Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS One ; 8(1): e54824, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23359812

RESUMEN

BACKGROUND: Blood and spleen expansion of immature myeloid cells (IMCs) might compromise the immune response to cancer. We studied in vivo circulating and splenic T lymphocyte and IMC subsets in patients with benign and malignant pancreatic diseases. We ascertained in vitro whether pancreatic adenocarcinoma (PDAC)-associated IMC subsets are induced by tumor-derived soluble factors and whether they are immunosuppressive focusing on the inhibitory co-stimulatory molecules PDL1 and CTLA4. METHODOLOGY AND PRINCIPAL FINDINGS: 103 pancreatic and/or splenic surgical patients were enrolled including 52 PDAC, 10 borderline and 10 neuroendocrine tumors (NETs). Lymphocytes and IMCs were analysed by flow cytometry in blood, in spleen and in three PDAC cell conditioned (CM) or non conditioned PBMC. PDL1 and CTLA4 were studied in 30 splenic samples, in control and conditioned PBMC. IMCs were FACS sorted and co-coltured with allogenic T lymphocytes. In PDAC a reduction was found in circulating CD8(+) lymphocytes (p = 0.004) and dendritic cells (p = 0.01), which were reduced in vitro by one PDAC CM (Capan1; p = 0.03). Blood myeloid derived suppressive cells (MDSCs) CD33(+)CD14(-)HLA-DR(-) were increased in PDAC (p = 0.022) and were induced in vitro by BxPC3 CM. Splenic dendritic cells had a higher PDL1 expression (p = 0.007), while CD33(+)CD14(+)HLA-DR(-) IMCs had a lower CTLA4 expression (p = 0.029) in PDAC patients. In vitro S100A8/A9 complex, one of the possible inflammatory mediators of immune suppression in PDAC, induced PDL1 (p = 0.018) and reduced CTLA4 expression (p = 0.028) among IMCs. IMCs not expressing CTLA4 were demonstrated to be immune suppressive. CONCLUSION: In PDAC circulating dendritic and cytotoxic T cells are reduced, while MDSCs are increased and this might favour tumoral growth and progression. The reduced CTLA4 expression found among splenic IMCs of PDAC patients was demonstrated to characterize an immune suppressive phenotype and to be consequent to the direct exposure of myeloid cells to pancreatic cancer derived products, S100A8/A9 complex in particular.


Asunto(s)
Antígeno B7-H1/inmunología , Antígeno CTLA-4/inmunología , Neoplasias Pancreáticas/inmunología , Bazo/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Separación Celular , Cartilla de ADN , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/patología , Adulto Joven
2.
Pancreas ; 40(7): 1131-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21792088

RESUMEN

OBJECTIVES: To verify whether the dysregulation of CD4 T cells concurs in worsening the outcome of pancreatic cancer, we compared the effects of pancreatic cancer and other gastrointestinal cancer cell-conditioned media on the (1) proliferation, migration, and differentiation of CD4 T cells and (2) expansion of CD4 memory (CD45RO), naive (CD45RA), activated (CD69), and regulatory (CD25) subsets. METHODS: After culture of CD4 T cells in control, pancreatic (BxPC3, Capan1, MiaPaCa2), or gastrointestinal cancer (AGS, HepG2, HT29) cell-conditioned media, we evaluated proliferation, migration, interferon γ (IFNγ) production, and CD45RA, CD45RO, CD69, and CD25 membrane expression in control and conditioned CD4 T cells. RESULTS: Only pancreatic cancer-conditioned media (1) inhibited CD4 T-cell proliferation (P < 0.001) and migration under human stromal cell-derived factor-α chemotaxis (P < 0.001) and (2) induced CD4 T-cell IFNγ production (P < 0.05) and the expansion of the CD69-positive subset (P < 0.001) with respect to the control, with no changes being found in the CD45RA, CD45RO, and CD25 subsets. CONCLUSIONS: The in vitro findings achieved in the present study demonstrate that pancreatic cancer cells inhibit CD4 T-cell proliferation and migration, induce IFNγ production, and favor a CD69 subset expansion, suggesting that CD4 T cells play an important role in pancreatic cancer immune evasion.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Pancreáticas/inmunología , Escape del Tumor , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Medios de Cultivo Condicionados/metabolismo , Células HT29 , Células Hep G2 , Humanos , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Factores de Tiempo
3.
J Cell Physiol ; 226(2): 456-68, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20717964

RESUMEN

After isolating NT-S100A8 from pancreatic cancer (PC) tissue of diabetic patients, we verified whether this peptide alters PC cell growth and invasion and/or insulin release and [Ca(2+)](i) oscillations of insulin secreting cells and/or insulin signaling. BxPC3, Capan1, MiaPaCa2, Panc1 (PC cell lines) cell growth, and invasion were assessed in the absence or presence of 50, 200, and 500 nM NT-S100A8. In NT-S100A8 stimulated ß-TC6 (insulinoma cell line) culture medium, insulin and [Ca(2+)] were measured at 2, 3, 5, 10, 15, 30, and 60 min, and [Ca(2+)](i) oscillations were monitored (epifluorescence) for 3 min. Five hundred nanomolars NT-S100A8 stimulated BxPC3 cell growth only and dose dependently reduced MiaPaCa2 and Panc1 invasion. Five hundred nanomolars NT-S100A8 induced a rapid insulin release and enhanced ß-TC6 [Ca(2+)](i) oscillations after both one (F = 6.05, P < 0.01) and 2 min (F = 7.42, P < 0.01). In the presence of NT-S100A8, [Ca(2+)] in ß-TC6 culture medium significantly decreased with respect to control cells (F = 6.3, P < 0.01). NT-S100A8 did not counteract insulin induced phosphorylation of the insulin receptor, Akt and IκB-α, but it independently activated Akt and NF-κB signaling in PC cells. In conclusion, NT-S100A8 exerts a mild effect on PC cell growth, while it reduces PC cell invasion, possibly by Akt and NF-κB signaling, NT-S100A8 enhances [Ca(2+)](i) oscillations and insulin release, probably by inducing Ca(2+) influx from the extracellular space, but it does not interfere with insulin signaling.


Asunto(s)
Calcio/metabolismo , Calgranulina A/metabolismo , Diabetes Mellitus/etiología , Neoplasias Pancreáticas/complicaciones , Péptidos/metabolismo , Animales , Calgranulina A/genética , Línea Celular Tumoral , Diabetes Mellitus/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/fisiopatología , Péptidos/genética , Péptidos/farmacología , Ratas , Transducción de Señal/fisiología
4.
Pancreas ; 39(5): 662-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20562578

RESUMEN

OBJECTIVES: alpha-Tocopheryl succinate (alpha-TOS) is thought to be toxic only for cancer cells. We ascertained in vitro alpha-TOS effects on pancreatic cancer (PC) and normal cell growth and verified whether the combination of nontoxic alpha-TOS and 5-fluorouracil (5-FU) doses causes cancer cell death and whether alpha-TOS effects are mediated by the proapoptotic proteins Bax/Bak and/or SMAD4/DPC4 status. METHODS: Five PC cell lines, myoblasts, normal monocytes, wild-type (WT) and Bax/Bak double knockout mouse embryonic fibroblast (MEF) cells, and permanently SMAD4/DPC4-transfected PSN1 cells were cultured in 1% and 10% fetal calf serums (FCSs), without or with alpha-TOS (5-500 micromol/L). Nontoxic 5-FU (0.0001 mmol/L) and alpha-TOS alone or in combination were also evaluated. RESULTS: Only PSN1 PC cell line, which had SMAD4/DPC4 homozygous deletion, was sensitive to nontoxic alpha-TOS doses (5 micromol/L in 1% FCS and 50 micromol/L in 10% FCS). A 20-micromol/L alpha-TOS inhibited MEF-WT, not MEF-double knockout growth. Only PSN1 cells were sensitive to nontoxic 5-FU and alpha-TOS combination. SMAD4/DPC4 transfection restored PSN1 resistance to the effects of combined 5-FU and alpha-TOS effects. CONCLUSIONS: Only a minority of PC cells are sensitive to the antiproliferative effects of alpha-TOS, any sensitivity appearing to be correlated with SMAD4/DPC4 homozygous deletion and Bax/Bak expression.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Vitamina E/análogos & derivados , alfa-Tocoferol/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Fibroblastos/efectos de los fármacos , Fluorouracilo/farmacología , Humanos , Ratones , Monocitos/efectos de los fármacos , Mioblastos/efectos de los fármacos , Proteína Smad4/análisis , alfa-Tocoferol/uso terapéutico , Proteína Destructora del Antagonista Homólogo bcl-2/análisis , Proteína X Asociada a bcl-2/análisis
5.
Clin Chem Lab Med ; 47(6): 713-23, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19426140

RESUMEN

BACKGROUND: Surface-enhanced laser desorption and ionization time-of-flight mass spectrometry (SELDI-TOF/MS), a laboratory-friendly technique, is used to identify biomarkers for cancer. The aim of the present study was to explore the application of SELDI proteomic patterns in serum for distinguishing between cases of pancreatic cancer, chronic pancreatitis, type 2 diabetes mellitus and healthy controls. METHODS: Sera from 12 healthy controls, 24 patients with type 2 diabetes mellitus, 126 with pancreatic cancer, including 84 with diabetes, and 61 with chronic pancreatitis, 32 of which were diabetics, were analyzed using SELDI-TOF/MS. Spectra (IMAC-30) were clustered and classified using Biomarker Wizard and Biomarker Pattern software. RESULTS: Two decision tree classification algorithms, one with and one without CA 19-9, were constructed. In the absence of CA 19-9, the splitting protein peaks were: m/z 1526, 1211, and 3519; when CA 19-9 was used in the analysis, it replaced the m/z 3519 splitter. The two algorithms performed equally for classifying patients. A classification tree that considered diabetic patients only was constructed; the main splitters were: 1211, CA 19-9, 7903, 3359, 1802. With this algorithm, 100% of patients with type 2 diabetes mellitus, 97% with chronic pancreatitis and 77% of patients with pancreatic cancer were correctly classified. SELDI-TOF/MS features improved the diagnostic accuracy of CA 19-9 (AUC = 0.883 for CA 19-9; AUC = 0.935 for CA 19-9 and SELDI-TOF/MS features combined). CONCLUSIONS: SELDI-TOF/MS allows identification of new peptides which, in addition to CA 19-9, enable the correct classification of the vast majority of patients with pancreatic cancer, which can be distinguished from patients with chronic pancreatitis or type 2 diabetes mellitus.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias Pancreáticas/diagnóstico , Péptidos/sangre , Proteómica/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Adulto , Anciano , Anciano de 80 o más Años , Antígeno CA-19-9/sangre , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/diagnóstico , Diagnóstico Diferencial , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/mortalidad , Pancreatitis Crónica/sangre , Pancreatitis Crónica/diagnóstico , Modelos de Riesgos Proporcionales
6.
J Gastrointest Surg ; 11(11): 1506-14; discussion 1514, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17846855

RESUMEN

Several bacterial and host-related factors concur in causing Helicobacter pylori eradication failure. We ascertained the role of bacterial virulence genes (cagA, vacA), clarithromycin resistance [Cla(R), 23S ribosomal RNA (rRNA) mutations], host polymorphism of CYP2C19 (polyphosphoinositide, PPI, metabolism) and of the cytokines IL-1B-31C>T, IL-1RN VNTR, IFN-gamma+874A>T, TNF-alpha-1031T>C, TNF-alpha-857C>T, TNF-alpha-376G>A, TNF-alpha-308G>A, TNF-alpha-238G>A, IL-10-1082A>G, IL-10-819C>T, IL-10-592C>A, IL-12A+6686G>A, IL-12B+15485A>C. Two groups of H. pylori-infected and H. pylori-treated patients were retrospectively identified: 45 not eradicated and 57 eradicated. Treatment failure was significantly correlated with Cla(R) (all resistant strains in non-eradicated patients); with TNF-alpha-238, IL10-819, IL10-592, IL-12B+15485 single nucleotide polymorphism (SNP); with IL10 ATA/ATA haplotype; and with antral inflammatory grade. On considering Cla(S)-infected patients only, logistic regression analysis (eradication = dependent; TNF-alpha-238, IL12B + 15485 genotypes, IL10 ATA/ATA as present or absent, antral gastritis grade = covariates) confirmed as significantly correlated with eradication antral gastritis grade only (Exp(B) = 6.48; 95% CI, 1.2-35.01). In conclusion, the bacterial determinant causing triple therapy failure is clarithromycin resistant, being virulence genes not involved. The host related factors that favor eradication are those linked to inflammation: a higher inflammatory infiltrate in the mucosa, possibly favored by genotypes able to down regulate the anti-inflammatory cytokine response, enhance the chance of eradication success.


Asunto(s)
Antibacterianos/farmacología , Claritromicina/farmacología , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori , Adolescente , Adulto , Anciano , Antígenos Bacterianos/genética , Hidrocarburo de Aril Hidroxilasas/genética , Proteínas Bacterianas/genética , Niño , Preescolar , Citocromo P-450 CYP2C19 , Farmacorresistencia Microbiana , Quimioterapia Combinada , Femenino , Mucosa Gástrica , Frecuencia de los Genes , Infecciones por Helicobacter/genética , Humanos , Interleucina-10/genética , Masculino , Persona de Mediana Edad , Oxigenasas de Función Mixta/genética , Farmacogenética , Mutación Puntual , Polimorfismo Genético , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/genética , Virulencia
7.
Pancreas ; 35(3): 224-31, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17895842

RESUMEN

OBJECTIVES: Suicide gene therapy with FCY1 gene, encoding cytosine deaminase (CD), together with FUR1, encoding uracil phosphoribosyltransferase (UPRT), has been proposed for pancreatic cancer therapy in vivo. We ascertained whether gene therapy with FCY1-FUR1 is effective in killing pancreatic cancer cells after 5-fluorocytosine (5-FC) treatment. METHODS: AsPC1, BxPC3, Capan1, MIA PaCa2, and Panc1 cell lines were transfected using 2 plasmid vectors expressing CD only (pRSV-CD) or the chimera CD-UPRT (pRSV-CD-UPRT). Control and pRSV-CD- or pRSV-CD-UPRT-transfected cell lines were treated with 0, 0.1, 0.5, 1, 5, and 10 mM of 5-FC for 1, 3, 6, 8, 10, and 13 days. RESULTS: FCY1 alone did not confer sensitivity to 5-FC. The CD-UPRT-transfected BxPC3 and Panc1 were sensitive to very low 5-FC doses (0.1 mM). 5-Fluorocytosine-sensitive transfected cell lines rapidly converted 5-FC into 5-fluorouracil, whereas the 5-FC resistant cell lines had an impaired 5-FC conversion. CONCLUSIONS: Suicide gene therapy with the FCY1 gene alone was ineffective in the treatment of pancreatic cancer in vitro. The pRSV-CD-UPRT construct conferred 5-FC sensitivity to some pancreatic cancer cell lines. Therefore, the application in vivo of suicide gene therapy with FCY1 alone or in combination with the FUR1 gene is probably destined to fail.


Asunto(s)
Adenocarcinoma/terapia , Antimetabolitos Antineoplásicos/farmacología , Citosina Desaminasa/genética , Flucitosina/farmacología , Genes Transgénicos Suicidas , Neoplasias Pancreáticas/terapia , Pentosiltransferasa/genética , Profármacos/farmacología , Proteínas de Saccharomyces cerevisiae/genética , Adenocarcinoma/patología , Antimetabolitos Antineoplásicos/metabolismo , Biotransformación , Línea Celular Tumoral/efectos de los fármacos , Neoplasias Colorrectales/patología , Citosina Desaminasa/metabolismo , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Flucitosina/metabolismo , Humanos , Técnicas In Vitro , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/patología , Pentosiltransferasa/metabolismo , Profármacos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Timidilato Sintasa/antagonistas & inhibidores , Transfección , Nucleótidos de Uracilo/biosíntesis
8.
Clin Chim Acta ; 381(1): 50-5, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17397816

RESUMEN

This work focuses on the main DNA repair pathways, highlighting their role in gastrointestinal carcinogenesis and the role of mitochondrial DNA (mtDNA), mutations being described in several tumor types, including those of the gastrointestinal tract. The mismatch repair (MMR) system is inherently altered in patients with hereditary non-polyposis colorectal cancer, and plays a role in carcinogenesis in a subset of sporadic colorectal, gastric and esophageal cancers. Alterations in homologous recombination (HR) and non-homologous end-joining (NHEJ) also contribute to the development of pancreatic cancer. Gene polymorphisms of some X-ray cross-complementing (XRCCs), cofactor proteins involved in the base excision repair pathway, have been investigated in relation to gastric, colorectal and pancreatic cancer. Yet only one polymorphism, XRCC1 Arg194Trp, appears to be involved in smoking-related cancers and in early onset pancreatic cancer. Although evidence in the literature indicates that mtDNA somatic mutations play a role in gastric and colorectal carcinogenesis, no sound conclusions have yet been drawn regarding this issue in pancreatic cancer, although an mtDNA variant at 16519 is believed to worsen the outcome of pancreatic cancer patients, possibly because it is involved in altering cellular metabolism.


Asunto(s)
Reparación del ADN/efectos de los fármacos , ADN Mitocondrial/genética , Neoplasias Gastrointestinales/genética , Mutación/fisiología , Transducción de Señal/efectos de los fármacos , Animales , Reparación de la Incompatibilidad de ADN , Neoplasias Gastrointestinales/patología , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA