Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Mol Cell ; 78(1): 96-111.e6, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32105612

RESUMO

Current models suggest that chromosome domains segregate into either an active (A) or inactive (B) compartment. B-compartment chromatin is physically separated from the A compartment and compacted by the nuclear lamina. To examine these models in the developmental context of C. elegans embryogenesis, we undertook chromosome tracing to map the trajectories of entire autosomes. Early embryonic chromosomes organized into an unconventional barbell-like configuration, with two densely folded B compartments separated by a central A compartment. Upon gastrulation, this conformation matured into conventional A/B compartments. We used unsupervised clustering to uncover subpopulations with differing folding properties and variable positioning of compartment boundaries. These conformations relied on tethering to the lamina to stretch the chromosome; detachment from the lamina compacted, and allowed intermingling between, A/B compartments. These findings reveal the diverse conformations of early embryonic chromosomes and uncover a previously unappreciated role for the lamina in systemic chromosome stretching.


Assuntos
Caenorhabditis elegans/genética , Cromossomos/química , Lâmina Nuclear/fisiologia , Animais , Caenorhabditis elegans/embriologia , Cromossomos/ultraestrutura , Embrião não Mamífero/ultraestrutura , Gastrulação/genética , Hibridização in Situ Fluorescente , Conformação Molecular
2.
PLoS Biol ; 22(3): e3002526, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38427703

RESUMO

Live imaging of RNA molecules constitutes an invaluable means to track the dynamics of mRNAs, but live imaging in Caenorhabditis elegans has been difficult to achieve. Endogenous transcripts have been observed in nuclei, but endogenous mRNAs have not been detected in the cytoplasm, and functional mRNAs have not been generated. Here, we have adapted live imaging methods to visualize mRNA in embryonic cells. We have tagged endogenous transcripts with MS2 hairpins in the 3' untranslated region (UTR) and visualized them after adjusting MS2 Coat Protein (MCP) expression. A reduced number of these transcripts accumulates in the cytoplasm, leading to loss-of-function phenotypes. In addition, during epithelial morphogenesis, MS2-tagged mRNAs for dlg-1 fail to associate with the adherens junction, as observed for untagged, endogenous mRNAs. These defects are reversed by inactivating the nonsense-mediated decay pathway. RNA accumulates in the cytoplasm, mutant phenotypes are rescued, and dlg-1 RNA associates with the adherens junction. These data suggest that MS2 repeats can induce the degradation of endogenous RNAs and alter their cytoplasmic distribution. Although our focus is RNAs expressed in epithelial cells during morphogenesis, we find that this method can be applied to other cell types and stages.


Assuntos
Caenorhabditis elegans , RNA , Animais , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , RNA/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Citosol/metabolismo
3.
Development ; 148(24)2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34846063

RESUMO

mRNA localization is an evolutionarily widespread phenomenon that can facilitate subcellular protein targeting. Extensive work has focused on mRNA targeting through 'zip-codes' within untranslated regions (UTRs), whereas much less is known about translation-dependent cues. Here, we examine mRNA localization in Caenorhabditis elegans embryonic epithelia. From an smFISH-based survey, we identified mRNAs associated with the cell membrane or cortex, and with apical junctions in a stage- and cell type-specific manner. Mutational analyses for one of these transcripts, dlg-1/discs large, revealed that it relied on a translation-dependent process and did not require its 5' or 3' UTRs. We suggest a model in which dlg-1 transcripts are co-translationally localized with the nascent protein: first the translating complex goes to the cell membrane using sequences located at the C-terminal/3' end, and then apically using N-terminal/5' sequences. These studies identify a translation-based process for mRNA localization within developing epithelia and determine the necessary cis-acting sequences for dlg-1 mRNA targeting.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Membrana Celular/genética , Desenvolvimento Embrionário/genética , Guanilato Quinases/genética , Biossíntese de Proteínas/genética , Junções Aderentes/genética , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Polaridade Celular/genética , Embrião não Mamífero/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Junções Intercelulares/genética , Proteínas de Membrana/genética , Transporte Proteico/genética , RNA Mensageiro/genética , Homologia de Sequência de Aminoácidos
4.
Annu Rev Cell Dev Biol ; 25: 597-628, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19575642

RESUMO

The digestive tracts of many animals are epithelial tubes with specialized compartments to break down food, remove wastes, combat infection, and signal nutrient availability. C. elegans possesses a linear, epithelial gut tube with foregut, midgut, and hindgut sections. The simple anatomy belies the developmental complexity that is involved in forming the gut from a pool of heterogeneous precursor cells. Here, I focus on the processes that specify cell fates and control morphogenesis within the embryonic foregut (pharynx) and the developmental roles of the pharynx after birth. Maternally donated factors in the pregastrula embryo converge on pha-4, a FoxA transcription factor that specifies organ identity for pharyngeal precursors. Positive feedback loops between PHA-4 and other transcription factors ensure commitment to pharyngeal fate. Binding-site affinity of PHA-4 for its target promoters contributes to the progression of the pharyngeal precursors towards differentiation. During morphogenesis, the pharyngeal precursors form an epithelial tube in a process that is independent of cadherins, catenins, and integrins but requires the kinesin zen-4/MKLP1. After birth, the pharynx and/or pha-4 are involved in repelling pathogens and controlling aging.


Assuntos
Caenorhabditis elegans/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Sistema Digestório/citologia , Sistema Digestório/embriologia , Embrião não Mamífero/metabolismo , Organogênese , Transativadores/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(26): 14636-14641, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32541064

RESUMO

Understanding the coordination of cell-division timing is one of the outstanding questions in the field of developmental biology. One active control parameter of the cell-cycle duration is temperature, as it can accelerate or decelerate the rate of biochemical reactions. However, controlled experiments at the cellular scale are challenging, due to the limited availability of biocompatible temperature sensors, as well as the lack of practical methods to systematically control local temperatures and cellular dynamics. Here, we demonstrate a method to probe and control the cell-division timing in Caenorhabditis elegans embryos using a combination of local laser heating and nanoscale thermometry. Local infrared laser illumination produces a temperature gradient across the embryo, which is precisely measured by in vivo nanoscale thermometry using quantum defects in nanodiamonds. These techniques enable selective, controlled acceleration of the cell divisions, even enabling an inversion of division order at the two-cell stage. Our data suggest that the cell-cycle timing asynchrony of the early embryonic development in C. elegans is determined independently by individual cells rather than via cell-to-cell communication. Our method can be used to control the development of multicellular organisms and to provide insights into the regulation of cell-division timings as a consequence of local perturbations.


Assuntos
Temperatura Corporal/fisiologia , Divisão Celular/fisiologia , Desenvolvimento Embrionário/fisiologia , Pontos Quânticos/química , Termometria , Animais , Caenorhabditis elegans/embriologia , Nanodiamantes/química , Termometria/instrumentação , Termometria/métodos
6.
Development ; 146(19)2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31540912

RESUMO

During the first hours of embryogenesis, formation of higher-order heterochromatin coincides with the loss of developmental potential. Here, we examine the relationship between these two events, and we probe the processes that contribute to the timing of their onset. Mutations that disrupt histone H3 lysine 9 (H3K9) methyltransferases reveal that the methyltransferase MET-2 helps terminate developmental plasticity, through mono- and di-methylation of H3K9 (me1/me2), and promotes heterochromatin formation, through H3K9me3. Although loss of H3K9me3 perturbs formation of higher-order heterochromatin, embryos are still able to terminate plasticity, indicating that the two processes can be uncoupled. Methylated H3K9 appears gradually in developing C. elegans embryos and depends on nuclear localization of MET-2. We find that the timing of H3K9me2 and nuclear MET-2 is sensitive to rapid cell cycles, but not to zygotic genome activation or cell counting. These data reveal distinct roles for different H3K9 methylation states in the generation of heterochromatin and loss of developmental plasticity by MET-2, and identify the cell cycle as a crucial parameter of MET-2 regulation.


Assuntos
Caenorhabditis elegans/embriologia , Caenorhabditis elegans/metabolismo , Desenvolvimento Embrionário , Histonas/metabolismo , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Contagem de Células , Embrião não Mamífero/citologia , Gastrulação , Heterocromatina/metabolismo , Interfase , Metilação , RNA Polimerase II/metabolismo , Fase S , Fatores de Tempo , Transcrição Gênica
7.
Dev Biol ; 403(1): 5-14, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25773364

RESUMO

Cell polarity is a fundamental characteristic of epithelial cells. Classical cell biological studies have suggested that establishment and orientation of polarized epithelia depend on outside-in cues that derive from interactions with either neighboring cells or the substratum (Akhtar and Streuli, 2013; Chen and Zhang, 2013; Chung and Andrew, 2008; McNeill et al., 1990; Nejsum and Nelson, 2007; Nelson et al., 2013; Ojakian and Schwimmer, 1994; Wang et al., 1990; Yu et al., 2005). This paradigm has been challenged by examples of epithelia generated in the absence of molecules that mediate cell-cell or cell-matrix interactions, notably E-cadherin and integrins (Baas et al., 2004; Choi et al., 2013; Costa et al., 1998; Harris and Peifer, 2004; Raich et al., 1999; Roote and Zusman, 1995; Vestweber et al., 1985; Williams and Waterston, 1994; Wu et al., 2009). Here we explore an alternative hypothesis, that cadherins and integrins function redundantly to substitute for one another during epithelium formation (Martinez-Rico et al., 2010; Ojakian et al., 2001; Rudkouskaya et al., 2014; Weber et al., 2011). We use C. elegans, which possesses a single E-cadherin (Costa et al., 1998; Hardin et al., 2013; Tepass, 1999) and a single ß-integrin (Gettner et al., 1995; Lee et al., 2001), and analyze the arcade cells, which generate an epithelium late in embryogenesis (Portereiko and Mango, 2001; Portereiko et al., 2004), after most maternal factors are depleted. Loss of E-cadherin(HMR-1) in combination with ß-integrin(PAT-3) had no impact on the onset or formation of the arcade cell epithelium, nor the epidermis or digestive tract. Moreover, ß-integrin(PAT-3) was not enriched at the basal surface of the arcades, and the candidate PAT-3 binding partner ß-laminin(LAM-1) was not detected until after arcade cell polarity was established and exhibited no obvious polarity defect when mutated. Instead, the polarity protein par-6 (Chen and Zhang, 2013; Watts et al., 1996) was required to polarize the arcade cells, and par-6 mutants exhibited mislocalized or absent apical and junctional proteins. We conclude that the arcade cell epithelium polarizes by a PAR-6-mediated pathway that is independent of E-cadherin, ß-integrin and ß-laminin.


Assuntos
Caderinas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Polaridade Celular/fisiologia , Cadeias beta de Integrinas/metabolismo , Animais , Caderinas/genética , Caenorhabditis elegans/embriologia , Proteínas de Caenorhabditis elegans/genética , Células Epiteliais/fisiologia , Cadeias beta de Integrinas/genética , Laminina/genética , Laminina/metabolismo , Interferência de RNA , RNA Interferente Pequeno
8.
Biochim Biophys Acta ; 1839(12): 1440-53, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24859459

RESUMO

Recent studies have discovered phenotypes induced by a transient treatment or mutation that persist for multiple generations without mutations in DNA. Both invertebrates and vertebrates exhibit such inheritance, and a range of environmental factors can act as a trigger. Now referred to as transgenerational epigenetic inheritance or TEI, this emerging field faces a big challenge-what molecular mechanisms account for inheritance of TEI phenotypes? This review describes examples of TEI and focuses on the possible role of histone methylation and small RNAs in mediating TEI.


Assuntos
Epigênese Genética/fisiologia , Interação Gene-Ambiente , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Padrões de Herança , Animais , Metilação de DNA , Aptidão Genética , Histona Metiltransferases , Humanos , Padrões de Herança/genética , Transdução de Sinais/genética
9.
Genome Biol ; 25(1): 71, 2024 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-38486337

RESUMO

Recent advances in microscopy have enabled studying chromosome organization at the single-molecule level, yet little is known about inherited chromosome organization. Here we adapt single-molecule chromosome tracing to distinguish two C. elegans strains (N2 and HI) and find that while their organization is similar, the N2 chromosome influences the folding parameters of the HI chromosome, in particular the step size, across generations. Furthermore, homologous chromosomes overlap frequently, but alignment between homologous regions is rare, suggesting that transvection is unlikely. We present a powerful tool to investigate chromosome architecture and to track the parent of origin.


Assuntos
Caenorhabditis elegans , Cromossomos , Animais , Hibridização in Situ Fluorescente , Caenorhabditis elegans/genética , Cromossomos/genética , DNA/genética
10.
PLoS Genet ; 6(8)2010 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-20714352

RESUMO

Central regulators of cell fate, or selector genes, establish the identity of cells by direct regulation of large cohorts of genes. In Caenorhabditis elegans, foregut (or pharynx) identity relies on the FoxA transcription factor PHA-4, which activates different sets of target genes at various times and in diverse cellular environments. An outstanding question is how PHA-4 distinguishes between target genes for appropriate transcriptional control. We have used the Nuclear Spot Assay and GFP reporters to examine PHA-4 interactions with target promoters in living embryos and with single cell resolution. While PHA-4 was found throughout the digestive tract, binding and activation of pharyngeally expressed promoters was restricted to a subset of pharyngeal cells and excluded from the intestine. An RNAi screen of candidate nuclear factors identified emerin (emr-1) as a negative regulator of PHA-4 binding within the pharynx, but emr-1 did not modulate PHA-4 binding in the intestine. Upon promoter association, PHA-4 induced large-scale chromatin de-compaction, which, we hypothesize, may facilitate promoter access and productive transcription. Our results reveal two tiers of PHA-4 regulation. PHA-4 binding is prohibited in intestinal cells, preventing target gene expression in that organ. PHA-4 binding within the pharynx is limited by the nuclear lamina component EMR-1/emerin. The data suggest that association of PHA-4 with its targets is a regulated step that contributes to promoter selectivity during organ formation. We speculate that global re-organization of chromatin architecture upon PHA-4 binding promotes competence of pharyngeal gene transcription and, by extension, foregut development.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/metabolismo , Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Transativadores/metabolismo , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Cromatina/genética , Sistema Digestório/crescimento & desenvolvimento , Sistema Digestório/metabolismo , Especificidade de Órgãos , Faringe/crescimento & desenvolvimento , Faringe/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Transativadores/genética
11.
PLoS Genet ; 6(2): e1000848, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20174564

RESUMO

Transcription factors are key components of regulatory networks that control development, as well as the response to environmental stimuli. We have established an experimental pipeline in Caenorhabditis elegans that permits global identification of the binding sites for transcription factors using chromatin immunoprecipitation and deep sequencing. We describe and validate this strategy, and apply it to the transcription factor PHA-4, which plays critical roles in organ development and other cellular processes. We identified thousands of binding sites for PHA-4 during formation of the embryonic pharynx, and also found a role for this factor during the starvation response. Many binding sites were found to shift dramatically between embryos and starved larvae, from developmentally regulated genes to genes involved in metabolism. These results indicate distinct roles for this regulator in two different biological processes and demonstrate the versatility of transcription factors in mediating diverse biological roles.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/genética , Meio Ambiente , Genoma Helmíntico/genética , Transativadores/metabolismo , Animais , Sítios de Ligação , Proteínas de Caenorhabditis elegans/genética , Imunoprecipitação da Cromatina , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes de Helmintos/genética , Proteínas de Fluorescência Verde/metabolismo , Larva/metabolismo , Ligação Proteica , RNA Polimerase II/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Inanição , Análise de Sobrevida , Transativadores/genética , Fatores de Transcrição/metabolismo
13.
Curr Opin Genet Dev ; 75: 101939, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35759905

RESUMO

Eukaryotic genome organization is ordered and multilayered, from the nucleosome to chromosomal scales. These layers are not static during development, but are remodeled over time and between tissues. Thus, animal model studies with high spatiotemporal resolution are necessary to understand the various forms and functions of genome organization in vivo. In C. elegans, sequencing- and imaging-based advances have provided insight on how histone modifications, regulatory elements, and large-scale chromosome conformations are established and changed. Recent observations include unexpected physiological roles for topologically associating domains, different roles for the nuclear lamina at different chromatin scales, cell-type-specific enhancer and promoter regulatory grammars, and prevalent compartment variability in early development. Here, we summarize these and other recent findings in C. elegans, and suggest future avenues of research to enrich our in vivo knowledge of the forms and functions of nuclear organization.


Assuntos
Caenorhabditis elegans , Cromatina , Animais , Caenorhabditis elegans/genética , Cromatina/genética , Genoma/genética , Nucleossomos/genética , Regiões Promotoras Genéticas
14.
Curr Biol ; 18(18): 1355-64, 2008 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-18804378

RESUMO

BACKGROUND: FoxA factors are critical regulators of embryonic development and postembryonic life, but little is know about the upstream pathways that modulate their activity. C. elegans pha-4 encodes a FoxA transcription factor that is required to establish the foregut in embryos and to control growth and longevity after birth. We previously identified the AAA+ ATPase homolog ruvb-1 as a potent suppressor of pha-4 mutations. RESULTS: Here we show that ruvb-1 is a component of the Target of Rapamycin (TOR) pathway in C. elegans (CeTOR). Both ruvb-1 and let-363/TOR control nucleolar size and promote localization of box C/D snoRNPs to nucleoli, suggesting a role in rRNA maturation. Inactivation of let-363/TOR or ruvb-1 suppresses the lethality associated with reduced pha-4 activity. The CeTOR pathway controls protein homeostasis and also contributes to adult longevity. We find that pha-4 is required to extend adult lifespan in response to reduced CeTOR signaling. Mutations in the predicted CeTOR target rsks-1/S6 kinase or in ife-2/eIF4E also reduce protein biosynthesis and extend lifespan, but only rsks-1 mutations require pha-4 for adult longevity. In addition, rsks-1, but not ife-2, can suppress the larval lethality associated with pha-4 loss-of-function mutations. CONCLUSIONS: The data suggest that pha-4 and the CeTOR pathway antagonize one another to regulate postembryonic development and adult longevity. We suggest a model in which nutrients promote TOR and S6 kinase signaling, which represses pha-4/FoxA, leading to a shorter lifespan. A similar regulatory hierarchy may function in other animals to modulate metabolism, longevity, or disease.


Assuntos
Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Transativadores/antagonistas & inibidores , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/genética , Larva/fisiologia , Longevidade , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/fisiologia , Fenótipo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , RNA/genética , Interferência de RNA , Ribonucleoproteínas/fisiologia , Transativadores/genética
15.
Sci Adv ; 7(34)2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34417172

RESUMO

Mothers contribute cytoplasmic components to their progeny in a process called maternal provisioning. Provisioning is influenced by the parental environment, but the molecular pathways that transmit environmental cues between generations are not well understood. Here, we show that, in Caenorhabditis elegans, social cues modulate maternal provisioning to regulate gene silencing in offspring. Intergenerational signal transmission depends on a pheromone-sensing neuron and neuronal FMRFamide (Phe-Met-Arg-Phe)-like peptides. Parental FMRFamide-like peptide signaling dampens oxidative stress resistance and promotes the deposition of mRNAs for translational components in progeny, which, in turn, reduces gene silencing. This study identifies a previously unknown pathway for intergenerational communication that links neuronal responses to maternal provisioning. We suggest that loss of social cues in the parental environment represents an adverse environment that stimulates stress responses across generations.

16.
STAR Protoc ; 1(3): 100107, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33377003

RESUMO

This protocol describes a high-throughput and multiplexed DNA fluorescence in situ hybridization method to trace chromosome conformation in Caenorhabditis elegans embryos. This approach generates single-cell and single-chromosome localization data that can be used to determine chromosome conformation and assess the heterogeneity of structures that exist in vivo. This strategy is flexible through modifications to the probe design steps to interrogate chromosome structure at the desired genomic scale (small-scale loops to whole-chromosome organization). For complete details on the use and execution of this protocol, please refer to Sawh et al. (2020).


Assuntos
Cromossomos/genética , Hibridização in Situ Fluorescente/métodos , Animais , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , DNA/genética , Primers do DNA/genética , Genoma/genética , Genômica/métodos , Conformação Molecular , Análise de Célula Única
17.
PLoS Genet ; 2(9): e161, 2006 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-17009877

RESUMO

The histone variant H2A.Z is evolutionarily conserved and plays an essential role in mice, Drosophila, and Tetrahymena. The essential function of H2A.Z is unknown, with some studies suggesting a role in transcriptional repression and others in activation. Here we show that Caenorhabditis elegans HTZ-1/H2A.Z and the remodeling complex MYS-1/ESA1-SSL-1/SWR1 synergize with the FoxA transcription factor PHA-4 to coordinate temporal gene expression during foregut development. We observe dramatic genetic interactions between pha-4 and htz-1, mys-1, and ssl-1. A survey of transcription factors reveals that this interaction is specific, and thus pha-4 is acutely sensitive to reductions in these three proteins. Using a nuclear spot assay to visualize HTZ-1 in living embryos as organogenesis proceeds, we show that HTZ-1 is recruited to foregut promoters at the time of transcriptional onset, and this recruitment requires PHA-4. Loss of htz-1 by RNAi is lethal and leads to delayed expression of a subset of foregut genes. Thus, the effects of PHA-4 on temporal regulation can be explained in part by recruitment of HTZ-1 to target promoters. We suggest PHA-4 and HTZ-1 coordinate temporal gene expression by modulating the chromatin environment.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/embriologia , Sistema Digestório/embriologia , Histonas/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Transativadores/fisiologia , Adenosina Trifosfatases/fisiologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ligação a DNA/metabolismo , Sistema Digestório/metabolismo , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Histona Acetiltransferases/fisiologia , Histonas/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Faringe/embriologia , Faringe/metabolismo , Regiões Promotoras Genéticas , Proteínas de Saccharomyces cerevisiae/metabolismo , Homologia de Sequência de Aminoácidos , Transativadores/metabolismo
18.
Curr Biol ; 15(10): 894-904, 2005 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-15916946

RESUMO

BACKGROUND: The Aurora kinases control multiple aspects of mitosis, among them centrosome maturation, spindle assembly, chromosome segregation, and cytokinesis. Aurora activity is regulated in part by a subset of Aurora substrates that, once phosphorylated, can enhance Aurora kinase activity. Aurora A substrate activators include TPX2 and Ajuba, whereas the only known Aurora B substrate activator is the chromosomal passenger INCENP. RESULTS: We report that the C. elegans Tousled kinase TLK-1 is a second substrate activator of the Aurora B kinase AIR-2. Tousled kinase (Tlk) expression and activity have been linked to ongoing DNA replication, and Tlk can phosphorylate the chromatin assembly factor Asf. Here, we show that TLK-1 is phosphorylated by AIR-2 during prophase/prometaphase and that phosphorylation increases TLK-1 kinase activity in vitro. Phosphorylated TLK-1 increases AIR-2 kinase activity in a manner that is independent of TLK-1 kinase activity but depends on the presence of ICP-1/INCENP. In vivo, TLK-1 and AIR-2 cooperate to ensure proper mitotic chromosome segregation. CONCLUSIONS: The C. elegans Tousled kinase TLK-1 is a substrate and activator of the Aurora B kinase AIR-2. These results suggest that Tousled kinases have a previously unrecognized role in mitosis and that Aurora B associates with discrete regulatory complexes that may impart distinct substrate specificities and functions to the Aurora B kinase.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Segregação de Cromossomos/fisiologia , Mitose/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Aurora Quinase B , Western Blotting , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas Cromossômicas não Histona/metabolismo , DNA Complementar/genética , Imuno-Histoquímica , Imunoprecipitação , Fosforilação , Plasmídeos/genética , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Análise de Sequência de DNA , Técnicas do Sistema de Duplo-Híbrido
19.
Genetics ; 177(2): 819-33, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17720918

RESUMO

FoxA transcription factors are critical regulators of gut development and function. FoxA proteins specify gut fate during early embryogenesis, drive gut differentiation and morphogenesis at later stages, and affect gut function to mediate nutritional responses. The level of FoxA is critical for these roles, yet we know relatively little about regulators for this family of proteins. To address this issue, we conducted a genetic screen for mutants that suppress a partial loss of pha-4, the sole FoxA factor of Caenorhabditis elegans. We identified 55 mutants using either chemical or insertional mutagenesis. Forty-two of these were informational suppressors that affected nonsense-mediated decay, while the remaining 13 were pha-4 suppressors. These 13 alleles defined at least six different loci. On the basis of mutational frequencies for C. elegans and the genetic dominance of four of the suppressors, we predict that many of the suppressors are either unusual loss-of-function mutations in negative regulators or rare gain-of-function mutations in positive regulators. We characterized one dominant suppressor molecularly and discovered the mutation alters a likely cis-regulatory region within pha-4 itself. A second suppressor defined a new locus, the predicted AAA+ helicase ruvb-1. These results indicate that our screen successfully found cis- or trans-acting regulators of pha-4.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , DNA Helicases/fisiologia , Transativadores/fisiologia , Animais , Proteínas de Caenorhabditis elegans/genética , Genes de Helmintos , Genes Supressores , Técnicas Genéticas
20.
Sci Adv ; 4(8): eaat6224, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30140741

RESUMO

Heterochromatin formation during early embryogenesis is timed precisely, but how this process is regulated remains elusive. We report the discovery of a histone methyltransferase complex whose nuclear accumulation and activation establish the onset of heterochromatin formation in Caenorhabditis elegans embryos. We find that the inception of heterochromatin generation coincides with the accumulation of the histone H3 lysine 9 (H3K9) methyltransferase MET-2 (SETDB) into nuclear hubs. The absence of MET-2 results in delayed and disturbed heterochromatin formation, whereas accelerated nuclear localization of the methyltransferase leads to precocious H3K9 methylation. We identify two factors that bind to and function with MET-2: LIN-65, which resembles activating transcription factor 7-interacting protein (ATF7IP) and localizes MET-2 into nuclear hubs, and ARLE-14, which is orthologous to adenosine 5'-diphosphate-ribosylation factor-like 14 effector protein (ARL14EP) and promotes stable association of MET-2 with chromatin. These data reveal that nuclear accumulation of MET-2 in conjunction with LIN-65 and ARLE-14 regulates timing of heterochromatin domains during embryogenesis.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Núcleo Celular/metabolismo , Metilação de DNA , Heterocromatina/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Animais , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Núcleo Celular/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Heterocromatina/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA