Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Ann Oncol ; 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38906254

RESUMEN

BACKGROUND: After surgical resection of pancreatic ductal adenocarcinoma (PDAC), patients are predominantly treated with adjuvant chemotherapy, commonly consisting of gemcitabine-based regimens or the modified FOLFIRINOX regimen (mFFX). While mFFX has been shown to be more effective than gemcitabine-based regimens, it is also associated with higher toxicity. Current treatment decisions are based on patient performance status rather than on the molecular characteristics of the tumor. To address this gap, the goal of this study was to develop drug-specific transcriptomic signatures for personalized chemotherapy treatment. PATIENTS AND METHODS: We used PDAC datasets from preclinical models, encompassing chemotherapy response profiles for the mFFX-regimen components. From them we identified specific gene transcripts associated with chemotherapy response. Three transcriptomic AI-signatures were obtained by combining Independent Component Analysis, Least Absolute Shrinkage and the Selection Operator-Random Forest approach. We integrated a previously developed gemcitabine signature with three newly developed ones. The machine learning strategy employed to enhance these signatures incorporates transcriptomic features from the tumor microenvironment, leading to the development of the Pancreas-View tool ultimately clinically validated in a cohort of 343 patients from the PRODIGE-24/CCTG PA6 trial. RESULTS: Patients who were predicted to be sensitive to the administered drugs (n=164; 47.8%) had longer disease-free survival (DFS) than the other patients. The median DFS in the mFFX sensitive group treated with mFFX was 50.0 months (stratified HR: 0.31; 95% CI, 0.21-0.44; p<0.001) and 33.7 months (stratified HR: 0.40; 95% CI, 0.17-0.59; p<0.001) in the gemcitabine sensitive group when treated with gemcitabine. Comparatively patients with signature predictions unmatched with the treatments (n=86; 25.1%) or those resistant to all drugs (n=93; 27.1%) had shorter DFS (10.6 and 10.8 months, respectively). CONCLUSIONS: This study presents a transcriptome-based tool that was developed using preclinical models and machine learning to accurately predict sensitivity to mFFX and gemcitabine.

2.
Transl Oncol ; 16: 101315, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34906890

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) patients are frequently treated by chemotherapy. Even if personalized therapy based on molecular analysis can be performed for some tumors, PDAC regimens selection is still mainly based on patients' performance status and expected efficacy. Therefore, the establishment of molecular predictors of chemotherapeutic efficacy could potentially improve prognosis by tailoring treatments. We have recently developed an RNA-based signature that predicts the efficacy of adjuvant gemcitabine using 38 PDAC primary cell cultures. While demonstrated its efficiency, a significant association with the classical/basal-like PDAC spectrum was observed. We hypothesized that this flaw was due to the basal-like biased phenotype of cellular models used in our strategy. To overcome this limitation, we generated a prospective cohort of 27 consecutive biopsied derived pancreatic organoids (BDPO) and include them in the signature identification strategy. As BDPO's do not have the same biased phenotype as primary cell cultures we expect they can compensate one with each other and cover a broader range of molecular phenotypes. We then obtained an improved signature predicting gemcitabine sensibility that was validated in a cohort of 300 resected PDAC patients that have or have not received adjuvant gemcitabine. We demonstrated a significant association between the improved signature and the overall and disease-free survival in patients predicted as sensitive and treated with adjuvant gemcitabine. We propose then that including BDPO along primary cell cultures represent a powerful strategy that helps to overcome primary cell cultures limitations producing unbiased RNA-based signatures predictive of adjuvant treatments in PDAC.

