Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rev ; 92(1): 75-99, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22298652

RESUMEN

Since the realization that embryonic stem cells are maintained in a pluripotent state through the interplay of a number of key signal transduction pathways, it is becoming increasingly clear that stemness and pluripotency are defined by the complex molecular convergence of these pathways. Perhaps this has most clearly been demonstrated by the capacity to induce pluripotency in differentiated cell types, so termed iPS cells. We are therefore building an understanding of how cells may be maintained in a pluripotent state, and how we may manipulate cells to drive them between committed and pluripotent compartments. However, it is less clear how cells normally pass in and out of the stem cell compartment under normal and diseased physiological states in vivo, and indeed, how important these pathways are in these settings. It is also clear that there is a potential "dark side" to manipulating the stem cell compartment, as deregulation of somatic stem cells is being increasingly implicated in carcinogenesis and the generation of "cancer stem cells." This review explores these relationships, with a particular focus on the role played by key molecular regulators of stemness in tissue repair, and the possibility that a better understanding of this control may open the door to novel repair strategies in vivo. The successful development of such strategies has the potential to replace or augment intervention-based strategies (cell replacement therapies), although it is clear they must be developed with a full understanding of how such approaches might also influence tumorigenesis.


Asunto(s)
Células Madre Embrionarias/fisiología , Células Madre Pluripotentes/fisiología , Regeneración/fisiología , Transducción de Señal/fisiología , Animales , Diferenciación Celular/fisiología , Transformación Celular Neoplásica , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos
2.
EMBO J ; 34(18): 2321-33, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26240067

RESUMEN

Wnt pathway deregulation is a common characteristic of many cancers. Only colorectal cancer predominantly harbours mutations in APC, whereas other cancer types (hepatocellular carcinoma, solid pseudopapillary tumours of the pancreas) have activating mutations in ß-catenin (CTNNB1). We have compared the dynamics and the potency of ß-catenin mutations in vivo. Within the murine small intestine (SI), an activating mutation of ß-catenin took much longer to achieve Wnt deregulation and acquire a crypt-progenitor cell (CPC) phenotype than Apc or Gsk3 loss. Within the colon, a single activating mutation of ß-catenin was unable to drive Wnt deregulation or induce the CPC phenotype. This ability of ß-catenin mutation to differentially transform the SI versus the colon correlated with higher expression of E-cadherin and a higher number of E-cadherin:ß-catenin complexes at the membrane. Reduction in E-cadherin synergised with an activating mutation of ß-catenin resulting in a rapid CPC phenotype within the SI and colon. Thus, there is a threshold of ß-catenin that is required to drive transformation, and E-cadherin can act as a buffer to sequester mutated ß-catenin.


Asunto(s)
Cadherinas/metabolismo , Transformación Celular Neoplásica , Neoplasias del Colon , Mutación , Proteínas de Neoplasias , Vía de Señalización Wnt , beta Catenina , Animales , Cadherinas/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
3.
J Virol ; 92(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30021891

RESUMEN

The early replication of certain prion strains within Peyer's patches in the small intestine is essential for the efficient spread of disease to the brain after oral exposure. Our data show that orally acquired prions utilize specialized gut epithelial cells known as M cells to enter Peyer's patches. M cells express the cellular isoform of the prion protein, PrPC, and this may be exploited by some pathogens as an uptake receptor to enter Peyer's patches. This suggested that PrPC might also mediate the uptake and transfer of prions across the gut epithelium into Peyer's patches in order to establish infection. Furthermore, the expression level of PrPC in the gut epithelium could influence the uptake of prions from the lumen of the small intestine. To test this hypothesis, transgenic mice were created in which deficiency in PrPC was specifically restricted to epithelial cells throughout the lining of the small intestine. Our data clearly show that efficient prion neuroinvasion after oral exposure occurred independently of PrPC expression in small intestinal epithelial cells. The specific absence of PrPC in the gut epithelium did not influence the early replication of prions in Peyer's patches or disease susceptibility. Acute mucosal inflammation can enhance PrPC expression in the intestine, implying the potential to enhance oral prion disease pathogenesis and susceptibility. However, our data suggest that the magnitude of PrPC expression in the epithelium lining the small intestine is unlikely to be an important factor which influences the risk of oral prion disease susceptibility.IMPORTANCE The accumulation of orally acquired prions within Peyer's patches in the small intestine is essential for the efficient spread of disease to the brain. Little is known of how the prions initially establish infection within Peyer's patches. Some gastrointestinal pathogens utilize molecules, such as the cellular prion protein PrPC, expressed on gut epithelial cells to enter Peyer's patches. Acute mucosal inflammation can enhance PrPC expression in the intestine, implying the potential to enhance oral prion disease susceptibility. We used transgenic mice to determine whether the uptake of prions into Peyer's patches was dependent upon PrPC expression in the gut epithelium. We show that orally acquired prions can establish infection in Peyer's patches independently of PrPC expression in gut epithelial cells. Our data suggest that the magnitude of PrPC expression in the epithelium lining the small intestine is unlikely to be an important factor which influences oral prion disease susceptibility.


Asunto(s)
Encéfalo/metabolismo , Intestino Delgado/metabolismo , Ganglios Linfáticos Agregados/metabolismo , Proteínas PrPC/genética , Enfermedades por Prión/metabolismo , Administración Oral , Animales , Encéfalo/patología , Mapeo Encefálico , Células Dendríticas Foliculares/metabolismo , Células Dendríticas Foliculares/patología , Susceptibilidad a Enfermedades , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Expresión Génica , Intestino Delgado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ganglios Linfáticos Agregados/patología , Proteínas PrPC/metabolismo , Enfermedades por Prión/mortalidad , Enfermedades por Prión/patología , Análisis de Supervivencia
4.
BMC Cancer ; 19(1): 677, 2019 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-31291912

RESUMEN

BACKGROUND: Canonical WNT signalling plays a critical role in the regulation of ovarian development; mis-regulation of this key pathway in the adult ovary is associated with subfertility and tumourigenesis. The roles of Adenomatous polyposis coli 2 (APC2), a little-studied WNT signalling pathway regulator, in ovarian homeostasis, fertility and tumourigenesis have not previously been explored. Here, we demonstrate essential roles of APC2 in regulating ovarian WNT signalling and ovarian homeostasis. METHODS: A detailed analysis of ovarian histology, gene expression, ovulation and hormone levels was carried out in 10 week old and in aged constitutive APC2-knockout (Apc2-/-) mice (mixed background). Statistical significance for qRT-PCR data was determined from 95% confidence intervals. Significance testing was performed using 2-tailed Student's t-test, when 2 experimental cohorts were compared. When more were compared, ANOVA test was used, followed by a post-hoc test (LSD or Games-Howell). P-values of < 0.05 were considered statistically significant. RESULTS: APC2-deficiency resulted in activation of ovarian WNT signalling and sub-fertility driven by intra-ovarian defects. Follicular growth was perturbed, resulting in a reduced rate of ovulation and corpora lutea formation, which could not be rescued by administration of gonadotrophins. Defects in steroidogenesis and follicular vascularity contributed to the subfertility phenotype. Tumour incidence was assessed in aged APC2-deficient mice, which also carried a hypomorphic Apc allele. APC2-deficiency in these mice resulted in predisposition to granulosa cell tumour (GCT) formation, accompanied by acute tumour-associated WNT-signalling activation and a histologic pattern and molecular signature seen in human adult GCTs. CONCLUSIONS: Our work adds APC2 to the growing list of WNT-signalling members that regulate ovarian homeostasis, fertility and suppress GCT formation. Importantly, given that the APC2-deficient mouse develops tumours that recapitulate the molecular signature and histological features of human adult GCTs, this mouse has excellent potential as a pre-clinical model to study ovarian subfertility and transitioning to GCT, tumour biology and for therapeutic testing.


Asunto(s)
Carcinogénesis/metabolismo , Proteínas del Citoesqueleto/metabolismo , Fertilidad , Ovario/metabolismo , Vía de Señalización Wnt , Análisis de Varianza , Animales , Proteínas del Citoesqueleto/genética , Femenino , Proteína Forkhead Box O1/metabolismo , Técnicas de Inactivación de Genes , Tumor de Células de la Granulosa/etiología , Tumor de Células de la Granulosa/metabolismo , Homeostasis , Infertilidad/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Folículo Ovárico/crecimiento & desarrollo , Neoplasias Ováricas/etiología , Neoplasias Ováricas/metabolismo , beta Catenina/metabolismo
5.
J Pathol ; 245(3): 270-282, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29603746

RESUMEN

Epigenetic regulation plays a key role in the link between inflammation and cancer. Here we examine Mbd2, which mediates epigenetic transcriptional silencing by binding to methylated DNA. In separate studies the Mbd2-/- mouse has been shown (1) to be resistant to intestinal tumourigenesis and (2) to have an enhanced inflammatory/immune response, observations that are inconsistent with the links between inflammation and cancer. To clarify its role in tumourigenesis and inflammation, we used constitutive and conditional models of Mbd2 deletion to explore its epithelial and non-epithelial roles in the intestine. Using a conditional model, we found that suppression of intestinal tumourigenesis is due primarily to the absence of Mbd2 within the epithelia. Next, we demonstrated, using the DSS colitis model, that non-epithelial roles of Mbd2 are key in preventing the transition from acute to tumour-promoting chronic inflammation. Combining models revealed that prior to inflammation the altered Mbd2-/- immune response plays a role in intestinal tumour suppression. However, following inflammation the intestine converts from tumour suppressive to tumour promoting. To summarise, in the intestine the normal function of Mbd2 is exploited by cancer cells to enable tumourigenesis, while in the immune system it plays a key role in preventing tumour-enabling inflammation. Which role is dominant depends on the inflammation status of the intestine. As environmental interactions within the intestine can alter DNA methylation patterns, we propose that Mbd2 plays a key role in determining whether these interactions are anti- or pro-tumourigenic and this makes it a useful new epigenetic model for inflammation-associated carcinogenesis. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Colitis/metabolismo , Proteínas de Unión al ADN/metabolismo , Mucosa Intestinal/metabolismo , Neoplasias Intestinales/metabolismo , Animales , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Metilación de ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Sulfato de Dextran , Modelos Animales de Enfermedad , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Genes APC , Mucosa Intestinal/patología , Neoplasias Intestinales/inducido químicamente , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Ratones Noqueados , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Transducción de Señal , Células TH1/metabolismo , Células TH1/patología , Células Th2/metabolismo , Células Th2/patología
6.
Semin Cell Dev Biol ; 52: 21-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26877140

RESUMEN

We describe in this review increasing evidence that loss of LKB1 kinase in Peutz-Jeghers syndrome (PJS) derails the existing natural balance between cell survival and tumour growth suppression. LKB1 deletion can plunge cells into an energy/oxidative stress-induced crisis which leads to the activation of alternative and often carcinogenic pathways to maintain cellular energy levels. It therefore appears that although LKB1 deficiency can suppress oncogenic transformation in the short term, it can ultimately lead to more progressed and malignant phenotypes by driving abnormal cell differentiation, genomic instability and increased tumour heterogeneity.


Asunto(s)
Neoplasias/enzimología , Síndrome de Peutz-Jeghers/enzimología , Proteínas Serina-Treonina Quinasas/deficiencia , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Humanos , Mutación , Neoplasias/genética , Neoplasias/patología , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
7.
J Pathol ; 243(4): 442-456, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29134654

RESUMEN

Aberrant phosphoinositide 3-kinase (PI3K), mitogen-activated protein kinase (MAPK) and WNT signalling are emerging as key events in the multistep nature of prostate tumourigenesis and progression. Here, we report a compound prostate cancer murine model in which these signalling pathways cooperate to produce a more aggressive prostate cancer phenotype. Using Cre-LoxP technology and the probasin promoter, we combined the loss of Pten (Ptenfl/fl ), to activate the PI3K signalling pathway, with either dominant stabilized ß-catenin [Catnb+/lox(ex3) ] or activated K-RAS (K-Ras+/V12 ) to aberrantly activate WNT and MAPK signalling, respectively. Synchronous activation of all three pathways (triple mutants) significantly reduced survival (median 96 days) as compared with double mutants [median: 140 days for Catnb+/lox(ex3) Ptenfl/fl ; 182 days for Catnb+/lox(ex3) K-Ras+/V12 ; 238 days for Ptenfl/fl K-Ras+/V12 ], and single mutants [median: 383 days for Catnb+/lox(ex3) ; 407 days for Ptenfl/fl ], reflecting the accelerated tumourigenesis. Tumours followed a stepwise progression from mouse prostate intraepithelial neoplasia to invasive adenocarcinoma, similar to that seen in human disease. There was significantly elevated cellular proliferation, tumour growth and percentage of invasive adenocarcinoma in triple mutants as compared with double mutants and single mutants. Triple mutants showed not only activated AKT, extracellular-signal regulated kinase 1/2, and nuclear ß-catenin, but also significantly elevated signalling through mechanistic target of rapamycin complex 1 (mTORC1). In summary, we show that combined deregulation of the PI3K, MAPK and WNT signalling pathways drives rapid progression of prostate tumourigenesis, and that deregulation of all three pathways results in tumours showing aberrant mTORC1 signalling. As mTORC1 signalling is emerging as a key driver of androgen deprivation therapy resistance, our findings are important for understanding the biology of therapy-resistant prostate cancer and identifying potential approaches to overcome this. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Adenocarcinoma/enzimología , Transformación Celular Neoplásica/metabolismo , Fosfohidrolasa PTEN/deficiencia , Neoplasia Intraepitelial Prostática/enzimología , Neoplasias de la Próstata/enzimología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , beta Catenina/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Mutación , Fosfohidrolasa PTEN/genética , Fenotipo , Fosfatidilinositol 3-Quinasa/metabolismo , Neoplasia Intraepitelial Prostática/genética , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Factores de Tiempo , Carga Tumoral , Vía de Señalización Wnt , beta Catenina/genética
8.
PLoS Genet ; 10(7): e1004453, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25010414

RESUMEN

Tumourigenesis within the intestine is potently driven by deregulation of the Wnt pathway, a process epigenetically regulated by the chromatin remodelling factor Brg1. We aimed to investigate this interdependency in an in vivo setting and assess the viability of Brg1 as a potential therapeutic target. Using a range of transgenic approaches, we deleted Brg1 in the context of Wnt-activated murine small intestinal epithelium. Pan-epithelial loss of Brg1 using VillinCreERT2 and AhCreERT transgenes attenuated expression of Wnt target genes, including a subset of stem cell-specific genes and suppressed Wnt-driven tumourigenesis improving animal survival. A similar increase in survival was observed when Wnt activation and Brg1 loss were restricted to the Lgr5 expressing intestinal stem cell population. We propose a mechanism whereby Brg1 function is required for aberrant Wnt signalling and ultimately for the maintenance of the tumour initiating cell compartment, such that loss of Brg1 in an Apc-deficient context suppresses adenoma formation. Our results highlight potential therapeutic value of targeting Brg1 and serve as a proof of concept that targeting the cells of origin of cancer may be of therapeutic relevance.


Asunto(s)
Adenoma/genética , Carcinogénesis/genética , ADN Helicasas/biosíntesis , Intestino Delgado/metabolismo , Proteínas Nucleares/biosíntesis , Factores de Transcripción/biosíntesis , Adenoma/patología , Animales , ADN Helicasas/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestino Delgado/patología , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Nucleares/genética , Factores de Transcripción/genética , Vía de Señalización Wnt/genética
9.
Proc Natl Acad Sci U S A ; 111(42): 15078-83, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25288747

RESUMEN

An unresolved issue in genotoxic stress response is identification of induced regulatory proteins and how these activate tumor suppressor p53 to determine appropriate cell responses. Transcription factor KAISO was previously described to repress transcription following binding to methylated DNA. In this study, we show that KAISO is induced by DNA damage in p53-expressing cells and then interacts with the p53-p300 complex to increase acetylation of p53 K320 and K382 residues, although decreasing K381 acetylation. Moreover, the p53 with this particular acetylation pattern shows increased DNA binding and potently induces cell cycle arrest and apoptosis by activating transcription of CDKN1A (cyclin-dependent kinase inhibitor 1) and various apoptotic genes. Analogously, in Kaiso KO mouse embryonic fibroblast cells, p53-to-promoter binding and up-regulation of p21 and apoptosis gene expression is significantly compromised. KAISO may therefore be a critical regulator of p53-mediated cell cycle arrest and apoptosis in response to various genotoxic stresses in mammalian cells.


Asunto(s)
Apoptosis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Animales , Ciclo Celular , Línea Celular , Proliferación Celular , ADN/química , Daño del ADN , Metilación de ADN , Proteína p300 Asociada a E1A/metabolismo , Femenino , Fibroblastos/citología , Células HCT116 , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Unión Proteica , Elementos de Respuesta
10.
BMC Cancer ; 16: 399, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27388476

RESUMEN

BACKGROUND: Increased numbers and improperly positioned centrosomes, aneuploidy or polyploidy, and chromosomal instability are frequently observed characteristics of cancer cells. While some aspects of these events and the checkpoint mechanisms are well studied, not all players have yet been identified. As the role of proteases other than the proteasome in tumorigenesis is an insufficiently addressed question, we investigated the epigenetic control of the widely conserved protease HTRA1 and the phenotypes of deregulation. METHODS: Mouse embryonal fibroblasts and HCT116 and SW480 cells were used to study the mechanism of epigenetic silencing of HTRA1. In addition, using cell biological and genetic methods, the phenotypes of downregulation of HTRA1 expression were investigated. RESULTS: HTRA1 is epigenetically silenced in HCT116 colon carcinoma cells via the epigenetic adaptor protein MBD2. On the cellular level, HTRA1 depletion causes multiple phenotypes including acceleration of cell growth, centrosome amplification and polyploidy in SW480 colon adenocarcinoma cells as well as in primary mouse embryonic fibroblasts (MEFs). CONCLUSIONS: Downregulation of HTRA1 causes a number of phenotypes that are hallmarks of cancer cells suggesting that the methylation state of the HtrA1 promoter may be used as a biomarker for tumour cells or cells at risk of transformation.


