Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 36(1): 109309, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34233193

RESUMEN

αvß8 integrin, a key activator of transforming growth factor ß (TGF-ß), inhibits anti-tumor immunity. We show that a potent blocking monoclonal antibody against αvß8 (ADWA-11) causes growth suppression or complete regression in syngeneic models of squamous cell carcinoma, mammary cancer, colon cancer, and prostate cancer, especially when combined with other immunomodulators or radiotherapy. αvß8 is expressed at the highest levels in CD4+CD25+ T cells in tumors, and specific deletion of ß8 from T cells is as effective as ADWA-11 in suppressing tumor growth. ADWA-11 increases expression of a suite of genes in tumor-infiltrating CD8+ T cells normally inhibited by TGF-ß and involved in tumor cell killing, including granzyme B and interferon-γ. The in vitro cytotoxic effect of tumor CD8 T cells is inhibited by CD4+CD25+ cells, and this suppressive effect is blocked by ADWA-11. These findings solidify αvß8 integrin as a promising target for cancer immunotherapy.


Asunto(s)
Inmunidad , Inmunoterapia , Integrinas/metabolismo , Modelos Biológicos , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Anticuerpos Antineoplásicos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Granzimas/metabolismo , Interferón gamma/metabolismo , Depleción Linfocítica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Neoplasias/genética , Neoplasias/patología , Transducción de Señal , Proteína smad3/metabolismo , Análisis de Supervivencia , Linfocitos T Citotóxicos/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
2.
Clin Cancer Res ; 25(6): 1889-1900, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30530704

RESUMEN

PURPOSE: Over 95% of human anal cancers are etiologically associated with high-risk HPVs, with HPV type 16 (HPV16) the genotype most commonly found. Activating mutations in the catalytic subunit of Phosphatidylinositol (3,4,5)-trisphosphate kinase (PI3K), encoded by the Pik3ca gene, are detected in approximately 20% of human anal cancers.Experimental Design: We asked if common activating mutations in Pik3ca contribute to anal carcinogenesis using an established mouse model for anal carcinogenesis in which mice are topically treated with the chemical carcinogen 7,12-Dimethylbenz(a)anthracene (DMBA). Mice expressing in their anal epithelium one of two activating mutations in Pik3ca genes, Pik3caH1047R or Pik3caE545K , were monitored for anal carcinogenesis in the presence or absence of transgenes expressing the HPV16 E6 and E7 oncogenes. RESULTS: Both mutant forms of Pik3ca increased susceptibility to anal carcinogenesis in the absence of HPV16 oncogenes, and cooperated with HPV16 oncogenes to induce the highest level and earliest onset of anal cancers. The combination of HPV16 oncogenes and Pik3ca mutations led to anal cancers even in the absence of treatment with DMBA. We further observed that the investigational mTOR1/2 dual inhibitor, TAK-228, significantly reduced the size of anal cancer-derived tumor spheroids in vitro and reduced the growth rates of anal cancer-derived tumor grafts in vivo. CONCLUSIONS: These data demonstrate that activating mutations in Pik3ca drive anal carcinogenesis together with HPV16 oncogenes, and that the PI3K/mTOR pathway is a relevant target for therapeutic intervention.


Asunto(s)
Neoplasias del Ano/genética , Carcinogénesis/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Papillomavirus Humano 16/patogenicidad , Neoplasias Experimentales/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Canal Anal/efectos de los fármacos , Canal Anal/patología , Animales , Neoplasias del Ano/inducido químicamente , Neoplasias del Ano/tratamiento farmacológico , Neoplasias del Ano/virología , Benzoxazoles/administración & dosificación , Carcinogénesis/efectos de los fármacos , Carcinógenos/toxicidad , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Femenino , Mutación con Ganancia de Función , Humanos , Ratones , Ratones Transgénicos , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/virología , Cultivo Primario de Células , Pirimidinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas
3.
Proc Natl Acad Sci U S A ; 111(21): 7723-8, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24812125

RESUMEN

Outcome of TGFß1 signaling is context dependent and differs between individuals due to germ-line genetic variation. To explore innate genetic variants that determine differential outcome of reduced TGFß1 signaling, we dissected the modifier locus Tgfbm3, on mouse chromosome 12. On a NIH/OlaHsd genetic background, the Tgfbm3b(C57) haplotype suppresses prenatal lethality of Tgfb1(-/-) embryos and enhances nuclear accumulation of mothers against decapentaplegic homolog 2 (Smad2) in embryonic cells. Amino acid polymorphisms within a disintegrin and metalloprotease 17 (Adam17) can account, at least in part, for this Tgfbm3b effect. ADAM17 is known to down-regulate Smad2 signaling by shedding the extracellular domain of TGFßRI, and we show that the C57 variant is hypomorphic for down-regulation of Smad2/3-driven transcription. Genetic variation at Tgfbm3 or pharmacological inhibition of ADAM17, modulates postnatal circulating endothelial progenitor cell (CEPC) numbers via effects on TGFßRI activity. Because CEPC numbers correlate with angiogenic potential, this suggests that variant Adam17 is an innate modifier of adult angiogenesis, acting through TGFßR1. To determine whether human ADAM17 is also polymorphic and interacts with TGFß signaling in human vascular disease, we investigated hereditary hemorrhagic telangiectasia (HHT), which is caused by mutations in TGFß/bone morphogenetic protein receptor genes, ENG, encoding endoglin (HHT1), or ACVRL1 encoding ALK1 (HHT2), and considered a disease of excessive abnormal angiogenesis. HHT manifests highly variable incidence and severity of clinical features, ranging from small mucocutaneous telangiectases to life-threatening visceral and cerebral arteriovenous malformations (AVMs). We show that ADAM17 SNPs associate with the presence of pulmonary AVM in HHT1 but not HHT2, indicating genetic variation in ADAM17 can potentiate a TGFß-regulated vascular disease.


Asunto(s)
Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Vasos Sanguíneos/patología , Regulación de la Expresión Génica/fisiología , Variación Genética , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Proteína ADAM17 , Animales , Regulación de la Expresión Génica/genética , Humanos , Inmunohistoquímica , Luciferasas , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Transducción de Señal/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/genética
4.
Breast Cancer Res ; 15(2): R36, 2013 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-23621987

RESUMEN

INTRODUCTION: Early pregnancy has a strong protective effect against breast cancer in humans and rodents, but the underlying mechanism is unknown. Because breast cancers are thought to arise from specific cell subpopulations of mammary epithelia, we studied the effect of parity on the transcriptome and the differentiation/proliferation potential of specific luminal and basal mammary cells in mice. METHODS: Mammary epithelial cell subpopulations (luminal Sca1-, luminal Sca1+, basal stem/progenitor, and basal myoepithelial cells) were isolated by flow cytometry from parous and age-matched virgin mice and examined by using a combination of unbiased genomics, bioinformatics, in vitro colony formation, and in vivo limiting dilution transplantation assays. Specific findings were further investigated with immunohistochemistry in entire glands of parous and age-matched virgin mice. RESULTS: Transcriptome analysis revealed an upregulation of differentiation genes and a marked decrease in the Wnt/Notch signaling ratio in basal stem/progenitor cells of parous mice. Separate bioinformatics analyses showed reduced activity for the canonical Wnt transcription factor LEF1/TCF7 and increased activity for the Wnt repressor TCF3. This finding was specific for basal stem/progenitor cells and was associated with downregulation of potentially carcinogenic pathways and a reduction in the proliferation potential of this cell subpopulation in vitro and in vivo. As a possible mechanism for decreased Wnt signaling in basal stem/progenitor cells, we found a more than threefold reduction in the expression of the secreted Wnt ligand Wnt4 in total mammary cells from parous mice, which corresponded to a similar decrease in the proportion of Wnt4-secreting and estrogen/progesterone receptor-positive cells. Because recombinant Wnt4 rescued the proliferation defect of basal stem/progenitor cells in vitro, reduced Wnt4 secretion appears to be causally related to parity-induced alterations of basal stem/progenitor cell properties in mice. CONCLUSIONS: By revealing that parity induces differentiation and downregulates the Wnt/Notch signaling ratio and the in vitro and in vivo proliferation potential of basal stem/progenitor cells in mice, our study sheds light on the long-term consequences of an early pregnancy. Furthermore, it opens the door to future studies assessing whether inhibitors of the Wnt pathway may be used to mimic the parity-induced protective effect against breast cancer.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Epitelio/patología , Glándulas Mamarias Animales/citología , Receptores Notch/metabolismo , Células Madre/citología , Proteínas Wnt/metabolismo , Animales , Antígenos Ly , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Epitelio/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Técnicas para Inmunoenzimas , Glándulas Mamarias Animales/metabolismo , Proteínas de la Membrana , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Paridad , Embarazo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Madre/metabolismo , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
5.
Breast Cancer Res ; 14(5): R131, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-23062209

RESUMEN

INTRODUCTION: The tyrosine kinase receptors HER2 and HER3 play an important role in breast cancer. The HER2/HER3 heterodimer is a critical oncogenic unit associated with reduced relapse-free and decreased overall survival. While signaling cascades downstream of HER2 and HER3 have been studied extensively at the level of post-translational modification, little is known about the effects of HER2/HER3 overexpression and activation on gene expression in breast cancer. We have now defined the genetic landscape induced by activation of the HER2/HER3 unit in mammary cells, and have identified interleukin (IL)8 and CXCR1 as potential therapeutic targets for the treatment of HER2/HER3-overexpressing breast cancers. METHODS: Three-dimensional (3D) cultures, invasion and migration assays were used to determine the effects of HER2 and HER3 co-expression and activation. Gene expression analysis was performed to identify the gene network induced by HER2/HER3 in 3D cultures. Bioinformatic analysis and neutralizing antibodies were used to identify key mediators of HER2/HER3-evoked invasion. RESULTS: Co-expression of the tyrosine kinase receptors HER2 and HER3 induced migration and invasion of MCF10A cells. Microarray analysis of these cells revealed a specific "HER2/HER3 signature" comprising 80 upregulated transcripts, with IL8 being the highest (11-fold upregulation). Notably, examination of public datasets revealed high levels of IL8 transcripts in HER2-enriched as well as basal-like primary breast tumors, two subtypes characterized by a particularly poor prognosis. Moreover, IL8 expression correlated with high tumor grade and ER-negative status. Importantly, treatment with IL8-neutralizing antibodies prevented invasion of MCF10A-HER2/HER3 and BT474 cells in 3D cultures, highlighting the importance of IL8 autocrine signaling upon HER2/HER3 activation. CONCLUSIONS: Our findings demonstrate that HER2 and HER3 co-expression induces IL8 autocrine signaling, leading to the invasion of mammary cells. Agents targeting IL8 or its receptor CXCR1 may be useful for the treatment of HER2/HER3/IL8-positive breast cancers with invasive traits.


Asunto(s)
Comunicación Autocrina , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Expresión Génica , Interleucina-8/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Conjuntos de Datos como Asunto , Femenino , Perfilación de la Expresión Génica , Humanos , Modelos Biológicos , Clasificación del Tumor , Neurregulina-1/farmacología , Transducción de Señal , Esferoides Celulares , Células Tumorales Cultivadas
6.
Cancer Res ; 71(13): 4344-51, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21482677

RESUMEN

The phosphoinositide 3-kinase (PI3K) signaling cascade, a key mediator of cellular survival, growth, and metabolism, is frequently altered in human cancer. Activating mutations in PIK3CA, which encodes the α-catalytic subunit of PI3K, occur in approximately 30% of breast cancers. These mutations result in constitutive activity of the enzyme and are oncogenic, but it is not known whether they are sufficient to induce mammary carcinomas in mice. In the present study, we show that the expression of mutant PIK3CA H1047R in the luminal mammary epithelium evokes heterogeneous tumors that express luminal and basal markers and are positive for the estrogen receptor. Our results suggest that the PIK3CA H1047R oncogene targets a multipotent progenitor cell and, furthermore, show that this model recapitulates features of human breast tumors with PIK3CA H1047R.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias Mamarias Experimentales/genética , Fosfatidilinositol 3-Quinasas/genética , Animales , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Fosfatidilinositol 3-Quinasa Clase I , Células Epiteliales/enzimología , Células Epiteliales/patología , Humanos , Glándulas Mamarias Animales/enzimología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Mutación , Fosfatidilinositol 3-Quinasas/biosíntesis , Receptores de Estrógenos/biosíntesis
7.
Breast Cancer Res ; 12(5): 315, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21062517

RESUMEN

Seventy percent of breast tumors are estrogen receptor (ER) positive. Although endocrine therapy is successful for the majority of patients with ER-positive tumors, approximately 30% show de novo or acquired resistance and the underlying molecular mechanisms and biomarkers that predict such resistance remain elusive. Two recent papers report that hyperactivation of the phosphatidylinositol 3-kinase (PI3K) pathway produces resistance to tamoxifen. This raises the possibility that combining endocrine therapy and PI3K inhibition may be more effective than monotherapy for treating ER-positive breast tumors, either as first-line therapy for tumors with high PI3K activity or after the development of resistance to endocrine therapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Tamoxifeno/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/prevención & control , Resistencia a Antineoplásicos , Femenino , Humanos , Receptores de Estrógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...