3.
Ann Oncol ; 32(2): 250-260, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33188873

RESUMEN

BACKGROUND: Chemotherapy is the only systemic treatment approved for pancreatic ductal adenocarcinoma (PDAC), with a selection of regimens based on patients' performance status and expected efficacy. The establishment of a potent stratification associated with chemotherapeutic efficacy could potentially improve prognosis by tailoring treatments. PATIENTS AND METHODS: Concomitant chemosensitivity and genome-wide RNA profiles were carried out on preclinical models (primary cell cultures and patient-derived xenografts) derived from patients with PDAC included in the PaCaOmics program (NCT01692873). The RNA-based stratification was tested in a monocentric cohort and validated in a multicentric cohort, both retrospectively collected from resected PDAC samples (67 and 368 patients, respectively). Forty-three (65%) and 203 (55%) patients received adjuvant gemcitabine in the monocentric and the multicentric cohorts, respectively. The relationships between predicted gemcitabine sensitivity and patients' overall survival (OS) and disease-free survival were investigated. RESULTS: The GemPred RNA signature was derived from preclinical models, defining gemcitabine sensitive PDAC as GemPred+. Among the patients who received gemcitabine in the test and validation cohorts, the GemPred+ patients had a higher OS than GemPred- (P = 0.046 and P = 0.00216). In both cohorts, the GemPred stratification was not associated with OS among patients who did not receive gemcitabine. Among gemcitabine-treated patients, GemPred+ patients had significantly higher OS than the GemPred-: 91.3 months [95% confidence interval (CI): 61.2-not reached] versus 33 months (95% CI: 24-35.2); hazard ratio 0.403 (95% CI: 0.221-0.735, P = 0.00216). The interaction test for gemcitabine and GemPred+ stratification was significant (P = 0.0245). Multivariate analysis in the gemcitabine-treated population retained an independent predictive value. CONCLUSION: The RNA-based GemPred stratification predicts the benefit of adjuvant gemcitabine in PDAC patients.


Asunto(s)
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Quimioterapia Adyuvante , Desoxicitidina/análogos & derivados , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Estudios Retrospectivos , Transcriptoma , Gemcitabina
4.
Cell Death Dis ; 7: e2295, 2016 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-27415425

RESUMEN

Both clinical and experimental evidence have firmly established that chronic pancreatitis, in particular in the context of Kras oncogenic mutations, predisposes to pancreatic ductal adenocarcinoma (PDAC). However, the repertoire of molecular mediators of pancreatitis involved in Kras-mediated initiation of pancreatic carcinogenesis remains to be fully defined. In this study we demonstrate a novel role for vacuole membrane protein 1 (VMP1), a pancreatitis-associated protein critical for inducible autophagy, in the regulation of Kras-induced PDAC initiation. Using a newly developed genetically engineered model, we demonstrate that VMP1 increases the ability of Kras to give rise to preneoplastic lesions, pancreatic intraepithelial neoplasias (PanINs). This promoting effect of VMP1 on PanIN formation is due, at least in part, by an increase in cell proliferation combined with a decrease in apoptosis. Using chloroquine, an inhibitor of autophagy, we show that this drug antagonizes the effect of VMP1 on PanIN formation. Thus, we conclude that VMP1-mediated autophagy cooperate with Kras to promote PDAC initiation. These findings are of significant medical relevance, molecules targeting autophagy are currently being tested along chemotherapeutic agents to treat PDAC and other tumors in human trials.


Asunto(s)
Carcinoma Ductal/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/genética , Neoplasias Pancreáticas/genética , Pancreatitis/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Autofagia/efectos de los fármacos , Carcinoma Ductal/etiología , Carcinoma Ductal/metabolismo , Carcinoma Ductal/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cloroquina/farmacología , Genes Reporteros , Células HEK293 , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Páncreas/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pancreatitis/complicaciones , Pancreatitis/metabolismo , Pancreatitis/patología , Proteínas Asociadas a Pancreatitis , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal
5.
Cell Death Dis ; 6: e1592, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25590802

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) is a critical health issue in the field of cancer, with few therapeutic options. Evidence supports an implication of the intratumoral microenvironment (stroma) on PDA progression. However, its contribution to the role of neuroplastic changes within the pathophysiology and clinical course of PDA, through tumor recurrence and neuropathic pain, remains unknown, neglecting a putative, therapeutic window. Here, we report that the intratumoral microenvironment is a mediator of PDA-associated neural remodeling (PANR), and we highlight factors such as 'SLIT2' (an axon guidance molecule), which is expressed by cancer-associated fibroblasts (CAFs), that impact on neuroplastic changes in human PDA. We showed that 'CAF-secreted SLIT2' increases neurite outgrowth from dorsal root ganglia neurons as well as from Schwann cell migration/proliferation by modulating N-cadherin/ß-catenin signaling. Importantly, SLIT2/ROBO signaling inhibition disrupts this stromal/neural connection. Finally, we revealed that SLIT2 expression and CAFs are correlated with neural remodeling within human and mouse PDA. All together, our data demonstrate the implication of CAFs, through the secretion of axon guidance molecule, in PANR. Furthermore, it provides rationale to investigate the disruption of the stromal/neural compartment connection with SLIT2/ROBO inhibitors for the treatment of pancreatic cancer recurrence and pain.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Cadherinas/metabolismo , Comunicación Celular/efectos de los fármacos , Compartimento Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Medios de Cultivo/farmacología , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones Desnudos , Modelos Biológicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neoplasias Pancreáticas/genética , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Células de Schwann/patología , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/patología , Transcriptoma/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , beta Catenina/metabolismo , Neoplasias Pancreáticas
6.
Cell Death Differ ; 22(1): 131-44, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25168244

RESUMEN

Tribbles pseudokinase-3 (TRIB3) has been proposed to act as an inhibitor of AKT although the precise molecular basis of this activity and whether the loss of TRIB3 contributes to cancer initiation and progression remain to be clarified. In this study, by using a wide array of in vitro and in vivo approaches, including a Trib3 knockout mouse, we demonstrate that TRIB3 has a tumor-suppressing role. We also find that the mechanism by which TRIB3 loss enhances tumorigenesis relies on the dysregulation of the phosphorylation of AKT by the mTORC2 complex, which leads to an enhanced phosphorylation of AKT on Ser473 and the subsequent hyperphosphorylation and inactivation of the transcription factor FOXO3. These observations support the notion that loss of TRIB3 is associated with a more aggressive phenotype in various types of tumors by enhancing the activity of the mTORC2/AKT/FOXO axis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Factores de Transcripción Forkhead/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Humanos , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Noqueados , Ratones Desnudos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Neoplasias/genética , Neoplasias/patología , Fosforilación/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Represoras/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/genética
7.
Cell Death Differ ; 21(7): 1107-18, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24608790

RESUMEN

Tumor Protein p53-Induced Nuclear Protein 1 (TP53INP1) is a tumor suppressor that modulates the p53 response to stress. TP53INP1 is one of the key mediators of p53 antioxidant function by promoting the p53 transcriptional activity on its target genes. TP53INP1 expression is deregulated in many types of cancers including pancreatic ductal adenocarcinoma in which its decrease occurs early during the preneoplastic development. In this work, we report that redox-dependent induction of p53 transcriptional activity is enhanced by the oxidative stress-induced SUMOylation of TP53INP1 at lysine 113. This SUMOylation is mediated by PIAS3 and CBX4, two SUMO ligases especially related to the p53 activation upon DNA damage. Importantly, this modification is reversed by three SUMO1-specific proteases SENP1, 2 and 6. Moreover, TP53INP1 SUMOylation induces its binding to p53 in the nucleus under oxidative stress conditions. TP53INP1 mutation at lysine 113 prevents the pro-apoptotic, antiproliferative and antioxidant effects of TP53INP1 by impairing the p53 response on its target genes p21, Bax and PUMA. We conclude that TP53INP1 SUMOylation is essential for the regulation of p53 activity induced by oxidative stress.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Choque Térmico/metabolismo , Estrés Oxidativo , Sumoilación , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Autofagia , Proliferación Celular , Células HEK293 , Humanos , Ligasas , Células MCF-7 , Chaperonas Moleculares/metabolismo , Oxidación-Reducción , Proteínas del Grupo Polycomb/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteína SUMO-1/metabolismo , Serina Endopeptidasas/metabolismo , Activación Transcripcional , Ubiquitina-Proteína Ligasas/metabolismo
8.
Cell Death Differ ; 19(9): 1525-35, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22421968

RESUMEN

TP53INP1 (tumor protein 53-induced nuclear protein 1) is a tumor suppressor, whose expression is downregulated in cancers from different organs. It was described as a p53 target gene involved in cell death, cell-cycle arrest and cellular migration. In this work, we show that TP53INP1 is also able to interact with ATG8-family proteins and to induce autophagy-dependent cell death. In agreement with this finding, we observe that TP53INP1, which is mainly nuclear, relocalizes in autophagosomes during autophagy where it is eventually degraded. TP53INP1-LC3 interaction occurs via a functional LC3-interacting region (LIR). Inactivating mutations of this sequence abolish TP53INP1-LC3 interaction, relocalize TP53INP1 in autophagosomes and decrease TP53INP1 ability to trigger cell death. Interestingly, TP53INP1 binds to ATG8-family proteins with higher affinity than p62, suggesting that it could partially displace p62 from autophagosomes, modifying thereby their composition. Moreover, silencing the expression of autophagy related genes (ATG5 or Beclin-1) or inhibiting caspase activity significantly decreases cell death induced by TP53INP1. These data indicate that cell death observed after TP53INP1-LC3 interaction depends on both autophagy and caspase activity. We conclude that TP53INP1 could act as a tumor suppressor by inducing cell death by caspase-dependent autophagy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia/fisiología , Proteínas Portadoras/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 5 Relacionada con la Autofagia , Familia de las Proteínas 8 Relacionadas con la Autofagia , Beclina-1 , Proteínas Portadoras/genética , Células HEK293 , Células HeLa , Proteínas de Choque Térmico/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/genética , Proteínas Asociadas a Microtúbulos/genética , Mutación , Fagosomas/genética , Fagosomas/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
9.
Oncogene ; 30(27): 3049-61, 2011 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-21339733

RESUMEN

Tumor protein 53 induced nuclear protein 1 (TP53INP1) is a p53 target gene that induces cell growth arrest and apoptosis by modulating p53 transcriptional activity. TP53INP1 interacts physically with p53 and is a major player in the p53-driven oxidative stress response. Previously, we demonstrated that TP53INP1 is downregulated in an early stage of pancreatic cancerogenesis and when restored is able to suppress pancreatic tumor development. TP53INP1 downregulation in pancreas is associated with an oncogenic microRNA miR-155. In the present work, we studied the effects of TP53INP1 on cell migration. We found that TP53INP1 inactivation correlates with increased cell migration both in vivo and in vitro. The impact of TP53INP1 expression on cell migration was studied in different cellular contexts: mouse embryonic fibroblast and different pancreatic cancer cell lines. Its expression decreases cell migration by the transcriptional downregulation of secreted protein acidic and rich in cysteine (SPARC). SPARC is a matrix cellular protein, which governs diverse cellular functions and has a pivotal role in regulating cell-matrix interactions, cellular proliferation and migration. SPARC was also showed to be upregulated in normal pancreas and in pancreatic intraepithelial neoplasia lesions in a pancreatic adenocarcinoma mouse model only in the TP53INP1-deficient animals. This novel TP53INP1 activity on the regulation of SPARC expression could explain in part its tumor suppressor function in pancreatic adenocarcinoma by modulating cellular spreading during the metastatic process.


Asunto(s)
Proteínas Portadoras/fisiología , Movimiento Celular/fisiología , Proteínas de Choque Térmico/fisiología , Osteonectina/metabolismo , Neoplasias Pancreáticas/patología , Regulación hacia Abajo , Humanos
10.
World J Surg ; 34(4): 830-5, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20145930

RESUMEN

BACKGROUND: Medullary thyroid cancer (MTC) is characterized by early regional lymph node metastasis, the presence of which represents a critical obstacle to cure. At present no molecular markers have been successfully integrated into the clinical care of sporadic MTC. The present study was designed to evaluate TP53INP1 expression in MTC and to assess its ability to guide the surgeon to the optimal extent of surgery performed with curative intent. METHODS: Thirty-eight patients with sporadic MTC were evaluated. TP53INP1 immunoexpression was studied on embedded paraffin material and on cytological smears. RESULTS: TP53INP1 was expressed in normal C cells, in C-cell hyperplasia, and in 57.9% of MTC. It was possible to identify two groups of MTC according to the proportion of TP53INP1 expressing tumor cells: group 1 from 0% to <50% and group 2 from 50% to 100% of positive cells. Patients with a decreased expression of TP53INP1 (group 1) had a lower rate of nodal metastasis (18.8% versus 63.4% in group 2; P = 0.009), with only minimal lymph node involvement per N1 patient (2.7% of positive lymph nodes versus 22.9%; P < 0.001) and better outcomes (100% of biochemical cure versus 55.5%; P < 0.001). Patients with distant metastases were only observed in group 2. Cytological samples exhibit similar results to their embedded counterparts. CONCLUSIONS: TP53INP1 immunoexpression appears to be a clinical predictor of lymph node metastasis in MTC. The evaluation of TP53INP1 expression may guide the extent of lymph node dissection in the clinically node-negative neck. These findings require prospective validation.


Asunto(s)
Carcinoma Medular/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Choque Térmico/metabolismo , Neoplasias de la Tiroides/metabolismo , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Western Blotting , Carcinoma Medular/patología , Carcinoma Medular/cirugía , Distribución de Chi-Cuadrado , Femenino , Humanos , Inmunohistoquímica , Modelos Logísticos , Escisión del Ganglio Linfático , Metástasis Linfática , Masculino , Persona de Mediana Edad , Estadísticas no Paramétricas , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/cirugía
11.
Oncogene ; 29(17): 2528-39, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20140013

RESUMEN

Vav proteins are guanine nucleotide exchange factors for Rho GTPases that regulate cell adhesion, motility, spreading and proliferation in response to growth factor signalling. In this work, we show that Vav2 expression delayed epidermal growth factor receptor (EGFR) internalization and degradation, and enhanced EGFR, ERK and Akt phosphorylations. This effect of Vav2 on EGFR degradation is dependent on its guanine nucleotide exchange function. Knockdown of Vav2 in HeLa cells enhanced EGFR degradation and reduced cell proliferation. epidermal growth factor stimulation led to co-localization of Vav2 with EGFR and Rab5 in endosomes. We further show that the effect of Vav2 on EGFR stability is modulated by its interaction with two endosome-associated proteins and require RhoA function. Thus, in this work, we report for the first time that Vav2 can regulate growth factors receptor signalling by slowing receptor internalization and degradation through its interaction with endosome-associated proteins.


Asunto(s)
Endocitosis , Receptores ErbB/metabolismo , Proteínas Proto-Oncogénicas c-vav/fisiología , Línea Celular Tumoral , Endosomas/metabolismo , Receptores ErbB/análisis , Humanos , Proteínas Proto-Oncogénicas c-vav/análisis , Proteínas de Unión al GTP rab5/análisis
12.
Cancer Lett ; 288(1): 116-23, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19631450

RESUMEN

ArgBP2 is a multi-adapter protein involved in signal transduction associated to the cytoskeleton and was shown to regulate the migration and adhesion of pancreatic cancer cells thereby modulating their tumorigenicity. Here we describe the interaction of ArgBP2 with CIP4, a new associated protein identified by yeast two-hybrid. We found that both proteins modulated their reciprocal tyrosine phosphorylation catalyzed by the non-receptor tyrosine kinase c-Abl. We observed that, like ArgBP2, CIP4 directly interacted with WAVE1 and could enhance its phosphorylation by c-Abl. ArgBP2 and CIP4 acted synergistically to increase WAVE1 tyrosine phosphorylation. Finally, we could show that CIP4 was dispensable for the ArgBP2 induced blockade of cell migration whereas its overexpression was deleterious for this important function of ArgBP2.


Asunto(s)
Movimiento Celular , Proteínas de Homeodominio/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Neoplasias Pancreáticas/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Sitios de Unión , Línea Celular Tumoral , Proteínas de Homeodominio/genética , Humanos , Proteínas Asociadas a Microtúbulos/genética , Antígenos de Histocompatibilidad Menor , Invasividad Neoplásica , Neoplasias Pancreáticas/patología , Fosforilación , Proteínas Proto-Oncogénicas c-abl/metabolismo , Interferencia de ARN , Proteínas de Unión al ARN , Transducción de Señal , Transfección , Técnicas del Sistema de Dos Híbridos , Dominios Homologos src
14.
Eur J Cell Biol ; 80(2): 156-63, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11302520

RESUMEN

Expression of the Cdx1 homeobox gene in epithelial intestinal cells promotes cellular growth and differentiation. Cdx1and the Pancreatitis Associated Protein I (PAP I) are concomitantly expressed in the epithelial cells of the lower part of the intestinal crypts. Because Cdx1 is a transcription factor and PAP I, in other tissues, is a proliferative factor, we looked for a relationship between these two proteins in the intestinal-derived IEC-6 cells. After stable transfection with a Cdx1 expression vector, they produce high levels of the PAP I transcript and protein indicating a functional link between the two genes. Demonstration of Cdx1 binding to the PAP I promoter region and suppression of PAP I induction after deletion of the corresponding sequence indicated that Cdx1 is a transcription factor controlling PAP I gene expression in intestinal cells. By infecting IEC-6 cells with adenoviruses expressing PAP I, we demonstrated that PAP I induces mitosis in these cells. On the other hand, inhibition of the PAP I expression in the IEC-6 Cdxl-expressing cells using an antisense strategy confirmed the requirement of this protein for the effect of Cdx1 on cell growth. Finally, addition of the immunopurified PAP I to the culture medium promotes cell growth of the IEC-6 cells in a dose-dependent manner. Maximal effect was obtained at 1 ng/ml. Taken together these results demonstrate that PAP I is a target of the Cdx1 homeobox gene in intestinal cells which participates in the regulation of intestinal cell growth via an autocrine and/or paracrine mechanism.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Antígenos de Neoplasias , Biomarcadores de Tumor , Proteínas de Homeodominio/metabolismo , Mucosa Intestinal/citología , Lectinas Tipo C , Factores de Transcripción/metabolismo , Proteínas de Fase Aguda/genética , Animales , División Celular , Línea Celular , Células Epiteliales/citología , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Mitosis , Proteínas Asociadas a Pancreatitis , Regiones Promotoras Genéticas , Ratas , Elementos de Respuesta , Factores de Transcripción/genética , Activación Transcripcional
15.
Gastroenterology ; 119(3): 816-28, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10982776

RESUMEN

BACKGROUND & AIMS: Tumor necrosis factor (TNF)-alpha contributes to the development of acute pancreatitis. Because TNF-alpha is involved in the control of apoptosis, we studied its interaction with the pancreatic apoptotic pathway. METHODS: Pancreatic acinar AR4-2J cells were used. Apoptosis was monitored by morphologic and biochemical criteria. RESULTS: TNF-alpha induced apoptosis in AR4-2J cells. Induction was strongly enhanced in cells treated with actinomycin D, suggesting that TNF-alpha activated concomitantly an antiapoptotic mechanism through newly synthesized proteins. This mechanism involved activation of nuclear factor-kappaB (NF-kappaB) and mitogen-activated protein (MAP) kinases because their inhibition worsened TNF-alpha-induced apoptosis. The antiapoptotic pancreatitis-associated protein (PAP) I is a candidate for mediating TNF-alpha activity. Its expression is induced by TNF-alpha, and cells overexpressing PAP I show significantly less apoptosis on exposure to TNF-alpha. We examined whether TNF-alpha induction of PAP I expression was mediated by NF-kappaB or MAP kinases by using specific inhibitors of both pathways. Inhibition of NF-kappaB had no effect. However, inhibitors of MEK1 eliminated PAP I induction. CONCLUSIONS: TNF-alpha induces concomitantly proapoptotic and antiapoptotic mechanisms in pancreatic AR4-2J cells. Antiapoptotic mechanisms are mediated by NF-kappaB and MAP kinases, and PAP I is one of the effectors of apoptosis inhibition.


Asunto(s)
Proteínas de Fase Aguda/fisiología , Antígenos de Neoplasias , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor , Lectinas Tipo C , FN-kappa B/antagonistas & inhibidores , Páncreas/efectos de los fármacos , Páncreas/fisiopatología , Factor de Necrosis Tumoral alfa/farmacología , Proteínas de Fase Aguda/metabolismo , Animales , Apoptosis/fisiología , Línea Celular , Dactinomicina/farmacología , Sinergismo Farmacológico , MAP Quinasa Quinasa 1 , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Páncreas/patología , Proteínas Asociadas a Pancreatitis , Proteínas Serina-Treonina Quinasas/fisiología , Ratas , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
16.
Exp Cell Res ; 246(2): 368-75, 1999 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9925752

RESUMEN

Endothelial cells form the inner lining of blood and lymphatic vessels. In mice, only tumors of the blood vessel endothelium (haemangiomas) have been thus far reported. Here we describe a highly reproducible method for the induction of benign tumors of the lymphatic endothelial cells (lymphangiomas) in mice by intraperitoneal injection of incomplete Freund's adjuvant. Morphological and histopathological studies of the lesions revealed the presence of cells at various levels of vascular development. The lymphangiomas developed in the peritoneal cavity and expressed the endothelial markers CD31/PECAM (platelet endothelial cell adhesion molecule), CD54/ICAM-1 (InterCellular Adhesion Molecule-1), and CD102/ICAM-2, as well as the vascular endothelial growth factor (VEGF) receptor Flk-1, the endothelial cell specific receptors Tie-1 and Tie-2 and the lymphatic endothelial cell specific Flt4 receptor as shown by in situ hybridization. The Flk-1 and Flt4 receptors were also identified in immunoblots of the tumors and in cells cultured from them. When induced in beta-galactosidase knock-in Flt4(+/-) mice, the tumor endothelia could be stained blue in a number of tumor cells although the staining was of lower intensity than in normal lymphatic vessels. The tumor-derived cells could be propagated in vitro and they spontaneously differentiated, forming vessel-like structures. Murine lymphangiomas thus represent a highly reproducible and convenient source of lymphatic endothelial cells.


Asunto(s)
Carcinógenos/toxicidad , Adyuvante de Freund/toxicidad , Linfangioma/inducido químicamente , Neoplasias Peritoneales/inducido químicamente , Animales , Biomarcadores de Tumor/biosíntesis , División Celular , Endotelio Linfático , Expresión Génica , Inyecciones Intraperitoneales , Linfangioma/metabolismo , Linfangioma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/patología , Proteínas Proto-Oncogénicas/genética , Conejos , Proteínas Tirosina Quinasas Receptoras/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular
17.
Biochem Biophys Res Commun ; 251(3): 710-3, 1998 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-9790974

RESUMEN

We report in this paper that cycloheximide induces PAP mRNA expression in the pancreatic acinar cell line AR4-2J in a dose- and time-dependent manner. We analyzed whether stabilization of the PAP mRNA or the direct induction of its transcription contributed to the induction of PAP mRNA expression by the drug. We first infected the cells, which do not express PAP mRNA constitutively, with a recombinant adenovirus in which the PAP cDNA was subcloned downstream of the CMV promotor, to obtain high levels of transcript. Then, transcription was pharmacologically blocked, the cells were treated with cycloheximide, and the PAP mRNA concentration was monitored over 8 h by Northern blot. PAP mRNA concentration remained unchanged for 4 h and then decreased in both cycloheximide-treated and control cells, ruling out a significant contribution of posttranscriptional regulation in cycloheximide induction. Direct regulation of gene transcription is therefore likely and we investigated whether it could involve ADP-ribosylation. Cycloheximide-induced cells were treated with two chemical inhibitors of poly(ADP-ribose) polymerase. 3-Aminobenzamide inhibited 75% of PAP gene induction and 4-hydroxyquinazolone, the highly specific inhibitor of the enzyme, blocked almost completely PAP expression, suggesting that ADP-ribosylation was indeed required for the upregulation of PAP gene expression by cycloheximide.


Asunto(s)
Proteínas de Fase Aguda/biosíntesis , Adenosina Difosfato Ribosa/metabolismo , Antígenos de Neoplasias , Biomarcadores de Tumor , Cicloheximida/farmacología , Lectinas Tipo C , Páncreas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas de Fase Aguda/genética , Benzamidas/farmacología , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Semivida , Modelos Genéticos , Páncreas/citología , Páncreas/efectos de los fármacos , Proteínas Asociadas a Pancreatitis , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Transcripción Genética , Activación Transcripcional
18.
Oncology ; 55(5): 421-5, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9732219

RESUMEN

This study evaluated the significance of serum pancreatitis-associated protein (PAP) assay, as a marker of hepatocellular carcinoma (HCC), in comparison and combined with alpha-fetoprotein (AFP) assay. Sixty-five patients with HCC, 59 with liver cirrhosis (LC) and 68 asymptomatic controls (C) were studied. PAP and AFP values significantly increased from C to LC and HCC group (p < 0.0001). The area under receiver-operating characteristic (ROC) curve for the two markers was not statistically different. At 100% specificity, ROC analysis gave a cut-off level for AFP of 166 IU/l with 40% sensitivity, and a cut-off level of 240 microg/l for PAP with 23% sensitivity. Diagnostic accuracy of combined AFP and PAP assay was significantly higher than AFP alone. Sensitivity according to tumor size also improved using the combined assay, especially for tumors <5 cm. Stepwise logistic regression indicated that AFP, but not PAP, was associated with an increased risk of developing HCC. These data confirm that PAP production is increased only in some cases of HCC and that the combined PAP and AFP assays do not significantly improve specificity over AFP assay alone. Consequently, PAP assay can only be recommended in cases of justified suspicion of HCC with negative AFP.


Asunto(s)
Proteínas de Fase Aguda/análisis , Antígenos de Neoplasias , Biomarcadores de Tumor/sangre , Carcinoma Hepatocelular/sangre , Lectinas Tipo C , Neoplasias Hepáticas/sangre , alfa-Fetoproteínas/análisis , Adulto , Anciano , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Proteínas Asociadas a Pancreatitis , Sensibilidad y Especificidad
19.
Gastroenterology ; 114(4): 808-16, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9516402

RESUMEN

BACKGROUND & AIMS: Free radicals are involved in the pathogenesis of acute pancreatitis, during which pancreatitis-associated protein (PAP)-I is overexpressed. We explored whether PAP-I expression could be induced by oxidative stress and whether it could affect apoptosis. METHODS: AR4-2J cells were exposed to H2O2 or menadione, and PAP-I messenger RNA (mRNA) expression was analyzed by Northern blotting. RESULTS: Maximal expression was observed with 0.1 mmol/L H2O2 or with 0.05 mmol/L menadione. Induction was detectable after 12 hours, reached a climax at 18 hours, and then decreased. Pretreatment of the cells with pyrrolidine dithiocarbamate completely abolished PAP-I mRNA induction, suggesting involvement of NFkappaB in the signaling pathway. These findings were confirmed in transient transfection assays using a plasmid containing the PAP-I promoter linked to the chloramphenicol acetyltransferase reporter gene. Then the relationship between PAP-I induction and protection against cell damage during oxidative stress was considered. Constitutive PAP-I expression in AR4-2J cells after transfection with PAP-I complementary DNA conferred significant resistance to apoptosis induced by low doses of H2O2 but not to necrosis induced by high doses of H2O2. CONCLUSIONS: These results suggest that during oxidative stress, PAP-I might be part of a mechanism of pancreatic cell protection against apoptosis.


Asunto(s)
Proteínas de Fase Aguda/biosíntesis , Antígenos de Neoplasias , Apoptosis , Biomarcadores de Tumor , Lectinas Tipo C , Páncreas/metabolismo , Proteínas de Fase Aguda/genética , Antioxidantes/farmacología , Línea Celular , Radicales Libres , Peróxido de Hidrógeno/toxicidad , Estrés Oxidativo , Páncreas/efectos de los fármacos , Páncreas/patología , Proteínas Asociadas a Pancreatitis , Regiones Promotoras Genéticas , ARN Mensajero/análisis
20.
Arch Biochem Biophys ; 340(1): 111-6, 1997 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-9126283

RESUMEN

Previous analysis of the rat PAP I promoter indicated that the region between nt -180 and -81 possessed silencer activity in cells that did not express PAP I. Based on this finding, we performed a series of experiments to characterize functionally that region and analyze the nuclear proteins interacting with it. Transient transfection assays were conducted in the fibroblast Rat2 cell line, in which PAP I is not expressed, and in the pancreatic cell line AR-42J, expressing PAP I, using the CAT gene as reporter. Experiments in Rat2 cells revealed that the sequence with silencer activity was located within the rep27 region (position -180/-153). Suppressor activity was observed when rep27 was inserted upstream from the core PAP I promoter, in both orientations. By contrast, inserting the rep27 region in front of the promoters of SV40 or thymidine kinase did not affect or weakly enhanced CAT activity. Suppressor activity is therefore position-independent and promoter-dependent. In pancreatic AR-42J cells, rep27 act as a positive element but did not alter CAT expression when inserted in front of the core PAP I promoter or heterologous promoters. Electrophoretic mobility shift assays allowed identification of specific DNA-protein complexes. The shifted complex migrated at the same position with both Rat2 and AR-42J nuclear extracts. Moreover, similar band shifts were obtained with rat nuclear extracts from healthy pancreas, pancreas with acute pancreatitis, liver, kidney, spleen, and small intestine. Results suggest that the rep27 cis-acting element contributes to the tissue specific expression of the PAP I gene. That activity could be mediated by the synergistic action of several transcription factors, one of which being present in all cells.


Asunto(s)
Proteínas de Fase Aguda/genética , Antígenos de Neoplasias , Biomarcadores de Tumor , Lectinas Tipo C , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Línea Celular , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Familia de Multigenes , Proteínas Nucleares/metabolismo , Pancreatitis/metabolismo , Proteínas Asociadas a Pancreatitis , Regiones Promotoras Genéticas , Ratas , Distribución Tisular , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA