Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
2.
EMBO Rep ; 21(7): e50861, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32496657

RESUMEN

Hamlet's question is the artist's expression of the meaning of life. The second law of thermodynamics is the physicist's equivalent.


Asunto(s)
Termodinámica
3.
EMBO J ; 32(16): 2248-63, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23860128

RESUMEN

Squamous cell carcinomas (SCCs) are highly heterogeneous tumours, resulting from deranged expression of genes involved in squamous cell differentiation. Here we report that microRNA-34a (miR-34a) functions as a novel node in the squamous cell differentiation network, with SIRT6 as a critical target. miR-34a expression increases with keratinocyte differentiation, while it is suppressed in skin and oral SCCs, SCC cell lines, and aberrantly differentiating primary human keratinocytes (HKCs). Expression of this miRNA is restored in SCC cells, in parallel with differentiation, by reversion of genomic DNA methylation or wild-type p53 expression. In normal HKCs, the pro-differentiation effects of increased p53 activity or UVB exposure are miR-34a-dependent, and increased miR-34a levels are sufficient to induce differentiation of these cells both in vitro and in vivo. SIRT6, a sirtuin family member not previously connected with miR-34a function, is a direct target of this miRNA in HKCs, and SIRT6 down-modulation is sufficient to reproduce the miR-34a pro-differentiation effects. The findings are of likely biological significance, as SIRT6 is oppositely expressed to miR-34a in normal keratinocytes and keratinocyte-derived tumours.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Diferenciación Celular/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Queratinocitos/fisiología , MicroARNs/metabolismo , Sirtuinas/metabolismo , Diferenciación Celular/efectos de la radiación , Cartilla de ADN/genética , Humanos , Queratinocitos/metabolismo , Análisis por Micromatrices , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Notch1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Rayos Ultravioleta
4.
EMBO Rep ; 21(10): e51505, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32924280

Asunto(s)
Autoria , Termodinámica
5.
Dermatol Surg ; 39(7): 1033-43, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23489371

RESUMEN

BACKGROUND: The use of carbon dioxide (CO2) laser-mediated ablative fractional resurfacing (AFR) is currently under extensive clinical investigation, but the molecular mechanisms underlying this process are unclear. OBJECTIVES: To determine the early expressed genes that are upregulated in human skin after treatment using a CO2 fractional laser. METHODS: Whole human skin was irradiated using an AFR CO2 laser, and changes in gene expression after 2 and 24 hours were analyzed using microarray analysis. The results were validated using reverse transcriptase polymerase chain reaction (RT-PCR). Laser scanning confocal microscopy (LSCM) was used to investigate the expression of the validated proteins after AFR CO2 laser treatment of skin that had been biopsied from seven Korean patients. RESULTS: Gene expression profiling showed that the most significantly upregulated genes in these skin samples were those encoding Wnt5a, cysteine-rich angiogenic inducer 61 (CYR61), and heat shock protein (HSP) 90. These results were confirmed using real-time RT-PCR and LSCM. CONCLUSIONS: Irradiation using an AFR laser may induce the expression of Wnt5a, CYR61, and HSP90 in human skin during the early remodeling phases, suggesting that the induction of proteins may be the preceding event that is associated with the clinical effects of laser treatment.


Asunto(s)
Terapia por Láser/métodos , Piel/efectos de la radiación , Cicatrización de Heridas/fisiología , Dióxido de Carbono , Colágeno/metabolismo , Proteína 61 Rica en Cisteína/metabolismo , Dermabrasión , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Metaloproteinasas de la Matriz/metabolismo , Microscopía Confocal , Proteínas Proto-Oncogénicas/metabolismo , Análisis de Matrices Tisulares , Regulación hacia Arriba/fisiología , Proteínas Wnt/metabolismo , Proteína Wnt-5a
6.
Cancer Cell ; 41(4): 637-640, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-37037612

RESUMEN

The multi-step process of carcinogenesis implies the existence of pre-malignant yet altered states that involve both the potentially carcinogenic cell as well as its surrounding microenvironment. Experts discuss some tumor types for which clear pre-cancerous stages have been identified and mention key biological alterations used for diagnosis and intervention strategies.


Asunto(s)
Neoplasias , Humanos , Neoplasias/diagnóstico , Neoplasias/terapia , Neoplasias/metabolismo , Carcinogénesis , Microambiente Tumoral
7.
Nat Commun ; 14(1): 6498, 2023 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-37838724

RESUMEN

Melanoma provides a primary benchmark for targeted drug therapy. Most melanomas with BRAFV600 mutations regress in response to BRAF/MEK inhibitors (BRAFi/MEKi). However, nearly all relapse within the first two years, and there is a connection between BRAFi/MEKi-resistance and poor response to immune checkpoint therapy. We reported that androgen receptor (AR) activity is required for melanoma cell proliferation and tumorigenesis. We show here that AR expression is markedly increased in BRAFi-resistant melanoma cells, and in sensitive cells soon after BRAFi exposure. Increased AR expression is sufficient to render melanoma cells BRAFi-resistant, eliciting transcriptional changes of BRAFi-resistant subpopulations, including elevated EGFR and SERPINE1 expression, of likely clinical significance. Inhibition of AR expression or activity blunts changes in gene expression and suppresses proliferation and tumorigenesis of BRAFi-resistant melanoma cells, promoting clusters of CD8+ T cells infiltration and cancer cells killing. Our findings point to targeting AR as possible co-therapeutical approach in melanoma treatment.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas B-raf , Humanos , Proteínas Proto-Oncogénicas B-raf/metabolismo , Linfocitos T CD8-positivos/metabolismo , Receptores Androgénicos/genética , Resistencia a Antineoplásicos/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos , Carcinogénesis , Inhibidores de Proteínas Quinasas/farmacología , Línea Celular Tumoral
8.
J Invest Dermatol ; 141(1): 152-163, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32619504

RESUMEN

Phenformin is a drug in the biguanide class that was previously used to treat type 2 diabetes. We have reported the antitumor activities of phenformin to enhance the efficacy of BRAF-MAPK kinase-extracellular signal-regulated kinase pathway inhibition and to inhibit myeloid-derived suppressor cells in various melanoma models. Here we demonstrate that phenformin suppresses tumor growth and promotes keratinocyte differentiation in the 7,12-dimethylbenz[a]anthracene/12-O-tetradecanoylphorbol-13-acetate two-stage skin carcinogenesis mouse model. Moreover, phenformin enhances the suspension-induced differentiation of mouse and human keratinocytes. Mechanistically, phenformin induces the nuclear translocation of NFATc1 in keratinocytes in an AMPK-dependent manner. Pharmacologic or genetic inhibition of calcineurin and NFAT signaling reverses the effects of phenformin on keratinocyte differentiation. Taken together, our study reveals an antitumor activity of phenformin to promote keratinocyte differentiation that warrants future translational efforts to repurpose phenformin for the treatment of cutaneous squamous cell carcinomas.


Asunto(s)
Calcineurina/metabolismo , Queratinocitos/patología , Melanoma/tratamiento farmacológico , Nitrofuranos/metabolismo , Fenformina/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Piel/patología , Animales , Diferenciación Celular , Humanos , Hipoglucemiantes/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Melanoma/metabolismo , Melanoma/patología , Ratones , Neoplasias Experimentales , Transducción de Señal , Piel/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
9.
EMBO Mol Med ; 13(7): e14133, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34185380

RESUMEN

Human populations of Black African ancestry have a relatively high risk of aggressive cancer types, including keratinocyte-derived squamous cell carcinomas (SCCs). We show that primary keratinocytes (HKCs) from Black African (Black) versus White Caucasian (White) individuals have on average higher oncogenic and self-renewal potential, which are inversely related to mitochondrial electron transfer chain activity and ATP and ROS production. HSD17B7 is the top-ranked differentially expressed gene in HKCs and Head/Neck SCCs from individuals of Black African versus Caucasian ancestries, with several ancestry-specific eQTLs linked to its expression. Mirroring the differences between Black and White HKCs, modulation of the gene, coding for an enzyme involved in sex steroid and cholesterol biosynthesis, determines HKC and SCC cell proliferation and oncogenicity as well as mitochondrial OXPHOS activity. Overall, the findings point to a targetable determinant of cancer susceptibility among different human populations, amenable to prevention and management of the disease.


Asunto(s)
Carcinoma de Células Escamosas , Proliferación Celular , Humanos , Queratinocitos , Oncogenes
10.
Heliyon ; 6(12): e05574, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33319088

RESUMEN

Mitogen-Activated Protein Kinases (MAPKs) control a wide array of cellular functions by transducing extracellular information into defined biological responses. In order to understand how these pathways are regulated, dynamic single cell measurements are highly needed. Fluorescence microscopy is well suited to perform these measurements. However, more dynamic and sensitive biosensors that allow the quantification of signaling activity in living mammalian cells are required. We have engineered a synthetic fluorescent substrate for human MAPKs (ERK, JNK and p38) that relocates from the nucleus to the cytoplasm when phosphorylated by the kinases. We demonstrate that this reporter displays an improved response compared to other relocation biosensors. This assay allows to monitor the heterogeneity in the MAPK response in a population of isogenic cells, revealing pulses of ERK activity upon a physiological EGFR stimulation. We show applicability of this approach to the analysis of multiple cancer cell lines and primary cells as well as its application in vivo to developing tumors. Using this ERK biosensor, dynamic single cell measurements with high temporal resolution can be obtained. These MAPK reporters can be widely applied to the analysis of molecular mechanisms of MAPK signaling in healthy and diseased state, in cell culture assays or in vivo.

11.
Clin Cancer Res ; 25(15): 4603-4610, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30890551

RESUMEN

The impact of sex hormones on anticancer immunity deserves attention due to the importance of the immune system in cancer therapy and the recognition of sex differences in immunity. Cancer is ultimately the result of failed immune surveillance, and the diverging effects of male and female sex hormones on anticancer immunity could contribute to the higher cancer incidence and poorer outcome in men. Estrogens and androgens affect the number and function of immune cells, an effect that depends on cell type, tumor microenvironment, and the age and reproductive status of the individual. Despite the recent progress in immuno-oncology, our current understanding of the interplay between sex hormones and anticancer immune responses is in its infancy. In this review, we will focus on the impact of sex hormones on anticancer immunity and immunotherapy. We will discuss the potential role of the changing hormone levels in anticancer immunity during aging and in the context of menopausal hormone therapies and oral contraception. We will review emerging data on sex differences in PD-L1 expression and potential biomarkers predictive for the efficacy of immune checkpoint inhibitors such as the microbiome and consider ongoing clinical trials evaluating the potential impact of hormone deprivation therapies to increase response to immune checkpoint inhibitors in breast and prostate cancer. Finally, we will point to areas of future research.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Neoplasias de la Mama/inmunología , Hormonas Esteroides Gonadales/inmunología , Inmunidad Celular , Neoplasias de la Próstata/inmunología , Microambiente Tumoral/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Femenino , Humanos , Inmunoterapia/métodos , Masculino , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/terapia
12.
Trends Cancer ; 3(3): 181-197, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28718431

RESUMEN

Epidemiological studies point to race as a determining factor in cancer susceptibility. In US registries recording cancer incidence and survival by race (distinguishing 'black versus white'), individuals of African ancestry have a globally increased risk of malignancies compared with Caucasians and Asian Americans. Differences in socioeconomic status and health-care access play a key role. However, the lesser disease susceptibility of Hispanic populations with comparable lifestyles and socioeconomic status as African Americans (Hispanic paradox) points to the concomitant importance of genetic determinants. Here, we overview the molecular basis of racial disparity in cancer susceptibility ranging from genetic polymorphisms and cancer-driver gene mutations to obesity, chronic inflammation, and immune responses. We discuss implications for race-adapted cancer screening programs and clinical trials to reduce disparities in cancer burden.


Asunto(s)
Predisposición Genética a la Enfermedad , Neoplasias/epidemiología , Neoplasias/genética , Grupos Raciales/genética , Asiático/genética , Disparidades en Atención de Salud , Hispánicos o Latinos/genética , Humanos , Neoplasias/patología , Clase Social , Población Blanca/genética
13.
PLoS One ; 12(4): e0173000, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28410392

RESUMEN

Squamous cell carcinoma of the skin (SCC) represents one of the most common cancers in the general population and is associated with a substantial risk of metastasis. Previous work uncovered the functional role of CYFIP1 in epithelial tumors as an invasion inhibitor. It was down-regulated in some cancers and correlated with the metastatic properties of these malignant cells. We investigated its role and expression mechanisms in SCC. We analyzed the expression of CYFIP1 in patient derived SCC, primary keratinocytes and SCC cell lines, and correlated it to the differentiation and NOTCH1 levels. We analyzed the effects of Notch1 manipulation on CYFIP1 expression and confirmed the biding of Notch1 to the CYFIP1 promoter. CYFIP1 expression was down-regulated in SCC and correlated inversely with histological differentiation of tumors. As keratinocyte differentiation depends on Notch1 signaling, we investigated the influence of Notch1 on CYFIP1 expression. CYFIP1 mRNA was highly increased in human Notch1-overexpressing keratinocytes. Further manipulation of the Notch1 pathway in keratinocytes impacted CYFIP1 levels and chromatin immunoprecipitation assay confirmed the direct binding of Notch1 to the CYFIP1 promoter. CYFIP1 may be a link between loss of differentiation and invasive potential in malignant keratinocytes of cutaneous squamous cell carcinoma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma de Células Escamosas/fisiopatología , Regulación hacia Abajo , Receptor Notch1/metabolismo , Neoplasias Cutáneas/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Diferenciación Celular , Línea Celular , Movimiento Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Tamoxifeno/farmacología , Factor de Transcripción HES-1/metabolismo
14.
PLoS One ; 12(9): e0185028, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28931048

RESUMEN

Cutaneous squamous cell carcinoma (SCC) is the second most common human skin cancer with a rapidly increasing incidence among the Caucasian population. Among the many regulators, responsible for cancer progression and growth, microRNAs (miRNA) are generally accepted as key players by now. In our current study we found that microRNA-181a (miR-181a) shows low abundance in SCC compared to normal epidermal skin. In vitro, miRNA downregulation in normal primary keratinocytes induced increased proliferation, while in vivo miR-181a downregulation in HaCaT normal keratinocytes showed tumor-like growth increase up to 50%. Inversely, upregulation of these miRNAs in cancer cells lead to reduced cellular proliferation and induction of apoptosis in vitro. An in vivo therapeutic model with induced miR-181a expression in SCC13 cancer cells reduced tumor formation in mice by 80%. Modulation of miR-181a levels showed an inverse correlation with the proto-oncogene KRAS both on mRNA and protein level by direct interaction. Knockdown of KRAS mimicked the anti-proliferative effects of miR-181a overexpression in patient-derived SCC cells and abolished the enhanced viability of HaCaT cells following miR-181a knockdown. Furthermore, phospho-ERK levels correlated with KRAS levels, suggesting that the observed effects were mediated via the MAPK signaling pathway. miR-181a seemed regulated during keratinocyte differentiation probably in order to amplify the tumor suppressive character of differentiation. Taken together, miR-181a plays a crucial tumor suppressive role in SCC by targeting KRAS and could be a promising candidate for a miRNA based therapy.


Asunto(s)
Carcinoma de Células Escamosas/patología , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias Cutáneas/patología , Piel/patología , Animales , Apoptosis , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Diferenciación Celular , Movimiento Celular , Femenino , Humanos , Ratones , Ratones Desnudos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal , Piel/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo
16.
Nat Commun ; 5: 5099, 2014 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-25283360

RESUMEN

The Nrf2 transcription factor controls the expression of genes involved in the antioxidant defense system. Here, we identified Nrf2 as a novel regulator of desmosomes in the epidermis through the regulation of microRNAs. On Nrf2 activation, expression of miR-29a and miR-29b increases in cultured human keratinocytes and in mouse epidermis. Chromatin immunoprecipitation identified the Mir29ab1 and Mir29b2c genes as direct Nrf2 targets in keratinocytes. While binding of Nrf2 to the Mir29ab1 gene activates expression of miR-29a and -b, the Mir29b2c gene is silenced by DNA methylation. We identified desmocollin-2 (Dsc2) as a major target of Nrf2-induced miR-29s. This is functionally important, since Nrf2 activation in keratinocytes of transgenic mice causes structural alterations of epidermal desmosomes. Furthermore, the overexpression of miR-29a/b or knockdown of Dsc2 impairs the formation of hyper-adhesive desmosomes in keratinocytes, whereas Dsc2 overexpression has the opposite effect. These results demonstrate that a novel Nrf2-miR-29-Dsc2 axis controls desmosome function and cutaneous homeostasis.


Asunto(s)
Desmocolinas/metabolismo , Desmosomas/metabolismo , Queratinocitos/citología , MicroARNs/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Islas de CpG , Metilación de ADN , Regulación de la Expresión Génica , Silenciador del Gen , Homeostasis , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Piel/metabolismo
17.
J Invest Dermatol ; 134(7): 1998-2004, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24509533

RESUMEN

Cutaneous squamous cell carcinoma (SCC) represents the most important cutaneous complication following organ transplantation. It develops mostly on sun-exposed areas. A recent study showed the role of activating transcription factor 3 (ATF3) in SCC development following treatment with calcineurin inhibitors. It has been reported that ATF3, which may act as an oncogene, is under negative calcineurin/nuclear factor of activated T cells (NFAT) control and is upregulated by calcineurin inhibitors. Still, these findings do not fully explain the preferential appearance of SCC on chronically sun-damaged skin. We analyzed the influence of UV radiation on ATF3 expression and its potential role in SCC development. We found that ATF3 is a specifically induced AP1 member in SCC of transplanted patients. Its expression was strongly potentiated by combination of cyclosporine A and UVA treatment. UVA induced ATF3 expression through reactive oxygen species-mediated nuclear factor erythroid 2-related factor 2 (NRF2) activation independently of calcineurin/NFAT inhibition. Activated NRF2 directly binds to ATF3 promoter, thus inducing its expression. These results demonstrate two mechanisms that independently induce and, when combined together, potentiate the expression of ATF3, which may then force SCC development. Taking into account the previously defined role of ATF3 in the SCC development, these findings may provide an explanation and a mechanism for the frequently observed burden on SCCs on sun-exposed areas of the skin in organ transplant recipients treated by calcineurin inhibitors.


Asunto(s)
Factor de Transcripción Activador 3/genética , Carcinoma de Células Escamosas/genética , Ciclosporina/farmacología , Neoplasias Inducidas por Radiación/genética , Neoplasias Cutáneas/genética , Rayos Ultravioleta/efectos adversos , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/patología , Humanos , Inmunosupresores/farmacología , Queratinocitos/citología , Queratinocitos/fisiología , Queratinocitos/efectos de la radiación , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Inducidas por Radiación/patología , Técnicas de Cultivo de Órganos , Trasplante de Órganos/efectos adversos , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Piel/patología , Piel/efectos de la radiación , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/patología , Células Tumorales Cultivadas
19.
J Clin Invest ; 120(5): 1561-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20424327

RESUMEN

Cleft palate is a common congenital disorder that affects up to 1 in 2,500 live human births and results in considerable morbidity to affected individuals and their families. The etiology of cleft palate is complex, with both genetic and environmental factors implicated. Mutations in the transcription factor-encoding genes p63 and interferon regulatory factor 6 (IRF6) have individually been identified as causes of cleft palate; however, a relationship between the key transcription factors p63 and IRF6 has not been determined. Here, we used both mouse models and human primary keratinocytes from patients with cleft palate to demonstrate that IRF6 and p63 interact epistatically during development of the secondary palate. Mice simultaneously carrying a heterozygous deletion of p63 and the Irf6 knockin mutation R84C, which causes cleft palate in humans, displayed ectodermal abnormalities that led to cleft palate. Furthermore, we showed that p63 transactivated IRF6 by binding to an upstream enhancer element; genetic variation within this enhancer element is associated with increased susceptibility to cleft lip. Our findings therefore identify p63 as a key regulatory molecule during palate development and provide a mechanism for the cooperative role of p63 and IRF6 in orofacial development in mice and humans.


Asunto(s)
Fisura del Paladar/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Mutación , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Animales , Sitios de Unión , Elementos de Facilitación Genéticos , Epistasis Genética , Variación Genética , Heterocigoto , Queratinocitos/citología , Ratones , Modelos Biológicos , Activación Transcripcional
20.
J Clin Invest ; 120(2): 446-56, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20071778

RESUMEN

The Rho family GTPases Cdc42 and Rac1 are critical regulators of the actin cytoskeleton and are essential for skin and hair function. Wiskott-Aldrich syndrome family proteins act downstream of these GTPases, controlling actin assembly and cytoskeletal reorganization, but their role in epithelial cells has not been characterized in vivo. Here, we used a conditional knockout approach to assess the role of neural Wiskott-Aldrich syndrome protein (N-WASP), the ubiquitously expressed Wiskott-Aldrich syndrome-like (WASL) protein, in mouse skin. We found that N-WASP deficiency in mouse skin led to severe alopecia, epidermal hyperproliferation, and ulceration, without obvious effects on epidermal differentiation and wound healing. Further analysis revealed that the observed alopecia was likely the result of a progressive and ultimately nearly complete block in hair follicle (HF) cycling by 5 months of age. N-WASP deficiency also led to abnormal proliferation of skin progenitor cells, resulting in their depletion over time. Furthermore, N-WASP deficiency in vitro and in vivo correlated with decreased GSK-3beta phosphorylation, decreased nuclear localization of beta-catenin in follicular keratinocytes, and decreased Wnt-dependent transcription. Our results indicate a critical role for N-WASP in skin function and HF cycling and identify a link between N-WASP and Wnt signaling. We therefore propose that N-WASP acts as a positive regulator of beta-catenin-dependent transcription, modulating differentiation of HF progenitor cells.


Asunto(s)
Folículo Piloso/fisiología , Fenómenos Fisiológicos de la Piel , Proteína Neuronal del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/fisiología , Alopecia/genética , Animales , Diferenciación Celular , División Celular , Células Epidérmicas , Epidermis/patología , Eliminación de Gen , Folículo Piloso/citología , Queratinocitos/citología , Ratones , Ratones Noqueados , Úlcera Cutánea/genética , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/genética , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética , Proteína Neuronal del Síndrome de Wiskott-Aldrich/fisiología , Cicatrización de Heridas , beta Catenina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA