Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Cell ; 179(1): 236-250.e18, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31495571

RESUMEN

Immunotherapy has revolutionized cancer treatment, yet most patients do not respond. Here, we investigated mechanisms of response by profiling the proteome of clinical samples from advanced stage melanoma patients undergoing either tumor infiltrating lymphocyte (TIL)-based or anti- programmed death 1 (PD1) immunotherapy. Using high-resolution mass spectrometry, we quantified over 10,300 proteins in total and ∼4,500 proteins across most samples in each dataset. Statistical analyses revealed higher oxidative phosphorylation and lipid metabolism in responders than in non-responders in both treatments. To elucidate the effects of the metabolic state on the immune response, we examined melanoma cells upon metabolic perturbations or CRISPR-Cas9 knockouts. These experiments indicated lipid metabolism as a regulatory mechanism that increases melanoma immunogenicity by elevating antigen presentation, thereby increasing sensitivity to T cell mediated killing both in vitro and in vivo. Altogether, our proteomic analyses revealed association between the melanoma metabolic state and the response to immunotherapy, which can be the basis for future improvement of therapeutic response.


Asunto(s)
Inmunoterapia/métodos , Melanoma/metabolismo , Melanoma/terapia , Mitocondrias/metabolismo , Proteómica/métodos , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/terapia , Traslado Adoptivo/métodos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Estudios de Cohortes , Femenino , Humanos , Metabolismo de los Lípidos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/inmunología , Resultado del Tratamiento , Adulto Joven
2.
J Pediatr Intensive Care ; 7(4): 207-209, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31073496

RESUMEN

Necrotizing pneumonia is a severe form of pneumonia that is mainly treated with conservative treatment, including antibiotics. We report a unique case of necrotizing pneumonia due to group A streptococcus infection in an 18-month-old boy who required extracorporeal membrane oxygenation (ECMO) support. Following surgical lobectomy, the child was weaned off ECMO and recovered uneventfully.

3.
Oncotarget ; 7(21): 30166-77, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-26885752

RESUMEN

As melanoma cells are immunogenic, they instigate an adaptive immune response and production of anti-tumor T-cells. A central factor in this interaction is CEACAM1 (carcinoembryonic antigen cell adhesion molecule 1), a transmembrane glycoprotein previously shown in our lab to protect melanoma cells from T cell-mediated killing. In this study, we examine the role of transcription factor SOX9 in the regulation of CEACAM1 expression and immune resistance in melanoma cells. Knockdown of endogenous SOX9 results in CEACAM1 up-regulation, while its overexpression leads to the opposite effect. We show that SOX9 controls CEACAM1 expression at a transcriptional level, but in an indirect manner, as regulation of the CEACAM1 promoter remains intact even when all eight potential SOX9-binding sites are abolished. A series of promoter truncations localizes the SOX9-controlled area to the proximal 200bp of the promoter. Point mutations in putative Sp1 and ETS1 binding sites identify these transcription factors as the primary SOX9-controlled mediators. Co-immunoprecipitation studies show that SOX9 and Sp1 physically interact in melanoma cells, while silencing of SOX9 down-regulates ETS1, but not Sp1, in the same cells. Finally, knockdown of SOX9 indeed renders melanoma cells resistant to T cell-mediated killing, in line with the increased CEACAM1 expression. In conclusion, we show that SOX9 regulates CEACAM1 expression in melanoma cells, and thereby their immune resistance. As CEACAM1 is a pivotal protein in melanoma biology and immune crosstalk, further understanding of its regulation can provide new insights and contribute to the development of novel approaches to therapy.


Asunto(s)
Inmunidad Adaptativa/genética , Antígenos CD/genética , Moléculas de Adhesión Celular/genética , Factor de Transcripción SOX9/genética , Antígenos CD/metabolismo , Sitios de Unión/genética , Western Blotting , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteína Proto-Oncogénica c-ets-1/metabolismo , Interferencia de ARN , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción Sp1/metabolismo
4.
Oncotarget ; 6(30): 28999-9015, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26338962

RESUMEN

The blossom of immunotherapy in melanoma highlights the need to delineate mechanisms of immune resistance. Recently, we have demonstrated that the RNA editing protein, adenosine deaminase acting on RNA-1 (ADAR1) is down-regulated during metastatic transition of melanoma, which enhances melanoma cell proliferation and tumorigenicity. Here we investigate the role of ADAR1 in melanoma immune resistance.Importantly, knockdown of ADAR1 in human melanoma cells induces resistance to tumor infiltrating lymphocytes in a cell contact-dependent mechanism. We show that ADAR1, in an editing-independent manner, regulates the biogenesis of miR-222 at the transcription level and thereby Intercellular Adhesion Molecule 1 (ICAM1) expression, which consequently affects melanoma immune resistance. ADAR1 thus has a novel, pivotal, role in cancer immune resistance. Corroborating with these results, the expression of miR-222 in melanoma tissue specimens was significantly higher in patients who had no clinical benefit from treatment with ipilimumab as compared to patients that responded clinically, suggesting that miR-222 could function as a biomarker for the prediction of response to ipilimumab.These results provide not only novel insights on melanoma immune resistance, but also pave the way to the development of innovative personalized tools to enable optimal drug selection and treatment.


Asunto(s)
Adenosina Desaminasa/metabolismo , Melanoma/enzimología , Proteínas de Unión al ARN/metabolismo , Neoplasias Cutáneas/enzimología , Adenosina Desaminasa/genética , Anticuerpos Monoclonales/uso terapéutico , Comunicación Celular , Línea Celular Tumoral , Resistencia a Antineoplásicos , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Ipilimumab , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , MicroARNs/genética , MicroARNs/metabolismo , Interferencia de ARN , Proteínas de Unión al ARN/genética , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Transcripción Genética , Transfección , Escape del Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA