Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 56, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397922

RESUMEN

RAC1 activity is critical for intestinal homeostasis, and is required for hyperproliferation driven by loss of the tumour suppressor gene Apc in the murine intestine. To avoid the impact of direct targeting upon homeostasis, we reasoned that indirect targeting of RAC1 via RAC-GEFs might be effective. Transcriptional profiling of Apc deficient intestinal tissue identified Vav3 and Tiam1 as key targets. Deletion of these indicated that while TIAM1 deficiency could suppress Apc-driven hyperproliferation, it had no impact upon tumourigenesis, while VAV3 deficiency had no effect. Intriguingly, deletion of either gene resulted in upregulation of Vav2, with subsequent targeting of all three (Vav2-/- Vav3-/- Tiam1-/-), profoundly suppressing hyperproliferation, tumourigenesis and RAC1 activity, without impacting normal homeostasis. Critically, the observed RAC-GEF dependency was negated by oncogenic KRAS mutation. Together, these data demonstrate that while targeting RAC-GEF molecules may have therapeutic impact at early stages, this benefit may be lost in late stage disease.


Asunto(s)
Carcinogénesis/metabolismo , Carcinogénesis/patología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Intestinos/patología , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Carcinogénesis/genética , Homeostasis , Intestinos/ultraestructura , Ratones Noqueados , Mutación/genética , Especificidad de Órganos , Fenotipo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/metabolismo , Regulación hacia Arriba , Vía de Señalización Wnt
2.
Nat Commun ; 11(1): 445, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974352

RESUMEN

The number of patients diagnosed with chronic bile duct disease is increasing and in most cases these diseases result in chronic ductular scarring, necessitating liver transplantation. The formation of ductular scaring affects liver function; however, scar-generating portal fibroblasts also provide important instructive signals to promote the proliferation and differentiation of biliary epithelial cells. Therefore, understanding whether we can reduce scar formation while maintaining a pro-regenerative microenvironment will be essential in developing treatments for biliary disease. Here, we describe how regenerating biliary epithelial cells express Wnt-Planar Cell Polarity signalling components following bile duct injury and promote the formation of ductular scars by upregulating pro-fibrogenic cytokines and positively regulating collagen-deposition. Inhibiting the production of Wnt-ligands reduces the amount of scar formed around the bile duct, without reducing the development of the pro-regenerative microenvironment required for ductular regeneration, demonstrating that scarring and regeneration can be uncoupled in adult biliary disease and regeneration.


Asunto(s)
Enfermedades de los Conductos Biliares/patología , Colangitis Esclerosante/patología , Cicatriz/patología , Vía de Señalización Wnt , Animales , Proteína Axina/genética , Proteína Axina/metabolismo , Enfermedades de los Conductos Biliares/inducido químicamente , Enfermedades de los Conductos Biliares/metabolismo , Conductos Biliares/citología , Polaridad Celular , Colangitis Esclerosante/metabolismo , Cicatriz/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Piridinas/toxicidad , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt-5a/metabolismo
5.
Oncogene ; 37(49): 6368-6382, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30061636

RESUMEN

The life expectancy for pancreatic cancer patients has seen no substantial changes in the last 40 years as very few and mostly just palliative treatments are available. As the five years survival rate remains around 5%, the identification of novel pharmacological targets and development of new therapeutic strategies are urgently needed. Here we demonstrate that inhibition of the G protein-coupled receptor GPR55, using genetic and pharmacological approaches, reduces pancreatic cancer cell growth in vitro and in vivo and we propose that this may represent a novel strategy to inhibit pancreatic ductal adenocarcinoma (PDAC) progression. Specifically, we show that genetic ablation of Gpr55 in the KRASWT/G12D/TP53WT/R172H/Pdx1-Cre+/+ (KPC) mouse model of PDAC significantly prolonged survival. Importantly, KPC mice treated with a combination of the GPR55 antagonist Cannabidiol (CBD) and gemcitabine (GEM, one of the most used drugs to treat PDAC), survived nearly three times longer compared to mice treated with vehicle or GEM alone. Mechanistically, knockdown or pharmacologic inhibition of GPR55 reduced anchorage-dependent and independent growth, cell cycle progression, activation of mitogen-activated protein kinase (MAPK) signalling and protein levels of ribonucleotide reductases in PDAC cells. Consistent with this, genetic ablation of Gpr55 reduced proliferation of tumour cells, MAPK signalling and ribonucleotide reductase M1 levels in KPC mice. Combination of CBD and GEM inhibited tumour cell proliferation in KPC mice and it opposed mechanisms involved in development of resistance to GEM in vitro and in vivo. Finally, we demonstrate that the tumour suppressor p53 regulates GPR55 protein expression through modulation of the microRNA miR34b-3p. Our results demonstrate the important role played by GPR55 downstream of p53 in PDAC progression. Moreover our data indicate that combination of CBD and GEM, both currently approved for medical use, might be tested in clinical trials as a novel promising treatment to improve PDAC patients' outcome.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/patología , Receptores de Cannabinoides/metabolismo , Animales , Antineoplásicos/farmacología , Cannabidiol/farmacología , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Ratones , Ratones Noqueados , Neoplasias Pancreáticas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Gemcitabina
6.
Nat Commun ; 9(1): 1132, 2018 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-29556067

RESUMEN

Many epithelial stem cell populations follow a pattern of stochastic stem cell divisions called 'neutral drift'. It is hypothesised that neutral competition between stem cells protects against the acquisition of deleterious mutations. Here we use a Porcupine inhibitor to reduce Wnt secretion at a dose where intestinal homoeostasis is maintained despite a reduction of Lgr5+ stem cells. Functionally, there is a marked acceleration in monoclonal conversion, so that crypts become rapidly derived from a single stem cell. Stem cells located further from the base are lost and the pool of competing stem cells is reduced. We tested whether this loss of stem cell competition would modify tumorigenesis. Reduction of Wnt ligand secretion accelerates fixation of Apc-deficient cells within the crypt leading to accelerated tumorigenesis. Therefore, ligand-based Wnt signalling influences the number of stem cells, fixation speed of Apc mutations and the speed and likelihood of adenoma formation.


Asunto(s)
Carcinogénesis/metabolismo , Transformación Celular Neoplásica/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Células Madre/citología , Células Madre/metabolismo , Vía de Señalización Wnt , Aciltransferasas/antagonistas & inhibidores , Adenoma/etiología , Adenoma/metabolismo , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Carcinogénesis/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Inhibidores Enzimáticos/farmacología , Mucosa Intestinal/efectos de los fármacos , Ligandos , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pirazinas/farmacología , Piridinas/farmacología , Células Madre/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
7.
Oncogene ; 37(8): 982-992, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29106388

RESUMEN

NUAK1 is a member of the AMPK-related family of kinases. Recent evidence suggests that NUAK1 is an important regulator of cell adhesion and migration, cellular and organismal metabolism, and regulation of TAU stability. As such, NUAK1 may play key roles in multiple diseases ranging from neurodegeneration to diabetes and metastatic cancer. Previous work revealed a crucial role for NUAK1 in supporting viability of tumour cells specifically when MYC is overexpressed. This role is surprising, given that NUAK1 is activated by the tumour suppressor LKB1. Here we show that, in tumour cells lacking LKB1, NUAK1 activity is maintained by an alternative pathway involving calcium-dependent activation of PKCα. Calcium/PKCα-dependent activation of NUAK1 supports engagement of the AMPK-TORC1 metabolic checkpoint, thereby protecting tumour cells from MYC-driven cell death, and indeed, MYC selects for this pathway in part via transcriptional regulation of PKCα and ITPR. Our data point to a novel role for calcium in supporting tumour cell viability and clarify the synthetic lethal interaction between NUAK1 and MYC.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Calcio/metabolismo , Regulación Neoplásica de la Expresión Génica , Osteosarcoma/patología , Proteína Quinasa C-alfa/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Señalización del Calcio , Proliferación Celular , Células HeLa , Humanos , Osteosarcoma/genética , Osteosarcoma/metabolismo , Fosforilación , Proteína Quinasa C-alfa/genética , Proteínas Quinasas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Represoras/genética , Células Tumorales Cultivadas
8.
Mucosal Immunol ; 10(2): 352-360, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27435106

RESUMEN

Thiopurines are commonly used drugs in the therapy of Crohn's disease, but unfortunately only show a 30% response rate. The biological basis for the thiopurine response is unclear, thus hampering patient selection prior to treatment. A genetic risk factor associated specifically with Crohn's disease is a variant in ATG16L1 that reduces autophagy. We have previously shown that autophagy is involved in dendritic cell (DC)-T-cell interactions and cytoskeletal regulation. Here we further investigated the role of autophagy in DC cytoskeletal modulation and cellular trafficking. Autophagy-deficient DC displayed loss of filopodia, altered podosome distribution, and increased membrane ruffling, all consistent with increased cellular adhesion. Consequently, autophagy-deficient DC showed reduced migration. The cytoskeletal aberrations were mediated through hyperactivation of Rac1, a known thiopurine target. Indeed thiopurines restored the migratory defects in autophagy-deficient DC. Clinically, the ATG16L1 risk variant associated with increased response to thiopurine treatment in patients with Crohn's disease but not ulcerative colitis. These results suggest that the association between ATG16L1 and Crohn's disease is mediated at least in part through Rac1 hyperactivation and subsequent defective DC migration. As this phenotype can be corrected using thiopurines, ATG16L1 genotyping may be useful in the identification of patients that will benefit most from thiopurine treatment.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia , Enfermedad de Crohn/inmunología , Células Dendríticas/fisiología , Proteína de Unión al GTP rac1/metabolismo , Alelos , Animales , Autofagia/genética , Proteínas Relacionadas con la Autofagia/genética , Estructuras de la Membrana Celular/patología , Movimiento Celular , Células Cultivadas , Colitis Ulcerosa/tratamiento farmacológico , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/genética , Citoesqueleto/metabolismo , Células Dendríticas/patología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Mercaptopurina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo Genético , ARN Interferente Pequeño/genética , Riesgo
9.
Br J Cancer ; 113(1): 1-5, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26110974

RESUMEN

Colorectal cancer (CRC) is one of the most common cancers in the western world and is characterised by deregulation of the Wnt signalling pathway. Mutation of the adenomatous polyposis coli (APC) tumour suppressor gene, which encodes a protein that negatively regulates this pathway, occurs in almost 80% of CRC cases. The progression of this cancer from an early adenoma to carcinoma is accompanied by a well-characterised set of mutations including KRAS, SMAD4 and TP53. Using elegant genetic models the current paradigm is that the intestinal stem cell is the origin of CRC. However, human histology and recent studies, showing marked plasticity within the intestinal epithelium, may point to other cells of origin. Here we will review these latest studies and place these in context to provide an up-to-date view of the cell of origin of CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Predisposición Genética a la Enfermedad , Intestinos/patología , Células Madre/patología , Neoplasias Colorrectales/genética , Humanos , Mutación
10.
Oncogene ; 34(31): 4118-29, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-25347740

RESUMEN

Disruption of Apc (adenomatous polyposis coli) within hepatocytes activates Wnt signalling, perturbs differentiation and ultimately leads to neoplasia. Apc negatively regulates Wnt signalling but is also involved in organizing the cytoskeleton and may have a role in chromosome segregation. In vitro studies have implicated Apc in the control of genomic stability. However, the relevance of this data has been questioned in vivo as Apc is lost earlier than the onset of genomic instability. Here we analyse the relationship between immediate loss of Apc and the acquisition of genomic instability in hepatocytes. We used Cre-lox technology to inactivate Apc and in combination with p53 in vivo, to define the consequences of gene loss on cell cycle regulation, proliferation, death and aneuploidy. We show that, although Apc loss leads to increased proliferation, it also leads to increased apoptosis, the accumulation of p53, p21 and markers of double-strand breaks and DNA repair. Flow cytometry revealed an increased 4N DNA content, consistent with a G2 arrest. Levels of anaphase bridges were also elevated, implicating failed chromosome segregation. This was accompanied by an increase in centrosome number, which demonstrates a role for Apc in maintaining euploidy. To address the role of p53 in these processes, we analysed combined loss of Apc and p53, which led to a further increase in proliferation, cell death, DNA damages and repair and a bypass of G2 arrest than was observed with Apc loss. However, we observed only a marginal effect on anaphase bridges and centrosome number, which could be due to increased cell death. Our data therefore establishes, in an in vivo setting, that APC loss leads to a DNA damage signature and genomic instability in the liver and that additional loss of p53 leads to an increase in the DNA damage signal but not to an immediate increase in the genomic instability phenotype.


Asunto(s)
Daño del ADN/genética , Epistasis Genética , Genes APC/fisiología , Genes p53/fisiología , Inestabilidad Genómica/genética , Hepatocitos/patología , Animales , Proliferación Celular/genética , Células Cultivadas , Reparación del ADN/genética , Femenino , Hepatomegalia/genética , Hepatomegalia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , beta-naftoflavona
11.
Cell Death Differ ; 21(6): 956-66, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24583641

RESUMEN

Recent studies have suggested that C-MYC may be an excellent therapeutic cancer target and a number of new agents targeting C-MYC are in preclinical development. Given most therapeutic regimes would combine C-MYC inhibition with genotoxic damage, it is important to assess the importance of C-MYC function for DNA damage signalling in vivo. In this study, we have conditionally deleted the c-Myc gene in the adult murine intestine and investigated the apoptotic response of intestinal enterocytes to DNA damage. Remarkably, c-Myc deletion completely abrogated the immediate wave of apoptosis following both ionizing irradiation and cisplatin treatment, recapitulating the phenotype of p53 deficiency in the intestine. Consistent with this, c-Myc-deficient intestinal enterocytes did not upregulate p53. Mechanistically, this was linked to an upregulation of the E3 Ubiquitin ligase Mdm2, which targets p53 for degradation in c-Myc-deficient intestinal enterocytes. Further, low level overexpression of c-Myc, which does not impact on basal levels of apoptosis, elicited sustained apoptosis in response to DNA damage, suggesting c-Myc activity acts as a crucial cell survival rheostat following DNA damage. We also identify the importance of MYC during DNA damage-induced apoptosis in several other tissues, including the thymus and spleen, using systemic deletion of c-Myc throughout the adult mouse. Together, we have elucidated for the first time in vivo an essential role for endogenous c-Myc in signalling DNA damage-induced apoptosis through the control of the p53 tumour suppressor protein.


Asunto(s)
Apoptosis/genética , Enterocitos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Enterocitos/efectos de los fármacos , Enterocitos/efectos de la radiación , Humanos , Ratones , Proteínas Proto-Oncogénicas c-myc/genética , Radiación Ionizante
12.
Oncogene ; 33(25): 3325-33, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23873029

RESUMEN

Many tumours harbour mutations in the p53 tumour-suppressor gene that result in the expression of a mutant p53 protein. This mutant p53 protein has, in most cases, lost wild-type transcriptional activity and can also acquire novel functions in promoting invasion and metastasis. One of the mechanisms underlying these novel functions involves the ability of the mutant p53 to interfere with other transcription factors, including the p53 family protein TAp63. To investigate whether simultaneous depletion of both p53 and TAp63 can recapitulate the effect of mutant p53 expression in vivo, we used a mouse model of pancreatic cancer in which the expression of mutant p53 resulted in the rapid appearance of primary tumours and metastases. As shown previously, loss of one allele of wild-type (WT) p53 accelerated tumour development. A change of one WT p53 allele into mutant p53 did not further accelerate tumour development, but did promote the formation of metastasis. By contrast, loss of TAp63 did not significantly accelerate tumour development or metastasis. However, simultaneous depletion of p53 and TAp63 led to both rapid tumour development and metastatic potential, although the incidence of metastases remained lower than that seen in mutant p53-expressing tumours. TAp63/p53-null cells derived from these mice also showed an enhanced ability to scatter and invade in tissue culture as was observed in mutant p53 cells. These data suggest that depletion of TAp63 in a p53-null tumour can promote metastasis and recapitulate-to some extent-the consequences of mutant p53 expression.


Asunto(s)
Neoplasias Pancreáticas/patología , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Animales , Línea Celular Tumoral , Humanos , Ratones , Mutación , Metástasis de la Neoplasia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo
13.
Br J Cancer ; 108(5): 997-1003, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23385734

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy associated with <5% 5-year survival, in which standard chemotherapeutics have limited benefit. The disease is associated with significant intra- and peritumoral inflammation and failure of protective immunosurveillance. Indeed, inflammatory signals are implicated in both tumour initiation and tumour progression. The major pathways regulating PDAC-associated inflammation are now being explored. Activation of leukocytes, and upregulation of cytokine and chemokine signalling pathways, both have been shown to modulate PDAC progression. Therefore, targeting inflammatory pathways may be of benefit as part of a multi-target approach to PDAC therapy. This review explores the pathways known to modulate inflammation at different stages of tumour development, drawing conclusions on their potential as therapeutic targets in PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/terapia , Vigilancia Inmunológica , Inflamación/terapia , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/terapia , Animales , Carcinoma Ductal Pancreático/patología , Quimiocinas/metabolismo , Citocinas/metabolismo , Humanos , Macrófagos/inmunología , Terapia Molecular Dirigida , Mutación , Metástasis de la Neoplasia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Pronóstico , Transducción de Señal , Escape del Tumor , Microambiente Tumoral/inmunología
14.
Oncogene ; 32(16): 2048-57, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22665058

RESUMEN

A role for WNT signalling in gastric carcinogenesis has been suggested due to two major observations. First, patients with germline mutations in adenomatous polyposis coli (APC) are susceptible to stomach polyps and second, in gastric cancer, WNT activation confers a poor prognosis. However, the functional significance of deregulated WNT signalling in gastric homoeostasis and cancer is still unclear. In this study we have addressed this by investigating the immediate effects of WNT signalling activation within the stomach epithelium. We have specifically activated the WNT signalling pathway within the mouse adult gastric epithelium via deletion of either glycogen synthase kinase 3 (GSK3) or APC or via expression of a constitutively active ß-catenin protein. WNT pathway deregulation dramatically affects stomach homoeostasis at very short latencies. In the corpus, there is rapid loss of parietal cells with fundic gland polyp (FGP) formation and adenomatous change, which are similar to those observed in familial adenomatous polyposis. In the antrum, adenomas occur from 4 days post-WNT activation. Taken together, these data show a pivotal role for WNT signalling in gastric homoeostasis, FGP formation and adenomagenesis. Loss of the parietal cell population and corresponding FGP formation, an early event in gastric carcinogenesis, as well as antral adenoma formation are immediate effects of nuclear ß-catenin translocation and WNT target gene expression. Furthermore, our inducible murine model will permit a better understanding of the molecular changes required to drive tumourigenesis in the stomach.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Transformación Celular Neoplásica/metabolismo , Neoplasias Gástricas/metabolismo , Proteínas Wnt/metabolismo , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Animales , Diferenciación Celular/fisiología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Transgenes , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
15.
Oncogene ; 32(10): 1252-65, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22580601

RESUMEN

Tumour-derived mutant p53 proteins promote invasion, in part, by enhancing Rab coupling protein (RCP)-dependent receptor recycling. Here we identified MET as an RCP-binding protein and showed that mutant p53 promoted MET recycling. Mutant p53-expressing cells were more sensitive to hepatocyte growth factor, the ligand for MET, leading to enhanced MET signalling, invasion and cell scattering that was dependent on both MET and RCP. In cells expressing the p53 family member TAp63, inhibition of TAp63 also lead to cell scattering and MET-dependent invasion. However, in cells that express very low levels of TAp63, the ability of mutant p53 to promote MET-dependent cell scattering was independent of TAp63. Taken together, our data show that mutant p53 can enhance MET signalling to promote cell scattering and invasion through both TAp63-dependent and -independent mechanisms. MET has a predominant role in metastatic progression and the identification of mechanisms through which mutations in p53 can drive MET signalling may help to identify and direct therapy.


Asunto(s)
Mutación , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Células HCT116 , Células HT29 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Invasividad Neoplásica , Fosforilación , Transducción de Señal , Factores de Transcripción/metabolismo , Transfección , Proteínas Supresoras de Tumor/metabolismo
16.
Br J Cancer ; 107(9): 1554-63, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23093296

RESUMEN

BACKGROUND: Cell line models suggest that activation of NFκB is associated with progression of prostate cancer. This pathway may be a therapeutic target if these observations translate to clinical specimens. METHODS: Immunohistochemistry measured NFκBp65 (p65), NFκBp65 nuclear localisation signal (NLS), NFκBp65 phosphorylated at ser 276 (p65(ser276)), NFκBp65 phosphorylated at ser 536 (p65(ser536)), IκBα phosphorylated at ser 32/36 (pIκBα(ser32/36)) and MMP-9 protein expression in 61 matched hormone naive prostate cancer (HNPC) and castrate-resistant prostate cancer (CRPC) tumours. Animal and cell models were used to investigate the role of NFκB inhibition in prostate carcinogenesis. RESULTS: In HNPC tumours, NLS expression significantly associated with a shorter time to disease recurrence and disease-specific death. In CRPC tumours p65, pIκBα(ser32/36) and MMP-9 expression significantly associated with shorter time to death from disease recurrence and shorter disease-specific death. MMP-9 and pIκBα(ser32/36) expression significantly associated with metastases at recurrence and were independent of Gleason sum and prostate-specific antigen at recurrence. Expression of phosphorylated Akt was associated with increased p65 activation in mouse models and inhibition of NFκB in LNCaP cells significantly reduced cellular proliferation and induced apoptosis. CONCLUSION: These results provide further evidence that the NFκB pathway could be exploited as a target for CRPC.


Asunto(s)
FN-kappa B/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Anciano , Animales , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Estudios de Cohortes , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Proteínas I-kappa B/metabolismo , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Inhibidor NF-kappaB alfa , FN-kappa B/biosíntesis , FN-kappa B/genética , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Fosforilación , Neoplasias de la Próstata/genética , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Regulación hacia Arriba
17.
Acta Physiol (Oxf) ; 204(1): 137-43, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21439026

RESUMEN

Adult stem cells are crucial for normal tissue homeostasis and regeneration upon damage. Deregulated stem cell proliferation and/or differentiation have been linked to the formation and progression of tumours. Due to its high regenerative potential, the adult intestinal epithelium is an excellent system to study the mechanisms that underpin regeneration and transformation. Since the discovery of stem cell markers in the mammalian intestine - such as Lgr5 - and the presence of Drosophila intestinal stem cells there have been field-changing discoveries that have revolutionized our understanding of intestinal homeostasis, stem cells and transformation. We will thus present a brief overview of these advances with an emphasis on the role of canonical Wnt signalling and the lessons learned from genetic tractable model systems.


Asunto(s)
Hiperplasia/patología , Intestinos/patología , Intestinos/fisiología , Regeneración/fisiología , Células Madre/fisiología , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Humanos , Intestinos/citología , Células Madre/citología
18.
Cell Death Differ ; 18(12): 1934-43, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21637293

RESUMEN

The transcription factor signal transducer and activator of transcription 3 (STAT3) is frequently activated in human cancers. Interestingly, STAT3 also maintains the pluripotency and self-renewal of murine embryonic stem cells, and several tissue stem cell types. To investigate whether STAT3 also maintains the small-intestine crypt stem cell, we conditionally inactivated a Floxed Stat3 allele (Stat3(fl)) in murine small-intestine crypt stem cells. Following Cre recombinase expression, apoptosis increased in Stat3(fl/-) experimental crypts relative to Stat3(wt/-) controls before declining. Control Stat3(wt/-) mice carrying a Flox-STOP LacZ reporter transgene stably expressed LacZ after Cre induction. In contrast, Stat3(fl/-) intestine LacZ expression initially increased modestly, before declining to background levels. Quantitative PCRs revealed a similar transient in recombined Stat3(fl) allele levels. Long-term bromodeoxyuridine labelling directly demonstrated that functional STAT3 is required for +4 to +6 region label-retaining small-intestine stem cell survival. Rapid clearance of recombined Stat3(fl/-) cells involves apoptosis potentially induced by elevated c-Myc in non-recombined cells and involves elevated p53 expression and caspase 3 activation. Intriguingly, Stat3(fl/-) intestine recombination triggered dramatically upregulated polycomb transcriptional repressor Bmi1 - potentially accelerating recombined crypt repopulation. In summary, STAT3 activity is absolutely required for small-intestine crypt stem cell survival at both the +4 to +6 label-retaining and crypt base columnar cell locations.


Asunto(s)
Supervivencia Celular , Mucosa Intestinal/citología , Intestino Delgado/citología , Factor de Transcripción STAT3/fisiología , Células Madre/fisiología , Animales , Apoptosis , Caspasa 3/metabolismo , Movimiento Celular , Proliferación Celular , Activación Enzimática , Expresión Génica , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
19.
Cell Death Dis ; 2: e124, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21368895

RESUMEN

Mutations in the Ras family of proteins (predominantly in H-Ras) occur in approximately 40% of urothelial cell carcinoma (UCC). However, relatively little is known about subsequent mutations/pathway alterations that allow tumour progression. Indeed, expressing mutant H-Ras within the mouse bladder does not lead to tumour formation, unless this is expressed at high levels. The Wnt signalling pathway is deregulated in approximately 25% of UCC, so we examined if this correlated with the activation of MAPK signalling in human UCC and found a significant correlation. To test the functional significance of this association we examined the impact of combining Ras mutation (H-Ras(Q61L) or K-Ras(G12D)) with an activating ß-catenin mutation within the mouse bladder using Cre-LoxP technology. Although alone, neither Ras mutation nor ß-catenin activation led to UCC (within 12 months), mice carrying both mutations rapidly developed UCC. Mechanistically this was associated with reduced levels of p21 with dependence on the MAPK signalling pathway. Moreover, tumours from these mice were sensitive to MEK inhibition. Importantly, in human UCC there was a negative correlation between levels of p-ERK and p21 suggesting that p21 accumulation may block tumour progression following Ras mutation. Taken together these data definitively show Ras pathway activation strongly cooperates with Wnt signalling to drive UCC in vivo.


Asunto(s)
Mutación , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , beta Catenina/metabolismo , Proteínas ras/genética , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Procesos Neoplásicos , Transducción de Señal , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , Proteínas ras/metabolismo
20.
Oncogene ; 30(2): 178-89, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20818428

RESUMEN

Although deregulation of the Wnt signalling pathway has been implicated in urothelial cell carcinoma (UCC), the functional significance is unknown. To test its importance, we have targeted expression of an activated form of ß-catenin to the urothelium of transgenic mice using Cre-Lox technology (UroIICRE(+) ß-catenin(exon3/+)). Expression of this activated form of ß-catenin led to the formation of localized hyperproliferative lesions by 3 months, which did not progress to malignancy. These lesions were characterized by a marked increase of the phosphatase and tensin homologue (PTEN) tumour suppressor protein. This appears to be a direct consequence of activating Wnt signalling in the bladder as conditional deletion of the adenomatous polyposis coli (Apc) gene within the adult bladder led rapidly to coincident ß-catenin and PTEN expression. This PTEN expression blocked proliferation. Next, we combined PTEN deficiency with ß-catenin activation and found that this caused papillary UCC. These tumours had increased pAKT signalling and were dependent on mammalian target of rapamycin (mTOR). Importantly, in human UCC, there was a significant correlation between high levels of ß-catenin and pAKT (and low levels of PTEN). Taken together these data show that deregulated Wnt signalling has a critical role in promoting UCC, and suggests that human UCC that have high levels of Wnt and PI3 kinase signalling may be responsive to mTOR inhibition.


Asunto(s)
Carcinoma Papilar/metabolismo , Transformación Celular Neoplásica/metabolismo , Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Vejiga Urinaria/inducido químicamente , beta Catenina/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Proliferación Celular , Estudios de Cohortes , Femenino , Humanos , Hiperplasia/inducido químicamente , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/análisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Eliminación de Secuencia , Serina-Treonina Quinasas TOR/metabolismo , Vejiga Urinaria/metabolismo , Urotelio/metabolismo , Proteínas Wnt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA