Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros













Base de dados
Intervalo de ano de publicação
1.
Eur J Neurosci ; 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38693604

RESUMO

The continually advancing landscape of neuroscientific and imaging research has broadened our comprehension of sex differences encoded in the human brain, expanding from the hypothalamus and sexual behaviour to encompass the entire brain, including its diverse lobes, structures, and functions. However, less is known about sex differences in the brains of neonates and infants, despite their relevance to various sex-linked diseases that develop early in life. In this review, we provide a synopsis of the literature evidence on sex differences in the brains of neonates and infants at the morphological, structural and network levels. We also briefly overview the present evidence on the sex bias in some brain disorders affecting infants and neonates.

2.
Mol Biol Rep ; 49(11): 10635-10652, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35716286

RESUMO

Medulloblastoma is the most common malignant brain tumor of childhood accounting for about 60% of all pediatric embryonal tumors. Despite improvements in the overall survival rate, this tumor still lacks an efficient, reliable, and less toxic therapeutic approach. Characterization of the molecular mechanisms involved in medulloblastoma initiation and progression is a crucial step for the development of effective therapies. Signal transducer and activator of transcription 3 is a convergence point for several signaling cascades that are implicated in medulloblastoma tumorigenesis. Accumulated evidence has revealed the pivotal role of signal transducer and activator of transcription 3 in medulloblastoma pathogenesis such as proliferation, survival, angiogenesis, and immunosuppression as well as maintenance, drug resistance, and recurrence. In this review, we focus on the role of signal transducer and activator of transcription 3 in medulloblastoma tumorigenesis and discuss the recent advances of signal transducer and activator of transcription 3 inhibition as a promising developed strategy for medulloblastoma therapy.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Humanos , Criança , Meduloblastoma/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/patologia , Carcinogênese
3.
Curr Treat Options Oncol ; 23(6): 864-886, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35412196

RESUMO

OPINION STATEMENT: Medulloblastoma (MB) is the most frequent pediatric brain tumor. Despite conventional therapy, MB patients have high mortality and morbidity rates mainly due to the incomplete understanding of the molecular and cellular processes involved in development of this cancer. Similar to other solid tumors, MB demonstrated high endothelial cell proliferation and angiogenic activity, wherein new blood vessels arise from the pre-existing vasculature, a process named angiogenesis. MB angiogenesis is considered a hallmark for MB development, progression, and metastasis emphasizing its potential target for antitumor therapy. However, angiogenesis is tightly regulated by a set of angiogenic factors making it a complex process to be targeted. Although agents targeting these factors and their receptors are early in development, the potential for their targeting may translate into improvement in the clinical care for MB patients. In this review, we focus on the most potent angiogenic factors and their corresponding receptors, highlighting their basic properties and expression in MB. We describe their contribution to MB tumorigenesis and angiogenesis and the potential therapeutic targeting of these factors.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Indutores da Angiogênese/uso terapêutico , Inibidores da Angiogênese/uso terapêutico , Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/etiologia , Criança , Humanos , Meduloblastoma/tratamento farmacológico , Meduloblastoma/etiologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo
4.
Curr Treat Options Oncol ; 22(9): 83, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34328587

RESUMO

OPINION STATEMENT: Medulloblastoma (MB) is the most common pediatric brain malignancy, with a 5-year overall survival (OS) rate of around 65%. The conventional MB treatment, comprising surgical resection followed by irradiation and adjuvant chemotherapy, often leads to impairment in normal body functions and poor quality of life, especially with the increased risk of recurrence and subsequent development of secondary malignancies. The development and progression of MB are facilitated by a variety of immune-evading mechanisms such as the secretion of immunosuppressive molecules, activation of immunosuppressive cells, inhibition of immune checkpoint molecules, impairment of adhesive molecules, downregulation of the major histocompatibility complex (MHC) molecules, protection against apoptosis, and activation of immunosuppressive pathways. Understanding the tumor-immune relationship in MB is crucial for effective development of immune-based therapeutic strategies. In this comprehensive review, we discuss the immunological aspect of the brain, focusing on the current knowledge tackling the mechanisms of MB immune suppression and evasion. We also highlight several key immunotherapeutic approaches developed to date for the treatment of MB.


Assuntos
Neoplasias Cerebelares/etiologia , Suscetibilidade a Doenças/imunologia , Tolerância Imunológica , Meduloblastoma/etiologia , Biomarcadores , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Cerebelares/diagnóstico , Neoplasias Cerebelares/epidemiologia , Neoplasias Cerebelares/terapia , Tomada de Decisão Clínica , Terapia Combinada/efeitos adversos , Terapia Combinada/métodos , Gerenciamento Clínico , Humanos , Hospedeiro Imunocomprometido , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Meduloblastoma/diagnóstico , Meduloblastoma/epidemiologia , Meduloblastoma/terapia , Especificidade de Órgãos/imunologia , Resultado do Tratamento , Microambiente Tumoral/imunologia
5.
Neoplasia ; 10(9): 996-1003, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18714401

RESUMO

The tyrosine kinase receptor c-kit and its ligand stem cell factor (SCF) have not been explored in prostate cancer (PC) bone metastasis. Herein, we found that three human PC cell lines and bone marrow stromal cells express a membrane-bound SCF isoform and release a soluble SCF. Bone marrow stromal cells revealed strong expression of c-kit, whereas PC cells showed very low levels of the receptor or did not express it all. Using an experimental model of PC bone metastasis, we found that intraosseous bone tumors formed by otherwise c-kit-negative PC3 cells strongly expressed c-kit, as demonstrated using immunohistochemical and Western blot analyses. Subcutaneous PC3 tumors were, however, c-kit-negative. Both bone and subcutaneous PC3 tumors were positive for SCF. Immunohistochemical analysis of human specimens revealed that the expression frequency of c-kit in epithelial cells was of 5% in benign prostatic hyperplasia, 14% in primary PC, and 40% in PC bone metastases, suggesting an overall trend of increased c-kit expression in clinical PC progression. Stem cell factor expression frequency was more than 80% in all the cases. Our data suggest that the bone microenvironment up-regulates c-kit expression on PC cells, favoring their intraosseous expansion.


Assuntos
Neoplasias Ósseas/genética , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas c-kit/biossíntese , Fator de Células-Tronco/metabolismo , Regulação para Cima , Animais , Neoplasias Ósseas/secundário , Técnicas de Cultura de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Ligantes , Masculino , Camundongos , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Neoplasia ; 10(5): 439-49, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18472961

RESUMO

A variety of proteases have been implicated in prostate cancer (PC) bone metastasis, but the individual contributions of these enzymes remain unclear. Urokinase-type plasminogen activator (uPA), a serine protease, can activate plasminogen and stimulate signaling events on binding its receptor uPAR. In the present study, we investigated the functional role of PC cell-associated uPA in intraosseous tumor growth and bone matrix degradation. Using a severe combined immunodeficient-human mouse model, we found that PC3 cells were the major source of uPA in the experimental bone tumor. Injection of uPA-silenced PC3 cells in bone xenografts resulted in significant reduction of bone tumor burdens and protection of trabecular bones from destruction. The suppressed tumor growth was associated with the level of uPA expression but not with its activity. An increase in the expression of PAI-1, the endogenous uPA inhibitor, was found during in vitro tumor-stromal interactions. Up-regulation of PAI-1 in bone stromal cells and preosteoclasts/osteoblasts was due to soluble factor(s) released by PC cells, and the enhanced PAI-1 expression in turn stimulated PC cell migration. Our results indicate that both tumor-derived uPA and tumor-stroma-induced PAI-1 play important roles in intraosseous metastatic PC growth through regulation of a uPA-uPAR-PAI-1 axis by autocrine/paracrine mechanisms.


Assuntos
Neoplasias Ósseas/metabolismo , Remodelação Óssea , Neoplasias da Próstata/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Neoplasias Ósseas/patologia , Movimento Celular , Proliferação de Células , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos SCID , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Neoplasias da Próstata/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/metabolismo , Células Estromais/patologia , Transplante Heterólogo
7.
Int J Cancer ; 122(11): 2482-90, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18324629

RESUMO

At the cellular level, the process of bone metastasis involves many steps. Circulating cancer cells enter the marrow, proliferate, induce neovascularization, and ultimately expand into a clinically detectable, often symptomatic, metastatic deposit. Although the initial establishment and later expansion of the metastatic deposit in bone require tumor cells to possess invasive capability, the exact proteases responsible for this phenotype are not well known. The objective of our study was to take an unbiased approach to determine which proteases were expressed and functional during the initial interactions between prostate cancer cells and bone marrow stromal (BMS) cells. We found that the combination of human prostate cancer PC3 and BMS cells stimulates the invasive ability of cancer cells through type I collagen. The use of inhibitors for each of the major protease families indicated that 1 or more MMPs was/were responsible for the BMS-induced invasion. Gene profiling and semiquantitative RT-PCR analysis revealed an increased expression of several MMP genes because of PC3/BMS cell interaction. However, only MMP-12 showed an increase in protein expression. Downregulation of MMP-12 expression in PC3 cells by siRNA inhibited the enhanced invasion induced by PC3/BMS cell interaction. In vivo, MMP-12 was found to be primarily expressed by prostate cancer cells growing in bone. Our data suggest that BMS cells induce MMP-12 expression in prostate cancer cells, which results in invasive cells capable of degradation of type I collagen.


Assuntos
Células da Medula Óssea/patologia , Colágeno Tipo I/metabolismo , Metaloproteinase 12 da Matriz/metabolismo , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Células Estromais/patologia , Western Blotting , Linhagem Celular Tumoral , Técnicas de Cocultura , Regulação para Baixo , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Queratinas/análise , Masculino , Metaloproteinases da Matriz/metabolismo , Invasividade Neoplásica , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Regulação para Cima
8.
BMC Cancer ; 7: 184, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17908302

RESUMO

BACKGROUND: Despite significant advancement in breast cancer therapy, there is a great need for a better understanding of the mechanisms involved in breast carcinogenesis and progression, as well as of the role of epigenetic contributions from stromal cells in mammary tumorigenesis. In this study, we isolated and characterized murine mammary tumor-derived epithelial and myofibroblast cell lines, and investigated the in vitro and in vivo effect of cellular soluble factors produced by the epithelial cell line on tumor cells. METHODS: Morphology, immunophenotype, cytogenetics, invasiveness, and tumorigenicity of epithelial (LM-234ep) and myofibroblast (LM-234mf) cell lines isolated from two murine mammary adenocarcinomas with common ancestor were studied. The in vitro effects of LM-234ep conditioned medium on proliferation, cell cycle distribution, and expression of cell cycle proteins, were investigated in LM-234mf cells, mouse melanoma cells (B16-F10), and human cervical adenocarcinoma cells (HeLa). The in vivo anti-tumor activity of LM-234ep conditioned media was evaluated in subcutaneous tumors formed in nude mice by B16-F10 and HeLa cells. RESULTS: LM-234ep cells were found to be cytokeratin positive and hipertriploid, whereas LM-234mf cells were alpha-smooth muscle actin positive and hypohexaploid. Chromosome aberrations were found in both cases. Only LM-234mf revealed to be invasive in vitro and to secrete active MMP-2, though neither of the cell types were able to produce progressing tumors. LM-234ep-derived factors were able to inhibit the in vitro growth of LM-234mf, B16-F10, and HeLa cells, inducing cell cycle arrest in G0/G1 phase. The administration of LM-234ep conditioned medium inhibited the growth of B16-F10 and HeLa tumors in nude mice. CONCLUSION: Our data suggest the existence of epithelial cell variants with tumor suppressive properties within mammary tumors. To our knowledge, this is the first report showing antiproliferative and antineoplastic activities induced by tumor-derived epithelial cells.


Assuntos
Adenocarcinoma/metabolismo , Proliferação de Células , Células Epiteliais/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Actinas/metabolismo , Adenocarcinoma/genética , Animais , Testes de Carcinogenicidade , Linhagem Celular Tumoral , Aberrações Cromossômicas , Meios de Cultivo Condicionados , Feminino , Gelatinases/metabolismo , Humanos , Queratinas/metabolismo , Neoplasias Mamárias Experimentais/genética , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Comunicação Parácrina , Neoplasias do Colo do Útero/metabolismo
9.
Urology ; 69(4): 795-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17445684

RESUMO

OBJECTIVES: To determine whether matrix metalloproteinase (MMP)-9 activation resulting from prostate cancer cell-bone interaction is dependent on the tumor cell type and/or the nature of the bone microenvironment. METHODS: In vitro co-cultures of human prostate cancer cells (PC3 and C4-2B) and mouse, human fetal, or human adult tissues were performed. In vivo the tumor cells were intratibially injected in SCID mice or intraosseously inoculated into fetal or adult bone xenografts in SCID mice. MMP-2 and MMP-9 expression and activation were determined by gelatin zymography in conditioned media obtained in vitro and in lysates derived from the in vivo studies at different time points. RESULTS: Activation of MMP-9 occurred when PC3 cells interacted with human adult or fetal bone, either in vitro or in vivo at early time points. With C4-2B cells, activation of MMP-9 only happened in the human adult bone microenvironment at early time points after intraosseous inoculation of tumor cells. No activation of MMP-9 occurred when PC3 or C4-2B cells interacted with mouse bone, either in vitro or in vivo. CONCLUSIONS: The results of our study have shown that the activation of MMP-9 when human prostate cancer cells interact with bone depends on the particular identity of the tumor cells and the type of bone tissue used. These findings have broad implications for experimental models attempting to define tumor-microenvironmental interactions in bone metastasis.


Assuntos
Osso e Ossos/citologia , Osso e Ossos/enzimologia , Metaloproteinase 9 da Matriz/fisiologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Animais , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos SCID
10.
Clin Exp Metastasis ; 23(7-8): 335-44, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17136575

RESUMO

Matrix metalloproteinases (MMPs) have been associated with initiation, progression and vascularization of a number of tumors. However, clinical trials using MMP inhibitors failed to meet expectations. Previously, we demonstrated the potential importance of MMP-9 activity in experimental prostate cancer bone tumor tissue. However, the particular roles of host- and tumor-derived MMP-9 remains to be defined. Herein, we examined the role of host MMP-9 in subcutaneous and intraosseous growth of the human androgen independent prostate cancer cell line PC3 in MMP-9 deficient mice. In the subcutaneous model, the tumor incidence in the control (RAG-1(ko/ko)) and experimental (RAG-1(ko/ko) /MMP-9(ko/ko)) group was 100%, with similar tumor growth kinetics and microvascular densities. In the intraosseous tumor model, the tumor incidence was higher in RAG-1(ko/ko) /MMP-9(ko/ko mice than in RAG-1(ko/ko) mice (67% and 39%, respectively), though no statistical differences were found. The intraosseous tumor areas were similar in both groups, and the number of tumor-associated osteoclasts did not differ significantly. However, the microvascular density of intraosseous tumors was higher in RAG-1(ko/ko) than in RAG-1(ko/ko)/MMP-9(ko/ko) mice, though no changes in tumor growth could be detected. In an in vitro assay, we found that bone marrow (BM) cells increased the invasiveness of PC3 cells, and that this enhancement was independent of MMP-9 expression by marrow cells. Our results with the RAG-1 model suggest that host-derived MMP-9 is neither necessary nor sufficient for subcutaneous or intraosseous PC3 tumor growth, osteoclastic response, or in vitro invasiveness of tumor cells.


Assuntos
Neoplasias Ósseas/secundário , Metaloproteinase 9 da Matriz/fisiologia , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/patologia , Animais , Movimento Celular , Proteínas de Homeodomínio/fisiologia , Humanos , Masculino , Camundongos , Invasividade Neoplásica
11.
Oncogene ; 24(19): 3166-76, 2005 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-15735693

RESUMO

Acquired resistance to tamoxifen (Tam) in breast cancer patients is a serious therapeutic problem. We have previously reported that protein kinase C-delta (PKC-delta) plays a major role in estrogen (E2)-mediated cell proliferation. To determine if PKC-delta is one of the major alternate signaling pathways that supports cell growth in the presence of Tam, we determined the levels of PKC isoforms in four different models of antiestrogen-resistant cells. Three out of four antiestrogen resistance cell lines (Tam/MCF-7, ICI/MCF-7 and HER-2/MCF-7) expressed significantly high levels of both total and activated PKC-delta levels compared to sensitive cells. Estrogen receptor (ER) alpha content and function are maintained in all the antiestrogen-resistant cell lines. Overexpressing active PKC-delta in Tam-sensitive MCF-7 cells (PKC-delta/MCF-7) led to Tam resistance both in vitro and in vivo. Inhibition of PKC-delta by rottlerin (a relatively specific inhibitor of PKC-delta) or siRNA significantly inhibited estrogen- and Tam-induced growth in antiestrogen-resistant cells. PKC-delta levels are significantly higher in Tam-resistant tumors compared to Tam-sensitive tumors in xenograft model (P<0.05). Taken together, these data suggest that PKC-delta plays a major role in antiestrogen resistance in breast tumor cells and thus provides a new target for treatment.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Moduladores de Receptor Estrogênico/farmacologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Mamárias Animais/metabolismo , Proteína Quinase C/biossíntese , Regulação para Cima , Acetofenonas/farmacologia , Animais , Benzopiranos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , DNA/metabolismo , Feminino , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Nus , Microscopia Confocal , Microscopia de Fluorescência , Transplante de Neoplasias , Fosforilação , Isoformas de Proteínas , Proteína Quinase C-delta , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção
12.
Cancer Metastasis Rev ; 22(4): 395-403, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12884914

RESUMO

Activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway is a frequent event in tumorigenesis. MAPKs have been implicated in cell migration, proteinase-induction, regulation of apoptosis, and angiogenesis, events that are essential for successful completion of metastasis. In this review, we discuss the potential role that MAPKs play in metastasis by regulating cell migration, proteinase-induction and apoptosis.


Assuntos
Sistema de Sinalização das MAP Quinases , Neoplasias/patologia , Apoptose , Movimento Celular , Progressão da Doença , Humanos , Modelos Biológicos , Invasividade Neoplásica , Neoplasias/enzimologia , Neovascularização Patológica , Transdução de Sinais
13.
Clin Cancer Res ; 8(8): 2735-41, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12171907

RESUMO

We have previously reported that combretastatin-A4 prodrug (CA4P), anantitubulin/antiangiogenic agent isolated from the South African willow tree Combretum caffrum, induced cell death primarily through mitotic catastrophe in a panel of human B-lymphoid tumors. In this study, we investigated the molecular aspects of the mitotic catastrophe and whether or not it shares the same pathways of apoptosis. For this we studied the effect of CA4P on selected markers of apoptosis [caspases 9 and 3, poly(ADP-ribose) polymerase (PARP), bcl-2, and bax] and G2-M protein regulators (p53, MDM2, 14-3-3sigma, GADD45, cdc2, cdc25, chk1, wee1, p21, and cyclin B1). The chronic lymphocytic leukemia cell line WSU-CLL was used for this purpose. Western blot analysis showed that 24 h of CA4P (5 nM) exposure induces caspase 9 activation and PARP cleavage. However, the addition of Z-Val-Ala-Asp-fluoromethylketone (a general caspase inhibitor) or Z-Leu-Glu(OMe)-His-Asp(OMe)-CH2F (a caspase 9 inhibitor) before CA4P treatment did not block cell death. No change in bcl-2 or bax protein expression was observed. Exposure of WSU-CLL cells to 4 and 5 nM CA4P was associated with overproduction of total p53 and no dramatic change in MDM2, 14-3-3sigma, GADD45, the cyclin-dependent kinase cdc2, its inhibitory phosphorylation, the cdc2-inhibitory kinase (wee1), chk1, or cdc25 hyperphosphorylation. The overaccumulation of p21 and cyclin B1 protein was obvious at 24 h. Furthermore, CA4P treatment showed an increase in the expression of a marker of mitosis (mitotic protein monoclonal-2 antibody) and an overaccumulation of the cyclin B in the nucleus. Our findings suggest that CA4P induces mitotic catastrophe and arrest of WSU-CLL cells mostly in the M phase independent of p53 and independent of chk1 and cdc2 phosphorylation pathways. Apoptosis is a secondary mechanism of death in a small proportion of cells through activation of caspase 9 and PARP cleavage. The two mechanisms of cell death, i.e., mitotic catastrophe and apoptosis, are independent of each other in our model.


Assuntos
Biomarcadores Tumorais , Caspases/metabolismo , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Proteínas de Neoplasias , Proteínas Nucleares , Poli(ADP-Ribose) Polimerases/metabolismo , Pró-Fármacos/farmacologia , Estilbenos/farmacologia , Proteínas 14-3-3 , Antineoplásicos Fitogênicos/farmacologia , Western Blotting , Proteína Quinase CDC2/metabolismo , Caspase 3 , Caspase 9 , Núcleo Celular/metabolismo , Sobrevivência Celular , Quinase 1 do Ponto de Checagem , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Exonucleases/metabolismo , Exorribonucleases , Citometria de Fluxo , Fase G2 , Marcadores Genéticos , Humanos , Mitose , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-mdm2 , Fatores de Tempo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA