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.
Oncogene ; 35(45): 5882-5892, 2016 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-27109098

RESUMO

Pancreatic cancer (PC) and associated pre-neoplastic lesions have been reported to be hypoxic, primarily due to hypovascular nature of PC. Though the presence of hypoxia under cancerous condition has been associated with the overexpression of oncogenic proteins (MUC1), multiple emerging reports have also indicated the growth inhibitory effects of hypoxia. In spite of being recognized as the top-most differentially expressed and established oncogenic protein in PC, MUC4 regulation in terms of micro-environmental stress has not been determined. Herein, for the first time, we are reporting that MUC4 protein stability is drastically affected in PC, under hypoxic condition in a hypoxia inducible factor 1α (HIF-1α)-independent manner. Mechanistically, we have demonstrated that hypoxia-mediated induction of reactive oxygen species (ROS) promotes autophagy by inhibiting pAkt/mTORC1 pathway, one of the central regulators of autophagy. Immunohistofluorescence analyses revealed significant negative correlation (P-value=0.017) between 8-hydroxy guanosine (8-OHG) and MUC4 in primary pancreatic tumors (n=25). Moreover, we found pronounced colocalization between MUC4 and LAMP1/LC3 (microtubule-associated protein 1A/1B-light chain 3) in PC tissues and also observed their negative relationship in their expression pattern, suggesting that areas with high autophagy rate had less MUC4 expression. We also found that hypoxia and ROS have negative impact on overall cell growth and viability, which was partially, though significantly (P<0.05), rescued in the presence of MUC4. Altogether, hypoxia-mediated oxidative stress induces autophagy in PC, leading to the MUC4 degradation to enhance survival, possibly by offering required metabolites to stressed cells.


Assuntos
Autofagia , Sobrevivência Celular , Hipóxia/metabolismo , Mucina-4/metabolismo , Estresse Oxidativo , Neoplasias Pancreáticas/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Modelos Biológicos , Mucina-4/genética , Complexos Multiproteicos/metabolismo , Neoplasias Pancreáticas/genética , Estabilidade Proteica , Proteólise , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo
2.
Oncogene ; 35(31): 4112-21, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-26751774

RESUMO

MUC5AC is a secretory mucin aberrantly expressed in various cancers. In lung cancer, MUC5AC is overexpressed in both primary and metastatic lesions; however, its functional role is not well understood. The present study was aimed at evaluating mechanistic role of MUC5AC on metastasis of lung cancer cells. Clinically, the overexpression of MUC5AC was observed in lung cancer patient tissues and was associated with poor survival. In addition, the overexpression of Muc5ac was also observed in genetically engineered mouse lung adenocarcinoma tissues (Kras(G12D); Trp53(R172H/+); AdCre) in comparison with normal lung tissues. Our functional studies showed that MUC5AC knockdown resulted in significantly decreased migration in two lung cancer cell lines (A549 and H1437) as compared with scramble cells. Expression of integrins (α5, ß1, ß3, ß4 and ß5) was decreased in MUC5AC knockdown cells. As both integrins and MUC5AC have a von Willebrand factor domain, we assessed for possible interaction of MUC5AC and integrins in lung cancer cells. MUC5AC strongly interacted only with integrin ß4. The co-localization of MUC5AC and integrin ß4 was observed both in A549 lung cancer cells as well as genetically engineered mouse adenocarcinoma tissues. Activated integrins recruit focal adhesion kinase (FAK) that mediates metastatic downstream signaling pathways. Phosphorylation of FAK (Y397) was decreased in MUC5AC knockdown cells. MUC5AC/integrin ß4/FAK-mediated lung cancer cell migration was confirmed through experiments utilizing a phosphorylation (Y397)-specific FAK inhibitor. In conclusion, overexpression of MUC5AC is a poor prognostic marker in lung cancer. MUC5AC interacts with integrin ß4 that mediates phosphorylation of FAK at Y397 leading to lung cancer cell migration.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Integrina beta4/fisiologia , Neoplasias Pulmonares/patologia , Mucina-5AC/fisiologia , Transdução de Sinais/fisiologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Humanos , Integrina beta4/análise , Masculino , Camundongos , Mucina-5AC/análise , Fosforilação
3.
Oncogene ; 34(37): 4879-89, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-25531332

RESUMO

Pancreatic cancer (PC) is characterized by aberrant overexpression of mucins that contribute to its pathogenesis. Although the inflammatory cytokines contribute to mucin overexpression, the mucin profile of PC is markedly distinct from that of normal or inflamed pancreas. We postulated that de novo expression of various mucins in PC involves chromatin modifications. Analysis of chromatin modifying enzymes by PCR array identified differential expression of NCOA3 in MUC4-expressing PC cell lines. Immunohistochemistry analysis in tumor tissues from patients and spontaneous mouse models, and microarray analysis following the knockdown of NCOA3 were performed to elucidate its role in mucin regulation and overall impact on PC. Silencing of NCOA3 in PC cell lines resulted in significant downregulation of two most differentially expressed mucins in PC, MUC4 and MUC1 (P<0.01). Immunohistochemistry analysis in PC tissues and metastatic lesions established an association between NCOA3 and mucin (MUC1 and MUC4) expression. Spontaneous mouse model of PC (K-ras(G12D); Pdx-1cre) showed early expression of Ncoa3 during pre-neoplastic lesions. Mechanistically, NCOA3 knockdown abrogated retinoic acid-mediated MUC4 upregulation by restricting MUC4 promoter accessibility as demonstrated by micrococcus nuclease digestion (P<0.05) and chromatin immuno-precipitation analysis. NCOA3 also created pro-inflammatory conditions by upregulating chemokines like CXCL1, 2, 5 and CCL20 (P<0.001). AKT, ubiquitin C, ERK1/2 and NF-κB occupied dominant nodes in the networks significantly modulated after NCOA3 silencing. In addition, NCOA3 stabilized mucins post translationally through fucosylation by FUT8, as the knockdown of FUT8 resulted in the downregulation of MUC4 and MUC1 at protein levels.


Assuntos
Mucina-1 , Mucina-4 , Coativador 3 de Receptor Nuclear/fisiologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Processamento de Proteína Pós-Traducional/genética , Animais , Transformação Celular Neoplásica/genética , Fucose/metabolismo , Fucosiltransferases/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Análise em Microsséries , Mucina-1/genética , Mucina-1/metabolismo , Mucina-4/genética , Mucina-4/metabolismo , Neoplasias Pancreáticas/patologia , Ativação Transcricional , Células Tumorais Cultivadas , Regulação para Cima/genética
4.
Br J Cancer ; 111(3): 486-96, 2014 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-25003666

RESUMO

BACKGROUND: Cancer stem cells (CSCs) contribute towards disease aggressiveness and drug resistance. Specific identification of CSC maintenance genes and targeting can improve the efficiency of currently available treatment modalities. Pancreatic differentiation 2 (PD2) has a major role in the self-renewal of mouse embryonic stem cells. In the present study, we investigated the role of PD2 in pancreatic CSCs. METHODS: Characterisation of CSCs and non-CSCs from mouse models, pancreatic cancer cells and human tissues by CSC and self-renewal marker analysis using confocal assay. Effect of PD2 knockdown in CSCs (after gemcitabine treatment) was studied by immunoblot and apoptosis assays. RESULTS: A subpopulation of cells displayed PD2 overexpression in mouse (Kras(G12D); Pdx1-Cre and Kras(G12D); Trp53(R172H/+); Pdx1-Cre) and human pancreatic tumours, which co-express CSC markers. Cancer stem cells exhibited elevated expression of PD2 and self-renewal markers, such as Oct3/4, Shh and ß-catenin. Gemcitabine treatment maintained the CSC population with simultaneous maintenance of PD2 and CSC marker expression. Knockdown of PD2 in CSCs resulted in reduced viability of cells and enhanced apoptosis along with abrogated expression of CD133 and MDR2. CONCLUSIONS: Our results suggest that PD2 is a novel CSC maintenance protein, loss of which renders the CSCs more susceptible to drug-induced cell death.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Células-Tronco Neoplásicas/metabolismo , Proteínas Nucleares/fisiologia , Neoplasias Pancreáticas/metabolismo , Animais , Antimetabólitos Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Transgênicos , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Pancreáticas/patologia , Células da Side Population/efeitos dos fármacos , Células da Side Population/metabolismo , Fatores de Transcrição , Gencitabina
5.
Br J Cancer ; 108(12): 2590-600, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23598299

RESUMO

BACKGROUND: Protein phosphatase 2A (PP2A) is a dephosphorylating enzyme, loss of which can contribute to prostate cancer (PCa) pathogenesis. The aim of this study was to analyse the transcriptional and translational expression patterns of individual subunits of the PP2A holoenzyme during PCa progression. METHODS: Immunohistochemistry (IHC), western blot, and real-time PCR was performed on androgen-dependent (AD) and androgen-independent (AI) PCa cells, and benign and malignant prostate tissues for all the three PP2A (scaffold, regulatory, and catalytic) subunits. Mechanistic and functional studies were performed using various biochemical and cellular techniques. RESULTS: Through immunohistochemical analysis we observed significantly reduced levels of PP2A-A and -B'γ subunits (P<0.001 and P=0.0002) in PCa specimens compared with benign prostate. Contemporarily, there was no significant difference in PP2A-C subunit expression between benign and malignant tissues. Similar to the expression pattern observed in tissues, the endogenous levels of PP2A-A and B'γ subunits were abrogated from the low metastatic to high metastatic and AD to AI cell line models, without any change in the catalytic subunit expression. Furthermore, using in vitro studies we demonstrated that PP2A-Aα scaffold subunit has a role in dampening AKT, ß-catenin, and FAK (focal adhesion kinase) signalling. CONCLUSION: We conclude that loss of expression of scaffold and regulatory subunits of PP2A is responsible for its altered function during PCa pathogenesis.


Assuntos
Adenocarcinoma/patologia , Proteína Oncogênica v-akt/metabolismo , Neoplasias da Próstata/patologia , Proteína Fosfatase 2/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo/fisiologia , Ativação Enzimática/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Masculino , Modelos Biológicos , Metástase Neoplásica , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteína Fosfatase 2/metabolismo , Proteína Fosfatase 2/fisiologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transdução de Sinais/genética
6.
Oncogene ; 32(11): 1384-95, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-22614008

RESUMO

Despite evidence that long-term smoking is the leading risk factor for pancreatic malignancies, the underlying mechanism(s) for cigarette-smoke (CS)-induced pancreatic cancer (PC) pathogenesis has not been well established. Our previous studies revealed an aberrant expression of the MUC4 mucin in PC as compared with the normal pancreas, and its association with cancer progression and metastasis. Interestingly, here we explore a potential link between MUC4 expression and smoking-mediated PC pathogenesis and report that both cigarette smoke extract and nicotine, which is the major component of CS, significantly upregulates MUC4 in PC cells. This nicotine-mediated MUC4 overexpression was via the α7 subunit of nicotinic acetylcholine receptor (nAChR) stimulation and subsequent activation of the JAK2/STAT3 downstream signaling cascade in cooperation with the MEK/ERK1/2 pathway; this effect was blocked by the α7nAChR antagonists, α-bungarotoxin and mecamylamine, and by specific siRNA-mediated STAT3 inhibition. In addition, we demonstrated that nicotine-mediated MUC4 upregulation promotes the PC cell migration through the activation of the downstream effectors, such as HER2, c-Src and FAK; this effect was attenuated by shRNA-mediated MUC4 abrogation, further implying that these nicotine-mediated pathological effects on PC cells are MUC4 dependent. Furthermore, the in vivo studies showed a marked increase in the mean pancreatic tumor weight (low dose (100 mg/m(3) total suspended particulate (TSP)), P=0.014; high dose (247 mg/m(3) TSP), P=0.02) and significant tumor metastasis to various distant organs in the CS-exposed mice, orthotopically implanted with luciferase-transfected PC cells, as compared with the sham controls. Moreover, the CS-exposed mice had elevated levels of serum cotinine (low dose, 155.88±35.96 ng/ml; high dose, 216.25±29.95 ng/ml) and increased MUC4, α7nAChR and pSTAT3 expression in the pancreatic tumor tissues. Altogether, our findings revealed for the first time that CS upregulates the MUC4 mucin in PC via the α7nAChR/JAK2/STAT3 downstream signaling cascade, thereby promoting metastasis of PC.


Assuntos
Carcinoma Ductal Pancreático/induzido quimicamente , Carcinoma Ductal Pancreático/patologia , Mucina-4/genética , Nicotina/toxicidade , Neoplasias Pancreáticas/induzido quimicamente , Neoplasias Pancreáticas/patologia , Receptores Nicotínicos/fisiologia , Fumaça/efeitos adversos , Produtos do Tabaco , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos SCID , Modelos Biológicos , Mucina-4/metabolismo , Metástase Neoplásica , Transplante de Neoplasias/patologia , Nicotina/farmacologia , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Produtos do Tabaco/toxicidade , Transplante Heterólogo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Receptor Nicotínico de Acetilcolina alfa7
7.
Oncogene ; 31(7): 805-17, 2012 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21785467

RESUMO

MUC16/CA125 is a tumor marker currently used in clinics for the follow-up of patients with ovarian cancer. However, MUC16 expression is not entirely restricted to ovarian malignancies and has been reported in other cancers including breast cancer. Although it is well established as a biomarker, function of MUC16 in cancer remains to be elucidated. In the present study, we investigated the role of MUC16 in breast cancer and its underlying mechanisms. Interestingly, our results showed that MUC16 is overexpressed in breast cancer tissues whereas not expressed in non-neoplastic ducts. Further, stable knockdown of MUC16 in breast cancer cells (MDA MB 231 and HBL100) resulted in significant decrease in the rate of cell growth, tumorigenicity and increased apoptosis. In search of a mechanism for breast cancer cell proliferation we found that MUC16 interacts with the ezrin/radixin/moesin domain-containing protein of Janus kinase (JAK2) as demonstrated by the reciprocal immunoprecipitation method. These interactions mediate phosphorylation of STAT3 (Tyr705), which might be a potential mechanism for MUC16-induced proliferation of breast cancer cells by a subsequent co-transactivation of transcription factor c-Jun. Furthermore, silencing of MUC16 induced G2/M arrest in breast cancer cells through downregulation of Cyclin B1 and decreased phosphorylation of Aurora kinase A. This in turn led to enhanced apoptosis in the MUC16-knockdown breast cancer cells through Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated extrinsic apoptotic pathway with the help of c-Jun N-terminal kinase signaling. Collectively, our results suggest that MUC16 has a dual role in breast cancer cell proliferation by interacting with JAK2 and by inhibiting the apoptotic process through downregulation of TRAIL.


Assuntos
Apoptose , Neoplasias da Mama/metabolismo , Antígeno Ca-125/metabolismo , Proliferação de Células , Janus Quinase 2/metabolismo , Proteínas de Membrana/metabolismo , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Antígeno Ca-125/genética , Divisão Celular , Linhagem Celular Tumoral , Ciclina B1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Feminino , Fase G2 , Humanos , Imuno-Histoquímica , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Janus Quinase 2/genética , Proteínas de Membrana/genética , Modelos Biológicos , Fosforilação , Ligação Proteica , Interferência de RNA , Fator de Transcrição STAT3/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
8.
Br J Cancer ; 104(6): 1038-48, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21364589

RESUMO

BACKGROUND: Pancreatic cancer (PC) harbours an activated point mutation (Kras(G12D)) in the Kras proto-oncogene that has been demonstrated to promote the development of PC. METHODS: This study was designed to investigate the effect of the oncogenic Kras(G12D) allele on aggressiveness and metastatic potential of PC cells. We silenced the oncogenic Kras(G12D) allele expression in CD18/HPAF and ASPC1 cell lines by stable expression of shRNA specific to the Kras(G12D)allele. RESULTS: The Kras(G12D) knockdown cells exhibited a significant decrease in motility (P<0.0001), invasion (P<0.0001), anchorage-dependent (P<0.0001) and anchorage-independent growth (P<0.0001), proliferation (P<0.005) and an increase in cell doubling time (P<0.005) in vitro and a decrease in the incidence of metastases upon orthotopic implantation into nude mice. The knockdown of the Kras(G12D) allele led to a significant increase in the expression of E-cadherin (mRNA and protein) both in vitro and in vivo. This was associated with a decrease in the expression of phoshpo-ERK-1/2, NF-κB and MMP-9, and transcription factors such as δEF1, Snail and ETV4. Furthermore, the expression of several proteins involved in cell survival, invasion and metastasis was decreased in the Kras(G12D) knockdown cells. CONCLUSIONS: The results of this study suggest that the Kras(G12D) allele promotes metastasis in PC cells partly through the downregulation of E-cadherin.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/patologia , Caderinas/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Substituição de Aminoácidos/genética , Substituição de Aminoácidos/fisiologia , Animais , Ácido Aspártico/genética , Caderinas/fisiologia , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicina/genética , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Mutação Puntual , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , RNA Interferente Pequeno/farmacologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/metabolismo
9.
Oncogene ; 29(42): 5741-54, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20697346

RESUMO

The acquisition of invasiveness in ovarian cancer (OC) is accompanied by the process of epithelial-to-mesenchymal transition (EMT). The MUC4 mucin is overexpressed in ovarian tumors and has a role in the invasiveness of OC cells. The present study was aimed at evaluating the potential involvement of MUC4 in the metastasis of OC cells by inducing EMT. Ectopic overexpression of MUC4 in OC cells (SKOV3-MUC4) resulted in morphological alterations along with a decreased expression of epithelial markers (E-cadherin and cytokeratin (CK)-18) and an increased expression of mesenchymal markers (N-cadherin and vimentin) compared with the control cells (SKOV3-vector). Also, pro-EMT transcription factors TWIST1, TWIST2 and SNAIL showed an upregulation in SKOV3-MUC4 cells. We further investigated the pathways upstream of N-cadherin, such as focal adhesion kinase (FAK), MKK7, JNK1/2 and c-Jun, which were also activated in the SKOV3-MUC4 cells compared with SKOV3-vector cells. Inhibition of phospho-FAK (pFAK) and pJNK1/2 decreased N-cadherin expression in the MUC4-overexpressing cells, which further led to a significant decrease in cellular motility. Knockdown of N-cadherin decreased the activation of extracellular signal-regulated kinase-1/2 (ERK1/2), AKT and matrix metalloproteinase 9 (MMP9), and inhibited the motility in the SKOV3-MUC4 cells. Upon in vivo tumorigenesis and metastasis analysis, the SKOV3-MUC4 cells produced significantly larger tumors and demonstrated a higher incidence of metastasis to distance organs (peritoneal wall, colon, intestine, stomach, lymph nodes, liver and diaphragm). Taken together, our study reveals a novel role for MUC4 in inducing EMT through the upregulation of N-cadherin and promoting metastasis of OC cells.


Assuntos
Transdiferenciação Celular/fisiologia , Mucina-4/metabolismo , Invasividade Neoplásica/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Transdução de Sinais/fisiologia , Animais , Caderinas/biossíntese , Linhagem Celular Tumoral , Células Epiteliais/patologia , Feminino , Imunofluorescência , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Immunoblotting , Imuno-Histoquímica , Mesoderma/patologia , Camundongos , Camundongos Nus , Microscopia Confocal , Invasividade Neoplásica/genética , Neoplasias Ovarianas/genética
10.
Minerva Med ; 100(5): 385-400, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19910891

RESUMO

Ovarian cancer, the most aggressive gynecologic cancer, is the foremost cause of death from gynecologic malignancies in the developed world. Over 90% of ovarian cancers arise from the surface epithelium, which are classified as epithelial ovarian cancer (EOC). EOCs can be categorized as serous, mucinous, endometrioid, clear cell, and transitional cell types. The molecular pathology of ovarian carcinomas is heterogeneous and involves various putative precursor lesions and multiple pathways of development. Furthermore, in another aspect, immune deficiencies that are present in the ovarian tumor environment enhance the progression of the tumor in the host. The presence of regulatory T cells, the inhibition of natural killer cytotoxic responses, the accumulation of myeloid suppressor cells in the tumor, deficiencies on interferon signaling, the secretion of cytokines that enhance tumor growth (i.e., IL-6, IL-10, CSF-1, TGF-b, TNF), and the expression of surface molecules (i.e., HLA-G, B7-H1, B7-H4, CD40, CD80) that have a role on immune suppression, are discussed in detail. The aim of this review is to provide insight of the evidence that supports the role of immunodeficiency in the progression of ovarian cancer and future directions for ovarian cancer therapies. It also discusses the genetic alterations in the subtypes of ovarian cancers.


Assuntos
Biomarcadores Tumorais/imunologia , Carcinoma/imunologia , Hospedeiro Imunocomprometido , Neoplasias Ovarianas/imunologia , Antígenos CD/imunologia , Antígeno B7-1/imunologia , Antígeno B7-H1 , Antígenos CD40/imunologia , Carcinoma/classificação , Carcinoma/genética , Carcinoma/patologia , Transformação Celular Neoplásica/imunologia , Citocinas/imunologia , Progressão da Doença , Feminino , Antígenos HLA/imunologia , Antígenos HLA-G , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Células Matadoras Naturais/imunologia , Neoplasias Ovarianas/classificação , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Linfócitos T/imunologia , Inibidor 1 da Ativação de Células T com Domínio V-Set
11.
Br J Cancer ; 99(3): 520-6, 2008 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-18665193

RESUMO

The mucin MUC4 is a high molecular weight transmembrane glycoprotein. It consists of a mucin-type subunit (MUC4alpha) and a transmembrane growth factor-like subunit (MUC4beta). The mucin MUC4 is overexpressed in many epithelial malignancies including ovarian cancer, suggesting a possible role in the pathogenesis of these cancers. In this study, we investigated the functional role of MUC4 in the human ovarian cancer cell line SKOV3. The mucin MUC4 was ectopically expressed by stable transfection, and its expression was examined by western blot and confocal microscopy analyses. The in vitro studies demonstrated an enhanced motility of MUC4-expressing SKOV3 cells compared with the vector-transfected cells. The mucin MUC4 expression was associated with apparent changes in actin organisation, leading to the formation of microspike, lammelopodia and filopodia-like cellular projections. An enhanced protein expression and activation of HER2, a receptor tyrosine kinase, was also seen, although no significant change was observed in HER-2 transcript levels in the MUC4-transfected SKOV3 cells. Reciprocal co-immunoprecipitation revealed an interaction of MUC4 with HER2. Further, the MUC4-overexpressing SKOV3 cells exhibited an increase in the phosphorylation of focal adhesion kinase (FAK), Akt and ERK, downstream effectors of HER2. Taken together, our findings demonstrate that MUC4 plays a role in ovarian cancer cell motility, in part, by altering actin arrangement and potentiating HER2 downstream signalling in these cells.


Assuntos
Mucinas/fisiologia , Neoplasias Ovarianas/patologia , Receptor ErbB-2/metabolismo , Transdução de Sinais , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Feminino , Humanos , Microscopia de Fluorescência , Mucina-4 , Metástase Neoplásica , Neoplasias Ovarianas/metabolismo , RNA Mensageiro/genética , Receptor ErbB-2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Oncogene ; 26(54): 7499-507, 2007 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-17599057

RESUMO

Genetic instabilities are believed to be one of the major causes of developing a cancer phenotype in humans. During the progression of cancer, aberrant expression of proteins, either owing to genetic (amplification, mutation and deletion) or epigenetic modifications (DNA methylation and histone deacetylation), contributes in different ways to the development of cancer. By differential screening analysis, an amplification of the 19q13 locus containing a novel pancreatic differentiation 2 (PD2) gene was identified. PD2 is the human homolog of the yeast RNA polymerase II-associated factor 1 (yPaf1) and is part of the human RNA polymerase II-associated factor (hPAF) complex. hPAF is comprised of five subunits that include PD2/hPaf1, parafibromin, hLeo1, hCtr9 and hSki8. This multifaceted complex was first identified in yeast (yPAF) and subsequently in Drosophila and human. Recent advances in the study on PAF have revealed various functions of the complex in human, which are similar to yPAF, including efficient transcription elongation, mRNA quality control and cell-cycle regulation. Although the precise function of this complex in cancer is not clearly known, some of its subunits have been linked to a malignant phenotype. Its core subunit, PD2/hPaf1, is amplified and overexpressed in many cancers. Further, an overexpression of PD2/hPaf1 results in the induction of a transformed phenotype, suggesting its possible involvement in tumorigenesis. The parafibromin subunit of the hPAF complex is a product of the HRPT-2 (hereditary hyperparathyroidism type 2) tumor suppressor gene, which is mutated in the germ line of hyperparathyroidism-jaw tumor patients. This review focuses on the functions of the PAF complex and its individual subunits, the interaction of the subunits with each other and/or with other molecules, and dysregulation of the complex, providing an insight into its potential involvement in the development of cancer.


Assuntos
Mapeamento Cromossômico , Neoplasias/genética , Proteínas Nucleares/genética , Oncogenes , RNA Polimerase II/genética , Linhagem Celular Tumoral , Instabilidade Genômica , Humanos , Neoplasias/enzimologia , Proteínas Nucleares/metabolismo , RNA Polimerase II/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
Oncogene ; 26(51): 7251-61, 2007 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-17525742

RESUMO

MUC4 is a transmembrane mucin, which is aberrantly expressed in pancreatic adenocarcinoma with no detectable expression in the normal pancreas. Here, we present a novel mechanism of IFN-gamma-induced expression of MUC4 in pancreatic cancer cells. Our studies highlight the upregulation of STAT-1 as a basis for MUC4 induction and demonstrate that its activation and upregulation by IFN-gamma are two distinct, albeit temporally integrated, signalling events that drive the selective induction of IRF-1 and MUC4, respectively, within a single cell system. The profile of interferon regulatory factor (IRF)-1 gene induction by IFN-gamma is consistent with its rapid transactivation by phospho-Y701-STAT-1. In contrast, the induction of the MUC4 mucin gene expression is relatively delayed, and occurs only in response to an increase in STAT-1 expression. A progressive binding of STAT-1 to various gamma-interferon-activated sequences (GAS) in the MUC4 promoter is observed in chromatin immunoprecipitation assay, indicating its direct association. Stimulation of STAT-1 expression by double-stranded polynucleotides or ectopic expression is shown to induce MUC4 expression, without Y701 phosphorylation of STAT-1. This effect is abrogated by short interfering RNA (siRNA)-mediated inhibition of STAT-1 expression, supporting further the relevance of STAT-1 in MUC4 regulation. In conclusion, our findings identify a novel mechanism for MUC4 regulation in pancreatic cancer cells and unfold new perspectives on the foundation of IFN-gamma-dependent gene regulation.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Interferon gama/farmacologia , Mucinas/genética , Neoplasias Pancreáticas/metabolismo , Fator de Transcrição STAT1/fisiologia , Regulação para Cima , Sequência de Bases , Linhagem Celular Tumoral , Humanos , Mucina-4 , Mucinas/biossíntese , Neoplasias Pancreáticas/patologia , Fosforilação , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...