Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35022236

RESUMO

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.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimologia , Proteínas de Ciclo Celular/metabolismo , Células Germinativas/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Ciclo Celular/genética , Proliferação de Células , Ativação Enzimática , Mutação/genética , Oócitos/citologia , Oócitos/metabolismo , Estágio Paquíteno , Fenótipo , Fosforilação , Proteínas Tirosina Fosfatases/genética , Especificidade por Substrato , Complexo Sinaptonêmico/metabolismo , Temperatura
2.
Proc Natl Acad Sci U S A ; 117(49): 31301-31308, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33229589

RESUMO

The function of the nucleus depends on the integrity of the nuclear lamina, an intermediate filament network associated with the linker of nucleoskeleton and cytoskeleton (LINC) complex. The LINC complex spans the nuclear envelope and mediates nuclear mechanotransduction, the process by which mechanical signals and forces are transmitted across the nuclear envelope. In turn, the AAA+ ATPase torsinA is thought to regulate force transmission from the cytoskeleton to the nucleus. In humans, mutations affecting nuclear envelope-associated proteins cause laminopathies, including progeria, myopathy, and dystonia, though the extent to which endogenous mechanical stresses contribute to these pathologies is unclear. Here, we use the Caenorhabditis elegans germline as a model to investigate mechanisms that maintain nuclear integrity as germ cell nuclei progress through meiotic development and migrate for gametogenesis-processes that require LINC complex function. We report that decreasing the function of the C. elegans torsinA homolog, OOC-5, rescues the sterility and premature aging caused by a null mutation in the single worm lamin homolog. We show that decreasing OOC-5/torsinA activity prevents nuclear collapse in lamin mutants by disrupting the function of the LINC complex. At a mechanistic level, OOC-5/torsinA promotes the assembly or maintenance of the lamin-associated LINC complex and this activity is also important for interphase nuclear pore complex insertion into growing germline nuclei. These results demonstrate that LINC complex-transmitted forces damage nuclei with a compromised nuclear lamina. Thus, the torsinA-LINC complex nexus might comprise a therapeutic target for certain laminopathies by preventing damage from endogenous cellular forces.


Assuntos
Caenorhabditis elegans/metabolismo , Núcleo Celular/metabolismo , Laminopatias/patologia , Mecanotransdução Celular , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Interfase , Longevidade , Meiose , Modelos Biológicos , Mutação/genética , Poro Nuclear/metabolismo , Prófase
3.
Cancer Immunol Immunother ; 70(4): 1127-1142, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33033851

RESUMO

Cytokine storm can result from cancer immunotherapy or certain infections, including COVID-19. Though short-term immune-related adverse events are routinely described, longer-term immune consequences and sequential immune monitoring are not as well defined. In 2006, six healthy volunteers received TGN1412, a CD28 superagonist antibody, in a first-in-man clinical trial and suffered from cytokine storm. After the initial cytokine release, antibody effect-specific immune monitoring started on Day + 10 and consisted mainly of evaluation of dendritic cell and T-cell subsets and 15 serum cytokines at 21 time-points over 2 years. All patients developed problems with concentration and memory; three patients were diagnosed with mild-to-moderate depression. Mild neutropenia and autoantibody production was observed intermittently. One patient suffered from peripheral dry gangrene, required amputations, and had persistent Raynaud's phenomenon. Gastrointestinal irritability was noted in three patients and coincided with elevated γδT-cells. One had pruritus associated with elevated IgE levels, also found in three other asymptomatic patients. Dendritic cells, initially undetectable, rose to normal within a month. Naïve CD8+ T-cells were maintained at high levels, whereas naïve CD4+ and memory CD4+ and CD8+ T-cells started high but declined over 2 years. T-regulatory cells cycled circannually and were normal in number. Cytokine dysregulation was especially noted in one patient with systemic symptoms. Over a 2-year follow-up, cognitive deficits were observed in all patients following TGN1412 infusion. Some also had signs or symptoms of psychological, mucosal or immune dysregulation. These observations may discern immunopathology, treatment targets, and long-term monitoring strategies for other patients undergoing immunotherapy or with cytokine storm.


Assuntos
Anticorpos Monoclonais Humanizados/efeitos adversos , Antígenos CD28/agonistas , COVID-19/imunologia , Disfunção Cognitiva/imunologia , Síndrome da Liberação de Citocina/imunologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/imunologia , Imunoterapia/efeitos adversos , SARS-CoV-2/fisiologia , Linfócitos T/imunologia , Adulto , Anticorpos Monoclonais Humanizados/farmacologia , Disfunção Cognitiva/etiologia , Estudos de Coortes , Síndrome da Liberação de Citocina/etiologia , Seguimentos , Humanos , Masculino , Adulto Jovem
4.
Semin Cell Dev Biol ; 84: 90-99, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29242146

RESUMO

In virtually all sexually reproducing animals, oocytes arrest in meiotic prophase and resume meiosis in a conserved biological process called meiotic maturation. Meiotic arrest enables oocytes, which are amongst the largest cells in an organism, to grow and accumulate the necessary cellular constituents required to support embryonic development. Oocyte arrest can be maintained for a prolonged period, up to 50 years in humans, and defects in the meiotic maturation process interfere with the faithful segregation of meiotic chromosomes, representing the leading cause of human birth defects and female infertility. Hormonal signaling and interactions with somatic cells of the gonad control the timing of oocyte meiotic maturation. Signaling activates the CDK1/cyclin B kinase, which plays a central role in regulating the nuclear and cytoplasmic events of meiotic maturation. Nuclear maturation encompasses nuclear envelope breakdown, meiotic spindle assembly, and chromosome segregation whereas cytoplasmic maturation involves major changes in oocyte protein translation and cytoplasmic organelles and is less well understood. Classically, meiotic maturation has been studied in organisms with large oocytes to facilitate biochemical analysis. Recently, the nematode Caenorhabditis elegans is emerging as a genetic paradigm for studying the regulation of oocyte meiotic maturation. Studies in this system have revealed conceptual, anatomical, and molecular links to oocytes in all animals including humans. This review focuses on the signaling mechanisms required to control oocyte growth and meiotic maturation in C. elegans and discusses how the downstream regulation of protein translation coordinates the completion of meiosis and the oocyte-to-embryo transition.


Assuntos
Desenvolvimento Embrionário/fisiologia , Meiose/fisiologia , Oócitos/citologia , Oogênese/fisiologia , Animais , Caenorhabditis elegans , Humanos , Transdução de Sinais/fisiologia
5.
Development ; 140(22): 4614-23, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24131629

RESUMO

The restricted spatiotemporal translation of maternal mRNAs, which is crucial for correct cell fate specification in early C. elegans embryos, is regulated primarily through the 3'UTR. Although genetic screens have identified many maternally expressed cell fate-controlling RNA-binding proteins (RBPs), their in vivo targets and the mechanism(s) by which they regulate these targets are less clear. These RBPs are translated in oocytes and localize to one or a few blastomeres in a spatially and temporally dynamic fashion unique for each protein and each blastomere. Here, we characterize the translational regulation of maternally supplied mom-2 mRNA, which encodes a Wnt ligand essential for two separate cell-cell interactions in early embryos. A GFP reporter that includes only the mom-2 3'UTR is translationally repressed properly in oocytes and early embryos, and then correctly translated only in the known Wnt signaling cells. We show that the spatiotemporal translation pattern of this reporter is regulated combinatorially by a set of nine maternally supplied RBPs. These nine proteins all directly bind the mom-2 3'UTR in vitro and function as positive or negative regulators of mom-2 translation in vivo. The net translational readout for the mom-2 3'UTR reporter is determined by competitive binding between positive- and negative-acting RBPs for the 3'UTR, along with the distinct spatiotemporal localization patterns of these regulators. We propose that the 3'UTR of maternal mRNAs contains a combinatorial code that determines the topography of associated RBPs, integrating positive and negative translational inputs.


Assuntos
Caenorhabditis elegans/embriologia , Caenorhabditis elegans/genética , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Biossíntese de Proteínas/genética , RNA Mensageiro Estocado/genética , Proteínas Wnt/genética , Regiões 3' não Traduzidas/genética , Animais , Ligação Competitiva , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Modelos Biológicos , Oócitos/metabolismo , Fenótipo , Ligação Proteica/genética , Transporte Proteico/genética , Transporte de RNA/genética , RNA Mensageiro Estocado/metabolismo , Transdução de Sinais/genética , Proteínas Wnt/metabolismo
7.
Exp Physiol ; 99(2): 442-53, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24142453

RESUMO

NEW FINDINGS: What is the central question of this study? Activation of angiotensin-converting enzyme 2, resulting in production of angiotensin-(1-7) and stimulation of its receptor, Mas, exerts beneficial actions in a number cardiovascular diseases, including ischaemic stroke. A potential beneficial role for angiotensin-(1-7) in haemorrhagic stroke has not previously been reported. What is the main finding and its importance? Central administration of angiotensin-(1-7) into stroke-prone spontaneously hypertensive rats, a model of haemorrhagic stroke, increases lifespan and improves the neurological status of these rats, as well as decreasing microglial numbers in the striatum (implying attenuation of cerebral inflammation). These actions of angiotensin-(1-7) have not previously been reported and identify this peptide as a potential new therapeutic target in haemorrhagic stroke. Angiotensin-(1-7) [Ang-(1-7)] exerts cerebroprotective effects in ischaemic stroke, and this action is associated with a blunting of intracerebral inflammatory processes and microglial activation. Given that intracerebral inflammation and microglial activation play key roles in the mechanism of injury and brain damage in both ischaemic and haemorrhagic stroke, we have investigated the potential beneficial actions of Ang-(1-7) in stroke-prone spontaneously hypertensive rats (spSHRs), an established animal model of hypertension-induced haemorrhagic stroke. Angiotensin-(1-7) was administered by continuous infusion via the intracerebroventricular route for 6 weeks into spSHRs fed a high-sodium (4%) diet, starting at 49 days of age. This treatment resulted in a significant increase in survival of the spSHRs. Median survival was 108 days in control, artificial cerebrospinal fluid-infused spSHRs and 154 days in Ang-(1-7)-treated spSHRs. This effect was partly reversed by intracerebroventricular infusion of the Mas receptor blocker, A779. This Ang-(1-7) treatment also decreased the number of haemorrhages in the striatum, improved neurological status (reduced lethargy), decreased the number of microglia in the striatum and tended to increase neuron survival at the same site. Importantly, infusions of Ang-(1-7) had no effect on kidney pathology, heart pathology, body weight, serum corticosterone levels or blood pressure. This study is the first to demonstrate the cerebroprotective actions of Ang-(1-7), including increased survival time, in spSHRs. As such, these data reveal a potential therapeutic target for haemorrhagic stroke.


Assuntos
Angiotensina I/farmacologia , Hipertensão/complicações , Fragmentos de Peptídeos/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/mortalidade , Animais , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Corticosterona/sangue , Coração/efeitos dos fármacos , Infusões Intraventriculares , Rim/efeitos dos fármacos , Masculino , Microglia/efeitos dos fármacos , Ratos , Ratos Endogâmicos SHR , Acidente Vascular Cerebral/sangue
8.
Alcohol Clin Exp Res ; 37(1): 86-95, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22691134

RESUMO

BACKGROUND: Disturbed gait and balance are common and important sequelae of chronic alcoholism. We present longitudinal data on recovery of gait and balance in alcoholics 6 to 15 weeks abstinent at baseline assessment through follow-up assessment 4 to 16 months after baseline. METHODS: We performed a follow-up assessment (4 to 16 months after baseline) of gait and balance functioning in 37 short-term (6 to 15 weeks) abstinent alcoholics (STAA), 25 of whom remained abstinent through the follow-up period. Fourteen non-substance-abusing controls (NSAC) were also brought back for a follow-up assessment to examine practice effects. RESULTS: Alcoholics showed gait and balance impairment versus controls at both the initial and follow-up assessments, showing no improvement in gait and balance measures over the follow-up period. At follow-up, NSAC showed improvement on the Walk on Floor eyes closed measure, possibly representing a practice effect not present in STAA. CONCLUSIONS: This study finds no improvement from about 10 weeks to about 1 year of abstinence in chronic alcoholics. The study is silent with regard to gait and balance recovery that occurs prior to 10 weeks abstinence, and after the first year of abstinence. Other studies suggest some recovery of gait and balance prior to 10 weeks abstinence, and our recent cross-sectional study (Smith and Fein, 2011, Alcohol Clin Exp Res 35:2184-2192) suggests that significant additional recovery occurs in the ensuing years.


Assuntos
Alcoolismo/fisiopatologia , Marcha , Equilíbrio Postural , Adulto , Consumo de Bebidas Alcoólicas/fisiopatologia , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fumar
9.
Adv Exp Med Biol ; 757: 277-320, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22872481

RESUMO

In sexually reproducing animals, oocytes arrest at diplotene or diakinesis and resume meiosis (meiotic maturation) in response to hormones. Chromosome segregation errors in female meiosis I are the leading cause of human birth defects, and age-related changes in the hormonal environment of the ovary are a suggested cause. Caenorhabditis elegans is emerging as a genetic paradigm for studying hormonal control of meiotic maturation. The meiotic maturation processes in C. elegans and mammals share a number of biological and molecular similarities. Major sperm protein (MSP) and luteinizing hormone (LH), though unrelated in sequence, both trigger meiotic resumption using somatic Gα(s)-adenylate cyclase pathways and soma-germline gap-junctional communication. At a molecular level, the oocyte responses apparently involve the control of conserved protein kinase pathways and post-transcriptional gene regulation in the oocyte. At a cellular level, the responses include cortical cytoskeletal rearrangement, nuclear envelope breakdown, assembly of the acentriolar meiotic spindle, chromosome segregation, and likely changes important for fertilization and the oocyte-to-embryo transition. This chapter focuses on signaling mechanisms required for oocyte growth and meiotic maturation in C. elegans and discusses how these mechanisms coordinate the completion of meiosis and the oocyte-to-embryo transition.


Assuntos
Caenorhabditis elegans/embriologia , Embrião não Mamífero , Meiose/fisiologia , Oócitos/crescimento & desenvolvimento , Oogênese/fisiologia , Animais , Caenorhabditis elegans/metabolismo
10.
Alcohol Clin Exp Res ; 36(9): 1550-62, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22390787

RESUMO

BACKGROUND: Disturbed gait and balance are among the most consistent sequelae of chronic alcoholism. However, although a majority of alcoholics have never sought treatment, most investigations showing ataxia in alcohol-dependent individuals have relied on samples drawn from treated populations. In addition, few studies have addressed the associations of codependence on other drugs with alcoholic gait and balance disturbance. METHODS: This study employed the Walk-a-Line Ataxia Battery (Fregly et al. Alcohol Clin Exp Res 1972;43:395-399) to assess gait and balance in treatment-naïve, actively drinking alcohol-dependent men and women (TNA; n = 69) who were dependent on alcohol only (ALC; n = 43), or who also had a lifetime drug dependence (ALC + DRG; n = 26; i.e., methamphetamine, cocaine, opiates, and/or marijuana), compared with nonsubstance abusing controls (NSAC; n = 74).We also examined associations between lifetime alcohol use and age with gait and balance measures. RESULTS: Our main findings were (i) no evidence of disturbed gait and balance in ALC versus NSAC and (ii) significantly disturbed gait and balance in ALC + DRG, relative to both NSAC and ALC, along with steeper age-associated decline in gait and balance performance in ALC versus ALC + DRG. CONCLUSIONS: Our results provide evidence consistent with previous studies that TNA (without a lifetime drug codependence) may represent a population that is different and less impaired (including in gait and balance) than treated alcoholics. Additionally, we provide evidence that ALC + DRG, with greater alcohol use and family drinking density than ALC, have an accelerated effect of age on gait and balance disturbance compared with both NSAC and ALC. The ALC + DRG group likely represents a subset of TNA with different characteristics than ALC.


Assuntos
Alcoolismo/fisiopatologia , Marcha/fisiologia , Equilíbrio Postural/fisiologia , Codependência Psicológica , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fumar/fisiopatologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia
11.
Genetics ; 221(1)2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35377419

RESUMO

The ubiquitin-mediated degradation of oocyte translational regulatory proteins is a conserved feature of the oocyte-to-embryo transition. In the nematode Caenorhabditis elegans, multiple translational regulatory proteins, including the TRIM-NHL RNA-binding protein LIN-41/Trim71 and the Pumilio-family RNA-binding proteins PUF-3 and PUF-11, are degraded during the oocyte-to-embryo transition. Degradation of each protein requires activation of the M-phase cyclin-dependent kinase CDK-1, is largely complete by the end of the first meiotic division and does not require the anaphase-promoting complex. However, only LIN-41 degradation requires the F-box protein SEL-10/FBW7/Cdc4p, the substrate recognition subunit of an SCF-type E3 ubiquitin ligase. This finding suggests that PUF-3 and PUF-11, which localize to LIN-41-containing ribonucleoprotein particles, are independently degraded through the action of other factors and that the oocyte ribonucleoprotein particles are disassembled in a concerted fashion during the oocyte-to-embryo transition. We develop and test the hypothesis that PUF-3 and PUF-11 are targeted for degradation by the proteasome-associated HECT-type ubiquitin ligase ETC-1/UBE3C/Hul5, which is broadly expressed in C. elegans. We find that several GFP-tagged fusion proteins that are degraded during the oocyte-to-embryo transition, including fusions with PUF-3, PUF-11, LIN-41, IFY-1/Securin, and CYB-1/Cyclin B, are incompletely degraded when ETC-1 function is compromised. However, it is the fused GFP moiety that appears to be the critical determinant of this proteolysis defect. These findings are consistent with a conserved role for ETC-1 in promoting proteasome processivity and suggest that proteasomal processivity is an important element of the oocyte-to-embryo transition during which many key oocyte regulatory proteins are rapidly targeted for degradation.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Meiose , Oócitos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Fatores de Transcrição/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética
12.
Elife ; 112022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36098634

RESUMO

Gap-junctional signaling mediates myriad cellular interactions in metazoans. Yet, how gap junctions control the positioning of cells in organs is not well understood. Innexins compose gap junctions in invertebrates and affect organ architecture. Here, we investigate the roles of gap-junctions in controlling distal somatic gonad architecture and its relationship to underlying germline stem cells in Caenorhabditis elegans. We show that a reduction of soma-germline gap-junctional activity causes displacement of distal sheath cells (Sh1) towards the distal end of the gonad. We confirm, by live imaging, transmission electron microscopy, and antibody staining, that bare regions-lacking somatic gonadal cell coverage of germ cells-are present between the distal tip cell (DTC) and Sh1, and we show that an innexin fusion protein used in a prior study encodes an antimorphic gap junction subunit that mispositions Sh1. We determine that, contrary to the model put forth in the prior study based on this fusion protein, Sh1 mispositioning does not markedly alter the position of the borders of the stem cell pool nor of the progenitor cell pool. Together, these results demonstrate that gap junctions can control the position of Sh1, but that Sh1 position is neither relevant for GLP-1/Notch signaling nor for the exit of germ cells from the stem cell pool.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Células Germinativas/metabolismo , Gônadas/metabolismo , Células-Tronco/metabolismo
13.
Curr Biol ; 18(10): 705-714, 2008 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-18472420

RESUMO

BACKGROUND: In C. elegans, a sperm-sensing mechanism regulates oocyte meiotic maturation and ovulation, tightly coordinating sperm availability and embryo production; sperm release the major sperm protein (MSP) signal to trigger meiotic resumption. Meiotic arrest depends on the parallel function of the oocyte VAB-1 MSP/Eph receptor and somatic G protein signaling. MSP promotes meiotic maturation by antagonizing Eph receptor signaling and counteracting inhibitory inputs from the gonadal sheath cells. RESULTS: Here, we present evidence suggesting that in the absence of the MSP ligand, the VAB-1 Eph receptor inhibits meiotic maturation while either in or in transit to the endocytic-recycling compartment. VAB-1::GFP localization to the RAB-11-positive endocytic-recycling compartment is independent of ephrins but is antagonized by MSP signaling. Two negative regulators of oocyte meiotic maturation, DAB-1/Disabled and RAN-1, interact with the VAB-1 receptor and are required for its accumulation in the endocytic-recycling compartment in the absence of MSP or sperm (hereafter referred to as MSP/sperm). Inactivation of the endosomal recycling regulators rme-1 or rab-11.1 causes a vab-1-dependent reduction in the meiotic-maturation rate in the presence of MSP/sperm. Further, we show that Galpha(s) signaling in the gonadal sheath cells, which is required for meiotic maturation in the presence of MSP/sperm, affects VAB-1::GFP trafficking in oocytes. CONCLUSIONS: Regulated endocytic trafficking of the VAB-1 MSP/Eph receptor contributes to the control of oocyte meiotic maturation in C. elegans. Eph receptor trafficking in other systems may be influenced by the conserved proteins DAB-1/Disabled and RAN-1 and by crosstalk with G protein signaling in neighboring cells.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Helminto/metabolismo , Meiose/fisiologia , Oócitos/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Caenorhabditis elegans/citologia , Endocitose , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Masculino , Proteínas Nucleares/metabolismo , Oócitos/citologia , Ovulação , Transdução de Sinais , Espermatozoides/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo
14.
Curr Biol ; 17(20): R890-2, 2007 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-17956750

RESUMO

It has long been appreciated that the oocyte cortex plays a key role in regulating fertilization and establishing embryonic polarity. Recent studies have identified the anti-phosphatase EGG-3 as a cortical anchor for regulatory proteins required for launching embryogenesis in Caenhorhabditis elegans.


Assuntos
Desenvolvimento Embrionário/fisiologia , Animais , Padronização Corporal/fisiologia , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/fisiologia , Quitina Sintase/fisiologia , Feminino , Modelos Biológicos , Oócitos/fisiologia , Proteínas Tirosina Quinases/fisiologia
15.
Biomolecules ; 10(12)2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33321846

RESUMO

In Caenorhabditis elegans, gap junctions couple cells of the somatic gonad with the germline to support germ cell proliferation and gametogenesis. A strong loss-of-function mutation (T239I) affects the second extracellular loop (EL2) of the somatic INX-8 hemichannel subunit. These mutant hemichannels form non-functional gap junctions with germline-expressed innexins. We conducted a genetic screen for suppressor mutations that restore germ cell proliferation in the T239I mutant background and isolated seven intragenic mutations, located in diverse domains of INX-8 but not the EL domains. These second-site mutations compensate for the original channel defect to varying degrees, from nearly complete wild-type rescue, to partial rescue of germline proliferation. One suppressor mutation (E350K) supports the innexin cryo-EM structural model that the channel pore opening is surrounded by a cytoplasmic dome. Two suppressor mutations (S9L and I36N) may form leaky channels that support germline proliferation but cause the demise of somatic sheath cells. Phenotypic analyses of three of the suppressors reveal an equivalency in the rescue of germline proliferation and comparable delays in gametogenesis but a graded rescue of fertility. The mutations described here may be useful for elucidating the biochemical pathways that produce the active biomolecules transiting through soma-germline gap junctions.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Conexinas/genética , Gametogênese/genética , Organismos Hermafroditas/genética , Mutação , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/citologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/metabolismo , Proliferação de Células , Conexinas/química , Conexinas/metabolismo , Fertilidade/genética , Junções Comunicantes/metabolismo , Junções Comunicantes/ultraestrutura , Gônadas/citologia , Gônadas/metabolismo , Organismos Hermafroditas/citologia , Organismos Hermafroditas/metabolismo , Masculino , Oócitos/citologia , Oócitos/metabolismo , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Espermatozoides/citologia , Espermatozoides/metabolismo , Homologia Estrutural de Proteína
16.
Elife ; 92020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32735213

RESUMO

Gap junctions are ubiquitous in metazoans and play critical roles in important biological processes, including electrical conduction and development. Yet, only a few defined molecules passing through gap junction channels have been linked to specific functions. We isolated gap junction channel mutants that reduce coupling between the soma and germ cells in the Caenorhabditis elegans gonad. We provide evidence that malonyl-CoA, the rate-limiting substrate for fatty acid synthesis (FAS), is produced in the soma and delivered through gap junctions to the germline; there it is used in fatty acid synthesis to critically support embryonic development. Separation of malonyl-CoA production from its site of utilization facilitates somatic control of germline development. Additionally, we demonstrate that loss of malonyl-CoA production in the intestine negatively impacts germline development independently of FAS. Our results suggest that metabolic outsourcing of malonyl-CoA may be a strategy by which the soma communicates nutritional status to the germline.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/embriologia , Desenvolvimento Embrionário/genética , Células Germinativas/crescimento & desenvolvimento , Malonil Coenzima A/genética , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Junções Comunicantes/fisiologia , Gônadas/crescimento & desenvolvimento , Malonil Coenzima A/metabolismo
17.
Genetics ; 214(4): 869-893, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32060018

RESUMO

Mutations affecting spliceosomal proteins are frequently found in hematological malignancies, including myelodysplastic syndromes and acute myeloid leukemia (AML). DDX41/Abstrakt is a metazoan-specific spliceosomal DEAD-box RNA helicase that is recurrently mutated in inherited myelodysplastic syndromes and in relapsing cases of AML. The genetic properties and genomic impacts of disease-causing missense mutations in DDX41 and other spliceosomal proteins have been uncertain. Here, we conduct a comprehensive analysis of the Caenorhabditis elegans DDX41 ortholog, SACY-1 Biochemical analyses defined SACY-1 as a component of the C. elegans spliceosome, and genetic analyses revealed synthetic lethal interactions with spliceosomal components. We used the auxin-inducible degradation system to analyze the consequence of SACY-1 depletion on the transcriptome using RNA sequencing. SACY-1 depletion impacts the transcriptome through splicing-dependent and splicing-independent mechanisms. Altered 3' splice site usage represents the predominant splicing defect observed upon SACY-1 depletion, consistent with a role for SACY-1 in the second step of splicing. Missplicing events appear more prevalent in the soma than the germline, suggesting that surveillance mechanisms protect the germline from aberrant splicing. The transcriptome changes observed after SACY-1 depletion suggest that disruption of the spliceosome induces a stress response, which could contribute to the cellular phenotypes conferred by sacy-1 mutant alleles. Multiple sacy-1/ddx41 missense mutations, including the R525H human oncogenic variant, confer antimorphic activity, suggesting that their incorporation into the spliceosome is detrimental. Antagonistic variants that perturb the function of the spliceosome may be relevant to the disease-causing mutations, including DDX41, affecting highly conserved components of the spliceosome in humans.


Assuntos
Proteínas de Caenorhabditis elegans/genética , RNA Helicases DEAD-box/genética , Mutação de Sentido Incorreto , Síndromes Mielodisplásicas/genética , Spliceossomos/genética , Animais , Caenorhabditis elegans , Sítios de Splice de RNA
18.
Curr Biol ; 16(3): R93-5, 2006 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-16461272

RESUMO

The transition from oocyte to embryo is among the most enthralling events in developmental biology. Recent studies of this transition in the nematode Caenorhabditis elegans have revealed how conserved kinases administer the destruction of key oocyte meiotic regulators to create an embryo.


Assuntos
Padronização Corporal/fisiologia , Caenorhabditis elegans/embriologia , Quinases Ciclina-Dependentes/metabolismo , Embrião não Mamífero/enzimologia , Meiose/fisiologia , Modelos Biológicos , Oócitos/enzimologia , Adenosina Trifosfatases/metabolismo , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Transporte/metabolismo , Feminino , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Tirosina Quinases/metabolismo
19.
Curr Biol ; 16(13): 1257-68, 2006 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-16824915

RESUMO

BACKGROUND: A conserved biological feature of sexual reproduction in animals is that oocytes arrest in meiotic prophase and resume meiosis in response to extraovarian signals. In C. elegans, sperm trigger meiotic resumption by means of the major sperm protein (MSP) signal. MSP promotes meiotic resumption by functioning as an ephrin-signaling antagonist and by counteracting inhibitory inputs from the somatic gonadal sheath cells. RESULTS: By using a genome-wide RNAi screen in a female-sterile genetic background, we identified 17 conserved genes that maintain meiotic arrest in the absence of the MSP signal. In vitro binding experiments show that MSP promotes oocyte mitogen-activated protein kinase activation and meiotic maturation in part through direct interaction with the VAB-1 Eph receptor. Four conserved proteins, including a disabled protein (DAB-1), a vav family GEF (VAV-1), a protein kinase C (PKC-1), and a STAM homolog (PQN-19), function with the VAB-1 Eph/MSP receptor in oocytes. We show that antagonistic Galphao/i and Galphas signaling pathways function in the soma to regulate meiotic maturation in parallel to the VAB-1 pathway. Galphas activity is necessary and sufficient to promote meiotic maturation, which it does in part by antagonizing inhibitory sheath/oocyte gap-junctional communication. CONCLUSIONS: Our findings show that oocyte Eph receptor and somatic cell G protein signaling pathways control meiotic diapause in C. elegans, highlighting contrasts and parallels between MSP signaling in C. elegans and luteinizing hormone signaling in mammals.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Proteínas de Helminto/metabolismo , Meiose/fisiologia , Receptores da Família Eph/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Animais , Caenorhabditis elegans/citologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Perfilação da Expressão Gênica , Mamíferos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Oócitos/metabolismo , Interferência de RNA , Receptores Proteína Tirosina Quinases/metabolismo
20.
Genetics ; 210(3): 1011-1037, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30206186

RESUMO

In the nematode Caenorhabditis elegans, the conserved LIN-41 RNA-binding protein is a translational repressor that coordinately controls oocyte growth and meiotic maturation. LIN-41 exerts these effects, at least in part, by preventing the premature activation of the cyclin-dependent kinase CDK-1 Here we investigate the mechanism by which LIN-41 is rapidly eliminated upon the onset of meiotic maturation. Elimination of LIN-41 requires the activities of CDK-1 and multiple SCF (Skp1, Cul1, and F-box protein)-type E3 ubiquitin ligase subunits, including the conserved substrate adaptor protein SEL-10/Fbw7/Cdc4, suggesting that LIN-41 is a target of ubiquitin-mediated protein degradation. Within the LIN-41 protein, two nonoverlapping regions, Deg-A and Deg-B, are individually necessary for LIN-41 degradation; both contain several potential phosphodegron sequences, and at least one of these sequences is required for LIN-41 degradation. Finally, Deg-A and Deg-B are sufficient, in combination, to mediate SEL-10-dependent degradation when transplanted into a different oocyte protein. Although LIN-41 is a potent inhibitor of protein translation and M phase entry, the failure to eliminate LIN-41 from early embryos does not result in the continued translational repression of LIN-41 oocyte messenger RNA targets. Based on these observations, we propose a model for the elimination of LIN-41 by the SEL-10 E3 ubiquitin ligase and suggest that LIN-41 is inactivated before it is degraded. Furthermore, we provide evidence that another RNA-binding protein, the GLD-1 tumor suppressor, is regulated similarly. Redundant mechanisms to extinguish translational repression by RNA-binding proteins may both control and provide robustness to irreversible developmental transitions, including meiotic maturation and the oocyte-to-embryo transition.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/metabolismo , Embrião não Mamífero/metabolismo , Oócitos/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Feminino , Edição de Genes , Regulação da Expressão Gênica no Desenvolvimento , Meiose , Fenótipo , Domínios Proteicos , Proteólise , Fatores de Transcrição/química , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA