Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Tumour Biol ; 41(3): 1010428319840473, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30915904

RESUMO

Human papillomavirus is detected in over 50% of oropharyngeal squamous cell carcinomas. Human papillomavirus-positive oropharyngeal squamous cell carcinomas differ from human papillomavirus-negative tumors, and both expression patterns are classified as distinct entities. The Bmi-1 oncogene is a well-known member of the mammalian polycomb-group family. HESC5:3 and HES77 are newly developed monoclonal antibodies produced against undifferentiated embryonic stem cells. Our aim was to explore their roles in both human papillomavirus-positive and -negative oropharyngeal squamous cell carcinomas. Our cohort comprised 202 consecutive oropharyngeal squamous cell carcinoma patients diagnosed and treated with curative intent. We used tissue microarray tumor blocks to study the immunohistochemical expression of Bmi-1, HESC5:3, and HES77. We compared the expressions of these stem cell markers with p16 immunoexpression and human papillomavirus status, as well as with other characteristics of the tumor, and with patients' clinical data and follow-up data. Human papillomavirus- and p16-positive tumors expressed less Bmi-1 and more HESC5:3 than the negative tumors. HES77 expression was high in human papillomavirus-positive oropharyngeal squamous cell carcinoma, but it did not correlate with p16 positivity. In our multivariable model, Bmi-1 and HESC5:3 were still associated with human papillomavirus, but the association between human papillomavirus and HES77 remained absent. In conclusion, Bmi-1, HESC5:3, and HES77 may have a different role in human papillomavirus-positive and human papillomavirus-negative tumors. There was no correlation between Bmi-1, HESC5:3, and HES77 expression and survival.


Assuntos
Carcinoma de Células Escamosas/patologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Neoplasias Orofaríngeas/patologia , Neoplasias Orofaríngeas/virologia , Papillomaviridae/isolamento & purificação , Complexo Repressor Polycomb 1/biossíntese , Anticorpos Monoclonais/imunologia , Carcinoma de Células Escamosas/virologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Transição Epitelial-Mesenquimal/genética , Humanos , Infecções por Papillomavirus/virologia
2.
Dev Biol ; 418(1): 124-134, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27475488

RESUMO

Cell fate specification during organogenesis is usually followed by a phase of cell proliferation to produce the required number of differentiated cells. The Caenorhabditis elegans vulva is an excellent model to study how cell fate specification and cell proliferation are coordinated. The six vulval precursor cells (VPCs) are born at the first larval stage, but they arrest in the G1 phase of the cell cycle until the beginning of the third larval stage, when their fates are specified and the three proximal VPCs proliferate to generate 22 vulval cells. An epidermal growth factor (EGF) signal from the gonadal anchor cell combined with lateral DELTA/NOTCH signaling between the VPCs determine the primary (1°) and secondary (2°) fates, respectively. The hox gene lin-39 plays a key role in integrating these spatial patterning signals and in maintaining the VPCs as polarized epithelial cells. Using a fusion-defective eff-1(lf) mutation to keep the VPCs polarized, we find that VPCs lacking lin-39 can neither activate lateral NOTCH signaling nor proliferate. LIN-39 promotes cell cycle progression through two distinct mechanisms. First, LIN-39 maintains the VPCs competent to proliferate by inducing cdk-4 cdk and cye-1 cyclinE expression via a non-canonical HOX binding motif. Second, LIN-39 activates in the adjacent VPCs the NOTCH signaling pathway, which promotes VPC proliferation independently of LIN-39. The hox gene lin-39 is therefore a central node in a regulatory network coordinating VPC differentiation and proliferation.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriologia , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Proteínas de Homeodomínio/metabolismo , Organogênese/genética , Receptores Notch/metabolismo , Vulva/embriologia , Animais , Padronização Corporal/genética , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/biossíntese , Proteínas de Caenorhabditis elegans/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Quinase 4 Dependente de Ciclina/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Células Epiteliais/citologia , Feminino , Glicoproteínas de Membrana/genética , Transdução de Sinais
3.
J Biol Chem ; 290(37): 22460-73, 2015 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-26224636

RESUMO

The pluripotency and self-renewal capacity of embryonic stem (ES) cells is regulated by several transcription factors. Here, we show that the ETS-related transcription factors Etv4 and Etv5 (Etv4/5) are specifically expressed in undifferentiated ES cells, and suppression of Oct3/4 results in down-regulation of Etv4/5. Simultaneous deletion of Etv4 and Etv5 (Etv4/5 double knock-out (dKO)) in ES cells resulted in a flat, epithelial cell-like appearance, whereas the morphology changed into compact colonies in a 2i medium (containing two inhibitors for GSK3 and MEK/ERK). Expression levels of self-renewal marker genes, including Oct3/4 and Nanog, were similar between wild-type and dKO ES cells, whereas proliferation of Etv4/5 dKO ES cells was decreased with overexpression of cyclin-dependent kinase inhibitors (p16/p19, p15, and p57). A differentiation assay revealed that the embryoid bodies derived from Etv4/5 dKO ES cells were smaller than the control, and expression of ectoderm marker genes, including Fgf5, Sox1, and Pax3, was not induced in dKO-derived embryoid bodies. Microarray analysis demonstrated that stem cell-related genes, including Tcf15, Gbx2, Lrh1, Zic3, and Baf60c, were significantly repressed in Etv4/5 dKO ES cells. The artificial expression of Etv4 and/or Etv5 in Etv4/5 dKO ES cells induced re-expression of Tcf15 and Gbx2. These results indicate that Etv4 and Etv5, potentially through regulation of Gbx2 and Tcf15, are involved in the ES cell proliferation and induction of differentiation-associated genes in ES cells.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Fatores de Transcrição/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/citologia , Quinase 3 da Glicogênio Sintase/biossíntese , Quinase 3 da Glicogênio Sintase/genética , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-ets/genética , Fatores de Transcrição/genética
4.
Circulation ; 131(11): 995-1005, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25596186

RESUMO

BACKGROUND: Intracranial aneurysm (IA) is a common vascular disorder that frequently leads to fatal vascular rupture. Although various acquired risk factors associated with IA have been identified, the hereditary basis of IA remains poorly understood. As a result, genetically modified animals accurately modeling IA and related pathogenesis have been lacking, and subsequent drug development has been delayed. METHODS AND RESULTS: The transcription factor Sox17 is robustly expressed in endothelial cells of normal intracerebral arteries. The combination of Sox17 deficiency and angiotensin II infusion in mice induces vascular abnormalities closely resembling the cardinal features of IA such as luminal dilation, wall thinning, tortuosity, and subarachnoid hemorrhages. This combination impairs junctional assembly, cell-matrix adhesion, regeneration capacity, and paracrine secretion in endothelial cells of intracerebral arteries, highlighting key endothelial dysfunctions that lead to IA pathogenesis. Moreover, human IA samples showed reduced Sox17 expression and impaired endothelial integrity, further strengthening the applicability of this animal model to clinical settings. CONCLUSIONS: Our findings demonstrate that Sox17 deficiency in mouse can induce IA under hypertensive conditions, suggesting Sox17 deficiency as a potential genetic factor for IA formation. The Sox17-deficient mouse model provides a novel platform to develop therapeutics for incurable IA.


Assuntos
Endotélio Vascular/patologia , Proteínas HMGB/deficiência , Aneurisma Intracraniano/genética , Fatores de Transcrição SOXF/deficiência , Fatores de Transcrição SOXF/fisiologia , Adulto , Idoso , Angiotensina II/toxicidade , Animais , Aorta/patologia , Células Cultivadas , Artérias Cerebrais/química , Artérias Cerebrais/patologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Dilatação Patológica/genética , Dilatação Patológica/patologia , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Feminino , Proteínas HMGB/genética , Proteínas HMGB/fisiologia , Humanos , Hipertensão/complicações , Aneurisma Intracraniano/etiologia , Aneurisma Intracraniano/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Miócitos de Músculo Liso/química , Comunicação Parácrina , Interferência de RNA , Fatores de Transcrição SOXF/análise , Fatores de Transcrição SOXF/genética , Organismos Livres de Patógenos Específicos , Hemorragia Subaracnóidea/etiologia , Transcrição Gênica , Regulação para Cima , Veias/química
5.
J Cutan Pathol ; 43(3): 200-10, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26349899

RESUMO

BACKGROUND: Actinic keratosis (AK) and Bowen's disease (squamous cell carcinoma in situ, SCCIS) are pre-invasive stages in the development of squamous cell carcinoma (SCC). METHODS: Immunohistochemical study of cyclin D1, cyclin E, p16(INK4a) and p21(Cip1) (/Waf1) in AK (53 cases), SCCIS (16 cases) and SCC (40 cases), in relation to the type of the lesion and SCC prognostic parameters (grade, diameter and thickness). RESULTS: Diffuse cyclin D1 distribution was more frequent in SCCIS and SCC than in AK (p = 0.03) and similar pattern was observed for p16(INK4a) . For cyclin E, central distribution dominated in SCC compared with the AK (p = 0.001) and SCCIS (p = 0.03). p21(Cip1) (/Waf1) displayed suprabasal distribution more frequently in AK than in SCCIS (p = 0.001) and SCC (p = 0.0004). Semiquantitative assessment showed more positive cells in AK (p = 0.04) and SCCIS (p = 0.04) than in SCC for cyclin E. SCC with diameter over 20 mm and those thicker than 6 mm revealed higher labeling index with p16(INK4a) and p21(Cip1) (/Waf1) , respectively. CONCLUSIONS: Our results suggest different alterations for p16(INK4a) and p21(Cip1) (/Waf1) in AK, SCCIS and SCC. Immunostaining distribution showed closer correlation with the type of the lesion, whereas percentage of positive cells displayed better association with the SCC prognostic parameters.


Assuntos
Carcinoma de Células Escamosas , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Ciclinas/biossíntese , Fase G1 , Regulação Neoplásica da Expressão Gênica , Ceratose Actínica , Proteínas de Neoplasias/biossíntese , Fase S , Neoplasias Cutâneas , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Feminino , Humanos , Ceratose Actínica/metabolismo , Ceratose Actínica/patologia , Masculino , Pessoa de Meia-Idade , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
6.
Int J Cancer ; 131(1): 18-29, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21796622

RESUMO

Apoptosis genes, such as TP53 and p16/CDKN2A, that mediate responses to cytotoxic chemotherapy, are frequently nonfunctional in melanoma. Differentiation may be an alternative to apoptosis for inducing melanoma cell cycle exit. Epigenetic mechanisms regulate differentiation, and DNA methylation alterations are associated with the abnormal differentiation of melanoma cells. The effects of the deoxycytidine analogue decitabine (5-aza-2'-deoxycytidine), which depletes DNA methyl transferase 1 (DNMT1), on melanoma differentiation were examined. Treatment of human and murine melanoma cells in vitro with concentrations of decitabine that did not cause apoptosis inhibited proliferation accompanied by cellular differentiation. A decrease in promoter methylation, and increase in expression of the melanocyte late-differentiation driver SOX9, was followed by increases in cyclin-dependent kinase inhibitors (CDKN) p27/CDKN1B and p21/CDKN1A that mediate cell cycle exit with differentiation. Effects were independent of the TP53, p16/CDKN2A and also the BRAF status of the melanoma cells. Resistance, when observed, was pharmacologic, characterized by diminished ability of decitabine to deplete DNMT1. Treatment of murine melanoma models in vivo with intermittent, low-dose decitabine, administered sub-cutaneously to limit high peak drug levels that cause cytotoxicity and increase exposure time for DNMT1 depletion, and with tetrahydrouridine to decrease decitabine metabolism and further increase exposure time, inhibited tumor growth and increased molecular and tumor stromal factors implicated in melanocyte differentiation. Modification of decitabine dose, schedule and formulation for differentiation rather than cytotoxic objectives inhibits the growth of melanoma cells in vitro and in vivo.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , DNA (Citosina-5-)-Metiltransferases/metabolismo , Epigênese Genética , Melanoma Experimental/tratamento farmacológico , Animais , Apoptose , Azacitidina/administração & dosagem , Azacitidina/farmacologia , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/análise , Metilação de DNA , Decitabina , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas B-raf/biossíntese , Fatores de Transcrição SOX9/biossíntese , Análise de Sequência de DNA , Tetra-Hidrouridina/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
7.
Biochem Biophys Res Commun ; 420(1): 29-35, 2012 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-22390936

RESUMO

Cyclin-dependent kinase inhibitor 3 (CDKN3) belongs to the protein phosphatases family and has a dual function in cell cycling. The function of this gene has been studied in several kinds of cancers, but its role in human hepatocellular carcinoma (HCC) remains to be elucidated. In this study, we found that CDKN3 was frequently overexpressed in both HCC cell lines and clinical samples, and this overexpression was correlated with poor tumor differentiation and advanced tumor stage. Functional studies showed that overexpression of CDKN3 could promote cell proliferation by stimulating G1-S transition but has no impact on cell apoptosis and invasion. Microarray-based co-expression analysis identified a total of 61 genes co-expressed with CDKN3, with most of them involved in cell proliferation, and BIRC5 was located at the center of CDKN3 co-expression network. These results suggest that CDKN3 acts as an oncogene in human hepatocellular carcinoma and antagonism of CDKN3 may be of interest for the treatment of HCC.


Assuntos
Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/patologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Fosfatases de Especificidade Dupla/biossíntese , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/patologia , Adulto , Proliferação de Células , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Fosfatases de Especificidade Dupla/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Adulto Jovem
8.
Biochim Biophys Acta Gene Regul Mech ; 1864(6-7): 194714, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33971368

RESUMO

Dependent on phosphate availability the yeast Saccharomyces cerevisiae expresses either low or high affinity phosphate transporters. In the presence of phosphate yeast cells still express low levels of the high affinity phosphate transporter Pho84. The regulator Spl2 is expressed in approximately 90% of the cells, and is not expressed in the remaining cells. Here we report that deletion of RRP6, encoding an exonuclease degrading non-coding RNA, or BMH1, encoding the major 14-3-3 isoform, resulted in less cells expressing SPL2 and in increased levels of RNA transcribed from sequences upstream of the SPL2 coding region. SPL2 stimulates its own expression and that of PHO84 ensuing a positive feedback. Upon deletion of the region responsible for upstream SPL2 transcription almost all cells express SPL2. These results indicate that the cell-to-cell variation in PHO84 and SPL2 expression is dependent on a specific part of the SPL2 promoter and is controlled by Bmh1 and Spl2.


Assuntos
Proteínas 14-3-3/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Regulação Fúngica da Expressão Gênica , Fosfatos/metabolismo , Proteínas de Saccharomyces cerevisiae/biossíntese , Saccharomyces cerevisiae/metabolismo , Transcrição Gênica , Proteínas 14-3-3/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
9.
Allergy ; 65(11): 1438-45, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20456314

RESUMO

BACKGROUND: Bronchial epithelium is considered a key player in coordinating airway wall remodelling. The function of epithelial cells can be modulated by the underlying fibroblasts through autocrine and paracrine mechanisms. OBJECTIVE: To investigate the effect of phenotypic changes in bronchial fibroblasts from asthmatic subjects on epithelial cell proliferation. METHODS: Epithelial cells and fibroblasts derived from bronchial biopsies of asthmatic and healthy controls were cultured in an engineered model. Proliferation was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium-bromid (MTT). Epidermal growth factor receptor (EGFR), cyclin-dependent kinase inhibitors p21 and p27 were measured by western blots. Total and active forms of transforming growth factor (TGF)-ß1 were measured using ELISA and bioassay. TGF-ß was inhibited using a recombinant TGF-ß soluble receptor II protein. RESULTS: Proliferation of epithelial cells from asthmatics (AE) is increased when cells were cultured with fibroblasts from normal controls (NF). Fibroblasts from asthmatics (AF) significantly decreased the proliferation of epithelial cells from healthy subjects (NE). Activation of p21, p27, EGFR and TGF-ß1 reflects the proliferation data by decreasing in AE cultured with NF and increasing in NE cultured with AF. Neutralization of TGF-ß increased proliferation of epithelial cells cultured in the asthmatic model. CONCLUSION: Fibroblasts from asthmatic subjects regulate epithelial cell prolifearation, and TGF-ß signalling may represent one of the pathway involved in these interactions.


Assuntos
Asma/metabolismo , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais/fisiologia , Asma/imunologia , Asma/patologia , Western Blotting , Brônquios/imunologia , Brônquios/metabolismo , Brônquios/patologia , Comunicação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/imunologia , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/imunologia , Células Epiteliais/ultraestrutura , Receptores ErbB/metabolismo , Fibroblastos/imunologia , Fibroblastos/ultraestrutura , Humanos , Microscopia Eletrônica de Varredura , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Engenharia Tecidual , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/imunologia
10.
Br J Nutr ; 101(9): 1306-15, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19079835

RESUMO

Oligosaccharides are present in human milk in large amounts and in a high variety. We have previously shown that these oligosaccharides are strong inhibitors of proliferation and inducers of differentiation in intestinal cell lines. To elucidate the molecular mechanism, we investigated the influence on cell cycle events via flow cytometry and expression levels by using quantitative real-time RT-PCR. Human intestinal cells, i.e. HT-29, HIEC and Caco-2 cells, were exposed to neutral or acidic human milk oligosaccharides. Both fractions induced a concentration-dependent G2/M arrest. Cell cycle analysis for HT-29 revealed 37 % of cells in G1 and 35 % in G2/M (neutral oligosaccharides) and incubation with acidic oligosaccharides led to 42 % cells in G1 and 40 % in G2/M. In control experiments without oligosaccharides we found 71 % of cells to be in G1 and 17 % in G2/M. This G2/M arrest was associated with changes in mRNA expression of cyclin A and B. A G2/M arrest with concomitant alterations of cell cycle gene expression could also be shown for HIEC and Caco-2 cells. Analysing the expression of cyclin-dependent kinase inhibitors p21(cip1) and p27(kip1) and the tumour suppressor p53 we observed that the expression of p21(cip1) was p53-independent and necessary for arresting cells in the G2/M phase, while p27(kip1) was associated with differentiation effects. Both neutral and acidic human milk oligosaccharides were able to induce epidermal growth factor receptor, extracellular signal-regulated kinase 1/2 and p38 phosphorylation. These results suggest that oligosaccharides from human milk inhibited intestinal cell proliferation and altered cell cycle dynamics by affecting corresponding regulator genes and mitogen-activated protein kinase signalling.


Assuntos
Carboidratos da Dieta/farmacologia , Genes cdc/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Leite Humano/química , Oligossacarídeos/farmacologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Ciclinas/biossíntese , Ciclinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética
11.
J Cutan Pathol ; 36(7): 753-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19519606

RESUMO

BACKGROUND: Loss of p16 in melanomas reflects worse outcomes for patients. It is associated with depth of invasion, ulceration, vascular invasion, lymph node metastases, metastases, recurrence of melanoma and decreased 5-year survival. Desmoplastic melanoma is an insidious malignant melanoma subtype that commonly occurs on sun-damaged skin of the head and neck area in elderly patients. The diagnostic conundrum occurs with confusion of desmoplastic melanoma with scars, hyalinizing blue nevi, desmoplastic Spitz nevi and diffuse neurofibromas. METHODS: The present study uses immunohistochemistry with a p16 antibody to differentiate desmoplastic Spitz nevi (n = 15 cases) from desmoplastic melanomas (n = 11). To date, no other studies have been published defining the expression pattern of p16 in desmoplastic melanoma. RESULTS: 81.8% of desmoplastic melanomas were negative for p16 and 18.2% were only weakly stained. In contrast, all desmoplastic Spitz nevi were moderately to strongly positive for p16. CONCLUSIONS: The staining pattern for p16 in desmoplastic melanomas and Spitz nevi in conjunction with the histopathologic features, S100 staining, Ki67 proliferation index and clinical scenario may aid in the difficult differential diagnosis between these two entities. Further confirmatory studies are indicated.


Assuntos
Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Melanoma/patologia , Nevo de Células Epitelioides e Fusiformes/metabolismo , Nevo de Células Epitelioides e Fusiformes/patologia , Adulto , Proliferação de Células , Diagnóstico Diferencial , Feminino , Humanos , Antígeno Ki-67/biossíntese , Metástase Linfática , Masculino , Melanoma/diagnóstico , Pessoa de Meia-Idade , Nevo de Células Epitelioides e Fusiformes/diagnóstico
12.
Cancer Res ; 67(1): 130-8, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17210692

RESUMO

The cyclin-dependent kinase (Cdk)-associated protein phosphatase KAP is a dual-specificity phosphatase of which the only known function is to dephosphorylate Cdk2 and inhibit cell cycle progression. Paradoxically, we find increased KAP mRNA expression in malignant astrocytomas, which correlates with increasing histologic grade and decreased patient survival. We have resolved this apparent paradox with the discovery of aberrant KAP splicing in malignant astrocytomas that leads to increased expression of KAP-related transcripts but decreased KAP protein expression. In addition, the aberrant splicing generates a dominant negative KAP variant that increases proliferation. We provide the first evidence that KAP not only regulates proliferation but also inhibits migration by decreasing cdc2 mRNA and protein expression. The effect of KAP on cdc2 expression requires its phosphatase activity but does not involve direct dephosphorylation of cdc2. Thus, KAP regulates both cdc2-dependent migration and Cdk2-dependent proliferation, and its loss due to aberrant splicing increases malignancy in human gliomas.


Assuntos
Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Movimento Celular/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Glioblastoma/enzimologia , Glioblastoma/patologia , Proteínas Tirosina Fosfatases/genética , Processamento Alternativo , Sequência de Bases , Neoplasias Encefálicas/genética , Proteína Quinase CDC2/biossíntese , Proteína Quinase CDC2/genética , Processos de Crescimento Celular/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Fosfatases de Especificidade Dupla , Glioblastoma/genética , Humanos , Isoenzimas/biossíntese , Isoenzimas/genética , Dados de Sequência Molecular , Proteínas Tirosina Fosfatases/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transfecção
13.
J Neurosci ; 27(6): 1434-44, 2007 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-17287518

RESUMO

Sensory hair cells of the auditory organ are generated during embryogenesis and remain postmitotic throughout life. Previous work has shown that inactivation of the cyclin-dependent kinase inhibitor (CKI) p19(Ink4d) leads to progressive hearing loss attributable to inappropriate DNA replication and subsequent apoptosis of hair cells. Here we show the synergistic action of another CKI, p21(Cip1), on cell cycle reactivation. The codeletion of p19(Ink4d) and p21(Cip1) triggered profuse S-phase entry of auditory hair cells during a restricted period in early postnatal life, leading to the transient appearance of supernumerary hair cells. In addition, we show that aberrant cell cycle reentry leads to activation of a DNA damage response pathway in these cells, followed by p53-mediated apoptosis. The majority of hair cells were absent in adult cochleas. These data, together with the demonstration of changing expression patterns of multiple CKIs in auditory hair cells during the stages of early postnatal maturation, show that the maintenance of the postmitotic state is an active, tissue-specific process, cooperatively regulated by several CKIs, and is critical for the lifelong survival of these sensory cells.


Assuntos
Apoptose/fisiologia , Cóclea/patologia , Inibidor de Quinase Dependente de Ciclina p19/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Genes cdc , Células Ciliadas Auditivas/patologia , Fase S , Animais , Animais Recém-Nascidos , Animais Lactentes , Contagem de Células , Cóclea/crescimento & desenvolvimento , Cruzamentos Genéticos , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p19/deficiência , Inibidor de Quinase Dependente de Ciclina p19/genética , Inibidor de Quinase Dependente de Ciclina p21/deficiência , Inibidor de Quinase Dependente de Ciclina p21/genética , Dano ao DNA , Reparo do DNA/genética , Progressão da Doença , Células Ciliadas Auditivas/metabolismo , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitose , Especificidade de Órgãos , Órgão Espiral/patologia , Transfecção , Proteína Supressora de Tumor p53/fisiologia
14.
Clin Sci (Lond) ; 114(1): 73-83, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17678500

RESUMO

Ras oncoproteins are probably implicated in normal and malignant cell growth in various organs. Inhibition of Ras interferes with cell proliferation of non-hepatic cells in vitro and in vivo. A potential role for Ras in normal and malignant hepatocyte proliferation prompted us to evaluate the impact of Ras inhibition by FTS (S-farnesylthiosalicylic acid) on hepatocyte proliferation in vitro in the human hepatic tumour cell line HepG2 and in vivo after PH (partial hepatectomy) in rats. Rats were administered with FTS intraperitoneally (1, 8 and 16 h after PH) and killed 12, 24 and 48 h after PH. Cell proliferation, phosphorlyation of members of the MAPK (mitogen-activated protein kinase) pathway and levels and activity of cell cycle effectors (cyclin D, cyclin E, Cdk2 and Cdk4) were assessed in FTS-treated rats compared with controls. FTS significantly decreased overall cell count, PCNA (proliferating-cell nuclear antigen) expression and BrdU (bromodeoxyuridine) incorporation into HepG2 cells after 7 days of culture. FTS treatment significantly reduced BrdU incorporation and PCNA expression in hepatocytes after PH. Unlike control rats, cell-membrane expression of Ras was decreased in FTS-treated animals after PH, resulting in decreased Raf membrane recruitment and phosphorylation and in reduced phosphorylation of ERK1/2 (extracellular-signal-regulated kinase 1/2). The antiproliferative effect of FTS was linked to a decrease in expression and activity of the cyclin E/Cdk2 complex, without affecting cyclin D and Cdk4. Ras inhibition by FTS significantly decreased proliferation of HepG2 cells and normal hepatocytes after a strong and highly synchronized proliferation stimulus elicited by PH. The inhibitory effect was at least partially mediated by inhibition of Ras/Raf/MAPK signalling. It appears worthwhile to evaluate the impact of Ras inhibition on the development of hepatocarcinomas in vivo in adequate animal models.


Assuntos
Farneseno Álcool/análogos & derivados , Hepatócitos/efeitos dos fármacos , Neoplasias Hepáticas Experimentais/patologia , Proteína Oncogênica p21(ras)/antagonistas & inibidores , Salicilatos/farmacologia , Animais , Índice de Massa Corporal , Proteínas de Ciclo Celular/metabolismo , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Citosol/metabolismo , Farneseno Álcool/farmacologia , Regulação da Expressão Gênica , Hepatectomia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Proteína Oncogênica p21(ras)/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/metabolismo , RNA Mensageiro , Ratos , Ratos Wistar , Células Tumorais Cultivadas
15.
Cell Death Dis ; 9(9): 893, 2018 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-30166528

RESUMO

The role of sirtuins (SIRTs) in cancer biology has been the focus of recent research. The similarities between underlying pathways involved in the induction of pluripotent stem cells and transformation of cancer cells revealed the role of SIRTs in cellular reprogramming. Seven SIRTs have been identified in mammals and downregulation of SIRT2 was found to facilitate the generation of primed pluripotent stem cells, such as human induced pluripotent stem cells. Herein, we evaluated the role of SIRT2 in naive pluripotent stem cell generation using murine cells. We found that absolute depletion of SIRT2 in mouse embryonic fibroblasts resulted in a notable reduction in reprogramming efficiency. SIRT2 depletion not only upregulated elements of the INK4/ARF locus, which in turn had an antiproliferative effect, but also significantly altered the expression of proteins related to the PI3K/Akt and Hippo pathways, which are important signaling pathways for stemness. Thus, this study demonstrated that SIRT2 is required for cellular reprogramming to naive states of pluripotency in contrast to primed pluripotency states.


Assuntos
Reprogramação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes/citologia , Sirtuína 2/genética , Animais , Ciclo Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Via de Sinalização Hippo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo
16.
Oncogene ; 25(16): 2304-17, 2006 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-16331265

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARgamma) agonists demonstrate antitumor activity likely through transactivating genes that regulate cell proliferation, apoptosis, and differentiation. The PAX8/PPARgamma fusion oncogene, which is common in human follicular thyroid carcinomas appears to act via dominant negative suppression of wild-type PPARgamma, suggesting that it may be a tumor suppressor gene in thyroid cells. We have identified a novel high-affinity PPARgamma agonist (RS5444) that is dependent upon PPARgamma for its biological activity. This is the first report of this molecule and its antitumor activity. In vitro, the IC50 for growth inhibition is approximately 0.8 nM while anaplastic thyroid carcinoma (ATC) tumor growth was inhibited three- to fourfold in nude mice. siRNA against PPARgamma and a pharmacological antagonist demonstrated that functional PPARgamma was required for growth inhibitory activity of RS5444. RS5444 upregulated the cell cycle kinase inhibitor, p21WAF1/CIP1. Silencing p21WAF1/CIP1 rendered cells insensitive to RS5444. RS5444 plus paclitaxel demonstrated additive antiproliferative activity in cell culture and minimal ATC tumor growth in vivo. RS5444 did not induce apoptosis but combined with paclitaxel, doubled the apoptotic index compared to that of paclitaxel. Our data indicate that functional PPARgamma is a molecular target for therapy in ATC. We demonstrated that RS5444, a thiazolidinedione (Tzd) derivative, alone or in combination with paclitaxel, may provide therapeutic benefit to patients diagnosed with ATC.


Assuntos
Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , PPAR gama/agonistas , Paclitaxel/administração & dosagem , Tiazolidinedionas/uso terapêutico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromanos/farmacologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Feminino , Humanos , Camundongos , PPAR gama/fisiologia , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/farmacologia , Neoplasias da Glândula Tireoide/patologia , Troglitazona
17.
Zhonghua Fu Chan Ke Za Zhi ; 42(9): 600-4, 2007 Sep.
Artigo em Zh | MEDLINE | ID: mdl-17983514

RESUMO

OBJECTIVE: To evaluate the relationship between combined multigene detection and response to chemotherapy and prognosis in epithelial ovarian carcinomas (EOCS). METHODS: A total of 80 ovarian tissue samples taken from the surgical specimens of patients with EOCS of our hospital in the last two decades who had received chemotherapy after surgery were paraffin-embedded. The samples were divided into 2 groups, good prognosis group (patients who survived more than 2 years, n = 46) and poor prognosis group (patients who survived less than 2 years, n = 34). The expression levels of ToPo-II, Ki-67, MGMT, PCNA, p27, p53, p16, P-gp, LRP, GST-pi, bcl-2, C-myc, Fas, bax, MSH2, MRP and BCRP were investigated by the combination of tissue arrays and immunohistochemical streptavidin-biotin peroxidase (SP) method in all samples. Data were analysed with SPSS 12.0 for windows. RESULTS: There were statistically significant differences in the positive expression levels of P-gp, BCRP, MGMT, MSH2, p27 and p16 (62%, 50% and 50% in poor prognosis group vs 33%, 28% and 28% respectively, P < 0.05) in the good prognosis group, suggesting that the positive expression levels of P-gp, BCRP, MGMT, MSH2, p27 and p16 were related to the response to chemotherapy and prognosis in EOCS. And the positive expression of P-gp, BCRP and MSH2 (43%, 54% and 43%) indicated poor prognosis, while the positive expression of MGMT, p27 and p16 (18%, 29% and 24%, P < 0.05) indicated good prognosis. Cox multigene expression analysis confirmed that the positive expression levels of MRP, C-myc, LRP, p16, p27, MGMT, ToPo-II, P-gp and GST-pi were related to the response to chemotherapy and prognosis in EOCS. And the positive expression of MRP, C-myc, LRP, ToPo-II, P-gp and GST-pi indicated poor prognosis, while the positive expression of MGMT, p27 and p16 indicated good prognosis. Combined multigene detections were conducted among P-gp, BCRP, MGMT, MSH2, p27 and p16, and there were statistically significant differences in the positive coexpression of P-gp plus MGMT in the two groups (P < 0.05); indicating that the combined multigene expression were related to the response to chemotherapy and prognosis in EOCS. The predictive value to response to chemotherapy and prognosis of the positive coexpression of P-gp plus MGMT was 70%. CONCLUSIONS: By univariate and multivariate analyses, the positive expression of P-gp, MGMT, p27 and p16 are related to the response to chemotherapy and prognosis in EOCS. The combined multigene expression of P-gp plus MGMT are related to the response to chemotherapy and prognosis and could predict prognosis more effectively.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , O(6)-Metilguanina-DNA Metiltransferase/biossíntese , Neoplasias Ovarianas/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Adulto , Idoso , Células Epiteliais/patologia , Feminino , Glutationa S-Transferase pi/biossíntese , Humanos , Imuno-Histoquímica , Modelos Logísticos , Pessoa de Meia-Idade , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Prognóstico , Análise de Sobrevida , Análise Serial de Tecidos
18.
Oncol Res ; 25(9): 1431-1440, 2017 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-28109073

RESUMO

Nasopharyngeal carcinoma (NPC) is a common malignancy of the head and neck that arises from the nasopharynx epithelium and is highly invasive. Cyclin-dependent kinase inhibitor 3 (CDKN3) belongs to the dual-specificity protein phosphatase family, which plays a key role in regulating cell division. Abnormal expression of CDKN3 has been found in numerous types of cancer. In the current study, we explored the possible role of CDKN3 in cell proliferation, ability to invade, and radiosensitivity in NPC cells. We reported that CDKN3 was upregulated and p27 was downregulated in NPC tissues and is associated with a worse prognosis for patients. In addition, downregulation of CDKN3 and upregulation of p27 decreased cell proliferation, induced cell cycle arrest, increased apoptosis, decreased cell invasion, and enhanced radiosensitivity. Silencing of p27 significantly inhibited the effects of the knockdown of CDKN3. Moreover, downregulation of CDKN3 and upregulation of p27 inhibited the increase in tumor volume and weight in implanted tumors, decreased the phosphorylation of Akt, and increased the expression of cleaved caspase 3 in tumors. CDKN3 expression was also inversely correlated with p27 expression in NPC patients. Knockdown of CDKN3 increased p27 expression. Silencing of p27 markedly inhibited the effects of CDKN3 on cell proliferation, cell cycle progression, apoptosis, invasion, and radiosensitivity. These results demonstrate that upregulation of p27 is involved in the knockdown of CDKN3-induced decrease in cell proliferation, increase in cell cycle arrest and apoptosis, decrease in invasion, and increase in radiosensitivity. The results demonstrate that the CDKN3/p27 axis may be a novel target in the treatment of NPC.


Assuntos
Carcinoma/metabolismo , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Animais , Apoptose/fisiologia , Carcinogênese , Carcinoma/genética , Carcinoma/patologia , Carcinoma/radioterapia , Pontos de Checagem do Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/genética , Fosfatases de Especificidade Dupla/biossíntese , Fosfatases de Especificidade Dupla/genética , Humanos , Camundongos , Camundongos Nus , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/radioterapia , Proteína Oncogênica v-akt/metabolismo , Transfecção , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
19.
PLoS One ; 11(4): e0154719, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27123979

RESUMO

Golgi phosphoprotein 3 (GOLPH3) has been implicated in the development of carcinomas in many human tissues, and is currently considered a bona fide oncoprotein. Importantly, several tumor types show overexpression of GOLPH3, which is associated with tumor progress and poor prognosis. However, the underlying molecular mechanisms that connect GOLPH3 function with tumorigenicity are poorly understood. Experimental evidence shows that depletion of GOLPH3 abolishes transformation and proliferation of tumor cells in GOLPH3-overexpressing cell lines. Conversely, GOLPH3 overexpression drives transformation of primary cell lines and enhances mouse xenograft tumor growth in vivo. This evidence suggests that overexpression of GOLPH3 could result in distinct features of GOLPH3 in tumor cells compared to that of non-tumorigenic cells. GOLPH3 is a peripheral membrane protein mostly localized at the trans-Golgi network, and its association with Golgi membranes depends on binding to phosphatidylinositol-4-phosphate. GOLPH3 is also contained in a large cytosolic pool that rapidly exchanges with Golgi-associated pools. GOLPH3 has also been observed associated with vesicles and tubules arising from the Golgi, as well as other cellular compartments, and hence it has been implicated in several membrane trafficking events. Whether these and other features are typical to all different types of cells is unknown. Moreover, it remains undetermined how GOLPH3 acts as an oncoprotein at the Golgi. Therefore, to better understand the roles of GOLPH3 in cancer cells, we sought to compare some of its biochemical and cellular properties in the human breast cancer cell lines MCF7 and MDA-MB-231 with that of the non-tumorigenic breast human cell line MCF 10A. We found unexpected differences that support the notion that in different cancer cells, overexpression of GOLPH3 functions in diverse fashions, which may influence specific tumorigenic phenotypes.


Assuntos
Neoplasias da Mama/patologia , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , 1-Fosfatidilinositol 4-Quinase/metabolismo , Animais , Linhagem Celular Tumoral , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Feminino , Células HeLa , Humanos , Células MCF-7 , Camundongos , Transplante de Neoplasias , Fosfatos de Fosfatidilinositol/metabolismo , Ratos , Transplante Heterólogo , Rede trans-Golgi/metabolismo
20.
Oncogene ; 34(11): 1432-41, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-24704824

RESUMO

Aberrant splicing of the cyclin-dependent kinase-associated phosphatase, KAP, promotes glioblastoma invasion in a Cdc2-dependent manner. However, the mechanism by which this occurs is unknown. Here we show that miR-26a, which is often amplified in glioblastoma, promotes invasion in phosphatase and tensin homolog (PTEN)-competent and PTEN-deficient glioblastoma cells by directly downregulating KAP expression. Mechanistically, we find that KAP binds and activates ROCK2. Thus, RNA-mediated downregulation of KAP leads to decreased ROCK2 activity and this, in turn, increases Rac1-mediated invasion. In addition, the decrease in KAP expression activates the cyclin-dependent kinase, Cdk2, and this directly promotes invasion by increasing retinoblastoma phosphorylation, E2F-dependent Cdc2 expression and Cdc2-mediated inactivation of the actomyosin inhibitor, caldesmon. Importantly, glioblastoma cell invasion mediated by this pathway can be antagonized by Cdk2/Cdc2 inhibitors in vitro and in vivo. Thus, two distinct RNA-based mechanisms activate this novel KAP/ROCK2/Cdk2-dependent invasion pathway in glioblastoma.


Assuntos
Quinase 2 Dependente de Ciclina/metabolismo , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Glioblastoma/patologia , MicroRNAs/fisiologia , Quinases Associadas a rho/metabolismo , Actomiosina/antagonistas & inibidores , Neoplasias Encefálicas/patologia , Proteína Quinase CDC2 , Proteínas de Ligação a Calmodulina/antagonistas & inibidores , Linhagem Celular Tumoral , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/biossíntese , Fosfatases de Especificidade Dupla/biossíntese , Fosfatases de Especificidade Dupla/genética , Fatores de Transcrição E2F/fisiologia , Ativação Enzimática , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Invasividade Neoplásica , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno , Proteína do Retinoblastoma/metabolismo , Proteínas rac1 de Ligação ao GTP/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA