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.
STAR Protoc ; 3(4): 101666, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36125932

RESUMEN

Tumor-associated macrophages (TAMs) are key contributors to antitumor immunity. Here, we present a protocol to drive human monocyte-macrophage differentiation using tumor-derived conditioned media, followed by phenotypic and functional characterization of TAMs in vitro. We describe CD14+ cell isolation from healthy human blood, and detail the procedure to induce macrophage polarization. Finally, we outline morphological assessment of macrophages, and validation of their functional behaviors with a tumor cell killing assay. This translatable-based approach can be applied to different cancer cell types. For complete details on the use and execution of this protocol, please refer to Georgouli et al. (2019).


Asunto(s)
Macrófagos , Monocitos , Humanos , Medios de Cultivo Condicionados/farmacología , Línea Celular Tumoral , Macrófagos/metabolismo , Separación Celular
3.
Cancer Cell ; 37(1): 85-103.e9, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31935375

RESUMEN

Despite substantial clinical benefit of targeted and immune checkpoint blockade-based therapies in melanoma, resistance inevitably develops. We show cytoskeletal remodeling and changes in expression and activity of ROCK-myosin II pathway during acquisition of resistance to MAPK inhibitors. MAPK regulates myosin II activity, but after initial therapy response, drug-resistant clones restore myosin II activity to increase survival. High ROCK-myosin II activity correlates with aggressiveness, identifying targeted therapy- and immunotherapy-resistant melanomas. Survival of resistant cells is myosin II dependent, regardless of the therapy. ROCK-myosin II ablation specifically kills resistant cells via intrinsic lethal reactive oxygen species and unresolved DNA damage and limits extrinsic myeloid and lymphoid immunosuppression. Efficacy of targeted therapies and immunotherapies can be improved by combination with ROCK inhibitors.


Asunto(s)
Citoesqueleto/metabolismo , Resistencia a Antineoplásicos , Melanoma/metabolismo , Miosina Tipo II/metabolismo , Animales , Antígeno B7-H1/metabolismo , Ciclo Celular , Línea Celular Tumoral , Daño del ADN , Femenino , Humanos , Inmunoterapia , Sistema de Señalización de MAP Quinasas , Masculino , Melanoma/inmunología , Melanoma/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Estrés Oxidativo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Especies Reactivas de Oxígeno , Linfocitos T Reguladores/inmunología , Resultado del Tratamiento , Microambiente Tumoral/inmunología , Quinasas Asociadas a rho/metabolismo
4.
Cell Rep ; 29(11): 3385-3393.e6, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31825823

RESUMEN

p21-Activated kinase 4 (PAK4), a serine/threonine kinase, is purported to localize to podosomes: transient adhesive structures that degrade the extracellular matrix to facilitate rapid myeloid cell migration. We find that treatment of transforming growth factor ß (TGF-ß)-differentiated monocytic (THP-1) cells with a PAK4-targeted inhibitor significantly reduces podosome formation and induces the formation of focal adhesions. This switch in adhesions confers a diminution of matrix degradation and reduced cell migration. Furthermore, reduced PAK4 expression causes a significant reduction in podosome number that cannot be rescued by kinase-dead PAK4, supporting a kinase-dependent role. Concomitant with PAK4 depletion, phosphorylation of Akt is perturbed, whereas a specific phospho-Akt signal is detected within the podosomes. Using superresolution analysis, we find that PAK4 specifically localizes in the podosome ring, nearer to the actin core than other ring proteins. We propose PAK4 kinase activity intersects with the Akt pathway at the podosome ring:core interface to drive regulation of macrophage podosome turnover.


Asunto(s)
Células Mieloides/metabolismo , Podosomas/metabolismo , Quinasas p21 Activadas/metabolismo , Células Cultivadas , Disulfuros/farmacología , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Células HEK293 , Humanos , Células Mieloides/efectos de los fármacos , Células Mieloides/ultraestructura , Naftoles/farmacología , Fosforilación , Podosomas/ultraestructura , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células THP-1 , Quinasas p21 Activadas/antagonistas & inhibidores
5.
F1000Res ; 82019.
Artículo en Inglés | MEDLINE | ID: mdl-31824652

RESUMEN

Image analysis in clinical research has evolved at fast pace in the last decade. This review discusses basic concepts ranging from immunohistochemistry to advanced techniques such as multiplex imaging, digital pathology, flow cytometry and intravital microscopy. Tissue imaging ex vivo is still one of the gold-standards in the field due to feasibility. We describe here different protocols and applications of digital analysis providing basic and clinical researchers with an overview on how to analyse tissue images. In vivo imaging is not accessible to researchers; however, it provides invaluable dynamic information easily. Overall, we discuss a plethora of techniques that - when combined - constitute a powerful platform for basic and translational cancer research.


Asunto(s)
Neoplasias , Humanos , Inmunohistoquímica , Microscopía Intravital , Investigación
6.
Cell ; 176(4): 757-774.e23, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30712866

RESUMEN

ROCK-Myosin II drives fast rounded-amoeboid migration in cancer cells during metastatic dissemination. Analysis of human melanoma biopsies revealed that amoeboid melanoma cells with high Myosin II activity are predominant in the invasive fronts of primary tumors in proximity to CD206+CD163+ tumor-associated macrophages and vessels. Proteomic analysis shows that ROCK-Myosin II activity in amoeboid cancer cells controls an immunomodulatory secretome, enabling the recruitment of monocytes and their differentiation into tumor-promoting macrophages. Both amoeboid cancer cells and their associated macrophages support an abnormal vasculature, which ultimately facilitates tumor progression. Mechanistically, amoeboid cancer cells perpetuate their behavior via ROCK-Myosin II-driven IL-1α secretion and NF-κB activation. Using an array of tumor models, we show that high Myosin II activity in tumor cells reprograms the innate immune microenvironment to support tumor growth. We describe an unexpected role for Myosin II dynamics in cancer cells controlling myeloid function via secreted factors.


Asunto(s)
Movimiento Celular/fisiología , Miosina Tipo II/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular/inmunología , Proteínas del Citoesqueleto , Femenino , Humanos , Interleucina-1alfa/metabolismo , Masculino , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Persona de Mediana Edad , FN-kappa B/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Fosforilación , Proteómica , Receptor Cross-Talk/fisiología , Transducción de Señal , Microambiente Tumoral/inmunología
7.
Nat Commun ; 9(1): 2951, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-30054470

RESUMEN

Tumour-associated macrophages (TAMs) play an important role in tumour progression, which is facilitated by their ability to respond to environmental cues. Here we report, using murine models of breast cancer, that TAMs expressing fibroblast activation protein alpha (FAP) and haem oxygenase-1 (HO-1), which are also found in human breast cancer, represent a macrophage phenotype similar to that observed during the wound healing response. Importantly, the expression of a wound-like cytokine response within the tumour is clinically associated with poor prognosis in a variety of cancers. We show that co-expression of FAP and HO-1 in macrophages results from an innate early regenerative response driven by IL-6, which both directly regulates HO-1 expression and licenses FAP expression in a skin-like collagen-rich environment. We show that tumours can exploit this response to facilitate transendothelial migration and metastatic spread of the disease, which can be pharmacologically targeted using a clinically relevant HO-1 inhibitor.


Asunto(s)
Neoplasias de la Mama/metabolismo , Macrófagos/metabolismo , Metástasis de la Neoplasia , Cicatrización de Heridas/fisiología , Animales , Línea Celular Tumoral , Supervivencia Celular , Colágeno/metabolismo , Citocinas/metabolismo , Endopeptidasas , Femenino , Gelatinasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Humanos , Interleucina-6/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Fenotipo , Pronóstico , Serina Endopeptidasas/metabolismo , Piel/metabolismo , Microambiente Tumoral/fisiología
8.
PLoS One ; 12(4): e0175712, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28410380

RESUMEN

Dendritic cells (DC) have the potential to instigate a tumour-specific immune response, but their ability to prime naïve lymphocytes depends on their activation status. Thus, for tumour immunotherapy to be effective, the provision of appropriate DC activation stimuli such as Toll-like receptor (TLR) agonists is crucial in order to overcome immunosuppression associated with the tumour microenvironment. To address this, we investigated how ovarian carcinoma (OC)-associated ascites impedes activation of DC by TLR agonists. Our results show that ascites reduces the TLR-mediated up-regulation of CD86 and partially inhibits the production of the pro-inflammatory cytokines interleukin 6 (IL-6), IL-12 and tumour necrosis factor α (TNFα) in monocyte-derived DC from healthy controls. We further observe an impaired T cell stimulatory capacity of DC upon activation with TLR agonists in the presence of ascites, indicating that their functionality is affected by the immunosuppressive factors. We identify IL-10 and prostaglandin E2 (PGE2) as the pivotal immunosuppressive components in OC-associated ascites compromising TLR-mediated DC activation. Interestingly, IL-10 is present in both ascites from patients with malignant OC and in peritoneal fluid from patients with benign ovarian conditions and both fluids have similar ability to reduce TLR-mediated DC activation. However, depletion of IL-10 from ascites revealed that the presence of paracrine IL-10 is not crucial for ascites-mediated suppression of DC activation in response to TLR activation. Unlike IL-10, PGE2 is absent from peritoneal fluid of patients with benign conditions and selectively reduces TNFα induction in response to TLR-mediated activation in the presence of OC-associated ascites. Our study highlights PGE2 as an immunosuppressive component of the malignant OC microenvironment rendering PGE2 a potentially important target for immunotherapy in OC.


Asunto(s)
Dinoprostona/metabolismo , Interleucina-10/metabolismo , Neoplasias Ováricas/patología , Receptores Toll-Like/metabolismo , Anticuerpos Neutralizantes/metabolismo , Ascitis/metabolismo , Antígeno B7-2/metabolismo , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Dinoprostona/inmunología , Femenino , Humanos , Imidazoles/toxicidad , Interleucina-10/inmunología , Interleucina-12/análisis , Interleucina-12/metabolismo , Interleucina-6/análisis , Interleucina-6/metabolismo , Lipopolisacáridos/toxicidad , Activación de Linfocitos/efectos de los fármacos , Monocitos/citología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Poli I-C/toxicidad , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Receptores Toll-Like/agonistas , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
9.
Cancer Res ; 77(5): 1127-1141, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28096174

RESUMEN

IgE antibodies are key mediators of antiparasitic immune responses, but their potential for cancer treatment via antibody-dependent cell-mediated cytotoxicity (ADCC) has been little studied. Recently, tumor antigen-specific IgEs were reported to restrict cancer cell growth by engaging high-affinity Fc receptors on monocytes and macrophages; however, the underlying therapeutic mechanisms were undefined and in vivo proof of concept was limited. Here, an immunocompetent rat model was designed to recapitulate the human IgE-Fcε receptor system for cancer studies. We also generated rat IgE and IgG mAbs specific for the folate receptor (FRα), which is expressed widely on human ovarian tumors, along with a syngeneic rat tumor model expressing human FRα. Compared with IgG, anti-FRα IgE reduced lung metastases. This effect was associated with increased intratumoral infiltration by TNFα+ and CD80+ macrophages plus elevated TNFα and the macrophage chemoattractant MCP-1 in lung bronchoalveolar lavage fluid. Increased levels of TNFα and MCP-1 correlated with IgE-mediated tumor cytotoxicity by human monocytes and with longer patient survival in clinical specimens of ovarian cancer. Monocytes responded to IgE but not IgG exposure by upregulating TNFα, which in turn induced MCP-1 production by monocytes and tumor cells to promote a monocyte chemotactic response. Conversely, blocking TNFα receptor signaling abrogated induction of MCP-1, implicating it in the antitumor effects of IgE. Overall, these findings show how antitumor IgE reprograms monocytes and macrophages in the tumor microenvironment, encouraging the clinical use of IgE antibody technology to attack cancer beyond the present exclusive reliance on IgG. Cancer Res; 77(5); 1127-41. ©2017 AACR.


Asunto(s)
Anticuerpos Antiidiotipos/inmunología , Receptor 1 de Folato/inmunología , Macrófagos/inmunología , Neoplasias Ováricas/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Línea Celular Tumoral , Femenino , Receptor 1 de Folato/antagonistas & inhibidores , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Ratas , Ratas Wistar , Transducción de Señal , Factor de Necrosis Tumoral alfa/biosíntesis
10.
J Autoimmun ; 79: 53-62, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28118944

RESUMEN

Monocytes and macrophages are key mediators of inflammation in rheumatoid arthritis (RA). Their persistence at the inflammatory site is likely to contribute to immunopathology. We sought to characterise one mechanism by which persistence may be achieved: resistance to apoptosis and the role of mir-155 in this process. CD14+ monocytes from peripheral blood (PBM) and synovial fluid (SFM) of RA patients were found to be resistant to spontaneous apoptosis relative to PBM from healthy control (HC) individuals. RA SFM were also resistant to anti-Fas-mediated apoptosis and displayed a gene expression profile distinct from HC and RA PBM populations. Gene expression profiling analysis revealed that the differentially expressed genes in RA SFM vs. PBM were enriched for apoptosis-related genes and showed increased expression of the mir-155 precursor BIC. Following identification of potential mir-155 target transcripts by bioinformatic methods, we show increased levels of mature mir-155 expression in RA PBM and SFM vs. HC PBM and a corresponding decrease in SFM of two predicted mir-155-target mRNAs, apoptosis mediators CASP10 and APAF1. Using miR mimics, we demonstrate that mir-155 over-expression in healthy CD14+ cells conferred resistance to spontaneous apoptosis, but not Fas-induced death in these cells, and resulted in increased production of cytokines and chemokines. Collectively our data indicate that CD14+ cells from patients with RA show enhanced resistance to apoptosis, and suggest that an increase in mir-155 may partially contribute to this phenotype.


Asunto(s)
Apoptosis/genética , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , MicroARNs/genética , Monocitos/inmunología , Monocitos/metabolismo , Artritis Reumatoide/metabolismo , Biomarcadores , Estudios de Casos y Controles , Supervivencia Celular/genética , Biología Computacional/métodos , Citocinas/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Inmunofenotipificación , Mediadores de Inflamación/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Fenotipo , Interferencia de ARN , Receptor fas/metabolismo
11.
Semin Cancer Biol ; 42: 60-69, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27586372

RESUMEN

The Transforming Growth Factor-beta (TGFß) pathway mediates a broad spectrum of cellular processes and is involved in several diseases, including cancer. TGFß has a dual role in tumours, acting as a tumour suppressor in the early phase of tumorigenesis and as a tumour promoter in more advanced stages. In this review, we discuss the effects of TGFß-driven transcription on all stages of tumour progression, with special focus on lung cancer. Since some TGFß target genes are specifically involved in promoting metastasis, we speculate that these genes might be good targets to block tumour progression without compromising the tumour suppressor effects of the TGFß pathway.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias/genética , Transcripción Genética , Factor de Crecimiento Transformador beta/genética , Genes Supresores de Tumor , Humanos , Estadificación de Neoplasias , Neoplasias/patología , Transducción de Señal/genética
12.
Biol Chem ; 397(6): 519-28, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26866879

RESUMEN

Invariant chain (Ii) or CD74 is a non-polymorphic glycoprotein, which apart from its role as a chaperone dedicated to MHCII molecules, is known to be a high-affinity receptor for macrophage migration inhibitory factor (MIF). The present study aimed to define the roles of CD74 and MIF in the immune surveillance escape process. Towards this direction, the cell lines HL-60, Raji, K562 and primary pre-B leukemic cells were examined for expression and secretion of MIF. Flow cytometry analysis detected high levels of MIF and intracellular/membrane CD74 expression in all leukemic cells tested, while MIF secretion was shown to be inversely proportional to intracellular HLA-DR (DR) expression. In the MHCII-negative cells, IFN-γ increased MIF expression and induced its secretion in HL-60 and K562 cells, respectively. In K562 cells, CD74 (Iip33Iip35) was shown to co-precipitate with HLA-DOß (DOß), inhibiting thus MIF or DR binding. Induced expression of DOα in K562 (DOα-DOß+) cells in different transfection combinations decreased MIF expression and secretion, while increasing surface DR expression. Thus, MIF could indeed be part of the antigen presentation process.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/metabolismo , Regulación Neoplásica de la Expresión Génica , Antígenos HLA-DR/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Leucemia/patología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/farmacología
13.
Immunobiology ; 218(8): 1019-25, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23462321

RESUMEN

The expression of DOß and not DOα, in addition to the high intracellular DR, low DM levels and absence of surface DR expression in K562 and HL-60 cells introduce alternative regulatory pathways in DR trafficking and consequently the antigen presentation process. The present study attempted to define the naturally occurring DOα negative state and explain the role of DOß in the intracellular DR accumulation in K562 and HL-60 cells. Despite the absence of DOα, the DOß chain was detected in the endosomal compartments. The lack of DOα was found to be partially responsible for the absence of DR from the cell membrane since stable K562-DOα transfectants allowed expression of membrane DR. This expression could be significantly increased upon DM induction by IFN-γ, indicating that DM was another limiting factor for the migration of DR to the cell surface of K562 and HL-60 cells. Furthermore, intracellular DR co-localized with the exosome specific marker CD9, while culture supernatants were shown to contain exosome-engaged and exosome free DR activity as evaluated by SDS-page followed by western blot, ELISA and transmission electron microscopy analysis. These findings indicated that in DOα⁻ß⁺ cells, DR molecules were programmed to secretion rather than surface expression. The presented results provide novel regulatory processes as to DR trafficking, avoiding expression to the cell surface.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos HLA-D/inmunología , Antígenos HLA-DR/inmunología , Transporte de Proteínas/inmunología , Línea Celular Tumoral , Exosomas , Células HL-60 , Antígenos HLA-D/biosíntesis , Antígenos HLA-D/metabolismo , Humanos , Interferón gamma , Células K562
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...