Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38916963

RESUMEN

Despite epidermal turnover, the skin is host to a complex array of microbes including viruses, such as the human papillomavirus (HPV), which must infect and manipulate skin keratinocyte stem cells (KSC) to survive. This crosstalk between the virome and KSC populations remains largely unknown. Here, we investigated the effect of HPV8 on KSCs using various mouse models. We observed that the HPV8 early region gene E6 specifically caused Lrig1+ hair follicle junctional zone KSC proliferation and expansion, which would facilitate viral transmission. Within Lrig1+ KSCs specifically, HPV8 E6 bound intracellular p300 to phosphorylate the STAT3 transcriptional regulatory node. This induces ΔNp63 expression, resulting in KSC expansion into the overlying epidermis. HPV8 was associated with 70% of human actinic keratoses (AK). Together these results define the "hit and run" mechanism for HPV8 in human actinic keratosis as an expansion of KSCs, which lacks melanosome protection and is thus susceptible to sun-light-induced malignant transformation.

2.
J Clin Invest ; 132(21)2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36074574

RESUMEN

The basis of immune evasion, a hallmark of cancer, can differ even when cancers arise from one cell type such as in the human skin keratinocyte carcinomas: basal and squamous cell carcinoma. Here we showed that the basal cell carcinoma tumor-initiating cell surface protein CD200, through ectodomain shedding, was responsible for the near absence of NK cells within the basal cell carcinoma tumor microenvironment. In situ, CD200 underwent ectodomain shedding by metalloproteinases MMP3 and MMP11, which released biologically active soluble CD200 into the basal cell carcinoma microenvironment. CD200 bound its cognate receptor on NK cells to suppress MAPK pathway signaling that in turn blocked indirect (IFN-γ release) and direct cell killing. In addition, reduced ERK phosphorylation relinquished negative regulation of PPARγ-regulated gene transcription and led to membrane accumulation of the Fas/FADD death receptor and its ligand, FasL, which resulted in activation-induced apoptosis. Blocking CD200 inhibition of MAPK or PPARγ signaling restored NK cell survival and tumor cell killing, with relevance to many cancer types. Our results thus uncover a paradigm for CD200 as a potentially novel and targetable NK cell-specific immune checkpoint, which is responsible for NK cell-associated poor outcomes in many cancers.


Asunto(s)
Carcinoma Basocelular , Carcinoma de Células Escamosas , Humanos , Microambiente Tumoral , PPAR gamma , Células Asesinas Naturales , Receptor fas , Apoptosis , Carcinoma de Células Escamosas/genética
3.
Int J Mol Sci ; 21(12)2020 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-32630372

RESUMEN

Oncogenic activation of the phosphatidylinositol-3-kinase (PI3K), protein kinase B (PKB/AKT), and mammalian target of rapamycin (mTOR) pathway is a frequent event in prostate cancer that facilitates tumor formation, disease progression and therapeutic resistance. Recent discoveries indicate that the complex crosstalk between the PI3K-AKT-mTOR pathway and multiple interacting cell signaling cascades can further promote prostate cancer progression and influence the sensitivity of prostate cancer cells to PI3K-AKT-mTOR-targeted therapies being explored in the clinic, as well as standard treatment approaches such as androgen-deprivation therapy (ADT). However, the full extent of the PI3K-AKT-mTOR signaling network during prostate tumorigenesis, invasive progression and disease recurrence remains to be determined. In this review, we outline the emerging diversity of the genetic alterations that lead to activated PI3K-AKT-mTOR signaling in prostate cancer, and discuss new mechanistic insights into the interplay between the PI3K-AKT-mTOR pathway and several key interacting oncogenic signaling cascades that can cooperate to facilitate prostate cancer growth and drug-resistance, specifically the androgen receptor (AR), mitogen-activated protein kinase (MAPK), and WNT signaling cascades. Ultimately, deepening our understanding of the broader PI3K-AKT-mTOR signaling network is crucial to aid patient stratification for PI3K-AKT-mTOR pathway-directed therapies, and to discover new therapeutic approaches for prostate cancer that improve patient outcome.


Asunto(s)
Fosfatidilinositol 3-Quinasa/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Antagonistas de Andrógenos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Humanos , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Recurrencia Local de Neoplasia/genética , Oncogenes , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/metabolismo , Neoplasias de la Próstata/genética , Receptores Androgénicos/metabolismo , Vía de Señalización Wnt/fisiología
4.
PLoS One ; 14(9): e0223041, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31536587

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0004264.].

5.
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
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.
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
8.
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
9.
Gut ; 61(2): 202-13, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21813469

RESUMEN

BACKGROUND AND AIMS: LKB1 is a serine-threonine kinase, mutation of which can lead to the development of multiple benign intestinal hamartomas (Peutz-Jeghers syndrome). In this study, the authors investigate the mechanisms underlying this phenotype by exploring the transcriptional changes associated with Lkb1 deletion in intestinal epithelium. METHODS: The authors used mice with Lkb1 deleted in the intestinal epithelium using a Cyp1a1-specific inducible Cre recombinase and used Affymetrix (Santa Clara, California, USA) microarray analysis to examine the transcriptional changes occurring immediately after Lkb1 loss. The authors also generated crypt-villus organoid culture to analyse Lkb1 role in intestinal responses to exogenous stimuli. RESULTS: Affymetrix analysis identified the most significant change to be in Ren1 expression, a gene encoding a protease involved in angiotensinogen processing. Lkb1 deletion also enhanced ACE expression and subsequently angiotensin II (AngII) production in the mouse intestine. Intestinal apoptosis induced by Lkb1 deficiency was suppressed by ACE inhibitor captopril. Lkb1-deficient intestinal epithelium showed dynamic changes in AngII receptor type 1, suggesting a possible compensatory response to elevated AngII levels. A similar reduction in epithelial AngII receptor type 1 was also observed in human Peutz-Jeghers syndrome tumours contrasting with high expression of the receptor in the tumour stroma. Mechanistically, the authors showed two pieces of data that position Lkb1 in renin expression regulation, and they implied the importance of Lkb1 in linking cell responses with nutrient levels. First, the authors showed that Lkb1 deletion in isolated epithelial organoid culture resulted in renin upregulation only when the organoids were challenged with external cues such as AngII; second, that renin upregulation was dependent upon the MEK/ERK pathway in a circadian fashion and corresponded to active feeding time when nutrient levels were high. CONCLUSIONS: Taken together, these data reveal a novel role for Lkb1 in regulation of the gastrointestinal renin-angiotensin system.


Asunto(s)
Angiotensina II/metabolismo , Eliminación de Gen , Mucosa Intestinal/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Sistema Renina-Angiotensina/fisiología , Renina/metabolismo , Proteínas Quinasas Activadas por AMP , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratones , Mucina 2/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Renina/genética , Sistema Renina-Angiotensina/genética
10.
PLoS One ; 6(1): e16209, 2011 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-21283818

RESUMEN

The AKT/PI3K/mTOR pathway is frequently altered in a range of human tumours, including bladder cancer. Here we report the phenotype of mice characterised by deletion of two key players in mTOR regulation, Pten and Lkb1, in a range of tissues including the mouse urothelium. Despite widespread recombination within the range of epithelial tissues, the primary phenotype we observe is the rapid onset of bladder tumorigenesis, with median onset of approximately 100 days. Single deletion of either Pten or Lkb1 had no effect on bladder cell proliferation or tumour formation. However, simultaneous deletion of Lkb1 and Pten led to an upregulation of the mTOR pathway and the hypoxia marker GLUT1, increased bladder epithelial cell proliferation and ultimately tumorigenesis. Bladder tissue also exhibited characteristic features of epithelial-mesenchymal transition, with loss of the epithelial markers E-cadherin and the tight junction protein ZO-1, and increases in the mesenchymal marker vimentin as well as nuclear localization of epithelial-mesenchymal transition (EMT) regulator Snail. We show that these effects were all dependent upon mTOR activity, as rapamycin treatment blocked both EMT and tumorigenesis. Our data therefore establish clear synergy between Lkb1 and Pten in controlling the mTOR pathway within bladder epithelium, and show that loss of this control leads to the disturbance of epithelial structure, EMT and ultimately tumorigenesis.


Asunto(s)
Transición Epitelial-Mesenquimal , Fosfohidrolasa PTEN/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Serina-Treonina Quinasas TOR/fisiología , Neoplasias de la Vejiga Urinaria/etiología , Proteínas Quinasas Activadas por AMP , Animales , Proliferación Celular , Eliminación de Gen , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/genética , Proteínas Serina-Treonina Quinasas/genética , Vejiga Urinaria/citología , Vejiga Urinaria/metabolismo
11.
FEBS Lett ; 585(7): 958-66, 2011 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-21241696

RESUMEN

Recent developments have placed the serine/threonine kinase LKB1 on the crossroads linking energy metabolism, cell structure and cancer progression and that its deletion can affect tumorigenesis, metastasis, cell adhesion and polarity. LKB1 can regulate a host of different functions which all have potential to impact upon the initiation and progression of neoplastic disease. To understand the phenotypic consequences of LKB1 loss in a range of different settings, a number of animal models of loss of function have been generated and analyzed. In this review we summarize recent data generated from a range of these models, which reveal clear tissue specific differences in LKB1 function in vivo and in the consequences of its loss.


Asunto(s)
Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Fenómenos Fisiológicos Celulares/genética , Modelos Animales de Enfermedad , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/fisiopatología , Proteínas Serina-Treonina Quinasas/deficiencia
12.
PLoS One ; 4(1): e4264, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19165340

RESUMEN

The Lkb1 tumour suppressor is a multitasking kinase participating in a range of physiological processes. We have determined the impact of Lkb1 deficiency on intestinal homeostasis, particularly focussing on secretory cell differentiation and development since we observe strong expression of Lkb1 in normal small intestine Paneth and goblet cells. We crossed mice bearing an Lkb1 allele flanked with LoxP sites with those carrying a Cyp1a1-specific inducible Cre recombinase. Lkb1 was efficiently deleted from the epithelial cells of the mouse intestine after intraperitoneal injection of the inducing agent beta-naphthoflavone. Bi-allelic loss of Lkb1 led to the perturbed development of Paneth and goblet cell lineages. These changes were characterised by the lack of Delta ligand expression in Lkb1-deficient secretory cells and a significant increase in the levels of the downstream Notch signalling effector Hes5 but not Hes1. Our data show that Lkb1 is required for the normal differentiation of secretory cell lineages within the intestine, and that Lkb1 deficiency modulates Notch signalling modulation in post-mitotic cells.


Asunto(s)
Regulación de la Expresión Génica , Células Caliciformes/citología , Intestino Delgado/citología , Intestino Delgado/patología , Células de Paneth/citología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Receptores Notch/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Alelos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Citocromo P-450 CYP1A1/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Proteínas Represoras/metabolismo , Transducción de Señal , Factor de Transcripción HES-1 , beta-naftoflavona/farmacología
13.
Dev Genes Evol ; 213(10): 514-8, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-13680226

RESUMEN

CLIC proteins are components or regulators of novel intracellular anion channels in mammalian cells, and previous studies have suggested that human nuclear membrane-associated CLIC1 and mouse inner mitochondrial membrane CLIC4 are involved in cell division and apoptosis. We have isolated Xenopus homologues of CLIC1 and CLIC4 and shown them to be well conserved during chordate evolution, but poorly conserved in invertebrates. Consistent with fundamental cellular roles, Xenopus CLIC genes are expressed at every stage of embryonic development. Expression is localised to mesodermal and ectodermal tissues, with particularly marked expression of xCLIC4 in the developing nervous system. This is the first description of non-mammalian CLIC expression, and use of Xenopus laevis as a model organism may provide insights into the role of CLIC-associated ion channels in animal development.


Asunto(s)
Canales de Cloruro/genética , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Xenopus laevis/embriología , Xenopus laevis/genética , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Secuencia de Bases , Cartilla de ADN , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...