Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(7): 1189-1207.e25, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35325594

RESUMEN

Macrophage infiltration is a hallmark of solid cancers, and overall macrophage infiltration correlates with lower patient survival and resistance to therapy. Tumor-associated macrophages, however, are phenotypically and functionally heterogeneous. Specific subsets of tumor-associated macrophage might be endowed with distinct roles on cancer progression and antitumor immunity. Here, we identify a discrete population of FOLR2+ tissue-resident macrophages in healthy mammary gland and breast cancer primary tumors. FOLR2+ macrophages localize in perivascular areas in the tumor stroma, where they interact with CD8+ T cells. FOLR2+ macrophages efficiently prime effector CD8+ T cells ex vivo. The density of FOLR2+ macrophages in tumors positively correlates with better patient survival. This study highlights specific roles for tumor-associated macrophage subsets and paves the way for subset-targeted therapeutic interventions in macrophages-based cancer therapies.


Asunto(s)
Neoplasias de la Mama , Macrófagos , Mama/inmunología , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos , Femenino , Receptor 2 de Folato , Humanos , Linfocitos Infiltrantes de Tumor , Pronóstico
2.
Cell ; 184(15): 4016-4031.e22, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34081922

RESUMEN

Cross-presentation of antigens from dead tumor cells by type 1 conventional dendritic cells (cDC1s) is thought to underlie priming of anti-cancer CD8+ T cells. cDC1 express high levels of DNGR-1 (a.k.a. CLEC9A), a receptor that binds to F-actin exposed by dead cell debris and promotes cross-presentation of associated antigens. Here, we show that secreted gelsolin (sGSN), an extracellular protein, decreases DNGR-1 binding to F-actin and cross-presentation of dead cell-associated antigens by cDC1s. Mice deficient in sGsn display increased DNGR-1-dependent resistance to transplantable tumors, especially ones expressing neoantigens associated with the actin cytoskeleton, and exhibit greater responsiveness to cancer immunotherapy. In human cancers, lower levels of intratumoral sGSN transcripts, as well as presence of mutations in proteins associated with the actin cytoskeleton, are associated with signatures of anti-cancer immunity and increased patient survival. Our results reveal a natural barrier to cross-presentation of cancer antigens that dampens anti-tumor CD8+ T cell responses.


Asunto(s)
Reactividad Cruzada/inmunología , Gelsolina/metabolismo , Inmunidad , Lectinas Tipo C/metabolismo , Neoplasias/inmunología , Receptores Inmunológicos/metabolismo , Receptores Mitogénicos/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Reactividad Cruzada/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Gelsolina/química , Gelsolina/deficiencia , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunidad/efectos de los fármacos , Ratones Endogámicos C57BL , Mutación/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Unión Proteica/efectos de los fármacos , Análisis de Supervivencia
3.
Nat Immunol ; 22(2): 140-153, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33349708

RESUMEN

Type 1 conventional dendritic (cDC1) cells are necessary for cross-presentation of many viral and tumor antigens to CD8+ T cells. cDC1 cells can be identified in mice and humans by high expression of DNGR-1 (also known as CLEC9A), a receptor that binds dead-cell debris and facilitates XP of corpse-associated antigens. Here, we show that DNGR-1 is a dedicated XP receptor that signals upon ligand engagement to promote phagosomal rupture. This allows escape of phagosomal contents into the cytosol, where they access the endogenous major histocompatibility complex class I antigen processing pathway. The activity of DNGR-1 maps to its signaling domain, which activates SYK and NADPH oxidase to cause phagosomal damage even when spliced into a heterologous receptor and expressed in heterologous cells. Our data reveal the existence of innate immune receptors that couple ligand binding to endocytic vesicle damage to permit MHC class I antigen presentation of exogenous antigens and to regulate adaptive immunity.


Asunto(s)
Presentación de Antígeno , Reactividad Cruzada , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Fagosomas/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Mitogénicos/metabolismo , Linfocitos T/metabolismo , Animales , Muerte Celular , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células HEK293 , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Lectinas Tipo C/genética , Ligandos , Ratones , NADPH Oxidasas/metabolismo , Fagosomas/genética , Fagosomas/inmunología , Fosforilación , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Receptores Inmunológicos/genética , Receptores Mitogénicos/genética , Transducción de Señal , Quinasa Syk/metabolismo , Linfocitos T/inmunología
4.
Cell ; 172(5): 1022-1037.e14, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29429633

RESUMEN

Conventional type 1 dendritic cells (cDC1) are critical for antitumor immunity, and their abundance within tumors is associated with immune-mediated rejection and the success of immunotherapy. Here, we show that cDC1 accumulation in mouse tumors often depends on natural killer (NK) cells that produce the cDC1 chemoattractants CCL5 and XCL1. Similarly, in human cancers, intratumoral CCL5, XCL1, and XCL2 transcripts closely correlate with gene signatures of both NK cells and cDC1 and are associated with increased overall patient survival. Notably, tumor production of prostaglandin E2 (PGE2) leads to evasion of the NK cell-cDC1 axis in part by impairing NK cell viability and chemokine production, as well as by causing downregulation of chemokine receptor expression in cDC1. Our findings reveal a cellular and molecular checkpoint for intratumoral cDC1 recruitment that is targeted by tumor-derived PGE2 for immune evasion and that could be exploited for cancer therapy.


Asunto(s)
Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Neoplasias/patología , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral , Quimiocina CCL5/metabolismo , Quimiocinas C/metabolismo , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/patología , Ratones , Mutación/genética , Pronóstico , Proteínas Proto-Oncogénicas B-raf/genética , Análisis de Supervivencia
5.
Cell ; 162(6): 1257-70, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26343581

RESUMEN

The mechanisms by which melanoma and other cancer cells evade anti-tumor immunity remain incompletely understood. Here, we show that the growth of tumors formed by mutant Braf(V600E) mouse melanoma cells in an immunocompetent host requires their production of prostaglandin E2, which suppresses immunity and fuels tumor-promoting inflammation. Genetic ablation of cyclooxygenases (COX) or prostaglandin E synthases in Braf(V600E) mouse melanoma cells, as well as in Nras(G12D) melanoma or in breast or colorectal cancer cells, renders them susceptible to immune control and provokes a shift in the tumor inflammatory profile toward classic anti-cancer immune pathways. This mouse COX-dependent inflammatory signature is remarkably conserved in human cutaneous melanoma biopsies, arguing for COX activity as a driver of immune suppression across species. Pre-clinical data demonstrate that inhibition of COX synergizes with anti-PD-1 blockade in inducing eradication of tumors, implying that COX inhibitors could be useful adjuvants for immune-based therapies in cancer patients.


Asunto(s)
Neoplasias/inmunología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Escape del Tumor , Inmunidad Adaptativa , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD/inmunología , Aspirina/administración & dosificación , Línea Celular Tumoral , Células Dendríticas/inmunología , Humanos , Inmunidad Innata , Inmunoterapia , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Cadenas alfa de Integrinas/inmunología , Interferones/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Ratones , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Prostaglandinas/inmunología , Proteínas Proto-Oncogénicas B-raf/metabolismo
6.
Immunity ; 53(6): 1215-1229.e8, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33220234

RESUMEN

Inflammation can support or restrain cancer progression and the response to therapy. Here, we searched for primary regulators of cancer-inhibitory inflammation through deep profiling of inflammatory tumor microenvironments (TMEs) linked to immune-dependent control in mice. We found that early intratumoral accumulation of interferon gamma (IFN-γ)-producing natural killer (NK) cells induced a profound remodeling of the TME and unleashed cytotoxic T cell (CTL)-mediated tumor eradication. Mechanistically, tumor-derived prostaglandin E2 (PGE2) acted selectively on EP2 and EP4 receptors on NK cells, hampered the TME switch, and enabled immune evasion. Analysis of patient datasets across human cancers revealed distinct inflammatory TME phenotypes resembling those associated with cancer immune control versus escape in mice. This allowed us to generate a gene-expression signature that integrated opposing inflammatory factors and predicted patient survival and response to immune checkpoint blockade. Our findings identify features of the tumor inflammatory milieu associated with immune control of cancer and establish a strategy to predict immunotherapy outcomes.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inflamación/inmunología , Neoplasias/inmunología , Escape del Tumor/inmunología , Animales , Dinoprostona/metabolismo , Humanos , Inmunoterapia , Inflamación/genética , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Ratones , Neoplasias/terapia , Fenotipo , Pronóstico , Prostaglandina-Endoperóxido Sintasas/genética , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/inmunología
8.
Cell ; 154(4): 843-58, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23953115

RESUMEN

Mononuclear phagocytes are classified as macrophages or dendritic cells (DCs) based on cell morphology, phenotype, or select functional properties. However, these attributes are not absolute and often overlap, leading to difficulties in cell-type identification. To circumvent this issue, we describe a mouse model to define DCs based on their ontogenetic descendence from a committed precursor. We show that precursors of mouse conventional DCs, but not other leukocytes, are marked by expression of DNGR-1. Genetic tracing of DNGR-1 expression history specifically marks cells traditionally ascribed to the DC lineage, and this restriction is maintained after inflammation. Notably, in some tissues, cells previously thought to be monocytes/macrophages are in fact descendants from DC precursors. These studies provide an in vivo model for fate mapping of DCs, distinguishing them from other leukocyte lineages, and thus help to unravel the functional complexity of the mononuclear phagocyte system.


Asunto(s)
Linaje de la Célula , Células Dendríticas/citología , Lectinas Tipo C/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Células Dendríticas/metabolismo , Hematopoyesis , Inflamación/patología , Riñón/citología , Lectinas Tipo C/genética , Células Progenitoras Linfoides/metabolismo , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Fagocitos/citología , Receptores de IgG/metabolismo , Receptores Inmunológicos/genética
9.
EMBO J ; 41(6): e109760, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35156720

RESUMEN

RNA editing by the adenosine deaminase ADAR1 prevents innate immune responses to endogenous RNAs. In ADAR1-deficient cells, unedited self RNAs form base-paired structures that resemble viral RNAs and inadvertently activate the cytosolic RIG-I-like receptor (RLR) MDA5, leading to an antiviral type I interferon (IFN) response. Mutations in ADAR1 cause Aicardi-Goutières Syndrome (AGS), an autoinflammatory syndrome characterized by chronic type I IFN production. Conversely, ADAR1 loss and the consequent type I IFN production restricts tumor growth and potentiates the activity of some chemotherapeutics. Here, we show that another RIG-I-like receptor, LGP2, also has an essential role in the induction of a type I IFN response in ADAR1-deficient human cells. This requires the canonical function of LGP2 as an RNA sensor and facilitator of MDA5-dependent signaling. Furthermore, we show that the sensitivity of tumor cells to ADAR1 loss requires LGP2 expression. Finally, type I IFN induction in tumor cells depleted of ADAR1 and treated with some chemotherapeutics fully depends on LGP2 expression. These findings highlight a central role for LGP2 in self RNA sensing with important clinical implications.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso , Malformaciones del Sistema Nervioso , ARN Helicasas/metabolismo , Enfermedades Autoinmunes del Sistema Nervioso/genética , Humanos , Malformaciones del Sistema Nervioso/genética , Edición de ARN , ARN Bicatenario
10.
Eur J Immunol ; 54(2): e2350635, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38059519

RESUMEN

Tumor immune escape is a major factor contributing to cancer progression and unresponsiveness to cancer therapies. Tumors can produce prostaglandin E2 (PGE2 ), an inflammatory mediator that directly acts on Natural killer (NK) cells to inhibit antitumor immunity. However, precisely how PGE2 influences NK cell tumor-restraining functions remains unclear. Here, we report that following PGE2 treatment, human NK cells exhibited altered expression of specific activating receptors and a reduced ability to degranulate and kill cancer targets. Transcriptional analysis uncovered that PGE2 also differentially modulated the expression of chemokine receptors by NK cells, inhibiting CXCR3 but increasing CXCR4. Consistent with this, PGE2-treated NK cells exhibited decreased migration to CXCL10 but increased ability to migrate toward CXCL12. Using live cell imaging, we showed that in the presence of PGE2 , NK cells were slower and less likely to kill cancer target cells following conjugation. Imaging the sequential stages of NK cell killing revealed that PGE2 impaired NK cell polarization, but not the re-organization of synaptic actin or the release of perforin itself. Together, these findings demonstrate that PGE2 affects multiple but select NK cell functions. Understanding how cancer cells subvert NK cells is necessary to more effectively harness the cancer-inhibitory function of NK cells in treatments.


Asunto(s)
Dinoprostona , Células Asesinas Naturales , Humanos , Dinoprostona/metabolismo , Línea Celular Tumoral , Inmunidad
11.
Immunity ; 42(6): 1197-211, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26084029

RESUMEN

Dendritic cells (DCs) are key players in the immune system. Much of their biology has been elucidated via culture systems in which hematopoietic precursors differentiate into DCs under the aegis of cytokines. A widely used protocol involves the culture of murine bone marrow (BM) cells with granulocyte-macrophage colony-stimulating factor (GM-CSF) to generate BM-derived DCs (BMDCs). BMDCs express CD11c and MHC class II (MHCII) molecules and share with DCs isolated from tissues the ability to present exogenous antigens to T cells and to respond to microbial stimuli by undergoing maturation. We demonstrate that CD11c(+)MHCII(+) BMDCs are in fact a heterogeneous group of cells that comprises conventional DCs and monocyte-derived macrophages. DCs and macrophages in GM-CSF cultures both undergo maturation upon stimulation with lipopolysaccharide but respond differentially to the stimulus and remain separable entities. These results have important implications for the interpretation of a vast array of data obtained with DC culture systems.


Asunto(s)
Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Macrófagos/inmunología , Animales , Presentación de Antígeno , Antígeno CD11c/metabolismo , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Inmunofenotipificación , Lipopolisacáridos/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Transcriptoma
12.
Nature ; 551(7678): 110-114, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29072292

RESUMEN

Interleukin-1 receptor 8 (IL-1R8, also known as single immunoglobulin IL-1R-related receptor, SIGIRR, or TIR8) is a member of the IL-1 receptor (ILR) family with distinct structural and functional characteristics, acting as a negative regulator of ILR and Toll-like receptor (TLR) downstream signalling pathways and inflammation. Natural killer (NK) cells are innate lymphoid cells which mediate resistance against pathogens and contribute to the activation and orientation of adaptive immune responses. NK cells mediate resistance against haematopoietic neoplasms but are generally considered to play a minor role in solid tumour carcinogenesis. Here we report that IL-1R8 serves as a checkpoint for NK cell maturation and effector function. Its genetic blockade unleashes NK-cell-mediated resistance to hepatic carcinogenesis, haematogenous liver and lung metastasis, and cytomegalovirus infection.


Asunto(s)
Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas/inmunología , Muromegalovirus/inmunología , Receptores de Interleucina-1/inmunología , Animales , Diferenciación Celular/genética , Femenino , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/inmunología , Humanos , Células Asesinas Naturales/metabolismo , Neoplasias Hepáticas/genética , Activación de Linfocitos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/inmunología , Receptores de Interleucina-1/genética
13.
Immunity ; 36(4): 635-45, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22483800

RESUMEN

Sterile inflammation can be initiated by innate immune recognition of markers of tissue injury termed damage-associated molecular patterns (DAMPs). DAMP recognition by dendritic cells (DCs) has also been postulated to lead to T cell responses to foreign antigens in tumors or allografts. Many DAMPs represent intracellular contents that are released upon cell damage, notably after necrosis. In this regard, we have previously described DNGR-1 (CLEC9A) as a DC-restricted receptor specific for an unidentified DAMP that is exposed by necrotic cells and is necessary for efficient priming of cytotoxic T cells against dead cell-associated antigens. Here, we have shown that the DNGR-1 ligand is preserved from yeast to man and corresponds to the F-actin component of the cellular cytoskeleton. The identification of F-actin as a DNGR-1 ligand suggests that cytoskeletal exposure is a universal sign of cell damage that can be targeted by the innate immune system to initiate immunity.


Asunto(s)
Actinas/metabolismo , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Necrosis/metabolismo , Receptores Mitogénicos/inmunología , Receptores Mitogénicos/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Citoesqueleto de Actina/metabolismo , Actinas/genética , Células Dendríticas/metabolismo , Células HeLa , Humanos , Inmunidad Innata , Necrosis/inmunología , Interferencia de ARN , ARN Interferente Pequeño , Saccharomyces cerevisiae/genética , Linfocitos T Citotóxicos/inmunología
15.
J Immunol ; 195(10): 5066-76, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26459350

RESUMEN

Dendritic cells (DCs) are powerful APCs that can induce Ag-specific adaptive immune responses and are increasingly recognized as important players in innate immunity to both infection and malignancy. Interestingly, although there are multiple described hematological malignancies, DC cancers are rarely observed in humans. Whether this is linked to the immunogenic potential of DCs, which might render them uniquely susceptible to immune control upon neoplastic transformation, has not been fully investigated. To address the issue, we generated a genetically engineered mouse model in which expression of Cre recombinase driven by the C-type lectin domain family 9, member a (Clec9a) locus causes expression of the Kirsten rat sarcoma viral oncogene homolog (Kras)(G12D) oncogenic driver and deletion of the tumor suppressor p53 within developing and differentiated DCs. We show that these Clec9a(Kras-G12D) mice rapidly succumb from disease and display massive accumulation of transformed DCs in multiple organs. In bone marrow chimeras, the development of DC cancer could be induced by a small number of transformed cells and was not prevented by the presence of untransformed DCs. Notably, activation of transformed DCs did not happen spontaneously but could be induced upon stimulation. Although Clec9a(Kras-G12D) mice showed altered thymic T cell development, peripheral T cells were largely unaffected during DC cancer development. Interestingly, transformed DCs were rejected upon adoptive transfer into wild-type but not lymphocyte-deficient mice, indicating that immunological control of DC cancer is in principle possible but does not occur during spontaneous generation in Clec9a(Kras-G12D) mice. Our findings suggest that neoplastic transformation of DCs does not by default induce anti-cancer immunity and can develop unhindered by immunological barriers.


Asunto(s)
Transformación Celular Neoplásica/inmunología , Células Dendríticas/inmunología , Neoplasias Experimentales/inmunología , Células Madre/inmunología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Dendríticas/patología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/inmunología , Ratas , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Células Madre/patología
16.
J Immunol ; 194(1): 307-15, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25411201

RESUMEN

Dendritic cells (DCs) are key regulators of innate and adaptive immunity. Our understanding of immune function has benefited greatly from mouse models allowing for selective ablation of DCs. Many such models rely on transgenic diphtheria toxin receptor (DTR) expression driven by DC-restricted promoters. This renders DCs sensitive to DT but is otherwise thought to have no effect on immune physiology. In this study, we report that, unexpectedly, mice in which DTR is expressed on conventional DCs display marked lymph node (LN) hypocellularity and reduced frequency of DCs in the same organs but not in spleen or nonlymphoid tissues. Intriguingly, in mixed bone marrow chimeras the phenotype conferred by DTR-expressing DCs is dominant over control bone marrow-derived cells, leading to small LNs and an overall paucity of DCs independently of the genetic ability to express DTR. The finding of alterations in LN composition and size independently of DT challenge suggests that caution must be exercised when interpreting results of experiments obtained with mouse models to inducibly deplete DCs. It further indicates that DTR, a member of the epidermal growth factor family, is biologically active in mice. Its use in cell ablation experiments needs to be considered in light of this activity.


Asunto(s)
Células de la Médula Ósea/citología , Células Dendríticas/inmunología , Factor de Crecimiento Similar a EGF de Unión a Heparina/inmunología , Ganglios Linfáticos/patología , Animales , Células de la Médula Ósea/inmunología , Antígeno CD11c/metabolismo , Células Dendríticas/citología , Toxina Diftérica/inmunología , Factor de Crecimiento Similar a EGF de Unión a Heparina/biosíntesis , Lectinas Tipo C/genética , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Receptores Inmunológicos/genética , Bazo/citología , Bazo/inmunología
17.
Trends Immunol ; 34(7): 329-35, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23608152

RESUMEN

We understand much about the agents, receptors, and signalling pathways that lead to immunity to pathogens. Less is known about how the process is initiated in apparently sterile conditions such as spontaneous immunity to certain tumours, tissue grafts, or autoimmune disorders. Proinflammatory molecules released by dying cells, termed damage-associated molecular patterns (DAMPs), have been proposed to activate dendritic cells (DCs) to promote T cell responses to antigens present in cell corpses. Surprisingly, rather than affecting activation, some recently identified DAMP receptors control specialised DC functions such as antigen acquisition and presentation. This selectivity reveals a new point of control in the regulation of adaptive immunity and, potentially, tolerance that renders DAMPs nonredundant players in responses to both sterile and nonsterile insults.


Asunto(s)
Apoptosis/inmunología , Células Dendríticas/inmunología , Infecciones/inmunología , Receptores Depuradores/inmunología , Heridas y Lesiones/inmunología , Inmunidad Adaptativa , Animales , Presentación de Antígeno , Antígenos/inmunología , Muerte Celular/inmunología , Humanos , Inmunidad Innata , Activación de Linfocitos
18.
Proc Natl Acad Sci U S A ; 110(16): 6494-9, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23576744

RESUMEN

Most Forkhead box P3(+) (Foxp3(+)) CD4 regulatory T cell (Treg) precursors are newly formed thymocytes that acquire Foxp3 expression on antigen encounter in the thymus. Differentiation of Treg, however, can also occur in the periphery. What limits this second layer of self- and nonself-reactive Treg production in physiological conditions remains to be understood. In this work, we tested the hypothesis that, similarly to thymic Treg, the precursors of peripheral Treg are immature T cells. We show that CD4(+)CD8(-)Foxp3(-) thymocytes and recent thymic emigrants (RTEs), contrarily to peripheral naïve mature cells, efficiently differentiate into Treg on transfer into lymphopenic mice. By varying donor and recipient mice and conducting ex vivo assays, we document that the preferential conversion of newly formed T cells does not require intrathymic preactivation, is cell-intrinsic, and correlates with low and high sensitivity to natural inhibitors and inducers of Foxp3 expression, such as IL-6, T-cell receptor triggering, and TGF-ß. Finally, ex vivo analysis of human thymocytes and peripheral blood T cells revealed that human RTE and newly developed T cells share an increased potential to acquire a FOXP3(bright)CD25(high) Treg phenotype. Our findings indicating that RTEs are the precursors of Tregs differentiated in the periphery should guide the design of Treg-based therapies.


Asunto(s)
Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Células Precursoras de Linfocitos T/citología , Linfocitos T Reguladores/citología , Timocitos/citología , Animales , Células Cultivadas , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Humanos , Interleucina-6/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/inmunología , Estadísticas no Paramétricas , Timocitos/inmunología , