Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Mol Cell ; 43(5): 834-42, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21884983

RESUMO

While lysine acetylation in the nucleus is well characterized, comparatively little is known about its significance in cytoplasmic signaling. Here we show that inhibition of the Sirt1 deacetylase, which is primarily cytoplasmic in cancer cell lines, sensitizes these cells to caspase-2-dependent death. To identify relevant Sirt1 substrates, we developed a proteomics strategy, enabling the identification of a range of putative substrates, including 14-3-3ζ, a known direct regulator of caspase-2. We show here that inhibition of Sirtuin activity accelerates caspase activation and overrides caspase-2 suppression by nutrient abundance. Furthermore, 14-3-3ζ is acetylated prior to caspase activation, and supplementation of Xenopus egg extract with glucose-6-phosphate, which promotes caspase-2/14-3-3ζ binding, enhances 14-3-3ζ-directed Sirtuin activity. Conversely, inhibiting Sirtuin activity promotes14-3-3ζ dissociation from caspase-2 in both egg extract and human cultured cells. These data reveal a role for Sirt1 in modulating apoptotic sensitivity, in response to metabolic changes, by antagonizing 14-3-3ζ acetylation.


Assuntos
Proteínas 14-3-3/genética , Biotina/genética , Caspase 2/genética , Sirtuína 1/metabolismo , Proteínas 14-3-3/metabolismo , Acetilação , Animais , Apoptose , Biotina/metabolismo , Caspase 2/metabolismo , Morte Celular , Linhagem Celular Tumoral , Citoplasma/metabolismo , Humanos , Proteômica , Sirtuína 1/genética
2.
EMBO J ; 31(5): 1279-92, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22246185

RESUMO

Many pro-apoptotic signals trigger mitochondrial cytochrome c release, leading to caspase activation and ultimate cellular breakdown. Cell survival pathways, including the mitogen-activated protein kinase (MAPK) cascade, promote cell viability by impeding mitochondrial cytochrome c release and by inhibiting subsequent caspase activation. Here, we describe a mechanism for the inhibition of cytochrome c-induced caspase activation by MAPK signalling, identifying a novel mode of apoptotic regulation exerted through Apaf-1 phosphorylation by the 90-kDa ribosomal S6 kinase (Rsk). Recruitment of 14-3-3ɛ to phosphorylated Ser268 impedes the ability of cytochrome c to nucleate apoptosome formation and activate downstream caspases. High endogenous levels of Rsk in PC3 prostate cancer cells or Rsk activation in other cell types promoted 14-3-3ɛ binding to Apaf-1 and rendered the cells insensitive to cytochrome c, suggesting a potential role for Rsk signalling in apoptotic resistance of prostate cancers and other cancers with elevated Rsk activity. Collectively, these results identify a novel locus of apoptosomal regulation wherein MAPK signalling promotes Rsk-catalysed Apaf-1 phosphorylation and consequent binding of 14-3-3ɛ, resulting in decreased cellular responsiveness to cytochrome c.


Assuntos
Proteínas 14-3-3/metabolismo , Apoptose , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Citocromos c/antagonistas & inibidores , Citocromos c/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Fosforilação , Ligação Proteica
3.
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
4.
EMBO J ; 28(20): 3216-27, 2009 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-19730412

RESUMO

The apoptotic initiator caspase-2 has been implicated in oocyte death, in DNA damage- and heat shock-induced death, and in mitotic catastrophe. We show here that the mitosis-promoting kinase, cdk1-cyclin B1, suppresses apoptosis upstream of mitochondrial cytochrome c release by phosphorylating caspase-2 within an evolutionarily conserved sequence at Ser 340. Phosphorylation of this residue, situated in the caspase-2 interdomain, prevents caspase-2 activation. S340 was susceptible to phosphatase 1 dephosphorylation, and an interaction between phosphatase 1 and caspase-2 detected during interphase was lost in mitosis. Expression of S340A non-phosphorylatable caspase-2 abrogated mitotic suppression of caspase-2 and apoptosis in various settings, including oocytes induced to undergo cdk1-dependent maturation. Moreover, U2OS cells treated with nocodazole were found to undergo mitotic catastrophe more readily when endogenous caspase-2 was replaced with the S340A mutant to lift mitotic inhibition. These data demonstrate that for apoptotic stimuli transduced by caspase-2, cell death is prevented during mitosis through the inhibitory phosphorylation of caspase-2 and suggest that under conditions of mitotic arrest, cdk1-cyclin B1 activity must be overcome for apoptosis to occur.


Assuntos
Apoptose/fisiologia , Caspase 2/metabolismo , Mitose/fisiologia , Animais , Apoptose/genética , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Caspase 2/genética , Linhagem Celular , Linhagem Celular Tumoral , Ciclina B/genética , Ciclina B/metabolismo , Ciclina B1 , Eletroforese em Gel de Poliacrilamida , Vetores Genéticos , Humanos , Lentivirus , Mitose/efeitos dos fármacos , Mitose/genética , Nocodazol/farmacologia , Oócitos , Fosforilação , RNA Interferente Pequeno , Serina/genética , Serina/metabolismo , Serina/fisiologia , Xenopus
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(14): 1181-9, 2007 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-17614281

RESUMO

BACKGROUND: Several checkpoint pathways employ Wee1-mediated inhibitory tyrosine phosphorylation of cyclin-dependent kinases (CDKs) to restrain cell-cycle progression. Whereas in vertebrates this strategy can delay both DNA replication and mitosis, in yeast cells only mitosis is delayed. This is particularly surprising because yeasts, unlike vertebrates, employ a single family of cyclins (B type) and the same CDK to promote both S phase and mitosis. The G2-specific arrest could be explained in two fundamentally different ways: tyrosine phosphorylation of cyclin/CDK complexes could leave sufficient residual activity to promote S phase, or S phase-promoting cyclin/CDK complexes could somehow be protected from checkpoint-induced tyrosine phosphorylation. RESULTS: We demonstrate that in Saccharomyces cerevisiae, several cyclin/CDK complexes are protected from inhibitory tyrosine phosphorylation, allowing Clb5,6p to promote DNA replication and Clb3,4p to promote spindle assembly, even under checkpoint-inducing conditions that block nuclear division. In vivo, S phase-promoting Clb5p/Cdc28p complexes were phosphorylated more slowly and dephosphorylated more effectively than were mitosis-promoting Clb2p/Cdc28p complexes. Moreover, we show that the CDK inhibitor (CKI) Sic1p protects bound Clb5p/Cdc28p complexes from tyrosine phosphorylation, allowing the accumulation of unphosphorylated complexes that are unleashed when Sic1p is degraded to promote S phase. The vertebrate CKI p27(Kip1) similarly protects Cyclin A/Cdk2 complexes from Wee1, suggesting that the antagonism between CKIs and Wee1 is evolutionarily conserved. CONCLUSIONS: In yeast cells, the combination of CKI binding and preferential phosphorylation/dephosphorylation of different B cyclin/CDK complexes renders S phase progression immune from checkpoints acting via CDK tyrosine phosphorylation.


Assuntos
Proteína Quinase CDC28 de Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/metabolismo , Mitose/fisiologia , Proteínas Tirosina Quinases/metabolismo , Fase S/fisiologia , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Actinas/antagonistas & inibidores , Actinas/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes , Ciclina B/metabolismo , Proteínas Inibidoras de Quinase Dependente de Ciclina , Replicação do DNA/fisiologia , Complexos Multiproteicos/metabolismo , Fosforilação , Proteínas Tirosina Fosfatases/metabolismo , Fuso Acromático/metabolismo , Tiazolidinas , Tirosina/metabolismo , ras-GRF1
7.
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
8.
Mol Biol Cell ; 17(4): 1779-89, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16467385

RESUMO

The Cdc25 phosphatase promotes entry into mitosis through the removal of inhibitory phosphorylations on the Cdc2 subunit of the Cdc2/CyclinB complex. During interphase, or after DNA damage, Cdc25 is suppressed by phosphorylation at Ser287 (Xenopus numbering; Ser216 of human Cdc25C) and subsequent binding of the small acidic protein, 14-3-3. As reported recently, at the time of mitotic entry, 14-3-3 protein is removed from Cdc25 and S287 is dephosphorylated by protein phosphatase 1 (PP1). After the initial activation of Cdc25 and consequent derepression of Cdc2/CyclinB, Cdc25 is further activated through a Cdc2-catalyzed positive feedback loop. Although the existence of such a loop has been appreciated for some time, the molecular mechanism for this activation has not been described. We report here that phosphorylation of S285 by Cdc2 greatly enhances recruitment of PP1 to Cdc25, thereby accelerating S287 dephosphorylation and mitotic entry. Moreover, we show that two other previously reported sites of Cdc2-catalyzed phosphorylation on Cdc25 are required for maximal biological activity of Cdc25, but they do not contribute to PP1 regulation and do not act solely through controlling S287 phosphorylation. Therefore, multiple mechanisms, including enhanced recruitment of PP1, are used to promote full activation of Cdc25 at the time of mitotic entry.


Assuntos
Proteína Quinase CDC2/metabolismo , Ciclina B/metabolismo , Retroalimentação Fisiológica , Mitose , Fosfoproteínas Fosfatases/fisiologia , Serina/metabolismo , Fosfatases cdc25/metabolismo , Proteínas 14-3-3/metabolismo , Animais , Ativação Enzimática , Mutação , Fosforilação , Proteína Fosfatase 1 , Serina/genética , Treonina/metabolismo , Xenopus , Fosfatases cdc25/química , Fosfatases cdc25/genética
9.
Invest Ophthalmol Vis Sci ; 48(1): 303-12, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17197547

RESUMO

PURPOSE: To employ Mie scattering theory to predict the light-scattering from micrometer-sized particles surrounded by lipid shells, called multilamellar bodies (MLBs), reported in human age-related nuclear cataracts. METHODS: Mie scattering theory is applicable to randomly distributed spherical and globular particles separated by distances much greater than the wavelength of incident light. With an assumed refractive index of 1.40 for nuclear cytoplasm, particle refractive indices from 1.33 to 1.58 were used to calculate scattering efficiencies for particle radii 0.05 to 3 microm and incident light with wavelengths (in vacuo) of 400, 550, and 700 nm. RESULTS: Surface plots of scattering efficiency versus particle radius and refractive index were calculated for coated spherical particles. Pronounced peaks and valleys identified combinations of particle parameters that produce high and low scattering efficiencies. Small particles (<0.3 microm radius) had low scattering efficiency over a wide range of particle refractive indices. Particles with radii 0.6 to 3 microm and refractive indices 0.08 to 0.10 greater (or less) than the surrounding cytoplasm had very high scattering efficiencies. This size range corresponds well to MLBs in cataractous nuclei (average MLB radius, 1.4 microm) and, at an estimated 4000 particles/mm(3) of tissue, up to 18% of the incident light was scattered primarily within a 20 degrees forward cone. CONCLUSIONS: The calculated size of spherical particles that scatter efficiently was close to the observed dimensions of MLBs in cataractous nuclei. Particle refractive indices only 0.02 units different from the surrounding cytoplasm scatter a significant amount of light. These results suggest that the MLBs observed in human age-related nuclear cataracts may be major sources of forward light scattering that reduces contrast of fine details, particularly under dim light.


Assuntos
Catarata/patologia , Corpos de Inclusão/efeitos da radiação , Núcleo do Cristalino/efeitos da radiação , Modelos Teóricos , Espalhamento de Radiação , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/fisiologia , Humanos , Corpos de Inclusão/ultraestrutura , Núcleo do Cristalino/ultraestrutura , Luz , Pessoa de Meia-Idade , Tamanho da Partícula
10.
Mol Biol Cell ; 15(7): 3497-508, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15133124

RESUMO

To study the dynamics of stress fiber components in cultured fibroblasts, we expressed alpha-actinin and the myosin II regulatory myosin light chain (MLC) as fusion proteins with green fluorescent protein. Myosin activation was stimulated by treatment with calyculin A, a serine/threonine phosphatase inhibitor that elevates MLC phosphorylation, or with LPA, another agent that ultimately stimulates phosphorylation of MLC via a RhoA-mediated pathway. The resulting contraction caused stress fiber shortening and allowed observation of changes in the spacing of stress fiber components. We have observed that stress fibers, unlike muscle myofibrils, do not contract uniformly along their lengths. Although peripheral regions shortened, more central regions stretched. We detected higher levels of MLC and phosphorylated MLC in the peripheral region of stress fibers. Fluorescence recovery after photobleaching revealed more rapid exchange of myosin and alpha-actinin in the middle of stress fibers, compared with the periphery. Surprisingly, the widths of the myosin and alpha-actinin bands in stress fibers also varied in different regions. In the periphery, the banding patterns for both proteins were shorter, whereas in central regions, where stretching occurred, the bands were wider.


Assuntos
Actinina/metabolismo , Cadeias Leves de Miosina/metabolismo , Miosina Tipo II/metabolismo , Fibras de Estresse/fisiologia , Fibras de Estresse/ultraestrutura , Actinina/análise , Actinina/genética , Animais , Fibroblastos/imunologia , Fibroblastos/metabolismo , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Lisofosfolipídeos/farmacologia , Toxinas Marinhas , Camundongos , Cadeias Leves de Miosina/análise , Cadeias Leves de Miosina/genética , Miosina Tipo II/análise , Miosina Tipo II/genética , Oxazóis/farmacologia , Fosforilação/efeitos dos fármacos , Fotodegradação , Células Swiss 3T3
11.
BMC Ophthalmol ; 3: 1, 2003 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-12515578

RESUMO

BACKGROUND: Compaction of human ocular lens fiber cells as a function of both aging and cataractogenesis has been demonstrated previously using scanning electron microscopy. The purpose of this investigation is to quantify morphological differences in the inner nuclear regions of cataractous and non-cataractous human lenses from individuals with diabetes. The hypothesis is that, even in the presence of the osmotic stress caused by diabetes, compaction rather than swelling occurs in the nucleus of diabetic lenses. METHODS: Transparent and nuclear cataractous lenses from diabetic patients were examined by scanning electron microscopy (SEM). Measurements of the fetal nuclear (FN) elliptical angles (anterior and posterior), embryonic nuclear (EN) anterior-posterior (A-P) axial thickness, and the number of EN fiber cell membrane folds over 20 microns were compared. RESULTS: Diabetic lenses with nuclear cataract exhibited smaller FN elliptical angles, smaller EN axial thicknesses, and larger numbers of EN compaction folds than their non-cataractous diabetic counterparts. CONCLUSION: As in non-diabetic lenses, the inner nuclei of cataractous lenses from diabetics were significantly more compacted than those of non-cataractous diabetics. Little difference between diabetic and non-diabetic compaction levels was found, suggesting that diabetes does not affect the degree of compaction. However, consistent with previous proposals, diabetes does appear to accelerate the formation of cataracts that are similar to age-related nuclear cataracts in non-diabetics. We conclude that as scattering increases in the diabetic lens with cataract formation, fiber cell compaction is significant.


Assuntos
Envelhecimento/patologia , Catarata/patologia , Diabetes Mellitus/patologia , Núcleo do Cristalino/ultraestrutura , Idoso , Idoso de 80 Anos ou mais , Junções Comunicantes/ultraestrutura , Humanos , Microscopia Eletrônica de Varredura , Pessoa de Meia-Idade , Pressão Osmótica , Espalhamento de Radiação
12.
Artigo em Inglês | MEDLINE | ID: mdl-23732469

RESUMO

Caspases are the primary drivers of apoptotic cell death, cleaving cellular proteins that are critical for dismantling the dying cell. Initially translated as inactive zymogenic precursors, caspases are activated in response to a variety of cell death stimuli. In addition to factors required for their direct activation (e.g., dimerizing adaptor proteins in the case of initiator caspases that lie at the apex of apoptotic signaling cascades), caspases are regulated by a variety of cellular factors in a myriad of physiological and pathological settings. For example, caspases may be modified posttranslationally (e.g., by phosphorylation or ubiquitylation) or through interaction of modulatory factors with either the zymogenic or active form of a caspase, altering its activation and/or activity. These regulatory events may inhibit or enhance enzymatic activity or may affect activity toward particular cellular substrates. Finally, there is emerging literature to suggest that caspases can participate in a variety of cellular processes unrelated to apoptotic cell death. In these settings, it is particularly important that caspases are maintained under stringent control to avoid inadvertent cell death. It is likely that continued examination of these processes will reveal new mechanisms of caspase regulation with implications well beyond control of apoptotic cell death.


Assuntos
Apoptose/fisiologia , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspases/metabolismo , Modelos Biológicos , Processamento de Proteína Pós-Traducional/fisiologia , Transdução de Sinais/fisiologia , Animais , Caspases/genética , Caspases/fisiologia , Humanos , Mapas de Interação de Proteínas/fisiologia
13.
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
14.
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
15.
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
16.
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
17.
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
18.
J Biol Chem ; 283(1): 367-379, 2008 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-17998202

RESUMO

Morphological hallmarks of apoptosis result from activation of the caspase family of cysteine proteases, which are opposed by a pro-survival family of inhibitors of apoptosis proteins (IAPs). In Drosophila, disruption of IAP function by Reaper, HID, and Grim (RHG) proteins is sufficient to induce cell death. RHG proteins have been reported to localize to mitochondria, which, in the case of both Reaper and Grim proteins, is mediated by an amphipathic helical domain known as the GH3. Through direct binding, Reaper can bring the Drosophila IAP (DIAP1) to mitochondria, concomitantly promoting IAP auto-ubiquitination and destruction. Whether this localization is sufficient to induce DIAP1 auto-ubiquitination has not been reported. In this study we characterize the interaction between Reaper and the mitochondria using both Xenopus and Drosophila systems. We find that Reaper concentrates on the outer surface of mitochondria in a nonperipheral manner largely mediated by GH3-lipid interactions. Importantly, we show that mitochondrial targeting of DIAP1 alone is not sufficient for degradation and requires Reaper binding. Conversely, Reaper able to bind IAPs, but lacking a mitochondrial targeting GH3 domain (DeltaGH3 Reaper), can induce DIAP1 turnover only if DIAP1 is otherwise targeted to membranes. Surprisingly, targeting DIAP1 to the endoplasmic reticulum instead of mitochondria is partially effective in allowing DeltaGH3 Reaper to promote DIAP1 degradation, suggesting that co-localization of DIAP and Reaper at a membrane surface is critical for the induction of DIAP degradation. Collectively, these data provide a specific function for the GH3 domain in conferring protein-lipid interactions, demonstrate that both Reaper binding and mitochondrial localization are required for accelerated IAP degradation, and suggest that membrane localization per se contributes to DIAP1 auto-ubiquitination and degradation.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Lipídeos de Membrana/metabolismo , Mitocôndrias/metabolismo , Animais , Sítios de Ligação , Western Blotting , Linhagem Celular , Drosophila , Proteínas de Drosophila/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas Inibidoras de Apoptose/genética , Lipossomos/metabolismo , Microscopia Confocal , Membranas Mitocondriais/metabolismo , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ubiquitinação , Xenopus
19.
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
20.
Cell ; 127(4): 759-73, 2006 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-17110335

RESUMO

DNA-responsive checkpoints prevent cell-cycle progression following DNA damage or replication inhibition. The mitotic activator Cdc25 is suppressed by checkpoints through inhibitory phosphorylation at Ser287 (Xenopus numbering) and docking of 14-3-3. Ser287 phosphorylation is a major locus of G2/M checkpoint control, although several checkpoint-independent kinases can phosphorylate this site. We reported previously that mitotic entry requires 14-3-3 removal and Ser287 dephosphorylation. We show here that DNA-responsive checkpoints also activate PP2A/B56delta phosphatase complexes to dephosphorylate Cdc25 at a site distinct from Ser287 (T138), the phosphorylation of which is required for 14-3-3 release. However, phosphorylation of T138 is not sufficient for 14-3-3 release from Cdc25. Our data suggest that creation of a 14-3-3 "sink," consisting of phosphorylated 14-3-3 binding intermediate filament proteins, including keratins, coupled with reduced Cdc25-14-3-3 affinity, contribute to Cdc25 activation. These observations identify PP2A/B56delta as a central checkpoint effector and suggest a mechanism for controlling 14-3-3 interactions to promote mitosis.


Assuntos
Proteínas 14-3-3/metabolismo , Mitose , Fosfoproteínas Fosfatases/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus/metabolismo , Fosfatases cdc25/metabolismo , Animais , Quinase 1 do Ponto de Checagem , Replicação do DNA , Ativação Enzimática , Células HCT116 , Células HeLa , Holoenzimas/metabolismo , Humanos , Filamentos Intermediários/metabolismo , Interfase , Queratinas/metabolismo , Fosforilação , Fosfotreonina/metabolismo , Proteínas Quinases/metabolismo , Proteína Fosfatase 2 , Subunidades Proteicas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA