Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nat Immunol ; 15(8): 749-757, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24973821

RESUMEN

Interleukin 15 (IL-15) controls both the homeostasis and the peripheral activation of natural killer (NK) cells. The molecular basis for this duality of action remains unknown. Here we found that the metabolic checkpoint kinase mTOR was activated and boosted bioenergetic metabolism after exposure of NK cells to high concentrations of IL-15, whereas low doses of IL-15 triggered only phosphorylation of the transcription factor STAT5. mTOR stimulated the growth and nutrient uptake of NK cells and positively fed back on the receptor for IL-15. This process was essential for sustaining NK cell proliferation during development and the acquisition of cytolytic potential during inflammation or viral infection. The mTORC1 inhibitor rapamycin inhibited NK cell cytotoxicity both in mice and humans; this probably contributes to the immunosuppressive activity of this drug in different clinical settings.


Asunto(s)
Interleucina-15/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Serina-Treonina Quinasas TOR/inmunología , Animales , Proliferación Celular , Células Cultivadas , Infecciones por Herpesviridae/inmunología , Humanos , Inmunosupresores/farmacología , Inflamación/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Células Asesinas Naturales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Muromegalovirus/inmunología , Infecciones por Orthomyxoviridae/inmunología , Poli I-C/inmunología , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/inmunología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética
2.
EMBO J ; 40(9): e108164, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33880795

RESUMEN

Cellular senescence is considered to be a major driver of aging, yet the mechanisms explaining the accumulation of senescent cells during life time remain unclear. In this issue, Lagnado et al (2021) show that neutrophils can trigger the senescence of neighboring cells by transmitting reactive oxygen species (ROS), which they normally produce to fight pathogens. The main genomic targets of the neutrophil-mediated ROS damage are telomeres, supporting an intimate interplay between telomere homeostasis and oxidative stress in senescence and consequently aging.


Asunto(s)
Senescencia Celular , Neutrófilos , Senescencia Celular/genética , Estrés Oxidativo , Especies Reactivas de Oxígeno , Telómero , Homeostasis del Telómero
3.
Mol Cell ; 61(2): 274-86, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26774283

RESUMEN

The shelterin proteins protect telomeres against activation of the DNA damage checkpoints and recombinational repair. We show here that a dimer of the shelterin subunit TRF2 wraps ∼ 90 bp of DNA through several lysine and arginine residues localized around its homodimerization domain. The expression of a wrapping-deficient TRF2 mutant, named Top-less, alters telomeric DNA topology, decreases the number of terminal loops (t-loops), and triggers the ATM checkpoint, while still protecting telomeres against non-homologous end joining (NHEJ). In Top-less cells, the protection against NHEJ is alleviated if the expression of the TRF2-interacting protein RAP1 is reduced. We conclude that a distinctive topological state of telomeric DNA, controlled by the TRF2-dependent DNA wrapping and linked to t-loop formation, inhibits both ATM activation and NHEJ. The presence of RAP1 at telomeres appears as a backup mechanism to prevent NHEJ when topology-mediated telomere protection is impaired.


Asunto(s)
ADN/química , Conformación de Ácido Nucleico , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Emparejamiento Base , ADN/metabolismo , Daño del ADN , Reparación del ADN por Unión de Extremidades , Células HeLa , Humanos , Lisina/metabolismo , Modelos Moleculares , Mutación , Estructura Terciaria de Proteína , Complejo Shelterina , Transducción de Señal , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/química
4.
EMBO J ; 38(11)2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31000523

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells with strong immunosuppressive activity that promote tumor growth. In this study, we describe a mechanism by which cancer cells control MDSCs in human cancers by upregulating TRF2, a protein required for telomere stability. Specifically, we showed that the TRF2 upregulation in cancer cells has extratelomeric roles in activating the expression of a network of genes involved in the biosynthesis of heparan sulfate proteoglycan, leading to profound changes in glycocalyx length and stiffness, as revealed by atomic force microscopy. This TRF2-dependent regulation facilitated the recruitment of MDSCs, their activation via the TLR2/MyD88/IL-6/STAT3 pathway leading to the inhibition of natural killer recruitment and cytotoxicity, and ultimately tumor progression and metastasis. The clinical relevance of these findings is supported by our analysis of cancer cohorts, which showed a correlation between high TRF2 expression and MDSC infiltration, which was inversely correlated with overall patient survival.


Asunto(s)
Glicocálix/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Proteína 2 de Unión a Repeticiones Teloméricas/fisiología , Escape del Tumor/fisiología , Animales , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica , Glicocálix/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Células 3T3 NIH , Neoplasias/genética , Neoplasias/mortalidad , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Escape del Tumor/genética
5.
Nucleic Acids Res ; 47(7): 3365-3382, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30698737

RESUMEN

The telomeric protein TRF2 is overexpressed in several human malignancies and contributes to tumorigenesis even though the molecular mechanism is not completely understood. By using a high-throughput approach based on the multiplexed Luminex X-MAP technology, we demonstrated that TRF2 dramatically affects VEGF-A level in the secretome of cancer cells, promoting endothelial cell-differentiation and angiogenesis. The pro-angiogenic effect of TRF2 is independent from its role in telomere capping. Instead, TRF2 binding to a distal regulatory element promotes the expression of SULF2, an endoglucosamine-6-sulfatase that impairs the VEGF-A association to the plasma membrane by inducing post-synthetic modification of heparan sulfate proteoglycans (HSPGs). Finally, we addressed the clinical relevance of our findings showing that TRF2/SULF2 expression is a worse prognostic biomarker in colorectal cancer (CRC) patients.


Asunto(s)
Neoplasias del Colon/metabolismo , Sulfotransferasas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/patología , Proteoglicanos de Heparán Sulfato/química , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Neovascularización Patológica , Sulfatasas , Sulfotransferasas/biosíntesis , Proteína 2 de Unión a Repeticiones Teloméricas/deficiencia , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Front Immunol ; 15: 1382236, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38571942

RESUMEN

Immune checkpoint therapies (ICT) have transformed the treatment of cancer over the past decade. However, many patients do not respond or suffer relapses. Successful immunotherapy requires epitope spreading, but the slow or inefficient induction of functional antitumoral immunity delays the benefit to patients or causes resistances. Therefore, understanding the key mechanisms that support epitope spreading is essential to improve immunotherapy. In this review, we highlight the major role played by B-cells in breaking immune tolerance by epitope spreading. Activated B-cells are key Antigen-Presenting Cells (APC) that diversify the T-cell response against self-antigens, such as ribonucleoproteins, in autoimmunity but also during successful cancer immunotherapy. This has important implications for the design of future cancer vaccines.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Epítopos , Autoantígenos , Autoinmunidad , Inmunoterapia , Neoplasias/terapia
7.
Cancer Cell ; 41(4): 757-775.e10, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-37037615

RESUMEN

Metastasis is the major cause of cancer death, and the development of therapy resistance is common. The tumor microenvironment can confer chemotherapy resistance (chemoresistance), but little is known about how specific host cells influence therapy outcome. We show that chemotherapy induces neutrophil recruitment and neutrophil extracellular trap (NET) formation, which reduces therapy response in mouse models of breast cancer lung metastasis. We reveal that chemotherapy-treated cancer cells secrete IL-1ß, which in turn triggers NET formation. Two NET-associated proteins are required to induce chemoresistance: integrin-αvß1, which traps latent TGF-ß, and matrix metalloproteinase 9, which cleaves and activates the trapped latent TGF-ß. TGF-ß activation causes cancer cells to undergo epithelial-to-mesenchymal transition and correlates with chemoresistance. Our work demonstrates that NETs regulate the activities of neighboring cells by trapping and activating cytokines and suggests that chemoresistance in the metastatic setting can be reduced or prevented by targeting the IL-1ß-NET-TGF-ß axis.


Asunto(s)
Neoplasias de la Mama , Resistencia a Antineoplásicos , Trampas Extracelulares , Neoplasias Pulmonares , Neutrófilos , Microambiente Tumoral , Neutrófilos/metabolismo , Neutrófilos/patología , Humanos , Animales , Ratones , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Metástasis de la Neoplasia , Trampas Extracelulares/metabolismo , Inflamación/patología
8.
Cancer Metastasis Rev ; 30(1): 13-25, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21271351

RESUMEN

Cancer initiation, progression, and invasion occur in a complex and dynamic microenvironment which depends on the hosts and sites where tumors develop. Tumors arising in mucosal tissues may progress in an inflammatory context linked to local viral and/or bacterial infections. At the opposite, tumors developing in immunoprivileged sites are protected from microorganisms and grow in an immunosuppressive environment. In the present review, we summarize and present our recent data on the influence of infectious context and immune cell infiltration organization in human Non-Small Cell Lung Cancers (NSCLC) progression. We show that stimulation of tumor cells by TLR for viral ssRNA, such as TLR7/8, or bacteria, such as TLR4, promotes cell survival and induces chemoresistance. On the opposite, stimulation by TLR3, receptor for double-stranded viral RNA, decreases tumor cell viability and induces chemosensitivity in some lung tumor cell lines. Since fresh lung tumor cells exhibit a gene expression profile characteristic of TLR-stimulated lung tumor cell lines, we suspect that viral and bacterial influence may not only act on the host immune system but also directly on tumor growth and sensitivity to chemotherapy. The stroma of NSCLC contains tertiary lymphoid structures (or Tumor-induced Bronchus-Associated Lymphoid Tissues (Ti-BALT)) with mature DC, follicular DC, and T and B cells. Two subsets of immature DC, Langerhans cells (LC) and interstitial DC (intDC), were detected in the tumor nests and the stroma, respectively. Here, we show that the densities of the three DC subsets, mature DC, LC, and intDC, are highly predictive of disease-specific survival in a series of 74 early-stage NSCLC patients. We hypothesize that the mature DC may derive from local activation and migration of the immature DC--and especially LC which contact the tumor cells--to the tertiary lymphoid structures, after sampling and processing of the tumor antigens. In view of the prominent role of DC in the immune response, we suggest that the microenvironment of early-stage NSCLC may allow the in situ activation of the adaptive response. Finally, we find that the eyes or brain of mice with growing B cell lymphoma are infiltrated with T cells and that the cytokines produced ex vivo by the tumoral tissues have an impaired Th1 cytokine profile. Our work illustrates that the host and external tumor microenvironments are multifaceted and strongly influence tumor progression and anti-tumor immune responses.


Asunto(s)
Microambiente Tumoral , Animales , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Células Dendríticas/inmunología , Humanos , Linfoma/inmunología , Linfoma/patología , Receptores Toll-Like/metabolismo
9.
Cancers (Basel) ; 13(12)2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203903

RESUMEN

Telomeric repeat-binding factor 2 (TRF2) is a subunit of the shelterin protein complex, which binds to and protects telomeres from unwanted DNA damage response (DDR) activation. TRF2 expression plays a pivotal role in aging and cancer, being downregulated during cellular senescence and overexpressed during oncogenesis. Cancers overexpressing TRF2 often exhibit a poor prognosis. In cancer cells, TRF2 plays multiple functions, including telomere protection and non-cell autonomous roles, promoting neo-angiogenesis and immunosuppression. We present here an original screening strategy, which enables identification of small molecules that decrease or increase TRF2 expression. By screening a small library of Food and Drug Agency (FDA)-approved drugs, we identified two molecules (AR-A014418 and alexidine·2HCl) that impaired tumor growth, neo-angiogenesis and immunosuppression by downregulating TRF2 expression in a mouse xenograft model. These results support the chemotherapeutic strategy of downregulating TRF2 expression to treat aggressive human tumors and validate this cell-based assay capable of screening for potential anti-cancer and anti-aging molecules by modulating TRF2 expression levels.

10.
Oncoimmunology ; 10(1): 1901446, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33796413

RESUMEN

The outcome of patients with cutaneous melanoma has been strongly modified by recent advances obtained with Immune Checkpoint Inhibitors (ICIs). However, despite this breakthrough, durable response to ICIs is limited to a subset of patients. We investigated whether the expression of TRF2, which preserves telomere integrity, and have an effect on tumor immunosurveillance notably by directly recruiting and activating myeloid-derived suppressor cells (MDSCs), could be a prognostic biomarker in patients with relapsed or metastatic melanoma based on different treatment regimens. We evaluated retrospectively the association of TRF2 expressed in melanoma cells in combination with intratumoral CD33+ CD15+ CD14- MDSCs, as detected by immunohistochemistry and quantified by digital analysis, to clinicopathological features and overall survival (OS) among 48 patients treated with ICIs and 77 patients treated with other treatment options. The densities/mm2 of TRF2+ cells (P=.003) and CD33+ cells (P=.004) were individually significantly related to poor OS. In addition, only the combined expression of CD33+/CD15+/CD14- cells/mm2 was significantly correlated to poor OS (P=.017) in the whole study population as well as in patients treated by ICIs (P=.023). There was no significant difference in OS when analyzing the other markers individually or in combination according to the treatment regimen. The pre-treatment assessment of TRF2 expression and CD33+ cells/mm2 along with the density of CD33+/CD15+/CD14- cells/mm2 could assess OS and better predict clinical response of patients with melanoma treated by ICIs.


Asunto(s)
Melanoma , Células Supresoras de Origen Mieloide , Neoplasias Cutáneas , Humanos , Inmunohistoquímica , Melanoma/tratamiento farmacológico , Estudios Retrospectivos , Neoplasias Cutáneas/tratamiento farmacológico
11.
Nat Commun ; 12(1): 653, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33510147

RESUMEN

Only a subpopulation of non-small cell lung cancer (NSCLC) patients responds to immunotherapies, highlighting the urgent need to develop therapeutic strategies to improve patient outcome. We develop a chemical positive modulator (HEI3090) of the purinergic P2RX7 receptor that potentiates αPD-1 treatment to effectively control the growth of lung tumors in transplantable and oncogene-induced mouse models and triggers long lasting antitumor immune responses. Mechanistically, the molecule stimulates dendritic P2RX7-expressing cells to generate IL-18 which leads to the production of IFN-γ by Natural Killer and CD4+ T cells within tumors. Combined with immune checkpoint inhibitor, the molecule induces a complete tumor regression in 80% of LLC tumor-bearing mice. Cured mice are also protected against tumor re-challenge due to a CD8-dependent protective response. Hence, combination treatment of small-molecule P2RX7 activator followed by immune checkpoint inhibitor represents a strategy that may be active against NSCLC.


Asunto(s)
Carcinoma Pulmonar de Lewis/terapia , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Receptores Purinérgicos P2X7/inmunología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/inmunología , Línea Celular Tumoral , Terapia Combinada , Femenino , Células HEK293 , Humanos , Inhibidores de Puntos de Control Inmunológico/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-18/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Estructura Molecular , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
12.
Med Sci (Paris) ; 36(12): 1113-1117, 2020 Dec.
Artículo en Francés | MEDLINE | ID: mdl-33296627

RESUMEN

Aging is an alteration of our physiological capacities that is accompanied by an increased susceptibility to develop a wide range of diseases and which determines in large part our longevity. Despite intensive research on the origin of aging, its etiology is still poorly understood. We discuss here the hypothesis that the telomere shortening, programmed to start at the end of embryogenesis in numerous tissues, couples development with aging by a time-dependent regulation of a set of interconnected processes essential for the somatic maintenance of genome, epigenome, metabolism, circadian clock and immunity.


TITLE: Les horloges de la longévité - Le rôle promoteur des télomères ? ABSTRACT: Le vieillissement est une altération de nos capacités physiologiques qui s'accompagne d'une susceptibilité accrue à un grand nombre de maladies et qui détermine en grande partie notre longévité. Cependant, son étiologie reste encore mal comprise. Nous discutons ici l'hypothèse que le raccourcissement des télomères, programmé pour débuter en fin d'embryogenèse dans de nombreux tissus, couple développement et vieillissement. Il existe en effet de nombreuses indications que des variations de la structure des télomères régulent dans le temps un ensemble interconnecté de processus essentiels à la maintenance somatique du génome, de l'épigénome, du métabolisme, du rythme circadien et de l'immunité.


Asunto(s)
Relojes Circadianos/fisiología , Longevidad/genética , Telómero/fisiología , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Senescencia Celular/genética , Relojes Circadianos/genética , Humanos
13.
EMBO Mol Med ; 11(7): e10845, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31273935

RESUMEN

In the context of tumorigenesis, telomere shortening is associated with apparent antagonistic outcomes: On one side, it favors cancer initiation through mechanisms involving genome instability, while on the other side, it prevents cancer progression, due to the activation of the DNA damage response (DDR) checkpoint behaving as a cell-intrinsic proliferation barrier. Consequently, telomerase, which can compensate for replicative erosion by adding telomeric DNA repeats at the chromosomal DNA extremities, is crucial for cancer progression and is upregulated in nearly 90% of human cancers. Therefore, telomeres are considered potential anti-cancer targets and, to date, most of the studies have focused on telomerase inhibition. However, the development of clinically efficient telomerase targeting therapies is still in its infancy. In this context, the findings reported in this issue of EMBO Molecular Medicine by Bejarano et al (2019) open new avenues for alternative telomere therapies.


Asunto(s)
Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Telómero/metabolismo , Proteína 1 de Unión a Repeticiones Teloméricas/antagonistas & inhibidores , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Transducción de Señal/genética , Telómero/genética , Telómero/patología , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo
14.
Immunology ; 124(4): 562-74, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18284467

RESUMEN

Tumour necrosis factor receptor associated factor 4 (TRAF4) is a member of the TRAF family of proteins which are cytoplasmic adaptor molecules strongly implicated in multiple immune functions. A previous investigation of TRAF4 biological functions by gene targeting in mice has shown a role for TRAF4 in embryonic development and neurulation in vivo. However, unlike other TRAF family members, the role of TRAF4 in the immune system is still unknown. To address this question, we performed an extensive characterization of the immune development and immune functions of TRAF4-deficient mice. Our analyses did not reveal any defects in development of T and B lymphocytes, granulocytes, macrophages and dendritic cells, and no defects in reactive oxygen species production and phagocytosis by neutrophils. Cellular and humoral responses against T-cell-dependent antigens were normal, as was dendritic cell maturation in response to microbial components and antigen uptake by dendritic cells. However, we demonstrated that dendritic cells from TRAF4-deficient mice exhibited reduced migration both in transwell experiments and in vivo. These results suggest that TRAF4 is not strictly required for immune development and functions but could participate in immune functions by facilitating immune cell migration.


Asunto(s)
Células Dendríticas/inmunología , Factor 4 Asociado a Receptor de TNF/inmunología , Animales , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Proliferación Celular , Células Cultivadas , Citocinas/biosíntesis , Sistema Inmunológico/crecimiento & desarrollo , Inmunidad Celular , Inmunoglobulina G/biosíntesis , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Ovalbúmina/inmunología , Toxinas Shiga/inmunología , Subgrupos de Linfocitos T/inmunología , Factor 4 Asociado a Receptor de TNF/deficiencia
15.
PLoS One ; 11(11): e0165639, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27812136

RESUMEN

γδ T cells play critical roles in host defense against infections and cancer. Although advances have been made in identifying γδ TCR ligands, it remains essential to understand molecular mechanisms responsible for in vivo expansion of γδ T cells in periphery. Recent findings identified the expression of the inducible NO synthase (NOS2) in lymphoid cells and highlighted novel immunoregulatory functions of NOS2 in αß T cell differentiation and B cell survival. In this context, we wondered whether NOS2 exerts an impact on γδ T cell properties. Here, we show that γδ T cells express NOS2 not only in vitro after TCR triggering, but also directly ex vivo. Nos2 deficient mice have fewer γδ T cells in peripheral lymph nodes (pLNs) than their wild-type counterparts, and these cells exhibit a reduced ability to produce IL-2. Using chemical NOS inhibitors and Nos2 deficient γδ T cells, we further evidence that the inactivation of endogenous NOS2 significantly reduced γδ T cell proliferation and glycolysis metabolism that can be restored in presence of exogenous IL-2. Collectively, we demonstrate the crucial role of endogenous NOS2 in promoting optimal IL-2 production, proliferation and glycolysis of γδ T cells that may contribute to their regulation at steady state.


Asunto(s)
Proliferación Celular/genética , Glucólisis/genética , Interleucina-2/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Subgrupos de Linfocitos T/metabolismo , Animales , Proliferación Celular/fisiología , Células Cultivadas , Glucólisis/fisiología , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo
17.
Cancer Res ; 75(5): 835-45, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25564520

RESUMEN

Colitis-associated cancer (CAC) is a complication of inflammatory bowel disease (IBD). Binding of extracellular ATP to the purinergic receptor P2RX7 has emerged as a critical event in controlling intestinal inflammation, acting to limit elevation of proinflammatory mast cells and cytokines and promote survival of regulatory T cells (Treg) and enteric neurons. In this study, we investigated the effect of P2RX7 blockade in an established mouse model of CAC. Using genetic and pharmacologic tools, we found unexpectedly that while P2RX7 mediated inflammatory responses, it also acted at an early time to suppress CAC development. P2RX7 blockade enhanced proliferation of intestinal epithelial cells and protected them from apoptosis. The proliferative effects of P2RX7 blockade were associated with an increased production of TGFß1 that was sufficient to stimulate the proliferation of intestinal epithelial cells. Finally, P2RX7 blockade also altered immune cell infiltration and promoted Treg accumulation within lesions of the digestive system. Taken together, our findings reveal an unexpected role for P2RX7 in preventing CAC, suggesting cautions in the use of P2RX7 inhibitors to treat IBD given the possibility of increasing risks CAC as a result.


Asunto(s)
Colitis/metabolismo , Colitis/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/metabolismo , Animales , Colitis/inmunología , Neoplasias del Colon/inmunología , Modelos Animales de Enfermedad , Inmunohistoquímica , Incidencia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Purinérgicos P2X7/genética , Transducción de Señal , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/biosíntesis
18.
Oncoimmunology ; 3(1): e27358, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24701373

RESUMEN

Here, we describe a model in which telomeric repeat-binding factor 2 (TERF2) can control tumorigenesis not only via cancer cell-intrinsic mechanisms but also via non-cancer cell autonomous pathways. Indeed, we have recently shown that TERF2 regulates tissue homeostasis as it promotes the elimination of aged, damaged, and neoplastic cells by the immune system, opening the way to new therapeutic options against cancer.

19.
Nat Commun ; 5: 5852, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25510679

RESUMEN

Angiogenesis, activation of metastasis and avoidance of immune destruction are important for cancer progression. These biological capabilities are, apart from cancer cells, mediated by different cell types, including endothelial, haematopoietic progenitor and myeloid-derived suppressor cells. We show here that all these cell types frequently express the Wilms' tumour suppressor Wt1, which transcriptionally controls expression of Pecam-1 (CD31) and c-kit (CD117). Inducible conditional knockout of Wt1 in endothelial, haematopoietic and myeloid-derived suppressor cells is sufficient to cause regression of tumour vascularization and an enhanced immune response, leading to decreased metastasis, regression of established tumours and enhanced survival. Thus, Wt1 is an important regulator of cancer growth via modulation of tumour vascularization, immune response and metastasis formation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma Experimental/genética , Neovascularización Patológica/genética , Neoplasias Cutáneas/genética , Proteínas WT1/genética , Animales , Progresión de la Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/patología , Femenino , Técnicas de Inactivación de Genes , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Humanos , Inmunidad Innata , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/inmunología , Melanoma Experimental/mortalidad , Ratones , Ratones Transgénicos , Células Mieloides/inmunología , Células Mieloides/patología , Trasplante de Neoplasias , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Análisis de Supervivencia , Carga Tumoral , Células Tumorales Cultivadas , Proteínas WT1/antagonistas & inhibidores , Proteínas WT1/inmunología
20.
Cancer Res ; 74(18): 5008-18, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25074614

RESUMEN

Toll-like receptors (TLR) recognize pathogen molecules and danger-associated signals that stimulate inflammatory processes. TLRs have been studied mainly in antigen-presenting cells, where they exert important immune regulatory functions, but they are also expressed by epithelial tumor cells, where they have been implicated in tumor progression. In this study, we demonstrate that the injection of TLR7 agonist in NOD/SCID mice, in C57BL/6 wild-type, and TLR7-deficient mice grafted with lung adenocarcinoma tumor cells leads to increased tumor progression and chemotherapeutic resistance. In patients with non-small cell lung cancer, expression analyses revealed that high TLR7 expression was strongly associated with resistance to neoadjuvant chemotherapy and poor clinical outcomes. Our findings delineate a crucial role for TLR7 in lung cancer physiopathology. Cancer Res; 74(18); 5008-18. ©2014 AACR.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Guanosina/análogos & derivados , Neoplasias Pulmonares/tratamiento farmacológico , Receptor Toll-Like 7/agonistas , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Guanosina/farmacología , Humanos , Neoplasias Pulmonares/metabolismo , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Transducción de Señal , Receptor Toll-Like 7/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA