Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
País/Região como assunto
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 109(51): 20931-6, 2012 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-23213260

RESUMO

Tripartite motif 39 (Trim39) is a RING domain-containing E3 ubiquitin ligase able to inhibit the anaphase-promoting complex (APC/C) directly. Through analysis of Trim39 function in p53-positive and p53-negative cells, we have found, surprisingly, that p53-positive cells lacking Trim39 could not traverse the G1/S transition. This effect did not result from disinhibition of the APC/C. Moreover, although Trim39 loss inhibited etoposide-induced apoptosis in p53-negative cells, apoptosis was enhanced by Trim39 knockdown in p53-positive cells. Furthermore, we show here that the Trim39 can directly bind and ubiquitylate p53 in vitro and in vivo, leading to p53 degradation. Depletion of Trim39 significantly increased p53 protein levels and cell growth retardation in multiple cell lines. We found that the relative importance of Trim39 and the well-characterized p53-directed E3 ligase, murine double minute 2 (MDM2), varied between cell types. In cells that were relatively insensitive to the MDM2 inhibitor, nutlin-3a, apoptosis could be markedly enhanced by siRNA directed against Trim39. As such, Trim39 may serve as a potential therapeutic target in tumors with WT p53 when MDM2 inhibition is insufficient to elevate p53 levels and apoptosis.


Assuntos
Proteínas de Transporte/química , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ubiquitina/química , Ubiquitinação , Motivos de Aminoácidos , Ciclossomo-Complexo Promotor de Anáfase , Apoptose , Ciclo Celular , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Replicação do DNA , Citometria de Fluxo/métodos , Fase G1 , Humanos , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases
2.
EMBO J ; 29(18): 3196-207, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20700104

RESUMO

Apoptosis ensures tissue homeostasis in response to developmental cues or cellular damage. Recently reported genome-wide RNAi screens have suggested that several metabolic regulators can modulate caspase activation in Drosophila. Here, we establish a previously unrecognized link between metabolism and Drosophila apoptosis by showing that cellular NADPH levels modulate the initiator caspase Dronc through its phosphorylation at S130. Depletion of NADPH removed this inhibitory phosphorylation, resulting in the activation of Dronc and subsequent cell death. Conversely, upregulation of NADPH prevented Dronc-mediated apoptosis upon DIAP1 RNAi or cycloheximide treatment. Furthermore, this CaMKII-mediated phosphorylation of Dronc hindered Dronc activation, but not its catalytic activity. Blockade of NADPH production aggravated the death-inducing activity of Dronc in specific neurons, but not in the photoreceptor cells of the eyes of transgenic flies; similarly, non-phosphorylatable Dronc was more potent than wild type in triggering specific neuronal apoptosis. Our observations reveal a novel regulatory circuitry in Drosophila apoptosis, and, as NADPH levels are elevated in cancer cells, also provide a genetic model to understand aberrations in cancer cell apoptosis resulting from metabolic alterations.


Assuntos
Apoptose , Caspases/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Neurônios/metabolismo , Animais , Animais Geneticamente Modificados , Western Blotting , Sobrevivência Celular , Células Cultivadas , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Ativação Enzimática , Imunoprecipitação , Malatos/metabolismo , NADP/metabolismo , Neurônios/citologia , RNA Interferente Pequeno/farmacologia
3.
Duke Law J ; 62(5): 1069-108, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25330553

RESUMO

The pharmaceutical industry relies on innovation. However, many innovative firms are cutting their research and development investments and seeing their new product pipelines dry up, due in part to a lack of sufficient patent protection. This Note identifies two major factors that have caused this inadequacy in patent protection. First, pharmaceutical patents are challenged early and often by generic manufacturers, as encouraged by the 1984 Hatch-Waxman Act. Second, the scope of pharmaceutical-patents is sometimes unduly restrained due to limited application of the doctrine of equivalents. Consequently, pharmaceutical patents, especially drug-product patents, are easily designed around and cannot offer the protection necessary for innovative firms to recoup their developmental costs. This Note argues for a wider application of means-plus-function clauses in pharmaceutical patents as a potential cure for this problem. Means-plus-function claims, although authorized by Congress in the 1952 Patent Act, have not been explored much in the pharmaceutical context. This Note argues that this claiming strategy is not only appropriate but also particularly effective for pharmaceutical patents. Means-plus-function claims would give drug-product patents adequate scope even with the limited use of the doctrine of equivalents and thus would provide the protection necessary for innovative firms to withstand frequent attacks by generic manufacturers. Finally, this Note examines issues anticipated with applying means-plus-function claims to pharmaceutical patents and proposes possible solutions.


Assuntos
Aprovação de Drogas/legislação & jurisprudência , Competição Econômica/legislação & jurisprudência , Patentes como Assunto/legislação & jurisprudência , Pesquisa/legislação & jurisprudência , Indústria Farmacêutica/legislação & jurisprudência , Medicamentos Genéricos/economia , História do Século XX , História do Século XXI , Humanos , Legislação de Medicamentos/economia , Legislação de Medicamentos/história , Pesquisa/economia , Estados Unidos , United States Food and Drug Administration
4.
Blood ; 116(1): 140-50, 2010 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-20351309

RESUMO

Arteriovenous-lymphatic endothelial cell fates are specified by the master regulators, namely, Notch, COUP-TFII, and Prox1. Whereas Notch is expressed in the arteries and COUP-TFII in the veins, the lymphatics express all 3 cell fate regulators. Previous studies show that lymphatic endothelial cell (LEC) fate is highly plastic and reversible, raising a new concept that all 3 endothelial cell fates may co-reside in LECs and a subtle alteration can result in a reprogramming of LEC fate. We provide a molecular basis verifying this concept by identifying a cross-control mechanism among these cell fate regulators. We found that Notch signal down-regulates Prox1 and COUP-TFII through Hey1 and Hey2 and that activated Notch receptor suppresses the lymphatic phenotypes and induces the arterial cell fate. On the contrary, Prox1 and COUP-TFII attenuate vascular endothelial growth factor signaling, known to induce Notch, by repressing vascular endothelial growth factor receptor-2 and neuropilin-1. We show that previously reported podoplanin-based LEC heterogeneity is associated with differential expression of Notch1 in human cutaneous lymphatics. We propose that the expression of the 3 cell fate regulators is controlled by an exquisite feedback mechanism working in LECs and that LEC fate is a consequence of the Prox1-directed lymphatic equilibrium among the cell fate regulators.


Assuntos
Fator II de Transcrição COUP/metabolismo , Células Endoteliais/metabolismo , Proteínas de Homeodomínio/metabolismo , Receptor Notch1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Fator II de Transcrição COUP/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Células Cultivadas , Regulação para Baixo , Células Endoteliais/citologia , Retroalimentação Fisiológica , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Ligação Proteica , Interferência de RNA , Receptor Notch1/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Serrate-Jagged , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
5.
Curr Biol ; 18(13): 933-42, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18571408

RESUMO

BACKGROUND: In response to DNA damage, cells undergo either cell-cycle arrest or apoptosis, depending on the extent of damage and the cell's capacity for DNA repair. Cell-cycle arrest induced by double-stranded DNA breaks depends on activation of the ataxia-telangiectasia (ATM) protein kinase, which phosphorylates cell-cycle effectors such as Chk2 and p53 to inhibit cell-cycle progression. ATM is recruited to double-stranded DNA breaks by a complex of sensor proteins, including Mre11/Rad50/Nbs1, resulting in autophosphorylation, monomerization, and activation of ATM kinase. RESULTS: In characterizing Aven protein, a previously reported apoptotic inhibitor, we have found that Aven can function as an ATM activator to inhibit G2/M progression. Aven bound to ATM and Aven overexpressed in cycling Xenopus egg extracts prevented mitotic entry and induced phosphorylation of ATM and its substrates. Immunodepletion of endogenous Aven allowed mitotic entry even in the presence of damaged DNA, and RNAi-mediated knockdown of Aven in human cells prevented autophosphorylation of ATM at an activating site (S1981) in response to DNA damage. Interestingly, Aven is also a substrate of the ATM kinase. Mutation of ATM-mediated phosphorylation sites on Aven reduced its ability to activate ATM, suggesting that Aven activation of ATM after DNA damage is enhanced by ATM-mediated Aven phosphorylation. CONCLUSIONS: These results identify Aven as a new ATM activator and describe a positive feedback loop operating between Aven and ATM. In aggregate, these findings place Aven, a known apoptotic inhibitor, as a critical transducer of the DNA-damage signal.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia , Proteína Quinase CDC2/metabolismo , Ciclina B/metabolismo , Ativação Enzimática , Retroalimentação Fisiológica , Células HeLa , Humanos , Fosforilação , Interferência de RNA , Xenopus
6.
Curr Biol ; 17(3): 213-24, 2007 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-17276914

RESUMO

BACKGROUND: Vertebrate oocytes are arrested in metaphase II of meiosis prior to fertilization by cytostatic factor (CSF). CSF enforces a cell-cycle arrest by inhibiting the anaphase-promoting complex (APC), an E3 ubiquitin ligase that targets Cyclin B for degradation. Although Cyclin B synthesis is ongoing during CSF arrest, constant Cyclin B levels are maintained. To achieve this, oocytes allow continuous slow Cyclin B degradation, without eliminating the bulk of Cyclin B, which would induce release from CSF arrest. However, the mechanism that controls this continuous degradation is not understood. RESULTS: We report here the molecular details of a negative feedback loop wherein Cyclin B promotes its own destruction through Cdc2/Cyclin B-mediated phosphorylation and inhibition of the APC inhibitor Emi2. Emi2 bound to the core APC, and this binding was disrupted by Cdc2/Cyclin B, without affecting Emi2 protein stability. Cdc2-mediated phosphorylation of Emi2 was antagonized by PP2A, which could bind to Emi2 and promote Emi2-APC interactions. CONCLUSIONS: Constant Cyclin B levels are maintained during a CSF arrest through the regulation of Emi2 activity. A balance between Cdc2 and PP2A controls Emi2 phosphorylation, which in turn controls the ability of Emi2 to bind to and inhibit the APC. This balance allows proper maintenance of Cyclin B levels and Cdc2 kinase activity during CSF arrest.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas F-Box/metabolismo , Oócitos/citologia , Fosfoproteínas Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-mos/metabolismo , Proteínas de Xenopus/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Animais , Proteínas Cdc20 , Proteínas de Ciclo Celular/metabolismo , Ciclina B/metabolismo , DNA Complementar , Inibidores Enzimáticos/farmacologia , Biblioteca Gênica , Humanos , Meiose , Ácido Okadáico/farmacologia , Oócitos/metabolismo , Fosforilação , Ligação Proteica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Xenopus
7.
J Cell Biol ; 197(3): 361-7, 2012 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-22529100

RESUMO

Proapoptotic Bcl-2 family members, such as Bax, promote release of cytochrome c from mitochondria, leading to caspase activation and cell death. It was previously reported that modulator of apoptosis protein 1 (MOAP-1), an enhancer of Bax activation induced by DNA damage, is stabilized by Trim39, a protein of unknown function. In this paper, we show that MOAP-1 is a novel substrate of the anaphase-promoting complex (APC/C(Cdh1)) ubiquitin ligase. The influence of Trim39 on MOAP-1 levels stems from the ability of Trim39 (a RING domain E3 ligase) to directly inhibit APC/C(Cdh1)-mediated protein ubiquitylation. Accordingly, small interfering ribonucleic acid-mediated knockdown of Cdh1 stabilized MOAP-1, thereby enhancing etoposide-induced Bax activation and apoptosis. These data identify Trim39 as a novel APC/C regulator and provide an unexpected link between the APC/C and apoptotic regulation via MOAP-1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína da Polipose Adenomatosa do Colo/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Caderinas/metabolismo , Proteínas de Transporte/metabolismo , Ubiquitina/metabolismo , Proteína X Associada a bcl-2/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteína da Polipose Adenomatosa do Colo/genética , Antígenos CD , Apoptose , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/genética , Western Blotting , Caderinas/antagonistas & inibidores , Caderinas/genética , Proteínas de Transporte/genética , Dano ao DNA , Citometria de Fluxo , Fase G1/fisiologia , Células HeLa , Humanos , Imunoprecipitação , RNA Interferente Pequeno/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinação
8.
Mol Biol Cell ; 22(8): 1207-16, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21325626

RESUMO

Homeostatic maintenance of cellular mitochondria requires a dynamic balance between fission and fusion, and controlled changes in morphology are important for processes such as apoptosis and cellular division. Interphase mitochondria have been described as an interconnected network that fragments as cells enter mitosis, and this mitotic mitochondrial fragmentation is known to be regulated by the dynamin-related GTPase Drp1 (dynamin-related protein 1), a key component of the mitochondrial division machinery. Loss of Drp1 function and the subsequent failure of mitochondrial division during mitosis lead to incomplete cytokinesis and the unequal distribution of mitochondria into daughter cells. During mitotic exit and interphase, the mitochondrial network reforms. Here we demonstrate that changes in mitochondrial dynamics as cells exit mitosis are driven in part through ubiquitylation of Drp1, catalyzed by the APC/C(Cdh1) (anaphase-promoting complex/cyclosome and its coactivator Cdh1) E3 ubiquitin ligase complex. Importantly, inhibition of Cdh1-mediated Drp1 ubiquitylation and proteasomal degradation during interphase prevents the normal G1 phase regrowth of mitochondrial networks following cell division.


Assuntos
Caderinas/metabolismo , Citocinese , Mitocôndrias/enzimologia , Proteínas Mitocondriais/metabolismo , Mitose , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Antígenos CD , Caderinas/antagonistas & inibidores , Caderinas/genética , Dinaminas , Estabilidade Enzimática , Fase G1/genética , GTP Fosfo-Hidrolases/deficiência , GTP Fosfo-Hidrolases/genética , Expressão Gênica , Inativação Gênica , Células HEK293 , Células HeLa , Humanos , Interfase/genética , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Mitocôndrias/genética , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Interferente Pequeno/metabolismo , Transfecção , Complexos Ubiquitina-Proteína Ligase/antagonistas & inibidores , Complexos Ubiquitina-Proteína Ligase/genética , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
9.
Mol Biol Cell ; 21(15): 2589-97, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20534816

RESUMO

Vertebrate eggs are arrested at Metaphase II by Emi2, the meiotic anaphase-promoting complex/cyclosome (APC/C) inhibitor. Although the importance of Emi2 during oocyte maturation has been widely recognized and its regulation extensively studied, its mechanism of action remained elusive. Many APC/C inhibitors have been reported to act as pseudosubstrates, inhibiting the APC/C by preventing substrate binding. Here we show that a previously identified zinc-binding region is critical for the function of Emi2, whereas the D-box is largely dispensable. We further demonstrate that instead of acting through a "pseudosubstrate" mechanism as previously hypothesized, Emi2 can inhibit Cdc20-dependent activation of the APC/C substoichiometrically, blocking ubiquitin transfer from the ubiquitin-charged E2 to the substrate. These findings provide a novel mechanism of APC/C inhibition wherein the final step of ubiquitin transfer is targeted and raise the interesting possibility that APC/C is inhibited by Emi2 in a catalytic manner.


Assuntos
Proteínas F-Box/química , Proteínas F-Box/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ubiquitina/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Xenopus/metabolismo , Motivos de Aminoácidos , Ciclossomo-Complexo Promotor de Anáfase , Animais , Biocatálise , Ativação Enzimática , Humanos , Ligação Proteica , Relação Estrutura-Atividade , Especificidade por Substrato , Complexos Ubiquitina-Proteína Ligase/antagonistas & inibidores
10.
Biosens Bioelectron ; 25(9): 2161-6, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20303735

RESUMO

Oligonucleotide probes on the sensor surface can be hybridized with single-strand DNA (ssDNA) that is formed from PCR products in ice bath after degeneration. Thus, detection of PCR products by piezoelectric sensors requires the participation of ssDNA PCR products in ice bath. When PCR products in ice bath are added into the buffer of the sensor well at room temperature, there will be a temperature change process during mixing. However, it still remains unclear whether the temperature change affects the frequency baseline stability of the sensor and the result judgment, which is the basic condition for detecting hybridization of nucleic acid. In this study, we detected the hybridization of HPV PCR products during temperature change process by a self-designed adjustable metal-clamping piezoelectric sensor. The study mainly involves sensor adjustment, probe immobilization and ice bath sample addition (at different concentrations and different volumes). The response curve of basic frequency in temperature change process showed three stages, i.e., increase, decrease to baseline, and continuous decrease to stability. The early increase of frequency and duration of the time can reach 55+/-7.4 Hz and 39 min when 40 microL sample (0-1 degrees C) was added into 110 microL buffer (25 degrees C). The frequency increase effect caused by temperature difference at early stage depends on the volume ratio of two liquids and on the temperature difference. The results indicate that we should pay more attention to possibly small volume of PCR products in ice bath and minor temperature difference of two liquids in operation.


Assuntos
Técnicas Biossensoriais/instrumentação , DNA de Cadeia Simples/análise , DNA de Cadeia Simples/genética , Sequência de Bases , Primers do DNA/genética , DNA de Cadeia Simples/química , Técnicas Eletroquímicas , Metais , Hibridização de Ácido Nucleico , Reação em Cadeia da Polimerase
11.
Nat Cell Biol ; 11(5): 644-51, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19396163

RESUMO

Loss of cell division cycle 2 (Cdc2, also known as Cdk1) activity after cyclin B degradation is necessary, but not sufficient, for mitotic exit. Proteins phosphorylated by Cdc2 and downstream mitotic kinases must be dephosphorylated. We report here that protein phosphatase-1 (PP1) is the main catalyst of mitotic phosphoprotein dephosphorylation. Suppression of PP1 during early mitosis is maintained through dual inhibition by Cdc2 phosphorylation and the binding of inhibitor-1. Protein kinase A (PKA) phosphorylates inhibitor-1, mediating binding to PP1. As Cdc2 levels drop after cyclin B degradation, auto-dephosphorylation of PP1 at its Cdc2 phosphorylation site (Thr 320) allows partial PP1 activation. This promotes PP1-regulated dephosphorylation at the activating site of inhibitor-1 (Thr 35) followed by dissociation of the inhibitor-1-PP1 complex and then full PP1 activation to promote mitotic exit. Thus, Cdc2 both phosphorylates multiple mitotic substrates and inhibits their PP1-mediated dephosphorylation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Mitose/fisiologia , Fosfoproteínas/metabolismo , Proteína Fosfatase 1/metabolismo , Proteínas/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Proteína Quinase CDC2 , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ciclina B/metabolismo , Ciclina B/farmacologia , Quinases Ciclina-Dependentes , Células HeLa , Humanos , Modelos Biológicos , Ácido Okadáico/farmacologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Fosforilação , Ligação Proteica/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Proteína Fosfatase 1/antagonistas & inibidores , Proteína Fosfatase 1/farmacologia , Proteínas/farmacologia , Purinas/farmacologia , Roscovitina , Treonina/metabolismo , Proteínas de Xenopus/antagonistas & inibidores , Proteínas de Xenopus/metabolismo , Xenopus laevis
12.
Dev Cell ; 16(6): 856-66, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19531356

RESUMO

Xenopus oocyte death is partly controlled by the apoptotic initiator caspase-2 (C2). We reported previously that oocyte nutrient depletion activates C2 upstream of mitochondrial cytochrome c release. Conversely, nutrient-replete oocytes inhibit C2 via S135 phosphorylation catalyzed by calcium/calmodulin-dependent protein kinase II. We now show that C2 phosphorylated at S135 binds 14-3-3zeta, thus preventing C2 dephosphorylation. Moreover, we determined that S135 dephosphorylation is catalyzed by protein phosphatase-1 (PP1), which directly binds C2. Although C2 dephosphorylation is responsive to metabolism, neither PP1 activity nor binding is metabolically regulated. Rather, release of 14-3-3zeta from C2 is controlled by metabolism and allows for C2 dephosphorylation. Accordingly, a C2 mutant unable to bind 14-3-3zeta is highly susceptible to dephosphorylation. Although this mechanism was initially established in Xenopus, we now demonstrate similar control of murine C2 by phosphorylation and 14-3-3 binding in mouse eggs. These findings provide an unexpected evolutionary link between 14-3-3 and metabolism in oocyte death.


Assuntos
Proteínas 14-3-3/metabolismo , Apoptose , Caspase 2/metabolismo , Oócitos/citologia , Oócitos/enzimologia , Proteína Fosfatase 1/metabolismo , Animais , Ativação Enzimática , Feminino , Camundongos , Fosforilação , Ligação Proteica , Xenopus
13.
Mol Biol Cell ; 19(8): 3536-43, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550795

RESUMO

The transition of oocytes from meiosis I (MI) to meiosis II (MII) requires partial cyclin B degradation to allow MI exit without S phase entry. Rapid reaccumulation of cyclin B allows direct progression into MII, producing a cytostatic factor (CSF)-arrested egg. It has been reported that dampened translation of the anaphase-promoting complex (APC) inhibitor Emi2 at MI allows partial APC activation and MI exit. We have detected active Emi2 translation at MI and show that Emi2 levels in MI are mainly controlled by regulated degradation. Emi2 degradation in MI depends not on Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), but on Cdc2-mediated phosphorylation of multiple sites within Emi2. As in MII, this phosphorylation is antagonized by Mos-mediated recruitment of PP2A to Emi2. Higher Cdc2 kinase activity in MI than MII allows sufficient Emi2 phosphorylation to destabilize Emi2 in MI. At MI anaphase, APC-mediated degradation of cyclin B decreases Cdc2 activity, enabling Cdc2-mediated Emi2 phosphorylation to be successfully antagonized by Mos-mediated PP2A recruitment. These data suggest a model of APC autoinhibition mediated by stabilization of Emi2; Emi2 proteins accumulate at MI exit and inhibit APC activity sufficiently to prevent complete degradation of cyclin B, allowing MI exit while preventing interphase before MII entry.


Assuntos
Ciclina B/fisiologia , Proteínas F-Box/fisiologia , Regulação da Expressão Gênica , Meiose , Proteínas Proto-Oncogênicas c-mos/fisiologia , Animais , Proteína Quinase CDC2 , Movimento Celular , Ciclina B/metabolismo , Quinases Ciclina-Dependentes , Endocitose , Células HL-60 , Humanos , Leucócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neutrófilos/metabolismo , Proteínas Proto-Oncogênicas c-mos/metabolismo
14.
J Proteome Res ; 6(12): 4624-33, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17988085

RESUMO

Previous investigations of plant responses to higher CO 2 levels were mostly based on physiological measurements and biochemical assays. In this study, a proteomic approach was employed to investigate plant response to higher CO 2 levels using rice as a model. Ten-day-old seedlings were progressively exposed to 760 ppm, 1140 ppm, and 1520 ppm CO 2 concentrations for 24 h each. The net photosynthesis rate ( P n), stomatal conductance ( G s), transpiration rate ( E), and intercellular to ambient CO 2 concentration ratio ( C i/ C a) were measured. P n, G s, and E showed a maximum increase at 1140 ppm CO 2, but further exposure to 1520 ppm for 24 h resulted in down regulation of these. Proteins extracted from leaves were subjected to 2-DE analysis, and 57 spots showing differential expression patterns, as detected by profile analysis, were identified by MALDI-TOF/TOF-MS. Most of the proteins belonged to photosynthesis, carbon metabolism, and energy pathways. Several molecular chaperones and ascorbate peroxidase were also found to respond to higher CO 2 levels. Concomitant with the down regulation of P n and G s, the levels of enzymes of the regeneration phase of the Calvin cycle were decreased. Correlations between the protein profiles and the photosynthetic measurements at the three CO 2 levels were explored.


Assuntos
Dióxido de Carbono/fisiologia , Oryza/metabolismo , Folhas de Planta/metabolismo , Proteoma/fisiologia , Plântula/metabolismo , Dióxido de Carbono/metabolismo , Eletroforese em Gel Bidimensional , Oryza/crescimento & desenvolvimento , Fotossíntese/fisiologia , Folhas de Planta/crescimento & desenvolvimento , Proteínas de Plantas/biossíntese , Proteínas de Plantas/química , Proteínas de Plantas/fisiologia , Plântula/crescimento & desenvolvimento
15.
Proc Natl Acad Sci U S A ; 104(42): 16564-9, 2007 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-17881560

RESUMO

Before fertilization, vertebrate eggs are arrested in meiosis II by cytostatic factor (CSF), which holds the anaphase-promoting complex (APC) in an inactive state. It was recently reported that Mos, an integral component of CSF, acts in part by promoting the Rsk-mediated phosphorylation of the APC inhibitor Emi2/Erp1. We report here that Rsk phosphorylation of Emi2 promotes its interaction with the protein phosphatase PP2A. Emi2 residues adjacent to the Rsk phosphorylation site were important for PP2A binding. An Emi2 mutant that retained Rsk phosphorylation but lacked PP2A binding could not be modulated by Mos. PP2A bound to Emi2 acted on two distinct clusters of sites phosphorylated by Cdc2, one responsible for modulating its stability during CSF arrest and one that controls binding to the APC. These findings provide a molecular mechanism for Mos action in promoting CSF arrest and also define an unusual mechanism, whereby protein phosphorylation recruits a phosphatase for dephosphorylation of distinct sites phosphorylated by another kinase.


Assuntos
Proteínas F-Box/metabolismo , Meiose , Óvulo/fisiologia , Fosforilase Fosfatase/metabolismo , Proteínas Proto-Oncogênicas c-mos/metabolismo , Proteínas de Xenopus/metabolismo , Sequência de Aminoácidos , Animais , Proteínas F-Box/genética , Humanos , Dados de Sequência Molecular , Fosforilação , Proteínas Proto-Oncogênicas c-mos/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Xenopus , Proteínas de Xenopus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA