factores de Transcripción/genética , Animales , Caenorhabditis elegans/citología , Proteínas de Caenorhabditis elegans/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Ciclina E/biosíntesis , Quinasa 2 Dependiente de la Ciclina/genética , Glucógeno Sintasa Quinasa 3/genética , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Transcripción Genética/genética
10.
Development ; 149(9)2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35535741

Asunto(s)
Gastrulación
12.
Mol Reprod Dev ; 84(6): 444-459, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28379636

RESUMEN

Insulin signaling regulates various aspects of physiology, such as glucose homeostasis and aging, and is a key determinant of female reproduction in metazoans. That insulin signaling is crucial for female reproductive health is clear from clinical data linking hyperinsulinemic and hypoinsulinemic condition with certain types of ovarian dysfunction, such as altered steroidogenesis, polycystic ovary syndrome, and infertility. Thus, understanding the signaling mechanisms that underlie the control of insulin-mediated ovarian development is important for the accurate diagnosis of and intervention for female infertility. Studies of invertebrate and vertebrate model systems have revealed the molecular determinants that transduce insulin signaling as well as which biological processes are regulated by the insulin-signaling pathway. The molecular determinants of the insulin-signaling pathway, from the insulin receptor to its downstream signaling components, are structurally and functionally conserved across evolution, from worms to mammals-yet, physiological differences in signaling still exist. Insulin signaling acts cooperatively with gonadotropins in mammals and lower vertebrates to mediate various aspects of ovarian development, mainly owing to evolution of the endocrine system in vertebrates. In contrast, insulin signaling in Drosophila and Caenorhabditis elegans directly regulates oocyte growth and maturation. In this review, we compare and contrast insulin-mediated regulation of ovarian functions in mammals, lower vertebrates, C. elegans, and Drosophila, and highlight conserved signaling pathways and regulatory mechanisms in general while illustrating insulin's unique role in specific reproductive processes.


Asunto(s)
Insulina/metabolismo , Oocitos/metabolismo , Transducción de Señal/fisiología , Animales , Caenorhabditis elegans , Drosophila melanogaster , Humanos
13.
Proc Natl Acad Sci U S A ; 110(40): 16033-8, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24046365

RESUMEN

How sexual regulators translate global sexual fate into appropriate local sexual differentiation events is perhaps the least understood aspect of sexual development. Here we have used ChIP followed by deep sequencing (ChIP-seq) to identify direct targets of the nematode global sexual regulator Transformer 1 (TRA-1), a transcription factor acting at the interface between organism-wide and cell-specific sexual regulation to control all sex-specific somatic differentiation events. We identified 184 TRA-1-binding sites in Caenorhabditis elegans, many with temporal- and/or tissue-specific TRA-1 association. We also identified 78 TRA-1-binding sites in the related nematode Caenorhabditis briggsae, 19 of which are conserved between the two species. Some DNA segments containing TRA-1-binding sites drive male-specific expression patterns, and RNAi depletion of some genes adjacent to TRA-1-binding sites results in defects in male sexual development. TRA-1 binds to sites adjacent to a number of heterochronic regulatory genes, some of which drive male-specific expression, suggesting that TRA-1 imposes sex specificity on developmental timing. We also found evidence for TRA-1 feedback regulation of the global sex-determination pathway: TRA-1 binds its own locus and those of multiple upstream masculinizing genes, and most of these associations are conserved in C. briggsae. Thus, TRA-1 coordinates sexual development by reinforcing the sex-determination decision and directing downstream sexual differentiation events.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Proteínas de Unión al ADN/fisiología , Retroalimentación Fisiológica/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Procesos de Determinación del Sexo/fisiología , Diferenciación Sexual/fisiología , Factores de Transcripción/fisiología , Animales , Proteínas de Caenorhabditis elegans/genética , Inmunoprecipitación de Cromatina , Biología Computacional , Proteínas de Unión al ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Interferencia de ARN , Análisis de Secuencia de ADN , Procesos de Determinación del Sexo/genética , Diferenciación Sexual/genética , Factores de Transcripción/genética
14.
Biophys J ; 109(11): 2436-45, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26636953

RESUMEN

The Caenorhabditis elegans germline is a well-studied model system for investigating the control of cell fate by signaling pathways. Cell signals at the distal tip of the germline promote cell proliferation; just before the loop, signals couple cell maturation to organism-level nutrient status; at the proximal end of the germline, signals coordinate oocyte maturation and fertilization in the presence of sperm. The latter two events require dual phosphorylation and activation of ERK, the effector molecule of the Ras/MAPK cascade. In C. elegans, ERK is known as MPK-1. At this point, none of today's methods for real-time monitoring of dually phosphorylated MPK-1 are working in the germline. Consequently, quantitative understanding of the MPK-1-dependent processes during germline development is limited. Here, we make a step toward advancing this understanding using a model-based framework that reconstructs the time course of MPK-1 activation from a snapshot of a fixed germline. Our approach builds on a number of recent studies for estimating temporal dynamics from fixed organisms, but takes advantage of the anatomy of the germline to simplify the analysis. Our model predicts that the MPK-1 signal turns on ∼30 h into germ cell progression and peaks ∼7 h later.


Asunto(s)
Caenorhabditis elegans/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Germinativas/metabolismo , Modelos Biológicos , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/enzimología , Activación Enzimática , Cinética , Sistema de Señalización de MAP Quinasas , Transporte de Proteínas
15.
MicroPubl Biol ; 20232023.
Artículo en Inglés | MEDLINE | ID: mdl-37206989

RESUMEN

Caenorhabditis elegans gene sart-3 was first identified as the homolog of human SART3 ( S quamous cell carcinoma A ntigen R ecognized by T -cells 3). In humans, expression of SART3 is associated with squamous cell carcinoma, thus most of the studies focus on its potential role as a target of cancer immunotherapy (Shichijo et al. 1998; Yang et al. 1999). Furthermore, SART3 is also known as Tip110 (Liu et al. 2002; Whitmill et al. 2016) in the context of HIV virus host activation pathway. Despite these disease related studies, the molecular function of this protein was not revealed until the yeast homolog was identified as spliceosome U4/U6 snRNP recycling factor (Bell et al. 2002). The function of SART3 in development, however, remains unknown. Here we report that the C. elegans sart-3 mutant hermaphrodites exhibit a Mog ( M asculinization O f the G ermline) phenotype in adulthood suggesting that sart-3 normally functions to regulate the switch from spermatogenic to oogenic gametic sex.

16.
Sci Adv ; 9(30): eadf6210, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37494452

RESUMEN

KRAS/ERK pathway phosphorylates DICER1, causing its nuclear translocation, and phosphomimetic Dicer1 contributes to tumorigenesis in mice. Mechanisms through which phospho-DICER1 regulates tumor progression remain undefined. While DICER1 canonically regulates microRNAs (miRNA) and epithelial-to-mesenchymal transition (EMT), we found that phosphorylated nuclear DICER1 (phospho-nuclear DICER1) promotes late-stage tumor progression in mice with oncogenic Kras, independent of miRNAs and EMT. Instead, we observe that the murine AT2 tumor cells exhibit altered chromatin compaction, and cells from disorganized advanced tumors, but not localized tumors, express gastric genes. Collectively, this results in subpopulations of tumor cells transitioning from a restricted alveolar to a broader endodermal lineage state. In human LUADs, we observed expression of phospho-nuclear DICER1 in advanced tumors together with the expression of gastric genes. We define a multimeric chromatin-DICER1 complex composed of the Mediator complex subunit 12, CBX1, MACROH2A.1, and transcriptional regulators supporting the model that phospho-nuclear DICER1 leads to lineage reprogramming of AT2 tumor cells to mediate lung cancer progression.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , MicroARNs , Humanos , Ratones , Animales , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Cromatina/genética , MicroARNs/genética , Adenocarcinoma del Pulmón/genética , Neoplasias Pulmonares/genética , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo
17.
Cell Rep ; 42(6): 112544, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37227820

RESUMEN

Maternal RNAs are stored from minutes to decades in oocytes throughout meiosis I arrest in a transcriptionally quiescent state. Recent reports, however, propose a role for nascent transcription in arrested oocytes. Whether arrested oocytes launch nascent transcription in response to environmental or hormonal signals while maintaining the meiosis I arrest remains undetermined. We test this by integrating single-cell RNA sequencing, RNA velocity, and RNA fluorescence in situ hybridization on C. elegans meiosis I arrested oocytes. We identify transcripts that increase as the arrested meiosis I oocyte ages, but rule out extracellular signaling through ERK MAPK and nascent transcription as a mechanism for this increase. We report transcript acquisition from neighboring somatic cells as a mechanism of transcript increase during meiosis I arrest. These analyses provide a deeper view at single-cell resolution of the RNA landscape of a meiosis I arrested oocyte and as it prepares for oocyte maturation and fertilization.


Asunto(s)
Caenorhabditis elegans , Oocitos , Animales , Caenorhabditis elegans/genética , Hibridación Fluorescente in Situ , Meiosis/genética , ARN
18.
Proc Natl Acad Sci U S A ; 106(12): 4776-81, 2009 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-19264959

RESUMEN

RAS-extracellular signal regulated kinase (ERK) signaling governs multiple aspects of cell fate specification, cellular transitions, and growth by regulating downstream substrates through phosphorylation. Understanding how perturbations to the ERK signaling pathway lead to developmental disorders and cancer hinges critically on identification of the substrates. Yet, only a limited number of substrates have been identified that function in vivo to execute ERK-regulated processes. The Caenorhabditis elegans germ line utilizes the well-conserved RAS-ERK signaling pathway in multiple different contexts. Here, we present an integrated functional genomic approach that identified 30 ERK substrates, each of which functions to regulate one or more of seven distinct biological processes during C. elegans germ-line development. Our results provide evidence for three themes that underlie the robustness and specificity of biological outcomes controlled by ERK signaling in C. elegans that are likely relevant to ERK signaling in other organisms: (i) multiple diverse ERK substrates function to control each individual biological process; (ii) different combinations of substrates function to control distinct biological processes; and (iii) regulatory feedback loops between ERK and its substrates help reinforce or attenuate ERK activation. Substrates identified here have conserved orthologs in humans, suggesting that insights from these studies will contribute to our understanding of human diseases involving deregulated ERK activity.


Asunto(s)
Caenorhabditis elegans/embriología , Caenorhabditis elegans/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Germinativas/enzimología , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/metabolismo , Biología Computacional , Activación Enzimática , Retroalimentación Fisiológica , Glucógeno Sintasa Quinasa 3/química , Glucógeno Sintasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas , Mamíferos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/química , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Datos de Secuencia Molecular , Fosforilación , Especificidad por Sustrato
19.
G3 (Bethesda) ; 12(5)2022 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-35389463

RESUMEN

Generation of functional gametes is accomplished through a multilayered and finely orchestrated succession of events during meiotic progression. In the Caenorhabditis elegans germline, the HORMA-domain-containing protein HTP-3 plays pivotal roles for the establishment of chromosome axes and the efficient induction of programmed DNA double-strand breaks, both of which are crucial for crossover formation. Double-strand breaks allow for accurate chromosome segregation during the first meiotic division and therefore are an essential requirement for the production of healthy gametes. Phosphorylation-dependent regulation of HORMAD protein plays important roles in controlling meiotic chromosome behavior. Here, we document a phospho-site in HTP-3 at Serine 285 that is constitutively phosphorylated during meiotic prophase I. pHTP-3S285 localization overlaps with panHTP-3 except in nuclei undergoing physiological apoptosis, in which pHTP-3 is absent. Surprisingly, we observed that phosphorylation of HTP-3 at S285 is independent of the canonical kinases that control meiotic progression in nematodes. During meiosis, the htp-3(S285A) mutant displays accelerated RAD-51 turnover, but no other meiotic abnormalities. Altogether, these data indicate that the Ser285 phosphorylation is independent of canonical meiotic protein kinases and does not regulate HTP-3-dependent meiotic processes. We propose a model wherein phosphorylation of HTP-3 occurs through noncanonical or redundant meiotic kinases and/or is likely redundant with additional phospho-sites for function in vivo.


Asunto(s)
Proteínas de Caenorhabditis elegans , Meiosis , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/genética , Segregación Cromosómica , Fosforilación , Serina/metabolismo , Complejo Sinaptonémico/metabolismo
20.
MicroPubl Biol ; 20212021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33688627

RESUMEN

CRISPR-Cas9 mediated genome editing is widely used for generating genetic lesions in C. elegans. Detection of single-site mutations in F1 progeny after CRISPR-Cas9 injections is currently labor intensive due to lack of a single step PCR-based detection method. Here we present CEPAD-PCR, an allele-specific PCR detection method based on generating silent mutations around the site of the desired genetic lesion during the CRISPR-Cas9 genome editing process. Detection of the desired allele is then performed by taking advantage of the tetra primer PCR method, based on the principle described in the ARMS-PCR. In the CEPAD-PCR, however, unlike ARMS-PCR, presence of additional silent mutations near the desired site-specific mutation in the genome results in PCR priming with high specificity resulting in a low false positive rate. As proof of concept, the method was successfully tested on point mutations in two different genes, daf-15 and raga-1.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA