Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Lett ; 332(2): 346-58, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-21078544

RESUMO

Pancreatic cancer - here in particular pancreatic ductal adenocarcinoma (PDAC) - is still a highly therapy refractory disease. Amongst the mechanisms by which PDAC cells could escape any non-surgical therapy, anti-apoptotic protection seems to be the most relevant one. PDAC cells have acquired resistance to apoptotic stimuli such as death ligands (FasL, TRAIL) or anti-cancer drugs (gemcitabine) by a great number of molecular alterations either disrupting an apoptosis inducing signal or counteracting the execution of apoptosis. Thus, PDAC cells exhibit alterations in the EGFR/MAPK/Ras/raf1-, PI3K/Akt-, TRAIL/TRAF2-, or IKK/NF-κB pathway accompanied by deregulations in the expression of apoptosis regulators such as cIAP, Bcl2, XIAP or survivin. Along with protection against apoptosis, PDAC cells also overexpress histone deacetylases (HDACs) giving rise to epigenetic patterns of chemoresistance and to acetylation of other regulatory proteins, as well. With respect to the multitude of anti-apoptotic pathways, a great number of molecular targets might be of high potential in novel therapy strategies. Thus, natural compounds as well as novel synthetic drugs are considered to be used in single or combined therapy of PDAC. A number of proteasome and HDAC inhibitors or selective inhibitors of IKK, EGFR, Akt and mTOR have been widely explored in preclinical settings and clinical studies. Even though these early studies encouraged an application in a clinical setting, most of the trials have been rather disappointing yet. Thus, new molecular targets and novel concepts of combination therapies need to get access into clinical trials - either in neoadjuvant/adjuvant or in palliative treatments.


Assuntos
Apoptose , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Animais , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Histona Desacetilases/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Inibidores de Proteassoma/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas ras/metabolismo , Gencitabina
2.
Int J Oncol ; 38(1): 257-66, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21109948

RESUMO

Members of the Slug/Snail family of transcription factors are thought to drive epithelial-mesenchymal-transition (EMT) in preneoplastic epithelial cells, thereby contributing to malignant transformation. One mediator in the EMT of pancreatic ductal adenocarcinoma (PDAC) cells and a potential target gene of Slug is the cellular adhesion molecule L1CAM. Using the human pancreatic ductal epithelial cell line H6c7 and the PDAC cell line Panc1, we could show that along with TGF-ß1-induced EMT, L1CAM expression is increased in a Slug- but not Snail-dependent fashion. Two E-box recognition motifs in the L1CAM promoter upstream of the most distal transcriptional start site could be verified by gel shift and supershift assay to interact with Slug. ChIP assays detected an increased interaction of Slug with both recognition motifs of the human L1CAM promoter in TGF-ß1-treated H6c7 cells, whereas binding of Snail was downregulated. Moreover, ChIP assays with Panc1 cells confirmed this interaction of Slug with the human L1CAM promoter and further detected an interaction of both recognition sites with RNA-polymerase II in a Slug-dependent fashion. Luciferase reporter gene assays using wild-type or single- and double-mutated variants of the L1CAM promoter confirmed transcriptional activation by Slug involving both recognition motifs. By demonstrating the direct transcriptional control of L1CAM expression through Slug during TGF-ß1-induced EMT of PDAC cells, our findings point to a novel mechanism by which Slug contributes quite early to tumorigenesis. Moreover, our study is the first one describing the control of the human L1CAM promoter in tumor cells.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Molécula L1 de Adesão de Célula Nervosa/biossíntese , Neoplasias Pancreáticas/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Humanos , Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transfecção , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/farmacologia
3.
Oncol Rep ; 24(4): 909-15, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20811670

RESUMO

The promigratory molecule L1CAM is overexpressed in various tumors, often representing an unfavorable prognostic marker. Recently, we identified L1CAM expression in pancreatic ductal adenocarcinoma (PDAC) cells accounting for chemoresistance and increased cell migration. Thus, the present study aims at further elucidating the role of L1CAM in a larger cohort of PDAC specimens including precursor lesions and metastasis. L1CAM expression was determined by immunohistochemistry in tissues of 123 patients including tissues of 110 primary PDACs, 15 lymph node metastases and 14 liver metastases. The immunohistochemical analyses revealed L1CAM expression in 92.7% of primary PDACs, 80% of lymph node metastases and 100% of liver metastases. Furthermore, we have investigated PDAC precursors, pancreatic intraepithelial neoplasia (PanIN) lesions, revealing a significant increase of L1CAM expression with the PanIN grade (6.4 and 6.8% in PanIN 1A and B, 35% in PanIN 2 and 20% in PanIN 3). The elevated expression of L1CAM already found in PanINs points to a role of L1CAM quite early in tumorigenesis of PDAC. Furthermore, its broad expression in primary tumors as well as in metastases of PDAC patients provide a rationale to further explore the value of L1CAM as a therapeutic target in the treatment of this highly malignant tumor.


Assuntos
Carcinoma in Situ/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Molécula L1 de Adesão de Célula Nervosa/biossíntese , Neoplasias Pancreáticas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/análise , Carcinoma in Situ/mortalidade , Carcinoma in Situ/patologia , Carcinoma Ductal Pancreático/mortalidade , Carcinoma Ductal Pancreático/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica/patologia , Estadiamento de Neoplasias , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Prognóstico
4.
Histopathology ; 56(4): 440-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20459551

RESUMO

AIMS: Undifferentiated (anaplastic) pancreatic cancer and undifferentiated pancreatic carcinoma with osteoclast-like giant cells (giant cell tumour) are rare variants of pancreatic ductal adenocarcinoma. Representing biologically highly aggressive neoplasms, they are frequently diagnosed at an advanced stage. The response to established chemo- or radiochemotherapeutic treatment regimens is poor, and undifferentiated pancreatic cancer generally has a dismal prognosis. As additional therapeutic options have not yet been investigated in undifferentiated pancreatic cancer, the aim was to analyse the expression of putative therapeutic targets that have shown promising results in various other neoplasms. METHODS AND RESULTS: Fifteen cases of undifferentiated pancreatic cancer (seven containing osteoclast-like giant cells) were investigated clinicopathologically and immunohistochemically for putative therapeutic targets. Whereas L1CAM, cyclooxygenase (COX)-2 and epidermal growth factor receptor (EGFR) were found to be significantly expressed in 80%, 93% and 87% of the investigated tumours, respectively, there was no substantial expression of c-kit (CD117) and there was no detectable expression of Her2/neu. CONCLUSIONS: The expression of L1CAM, COX-2 and EGFR in the majority of undifferentiated pancreatic carcinomas suggests that they might represent targets for adjuvant therapy in anaplastic pancreatic cancer. On the other hand, c-kit and Her2/neu seem to have no relevance for the therapy of these tumours.


Assuntos
Adenocarcinoma/metabolismo , Ciclo-Oxigenase 2/metabolismo , Receptores ErbB/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptor ErbB-2/metabolismo , Adenocarcinoma/patologia , Idoso , Receptores ErbB/biossíntese , Receptores ErbB/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Prognóstico , Estudos Retrospectivos
5.
Cancer Res ; 70(8): 3299-308, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20388803

RESUMO

The chemokine CXCL12/stromal cell-derived factor-1 and its receptor CXCR4 play a major role in tumor invasion, proliferation, and metastasis. Recently, CXCR7 was identified as a novel, alternate receptor for CXCL12 and CXCL11/I-TAC. Because both chemokines are expressed abundantly in human astrocytomas and glioblastomas, we investigated the occurrence and function of both receptors in astroglial tumors. In situ, CXCR7 is highly expressed on tumor endothelial, microglial, and glioma cells whereas CXCR4 has a much more restricted localization; CXCL12 is often colocalized with CXCR7. CXCR7 transcription in tumor homogenates increased with malignancy. In vitro, CXCR7 was highly expressed in all glioma cell lines investigated whereas CXCR4 was only scarcely transcribed on one of eight lines. In contrast, a tumor stem-like cell line preferentially expressed CXCR4 which diminished upon differentiation, whereas CXCR7 increased drastically. Stimulation of CXCR7-positive glioma cells (CXCR4- and CXCR3-negative) by CXCL12 induced transient phosphorylation of extracellular signal-regulated kinases Erk1/2, indicating that the receptor is functionally active. The phosphoinositide-specific phospholipase C inhibitor U73122 effectively inhibited Erk activation and suggests that the mitogen-activated protein kinase pathway is activated indirectly. Whereas proliferation and migration were little influenced, chemokine stimulation prevented camptothecin- and temozolomide-induced apoptosis. The selective CXCR7 antagonist CCX733 reduced the antiapoptotic effects of CXCL12 as shown by nuclear (Nicoletti) staining, caspase-3/7 activity assays, and cleavage of poly(ADP-ribose) polymerase-1. Thus, CXCR7 is a functional receptor for CXCL12 in astrocytomas/glioblastomas and mediates resistance to drug-induced apoptosis. Whereas CXCR7 is found on "differentiated" glioma cells, the alternate receptor CXCR4 is also localized on glioma stem-like cells.


Assuntos
Apoptose , Neoplasias Encefálicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Receptores CXCR/biossíntese , Receptores CXCR/química , Proteínas Reguladoras de Apoptose/farmacologia , Camptotecina/farmacologia , Movimento Celular , Proliferação de Células , Quimiocina CXCL12/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Ativação Enzimática , Estrenos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Pirrolidinonas/farmacologia , Temozolomida
6.
Exp Cell Res ; 316(9): 1553-66, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20184883

RESUMO

The transmembrane chemokine CX3CL1 and its receptor CX3CR1 are thought to be involved in the trafficking of immune cells during an immune response and in the pathology of various human diseases including cancer. However, little is known about the expression and function of CX3CR1 in human glioma-infiltrating microglia/macrophages (GIMs), representing the major cellular stroma component of highly malignant gliomas. Here, we show that CX3CR1 is overexpressed at both the mRNA and protein level in solid human astrocytomas of different malignancy grades and in glioblastomas. CX3CR1 was localized in ionized calcium-binding adapter molecule 1 (Iba1) and CD11b/c positive GIMs in situ as shown by fluorescence microscopy. In accordance with this, freshly isolated human GIM-enriched fractions separated by CD11b MACS technology displayed high Iba1 and CX3CR1 mRNA expression levels in vitro. Moreover, cultured human GIMs responded to CX3CL1-triggered activation of CX3CR1 with adhesion and migration in vitro. Besides an increase in motility, CX3CL1 also enhanced expression of matrix metalloproteases 2, 9, and 14 in GIM fractions in vitro. These data indicate that the CX3CL1/CX3CR1 system has a crucial tumor-promoting role in human glioblastomas via its impact on glioma-infiltrating immune subsets.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Macrófagos/metabolismo , Microglia/metabolismo , Receptores de Quimiocinas/metabolismo , Western Blotting , Encéfalo/citologia , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Receptor 1 de Quimiocina CX3C , Proteínas de Ligação ao Cálcio , Estudos de Casos e Controles , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Imunofluorescência , Glioma/genética , Glioma/patologia , Humanos , Macrófagos/patologia , Masculino , Metaloproteinases da Matriz/metabolismo , Proteínas dos Microfilamentos , Microglia/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Quimiocinas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esferoides Celulares/metabolismo
7.
Inflamm Bowel Dis ; 16(2): 320-331, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19714745

RESUMO

BACKGROUND: Inflammatory bowel diseases (IBDs) result from environmental and genetic factors and are characterized by an imbalanced immune response in the gut and deregulated activation of the transcription factor NF-kappaB. Addressing the potential role of gly96/iex-1 in the regulation of NF-kappaB in IBD, we used the dextran sodium sulfate (DSS) colitis model in mice in which the gly96/iex-1 gene had been deleted. METHODS: C57BL/6 mice of gly96/iex-1(-/-) or gly96/iex-1(+/+) genotype were treated continuously with 4% DSS (5 days) and repeatedly with 2% DSS (28 days) for inducing acute and chronic colitis, respectively. In addition to clinical and histological exploration, colon organ culture and bone marrow-derived cells (BMCs) were analyzed for chemo/cytokine expression and NF-kappaB activation. RESULTS: Compared to wildtype littermates, gly96/iex-1(-/-) mice exhibited an aggravated phenotype of both acute and chronic colitis, along with a greater loss of body weight and colon length. Colonic endoscopy revealed a higher degree of hyperemia, edema, and bleeding in gly96/iex-1(-/-) mice, and immunohistochemistry detected massive mucosal infiltration of leukocytes and marked histological changes. The expression of proinflammatory chemo- and cytokines was higher in the colon of DSS-treated gly96/iex-1(-/-) mice, and the NF-kappaB activation was enhanced particularly in the distal colon. In cultured BMCs from gly96/iex-1(-/-) mice, Pam(3)Cys(4) treatment induced expression of proinflammatory mediators to a higher degree than in gly96/iex-1(+/+) BMCs, along with greater NF-kappaB activation. CONCLUSIONS: Based on the observation that genetic ablation of gly96/iex-1 triggers intestinal inflammation in mice, we demonstrate for the first time that gly96/iex-1 exerts strong antiinflammatory activity via its NF-kappaB-counterregulatory effect.


Assuntos
Colite/genética , Proteínas Imediatamente Precoces/fisiologia , NF-kappa B/fisiologia , Animais , Quimiocinas/fisiologia , Colite/patologia , Colite/fisiopatologia , Colo/patologia , Citocinas/fisiologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Proteínas Imediatamente Precoces/genética , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neutrófilos/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/fisiologia
8.
Cancer Res ; 69(10): 4517-26, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19435915

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is thought to originate from ductal structures, exhibiting strong desmoplastic reaction with stromal pancreatic myofibroblasts (PMF), which are supposed to drive PDAC tumorigenesis. Previously, we observed high expression of the adhesion molecule L1CAM (CD171) in PDAC cells accounting for chemoresistance. Thus, this study aimed to investigate whether PMFs are involved in the induction of tumoral L1CAM and whether this contributes to malignant transformation of pancreatic ductal cells and PDAC tumorigenesis. Immunohistochemistry of tissues from chronic pancreatitis specimens revealed considerable L1CAM expression in ductal structures surrounded by dense fibrotic tissue, whereas no L1CAM staining was seen in normal pancreatic tissues. Using the human pancreatic duct cell line H6c7, we show that coculture with PMFs led to a transforming growth factor-beta1 (TGF-beta1)-dependent up-regulation of L1CAM expression. Similarly, L1CAM expression increased in monocultured H6c7 cells after administration of exogenous TGF-beta1. Both TGF-beta1- and PMF-induced L1CAM expression were independent of Smad proteins but required c-Jun NH(2)-terminal kinase activation leading to the induction of the transcription factor Slug. Moreover, Slug interacted with the L1CAM promoter, and its knockdown abrogated the TGF-beta1- and PMF-induced L1CAM expression. As a result of L1CAM expression, H6c7 cells acquired a chemoresistant and migratory phenotype. This mechanism of TGF-beta1-induced L1CAM expression and the resulting phenotype could be verified in the TGF-beta1-responsive PDAC cell lines Colo357 and Panc1. Our data provide new insights into the mechanisms of tumoral L1CAM induction and how PMFs contribute to malignant transformation of pancreatic duct cells early in PDAC tumorigenesis.


Assuntos
Carcinoma Ductal Pancreático/patologia , Molécula L1 de Adesão de Célula Nervosa/genética , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/patologia , Fatores de Transcrição/fisiologia , Fator de Crescimento Transformador beta1/fisiologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Carcinoma Ductal Pancreático/genética , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Transformação Celular Neoplásica , Técnicas de Cocultura , Humanos , Camundongos , Ductos Pancreáticos/fisiopatologia , Neoplasias Pancreáticas/genética , Pancreatite/patologia , Pancreatite/cirurgia , RNA Interferente Pequeno/genética , Fatores de Transcrição da Família Snail , Transfecção , Regulação para Cima
9.
J Mol Med (Berl) ; 87(1): 99-112, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18931829

RESUMO

L1 cell adhesion molecule (L1CAM) is a transmembrane cell adhesion molecule initially defined as a promigratory molecule in the developing nervous system that appears to be also expressed in some endothelial cells. However, little is known about the functional role of L1CAM on endothelial cells. We observed that L1CAM expression was selectively enhanced on endothelium associated with pancreatic adenocarcinoma in situ and on cultured pancreatic tumor-derived endothelial cells in vitro. L1CAM expression of endothelial cells could be augmented by incubation with immunomodulatory cytokines such as tumor necrosis factor alpha, interferon gamma, or transforming growth factor beta 1. Antibodies to L1CAM and the respective ligand neuropilin-1 blocked tube formation and stromal cell-derived factor 1beta induced transmigration of tumor endothelial cells in vitro. L1CAM expression on tumor-derived-endothelial cells enhanced Panc1 carcinoma cell adhesion to endothelial cell monolayers and transendothelial migration. Our data demonstrate a functional role of L1CAM expression on tumor endothelium that could favor metastasis and angiogenesis during tumor progression.


Assuntos
Adenocarcinoma/metabolismo , Movimento Celular , Endotélio/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Molécula L1 de Adesão de Célula Nervosa/biossíntese , Neoplasias Pancreáticas/metabolismo , Adenocarcinoma/patologia , Anticorpos Antineoplásicos/farmacologia , Citocinas/farmacologia , Endotélio/patologia , Humanos , Metástase Neoplásica , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neuropilina-1/antagonistas & inibidores , Neuropilina-1/metabolismo , Neoplasias Pancreáticas/patologia , Células Tumorais Cultivadas
10.
Int J Oncol ; 34(1): 243-53, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19082495

RESUMO

We recently showed that the adhesion molecule L1CAM (CD171) is overexpressed in pancreatic adenocarcinoma (PDAC) essentially contributing to chemoresistance of PDAC cells. In search of the mechanisms of this effect we now identified alpha5-integrin as the L1CAM ligand being essential for L1CAM-mediated chemoresistance of these highly malignant tumor cells. Thus, blockade or knock-down of alpha5-integrin in the L1CAM expressing PDAC cell lines PT45-P1res, Colo357 and Panc1 increased anti-cancer drug sensitivity. In line with the previously reported NO-dependent caspase inhibition resulting from L1CAM induced iNOS expression, the loss of chemoresistance upon alpha5-integrin inhibition was preceded by decreased iNOS expression and enhanced caspase-3/-7 activation. Accordingly, the loss of anti-cancer drug protection by alpha5-integrin inhibition could be overcome by administration of the NO-donor SNAP. Moreover, the gain of chemoresistance of parental PT45-P1 cells when transfected with L1CAM was abrogated by alpha5-integrin inhibition, whereas transfection of PT45-P1 cells with an integrin binding-deficient L1CAM mutant (L1mutRGE) did neither induce chemoresistance or iNOS expression nor conferred sensitivity to alpha5-integrin inhibition as seen upon transfection with wild-type L1CAM. Thus, mutational loss of the integrin binding site in the L1CAM molecule or the blockade of alpha5-integrin abolished the induction of iNOS expression and chemoresistance by L1CAM, indicating that both a functional L1CAM and alpha5-integrin are indispensable of L1CAM-induced drug resistance in PDAC cells.


Assuntos
Adenocarcinoma/patologia , Carcinoma Ductal Pancreático/patologia , Resistencia a Medicamentos Antineoplásicos , Integrina alfa5/fisiologia , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neoplasias Pancreáticas/patologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Western Blotting , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Caspases/metabolismo , Etoposídeo/uso terapêutico , Citometria de Fluxo , Humanos , Mutagênese Sítio-Dirigida , Molécula L1 de Adesão de Célula Nervosa/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas
11.
Int J Cancer ; 123(8): 1751-60, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18649362

RESUMO

We recently reported on continuous tumor-stroma interactions essentially contributing to chemoresistance of pancreatic ductal adenocarcinoma (PDAC) cells. As demonstrated here, long-term coculture with pancreatic myofibroblasts representing the main stromal compartment of PDAC resulted in a chemoresistant phenotype in the pancreatic ductal epithelial cell line H6c7 as well as in the chemosensitive PDAC cell line T3M4. This involved a reduced expression of caspases and the caspase inducing transcription factor STAT1, both caused by diminished gene transcription. The DNA-methylation inhibitor 5-azadeoxycytidine enhanced caspase and STAT1 expression in cocultured H6c7 and T3M4 cells along with an increased chemosensitivity, indicating a role for CpG DNA-hypermethylation in the downregulation of these crucial apoptosis mediators. Cocultured H6c7 and T3M4 cells exhibited elevated nuclear levels of DNA-methyltransferase-1 (DNMT1). Silencing of DNMT1 expression by siRNA increased expression of caspases and STAT1 and restored chemosensitivity. In SCID mice, tumors arising from coinoculated T3M4 cells and myofibroblasts (co-tumors) responded less towards chemotherapy than mono-tumors, exhibiting decreased apoptosis, no remission and reduced expression of caspases and STAT1. These data underscore the role of myofibroblasts in chemoresistance of PDAC and point to the importance of caspases as central target structures of epigenetic regulation in this scenario. Furthermore, an activated microenvironment might apparently promote the manifestation of chemoresistance already in premalignant precursor cells at early stages of PDAC tumorigenesis.


Assuntos
Caspases/biossíntese , Fibroblastos/patologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/enzimologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Caspases/genética , Linhagem Celular Tumoral , Núcleo Celular/enzimologia , Técnicas de Cocultura , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/biossíntese , DNA (Citosina-5-)-Metiltransferases/metabolismo , Decitabina , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Epigênese Genética , Etoposídeo/farmacologia , Feminino , Fibroblastos/enzimologia , Humanos , Camundongos , Camundongos SCID , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/patologia , Fator de Transcrição STAT1/biossíntese , Fator de Transcrição STAT1/genética , Transfecção , Gencitabina
12.
Exp Cell Res ; 314(13): 2411-8, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18555990

RESUMO

L1 cell adhesion molecule (L1-CAM) is a transmembrane cell adhesion molecule initially defined as a promigratory molecule in the developing nervous system. L1 is also overexpressed in a variety of human carcinomas and is associated with bad prognosis. In carcinoma cell lines L1 augments cell motility and metastasis, tumor growth in nude mice and induces expression of L1-dependent genes. It is not known whether L1-signaling requires ligand binding. The RGD motif in the sixth Ig domain of L1 is a binding site for integrins. In the present study we analyzed the role of RGDs in L1-signaling using site-directed mutagenesis combined with antibody blocking studies. We observed that L1-RGE expressing HEK293 cells showed reduced cell-cell binding, cell motility, invasiveness and tumor growth in NOD/SCID mice. The RGE-mutation impaired L1-dependent gene regulation and antibodies to alphavbeta5 integrin had similar effects. Mutant L1 was unable to translocate to the nucleus. Our findings highlight the importance of the RGD site in L1 for human tumors and suggest that nuclear signaling of L1 is dependent on integrins.


Assuntos
Núcleo Celular/metabolismo , Integrinas/metabolismo , Molécula L1 de Adesão de Célula Nervosa/química , Oligopeptídeos/fisiologia , Transporte Ativo do Núcleo Celular/genética , Animais , Sítios de Ligação/genética , Sítios de Ligação/fisiologia , Células CHO , Adesão Celular/genética , Comunicação Celular/genética , Movimento Celular/genética , Células Cultivadas , Cricetinae , Cricetulus , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Molécula L1 de Adesão de Célula Nervosa/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Molécula L1 de Adesão de Célula Nervosa/fisiologia , Oligopeptídeos/genética , Transdução de Sinais , Transplante Heterólogo
13.
Int J Cancer ; 120(8): 1641-51, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17230504

RESUMO

In this study, the authors investigated the expression of activin receptor-like kinase 1 (ALK1) in pancreatic carcinoma and evaluated its potential role as a tumor suppressor in vitro and in vivo. Endogenous ALK1 expression was demonstrated by immunohistochemistry in both pancreatic tumor tissue and peritumoral normal tissue from 6 patients and by RT-PCR in 8/12 established pancreatic cancer cell lines. Ectopic expression of a constitutively active (ca) ALK1 mutant in TGF-beta sensitive PANC-1 and COLO-357 cells augmented transcriptional activation of a Smad2/3 responsive reporter, and slowed down basal growth in vitro. Both effects were further enhanced by TGF-beta/ALK5 stimulation, suggesting largely independent nuclear Smad signaling by both type I receptors. Upon orthotopic transplantation of PANC-1-caALK1 into immunodeficient mice, tumor size was strongly reduced and was associated with a lower microvessel density in the PANC-1-caALK1-derived tumors. In vitro, this mutant efficiently blocked TGF-beta-induced epithelial-to-mesenchymal transdifferentiation and suppressed TGF-beta/ALK5-mediated activation of the p38 MAPK pathway. Mechanistically, caALK1 silenced MyD118, an immediate TGF-beta target gene whose protein product, GADD45beta, couples Smad signaling to p38 activation. These results show that ALK1 activation in pancreatic tumor cells is antioncogenic by inducing ALK5-independent growth inhibition and by blocking TGF-beta/ALK5-mediated epithelial-to-mesenchymal transdifferentiation and, possibly, invasion and metastatic progression.


Assuntos
Receptores de Ativinas Tipo I/genética , Adenocarcinoma/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Supressores de Tumor/fisiologia , Neoplasias Pancreáticas/genética , Receptores de Ativinas Tipo I/metabolismo , Adenocarcinoma/terapia , Animais , Antígenos de Diferenciação/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células Epiteliais/metabolismo , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/terapia , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteínas Smad/metabolismo , Ativação Transcricional , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Gastroenterology ; 129(3): 952-68, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16143134

RESUMO

BACKGROUND & AIMS: The role of amidated gastrin17 (G17) and the gastrin/CCKB/CCK2 receptor in colorectal carcinogenesis is still a controversial issue. Here, we investigated the effect of G17 on proliferation and apoptosis of CCK2 receptor-expressing human colon cancer cell lines in vitro and in vivo. METHODS: Proliferation was determined by cell counting and cell cycle analysis. Apoptosis was analyzed by annexin V staining, TUNEL staining, caspase-3/7 assay, and JC1 (delta psi) assay. Signal-transduction pathways were analyzed by Western blotting and gel-shift and luciferase assays. An in vivo tumor model with subcutaneously inoculated colon cancer cells in SCID mice was used, and systemic hypergastrinemia was induced by omeprazole. RESULTS: In Colo320 cells stably transfected with the wild-type CCK2 receptor (Colo320wt) or in Lovo cells endogenously expressing CCK2 receptors, G17 treatment inhibited proliferation along with a G2/M cell cycle arrest. Furthermore, the administration of G17 significantly augmented apoptosis of CCK2 receptor-expressing cells. In contrast, G17 had no effect on proliferation and apoptosis in Colo320 cells stably transfected with a tumor-derived CCK2 receptor mutant (Colo320mut) or in cells lacking CCK2 receptor expression. Systemic hypergastrinemia in severe combined immunodeficiency (SCID) mice suppressed the growth of Colo320wt tumors accompanied by enhanced apoptosis as compared with untreated tumors. In contrast, omeprazole did not affect Colo320mut tumors reflecting a loss-of-function state of the CCK2(mut) receptor. This is supported by the observation that, in Colo320wt cells, but not in Colo320mut cells, G17 treatment induced the MAPK/ERK/AP-1 pathway and inhibited the activity of NF-kappaB. CONCLUSIONS: G17 exerts an antiproliferative and proapoptotic effect on human colon cancer cells expressing the wild-type CCK2 receptor. This supports the view that amidated gastrin prevents rather than promotes colorectal carcinogenesis.


Assuntos
Neoplasias do Colo/genética , Gastrinas/farmacologia , Receptor de Colecistocinina B/genética , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , RNA Neoplásico/genética , RNA Neoplásico/isolamento & purificação , Receptor de Colecistocinina B/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
15.
Cell Signal ; 17(12): 1505-15, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15951156

RESUMO

Although expression of the gastrin/cholecystokinin-2 receptor (CCK2R) is widely reported in human colorectal cancer, little is known on its role in mediating mature amidated gastrin (gastrin-17 amide, G-17) induced intracellular signal transduction in colon cancer cells. The purpose of this study was to explore the intracellular events of colorectal cancer cells after gastrin binding to CCK2R. Meanwhile, the influence of a natural point mutation 286V-->F in the third intracellular loop of CCK2R on gastrin-envoked intracellular signal transduction was also investigated. Firstly, Colo320 cells were stably transfected with wild type (Colo320 WT) and mutant CCK2R (Colo320 M), respectively. The intracellular signal transduction events in response to gastrin were investigated in both Colo320 WT and Colo320 M cells. In Colo320 WT cells, G-17 induced formation of intracellular cyclic AMP and inositol 1,4,5-trisphosphate, and stimulated intracellular calcium mobilization. G-17 also stimulated tyrosine phosphorylation of ERKl/2, p38, FAK, and paxillin, and up-regulated the mRNA expression of early response gene c-Jun and c-Fos. However, G-17 inhibited proliferation and induced apoptosis in Colo320 WT cells. Mutation 286V-->F in the third intracellular loop of CCK2R blocked G-17 induced biological without affecting binding affinity of CCK2R to G-17. Our results suggest that activation of CCK2R by gastrin stimulates heterotrimeric G-protein Gq and G(12/13) mediated intracellular signal transduction pathway in colon cancer cells. The valine-287 residue in third intracellular loop of CCK2R plays a pivotal role in CCK2R mediated intracellular signal transduction.


Assuntos
Neoplasias do Colo/metabolismo , Receptor de Colecistocinina B/metabolismo , Transdução de Sinais , Valina/metabolismo , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , AMP Cíclico/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Gastrinas/metabolismo , Genes Precoces , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Paxilina/metabolismo , Fosforilação , Mutação Puntual , Estrutura Secundária de Proteína , RNA Mensageiro/metabolismo , Receptor de Colecistocinina B/química , Receptor de Colecistocinina B/genética , Transfecção , Tirosina/metabolismo , Regulação para Cima
16.
Cancer Res ; 65(4): 1316-24, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15735017

RESUMO

The permanent activation of the transcription factor nuclear factor-kappaB (NF-kappaB) in pancreatic cancer cells is associated with a profound resistance towards chemotherapy. In the present study, we show that chemoresistant pancreatic cancer cell lines exhibiting constitutive NF-kappaB activity (i.e., PancTu-1, BxPc3, and Capan-1) express significantly elevated levels of the E3-ubiquitin ligase receptor subunit betaTRCP1, compared with pancreatic carcinoma cell lines lacking constitutive NF-kappaB activity and chemoresistance (i.e., PT45-P1 and T3M4). If transfected with betaTRCP1, PT45-P1 cells exhibit an elevated NF-kappaB activity and become less sensitive towards anticancer drug treatment (i.e., etoposide). Conversely, blockade of betaTRCP1 expression in PancTu-1 cells by transfection with a vector-expressed small interfering RNA reduces NF-kappaB activation and chemoresistance. In PancTu-1 cells, betaTRCP1 expression is inhibited, at least in part, by the interleukin-1 (IL-1) receptor(I) antagonist, whereas stimulation of PT45-P1 cells with IL-1beta resulted in an increased expression of betaTRCP1, and transfection of this cell line with betaTRCP1 induced IL-1beta secretion in a NF-kappaB-dependent fashion. Thus, via its close and mutual link to IL-1beta secretion, betaTRCP1 expression might substantially contribute to the persistent, IL-1beta-dependent activation of NF-kappaB in pancreatic carcinoma cells. In support of this, betaTRCP1 expression is detectable at considerable levels in a great number of pancreatic ductal adenocarcinoma specimens, along with an intense staining for activated NF-kappaB. Altogether, our findings of the elevated betaTRCP1 expression in pancreatic carcinoma cells pinpoint to another important mediator of constitutive NF-kappaB activation and thereby of chemoresistance.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , NF-kappa B/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Contendo Repetições de beta-Transducina/biossíntese , Antineoplásicos Fitogênicos/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Etoposídeo/farmacologia , Humanos , Proteínas I-kappa B/metabolismo , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Sialoglicoproteínas/farmacologia , Transfecção , Proteínas Contendo Repetições de beta-Transducina/antagonistas & inibidores , Proteínas Contendo Repetições de beta-Transducina/genética
17.
FASEB J ; 19(6): 620-2, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15670977

RESUMO

Pancreatic adenocarcinoma represents a tumor type with extremely poor prognosis. High apoptosis resistance and a strong invasive and early metastatic potential contribute to its highly malignant phenotype. Here we identified the death receptor adaptor molecule TRAF2 as a key player in pancreatic cancer pathophysiology. Using immunohistochemistry and Western blot analysis we found TRAF2 overexpressed in 34 of 36 pancreatic tumor samples as well as in pancreatic tumor cell lines resistant to CD95-mediated apoptosis. The high TRAF2 protein level was not related to chromosomal changes, as monitored by FISH analysis. Instead, the NF-kappaB- and MEK-signaling pathways were involved. Introduction of a TRAF2 expression vector in CD95-sensitive Colo357 cells resulted in (i) resistance to CD95-induced apoptosis; (ii) increased constitutive NF-kappaB and AP-1 activity; and (iii) higher basal secretion of matrix metalloproteinases (MMPs), urokinase-type plasminogen activator (uPA), and IL-8, leading to increased invasiveness. High apoptosis resistance and uPA secretion could be reverted by TRAF2-specific siRNA. Stimulation of TRAF2-overexpressing cells with CD95 ligand led to induction of NF-kappaB and AP-1, enhanced IL-8- and uPA-secretion, and a further increased invasiveness. Thus, TRAF2 overexpression does not only block apoptosis induction by CD95 but also converts this death receptor into a mediator of invasiveness.


Assuntos
Adenocarcinoma/patologia , Invasividade Neoplásica , Neoplasias Pancreáticas/patologia , Fator 2 Associado a Receptor de TNF/fisiologia , Receptor fas/fisiologia , Apoptose , Linhagem Celular Tumoral , Ensaio de Desvio de Mobilidade Eletroforética , Expressão Gênica , Humanos , Hibridização in Situ Fluorescente , Interleucina-8/análise , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , NF-kappa B/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fator 2 Associado a Receptor de TNF/genética , Transfecção , Ativador de Plasminogênio Tipo Uroquinase/análise
18.
Nat Med ; 10(6): 602-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15146177

RESUMO

Suramin is a polysulfonated derivative of urea and has been widely used both to treat infections and as a chemotherapeutic drug. Suramin has been shown to inhibit growth factor signaling pathways; however, its effect on apoptosis is unknown. Here we show that suramin inhibits apoptosis induced through death receptors in hepatoma and lymphoma cells. It also inhibits the proapoptotic effect of chemotherapeutic drugs. The antiapoptotic mechanism is specific to cell type and is caused by reduced activation, but not altered composition, of the death-inducing signaling complex (DISC), and by inhibition of the initiator caspases 8, 9 and 10. Suramin also shows similar effects in in vivo models: apoptotic liver damage induced by CD95 stimulation and endotoxic shock mediated by tumor-necrosis factor (TNF) are inhibited in mice, but necrotic liver damage is not inhibited in a rat model of liver transplantation. Thus, the antiapoptotic property of suramin in the liver may be therapeutically exploited.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Receptores do Fator de Necrose Tumoral/metabolismo , Suramina/farmacologia , Receptor fas/metabolismo , Animais , Apoptose/fisiologia , Caspases/metabolismo , Linhagem Celular , Dexametasona/farmacologia , Ativação Enzimática , Raios gama , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Tripanossomicidas/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
19.
Med Klin (Munich) ; 99(4): 185-90, 2004 Apr 15.
Artigo em Alemão | MEDLINE | ID: mdl-15085288

RESUMO

BACKGROUND AND PURPOSE: The pancreatic ductal adenocarcinoma (PDAC) is characterized by a highly malignant phenotype and a profound chemoresistance. Thus, options for an effective treatment of this disease are still quite poor. In this study, it was investigated whether the autocrine secretion of interleukin-(IL-)1beta is related to a chemoresistant phenotype of PDAC cells in vivo. MATERIAL AND METHODS: Human PancTu1 PDAC cells were inoculated subcutaneously into female SCID mice. After 10 days of outgrowth, animals were randomized and left untreated or treated with an IL-1beta-RI antibody, etoposide, or a combination of both. After treatment for 14 days, tumor sizes were determined and each tumor was analyzed immunohistochemically for apoptosis (TUNEL), activated NF-kappaB (p65), and vascularization (CD31 staining). RESULTS: The combination of IL-1beta-RI antibody and etoposide led to a significantly reduced outgrowth of PancTu1 tumors in comparison to the monotherapies or no treatment. Accordingly, the number of apoptotic cells was significantly elevated in tumors of the combination group. After treatment with the IL-1beta-RI antibody, less activated NF-kappaB was present in tumors compared to the control group. Moreover, tumors of the combination group showed a clearly reduced vascularization. CONCLUSION: The autocrine secretion of IL-1beta contributes to a constitutively increased NF-kappaB activity in PDAC cells along with a chemoresistant phenotype.


Assuntos
Comunicação Autócrina/fisiologia , Carcinoma Ductal Pancreático/patologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistência a Múltiplos Medicamentos/fisiologia , Interleucina-1/antagonistas & inibidores , Interleucina-1/metabolismo , NF-kappa B/metabolismo , Neoplasias Pancreáticas/patologia , Sialoglicoproteínas/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Etoposídeo/farmacologia , Feminino , Humanos , Marcação In Situ das Extremidades Cortadas , Proteína Antagonista do Receptor de Interleucina 1 , Camundongos , Camundongos SCID , Transplante de Neoplasias , Proteínas Recombinantes/farmacologia , Células Tumorais Cultivadas/patologia
20.
Cancer Res ; 64(4): 1331-7, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14973050

RESUMO

Pancreatic ductal carcinoma is characterized by a profound chemoresistance. As we have shown previously, these tumor cells can develop chemoresistance by interleukin (IL)-1beta in an autocrine and nuclear factor-kappaB-dependent fashion. Because pancreatic ductal carcinoma contains many mesenchymal stromal cells, we further investigated how tumor-stroma interactions contribute to chemoresistance by using a transwell coculture model, including murine pancreatic fibroblasts and the chemosensitive human pancreatic carcinoma cell lines T3M4 and PT45-P1. If cultured with fibroblast-conditioned medium or kept in coculture with fibroblasts, both cell lines became much less sensitive toward treatment with etoposide than cells cultured under standard conditions. Furthermore, the secretion of IL-1beta in T3M4 and PT45-P1 cells was increased by the fibroblasts, and IL-1beta-receptor blockade abolished the resistance-inducing effect during cocultivation. This stimulated IL-1beta secretion could be attributed to nitric oxide (NO) released by the fibroblasts as an IL-1beta-inducing factor. Although both tumor cells secreted only little NO, which was in line with undetectable inducible nitric oxide synthase (iNOS) expression, fibroblasts exhibited significant iNOS expression and NO secretion that could be further induced by the tumor cells. Incubation of T3M4 and PT45-P1 cells with the NO donor S-Nitroso-N-acetyl-D,L-penicillamine up-regulated IL-1beta secretion and conferred resistance toward etoposide-induced apoptosis. Conversely, the resistance-inducing effect of the fibroblasts was significantly abolished, when the specific iNOS inhibitor aminoguanidine was added during coculture. Immunohistochemistry on tissue sections from human pancreatic ductal carcinoma also revealed iNOS expression in stromal cells and IL-1beta expression in tumor cells, thus supporting the in vitro findings. These data clearly demonstrate that fibroblasts contribute to the development of chemoresistance in pancreatic carcinoma cells via increased secretion of NO, which in turn leads to an elevated release of IL-1beta by the tumor cells. These findings substantiate the implication of tumor-stromal interactions in the chemoresistance of pancreatic carcinoma.


Assuntos
Carcinoma Ductal Pancreático/tratamento farmacológico , Fibroblastos/fisiologia , Interleucina-1/metabolismo , Óxido Nítrico/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Penicilamina/análogos & derivados , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Comunicação Celular , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Penicilamina/farmacologia , Células Estromais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...