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 ; 11(4): 564-76, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25892239

RESUMEN

Understanding the molecular pathways that contribute to the aggressive behavior of HER2-positive breast cancers may aid in the development of novel therapeutic interventions. Here, we show that CDCP1 and HER2 are frequently co-overexpressed in metastatic breast tumors and associated with poor patient prognosis. HER2 and CDCP1 co-overexpression leads to increased transformation ability, cell migration, and tumor formation in vivo, and enhanced HER2 activation and downstream signaling in different breast cancer cell lines. Mechanistically, we demonstrate that CDCP1 binds to HER2 through its intracellular domain, thereby increasing HER2 interaction with the non-receptor tyrosine kinase c-SRC (SRC), leading to trastuzumab resistance. Taken together, our findings establish that CDCP1 is a modulator of HER2 signaling and a biomarker for the stratification of breast cancer patients with poor prognosis. Our results also provide a rationale for therapeutic targeting of CDCP1 in HER2-positive breast cancer patients.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias de la Mama/metabolismo , Carcinogénesis/metabolismo , Moléculas de Adhesión Celular/metabolismo , Resistencia a Antineoplásicos , Proteínas de Neoplasias/metabolismo , Receptor ErbB-2/metabolismo , Animales , Antígenos CD/genética , Antígenos de Neoplasias , Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Carcinogénesis/genética , Moléculas de Adhesión Celular/genética , Femenino , Humanos , Células MCF-7 , Ratones , Proteínas de Neoplasias/genética , Unión Proteica , Receptor ErbB-2/genética , Trastuzumab/farmacología , Familia-src Quinasas/metabolismo
2.
Cell Rep ; 9(1): 75-89, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25263564

RESUMEN

Prosenescence therapy has recently emerged as a novel therapeutic approach for treating cancer. However, this concept is challenged by conflicting evidence showing that the senescence-associated secretory phenotype (SASP) of senescent tumor cells can have pro- as well as antitumorigenic effects. Herein, we report that, in Pten-null senescent tumors, activation of the Jak2/Stat3 pathway establishes an immunosuppressive tumor microenvironment that contributes to tumor growth and chemoresistance. Activation of the Jak2/Stat3 pathway in Pten-null tumors is sustained by the downregulation of the protein tyrosine phosphatase PTPN11/SHP2, providing evidence for the existence of a novel PTEN/SHP2 axis. Importantly, treatment with docetaxel in combination with a JAK2 inhibitor reprograms the SASP and improves the efficacy of docetaxel-induced senescence by triggering a strong antitumor immune response in Pten-null tumors. Altogether, these data demonstrate that immune surveillance of senescent tumor cells can be suppressed in specific genetic backgrounds but also evoked by pharmacological treatments.


Asunto(s)
Antineoplásicos/farmacología , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/inmunología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/inmunología , Animales , Senescencia Celular/inmunología , Citocinas/inmunología , Docetaxel , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Transgénicos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transducción de Señal , Taxoides/farmacología , Microambiente Tumoral
3.
Nature ; 515(7525): 134-7, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25156255

RESUMEN

Aberrant activation of oncogenes or loss of tumour suppressor genes opposes malignant transformation by triggering a stable arrest in cell growth, which is termed cellular senescence. This process is finely tuned by both cell-autonomous and non-cell-autonomous mechanisms that regulate the entry of tumour cells to senescence. Whether tumour-infiltrating immune cells can oppose senescence is unknown. Here we show that at the onset of senescence, PTEN null prostate tumours in mice are massively infiltrated by a population of CD11b(+)Gr-1(+) myeloid cells that protect a fraction of proliferating tumour cells from senescence, thus sustaining tumour growth. Mechanistically, we found that Gr-1(+) cells antagonize senescence in a paracrine manner by interfering with the senescence-associated secretory phenotype of the tumour through the secretion of interleukin-1 receptor antagonist (IL-1RA). Strikingly, Pten-loss-induced cellular senescence was enhanced in vivo when Il1ra knockout myeloid cells were adoptively transferred to PTEN null mice. Therapeutically, docetaxel-induced senescence and efficacy were higher in PTEN null tumours when the percentage of tumour-infiltrating CD11b(+)Gr-1(+) myeloid cells was reduced using an antagonist of CXC chemokine receptor 2 (CXCR2). Taken together, our findings identify a novel non-cell-autonomous network, established by innate immunity, that controls senescence evasion and chemoresistance. Targeting this network provides novel opportunities for cancer therapy.


Asunto(s)
Movimiento Celular , Senescencia Celular , Células Mieloides/citología , Células Mieloides/metabolismo , Neoplasias de la Próstata/patología , Receptores de Quimiocina/metabolismo , Animales , Senescencia Celular/efectos de los fármacos , Progresión de la Enfermedad , Docetaxel , Resistencia a Antineoplásicos , Humanos , Inmunidad Innata , Proteína Antagonista del Receptor de Interleucina 1/deficiencia , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-1alfa/inmunología , Interleucina-1alfa/metabolismo , Masculino , Ratones , Células Mieloides/trasplante , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Receptores de Interleucina-8B/antagonistas & inhibidores , Taxoides/farmacología , Escape del Tumor , Microambiente Tumoral
4.
FEBS J ; 280(23): 6045-60, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24028392

RESUMEN

The protein dysferlin is abundantly expressed in skeletal and cardiac muscles, where its main function is membrane repair. Mutations in the dysferlin gene are involved in two autosomal recessive muscular dystrophies: Miyoshi myopathy and limb-girdle muscular dystrophy type 2B. Development of effective therapies remains a great challenge. Strategies to repair the dysferlin gene by skipping mutated exons, using antisense oligonucleotides (AONs), may be suitable only for a subset of mutations, while cell and gene therapy can be extended to all mutations. AON-treated blood-derived CD133+ stem cells isolated from patients with Miyoshi myopathy led to partial dysferlin reconstitution in vitro but failed to express dysferlin after intramuscular transplantation into scid/blAJ dysferlin null mice. We thus extended these experiments producing the full-length dysferlin mediated by a lentiviral vector in blood-derived CD133+ stem cells isolated from the same patients. Transplantation of engineered blood-derived CD133+ stem cells into scid/blAJ mice resulted in sufficient dysferlin expression to correct functional deficits in skeletal muscle membrane repair. Our data suggest for the first time that lentivirus-mediated delivery of full-length dysferlin in stem cells isolated from Miyoshi myopathy patients could represent an alternative therapeutic approach for treatment of dysferlinopathies.


Asunto(s)
Antígenos CD/metabolismo , Miopatías Distales/terapia , Glicoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/terapia , Oligonucleótidos Antisentido/farmacología , Péptidos/metabolismo , Trasplante de Células Madre , Células Madre/citología , Antígeno AC133 , Adulto , Animales , Western Blotting , Células Cultivadas , Miopatías Distales/genética , Miopatías Distales/patología , Disferlina , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Hibridación Fluorescente in Situ , Inyecciones Intramusculares , Lentivirus/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos A , Ratones SCID , Proteínas Musculares/genética , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patología , Mutación/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo
5.
Blood ; 121(20): 4110-4, 2013 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-23550036

RESUMEN

Plasma cells (PCs) are terminally differentiated cells of the B-cell lineage that secrete antibodies at a high rate and are thought to lack the expression of the B-cell receptor (BCR). Here, we report that human IgA and IgM, unlike IgG, PCs express a membrane functional BCR associated with the Igα/Igß heterodimer. BCR cross-linking on IgA and IgM PCs led to Ca(2+) mobilization and extracellular signal-regulated kinase 1/2 and AKT phosphorylation and impacted survival of IgA PCs. These findings demonstrate a significant difference between human IgG, IgM, and IgA PCs and suggest that the IgA PC repertoire may be modulated by specific antigens with implications for the regulation of the mucosal immune system.


Asunto(s)
Inmunoglobulina A/metabolismo , Inmunoglobulina M/metabolismo , Células Plasmáticas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , ADP-Ribosil Ciclasa 1/metabolismo , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Antígenos CD79/metabolismo , Supervivencia Celular/inmunología , Células Cultivadas , Humanos , Memoria Inmunológica/inmunología , Memoria Inmunológica/fisiología , Glicoproteínas de Membrana/metabolismo , Células Plasmáticas/inmunología
6.
J Neurosci Rural Pract ; 3(3): 301-10, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23188983

RESUMEN

BACKGROUND: there is an intimate relation between transition metals and cell homeostasis due to the physiological necessity of metals in vivo. Particularly, iron (ferrous and ferric state) is utilized in many physiological processes of the cell but in excess has been linked with negative role contributing in many neurodegenerative processes. OBJECTIVE: the aim of this study was to investigate which oxidation state of ionic iron (Ferrous (II) versus Ferric (III)) is more toxic to neuronal cells (SHSY(5)Y). MATERIALS AND METHODS: The neuroblastoma (SHSY(5)Y) cells were exposed to varying concentration of ferric and ferrous iron. Morphological studies using immunofluorescence staining and microscopic analysis as confirmed by intracellular glutathione (GSH) test demonstrated oxidative stress to cells in iron microenvironment. In addition, MTT assay was performed to evaluate the viability and metabolic state of the cells. RESULTS: the results showed that ferrous form has significantly higher toxicity compared to the ferric ionic state of higher concentration. In addition, microscopic analysis shows cell fenestration at higher concentrations and swelling at intermediate ferric dosages as demonstrated by atomic force microscopy (AFM). Interestingly, the addition of a differentiation inducing factor, trans-retinoic rcid (RA) retains significant viability and morphological features of the cells irrespective of the ionic state of the iron. AFM images revealed clustered aggregates arising from iron chelation with RA. CONCLUSIONS: the results indicate that Fe (II) has more toxic effects on cells. In addition, it could be an interesting finding with respect to the antioxidant properties of RA as a chelating agent for the neurodegenerative therapeutics.

7.
Nat Cell Biol ; 13(3): 292-302, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21336312

RESUMEN

Two major mechanisms have been causally implicated in the establishment of cellular senescence: the activation of the DNA damage response (DDR) pathway and the formation of senescence-associated heterochromatic foci (SAHF). Here we show that in human fibroblasts resistant to premature p16(INK4a) induction, SAHF are preferentially formed following oncogene activation but are not detected during replicative cellular senescence or on exposure to a variety of senescence-inducing stimuli. Oncogene-induced SAHF formation depends on DNA replication and ATR (ataxia telangiectasia and Rad3-related). Inactivation of ATM (ataxia telangiectasia mutated) or p53 allows the proliferation of oncogene-expressing cells that retain increased heterochromatin induction. In human cancers, levels of heterochromatin markers are higher than in normal tissues, and are independent of the proliferative index or stage of the tumours. Pharmacological and genetic perturbation of heterochromatin in oncogene-expressing cells increase DDR signalling and lead to apoptosis. In vivo, a histone deacetylase inhibitor (HDACi) causes heterochromatin relaxation, increased DDR, apoptosis and tumour regression. These results indicate that heterochromatin induced by oncogenic stress restrains DDR and suggest that the use of chromatin-modifying drugs in cancer therapies may benefit from the study of chromatin and DDR status of tumours.


Asunto(s)
Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Daño del ADN , Heterocromatina/genética , Neoplasias/metabolismo , Oncogenes , Animales , Apoptosis , Línea Celular Tumoral , Cromatina/metabolismo , Replicación del ADN , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Microscopía Fluorescente/métodos , Trasplante de Neoplasias , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...