Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
1.
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
2.
Br J Cancer ; 111(6): 1139-49, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25025965

RESUMO

BACKGROUND: Despite its promise as a highly useful therapy for pancreatic cancer (PC), the addition of external beam radiation therapy to PC treatment has shown varying success in clinical trials. Understanding PC radioresistance and discovery of methods to sensitise PC to radiation will increase patient survival and improve quality of life. In this study, we identified PC radioresistance-associated pathways using global, unbiased techniques. METHODS: Radioresistant cells were generated by sequential irradiation and recovery, and global genome cDNA microarray analysis was performed to identify differentially expressed genes in radiosensitive and radioresistant cells. Ingenuity pathway analysis was performed to discover cellular pathways and functions associated with differential radioresponse and identify potential small-molecule inhibitors for radiosensitisation. The expression of FDPS, one of the most differentially expressed genes, was determined in human PC tissues by IHC and the impact of its pharmacological inhibition with zoledronic acid (ZOL, Zometa) on radiosensitivity was determined by colony-forming assays. The radiosensitising effect of Zol in vivo was determined using allograft transplantation mouse model. RESULTS: Microarray analysis indicated that 11 genes (FDPS, ACAT2, AG2, CLDN7, DHCR7, ELFN2, FASN, SC4MOL, SIX6, SLC12A2, and SQLE) were consistently associated with radioresistance in the cell lines, a majority of which are involved in cholesterol biosynthesis. We demonstrated that knockdown of farnesyl diphosphate synthase (FDPS), a branchpoint enzyme of the cholesterol synthesis pathway, radiosensitised PC cells. FDPS was significantly overexpressed in human PC tumour tissues compared with healthy pancreas samples. Also, pharmacologic inhibition of FDPS by ZOL radiosensitised PC cell lines, with a radiation enhancement ratio between 1.26 and 1.5. Further, ZOL treatment resulted in radiosensitisation of PC tumours in an allograft mouse model. CONCLUSIONS: Unbiased pathway analysis of radioresistance allowed for the discovery of novel pathways associated with resistance to ionising radiation in PC. Specifically, our analysis indicates the importance of the cholesterol synthesis pathway in PC radioresistance. Further, a novel radiosensitiser, ZOL, showed promising results and warrants further study into the universality of these findings in PC, as well as the true potential of this drug as a clinical radiosensitiser.


Assuntos
Adenocarcinoma/radioterapia , Colesterol/biossíntese , Difosfonatos/farmacologia , Geraniltranstransferase/genética , Imidazóis/farmacologia , Neoplasias Pancreáticas/radioterapia , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animais , Linhagem Celular Tumoral , DNA Complementar/análise , Difosfonatos/uso terapêutico , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Geraniltranstransferase/análise , Humanos , Imidazóis/uso terapêutico , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Tolerância a Radiação/genética , Radiossensibilizantes/uso terapêutico , Ácido Zoledrônico
3.
Br J Cancer ; 108(5): 1079-91, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-23449353

RESUMO

BACKGROUND: Overexpression of macrophage inhibitory cytokine-1 (MIC-1) frequently occurs during the progression of prostate cancer (PC) to androgen-independent (AI) and metastatic disease states and is associated with a poor outcome of patients. METHODS: The gain- and loss-of-function analyses of MIC-1 were performed to establish its implications for aggressive and chemoresistant phenotypes of metastatic and AI PC cells and the benefit of its downregulation for reversing docetaxel resistance. RESULTS: The results have indicated that an enhanced level of secreted MIC-1 protein in PC3 cells is associated with their acquisition of epithelial-mesenchymal transition features and higher invasive capacity and docetaxel resistance. Importantly, the downregulation of MIC-1 in LNCaP-LN3 and PC3M-LN4 cells significantly decreased their invasive capacity and promoted the antiproliferative, anti-invasive and mitochrondrial- and caspase-dependent apoptotic effects induced by docetaxel. The downregulation of MIC-1 in PC3M-LN4 cells was also effective in promoting the cytotoxic effects induced by docetaxel on the side population (SP) endowed with stem cell-like properties and the non-SP cell fraction from PC3M-LN4 cells. CONCLUSION: These data suggest that the downregulation of MIC-1 may constitute a potential therapeutic strategy for improving the efficacy of current docetaxel-based chemotherapies, eradicating the total mass of PC cells and thereby preventing disease relapse and the death of PC patients.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Fator 15 de Diferenciação de Crescimento/genética , Neoplasias da Próstata/tratamento farmacológico , Taxoides/farmacologia , Androgênios , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Docetaxel , Regulação para Baixo , Transição Epitelial-Mesenquimal , Fator 15 de Diferenciação de Crescimento/metabolismo , Humanos , Masculino , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia
4.
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
5.
Br J Cancer ; 107(3): 501-7, 2012 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-22782341

RESUMO

BACKGROUND: Our earlier reports demonstrated that membrane-bound semaphorin 5A (SEMA5A) is expressed in aggressive pancreatic cancer cells and tumours, and promotes tumour growth and metastasis. In this study, we examine whether (1) pancreatic cancer cells secrete SEMA5A and (2) that secreted SEMA5A modulates certain phenotypes associated with tumour progression, angiogenesis and metastasis through various other molecular factors and signalling proteins. METHODS AND RESULTS: In this study, we show that human pancreatic cancer cell lines secrete the extracellular domain (ECD) of SEMA5A (SEMA5A-ECD) and overexpression of mouse Sema5A-ECD in Panc1 cells (not expressing SEMA5A; Panc1-Sema5A-ECD; control cells - Panc1-control) significantly increases their invasion in vitro via enhanced ERK phosphorylation. Interestingly, orthotopic injection of Panc1-Sema5A-ECD cells into athymic nude mice results in a lower primary tumour burden, but enhances the micrometastases to the liver as compared with Panc1-control cells. Furthermore, there is a significant increase in proliferation of endothelial cells treated with conditioned media (CM) from Panc1-Sema5A-ECD cells and a significant increase in microvessel density in Panc1-Sema5A-ECD orthotopic tumours compared with those from Panc1-control cells, suggesting that the increase in liver micrometastases is probably due to increased tumour angiogenesis. In addition, our data demonstrate that this increase in endothelial cell proliferation by Sema5A-ECD is mediated through the angiogenic molecules - interleukin-8 and vascular endothelial growth factor. CONCLUSION: Taken together, these results suggest that a bioactive, secreted form of Sema5A-ECD has an intriguing and potentially important role in its ability to enhance pancreatic tumour invasiveness, angiogenesis and micrometastases.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Indutores da Angiogênese/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Progressão da Doença , Células Endoteliais/metabolismo , Células Endoteliais/patologia , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Neoplasias Hepáticas/secundário , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Micrometástase de Neoplasia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Proteínas do Tecido Nervoso/genética , Neoplasias Pancreáticas/genética , Fosforilação/genética , Semaforinas , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Minerva Gastroenterol Dietol ; 58(4): 283-97, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23207606

RESUMO

Pancreatic cancer (PC) remains a complex malignancy with the worst prognosis, lack of early diagnostic symptoms and resistance to conventional chemo- and radiotherapies. A better understanding of the etiology and early developmental events of PC requires profound attention. The evolution of fully blown PC from initial pancreatic injury is a multi-factorial phenomenon with a series of sequential events. The initial acute infection or tissue damage triggers inflammation that, in conjunction with innate immunity, establishes a state of homeostasis to limit harm to the body. Recurrent pancreatic injuries due to genetic susceptibility, smoking, unhealthy diet, and alcohol abuse induces a pro-inflammatory milieu, consisting of various types of immune cells, cytokines, chemokines, growth factors and restructured extracellular matrix, leading to prolonged inflammatory/chronic conditions. Cells having sustained DNA damage and/or mutagenic assault take advantage of this prolonged inflammatory response and aid in the initiation and development of neoplastic/fibrotic events. Eventually, many tumor-stromal interactions result in a chaotic environment accompanied by a loss of immune surveillance and repair response, thereby leading to PC. A better understanding of the inflammatory markers defining this "injury-inflammation-cancer" pathway would help to identify novel molecular targets for early screening and therapeutic intervention for this lethal malignancy.


Assuntos
Consumo de Bebidas Alcoólicas/efeitos adversos , Quimiocinas/sangue , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/etiologia , Pancreatite Crônica/complicações , Fumar/efeitos adversos , Biomarcadores/sangue , Índice de Massa Corporal , Complicações do Diabetes/fisiopatologia , Diagnóstico Precoce , Comportamento Alimentar , Humanos , Imunidade Inata , Inflamação/complicações , Obesidade/complicações , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/terapia , Pancreatite Crônica/sangue , Prognóstico , Receptores de Fatores de Crescimento do Endotélio Vascular/sangue , Fatores de Risco
7.
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
8.
Br J Cancer ; 101(7): 1155-61, 2009 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-19738614

RESUMO

BACKGROUND: A major obstacle to the successful management of pancreatic cancer is to acquire resistance to the existing chemotherapeutic agents. Resistance to gemcitabine, the standard first-line chemotherapeutic agent for advanced and metastatic pancreatic cancer, is mainly attributed to an altered apoptotic threshold in the pancreatic cancer. The MUC4 transmembrane glycoprotein is aberrantly overexpressed in the pancreatic cancer and recently, has been shown to increase pancreatic tumour cell growth by the inhibition of apoptosis. METHODS: Effect of MUC4 on pancreatic cancer cells resistance to gemcitabine was studied in MUC4-expressing and MUC4-knocked down pancreatic cancer cell lines after treatment with gemcitabine by Annexin-V staining, DNA fragmentation assay, assessment of mitochondrial cytochrome c release, immunoblotting and co-immunoprecipitation techniques. RESULTS: Annexin-V staining and DNA fragmentation experiment demonstrated that MUC4 protects CD18/HPAF pancreatic cancer cells from gemcitabine-induced apoptosis. In concert with these results, MUC4 also attenuated mitochondrial cytochrome c release and the activation of caspase-9. Further, our results showed that MUC4 exerts anti-apoptotic function through HER2/extracellular signal-regulated kinase-dependent phosphorylation and inactivation of the pro-apoptotic protein Bad. CONCLUSION: Our results elucidate the function of MUC4 in imparting resistance to pancreatic cancer cells against gemcitabine through the activation of anti-apoptotic pathways and, thereby, promoting cell survival.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Desoxicitidina/análogos & derivados , Mucina-4/fisiologia , Neoplasias Pancreáticas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Citosol/metabolismo , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Neoplasias Pancreáticas/patologia , Receptor ErbB-2/fisiologia , Transdução de Sinais , Proteína de Morte Celular Associada a bcl/metabolismo , Proteína bcl-X/metabolismo , Gencitabina
9.
Panminerva Med ; 51(2): 57-79, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19776709

RESUMO

The fascinating discovery of tissue-resident adult stem/progenitor cells in recent years led to an explosion of interest in the development of novel stem cell-based therapies for improving the regenerative capacity of these endogenous immature cells or transplanted cells for the repair of damaged and diseased tissues. In counterbalance, a growing body of evidence has revealed that the changes in phenotypic and functional properties of human adult stem/progenitor cells may occur during chronological aging and have severe pathological consequences. Especially, intense oxidative and metabolic stress and chronic inflammation, enhanced telomere attrition and defects in DNA repair mechanisms may lead to severe DNA damages and genomic instability in adult stem/progenitor cells with advancing age that may in turn trigger their replicative senescence and/or programmed cell death. Moreover, the changes in the intrinsic and extrinsic factors involved in the stringent control of self-renewal and multilineage differentiation capacities of these regenerative cells, including deregulated signals from the aged niche, may also contribute to their dysfunctions or loss during chronological aging. This age-associated decline in the regenerative capacity and number of functional adult stem/progenitor cells may increase the risk to develop certain diseases. At opposed end, the telomerase reactivation and accumulation of genetic alterations leading to a down-regulation of numerous tumor suppressor genes concomitant with the enhanced expression of diverse oncogenic products may result in their malignant transformation into cancer-initiating cells. Therefore, the rescue or replacement of aged and dysfunctional endogenous adult stem/progenitor cells or molecular targeting of their malignant counterpart, cancer stem/progenitor cells may constitute potential anti-aging and cancer therapies. These therapeutic strategies could be used for treating diverse devastating premature aging and age-related disorders including hematopoietic and immune disorders, heart failure and cardiovascular diseases, neurodegenerative, muscular and gastrointestinal diseases, atherosclerosis and aggressive and lethal cancers.


Assuntos
Células-Tronco Adultas/patologia , Envelhecimento/patologia , Senescência Celular , Transplante de Células-Tronco , Células-Tronco Adultas/transplante , Animais , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Transformação Celular Neoplásica/patologia , Dano ao DNA , Células-Tronco Hematopoéticas/patologia , Humanos , Células-Tronco Neoplásicas/patologia , Estresse Oxidativo , Fenótipo , Regeneração , Transplante de Células-Tronco/efeitos adversos
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.
Gut ; 57(10): 1456-68, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18791122

RESUMO

Recent progress on pancreatic stem/progenitor cell research has revealed that the putative multipotent pancreatic stem/progenitor cells and/or more committed beta cell precursors may persist in the pancreatic gland in adult life. The presence of immature pancreatic cells with stem cell-like properties offers the possibility of stimulating their in vivo expansion and differentiation or to use their ex vivo expanded progenies for beta cell replacement-based therapies for type 1 or 2 diabetes mellitus in humans. In addition, the transplantation of either insulin-producing beta cells derived from embryonic, fetal and other tissue-resident adult stem/progenitor cells or genetically modified adult stem/progenitor cells may also constitute alternative promising therapies for treating diabetic patients. The genetic and/or epigenetic alterations in putative pancreatic adult stem/progenitor cells and/or their early progenies may, however, contribute to their acquisition of a dysfunctional behaviour as well as their malignant transformation into pancreatic cancer stem/progenitor cells. More particularly, the activation of distinct tumorigenic signalling cascades, including the hedgehog, epidermal growth factor-epidermal growth factor receptor (EGF-EGFR) system, wingless ligand (Wnt)/beta-catenin and/or stromal cell-derived factor-1 (SDF-1)-CXC chemokine receptor 4 (CXCR4) pathways may play a major role in the sustained growth, survival, metastasis and/or drug resistance of pancreatic cancer stem/progenitor cells and their further differentiated progenies. The combination of drugs that target the oncogenic elements in pancreatic cancer stem/progenitor cells and their microenvironment, with the conventional chemotherapeutic regimens, could represent promising therapeutic strategies. These novel targeted therapies should lead to the development of more effective treatments of locally advanced and metastatic pancreatic cancers, which remain incurable with current therapies.


Assuntos
Transformação Celular Neoplásica/patologia , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/terapia , Neoplasias Pancreáticas/terapia , Transplante de Células-Tronco/métodos , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Humanos , Transplante das Ilhotas Pancreáticas/métodos , Transplante de Pâncreas/métodos
12.
Oncogene ; 26(1): 30-41, 2007 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-16799633

RESUMO

MUC4 mucin is a high molecular weight transmembrane glycoprotein that plays important roles in tumour biology. It is aberrantly expressed in pancreatic adenocarcinoma, while not being detectable in the normal pancreas. Previous studies have demonstrated that the cystic fibrosis transmembrane conductance regulator (CFTR), a chloride channel that is defective in CF, is implicated in multiple cellular functions, including gene regulation. In the present study, using a CFTR-defective pancreatic cancer cell line and its derived subline expressing functional CFTR, we report that MUC4 expression is negatively regulated by CFTR. Short-interfering RNA (siRNA)-mediated silencing of CFTR also leads to an increased expression of MUC4. Additionally, our results suggest that CFTR-mediated regulation of MUC4 is cell density-dependent and is achieved by transcriptional and posttranslational mechanisms. Moreover, in a panel of pancreatic cancer cell lines and normal pancreas, we observed that CFTR was downregulated in pancreatic cancer cells and negatively correlated with MUC4 in most cases. An aberrant expression of MUC4 was also detected in the CF pancreas. Downregulation of CFTR in pancreatic adenocarcinoma and its inverse association with the tumour-linked mucin, MUC4, indicate novel function(s) of CFTR in pancreatic tumour biology and suggest the implication of new signalling pathway(s) in MUC4 regulation.


Assuntos
Adenocarcinoma/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Mucinas/genética , Neoplasias Pancreáticas/metabolismo , Processamento de Proteína Pós-Traducional , Adenocarcinoma/patologia , Sequência de Bases , Northern Blotting , Linhagem Celular Tumoral , Primers do DNA , Humanos , Mucina-4 , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa
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
14.
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
15.
Br J Cancer ; 98(10): 1675-81, 2008 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-18475301

RESUMO

Alterations in epithelial mucin expression are associated with carcinogenesis, but there are few data in biliary tract cancer (BTC). In pancreatic malignancy, MUC4 is a diagnostic and prognostic tumour marker, whereas MUC5AC has been proposed as a sensitive serological marker for BTC. We assessed MUC4 and MUC5AC expression in (i) prospectively collected bile and serum specimens from 72 patients with biliary obstruction (39 BTC) by real-time reverse transcriptase-PCR (qPCR) and western blot analysis, and (ii) 79 archived biliary tissues (69 BTC) by immunohistochemistry. In bile, MUC4 protein was detected in 27% of BTC and 29% of primary sclerosing cholangitis (PSC) cases, but not in other benign and malignant biliary diseases (P<0.01 and P=0.06). qPCR revealed a 1.9-fold increased MUC4 mRNA expression in BTC patients' bile compared with benign disease. In archived tissues, MUC4 protein was detected in 37% of BTC but in none of the benign samples (P=0.03). In serum, MUC5AC was found exclusively in BTC and PSC sera (44% and 13%, respectively; P<0.001 for BTC vs non-BTC) and correlated negatively with BTC survival. Biliary MUC4 and serum MUC5AC are highly specific tumour-associated mucins that may be useful in the diagnosis and formulation of therapeutic strategies in BTC.


Assuntos
Bile/metabolismo , Neoplasias do Sistema Biliar/metabolismo , Biomarcadores Tumorais/metabolismo , Mucinas/sangue , Mucinas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias do Sistema Biliar/sangue , Neoplasias do Sistema Biliar/patologia , Biomarcadores Tumorais/sangue , Western Blotting , Feminino , Seguimentos , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Mucina-5AC , Mucina-4 , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Estudos Prospectivos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Br J Cancer ; 98(9): 1540-7, 2008 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-18392050

RESUMO

Pancreatic cancer is a highly lethal malignancy with a dismal 5-year survival of less than 5%. The scarcity of early biomarkers has considerably hindered our ability to launch preventive measures for this malignancy in a timely manner. Neutrophil gelatinase-associated lipocalin (NGAL), a 24-kDa glycoprotein, was reported to be upregulated nearly 27-fold in pancreatic cancer cells compared to normal ductal cells in a microarray analysis. Given the need for biomarkers in the early diagnosis of pancreatic cancer, we investigated the expression of NGAL in tissues with the objective of examining if NGAL immunostaining could be used to identify foci of pancreatic intraepithelial neoplasia, premalignant lesions preceding invasive cancer. To examine a possible correlation between NGAL expression and the degree of differentiation, we also analysed NGAL levels in pancreatic cancer cell lines with varying grades of differentiation. Although NGAL expression was strongly upregulated in pancreatic cancer, and moderately in pancreatitis, only a weak expression could be detected in the healthy pancreas. The average composite score for adenocarcinoma (4.26+/-2.44) was significantly higher than that for the normal pancreas (1.0) or pancreatitis (1.0) (P<0.0001). Further, although both well- and moderately differentiated pancreatic cancer were positive for NGAL, poorly differentiated adenocarcinoma was uniformly negative. Importantly, NGAL expression was detected as early as the PanIN-1 stage, suggesting that it could be a marker of the earliest premalignant changes in the pancreas. Further, we examined NGAL levels in serum samples. Serum NGAL levels were above the cutoff for healthy individuals in 94% of pancreatic cancer and 62.5% each of acute and chronic pancreatitis samples. However, the difference between NGAL levels in pancreatitis and pancreatic cancer was not significant. A ROC curve analysis revealed that ELISA for NGAL is fairly accurate in distinguishing pancreatic cancer from non-cancer cases (area under curve=0.75). In conclusion, NGAL is highly expressed in early dysplastic lesions in the pancreas, suggesting a possible role as an early diagnostic marker for pancreatic cancer. Further, serum NGAL measurement could be investigated as a possible biomarker in pancreatitis and pancreatic adenocarcinoma.


Assuntos
Proteínas de Fase Aguda/análise , Adenocarcinoma/diagnóstico , Biomarcadores Tumorais/análise , Carcinoma Ductal Pancreático/diagnóstico , Lipocalinas/análise , Neoplasias Pancreáticas/diagnóstico , Proteínas Proto-Oncogênicas/análise , Proteínas de Fase Aguda/genética , Adenocarcinoma/sangue , Adenocarcinoma/química , Adulto , Idoso , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Western Blotting , Carcinoma Ductal Pancreático/sangue , Carcinoma Ductal Pancreático/química , Linhagem Celular Tumoral , Diagnóstico Precoce , Ensaio de Imunoadsorção Enzimática , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Lipocalina-2 , Lipocalinas/sangue , Lipocalinas/genética , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/química , Proteínas Proto-Oncogênicas/sangue , Proteínas Proto-Oncogênicas/genética , RNA Neoplásico/análise , Curva ROC , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Panminerva Med ; 50(1): 3-18, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18427384

RESUMO

The rapid progression from aggressive primary cancers into locally advanced and invasive and/or metastatic diseases remains a big obstacle for an early diagnosis and curative therapeutic intervention for cancer patients. The late-stage leukemias and disseminated and metastatic sarcomas, melanomas, brain tumors and epithelial cancers are the devastating diseases associated with a high rate of recurrence after treatment with the conventional clinical therapies including surgery, ionizing radiation, hormonal therapy and systemic chemotherapy, which generally lead to the death of patients. Therefore, the establishment of the molecular events underlying cancer initiation and progression into locally invasive and metastatic diseases is of major interest in basic cancer research as well as for the development of new effective clinical therapeutic options against the recurrent and lethal cancers. Recent advances have led to the identification of specific oncogenic products that are implicated in the malignant transformation of adult stem/progenitor cells into leukemic or tumorigenic and migrating cancer stem/progenitor cells during cancer progression. Of therapeutic interest, the molecular targeting of deregulated signaling elements in cancer stem/progenitor cells and their local microenvironment represents a new potential strategy for the development of more effective clinical treatments against aggressive cancers. Particularly, the combined use of chemotherapeutic drugs to eradicate cancer-initiating cells with hematopoietic stem cell or genetically-modified stem cell transplant is emerging as potential cancer treatments that hold great promise in the area of clinical cancer research. These targeting and stem cell-based therapies may offer the ultimate hope for treating and even curing the patients diagnosed with locally advanced cancers at high risk of recurrence, metastatic and/or relapsed cancers in the clinics.


Assuntos
Recidiva Local de Neoplasia/terapia , Neoplasias/terapia , Células-Tronco Neoplásicas/patologia , Transplante de Células-Tronco , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Progressão da Doença , Humanos , Leucemia/patologia , Leucemia/terapia , Melanoma/patologia , Melanoma/terapia , Recidiva Local de Neoplasia/patologia , Neoplasias/patologia , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Epiteliais e Glandulares/terapia , Sarcoma/patologia , Sarcoma/terapia
18.
Oncogene ; 25(23): 3247-57, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16491129

RESUMO

The 19q13 amplicon in pancreatic cancer cells contains a novel pancreatic differentiation 2 (PD2) gene (accession number AJ401156), which was identified by differential screening analysis. PD2 is the human homologue of the RNA polymerase II-associated factor 1 (hPaf1). In yeast, Paf1 is part of the transcription machinery, acting as a docking protein in between the complexes Rad6-Bre1, COMPASS-Dot1p, and the phosphorylated carboxyl terminal domain of the RNA polymerase II. As such, Paf1 is directly involved in transcription elongation via histone H2B ubiquitination and histone H3 methylation. The PD2 sequence is highly conserved from Drosophila to humans with up to 98% identity between rodent and human, suggesting the functional importance of PD2/hPaf1 to maintain cellular homeostasis. PD2 is a modular protein composed of RNA recognition motif, DEAD-boxes, an aspartic/serine (DS)-domain, a regulator of the chromosome condensation domain and myc-type helix-loop-helix domains. Our results further showed that PD2 is a nuclear 80 kDa protein, which interacts with RNA polymerase II. In addition, we have demonstrated that the overexpression of PD2 in the NIH 3T3 cells result in enhanced growth rates in vitro and tumor formation in vivo. Altogether, this paper presents strong evidence that the overexpression of PD2/hPaf1 is involved in cancer development.


Assuntos
Transformação Celular Neoplásica/metabolismo , Cromossomos Humanos Par 19 , Amplificação de Genes , Proteínas Nucleares/fisiologia , RNA Polimerase II/metabolismo , Homologia de Sequência de Aminoácidos , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Dados de Sequência Molecular , Células NIH 3T3 , Proteínas Nucleares/genética , Alinhamento de Sequência , Fatores de Transcrição
19.
Clin Pharmacol Ther ; 82(3): 252-64, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17671448

RESUMO

Basic and clinical research accomplished during the last few years on embryonic, fetal, amniotic, umbilical cord blood, and adult stem cells has constituted a revolution in regenerative medicine and cancer therapies by providing the possibility of generating multiple therapeutically useful cell types. These new cells could be used for treating numerous genetic and degenerative disorders. Among them, age-related functional defects, hematopoietic and immune system disorders, heart failures, chronic liver injuries, diabetes, Parkinson's and Alzheimer's diseases, arthritis, and muscular, skin, lung, eye, and digestive disorders as well as aggressive and recurrent cancers could be successfully treated by stem cell-based therapies. This review focuses on the recent advancements in adult stem cell biology in normal and pathological conditions. We describe how these results have improved our understanding on critical and unique functions of these rare sub-populations of multipotent and undifferentiated cells with an unlimited self-renewal capacity and high plasticity. Finally, we discuss some major advances to translate the experimental models on ex vivo and in vivo expanded and/or differentiated stem cells into clinical applications for the development of novel cellular therapies aimed at repairing genetically altered or damaged tissues/organs in humans. A particular emphasis is made on the therapeutic potential of different tissue-resident adult stem cell types and their in vivo modulation for treating and curing specific pathological disorders.


Assuntos
Neoplasias/terapia , Medicina Regenerativa/tendências , Transplante de Células-Tronco , Células-Tronco/fisiologia , Células-Tronco Adultas/fisiologia , Animais , Transplante de Medula Óssea , Células-Tronco Embrionárias/fisiologia , Células-Tronco Fetais/fisiologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Regeneração/fisiologia , Células Estromais/transplante
20.
Ann Oncol ; 18(10): 1605-19, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17355951

RESUMO

In this review, we describe the critical functions assumed by the interplay of epidermal growth factor, hedgehog, Wnt/beta-catenin, tumor growth factor-beta and integrin signaling cascades in tumorigenic and migrating cancer progenitor cells and activated stromal cells during carcinogenesis. These growth factors provide an important role for the sustained growth and survival of tumorigenic cancer progenitor cells and their progeny by up-regulating numerous mitotic and antiapoptotic signaling cascades. Furthermore, these potent morphogens may cooperate for inducing the molecular events associated with the epithelial-mesenchymal program in cancer cells including the alterations in epithelial cell shape and motility through the dissociation of intercellular adherens junctions. Of therapeutic interest, new strategies for the development of more effective clinical treatments against the locally aggressive and invasive cancers based on the molecular targeting of deregulated signaling elements in tumorigenic and migrating cancer cells and their local microenvironment are also described.


Assuntos
Transformação Celular Neoplásica , Células Epiteliais/patologia , Substâncias de Crescimento/fisiologia , Mesoderma/patologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Células-Tronco Neoplásicas/fisiologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proliferação de Células , Sobrevivência Celular , Citocinas/fisiologia , Fator de Crescimento Epidérmico/fisiologia , Receptores ErbB/fisiologia , Proteínas Hedgehog/fisiologia , Humanos , Integrinas/fisiologia , Metástase Neoplásica , Células-Tronco Neoplásicas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , beta Catenina/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa