Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Hum Reprod ; 37(3): 612-620, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34997960

RESUMO

STUDY QUESTION: Are mutations in MOS (MOS proto-oncogene, serine/threonine kinase) involved in early embryonic arrest in infertile women? SUMMARY ANSWER: We identified mutations in MOS that may cause human female infertility characterized by preimplantation embryonic arrest (PREMBA), and the effects of the mutations in human embryonic kidney 293T (HEK293T cells) and mouse oocytes provided evidence for a causal relation between MOS and female infertility. WHAT IS KNOWN ALREADY: MOS, an activator of mitogen-activated protein kinase, mediates germinal vesicle breakdown and metaphase II arrest. Female MOS knockout mice are viable but sterile. Thus, MOS seems to be an important part of the mammalian cell cycle mechanism that regulates female meiosis. STUDY DESIGN, SIZE, DURATION: Whole-exome sequencing, bioinformatics filtering analysis and genetic analysis were performed to identify two different biallelic mutations in MOS in two independent families. The infertile patients presenting with early embryonic arrest were recruited from October 2018 to June 2020. PARTICIPANTS/MATERIALS, SETTING, METHODS: The female patients diagnosed with primary infertility were recruited from the reproduction centres of local hospitals. Genomic DNA from the affected individuals, their family members and healthy controls was extracted from peripheral blood. We performed whole-exome sequencing in patients diagnosed with PREMBA. Functional effects of the mutations were investigated in HEK293T cells by western blotting and in mouse oocytes by microinjection and immunofluorescence. MAIN RESULTS AND THE ROLE OF CHANCE: We identified the homozygous missense mutation c.285C>A (p.(Asn95Lys)) and the compound heterozygous mutations c.467delG (p.(Gly156Alafs*18)) and c.956G>A (p.(Arg319His)) in MOS in two independent patients. The mutations c.285C>A (p.(Asn95Lys)) and c.467delG (p.(Gly156Alafs*18)) reduced the protein level of MOS, and all mutations reduced the ability of MOS to phosphorylate its downstream target, extracellular signal-regulated kinase1/2. In addition, the identified mutations reduced the capacity of exogenous human MOS to rescue the metaphase II exit phenotype, and the F-actin cytoskeleton of mouse oocytes was affected by the patient-derived mutations. LIMITATIONS, REASONS FOR CAUTION: Owing to the lack of in vivo data from patient oocytes, the exact molecular mechanism affected by MOS mutations and leading to PREMBA is still unknown and should be further investigated using knock-out or knock-in mice. WIDER IMPLICATIONS OF THE FINDINGS: We identified recessive mutations in MOS in two independent patients with the PREMBA phenotype. Our findings reveal the important role of MOS during human oocyte meiosis and embryonic development and suggest that mutations in MOS may be precise diagnostic markers for clinical genetic counselling. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the National Natural Science Foundation of China (81725006, 81822019, 81771581, 81971450, 81971382,82001538 and 82071642), the project supported by the Shanghai Municipal Science and Technology Major Project (2017SHZDZX01), the Project of the Shanghai Municipal Science and Technology Commission (19JC1411001), the Natural Science Foundation of Shanghai (19ZR1444500 and 21ZR1404800), the Shuguang Program of the Shanghai Education Development Foundation and the Shanghai Municipal Education Commission (18SG03), the Foundation of the Shanghai Health and Family Planning Commission (20154Y0162), the Capacity Building Planning Program for Shanghai Women and Children's Health Service and the collaborative innovation centre project construction for Shanghai Women and Children's Health. The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Infertilidade Feminina , Proteínas Oncogênicas v-mos/genética , Animais , China , Feminino , Células HEK293 , Humanos , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Mamíferos , Camundongos , Mutação , Oócitos/metabolismo , Gravidez
2.
J Cell Sci ; 127(Pt 12): 2749-60, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24741069

RESUMO

Egg activation at fertilization in mammals is initiated by prolonged Ca(2+) oscillations that trigger the completion of meiosis and formation of pronuclei. A fall in mitogen-activated protein kinase (MAPK) activity is essential for pronuclear formation, but the precise timing and mechanism of decline are unknown. Here, we have measured the dynamics of MAPK pathway inactivation during fertilization of mouse eggs using novel chemiluminescent MAPK activity reporters. This reveals that the MAPK activity decrease begins during the Ca(2+) oscillations, but MAPK does not completely inactivate until after pronuclear formation. The MAPKs present in eggs are Mos, MAP2K1 and MAP2K2 (MEK1 and MEK2, respectively) and MAPK3 and MAPK1 (ERK1 and ERK2, respectively). Notably, the MAPK activity decline at fertilization is not explained by upstream destruction of Mos, because a decrease in the signal from a Mos-luciferase reporter is not associated with egg activation. Furthermore, Mos overexpression does not affect the timing of MAPK inactivation or pronuclear formation. However, the late decrease in MAPK could be rapidly reversed by the protein phosphatase inhibitor, okadaic acid. These data suggest that the completion of meiosis in mouse zygotes is driven by an increased phosphatase activity and not by a decline in Mos levels or MEK activity.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Óvulo/enzimologia , Animais , Sinalização do Cálcio , Inibidores Enzimáticos/farmacologia , Feminino , Fertilização , Genes Reporter , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Ácido Okadáico/farmacologia , Proteínas Oncogênicas v-mos/genética , Proteínas Oncogênicas v-mos/metabolismo , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Espermatozoides/fisiologia
3.
Mol Carcinog ; 53(7): 578-88, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23475563

RESUMO

AMR-Me, a C-28 methylester derivative of triterpenoid compound Amooranin isolated from Amoora rohituka stem bark and the plant has been reported to possess multitude of medicinal properties. Our previous studies have shown that AMR-Me can induce apoptosis through mitochondrial apoptotic and MAPK signaling pathways by regulating the expression of apoptosis related genes in human breast cancer MCF-7 cells. However, the molecular mechanism of AMR-Me induced apoptotic cell death remains unclear. Our results showed that AMR-Me dose-dependently inhibited the proliferation of MCF-7 and MDA-MB-231 cells under serum-free conditions supplemented with 1 nM estrogen (E2) with an IC50 value of 0.15 µM, 0.45 µM, respectively. AMR-Me had minimal effects on human normal breast epithelial MCF-10A + ras and MCF-10A cells with IC50 value of 6 and 6.5 µM, respectively. AMR-Me downregulated PI3K p85, Akt1, and p-Akt in an ERα-independent manner in MCF-7 cells and no change in expression levels of PI3K p85 and Akt were observed in MDA-MB-231 cells treated under similar conditions. The PI3K inhibitor LY294002 suppressed Akt activation similar to AMR-Me and potentiated AMR-Me induced apoptosis in MCF-7 cells. EMSA revealed that AMR-Me inhibited nuclear factor-kappaB (NF-κB) DNA binding activity in MDA-MB-231 cells in a time-dependent manner and abrogated EGF induced NF-κB activation. From these studies we conclude that AMR-Me decreased ERα expression and effectively inhibited Akt phosphorylation in MCF-7 cells and inactivate constitutive nuclear NF-κB and its regulated proteins in MDA-MB-231 cells. Due to this multifactorial effect in hormone-dependent and independent breast cancer cells AMR-Me deserves attention for use in breast cancer prevention and therapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , NF-kappa B/antagonistas & inibidores , Ácido Oleanólico/análogos & derivados , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Fator de Transcrição RelA/antagonistas & inibidores , Mama/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Cromonas/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/farmacologia , Inibidores Enzimáticos/farmacologia , Estrogênios/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Mitocôndrias/efeitos dos fármacos , Morfolinas/farmacologia , Proteínas Nucleares/antagonistas & inibidores , Ácido Oleanólico/farmacologia , Proteínas Oncogênicas v-mos/biossíntese , Fosfatidilinositol 3-Quinases/biossíntese , Fosforilação , Extratos Vegetais/farmacologia , Proteínas Proto-Oncogênicas c-akt/biossíntese , Proteínas Proto-Oncogênicas c-rel , Fator de Transcrição RelB/antagonistas & inibidores
4.
Mol Reprod Dev ; 76(3): 289-300, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18671273

RESUMO

Resumption of meiosis from diplotene arrest during the first meiotic prophase in vertebrate oocytes is universally controlled by MPF, a heterodimer of Cdk1 and cyclin B. Activation of MPF depends on the withdrawal of Cdk1 inhibition by Wee1/Myt1 kinase on the one hand and the activation of Cdk1 by Cdc25 phosphatase on the other. It is relevant to know whether both these pathways are necessary to rescue diplotene arrest or if either one of them is sufficient. In MIH (17alpha, 20beta dihydroxy-4-pregnen-3-one) incubated perch (Anabas testudineus) oocytes we have examined these possibilities. Perch oocyte extract following MIH incubation showed a significant increase in Myt1 phosphorylation from 12 to 16 hr indicating its progressive deactivation. MIH induced Mos expression markedly increased at 16 hr effecting 95% GVBD. Cycloheximide inhibited MIH induced Mos expression and its phosphorylation, which in turn reduced Myt1 phosphorylation and GVBD. Myt1 phosphorylation was blocked in Mos immunodepleted oocytes. All these suggest the involvement of Mos in Myt1 phosphorylation. Oocytes incubated in MIH for 16 hr activated Cdc25, but such activation could not rescue the inhibition of GVBD due to Myt1 in Mos immunodepleted oocytes. Blocking Cdc25 with an antisense oligo significantly inhibited GVBD even though Myt1 remained deactivated during this period. Taken together, our findings indicate that MIH requires both pathways for perch oocyte maturation: the expression and activation of Mos, which is linked to Myt1 deactivation on the one hand, and the activation of Cdc25 on the other, as blocking either pathway compromised G2-M transition in perch oocytes.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Fase G2/fisiologia , Proteínas Oncogênicas v-mos/metabolismo , Oócitos/crescimento & desenvolvimento , Percas/fisiologia , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/genética , Cicloeximida/farmacologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hidroxiprogesteronas/metabolismo , Fator Promotor de Maturação/genética , Fator Promotor de Maturação/metabolismo , Proteínas Oncogênicas v-mos/genética , Oogênese/efeitos dos fármacos , Oogênese/fisiologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fosfatases cdc25/metabolismo
5.
J Cell Biol ; 111(2): 533-41, 1990 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2143197

RESUMO

Previous work has demonstrated that the Xenopus protooncogene mosxe can induce the maturation of prophase-arrested Xenopus oocytes. Recently, we showed that mosxe can transform murine NIH3T3 fibroblasts, although it exhibited only 1-2% of the transforming activity of the v-mos oncogene. In this study we have investigated the ability of the v-mos protein to substitute for the mosxe protein in stimulating Xenopus oocytes to complete meiosis. Microinjection of in vitro synthesized RNAs encoding either the mosxe or v-mos proteins stimulates resting oocytes to undergo germinal vesicle breakdown. Microinjection of an antisense oligonucleotide spanning the initiation codon of the mosxe gene blocked progesterone-induced oocyte maturation. When oocytes were microinjected first with the mosxe antisense oligonucleotide, and subsequently with in vitro synthesized v-mos RNA, meiotic maturation was rescued as evidenced by germinal vesicle breakdown. The v-mos protein exhibited in vitro kinase activity when recovered by immunoprecipitation from either microinjected Xenopus oocytes or transfected monkey COS-1 cells; however, in parallel experiments, we were unable to detect in vitro kinase activity associated with the mosxe protein. Microinjection of in vitro synthesized v-mos RNA into cleaving Xenopus embryos resulted in mitotic arrest, demonstrating that the v-mos protein can function like the mosxe protein as a component of cytostatic factor. These results exemplify the apparently conflicting effects of the v-mos protein, namely, its ability to induce maturation of oocytes, its ability to arrest mitotic cleavage of Xenopus embryo, and its ability to transform mammalian fibroblasts.


Assuntos
Oncogenes , Oócitos/citologia , Proteínas Oncogênicas de Retroviridae/metabolismo , Animais , Divisão Celular , Linhagem Celular , Embrião não Mamífero/citologia , Feminino , Expressão Gênica , Meiose , Mitose , Proteínas Oncogênicas v-mos , Oócitos/enzimologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Oncogênicas de Retroviridae/genética , Transfecção , Xenopus
6.
J Cell Biol ; 163(6): 1231-42, 2003 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-14691134

RESUMO

In cells containing disrupted spindles, the spindle assembly checkpoint arrests the cell cycle in metaphase. The budding uninhibited by benzimidazole (Bub) 1, mitotic arrest-deficient (Mad) 1, and Mad2 proteins promote this checkpoint through sustained inhibition of the anaphase-promoting complex/cyclosome. Vertebrate oocytes undergoing meiotic maturation arrest in metaphase of meiosis II due to a cytoplasmic activity termed cytostatic factor (CSF), which appears not to be regulated by spindle dynamics. Here, we show that microinjection of Mad1 or Mad2 protein into early Xenopus laevis embryos causes metaphase arrest like that caused by Mos. Microinjection of antibodies to either Mad1 or Mad2 into maturing oocytes blocks the establishment of CSF arrest in meiosis II, and immunodepletion of either protein blocked the establishment of CSF arrest by Mos in egg extracts. A Mad2 mutant unable to oligomerize (Mad2 R133A) did not cause cell cycle arrest in blastomeres or in egg extracts. Once CSF arrest has been established, maintenance of metaphase arrest requires Mad1, but not Mad2 or Bub1. These results suggest a model in which CSF arrest by Mos is mediated by the Mad1 and Mad2 proteins in a manner distinct from the spindle checkpoint.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Meiose/fisiologia , Metáfase/fisiologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-mos/metabolismo , Proteínas Repressoras/metabolismo , Animais , Anticorpos/farmacologia , Proteínas de Ligação ao Cálcio/farmacologia , Proteínas de Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Feminino , Genes cdc/efeitos dos fármacos , Genes cdc/fisiologia , Proteínas Mad2 , Meiose/efeitos dos fármacos , Metáfase/efeitos dos fármacos , Mutação/genética , Proteínas Nucleares , Proteínas Oncogênicas v-mos/genética , Proteínas Oncogênicas v-mos/metabolismo , Oócitos/citologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oogênese/efeitos dos fármacos , Oogênese/fisiologia , Fosfoproteínas/farmacologia , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Repressoras/farmacologia , Xenopus laevis
7.
J Cell Biol ; 157(4): 603-13, 2002 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-12011110

RESUMO

Vertebrate oocytes arrest in the second metaphase of meiosis (metaphase II [MII]) by an activity called cytostatic factor (CSF), with aligned chromosomes and stable spindles. Segregation of chromosomes occurs after fertilization. The Mos/.../MAPK (mitogen-activated protein kinases) pathway mediates this MII arrest. Using a two-hybrid screen, we identified a new MAPK partner from a mouse oocyte cDNA library. This protein is unstable during the first meiotic division and accumulates only in MII, where it localizes to the spindle. It is a substrate of the Mos/.../MAPK pathway. The depletion of endogenous RNA coding for this protein by three different means (antisense RNA, double-stranded [ds] RNA, or morpholino oligonucleotides) induces severe spindle defects specific to MII oocytes. Overexpressing the protein from an RNA not targeted by the morpholino rescues spindle destabilization. However, dsRNA has no effect on the first two mitotic divisions. We therefore have discovered a new MAPK substrate involved in maintaining spindle integrity during the CSF arrest of mouse oocytes, called MISS (for MAP kinase-interacting and spindle-stabilizing protein).


Assuntos
Proteínas de Transporte/isolamento & purificação , Proteínas de Ciclo Celular/isolamento & purificação , Peptídeos e Proteínas de Sinalização Intracelular , Sistema de Sinalização das MAP Quinases/fisiologia , Meiose/fisiologia , Proteínas Nucleares/isolamento & purificação , Oócitos/metabolismo , Fuso Acromático/metabolismo , Sequência de Aminoácidos/genética , Animais , Sequência de Bases/genética , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Clonagem Molecular , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Genes myc/genética , Camundongos , Camundongos Knockout , Mitose/genética , Dados de Sequência Molecular , Mutação/genética , Proteínas Nucleares/genética , Sondas de Oligonucleotídeos/farmacologia , Proteínas Oncogênicas v-mos/genética , Oócitos/citologia , Fenótipo , Estrutura Terciária de Proteína/genética , RNA Mensageiro/genética , RNA Mensageiro/isolamento & purificação
8.
J Cell Biol ; 165(1): 63-75, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15067021

RESUMO

Ca(2+) is a ubiquitous intracellular messenger that is important for cell cycle progression. Genetic and biochemical evidence support a role for Ca(2+) in mitosis. In contrast, there has been a long-standing debate as to whether Ca(2+) signals are required for oocyte meiosis. Here, we show that cytoplasmic Ca(2+) (Ca(2+)(cyt)) plays a dual role during Xenopus oocyte maturation. Ca(2+) signals are dispensable for meiosis entry (germinal vesicle breakdown and chromosome condensation), but are required for the completion of meiosis I. Interestingly, in the absence of Ca(2+)(cyt) signals oocytes enter meiosis more rapidly due to faster activation of the MAPK-maturation promoting factor (MPF) kinase cascade. This Ca(2+)-dependent negative regulation of the cell cycle machinery (MAPK-MPF cascade) is due to Ca(2+)(cyt) acting downstream of protein kinase A but upstream of Mos (a MAPK kinase kinase). Therefore, high Ca(2+)(cyt) delays meiosis entry by negatively regulating the initiation of the MAPK-MPF cascade. These results show that Ca(2+) modulates both the cell cycle machinery and nuclear maturation during meiosis.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Diferenciação Celular/fisiologia , Citoplasma/metabolismo , Meiose/genética , Oócitos/metabolismo , Animais , Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Ciclo Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Canais de Cloreto/efeitos dos fármacos , Canais de Cloreto/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citoplasma/genética , Vesículas Citoplasmáticas/metabolismo , Retroalimentação Fisiológica/efeitos dos fármacos , Retroalimentação Fisiológica/fisiologia , Feminino , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Oncogênicas v-mos/metabolismo , Oócitos/citologia , Oócitos/efeitos dos fármacos , Fuso Acromático/genética , Fuso Acromático/metabolismo , Xenopus laevis
9.
Science ; 253(5015): 74-6, 1991 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-1829549

RESUMO

The endogenous mos proto-oncogene product (Mos) is required for meiotic maturation. In Xenopus oocytes, the ras oncogene product (Ras) can induce meiotic maturation and high levels of M-phase--promoting factor (MPF) independent of endogenous Mos, indicating that a parallel pathway to metaphase exists. In addition, Ras, like Mos and cytostatic factor, can arrest Xenopus embryonic cell cleavage in mitosis and maintain high levels of MPF. Thus, in the Xenopus oocyte and embryo systems Ras functions in the M phase of the cell cycle. The embryonic cleavage arrest assay is a rapid and sensitive test for Ras function.


Assuntos
Fator Promotor de Maturação/metabolismo , Meiose/efeitos dos fármacos , Proteína Oncogênica p21(ras)/farmacologia , Oogênese/efeitos dos fármacos , Animais , Células Cultivadas , Técnicas In Vitro , Proteínas Oncogênicas v-mos , Progesterona/farmacologia , Proteínas Oncogênicas de Retroviridae/farmacologia , Xenopus laevis
10.
Science ; 256(5058): 825-7, 1992 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-1317056

RESUMO

Controversy exists as to whether the interaction of a guanosine triphosphatase activating protein (GAP) with Ras proteins functions both to initiate and to terminate Ras-dependent signaling events or only to terminate them. GAP-C, a carboxyl-terminal fragment of GAP that is sufficient to stimulate GTPase activity, inhibited the stimulation of transcription produced by some oncoproteins (v-Src, polyoma middle T, wild-type Ras, and oncogenic Ras) but not that produced by v-Mos. Wild-type GAP did not affect transcription induced by oncogenic Ras but reversed the inhibitory effect of GAP-C on transcription induced by oncogenic Ras. These results indicate that GAP is a negative regulator of wild-type Ras and elicits a downstream signal by interacting with Ras-GTP (guanosine triphosphate).


Assuntos
Proteínas de Ciclo Celular , Transformação Celular Neoplásica , Elementos Facilitadores Genéticos , Genes ras , Polyomavirus/genética , Proteínas/metabolismo , Ativação Transcricional , ras-GRF1 , Células 3T3 , Animais , Antígenos Transformantes de Poliomavirus/genética , Células CHO , Cloranfenicol O-Acetiltransferase/genética , Cloranfenicol O-Acetiltransferase/metabolismo , Cricetinae , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Proteínas Ativadoras de GTPase , Humanos , Camundongos , Proteínas Oncogênicas v-mos , Oncogenes , Regiões Promotoras Genéticas , Proteínas Tirosina Quinases/genética , Proteínas Oncogênicas de Retroviridae/genética , Transdução de Sinais , Vírus 40 dos Símios/genética , Transcrição Gênica , Transfecção , Proteínas Ativadoras de ras GTPase
11.
Curr Opin Genet Dev ; 3(1): 19-25, 1993 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8384034

RESUMO

The product of the mos proto-oncogene is a serine/threonine kinase that is expressed at high levels in germ cells. Mos is a regulator of meiotic maturation, and is required for the initiation and progression of oocyte meiotic maturation that leads to the production of unfertilized eggs. Mos is also a component of cytostatic factor, an activity that is believed to arrest oocyte maturation at meiotic metaphase II. There is evidence showing that the Mos protein is associated with tubulin in unfertilized eggs and transformed cells, raising the possibility that it is involved in the microtubular reorganization that occurs during M-phase. Inappropriate expression of its M-phase activity during interphase of the cell cycle may be responsible for its transforming activity.


Assuntos
Ciclo Celular/genética , Transformação Celular Neoplásica/genética , Proteínas Oncogênicas v-mos/fisiologia , Proteínas Proto-Oncogênicas c-mos/fisiologia , Proto-Oncogenes , Proteínas Quinases Dependentes de Cálcio-Calmodulina , Transformação Celular Neoplásica/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Fator Promotor de Maturação/fisiologia , Meiose , Vírus do Sarcoma Murino de Moloney/genética , Vírus do Sarcoma Murino de Moloney/fisiologia , Proteínas Oncogênicas v-mos/genética , Oócitos/metabolismo , Fenótipo , Proteínas Quinases/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-mos/genética , Fuso Acromático/metabolismo
12.
Biomed Res Int ; 2018: 3693602, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29682539

RESUMO

Recent studies showed the modulatory effect of kisspeptin (KP) on calcium waves through the cell membrane and inside the cell. Spermatozoon can induce similar ooplasmic calcium oscillations at fertilization to trigger meiosis II. Here, we evaluated the effect of KP supplementation with 6-dimethylaminopurine (6-DMAP) for 4 h on embryonic development after oocyte activation with single electric pulse, 5 µM ionomycin, or 8% ethanol. Compared to control nonsupplemented groups, KP significantly improved embryo developmental competence electric- and ethanol-activated oocytes in terms of cleavage (75.3% and 58.6% versus 64% and 48%, respectively, p < 0.05) and blastocyst development (31.3% and 10% versus 19.3% and 4%, respectively, p < 0.05). MOS expression was increased in electrically activated oocytes in presence of KP while it significantly reduced CCNB1 expression. In ionomycin treated group, both MOS and CCNB1 showed significant increase with no difference between KP and control groups. In ethanol-treated group, KP significantly reduced CCNB1 but no effect was observed on MOS expression. The early alterations in MOS and CCNB1 mRNA transcripts caused by KP may explain the significant differences in the developmental competence between the experimental groups. Kisspeptin supplementation may be adopted in protocols for porcine oocyte activation through electric current and ethanol to improve embryonic developmental competence.


Assuntos
Kisspeptinas/metabolismo , Oócitos/metabolismo , Partenogênese/fisiologia , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Blastocisto/efeitos dos fármacos , Blastocisto/metabolismo , Ciclina B1/metabolismo , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/fisiologia , Ionomicina/farmacologia , Proteínas Oncogênicas v-mos/metabolismo , Oócitos/efeitos dos fármacos , Partenogênese/efeitos dos fármacos , RNA Mensageiro/metabolismo , Suínos
13.
FEBS Lett ; 581(9): 1735-41, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17418138

RESUMO

Src-homology (SH3) domain belongs to a class of ubiquitous modular protein domains found in nature. SH3 domains have a conserved surface that recognises proline-rich peptides in ligand proteins, but additional contacts also contribute to binding. Using the SH3 domain of hematopoietic cell kinase as a test case, we show that SH3 binding properties can be profoundly altered by modifications within a hexapeptide sequence in the RT-loop region that is not involved in recognition of currently known consensus SH3 target peptides. These results highlight the role of non-conserved regions in SH3 target selection, and introduce a strategy that may be generally feasible for generating artificial SH3 domains with desired ligand binding properties.


Assuntos
Engenharia de Proteínas/métodos , Domínios de Homologia de src , Proteínas ADAM/química , Proteínas ADAM/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Complexo CD3/química , Complexo CD3/metabolismo , Sequência Conservada , Produtos do Gene nef/química , Produtos do Gene nef/metabolismo , Imunoensaio/métodos , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Modelos Biológicos , Modelos Moleculares , Proteínas Oncogênicas v-mos/química , Proteínas Oncogênicas v-mos/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Secundária de Proteína , Transporte Proteico , Proteína SOS1/química , Proteína SOS1/metabolismo , Quinases Ativadas por p21
14.
Mol Cell Biol ; 16(3): 800-9, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8622681

RESUMO

We investigated the effect of cyclic AMP-dependent protein kinase (PKA ) on v-Mos kinase activity. Increase in PKA activity in vivo brought about either by forskolin treatment or by overexpression of PKA catalytic subunit resulted in a significant inhibition of v-Mos kinase activity. The purified PKA catalytic subunit was able to phosphorylate recombinant p37v-mos in vitro, suggesting that the mechanism of in vivo inhibition of v-Mos kinase involves direct phosphorylation by PKA. Combined tryptic phosphopeptide two-dimensional mapping analysis and in vitro mutagenesis studies indicated that Ser-56 is the major in vivo phosphorylation site on v-Mos. In vivo phosphorylation at Ser-56 correlated with slower migration of the v-Mos protein during sodium dodecyl sulfate-polyacrylamide gel electrophoresis. However, even though Ser-56 was phosphorylated by PKA, this phosphorylation was not involved in the inhibition of v-Mos kinase. The alanine-for-serine substitution at residue 56 did not affect the ability of v-Mos to autophosphorylate in vitro or, more importantly, to activate MEK1 in transformed NIH 3T3 cells. We identified Ser-263 phosphorylation, the Ala-263 mutant of v-Mos was not inhibited by forskolin treatment. From our results, we propose that the known inhibitory role of PKA in the initiation of oocyte maturation in mice could be explained at least in part by its inhibition of Mos kinase.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Oncogênicas v-mos/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Camundongos , Dados de Sequência Molecular , Mutação , Proteínas Oncogênicas v-mos/antagonistas & inibidores , Proteínas Oncogênicas v-mos/genética , Fosforilação , Serina/metabolismo
15.
Mol Cell Biol ; 8(11): 4685-91, 1988 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-2974919

RESUMO

We transfected Chinese hamster ovary (CHO) cells with a cloned v-mos gene (pHT25). The mos family of oncogenes has previously been shown to have serine-threonine kinase activity. This kinase activity may be required for oncogenic transformation, although its exact biological role is unknown. We found that the transfected cells had an altered morphology, a slower doubling time, and an apparent increase in the amount of a 25-kilodalton (kDa) phosphoprotein that appeared to be of low abundance. Transfection of CHO cells with a cloned temperature-sensitive mos gene (ts159) led to isolation of a cell line that showed the presence of the 25-kDa phosphoprotein at the permissive but not at the nonpermissive temperature, suggesting a direct relationship between mos activity and the presence of this phosphoprotein. The characteristics of altered morphology and depressed growth rate were reminiscent of changes seen after the activation of the cyclic AMP-dependent protein kinase (PKA) in CHO cells. However, PKA activation did not stimulate phosphorylation of this 25-kDa protein, nor was there a change in total PKA activity in these cells. We suggest that the increased presence of the 25-kDa phosphoprotein is a consequence of the v-mos transfection and that it may be involved in the change of morphology and growth rate seen in the CHO cells. Phosphorylation of this protein may be a useful marker of mos and have some functional importance in the transformation of cells by the v-mos oncogene.


Assuntos
Oncogenes , Fosfoproteínas/metabolismo , Proteínas dos Retroviridae/metabolismo , Transfecção , Animais , Linhagem Celular Transformada , Proteínas Oncogênicas v-mos , Fosforilação , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas dos Retroviridae/genética
16.
Mol Cell Biol ; 9(9): 4087-90, 1989 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2528689

RESUMO

We investigated the importance of specific serine residues for autophosphorylation and transformation by serine-threonine protein kinase p37mos. When either serine 326 or 358 was replaced with alanine, the resulting mutant protein retained the ability to transform NIH 3T3 cells but failed to autophosphorylate in vitro. These studies represent the first functional uncoupling of these two activities for p37mos.


Assuntos
Proteínas Quinases/genética , Proteínas dos Retroviridae/genética , Animais , Transformação Celular Viral , Técnicas In Vitro , Vírus do Sarcoma Murino de Moloney/genética , Mutação , Proteínas Oncogênicas v-mos , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas dos Retroviridae/metabolismo , Transformação Genética
17.
Reprod Toxicol ; 23(1): 20-31, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17055699

RESUMO

The widely used hormonal herbicide, 2,4-dichlorophenoxyacetic acid, blocks meiotic maturation in vitro and is thus a potential environmental endocrine disruptor with early reproductive effects. To test whether maturation inhibition was dependent on protein kinase A, an endogenous maturation inhibitor, oocytes were microinjected with PKI, a specific PKA inhibitor, and exposed to 2,4-D. Oocytes failed to mature, suggesting that 2,4-D is not dependent on PKA activity and likely acts on a downstream target, such as Mos. De novo synthesis of Mos, which is triggered by mRNA poly(A) elongation, was examined. Oocytes were microinjected with radiolabelled in vitro transcripts of Mos RNA and exposed to progesterone and 2,4-D. RNA analysis showed progesterone-induced polyadenylation as expected but none with 2,4-D. 2,4-D-activated MAPK was determined to be cytoplasmic in localization studies but poorly induced Rsk2 phosphorylation and activation. In addition to inhibition of the G2/M transition, 2,4-D caused abrupt reduction of H1 kinase activity in MII phase oocytes. Attempts to rescue maturation in oocytes transiently exposed to 2,4-D failed, suggesting that 2,4-D induces irreversible dysfunction of the meiotic signaling mechanism.


Assuntos
Ácido 2,4-Diclorofenoxiacético/toxicidade , Herbicidas/toxicidade , Meiose/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Xenopus laevis , Animais , Ciclo Celular/efeitos dos fármacos , Quimioterapia Combinada , Feminino , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Meiose/fisiologia , Proteínas Oncogênicas v-mos/biossíntese , Proteínas Oncogênicas v-mos/genética , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Progesterona/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , RNA Mensageiro/metabolismo , RNA Mensageiro/farmacologia
18.
Theriogenology ; 67(3): 475-85, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17027076

RESUMO

In matured rat oocytes, spontaneous activation from the metaphase-II (MII) stage occurred after collection from the oviducts. It is well known that the mitogen-activated protein kinase (MAPK) pathway and p34(cdc2) kinase play an important role in the arrest at MII in other species. However, there is no information about the difference in these factors among strains of rats. In the present study, in spontaneously activated oocytes from the Wistar rat, the Mos protein level and the activity of MAPK kinase (MEK)/MAPK were decreased at 120 min (13.8, 25.7, and 19.3, respectively, P<0.05), whereas Sprague-Dawley (SD) oocytes, which were not spontaneously activated, had a high level of Mos protein and MEK/MAPK activity (75.9, 76.2, and 87.9, respectively, P<0.05). Phosphorylation of MAPK in the SD oocytes was significantly suppressed by MEK inhibitor, U0126 at 60 min; this treatment decreased p34(cdc2) kinase activity via cyclin B1 degradation in a time-dependent manner. The treatment with proteasome inhibitor, MG132 or Ca2+-chelator, BAPTA-AM, overcame the spontaneous degradation of both Mos and cyclin B1 in a dose-dependent manner in Wistar oocytes. More than 90% of Wistar oocytes treated with BAPTA-AM were arrested at MII until 120 min. In conclusion, SD oocytes carrying Mos/MEK/MAPK, maintained a high activity of p34(cdc2) kinase by stabilizing cyclin B1, thus involved in their meiotic arrest. In contrast, Wistar oocytes had a relatively low cytostatic factor activity; rapid decrease of Mos/MEK/MAPK failed to stabilize both cyclin B1 and Mos, and these oocytes were likely to spontaneously activate.


Assuntos
Ciclina B/metabolismo , Proteínas Oncogênicas v-mos/metabolismo , Oócitos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Cálcio/metabolismo , Proliferação de Células , Ciclina B1 , Quinases Ciclina-Dependentes/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Feminino , Leupeptinas/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Oócitos/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Fatores de Tempo , Quinase Ativadora de Quinase Dependente de Ciclina
19.
J Leukoc Biol ; 78(1): 220-30, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15809290

RESUMO

We have shown previously that ulcerogenic (type I) strains of Helicobacter pylori (Hp) retard their entry into macrophages. However, the signaling pathways that regulate Hp phagocytosis are largely undefined. We show here that Hp strongly activated class IA phosphoinositide3-kinases (PI3Ks) in macrophages, coincident with phagocytosis, and endogenous p85 and active protein kinase Balpha accumulated on forming phagosomes. PI3K inhibitors, wortmannin and LY294002, inhibited phagocytosis of Hp in a dose-dependent manner, and blockade of engulfment correlated directly with loss of 3'-phosphoinositides in the membrane subjacent to attached bacteria. During uptake of large immunoglobulin G (IgG)-coated particles, PI3Ks regulate pseudopod extension and phagosome closure. In marked contrast, we show here that 3'-phosphoinositides regulated actin polymerization at sites of Hp uptake. Moreover, Hp and IgG beads activated distinct PI3K isoforms. Phagosomes containing IgG-coated particles accumulated 3'-phosphatase and tensin homologue deleted on chromosome 10 and Src homology 2 domain-containing inositol 5'-phosphatase, yet Hp phagosomes did not. Finally, rapid uptake of IgG-opsonized Hp or a less-virulent type II Hp was PI3K-independent. We conclude that Hp and IgG beads are ingested by distinct mechanisms and that PI3Ks regulate the actin cytoskeleton during slow phagocytosis of ulcerogenic Hp.


Assuntos
Actinas/metabolismo , Infecções por Helicobacter/enzimologia , Helicobacter pylori/fisiologia , Macrófagos/metabolismo , Fagocitose/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Membrana Celular/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Proteínas Oncogênicas v-mos/metabolismo , Fagocitose/efeitos dos fármacos , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilinositóis/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Monoéster Fosfórico Hidrolases/metabolismo , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo
20.
Cancer Res ; 51(3): 893-6, 1991 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-1824827

RESUMO

Metallothionein has been implicated in resistance to anticancer drugs. We examined whether transient induction of metallothionein by dexamethasone causes resistance to cis-diamminedichloroplatinum(II) (cis-DDP) in malignant and nonmalignant cells. Normal rat kidney cells (6m2) were infected with a modified v-mos oncogene construct in which expression of v-mos and consequently transformation was temperature-sensitive occurring at the permissive temperature of less than 33 degrees C and not at the nonpermissive temperature of 37 degrees C. Temperature-sensitive oncogenic transformation by v-mos attenuated induction of metallothionein by dexamethasone. No induction of metallothionein was observed in a revertant 6m2 cell line (54-5A4), which expressed v-mos and was transformed at 37 degrees C. Only nontransformed 6m2 cells displayed resistance to cis-DDP after dexamethasone pretreatment for 24 h. Dexamethasone pretreatment did not cause marked resistance to doxorubicin or melphalan in nontransformed 6m2 cells. When 6m2 cells (37 degrees C) were pretreated with dexamethasone (0.5 microM) for 24 h and then incubated in dexamethasone-free medium for 24 h, both metallothionein levels and resistance to cis-DDP decreased significantly. Thus, transient resistance to cis-DDP can be produced by a nonmetal inducer of metallothionein in nontransformed cells. Glucocorticoid-induced protection is suppressed in cells expressing v-mos and this might form the basis of future strategies to improve the therapeutic index of cis-DDP.


Assuntos
Transformação Celular Neoplásica/genética , Dexametasona/antagonistas & inibidores , Rim/metabolismo , Metalotioneína/biossíntese , Compostos Organoplatínicos/metabolismo , Proteínas Oncogênicas de Retroviridae/genética , Animais , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/metabolismo , Resistência a Medicamentos , Rim/efeitos dos fármacos , Proteínas Oncogênicas v-mos , Compostos Organoplatínicos/antagonistas & inibidores , Ratos , Proteínas Oncogênicas de Retroviridae/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA