Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(40): e2209471119, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36161922

RESUMO

The transmission of chromatin states from parent cells to daughter cells preserves cell-specific transcriptional states and thus cell identity through cell division. The mechanism that underpins this process is not fully understood. The role that chromatin states serve in transmitting gene expression information across generations via sperm and oocytes is even less understood. Here, we utilized a model in which Caenorhabditis elegans sperm and oocyte alleles were inherited in different states of the repressive mark H3K27me3. This resulted in the alleles achieving different transcriptional states within the nuclei of offspring. Using this model, we showed that sperm alleles inherited without H3K27me3 were sensitive to up-regulation in offspring somatic and germline tissues, and tissue context determined which genes were up-regulated. We found that the subset of sperm alleles that were up-regulated in offspring germlines retained the H3K27me3(-) state and were transmitted to grandoffspring as H3K27me3(-) and up-regulated epialleles, demonstrating that H3K27me3 can serve as a transgenerational epigenetic carrier in C. elegans.


Assuntos
Alelos , Caenorhabditis elegans , Epigênese Genética , Histonas , Espermatozoides , Animais , Caenorhabditis elegans/genética , Cromatina/metabolismo , Histonas/genética , Masculino , Oócitos/metabolismo , Sêmen/metabolismo , Espermatozoides/metabolismo
2.
Cell Rep ; 30(13): 4321-4331.e4, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32234470

RESUMO

Cellular translation surveillance rescues ribosomes that stall on problematic mRNAs. During translation surveillance, endonucleolytic cleavage of the problematic mRNA is a critical step in rescuing stalled ribosomes. Here we identify NONU-1 as a factor required for translation surveillance pathways including no-go and nonstop mRNA decay. We show that (1) NONU-1 reduces nonstop and no-go mRNA levels; (2) NONU-1 contains an Smr RNase domain required for mRNA decay; (3) the domain architecture and catalytic residues of NONU-1 are conserved throughout metazoans and eukaryotes, respectively; and (4) NONU-1 is required for the formation of mRNA cleavage fragments in the vicinity of stalled ribosomes. We extend our results in C. elegans to homologous factors in S. cerevisiae, showing the evolutionarily conserved function of NONU-1. Our work establishes the identity of a factor critical to translation surveillance and will inform mechanistic studies at the intersection of translation and mRNA decay.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Sequência Conservada , Endonucleases/metabolismo , Biossíntese de Proteínas , Sequência de Aminoácidos , Animais , Biocatálise , Evolução Molecular , Domínios Proteicos , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribossomos/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo
3.
Genetics ; 212(1): 125-140, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30910798

RESUMO

Repression of germline-promoting genes in somatic cells is critical for somatic development and function. To study how germline genes are repressed in somatic tissues, we analyzed key histone modifications in three Caenorhabditis elegans synMuv B mutants, lin-15B, lin-35, and lin-37-all of which display ectopic expression of germline genes in the soma. LIN-35 and LIN-37 are members of the conserved DREAM complex. LIN-15B has been proposed to work with the DREAM complex but has not been shown biochemically to be a member of the complex. We found that, in wild-type worms, synMuv B target genes and germline genes are enriched for the repressive histone modification dimethylation of histone H3 on lysine 9 (H3K9me2) at their promoters. Genes with H3K9me2 promoter localization are evenly distributed across the autosomes, not biased toward autosomal arms, as are the broad H3K9me2 domains. Both synMuv B targets and germline genes display a dramatic reduction of H3K9me2 promoter localization in lin-15B mutants, but much weaker reduction in lin-35 and lin-37 mutants. This difference between lin-15B and DREAM complex mutants likely represents a difference in molecular function for these synMuv B proteins. In support of the pivotal role of H3K9me2 in regulation of germline genes by LIN-15B, global loss of H3K9me2 but not H3K9me3 results in phenotypes similar to synMuv B mutants, high-temperature larval arrest, and ectopic expression of germline genes in the soma. We propose that LIN-15B-driven enrichment of H3K9me2 at promoters of germline genes contributes to repression of those genes in somatic tissues.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Animais , Caenorhabditis elegans/metabolismo , Células Germinativas , Metilação
4.
Nat Commun ; 9(1): 4310, 2018 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-30333496

RESUMO

Paternal contributions to epigenetic inheritance are not well understood. Paternal contributions via marked nucleosomes are particularly understudied, in part because sperm in some organisms replace the majority of nucleosome packaging with protamine packaging. Here we report that in Caenorhabditis elegans sperm, the genome is packaged in nucleosomes and carries a histone-based epigenetic memory of genes expressed during spermatogenesis, which unexpectedly include genes well known for their expression during oogenesis. In sperm, genes with spermatogenesis-restricted expression are uniquely marked with both active and repressive marks, which may reflect a sperm-specific chromatin signature. We further demonstrate that epigenetic information provided by sperm is important and in fact sufficient to guide proper germ cell development in offspring. This study establishes one mode of paternal epigenetic inheritance and offers a potential mechanism for how the life experiences of fathers may impact the development and health of their descendants.


Assuntos
Caenorhabditis elegans/metabolismo , Epigênese Genética , Histonas/metabolismo , Nucleossomos/metabolismo , Espermatozoides/metabolismo , Animais , Caenorhabditis elegans/crescimento & desenvolvimento , Fertilidade , Masculino , Oogênese , Espermatogênese
5.
Nature ; 512(7515): 449-52, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25164756

RESUMO

Genome function is dynamically regulated in part by chromatin, which consists of the histones, non-histone proteins and RNA molecules that package DNA. Studies in Caenorhabditis elegans and Drosophila melanogaster have contributed substantially to our understanding of molecular mechanisms of genome function in humans, and have revealed conservation of chromatin components and mechanisms. Nevertheless, the three organisms have markedly different genome sizes, chromosome architecture and gene organization. On human and fly chromosomes, for example, pericentric heterochromatin flanks single centromeres, whereas worm chromosomes have dispersed heterochromatin-like regions enriched in the distal chromosomal 'arms', and centromeres distributed along their lengths. To systematically investigate chromatin organization and associated gene regulation across species, we generated and analysed a large collection of genome-wide chromatin data sets from cell lines and developmental stages in worm, fly and human. Here we present over 800 new data sets from our ENCODE and modENCODE consortia, bringing the total to over 1,400. Comparison of combinatorial patterns of histone modifications, nuclear lamina-associated domains, organization of large-scale topological domains, chromatin environment at promoters and enhancers, nucleosome positioning, and DNA replication patterns reveals many conserved features of chromatin organization among the three organisms. We also find notable differences in the composition and locations of repressive chromatin. These data sets and analyses provide a rich resource for comparative and species-specific investigations of chromatin composition, organization and function.


Assuntos
Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Cromatina/genética , Cromatina/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Animais , Linhagem Celular , Centrômero/genética , Centrômero/metabolismo , Cromatina/química , Montagem e Desmontagem da Cromatina/genética , Replicação do DNA/genética , Elementos Facilitadores Genéticos/genética , Epigênese Genética , Heterocromatina/química , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Anotação de Sequência Molecular , Lâmina Nuclear/metabolismo , Nucleossomos/química , Nucleossomos/genética , Nucleossomos/metabolismo , Regiões Promotoras Genéticas/genética , Especificidade da Espécie
6.
Curr Biol ; 24(9): 970-5, 2014 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-24746798

RESUMO

Specialized ribonucleoprotein organelles collectively known as germ granules are found in the germline cytoplasm from worms to humans [1]. In Drosophila, germ granules have been implicated in germline determination [2]. C. elegans germ granules, known as P granules, do not appear to be required for primordial germ cell (PGC) determination [3], but their components are still needed for fertility [4-6]. One potential role for P granules is to maintain germline fate and totipotency. This is suggested by the loss of P granules from germ cells that transform into somatic cell types, e.g., in germlines lacking MEX-3 and GLD-1 or upon neuronal induction by CHE-1 [7, 8]. However, it has not been established whether loss of P granules is the cause or effect of cell fate transformation. To test cause and effect, we severely compromised P granules by simultaneously knocking down factors that nucleate granule formation (PGL-1 and PGL-3) and promote their perinuclear localization (GLH-1 and GLH-4) [9] and investigated whether this causes germ cells to lose totipotency and initiate somatic reprogramming. We found that compromising P granules causes germ cells to express neuronal and muscle markers and send out neurite-like projections, suggesting that P granules maintain totipotency and germline identity by antagonizing somatic fate.


Assuntos
Caenorhabditis elegans/embriologia , Grânulos Citoplasmáticos/genética , Grânulos Citoplasmáticos/metabolismo , Células Germinativas/crescimento & desenvolvimento , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/biossíntese , Proteínas de Caenorhabditis elegans/genética , RNA Helicases DEAD-box/biossíntese , RNA Helicases DEAD-box/genética , Proteínas do Tecido Nervoso/genética , Interferência de RNA , RNA Interferente Pequeno , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Ribonucleoproteínas , Fatores de Transcrição/genética
7.
Genome Res ; 23(8): 1339-47, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23550086

RESUMO

RNA polymerase transcription initiation sites are largely unknown in Caenorhabditis elegans. The initial 5' end of most protein-coding transcripts is removed by trans-splicing, and noncoding initiation sites have not been investigated. We characterized the landscape of RNA Pol II transcription initiation, identifying 73,500 distinct clusters of initiation. Bidirectional transcription is frequent, with a peak of transcriptional pairing at 120 bp. We assign transcription initiation sites to 7691 protein-coding genes and find that they display features typical of eukaryotic promoters. Strikingly, the majority of initiation events occur in regions with enhancer-like chromatin signatures. Based on the overlap of transcription initiation clusters with mapped transcription factor binding sites, we define 2361 transcribed intergenic enhancers. Remarkably, productive transcription elongation across these enhancers is predominantly in the same orientation as that of the nearest downstream gene. Directed elongation from an upstream enhancer toward a downstream gene could potentially deliver RNA polymerase II to a proximal promoter, or alternatively might function directly as a distal promoter. Our results provide a new resource to investigate transcription regulation in metazoans.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Elementos Facilitadores Genéticos , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Cromatina/genética , Anotação de Sequência Molecular , Análise de Sequência de DNA , Sítio de Iniciação de Transcrição , Ativação Transcricional
8.
Cell Rep ; 2(5): 1169-77, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-23103171

RESUMO

The Caenorhabditis elegans MES proteins are key chromatin regulators of the germline. MES-2, MES-3, and MES-6 form the C. elegans Polycomb repressive complex 2 and generate repressive H3K27me3. MES-4 generates H3K36me3 on germline-expressed genes. Transcript profiling of dissected mutant germlines revealed that MES-2/3/6 and MES-4 cooperate to promote the expression of germline genes and repress the X chromosomes and somatic genes. Results from genome-wide chromatin immunoprecipitation showed that H3K27me3 and H3K36me3 occupy mutually exclusive domains on the autosomes and that H3K27me3 is enriched on the X. Loss of MES-4 from germline genes causes H3K27me3 to spread to germline genes, resulting in reduced H3K27me3 elsewhere on the autosomes and especially on the X. Our findings support a model in which H3K36me3 repels H3K27me3 from germline genes and concentrates it on other regions of the genome. This antagonism ensures proper patterns of gene expression for germ cells, which includes repression of somatic genes and the X chromosomes.


Assuntos
Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Células Germinativas/metabolismo , Complexo Repressor Polycomb 2/antagonistas & inibidores , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Histonas/metabolismo , Metilação , Mutação , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Proteínas do Grupo Polycomb , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Cromossomo X/genética , Cromossomo X/metabolismo
9.
Nat Struct Mol Biol ; 18(1): 91-3, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21131980

RESUMO

We have tested the specificity and utility of more than 200 antibodies raised against 57 different histone modifications in Drosophila melanogaster, Caenorhabditis elegans and human cells. Although most antibodies performed well, more than 25% failed specificity tests by dot blot or western blot. Among specific antibodies, more than 20% failed in chromatin immunoprecipitation experiments. We advise rigorous testing of histone-modification antibodies before use, and we provide a website for posting new test results (http://compbio.med.harvard.edu/antibodies/).


Assuntos
Especificidade de Anticorpos , Histonas/imunologia , Animais , Anticorpos/química , Western Blotting , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/química , Imunoprecipitação da Cromatina , Proteínas de Drosophila/química , Drosophila melanogaster/genética , Histonas/química , Histonas/metabolismo , Immunoblotting , Processamento de Proteína Pós-Traducional , Controle de Qualidade , Reprodutibilidade dos Testes
10.
Genome Res ; 21(2): 227-36, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21177964

RESUMO

Chromatin immunoprecipitation identifies specific interactions between genomic DNA and proteins, advancing our understanding of gene-level and chromosome-level regulation. Based on chromatin immunoprecipitation experiments using validated antibodies, we define the genome-wide distributions of 19 histone modifications, one histone variant, and eight chromatin-associated proteins in Caenorhabditis elegans embryos and L3 larvae. Cluster analysis identified five groups of chromatin marks with shared features: Two groups correlate with gene repression, two with gene activation, and one with the X chromosome. The X chromosome displays numerous unique properties, including enrichment of monomethylated H4K20 and H3K27, which correlate with the different repressive mechanisms that operate in somatic tissues and germ cells, respectively. The data also revealed striking differences in chromatin composition between the autosomes and between chromosome arms and centers. Chromosomes I and III are globally enriched for marks of active genes, consistent with containing more highly expressed genes, compared to chromosomes II, IV, and especially V. Consistent with the absence of cytological heterochromatin and the holocentric nature of C. elegans chromosomes, markers of heterochromatin such as H3K9 methylation are not concentrated at a single region on each chromosome. Instead, H3K9 methylation is enriched on chromosome arms, coincident with zones of elevated meiotic recombination. Active genes in chromosome arms and centers have very similar histone mark distributions, suggesting that active domains in the arms are interspersed with heterochromatin-like structure. These data, which confirm and extend previous studies, allow for in-depth analysis of the organization and deployment of the C. elegans genome during development.


Assuntos
Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Cromossomos/metabolismo , Histonas/metabolismo , Animais , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Genes Ligados ao Cromossomo X/genética , Histonas/genética , Metilação , Metiltransferases/metabolismo , Membrana Nuclear/metabolismo , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , Sequências Repetitivas de Ácido Nucleico/genética
11.
Genome Biol ; 11(12): R120, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21176223

RESUMO

BACKGROUND: Although Caenorhabditis elegans was the first multicellular organism with a completely sequenced genome, how this genome is arranged within the nucleus is not known. RESULTS: We determined the genomic regions associated with the nuclear transmembrane protein LEM-2 in mixed-stage C. elegans embryos via chromatin immunoprecipitation. Large regions of several megabases on the arms of each autosome were associated with LEM-2. The center of each autosome was mostly free of such interactions, suggesting that they are largely looped out from the nuclear membrane. Only the left end of the X chromosome was associated with the nuclear membrane. At a finer scale, the large membrane-associated domains consisted of smaller subdomains of LEM-2 associations. These subdomains were characterized by high repeat density, low gene density, high levels of H3K27 trimethylation, and silent genes. The subdomains were punctuated by gaps harboring highly active genes. A chromosome arm translocated to a chromosome center retained its association with LEM-2, although there was a slight decrease in association near the fusion point. CONCLUSIONS: Local DNA or chromatin properties are the main determinant of interaction with the nuclear membrane, with position along the chromosome making a minor contribution. Genes in small gaps between LEM-2 associated regions tend to be highly expressed, suggesting that these small gaps are especially amenable to highly efficient transcription. Although our data are derived from an amalgamation of cell types in mixed-stage embryos, the results suggest a model for the spatial arrangement of C. elegans chromosomes within the nucleus.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Mapeamento Cromossômico , Proteínas de Membrana/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Animais , Proteínas de Caenorhabditis elegans/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Imunoprecipitação da Cromatina , Expressão Gênica , Perfilação da Expressão Gênica , Inativação Gênica , Genoma , Hibridização in Situ Fluorescente , Proteínas de Membrana/genética , Análise em Microsséries , Proteínas Nucleares/genética , Interferência de RNA
12.
PLoS Genet ; 6(9): e1001091, 2010 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-20824077

RESUMO

Methylation of histone H3K36 in higher eukaryotes is mediated by multiple methyltransferases. Set2-related H3K36 methyltransferases are targeted to genes by association with RNA Polymerase II and are involved in preventing aberrant transcription initiation within the body of genes. The targeting and roles of the NSD family of mammalian H3K36 methyltransferases, known to be involved in human developmental disorders and oncogenesis, are not known. We used genome-wide chromatin immunoprecipitation (ChIP) to investigate the targeting and roles of the Caenorhabditis elegans NSD homolog MES-4, which is maternally provided to progeny and is required for the survival of nascent germ cells. ChIP analysis in early C. elegans embryos revealed that, consistent with immunostaining results, MES-4 binding sites are concentrated on the autosomes and the leftmost approximately 2% (300 kb) of the X chromosome. MES-4 overlies the coding regions of approximately 5,000 genes, with a modest elevation in the 5' regions of gene bodies. Although MES-4 is generally found over Pol II-bound genes, analysis of gene sets with different temporal-spatial patterns of expression revealed that Pol II association with genes is neither necessary nor sufficient to recruit MES-4. In early embryos, MES-4 associates with genes that were previously expressed in the maternal germ line, an interaction that does not require continued association of Pol II with those loci. Conversely, Pol II association with genes newly expressed in embryos does not lead to recruitment of MES-4 to those genes. These and other findings suggest that MES-4, and perhaps the related mammalian NSD proteins, provide an epigenetic function for H3K36 methylation that is novel and likely to be unrelated to ongoing transcription. We propose that MES-4 transmits the memory of gene expression in the parental germ line to offspring and that this memory role is critical for the PGCs to execute a proper germline program.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Embrião não Mamífero/enzimologia , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/enzimologia , Histona-Lisina N-Metiltransferase/metabolismo , Animais , Sítios de Ligação , Biocatálise , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/enzimologia , Proteínas de Caenorhabditis elegans/genética , Embrião não Mamífero/citologia , Feminino , Genes de Helmintos/genética , Células Germinativas/citologia , Histona Metiltransferases , Histonas/metabolismo , Lisina/metabolismo , Metilação , Fases de Leitura Aberta/genética , Ligação Proteica , Interferência de RNA , RNA Polimerase II/metabolismo , Cromossomo X/metabolismo
13.
PLoS One ; 3(4): e2022, 2008 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-18431499

RESUMO

The septins are a conserved family of proteins that have been proposed to carry out diverse functions. In budding yeast, the septins become localized to the site of bud emergence in G1 but have not been thought to carry out important functions at this stage of the cell cycle. We show here that the septins function in redundant mechanisms that are required for formation of the bud neck and for the normal pattern of cell growth early in the cell cycle. The Shs1 septin shows strong genetic interactions with G1 cyclins and is directly phosphorylated by G1 cyclin-dependent kinases, consistent with a role in early cell cycle events. However, Shs1 phosphorylation site mutants do not show genetic interactions with the G1 cyclins or obvious defects early in the cell cycle. Rather, they cause an increased cell size and aberrant cell morphology that are dependent upon inhibitory phosphorylation of Cdk1 at the G2/M transition. Shs1 phosphorylation mutants also show defects in interaction with the Gin4 kinase, which associates with the septins during G2/M and plays a role in regulating inhibitory phosphorylation of Cdk1. Phosphorylation of Shs1 by G1 cyclin-dependent kinases plays a role in events that influence Cdk1 inhibitory phosphorylation.


Assuntos
Fase G1 , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomycetales/citologia , Sequência de Aminoácidos , Proliferação de Células , Sequência Consenso , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Dados de Sequência Molecular , Mutação/genética , Mapeamento de Peptídeos , Fosforilação , Ligação Proteica , Transporte Proteico , Proteínas de Saccharomyces cerevisiae/química , Saccharomycetales/enzimologia
14.
Nat Cell Biol ; 9(5): 506-15, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17417630

RESUMO

The mechanisms that control cell growth during the cell cycle are poorly understood. In budding yeast, cyclin dependent kinase 1 (Cdk1) triggers polarization of the actin cytoskeleton and bud emergence in late G1 through activation of the Cdc42 GTPase. However, Cdk1 is not thought to be required for subsequent growth of the bud. Here, we show that Cdk1 has an unexpected role in controlling bud growth after bud emergence. Moreover, we show that G1 cyclin-Cdk1 complexes specifically phosphorylate multiple proteins associated with Cdc24, the guanine nucleotide-exchange factor (GEF) that activates the Cdc42 GTPase. A mutant form of a Cdc24-associated protein that fails to undergo Cdk1-dependent phosphorylation causes defects in bud growth. These results provide a direct link between Cdk1 activity and the control of polarized cell growth.


Assuntos
Proteína Quinase CDC2/metabolismo , Ciclo Celular/fisiologia , Polaridade Celular , Proteínas Fúngicas/metabolismo , Saccharomycetales/crescimento & desenvolvimento , Proteína cdc42 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína Quinase CDC2/antagonistas & inibidores , Proteína Quinase CDC2/genética , Ciclo Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Ciclina G , Ciclinas/metabolismo , Inibidores Enzimáticos/farmacologia , Exocitose , Proteínas Fúngicas/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Kluyveromyces/crescimento & desenvolvimento , Complexos Multiproteicos/metabolismo , Mutação , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Saccharomycetales/efeitos dos fármacos , Saccharomycetales/genética , Saccharomycetales/metabolismo , Vesículas Secretórias/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...