Asunto(s)
Neoplasias del Colon/genética , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Fibroblastos/citología , Serina Endopeptidasas/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Centrosoma/metabolismo , Neoplasias del Colon/patología , Regulación hacia Abajo , Epigénesis Genética , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Serina Peptidasa A1 que Requiere Temperaturas Altas , Humanos , Ratones , Trasplante de Neoplasias , Poliploidía , Regiones Promotoras Genéticas
11.
PLoS Genet ; 9(8): e1003638, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935526

RESUMEN

Conditional deletion of Apc in the murine intestine alters crypt-villus architecture and function. This process is accompanied by multiple changes in gene expression, including upregulation of Cited1, whose role in colorectal carcinogenesis is unknown. Here we explore the relevance of Cited1 to intestinal tumorigenesis. We crossed Cited1 null mice with Apc(Min/+) and AhCre(+)Apc(fl/fl) mice and determined the impact of Cited1 deficiency on tumour growth/initiation including tumour multiplicity, cell proliferation, apoptosis and the transcriptome. We show that Cited1 is up-regulated in both human and murine tumours, and that constitutive deficiency of Cited1 increases survival in Apc(Min/+) mice from 230.5 to 515 days. However, paradoxically, Cited1 deficiency accentuated nearly all aspects of the immediate phenotype 4 days after conditional deletion of Apc, including an increase in cell death and enhanced perturbation of differentiation, including of the stem cell compartment. Transcriptome analysis revealed multiple pathway changes, including p53, PI3K and Wnt. The activation of Wnt through Cited1 deficiency correlated with increased transcription of ß-catenin and increased levels of dephosphorylated ß-catenin. Hence, immediately following deletion of Apc, Cited1 normally restrains the Wnt pathway at the level of ß-catenin. Thus deficiency of Cited1 leads to hyper-activation of Wnt signaling and an exaggerated Wnt phenotype including elevated cell death. Cited1 deficiency decreases intestinal tumourigenesis in Apc(Min/+) mice and impacts upon a number of oncogenic signaling pathways, including Wnt. This restraint imposed by Cited1 is consistent with a requirement for Cited1 to constrain Wnt activity to a level commensurate with optimal adenoma formation and maintenance, and provides one mechanism for tumour repression in the absence of Cited1.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Carcinogénesis , Perfilación de la Expresión Génica , Proteínas Nucleares/genética , Factores de Transcripción/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Diferenciación Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Células Madre Neoplásicas , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Transactivadores , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/genética
12.
Am J Pathol ; 184(1): 86-91, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24200851

RESUMEN

Cowden syndrome (CS) is a rare autosomal dominant cancer-prone disorder caused by germ-line mutation of the phosphatase and tensin homolog mutated on chromosome 10 (PTEN) tumor-suppressor gene. Affected patients commonly develop juvenile polyps, and show an elevated risk of developing colorectal cancers. The etiology of these peculiar polyps remains unclear, although previous work has suggested somatic PTEN alterations in the stroma of juvenile polyps. After a long latency period, we find epithelial-specific PTEN deletion to cause formation of juvenile polyps in the colorectum without stromal PTEN loss. More important, we find that these lesions closely recapitulate all of the characteristic histopathological features of juvenile polyps seen in patients with CS, including stromal alterations and dysplastic transformation to colorectal carcinoma. The stromal alterations we identify after epithelial-specific PTEN loss suggest that PTEN may be involved in altered epithelial-mesenchymal cross talk, which, in turn, predisposes to colorectal neoplasia and polyposis. Our transgenic model is the first to recapitulate colorectal juvenile polyposis in patients with CS. We conclude that stromal PTEN loss is not a prerequisite for the formation of juvenile polyps, and that colorectal juvenile polyps in CS are bona fide neoplastic precursor lesions.


Asunto(s)
Neoplasias Colorrectales/genética , Modelos Animales de Enfermedad , Pólipos Intestinales/genética , Fosfohidrolasa PTEN/genética , Lesiones Precancerosas/genética , Animales , Neoplasias Colorrectales/patología , Síndrome de Hamartoma Múltiple/genética , Síndrome de Hamartoma Múltiple/patología , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Poliposis Intestinal/congénito , Poliposis Intestinal/genética , Poliposis Intestinal/patología , Pólipos Intestinales/patología , Ratones , Ratones Transgénicos , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/patología , Lesiones Precancerosas/patología , Células del Estroma/metabolismo , Células del Estroma/patología
13.
BMC Cancer ; 15: 110, 2015 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-25881306

RESUMEN

BACKGROUND: Conditional deletion of the tumour suppressor gene Apc within the murine intestine results in acute Wnt signalling activation. The associated over-expression of a myriad of Wnt signalling target genes yields phenotypic alterations that encompass many of the hallmarks of neoplasia. Previous transcriptomic analysis aimed at identifying genes that potentially play an important role in this process, inferred the Hormonally upregulated Neu-associated kinase (HUNK/Mak-v/Bstk1) gene as a possible candidate. Hunk is a SNF1 (sucrose non fermenting 1)-related serine/threonine kinase with a proposed association with many different tumour types, including colorectal cancer. METHODS: Here we describe the generation of a novel Hunk kinase deficient mouse which has been used to investigate the involvement of Hunk-kinase activity in intestinal homeostasis and tumourigenesis. RESULTS: We show that in the morphologically normal intestine, Hunk-kinase negatively regulates epithelial cell proliferation. However, the increase in cell proliferation observed in the Hunk kinase deficient intestine is counteracted by increased cell migration, thereby maintaining intestinal homeostasis. Using qRT-PCR, we further demonstrate that Hunk is significantly over-expressed in Apc deficient / Wnt-signalling activated intestinal tissue. Using the classical intestinal tumourigenesis Apc (Min) mouse model we show that loss of Hunk-kinase activity significantly reduced tumour initiation rates in the small intestine. However, an accompanying increase in the size of the tumours counteracts the impact this has on overall tumour burden or subsequently survival. CONCLUSIONS: In the intestinal setting we demonstrate that Hunk has a role in normal intestinal proliferation and homeostasis and, although it does not alter overall survival rates, activity of this kinase does impact on tumour initiation rates during the early stages in tumourigenesis in the small intestine.


Asunto(s)
Mucosa Intestinal/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Células Madre Embrionarias/metabolismo , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Marcación de Gen , Sitios Genéticos , Masculino , Ratones , Datos de Secuencia Molecular , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/mortalidad , Neoplasias/patología , Proteínas Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas , Carga Tumoral , Regulación hacia Arriba , Vía de Señalización Wnt
14.
J Pathol ; 233(1): 27-38, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24293351

RESUMEN

Mutation or loss of the genes PTEN and KRAS have been implicated in human colorectal cancer (CRC), and have been shown to co-occur despite both playing a role in the PI3' kinase (PI3'K) pathway. We investigated the role of these genes in intestinal tumour progression in vivo, using genetically engineered mouse models, with the aim of generating more representative models of human CRC. Intestinal-specific deletion of Pten and activation of an oncogenic allele of Kras was induced in wild-type (WT) mice and mice with a predisposition to adenoma development (Apc(fl/+) ). The animals were euthanized when they became symptomatic of a high tumour burden. Histopathological examination of the tissues was carried out, and immunohistochemistry used to characterize signalling pathway activation. Mutation of Pten and Kras resulted in a significant life-span reduction of mice predisposed to adenomas. Invasive adenocarcinoma was observed in these animals, with evidence of activation of the PI3'K pathway but no metastasis. However, mutation of Pten and Kras in WT animals not predisposed to adenomas led to perturbed homeostasis of the intestinal epithelium and the development of hyperplastic polyps, dysplastic sessile serrated adenomas and metastasizing adenocarcinomas with serrated features. These studies demonstrate synergism between Pten and Kras mutations in intestinal tumour progression, in an autochthonous and immunocompetent murine model, with potential application to preclinical drug testing. In particular, they show that Pten and Kras mutations alone predispose mice to the spectrum of serrated lesions that reflect the serrated pathway of CRC progression in humans.


Asunto(s)
Adenocarcinoma/metabolismo , Adenoma/metabolismo , Neoplasias Intestinales/metabolismo , Pólipos Intestinales/metabolismo , Intestino Delgado/metabolismo , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/secundario , Adenoma/genética , Adenoma/patología , Animales , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Femenino , Genes APC , Predisposición Genética a la Enfermedad , Hiperplasia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Pólipos Intestinales/genética , Pólipos Intestinales/patología , Intestino Delgado/patología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Invasividad Neoplásica , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Fenotipo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal , Factores de Tiempo , Carga Tumoral
15.
Nature ; 457(7229): 608-11, 2009 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-19092804

RESUMEN

Intestinal cancer is initiated by Wnt-pathway-activating mutations in genes such as adenomatous polyposis coli (APC). As in most cancers, the cell of origin has remained elusive. In a previously established Lgr5 (leucine-rich-repeat containing G-protein-coupled receptor 5) knockin mouse model, a tamoxifen-inducible Cre recombinase is expressed in long-lived intestinal stem cells. Here we show that deletion of Apc in these stem cells leads to their transformation within days. Transformed stem cells remain located at crypt bottoms, while fuelling a growing microadenoma. These microadenomas show unimpeded growth and develop into macroscopic adenomas within 3-5weeks. The distribution of Lgr5(+) cells within stem-cell-derived adenomas indicates that a stem cell/progenitor cell hierarchy is maintained in early neoplastic lesions. When Apc is deleted in short-lived transit-amplifying cells using a different cre mouse, the growth of the induced microadenomas rapidly stalls. Even after 30weeks, large adenomas are very rare in these mice. We conclude that stem-cell-specific loss of Apc results in progressively growing neoplasia.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Proteína de la Poliposis Adenomatosa del Colon/genética , Linaje de la Célula , Transformación Celular Neoplásica , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Células Madre Neoplásicas/patología , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Genes APC , Neoplasias Intestinales/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Receptores Acoplados a Proteínas G/análisis , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , beta Catenina/metabolismo
16.
BMC Bioinformatics ; 15: 15, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24428888

RESUMEN

BACKGROUND: Given the estimate that 30% of our genes are controlled by microRNAs, it is essential that we understand the precise relationship between microRNAs and their targets. OncomiRs are microRNAs (miRNAs) that have been frequently shown to be deregulated in cancer. However, although several oncomiRs have been identified and characterized, there is as yet no comprehensive compilation of this data which has rendered it underutilized by cancer biologists. There is therefore an unmet need in generating bioinformatic platforms to speed the identification of novel therapeutic targets. DESCRIPTION: We describe here OncomiRdbB, a comprehensive database of oncomiRs mined from different existing databases for mouse and humans along with novel oncomiRs that we have validated in human breast cancer samples. The database also lists their respective predicted targets, identified using miRanda, along with their IDs, sequences, chromosome location and detailed description. This database facilitates querying by search strings including microRNA name, sequence, accession number, target genes and organisms. The microRNA networks and their hubs with respective targets at 3'UTR, 5'UTR and exons of different pathway genes were also deciphered using the 'R' algorithm. CONCLUSION: OncomiRdbB is a comprehensive and integrated database of oncomiRs and their targets in breast cancer with multiple query options which will help enhance both understanding of the biology of breast cancer and the development of new and innovative microRNA based diagnostic tools and targets of therapeutic significance. OncomiRdbB is freely available for download through the URL link http://tdb.ccmb.res.in/OncomiRdbB/index.htm.


Asunto(s)
Neoplasias de la Mama/genética , Biología Computacional/métodos , Bases de Datos de Ácidos Nucleicos , MicroARNs/genética , Regiones no Traducidas 3' , Algoritmos , Animales , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Ratones , MicroARNs/metabolismo
17.
Stem Cells ; 31(4): 776-85, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23335179

RESUMEN

The identification of the intestinal stem cell (ISC) markers Lgr5 and Bmi-1 has furthered our understanding of how they accomplish homeostasis in this rapidly self-renewing tissue. Recent work indicates that these markers identify a cycling Lgr5(+) ISC which can be replaced by a quiescent Bmi-1(+) ISC. Currently, there is little data on how these cells interact to control intestinal crypt homeostasis and regeneration. This interaction likely involves other differentiated cells within the niche as it has previously been demonstrated that the "stemness" of the Lgr5 ISC is closely tied to the presence of their neighboring Paneth cells. To investigate this, we used two conditional mouse models to delete the transcription factor ß-catenin within the intestinal crypt. Critically these differ in their ability to drive recombination within Paneth cells and therefore allow us to compare the effect of deleting the majority of active ISCs in the presence or absence of the Paneth cells. After gene deletion, the intestines in the model in which Paneth cells were retained showed a rapid recovery and repopulation of the crypt-villus axis presumably from either a spared ISC or the hypothetical quiescent ISCs. However, in the absence of Paneth cells the recovery ability was compromised resulting in complete loss of intestinal epithelial integrity. This data indicates that the Paneth cells play a crucial role within the in vivo ISC niche in aiding recovery following substantial insult.


Asunto(s)
Intestinos/citología , Intestinos/lesiones , Células de Paneth/citología , Células de Paneth/metabolismo , Alelos , Animales , Inmunohistoquímica , Ratones , Ratones Transgénicos , Células Madre/citología , Células Madre/metabolismo , beta Catenina/deficiencia , beta Catenina/genética , beta Catenina/metabolismo
18.
Stem Cells ; 31(11): 2457-66, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23922304

RESUMEN

Brg1 is a chromatin remodeling factor involved in mediation of a plethora of signaling pathways leading to its participation in various physiological processes both during development and in adult tissues. Among other signaling pathways, the Wnt pathway has been proposed to require Brg1 for transactivation of its target genes. Given the pivotal role of the Wnt pathway in the maintenance of normal intestinal homeostasis, we aimed to investigate the effects of Brg1 loss on the intestinal physiology. To this end, we deleted Brg1 in the murine small and large intestinal epithelia using a range of transgenic approaches. Pan-epithelial loss of Brg1 in the small intestine resulted in crypt ablation, while partial Brg1 deficiency led to gradual repopulation of the intestinal mucosa with wild-type cells. In contrast, Brg1 loss in the large intestinal epithelium was compensated by upregulation of Brm. We propose that while Brg1 is dispensable for the survival and function of the progenitor and differentiated cells in the murine intestinal epithelium, it is essential for the maintenance of the stem cell population in a tissue-specific manner.


Asunto(s)
ADN Helicasas/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Nucleares/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , ADN Helicasas/genética , Mucosa Intestinal/citología , Ratones , Análisis por Micromatrices , Proteínas Nucleares/genética , Transducción de Señal , Células Madre/citología , Factores de Transcripción/genética
19.
J Pathol ; 230(2): 165-73, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23483557

RESUMEN

Carcinomas of the biliary tract are aggressive malignancies in humans. Loss of the tumour suppressor PTEN has previously been associated with cholangiocarcinoma development in a murine model. Activation of KRAS is reported in up to one-third of human cholangiocarcinomas and 50% of gall bladder carcinomas. In this study we aimed to test the potential interaction between PTEN and KRAS mutation in biliary tract malignancy. We used an inducible Cre-LoxP-based approach to coordinately delete PTEN and activate KRAS within the adult mouse biliary epithelium. We found that activation of KRAS alone has little effect upon biliary epithelium. Loss of PTEN alone results in the development of low-grade neoplastic lesions, following long latency and at low incidence. Combination of both mutations causes rapid development of biliary epithelial proliferative lesions, which progress through dysplasia to invasive carcinoma. We conclude that activation of the PI3'K pathway following loss of PTEN is sufficient to drive slow development of low-grade biliary lesions in mice. In contrast, mutational activation of KRAS does not result in a similar phenotype, despite a prediction that this should activate both the RAF-MEK-ERK and PI3'-kinase pathways. However, mutation of both genes results in rapid tumourigenesis, arguing that PTEN normally functions as a 'brake' on the PI3'-kinase pathway, limiting the influence of KRAS activation. Mutation of both genes creates a 'permissive' environment, allowing the full effects of both mutations to be manifested. These data reveal an in vivo synergy between these mutations and provides a new mouse model of biliary tract malignancy.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Colangiocarcinoma/genética , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Animales no Consanguíneos , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/mortalidad , Sistema Biliar/citología , Sistema Biliar/metabolismo , Colangiocarcinoma/metabolismo , Colangiocarcinoma/mortalidad , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Femenino , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Estimación de Kaplan-Meier , Masculino , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Mutación , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/biosíntesis , Tasa de Supervivencia
20.
Gastroenterology ; 143(6): 1650-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22960659

RESUMEN

BACKGROUND & AIMS: Mutations in components of the Wnt signaling pathway, including ß-catenin and AXIN1, are found in more than 50% of human hepatocellular carcinomas (HCCs). Disruption of Axin1 causes embryonic lethality in mice. We generated mice with conditional disruption of Axin1 to study its function specifically in adult liver. METHODS: Mice with a LoxP-flanked allele of Axin1 were generated by homologous recombination. Mice homozygous for the Axin1fl/fl allele were crossed with AhCre mice; in offspring, Axin1 was disrupted in liver following injection of ß-naphthoflavone (Axin1fl/fl/Cre mice). Liver tissues were collected and analyzed by quantitative real-time polymerase chain reaction and immunoprecipitation, histology, and immunoblot assays. RESULTS: Deletion of Axin1 from livers of adult mice resulted in an acute and persistent increase in hepatocyte cell volume, proliferation, and transcription of genes that induce the G(2)/M transition in the cell cycle and cytokinesis. A subset of Wnt target genes was activated, including Axin2, c-Myc, and cyclin D1. However, loss of Axin1 did not increase nuclear levels of ß-catenin or cause changes in liver zonation that have been associated with loss of the adenomatous polyposis coli (APC) or constitutive activation of ß-catenin. After 1 year, 5 of 9 Axin1fl/fl/Cre mice developed liver tumors with histologic features of HCC. CONCLUSIONS: Hepatocytes from adult mice with conditional disruption of Axin1 in liver have a transcriptional profile that differs from that associated with loss of APC or constitutive activation of ß-catenin. It might be similar to a proliferation profile observed in a subset of human HCCs with mutations in AXIN1. Axin1fl/fl mice could be a useful model of AXIN1-associated tumorigenesis and HCC.


Asunto(s)
Proteína Axina/genética , Proteína Axina/fisiología , Carcinoma Hepatocelular/fisiopatología , Eliminación de Gen , Neoplasias Hepáticas/fisiopatología , Alelos , Animales , Carcinoma Hepatocelular/patología , Ciclo Celular/fisiología , Proliferación Celular , Modelos Animales de Enfermedad , Hepatocitos/patología , Neoplasias Hepáticas/patología , Ratones , Ratones Mutantes , Proteínas Wnt/fisiología , beta Catenina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA