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.
J Invest Dermatol ; 144(1): 73-83.e10, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37543242

RESUMEN

The p63 transcription factor is critical for epidermis formation in embryonic development, but its role in the adult epidermis is poorly understood. In this study, we show that acute genetic ablation of ΔNp63, the main p63 isoform, in adult epidermis disrupts keratinocyte proliferation and self-maintenance and, unexpectedly, triggers an inflammatory psoriasis-like condition. Mechanistically, single-cell RNA sequencing revealed the downregulation of cell cycle genes, upregulation of differentiation markers, and induction of several proinflammatory pathways in ΔNp63-ablated keratinocytes. Intriguingly, ΔNp63-ablated cells disappear by 3 weeks after ablation, at the expense of the remaining nonablated cells. This is not associated with active cell death and is likely due to reduced self-maintenance and enhanced differentiation. Indeed, in vivo wound healing, a physiological readout of the epidermal stem cell function, is severely impaired upon ΔNp63 ablation. We found that the Wnt signaling pathway (Wnt10A, Fzd6, Fzd10) and the activator protein 1 (JunB, Fos, FosB) factors are the likely ΔNp63 effectors responsible for keratinocyte proliferation/stemness and suppression of differentiation, respectively, whereas IL-1a, IL-18, IL-24, and IL-36γ are the likely negative effectors responsible for suppression of inflammation. These data establish ΔNp63 as a critical node that coordinates epidermal homeostasis, stemness, and suppression of inflammation, upstream of known regulatory pathways.


Asunto(s)
Células Epidérmicas , Epidermis , Humanos , Adulto , Epidermis/metabolismo , Queratinocitos/metabolismo , Homeostasis , Inflamación/genética , Inflamación/metabolismo
3.
Cell Death Dis ; 12(6): 525, 2021 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-34023861

RESUMEN

While pregnancy is known to reduce a woman's life-long risk of breast cancer, clinical data suggest that it can specifically promote HER2 (human EGF receptor 2)-positive breast cancer subtype (HER2+ BC). HER2+ BC, characterized by amplification of HER2, comprises about 20% of all sporadic breast cancers and is more aggressive than hormone receptor-positive breast cancer (the majority of cases). Consistently with human data, pregnancy strongly promotes HER2+ BC in genetic mouse models. One proposed mechanism of this is post-pregnancy accumulation of PIMECs (pregnancy-identified mammary epithelial cells), tumor-initiating cells for HER2+ BC in mice. We previously showed that p63, a homologue of the tumor suppressor p53, is required to maintain the post-pregnancy number of PIMECs and thereby promotes HER2+ BC. Here we set to test whether p63 also affects the intrinsic tumorigenic properties of PIMECs. To this end, we FACS-sorted YFP-labeled PIMECs from p63+/-;ErbB2 and control p63+/+;ErbB2 females and injected their equal amounts into immunodeficient recipients. To our surprise, p63+/- PIMECs showed increased, rather than decreased, tumorigenic capacity in vivo, i.e., significantly accelerated tumor onset and tumor growth, as well as increased self-renewal in mammosphere assays and proliferation in vitro and in vivo. The underlying mechanism of these phenotypes seems to be a specific reduction of the tumor suppressor TAp63 isoform in p63+/- luminal cells, including PIMECs, with concomitant aberrant upregulation of the oncogenic ΔNp63 isoform, as determined by qRT-PCR and scRNA-seq analyses. In addition, scRNA-seq revealed upregulation of several cancer-associated (Il-4/Il-13, Hsf1/HSP), oncogenic (TGFß, NGF, FGF, MAPK) and self-renewal (Wnt, Notch) pathways in p63+/-;ErbB2 luminal cells and PIMECs per se. Altogether, these data reveal a complex role of p63 in PIMECs and pregnancy-associated HER2+ BC: maintaining the amount of PIMECs while suppressing their intrinsic tumorigenic capacity.


Asunto(s)
Neoplasias de la Mama/patología , Células Madre Neoplásicas/fisiología , Embarazo/fisiología , Transactivadores/fisiología , Animales , Mama/patología , Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Epiteliales/patología , Células Epiteliales/fisiología , Femenino , Genes erbB-2 , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Células Madre Neoplásicas/patología , Transactivadores/genética
4.
Cell Death Dis ; 12(1): 126, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33500390

RESUMEN

Breast cancer is the leading cause of cancer-related death in women worldwide. Human epidermal growth factor receptor 2 (HER2)-positive subtype comprises 20% of sporadic breast cancers and is an aggressive disease. While targeted therapies have greatly improved its management, primary and acquired resistance remain a major roadblock to making it a curable malignancy. Ganetespib, an Hsp90 (Heat shock protein 90) small molecule inhibitor, shows preferential efficacy in HER2-positive breast cancer, including therapy-refractory cases, and has an excellent safety profile in ongoing clinical trials (38 in total, six on breast cancer). However, Ganetespib itself evokes acquired resistance, which is a significant obstacle to its clinical advancement. Here, we show that Ganetespib potently, albeit temporarily, suppresses HER2-positive breast cancer in genetic mouse models, but the animals eventually succumb via acquired resistance. We found that Ganetespib-resistant tumors upregulate several compensatory HSPs, as well as a wide network of phospho-activated receptor tyrosine kinases (RTKs), many of which are HSP clients. Downstream of p-RTKs, the MAPK pathway remains suppressed in the resistant tumors, as is HER2 itself. In contrast, the p-RTK effector Akt is stabilized and phospho-activated. Notably, pharmacological inhibition of Akt significantly delays acquired Ganetespib resistance, by 50%. These data establish Akt as a unifying actionable node downstream of the broadly upregulated HSP/p-RTK resistance program and suggests that Akt co-targeting with Ganetespib may be a superior therapeutic strategy in the clinic.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/uso terapéutico , Triazoles/uso terapéutico , Animales , Neoplasias de la Mama/patología , Femenino , Proteínas HSP90 de Choque Térmico/farmacología , Humanos , Ratones , Triazoles/farmacología
5.
Cell Death Dis ; 9(6): 621, 2018 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-29799521

RESUMEN

Despite success of ERBB2-targeted therapies such as lapatinib, resistance remains a major clinical concern. Multiple compensatory receptor tyrosine kinase (RTK) pathways are known to contribute to lapatinib resistance. The heterogeneity of these adaptive responses is a significant hurdle for finding most effective combinatorial treatments. The goal of this study was to identify a unifying molecular mechanism whose targeting could help prevent and/or overcome lapatinib resistance. Using the MMTV-ERBB2;mutant p53 (R175H) in vivo mouse model of ERBB2-positive breast cancer, together with mouse and human cell lines, we compared lapatinib-resistant vs. lapatinib-sensitive tumor cells biochemically and by kinome arrays and evaluated their viability in response to a variety of compounds affecting heat shock response. We found that multiple adaptive RTKs are activated in lapatinib-resistant cells in vivo, some of which have been previously described (Axl, MET) and some were novel (PDGFRα, PDGFRß, VEGFR1, MUSK, NFGR). Strikingly, all lapatinib-resistant cells show chronically activated HSF1 and its transcriptional targets, heat shock proteins (HSPs), and, as a result, superior tolerance to proteotoxic stress. Importantly, lapatinib-resistant tumors and cells retained sensitivity to Hsp90 and HSF1 inhibitors, both in vitro and in vivo, thus providing a unifying and actionable therapeutic node. Indeed, HSF1 inhibition simultaneously downregulated ERBB2, adaptive RTKs and mutant p53, and its combination with lapatinib prevented development of lapatinib resistance in vitro. Thus, the kinome adaptation in lapatinib-resistant ERBB2-positive breast cancer cells is governed, at least in part, by HSF1-mediated heat shock pathway, providing a novel potential intervention strategy to combat resistance.


Asunto(s)
Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Factores de Transcripción del Choque Térmico/metabolismo , Lapatinib/farmacología , Receptor ErbB-2/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Ratones Endogámicos C57BL , Proteolisis/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
6.
Cell Death Dis ; 8(3): e2661, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28277540

RESUMEN

Missense mutations in TP53 comprise >75% of all p53 alterations in cancer, resulting in highly stabilized mutant p53 proteins that not only lose their tumor-suppressor activity, but often acquire oncogenic gain-of-functions (GOFs). GOF manifests itself in accelerated tumor onset, increased metastasis, increased drug resistance and shortened survival in patients and mice. A known prerequisite for GOF is mutant p53 protein stabilization, which itself is linked to aberrant protein conformation. However, additional determinants for mutant p53 stabilization likely exist. Here we show that in initially heterozygous mouse tumors carrying the hotspot GOF allele R248Q (p53Q/+), another necessary prerequisite for mutant p53 stabilization and GOF in vivo is loss of the remaining wild-type p53 allele, termed loss-of-heterozygosity (LOH). Thus, in mouse tumors with high frequency of p53 LOH (osteosarcomas and fibrosarcomas), we find that mutant p53 protein is stabilized (16/17 cases, 94%) and tumor onset is significantly accelerated compared with p53+/- tumors (GOF). In contrast, in mouse tumors with low frequency of p53 LOH (MMTV-Neu breast carcinomas), mutant p53 protein is not stabilized (16/20 cases, 80%) and GOF is not observed. Of note, human genomic databases (TCGA, METABRIC etc.) show a high degree of p53 LOH in all examined tumor types that carry missense p53 mutations, including sarcomas and breast carcinomas (with and without HER2 amplification). These data - while cautioning that not all genetic mouse models faithfully represent the human situation - demonstrate for the first time that p53 LOH is a critical prerequisite for missense mutant p53 stabilization and GOF in vivo.


Asunto(s)
Neoplasias de la Mama/genética , Pérdida de Heterocigocidad/genética , Proteína p53 Supresora de Tumor/genética , Alelos , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Mutación Missense/genética
7.
Cell Death Dis ; 8(3): e2683, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28300840

RESUMEN

The DNA-alkylating cytotoxic agent cyclophosphamide (CTX) is commonly used in the clinic to treat hematological malignancies like lymphomas and leukemias as well as solid tumors, but shows dose-dependent potentially life-threatening toxicities and can induce secondary malignancies. Thus, the clinical utility of CTX would be improved if a companion drug could be identified that allows lowering the CTX dose, while maintaining or even increasing its antineoplastic therapeutic efficacy. In mouse models, high-dose CTX (300 mg/kg) is effective in treating T-lymphomas, while low dose (defined here as 100 mg/kg) is ineffective. We previously showed that the HSP90 inhibitor ganetespib potently suppresses T-lymphoma initiation and progression and extends overall survival (OS) in hotspot knockin mice expressing the p53 gain-of-function mutants R175H and R248Q (mutp53) by 30-59%. Here we asked whether ganetespib could potentiate the effect of low-dose CTX (100 mg/kg) in the autochthonous T-lymphoma-bearing mutp53 R248Q mouse model. Indeed, combinatorial CTX/ganetespib synergistically suppresses growth of autochthonous T-lymphomas in R248Q (p53Q/-) but not p53-/- control mice by reducing mutp53 levels and triggering apoptosis. Combinatorial treatment extends progression-free (PFS) and OS in p53Q/- mice significantly longer than in p53-/- mice. Specifically, PFS of p53Q/- mice improves 8.9-fold over CTX alone versus 3.6-fold in p53-/- mice. Likewise, OS of R248Q/- mice improves 3.6-fold, but worsens in p53-/- mice (0.85-fold) over CTX alone. Moreover, half of the p53Q/- mice on combinatorial treatment lived over 60 days, and one animal reached 121 days. In contrast, p53Q/- mice on single-drug treatment and p53-/- mice on any treatment lived less than 24 days. In sum, ganetespib synergizes with a sub-effective dose of CTX in mutp53 T-lymphomas by suppressing tumor growth and extending survival. Our results provide a potential strategy to reduce the effective clinical dose of CTX in mutant p53-bearing malignancies and attenuate CTX toxicity.


Asunto(s)
Antineoplásicos/farmacología , Ciclofosfamida/farmacología , Linfoma/tratamiento farmacológico , Triazoles/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Proteínas HSP90 de Choque Térmico/metabolismo , Linfoma/genética , Ratones , Proteína p53 Supresora de Tumor/genética
9.
Artículo en Inglés | MEDLINE | ID: mdl-25954247

RESUMEN

The main tumor suppressor function of p53 as a "guardian of the genome" is to respond to cellular stress by transcriptional activation of apoptosis, growth arrest, or senescence in damaged cells. Not surprisingly, mutations in the p53 gene are the most frequent genetic alteration in human cancers. Importantly, mutant p53 (mutp53) proteins not only lose their wild-type tumor suppressor activity but also can actively promote tumor development. Two main mechanisms accounting for mutp53 proto-oncogenic activity are inhibition of the wild-type p53 in a dominant-negative fashion and gain of additional oncogenic activities known as gain-of-function (GOF). Here, we discuss a novel mechanism of mutp53 GOF, which relies on its oncogenic cooperation with the heat shock machinery. This coordinated adaptive mechanism renders cancer cells more resistant to proteotoxic stress and provides both, a strong survival advantage to cancer cells and a promising means for therapeutic intervention.

10.
Methods Mol Biol ; 962: 157-64, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150445

RESUMEN

Here we describe a method for generating induced pluripotent stem (iPS) cells from mouse embryonic fibroblasts (MEFs). Recombinant retroviruses carrying human transcription factors for Klf4, Oct3/4, Sox2, with or without c-Myc, are used to transduce early passage MEFs several times. Based on morphologic criteria, the resulting iPS colonies are picked manually at first, and then propagated and expanded by standard methods. iPS cells can then be differentiated into virtually any cell type or lineage, thus allowing for discoveries of new functions of p53 and mutant p53.


Asunto(s)
Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Diferenciación Celular , Criopreservación , Fibroblastos/citología , Genes myc , Vectores Genéticos , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
11.
Leuk Lymphoma ; 53(11): 2116-29, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22497596

RESUMEN

p53, mutated in over half of human cancers and about 13% of all hematological malignancies, maintains genomic integrity and triggers cellular senescence and apoptosis of damaged cells. In contrast to p53, the homologs p73 and p63 play critical roles in development of the central nervous system and skin/limbs, respectively. Moreover, dependent on the context they can exert tumor suppressor activities that cooperate with p53. Unlike p53, p73 and p63 are rarely mutated in cancers. Instead, up-regulation of the anti-apoptotic dominant-negative ΔNp73 and ΔNp63 isoforms is the most frequent abnormality in solid cancers. In hematological malignancies the most frequent p73 defect is promoter methylation and loss of expression, associated with unfavorable clinical outcomes. This suggests an essential tumor suppressor role of p73 in blood cells, also supported by genetic mouse models. Many therapeutic approaches aiming to restore p73 activity are currently being investigated. In contrast, the most frequent p63 abnormality is protein overexpression, associated with higher disease grade and poorer prognosis. Surprisingly, although available data are still scarce, the emerging picture is up-regulation of transactivation-competent TAp63 isoforms, suggesting a tumor-promoting role in this context.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Leucemia/etiología , Linfoma/etiología , Proteínas de la Membrana/fisiología , Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Metilación de ADN , Proteínas de Unión al ADN/genética , Humanos , Leucemia/genética , Linfoma/genética , Proteínas de la Membrana/genética , Ratones , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/genética
12.
Dev Dyn ; 239(1): 261-70, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19842147

RESUMEN

The Wnt/beta-catenin signaling pathway regulates cell proliferation and cell fate determination in multiple systems. However, the subcellular localization of Wnt pathway components and the significance of this localization for the pathway regulation have not been extensively analyzed. Here we report that Xenopus Axin-related protein (XARP), a component of the beta-catenin destruction complex, is localized to the centrosome. This localization of XARP requires the presence of the DIX domain and an adjacent region. Since other components of the Wnt pathway have also been shown to associate with the centrosome, we tested a hypothesis that the beta-catenin destruction complex operates at the centrosome. However, XARP mutants with poor centrosomal localization revealed an enhanced rather than decreased ability to antagonize the Wnt/beta-catenin pathway. Our data are consistent with the idea that the inactivation of XARP at the centrosome is an important regulatory point in Wnt signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Centrosoma/metabolismo , Transducción de Señal/genética , Proteínas Wnt/metabolismo , Proteínas de Xenopus/metabolismo , beta Catenina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Animales , Embrión no Mamífero/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Luciferasas , Microinyecciones , Datos de Secuencia Molecular , Alineación de Secuencia , Transducción de Señal/fisiología , Proteínas de Xenopus/genética , Xenopus laevis
13.
Dev Biol ; 299(2): 398-410, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16973150

RESUMEN

The ubiquitin ligase Smurf1 can target a handful of signaling proteins for ubiquitin-mediated proteasomal destruction or functional modification, including TGF-beta receptors, Smads, transcription factors, RhoA and MEKK2. Smurf1 was initially implicated in BMP pathway regulation in embryonic development, but its potential role in vertebrate embryogenesis has yet to be clarified. Here we demonstrate that inhibition of Smurf1 in Xenopus laevis embryos with an antisense morpholino oligonucleotide or a dominant-negative protein disrupts early development, with the nervous system being the principal target. Smurf1 is enriched on the dorsal side of gastrula stage embryos, and blocking Smurf1 disturbs neural folding and neural, but not mesoderm differentiation, enhances BMP/Smad1 signaling, and elevates phospho-Smad1 levels in the dorsal ectoderm. We conclude that in Xenopus embryos, the BMP pathway is a major physiological target of Smurf1, and we propose that in normal development Smurf1 cooperates with secreted BMP antagonists to limit BMP signaling in dorsal ectoderm. Our data also reveal a novel role for Smurf1 and Smad1 in neural plate morphogenesis.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Gástrula/metabolismo , Sistema Nervioso/embriología , Proteína Smad1/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Xenopus laevis/embriología , Animales , Secuencia de Bases , Regulación del Desarrollo de la Expresión Génica , Sistema Nervioso/metabolismo , Alineación de Secuencia , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Xenopus laevis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...