Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Genet ; 18(2): e1010084, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35202393

RESUMO

Fragile X Syndrome (FXS) is the most prevalent cause of inherited mental deficiency and is the most common monogenetic cause of autism spectral disorder (ASD). Here, we demonstrate that disease-causing missense mutations in the conserved K homology (KH) RNA binding domains (RBDs) of FMRP cause defects in its ability to form RNA transport granules in neurons. Using molecular, genetic, and imaging approaches in the Drosophila FXS model system, we show that the KH1 and KH2 domains of FMRP regulate distinct aspects of neuronal FMRP granule formation, dynamics, and transport. Furthermore, mutations in the KH domains disrupt translational repression in cells and the localization of known FMRP target mRNAs in neurons. These results suggest that the KH domains play an essential role in neuronal FMRP granule formation and function which may be linked to the molecular pathogenesis of FXS.


Assuntos
Síndrome do Cromossomo X Frágil , Animais , Drosophila/genética , Drosophila/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Mutação de Sentido Incorreto , Neurônios/metabolismo
2.
Aging Cell ; 20(11): e13481, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34674371

RESUMO

As organisms age, they often accumulate protein aggregates that are thought to be toxic, potentially leading to age-related diseases. This accumulation of protein aggregates is partially attributed to a failure to maintain protein homeostasis. A variety of genetic factors have been linked to longevity, but how these factors also contribute to protein homeostasis is not completely understood. In order to understand the relationship between aging and protein aggregation, we tested how a gene that regulates lifespan and age-dependent locomotor behaviors, p38 MAPK (p38Kb), influences protein homeostasis as an organism ages. We find that p38Kb regulates age-dependent protein aggregation through an interaction with starvin, a regulator of muscle protein homeostasis. Furthermore, we have identified Lamin as an age-dependent target of p38Kb and starvin.


Assuntos
Envelhecimento/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Longevidade/genética , Sistema de Sinalização das MAP Quinases/genética , Proteostase/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Envelhecimento/genética , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Feminino , Deleção de Genes , Laminas/metabolismo , Locomoção/genética , Macroautofagia/genética , Músculos/metabolismo , Estresse Oxidativo/genética , Fenótipo , Proteólise , Interferência de RNA , Proteínas Quinases p38 Ativadas por Mitógeno/genética
3.
Front Genet ; 12: 762012, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34712272

RESUMO

Vps54 is an integral subunit of the Golgi-associated retrograde protein (GARP) complex, which is involved in tethering endosome-derived vesicles to the trans-Golgi network (TGN). A destabilizing missense mutation in Vps54 causes the age-progressive motor neuron (MN) degeneration, muscle weakness, and muscle atrophy observed in the wobbler mouse, an established animal model for human MN disease. It is currently unclear how the disruption of Vps54, and thereby the GARP complex, leads to MN and muscle phenotypes. To develop a new tool to address this question, we have created an analogous model in Drosophila by generating novel loss-of-function alleles of the fly Vps54 ortholog (scattered/scat). We find that null scat mutant adults are viable but have a significantly shortened lifespan. Like phenotypes observed in the wobbler mouse, we show that scat mutant adults are male sterile and have significantly reduced body size and muscle area. Moreover, we demonstrate that scat mutant adults have significant age-progressive defects in locomotor function. Interestingly, we see sexually dimorphic effects, with scat mutant adult females exhibiting significantly stronger phenotypes. Finally, we show that scat interacts genetically with rab11 in MNs to control age-progressive muscle atrophy in adults. Together, these data suggest that scat mutant flies share mutant phenotypes with the wobbler mouse and may serve as a new genetic model system to study the cellular and molecular mechanisms underlying MN disease.

5.
Biol Open ; 9(8)2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32747448

RESUMO

Vps54 is a subunit of the Golgi-associated retrograde protein (GARP) complex, which is involved in tethering endosome-derived vesicles to the trans-Golgi network (TGN). In the wobbler mouse, a model for human motor neuron (MN) disease, reduction in the levels of Vps54 causes neurodegeneration. However, it is unclear how disruption of the GARP complex leads to MN dysfunction. To better understand the role of Vps54 in MNs, we have disrupted expression of the Vps54 ortholog in Drosophila and examined the impact on the larval neuromuscular junction (NMJ). Surprisingly, we show that both null mutants and MN-specific knockdown of Vps54 leads to NMJ overgrowth. Reduction of Vps54 partially disrupts localization of the t-SNARE, Syntaxin-16, to the TGN but has no visible impact on endosomal pools. MN-specific knockdown of Vps54 in MNs combined with overexpression of the small GTPases Rab5, Rab7, or Rab11 suppresses the Vps54 NMJ phenotype. Conversely, knockdown of Vps54 combined with overexpression of dominant negative Rab7 causes NMJ and behavioral abnormalities including a decrease in postsynaptic Dlg and GluRIIB levels without any effect on GluRIIA. Taken together, these data suggest that Vps54 controls larval MN axon development and postsynaptic density composition through a mechanism that requires Rab7.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Epistasia Genética , Junção Neuromuscular/metabolismo , Densidade Pós-Sináptica/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Axônios/metabolismo , Proteínas de Drosophila/genética , Larva/metabolismo , Neurônios Motores/metabolismo , Músculos/metabolismo , Proteínas Mutantes/metabolismo , Neuroglia/metabolismo , Sintaxina 16/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas rab de Ligação ao GTP/genética , proteínas de unión al GTP Rab7 , Rede trans-Golgi/metabolismo
6.
Proteins ; 88(1): 166-174, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31295370

RESUMO

Lactoferrin (LF) is a multifunctional protein that plays important physiological roles as one of the most concentrated proteins in many human and other mammalian fluids and tissues. In particular, LF provides antibacterial properties to human milk, saliva, and tear fluid. LF also protects against stress-induced lipid peroxidation at inflammation sites through its iron-binding ability. Previous studies have shown that LF can be efficiently nitrated via biologically relevant mediators such as peroxynitrite (ONOO- ), which are also present at high intracellular concentrations during inflammation and nitrosative stress. Here, we examine changes in antibacterial properties and structure of LF following ONOO- treatment. The reaction induces nitration of tyrosine and tryptophan residues, which are commonly used as biomarker molecules for several diseases. Treatment with ONOO- at a 10/1 M ratio of ONOO- to tyrosine inhibited all antibacterial activity exhibited by native LF. Secondary structural changes in LF were assessed using circular dichroism spectroscopy. Nitration products with and without the addition of Fe3+ show significant reduction in alpha-helical properties, suggesting partial protein unfolding. Iron-binding capacity of LF was also reduced after treatment with ONOO- , suggesting a decreased ability of LF to protect against cellular damage. LC-MS/MS spectrometry was used to identify LF peptide fragments nitrated by ONOO- , including tyrosine residue Y92 located in the iron-binding region. These results suggest that posttranslational modification of LF by ONOO- could be an important pathway to exacerbate infection, for example, in inflamed tissues and to reduce the ability of LF to act as an immune responder and decrease oxidative damage.


Assuntos
Antibacterianos/metabolismo , Lactoferrina/metabolismo , Ácido Peroxinitroso/metabolismo , Sequência de Aminoácidos , Antibacterianos/química , Humanos , Ferro/metabolismo , Lactoferrina/química , Ligação Proteica , Processamento de Proteína Pós-Traducional , Estrutura Secundária de Proteína , Desdobramento de Proteína
7.
Mol Cell Neurosci ; 76: 33-41, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27567686

RESUMO

Spaced synaptic depolarization induces rapid axon terminal growth and the formation of new synaptic boutons at the Drosophila larval neuromuscular junction (NMJ). Here, we identify a novel presynaptic function for the Calcium/Calmodulin-dependent Kinase II (CamKII) protein in the control of activity-dependent synaptic growth. Consistent with this function, we find that both total and phosphorylated CamKII (p-CamKII) are enriched in axon terminals. Interestingly, p-CamKII appears to be enriched at the presynaptic axon terminal membrane. Moreover, levels of total CamKII protein within presynaptic boutons globally increase within one hour following stimulation. These effects correlate with the activity-dependent formation of new presynaptic boutons. The increase in presynaptic CamKII levels is inhibited by treatment with cyclohexamide suggesting a protein-synthesis dependent mechanism. We have previously found that acute spaced stimulation rapidly downregulates levels of neuronal microRNAs (miRNAs) that are required for the control of activity-dependent axon terminal growth at this synapse. The rapid activity-dependent accumulation of CamKII protein within axon terminals is inhibited by overexpression of activity-regulated miR-289 in motor neurons. Experiments in vitro using a CamKII translational reporter show that miR-289 can directly repress the translation of CamKII via a sequence motif found within the CamKII 3' untranslated region (UTR). Collectively, our studies support the idea that presynaptic CamKII acts downstream of synaptic stimulation and the miRNA pathway to control rapid activity-dependent changes in synapse structure.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Potenciais da Membrana , Crescimento Neuronal , Terminações Pré-Sinápticas/metabolismo , Regiões 3' não Traduzidas , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Linhagem Celular , Drosophila , MicroRNAs/genética , Junção Neuromuscular/metabolismo , Junção Neuromuscular/fisiologia , Terminações Pré-Sinápticas/fisiologia
8.
PLoS One ; 11(3): e0150291, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26930655

RESUMO

Non-translating RNAs that have undergone active translational repression are culled from the cytoplasm into P-bodies for decapping-dependent decay or for sequestration. Organisms that use microRNA-mediated RNA silencing have an additional pathway to remove RNAs from active translation. Consequently, proteins that govern microRNA-mediated silencing, such as GW182/Gw and AGO1, are often associated with the P-bodies of higher eukaryotic organisms. Due to the presence of Gw, these structures have been referred to as GW-bodies. However, several reports have indicated that GW-bodies have different dynamics to P-bodies. Here, we use live imaging to examine GW-body and P-body dynamics in the early Drosophila melanogaster embryo. While P-bodies are present throughout early embryonic development, cytoplasmic GW-bodies only form in significant numbers at the midblastula transition. Unlike P-bodies, which are predominantly cytoplasmic, GW-bodies are present in both nuclei and the cytoplasm. RNA decapping factors such as DCP1, Me31B, and Hpat are not associated with GW-bodies, indicating that P-bodies and GW-bodies are distinct structures. Furthermore, known Gw interactors such as AGO1 and the CCR4-NOT deadenylation complex, which have been shown to be important for Gw function, are also not present in GW-bodies. Use of translational inhibitors puromycin and cycloheximide, which respectively increase or decrease cellular pools of non-translating RNAs, alter GW-body size, underscoring that GW-bodies are composed of non-translating RNAs. Taken together, these data indicate that active translational silencing most likely does not occur in GW-bodies. Instead GW-bodies most likely function as repositories for translationally silenced RNAs. Finally, inhibition of zygotic gene transcription is unable to block the formation of either P-bodies or GW-bodies in the early embryo, suggesting that these structures are composed of maternal RNAs.


Assuntos
Biossíntese de Proteínas/genética , RNA Longo não Codificante/genética , Animais , Proteínas Argonautas/genética , Citoplasma/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , MicroRNAs/genética , Interferência de RNA/fisiologia , Transcrição Gênica/genética
9.
PLoS One ; 8(7): e68385, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23844193

RESUMO

It is widely accepted that long-term changes in synapse structure and function are mediated by rapid activity-dependent gene transcription and new protein synthesis. A growing amount of evidence suggests that the microRNA (miRNA) pathway plays an important role in coordinating these processes. Despite recent advances in this field, there remains a critical need to identify specific activity-regulated miRNAs as well as their key messenger RNA (mRNA) targets. To address these questions, we used the larval Drosophila melanogaster neuromuscular junction (NMJ) as a model synapse in which to identify novel miRNA-mediated mechanisms that control activity-dependent synaptic growth. First, we developed a screen to identify miRNAs differentially regulated in the larval CNS following spaced synaptic stimulation. Surprisingly, we identified five miRNAs (miRs-1, -8, -289, -314, and -958) that were significantly downregulated by activity. Neuronal misexpression of three miRNAs (miRs-8, -289, and -958) suppressed activity-dependent synaptic growth suggesting that these miRNAs control the translation of biologically relevant target mRNAs. Functional annotation cluster analysis revealed that putative targets of miRs-8 and -289 are significantly enriched in clusters involved in the control of neuronal processes including axon development, pathfinding, and growth. In support of this, miR-8 regulated the expression of a wingless 3'UTR (wg 3' untranslated region) reporter in vitro. Wg is an important presynaptic regulatory protein required for activity-dependent axon terminal growth at the fly NMJ. In conclusion, our results are consistent with a model where key activity-regulated miRNAs are required to coordinate the expression of genes involved in activity-dependent synaptogenesis.


Assuntos
Drosophila melanogaster/genética , Regulação da Expressão Gênica , MicroRNAs/genética , Junção Neuromuscular/genética , Sinapses/genética , Animais , Animais Geneticamente Modificados , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiologia , Perfilação da Expressão Gênica , Ontologia Genética , Larva/genética , Larva/metabolismo , Larva/fisiologia , Neurônios Motores/metabolismo , Junção Neuromuscular/metabolismo , Junção Neuromuscular/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Sinapses/metabolismo , Sinapses/fisiologia
10.
J Cell Sci ; 125(Pt 24): 6105-16, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23097047

RESUMO

The temporal and spatial regulation of protein synthesis plays an important role in the control of neural physiology. In axons and dendrites, translationally repressed mRNAs are actively transported to their destinations in a variety of ribonucleoprotein particles (RNPs). A subset of these neuronal RNPs has been shown to contain proteins associated with mRNA processing bodies (P bodies). P bodies are a class of highly conserved cytoplasmic granules that have been linked to both mRNA decay and translational repression via general and miRNA-mediated pathways. Here, we characterize functions for HPat/Pat1 (also known as Patr-1), a core component of P bodies, at the glutamatergic larval Drosophila neuromuscular junction (NMJ). We show that hpat mutants exhibit a strong synaptic hyperplasia at the NMJ. The synaptic defects observed in hpat mutants are associated with rearrangement of the axonal microtubule cytoskeleton suggesting that HPat negatively regulates presynaptic microtubule-based growth during NMJ development. Consistent with this, overexpression of HPat also blocks the rapid growth of presynaptic boutons induced by spaced depolarization. Finally, we demonstrate that HPat interacts genetically with the catalytic subunit of the deadenylase complex (twin/CCR4) and the miRNA pathway (Argonaute 1) to control bouton formation. We propose that HPat is required to target mRNAs involved in the control of microtubule architecture and synaptic terminal growth for repression, presumably in P bodies, via both general and miRNA-mediated mechanisms.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila/crescimento & desenvolvimento , Junção Neuromuscular/crescimento & desenvolvimento , Terminações Pré-Sinápticas/fisiologia , Proteínas de Ligação a RNA/fisiologia , Animais , Proteínas de Transporte/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Drosophila/embriologia , Proteínas de Drosophila/metabolismo , Feminino , Masculino , Junção Neuromuscular/metabolismo , Terminações Pré-Sinápticas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transmissão Sináptica
11.
Genetics ; 182(4): 1051-60, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19487564

RESUMO

Mechanisms of neuronal mRNA localization and translation are of considerable biological interest. Spatially regulated mRNA translation contributes to cell-fate decisions and axon guidance during development, as well as to long-term synaptic plasticity in adulthood. The Fragile-X Mental Retardation protein (FMRP/dFMR1) is one of the best-studied neuronal translational control molecules and here we describe the identification and early characterization of proteins likely to function in the dFMR1 pathway. Induction of the dFMR1 in sevenless-expressing cells of the Drosophila eye causes a disorganized (rough) eye through a mechanism that requires residues necessary for dFMR1/FMRP's translational repressor function. Several mutations in dco, orb2, pAbp, rm62, and smD3 genes dominantly suppress the sev-dfmr1 rough-eye phenotype, suggesting that they are required for dFMR1-mediated processes. The encoded proteins localize to dFMR1-containing neuronal mRNPs in neurites of cultured neurons, and/or have an effect on dendritic branching predicted for bona fide neuronal translational repressors. Genetic mosaic analyses indicate that dco, orb2, rm62, smD3, and dfmr1 are dispensable for translational repression of hid, a microRNA target gene, known to be repressed in wing discs by the bantam miRNA. Thus, the encoded proteins may function as miRNA- and/or mRNA-specific translational regulators in vivo.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/fisiologia , MicroRNAs/metabolismo , RNA Mensageiro/metabolismo , Alelos , Animais , Transporte Biológico , Células Cultivadas , Olho/patologia , Mutação , Neurônios/citologia , Ribonucleoproteínas
12.
Proc Natl Acad Sci U S A ; 105(38): 14644-9, 2008 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-18780789

RESUMO

The formation of long-term memory is believed to require translational control of localized mRNAs. In mammals, dendritic mRNAs are maintained in a repressed state and are activated upon repetitive stimulation. Several regulatory proteins required for translational control in early development are thought to be required for memory formation, suggesting similar molecular mechanisms. Here, using Drosophila, we identify the enzyme responsible for poly(A) elongation in the brain and demonstrate that its activity is required specifically for long-term memory. These findings provide strong evidence that cytoplasmic polyadenylation is critical for memory formation, and that GLD2 is the enzyme responsible.


Assuntos
Drosophila melanogaster/enzimologia , Memória/fisiologia , Polinucleotídeo Adenililtransferase/metabolismo , Animais , Animais Geneticamente Modificados , Células Cultivadas , Citoplasma/enzimologia , Proteínas de Drosophila/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Neurônios Motores/enzimologia , Neuritos/enzimologia , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo
13.
ScientificWorldJournal ; 7: 178-90, 2007 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-17982591

RESUMO

What is the protein apparatus required for microRNA (miRNA) function and translational repression in neurons? This article reviews our recent work on Me31B, a conserved P-body protein present on Staufen-containing neuronal and maternal ribonucleoprotein (RNP) particles, which is required for dendrite morphogenesis and miRNA function in vivo. In addition, it provides new data to show that Me31B is present on and regulates formation of P-bodies in the Drosophila wing disc, where it has a general role in the regulation of miRNA function. While illuminating the function of this important RNA regulatory molecule, it also brings into focus a hypothesis of potentially broad significance. Namely, that P-body proteins may play important roles in regulation of dendrite-localized mRNAs and, thereby, in synaptic plasticity. A wide range of protein localization and early functional data support this hypothesis. We also discuss current knowledge of RNP particles that mediate translational repression and the implications of these findings for understanding translational control in neurons.


Assuntos
Regulação da Expressão Gênica/genética , Membranas Intracelulares/metabolismo , MicroRNAs/genética , Plasticidade Neuronal/genética , Sinapses/metabolismo , Animais , Humanos , Biossíntese de Proteínas/genética
14.
Neuron ; 52(6): 997-1009, 2006 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-17178403

RESUMO

Local control of mRNA translation modulates neuronal development, synaptic plasticity, and memory formation. A poorly understood aspect of this control is the role and composition of ribonucleoprotein (RNP) particles that mediate transport and translation of neuronal RNAs. Here, we show that staufen- and FMRP-containing RNPs in Drosophila neurons contain proteins also present in somatic "P bodies," including the RNA-degradative enzymes Dcp1p and Xrn1p/Pacman and crucial components of miRNA (argonaute), NMD (Upf1p), and general translational repression (Dhh1p/Me31B) pathways. Drosophila Me31B is shown to participate (1) with an FMRP-associated, P body protein (Scd6p/trailer hitch) in FMRP-driven, argonaute-dependent translational repression in developing eye imaginal discs; (2) in dendritic elaboration of larval sensory neurons; and (3) in bantam miRNA-mediated translational repression in wing imaginal discs. These results argue for a conserved mechanism of translational control critical to neuronal function and open up new experimental avenues for understanding the regulation of mRNA function within neurons.


Assuntos
Proteínas de Drosophila/fisiologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Neurônios/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/fisiologia , Animais , Animais Geneticamente Modificados , Northern Blotting , Western Blotting/métodos , Caspases/metabolismo , Células Cultivadas , Sistema Nervoso Central/citologia , Dendritos/metabolismo , Dendritos/fisiologia , Drosophila , Proteínas de Drosophila/metabolismo , Exorribonucleases/metabolismo , Olho/metabolismo , Olho/ultraestrutura , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica/métodos , Larva , MicroRNAs/metabolismo , Microscopia Eletrônica de Varredura/métodos , Neurônios/citologia , Biossíntese de Proteínas/fisiologia , Transporte Proteico/fisiologia , Complexo de Inativação Induzido por RNA/metabolismo
15.
Dev Biol ; 291(1): 132-43, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16413530

RESUMO

Sm and Sm-like proteins are core components of the splicesome but have other functions distinct from pre-mRNA processing. Here, we show that Sm proteins also regulate germ cell specification during early C. elegans embryogenesis. SmE and SmG were required to maintain transcriptional quiescence in embryonic germ cell precursors. In addition, depletion of SmE inhibited expression of the germ lineage-specific proteins PIE-1, GLD-1, and NOS-2, but did not affect maintenance of several maternal mRNAs. PIE-1 had previously been shown to activate transcriptional silencing and NOS-2 expression. We found that PIE-1 also promotes GLD-1 expression by a process that is independent of transcriptional silencing. Thus, Sm proteins could control transcriptional silencing and maternal protein expression by regulating PIE-1. However, loss of SmE function also caused defects in P granule localization and premature division in early germline blastomeres, processes that are independent of PIE-1 function. Therefore, the Sm proteins control multiple aspects of germ cell precursor development. Because depletion of several other core splicing factors did not affect these events, these Sm functions are likely distinct from pre-mRNA splicing. Sm family proteins assemble into ribonucleoprotein complexes (RNPs) that control RNA activities. We suggest that novel Sm RNPs directly or indirectly influence posttranscriptional control of maternal mRNAs to promote germ cell specification in the early C. elegans embryo.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/embriologia , Desenvolvimento Embrionário , Células Germinativas/citologia , Ribonucleoproteínas Nucleares Pequenas/fisiologia , Animais , Caenorhabditis elegans/citologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/biossíntese , Proteínas de Caenorhabditis elegans/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Proteínas Nucleares/fisiologia , Interferência de RNA , Splicing de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro Estocado/biossíntese , Ribonucleoproteínas Nucleares Pequenas/genética
16.
Curr Biol ; 12(17): 1502-6, 2002 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-12225665

RESUMO

General mRNA processing factors are traditionally thought to function only in the control of global gene expression. Here we show that the Sm proteins, core components of the splicesome, also regulate germ granules during early C. elegans development. Germ granules are large cytoplasmic particles that localize to germ cells and their precursors during embryogenesis of diverse organisms. In C. elegans, germ granules, called P granules, are segregated to the germline precursor cells during embryogenesis by asymmetric cell division, and they remain in germ cells at all stages of development. We found that at least some Sm proteins are components of P granules. Moreover, disruption of Sm activity caused defects in P granule localization to the germ cell precursors during early embryogenesis. In contrast, loss of other splicing factor activities had no effect on germ granule control in the embryo. These observations suggest that the Sm proteins control germ granule integrity and localization in the early C. elegans embryo and that this role is independent of pre-mRNA splicing. Thus, a highly conserved splicing factor may have been adapted to control both snRNP biogenesis and the localization of components important for germ cell function.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/embriologia , Grânulos Citoplasmáticos/química , Células Germinativas/citologia , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/ultraestrutura , Proteínas de Caenorhabditis elegans/genética , Divisão Celular , Grânulos Citoplasmáticos/ultraestrutura , Substâncias Macromoleculares , Morfogênese , Splicing de RNA , RNA Interferente Pequeno/farmacologia , Spliceossomos/fisiologia , Spliceossomos/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...