Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
2.
FASEB J ; 35(11): e21959, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34605572

RESUMO

The proliferation and migration of vascular smooth muscle cells (VSMCs) are essential events in venous neointimal hyperplasia (VNH), a culprit of arteriovenous fistula (AVF) malfunction. Mitotic arrest-deficient protein 2B (MAD2B) is a critical regulator of cell proliferation and differentiation in many scenarios. To address the role of MAD2B in VSMCs proliferation and migration during VNH, AVFs from patients with end-stage renal disease (ESRD) and chronic kidney disease (CKD) mice were used to evaluate MAD2B expression. In cultured VSMCs treated with platelet-derived growth factor-BB (PDGF-BB), the effect of MAD2B on VSMCs proliferation and migration was detected by cell counting kit-8 (CCK8) assay, immunofluorescence, wound-healing scratch and transwell assays. Besides, we exploited different small interfering RNAs (siRNAs) to explore the potential mechanisms in the issue. Furthermore, rapamycin was applied to reveal whether MAD2B-associated pathways were involved in its inhibitory effect on VSMCs proliferation and migration. Accordingly, we found that MAD2B expression was enhanced in AVFs from patients with ESRD, CKD mice and VSMCs stimulated by PDGF-BB. Meanwhile, inhibition of MAD2B alleviated VSMCs proliferation and migration while the number of ski-related novel gene (SnoN)-positive VSMCs was also increased in vivo and in vitro. Moreover, gene deletion of MAD2B decreased the level of SnoN protein in PDGF-BB-stimulated VSMCs. Furthermore, rapamycin suppressed the increased expressions of MAD2B and SnoN induced by PDGF-BB. Thus, our study demonstrates that inhibition of MAD2B suppresses the proliferation and migration of VSMCs during VNH via reducing SnoN expression. Moreover, rapamycin exerts an inhibitory effect on intimal hyperplasia, possibly via the MAD2B-SnoN axis.


Assuntos
Hiperplasia , Falência Renal Crônica/metabolismo , Proteínas Mad2/fisiologia , Neointima , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Humanos , Hiperplasia/metabolismo , Hiperplasia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neointima/metabolismo , Neointima/patologia
3.
J Obstet Gynaecol Res ; 45(12): 2407-2418, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31523901

RESUMO

AIM: To explore the involvement of Mad2 and BubR1 in cervical carcinogenesis. METHODS: The expressions of Mad2 and BubR1 in tissues of high-grade squamous intraepithelial lesions (HSIL), low-grade squamous intraepithelial lesions (LSIL) and chronic cervicitis were analyzed immunohistochemistrily and compared with those of p16INK4A . PEGFP-Mad2 and pEGFP-BubR1 were transfected into SiHa cells to overexpress Mad2 and BubR1 and Si-RNAs to knockdown. Cell viability was measured by cell counting kit-8 (CCK-8) assay. Migration and invasion capabilities were detected by Transwell. Propidium iodide staining with flow cytometry was used for cell cycle analysis and apoptosis was detected using Annexin V/7-AAD staining after nocodazole treatment. RESULTS: The expression of Mad2 was significantly lower in HSIL than those in chronic cervicitis and LSIL, however, the expression of BubR1 showed no significant differences. To detect HSIL in cervical lesions, Mad2 had a sensitivity of 88.44% and a specificity of 87.23%, Mad2 was less sensitive and more specific than p16INK4a . In SiHa cells, knockdown of Mad2 and BubR1 increased cell growth, reinforced invasion capacity and migration potency, inhibited apoptosis and decreased G2-phase distribution after nocodazole treatment. Oppositely, the overexpression strategies made cells show decreased malignant behaviors, raised apoptosis and increased G2-phase distribution. CONCLUSION: Mad2 negativity was specific to identify HSIL immunohistochemistrily. Downregulation of Mad2 and BubR1 increase the malignant behavior and nocodazole resistance of SiHa cells via causing spindle assembly checkpoint defect. This mechanism may contribute to cervical carcinogenesis and resistance to microtubule-targeting drugs.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas Mad2/fisiologia , Nocodazol/uso terapêutico , Proteínas Serina-Treonina Quinases/fisiologia , Neoplasias do Colo do Útero/tratamento farmacológico , Adulto , Apoptose/efeitos dos fármacos , Células Cultivadas , Colo do Útero/química , Inibidor p16 de Quinase Dependente de Ciclina/análise , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Proteínas Mad2/análise , Proteínas Mad2/antagonistas & inibidores , Pessoa de Meia-Idade , Invasividade Neoplásica , Proteínas Serina-Treonina Quinases/análise , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Neoplasias do Colo do Útero/patologia
4.
J Cell Physiol ; 231(2): 336-44, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25752701

RESUMO

Weakening the Spindle Assembly Checkpoint by reduced expression of its components induces chromosome instability and aneuploidy that are hallmarks of cancer cells. The tumor suppressor p14(ARF) is overexpressed in response to oncogenic stimuli to stabilize p53 halting cell progression. Previously, we found that lack or reduced expression of p14(ARF) is involved in the maintenance of aneuploid cells in primary human cells, suggesting that it could be part of a pathway controlling their proliferation. To investigate this aspect further, p14(ARF) was ectopically expressed in HCT116 cells after depletion of the Spindle Assembly Checkpoint MAD2 protein that was used as a trigger for aneuploidy. p14(ARF) Re-expression reduced the number of aneuploid cells in MAD2 post-transcriptionally silenced cells. Also aberrant mitoses, frequently displayed in MAD2-depleted cells, were decreased when p14(ARF) was expressed at the same time. In addition, p14(ARF) ectopic expression in MAD2-depleted cells induced apoptosis associated with increased p53 protein levels. Conversely, p14(ARF) ectopic expression did not induce apoptosis in HCT116 p53KO cells. Collectively, our results suggest that the tumor suppressor p14(ARF) may have an important role in counteracting proliferation of aneuploid cells by activating p53-dependent apoptosis.


Assuntos
Aneuploidia , Apoptose/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Apoptose/genética , Proliferação de Células/genética , Proliferação de Células/fisiologia , Técnicas de Inativação de Genes , Células HCT116 , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/genética , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Proteínas Mad2/genética , Proteínas Mad2/fisiologia , Mitose/genética , Mitose/fisiologia , Interferência de RNA , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
5.
Mol Pharm ; 11(10): 3515-27, 2014 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-25256346

RESUMO

RNA interference has emerged as a powerful strategy in cancer therapy because it allows silencing of specific genes associated with tumor progression and resistance. Mad2 is an essential mitotic checkpoint component required for accurate chromosome segregation during mitosis, and its complete abolition leads to cell death. We have developed an epidermal growth factor receptor (EGFR)-targeted chitosan system for silencing the Mad2 gene as a strategy to efficiently induce cell death in EGFR overexpressing human A549 non-small cell lung cancer cells. Control and EGFR-targeted chitosan nanoparticles loaded with small interfering RNAs (siRNAs) against Mad2 were formulated and characterized for size, charge, morphology, and encapsulation efficiency. Qualitative and quantitative intracellular uptake studies by confocal imaging and flow cytometry, respectively, showed time-dependent enhanced and selective intracellular internalization of EGFR-targeted nanoparticles compared to nontargeted system. Targeted nanoparticles showed nearly complete depletion of Mad2 expression in A549 cells contrasting with the partial depletion in the nontargeted system. Accordingly, Mad2-silencing-induced apoptotic cell death was confirmed by cytotoxicity assay and flow cytometry. Our results demonstrate that EGFR-targeted chitosan loaded with Mad2 siRNAs is a potent delivery system for selective killing of cancer cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Quitosana/química , Receptores ErbB/metabolismo , Proteínas Mad2/genética , Nanopartículas/química , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Citometria de Fluxo , Inativação Gênica/fisiologia , Humanos , Proteínas Mad2/fisiologia
6.
Sci Rep ; 11(1): 68, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33420244

RESUMO

Chromosomal instability (CIN) is a hallmark of cancer, leading to aneuploid cells. To study the role that CIN plays in tumor evolution, several mouse models have been engineered over the last 2 decades. These models have unequivocally shown that systemic high-grade CIN is embryonic lethal. We and others have previously shown that embryonic lethality can be circumvented by provoking CIN in a tissue-specific fashion. In this study, we provoke systemic high-grade CIN in adult mice as an alternative to circumvent embryonic lethality. For this, we disrupt the spindle assembly checkpoint (SAC) by alleviating Mad2 or truncating Mps1, both essential genes for SAC functioning, with or without p53 inactivation. We find that disruption of the SAC leads to rapid villous atrophy, atypia and apoptosis of the epithelia of the jejunum and ileum, substantial weight loss, and death within 2-3 weeks after the start of the CIN insult. Despite this severe intestinal phenotype, most other tissues are unaffected, except for minor abnormalities in spleen, presumably due to the lower proliferation rate in these tissues. We conclude that high-grade CIN in vivo in adult mice is most toxic to the high cell turnover intestinal epithelia.


Assuntos
Intestinos/patologia , Proteínas Mad2/fisiologia , Animais , Atrofia , Pontos de Checagem da Fase M do Ciclo Celular , Proteínas Mad2/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
Cancer Cell ; 32(4): 444-459.e7, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-29017056

RESUMO

Proper organization of the mitotic spindle is key to genetic stability, but molecular components of inter-microtubule bridges that crosslink kinetochore fibers (K-fibers) are still largely unknown. Here we identify a kinase-independent function of class II phosphoinositide 3-OH kinase α (PI3K-C2α) acting as limiting scaffold protein organizing clathrin and TACC3 complex crosslinking K-fibers. Downregulation of PI3K-C2α causes spindle alterations, delayed anaphase onset, and aneuploidy, indicating that PI3K-C2α expression is required for genomic stability. Reduced abundance of PI3K-C2α in breast cancer models initially impairs tumor growth but later leads to the convergent evolution of fast-growing clones with mitotic checkpoint defects. As a consequence of altered spindle, loss of PI3K-C2α increases sensitivity to taxane-based therapy in pre-clinical models and in neoadjuvant settings.


Assuntos
Neoplasias da Mama/patologia , Instabilidade Genômica , Fosfatidilinositol 3-Quinases/fisiologia , Fuso Acromático/fisiologia , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Proteínas de Ciclo Celular/fisiologia , Proliferação de Células , Humanos , Células MCF-7 , Proteínas Mad2/fisiologia , Camundongos , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas Nucleares/fisiologia , Taxoides/uso terapêutico
8.
Trends Cell Biol ; 25(1): 21-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25220181

RESUMO

Error-free chromosome segregation relies on stable connections between kinetochores and spindle microtubules. The spindle assembly checkpoint (SAC) monitors such connections and relays their absence to the cell cycle machinery to delay cell division. The molecular network at kinetochores that is responsible for microtubule binding is integrated with the core components of the SAC signaling system. Molecular-mechanistic understanding of how the SAC is coupled to the kinetochore-microtubule interface has advanced significantly in recent years. The latest insights not only provide a striking view of the dynamics and regulation of SAC signaling events at the outer kinetochore but also create a framework for understanding how that signaling may be terminated when kinetochores and microtubules connect.


Assuntos
Cinetocoros/fisiologia , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Microtúbulos/fisiologia , Transdução de Sinais/fisiologia , Animais , Proteínas de Ciclo Celular/fisiologia , Segregação de Cromossomos/fisiologia , Humanos , Proteínas Mad2/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia
9.
Mol Biol Cell ; 26(6): 1129-40, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25631818

RESUMO

Ploidy variation is found in contexts as diverse as solid tumors, drug resistance in fungal infection, and normal development. Altering chromosome or genome copy number supports adaptation to fluctuating environments but is also associated with fitness defects attributed to protein imbalances. Both aneuploidy and polyploidy can arise from multinucleate states after failed cytokinesis or cell fusion. The consequences of ploidy variation in syncytia are difficult to predict because protein imbalances are theoretically buffered by a common cytoplasm. We examined ploidy in a naturally multinucleate fungus, Ashbya gossypii. Using integrated lac operator arrays, we found that chromosome number varies substantially among nuclei sharing a common cytoplasm. Populations of nuclei range from 1N to >4N, with different polyploidies in the same cell and low levels of aneuploidy. The degree of ploidy variation increases as cells age. In response to cellular stress, polyploid nuclei diminish and haploid nuclei predominate. These data suggest that mixed ploidy is tolerated in these syncytia; however, there may be costs associated with variation as stress homogenizes the genome content of nuclei. Furthermore, the results suggest that sharing of gene products is limited, and thus there is incomplete buffering of ploidy variation despite a common cytosol.


Assuntos
Ascomicetos/genética , Ascomicetos/citologia , Ascomicetos/crescimento & desenvolvimento , Núcleo Celular/genética , Segregação de Cromossomos , Cromossomos Fúngicos/genética , DNA Fúngico/genética , Proteínas Fúngicas/fisiologia , Dosagem de Genes , Genes Fúngicos , Proteínas Mad2/fisiologia , Poliploidia , Estresse Fisiológico
10.
J Cell Biol ; 211(4): 745-55, 2015 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-26598612

RESUMO

The HORMA domain is a multifunctional protein-protein interaction module found in diverse eukaryotic signaling pathways including the spindle assembly checkpoint, numerous DNA recombination/repair pathways, and the initiation of autophagy. In all of these pathways, HORMA domain proteins occupy key signaling junctures and function through the controlled assembly and disassembly of signaling complexes using a stereotypical "safety belt" peptide interaction mechanism. A recent explosion of structural and functional work has shed new light on these proteins, illustrating how strikingly similar structural mechanisms give rise to radically different functional outcomes in each family of HORMA domain proteins.


Assuntos
Proteínas Mad2/fisiologia , Transdução de Sinais , Animais , Autofagia , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/fisiologia , DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/fisiologia , Humanos , Pontos de Checagem da Fase M do Ciclo Celular , Proteínas Mad2/química , Domínios e Motivos de Interação entre Proteínas , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/fisiologia , Homologia Estrutural de Proteína
11.
Mol Biol Cell ; 25(18): 2774-87, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25057014

RESUMO

The binucleate pathogen Giardia intestinalis is a highly divergent eukaryote with a semiopen mitosis, lacking an anaphase-promoting complex/cyclosome (APC/C) and many of the mitotic checkpoint complex (MCC) proteins. However, Giardia has some MCC components (Bub3, Mad2, and Mps1) and proteins from the cohesin system (Smc1 and Smc3). Mad2 localizes to the cytoplasm, but Bub3 and Mps1 are either located on chromosomes or in the cytoplasm, depending on the cell cycle stage. Depletion of Bub3, Mad2, or Mps1 resulted in a lowered mitotic index, errors in chromosome segregation (including lagging chromosomes), and abnormalities in spindle morphology. During interphase, MCC knockdown cells have an abnormal number of nuclei, either one nucleus usually on the left-hand side of the cell or two nuclei with one mislocalized. These results suggest that the minimal set of MCC proteins in Giardia play a major role in regulating many aspects of mitosis, including chromosome segregation, coordination of mitosis between the two nuclei, and subsequent nuclear positioning. The critical importance of MCC proteins in an organism that lacks their canonical target, the APC/C, suggests a broader role for these proteins and hints at new pathways to be discovered.


Assuntos
Segregação de Cromossomos , Giardia lamblia/enzimologia , Proteínas Mad2/fisiologia , Mitose , Proteínas Tirosina Quinases/fisiologia , Proteínas de Protozoários/fisiologia , Núcleo Celular/metabolismo , DNA de Protozoário/genética , DNA de Protozoário/metabolismo , Técnicas de Silenciamento de Genes , Transporte Proteico , Polos do Fuso/metabolismo
12.
Curr Biol ; 24(22): 2687-92, 2014 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-25447996

RESUMO

Mitotic arrest deficient 1 (Mad1) plays a well-characterized role in the major cell-cycle checkpoint that regulates chromosome segregation during mitosis, the mitotic checkpoint (also known as the spindle assembly checkpoint). During mitosis, Mad1 recruits Mad2 to unattached kinetochores, where Mad2 is converted into an inhibitor of the anaphase-promoting complex/cyclosome bound to its specificity factor, Cdc20. During interphase, Mad1 remains tightly bound to Mad2, and both proteins localize to the nucleus and nuclear pores, where they interact with Tpr (translocated promoter region). Recently, it has been shown that interaction with Tpr stabilizes both proteins and that Mad1 binding to Tpr permits Mad2 to associate with Cdc20. However, interphase functions of Mad1 that do not directly affect the mitotic checkpoint have remained largely undefined. Here we identify a previously unrecognized interphase distribution of Mad1 at the Golgi apparatus. Mad1 colocalizes with multiple Golgi markers and cosediments with Golgi membranes. Although Mad1 has previously been thought to constitutively bind Mad2, Golgi-associated Mad1 is Mad2 independent. Depletion of Mad1 impairs secretion of α5 integrin and results in defects in cellular attachment, adhesion, and FAK activation. Additionally, reduction of Mad1 impedes cell motility, while its overexpression accelerates directed cell migration. These results reveal an unexpected role for a mitotic checkpoint protein in secretion, adhesion, and motility. More generally, they demonstrate that, in addition to generating aneuploidy, manipulation of mitotic checkpoint genes can have unexpected interphase effects that influence tumor phenotypes.


Assuntos
Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/fisiologia , Movimento Celular/genética , Complexo de Golgi/metabolismo , Integrinas/metabolismo , Proteínas Nucleares/fisiologia , Proteínas de Ciclo Celular/análise , Proteínas de Ciclo Celular/metabolismo , Citoplasma/metabolismo , Células HeLa , Humanos , Proteínas Mad2/genética , Proteínas Mad2/metabolismo , Proteínas Mad2/fisiologia , Proteínas Nucleares/análise , Proteínas Nucleares/metabolismo , Transdução de Sinais
13.
J Cell Biol ; 204(5): 647-57, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24567362

RESUMO

Recruitment of Mad1-Mad2 complexes to unattached kinetochores is a central event in spindle checkpoint signaling. Despite its importance, the mechanism that recruits Mad1-Mad2 to kinetochores is unclear. In this paper, we show that MAD-1 interacts with BUB-1 in Caenorhabditis elegans. Mutagenesis identified specific residues in a segment of the MAD-1 coiled coil that mediate the BUB-1 interaction. In addition to unattached kinetochores, MAD-1 localized between separating meiotic chromosomes and to the nuclear periphery. Mutations in the MAD-1 coiled coil that selectively disrupt interaction with BUB-1 eliminated MAD-1 localization to unattached kinetochores and between meiotic chromosomes, both of which require BUB-1, and abrogated checkpoint signaling. The identified MAD-1 coiled-coil segment interacted with a C-terminal region of BUB-1 that contains its kinase domain, and mutations in this region prevented MAD-1 kinetochore targeting independently of kinase activity. These results delineate an interaction between BUB-1 and MAD-1 that targets MAD-1-MAD-2 complexes to kinetochores and is essential for spindle checkpoint signaling.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/citologia , Proteínas de Ciclo Celular/fisiologia , Cinetocoros/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular , Proteínas Nucleares/fisiologia , Anáfase/fisiologia , Animais , Sítios de Ligação , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromossomos/metabolismo , Cinetocoros/fisiologia , Proteínas Mad2/genética , Proteínas Mad2/metabolismo , Proteínas Mad2/fisiologia , Meiose/fisiologia , Modelos Biológicos , Mutagênese , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Estrutura Terciária de Proteína , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
14.
J Cell Biol ; 203(6): 883-93, 2013 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-24344181

RESUMO

Tpr is a conserved nuclear pore complex (NPC) protein implicated in the spindle assembly checkpoint (SAC) by an unknown mechanism. Here, we show that Tpr is required for normal SAC response by stabilizing Mad1 and Mad2 before mitosis. Tpr coimmunoprecipitated with Mad1 and Mad2 (hereafter designated as Tpr/Mad1/Mad2 or TM2 complex) during interphase and mitosis, and is required for Mad1­c-Mad2 recruitment to NPCs. Interestingly, Tpr was normally undetectable at kinetochores and dispensable for Mad1, but not for Mad2, kinetochore localization, which suggests that SAC robustness depends on Mad2 levels at kinetochores. Protein half-life measurements demonstrate that Tpr stabilizes Mad1 and Mad2, ensuring normal Mad1­c-Mad2 production in an mRNA- and kinetochore-independent manner. Overexpression of GFP-Mad2 restored normal SAC response and Mad2 kinetochore levels in Tpr-depleted cells. Mechanistically, we provide evidence that Tpr might spatially regulate SAC proteostasis through the SUMO-isopeptidases SENP1 and SENP2 at NPCs. Thus, Tpr is a kinetochore-independent, rate-limiting factor required to mount and sustain a robust SAC response.


Assuntos
Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/fisiologia , Proteínas Mad2/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Células HeLa , Humanos , Proteínas Mad2/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA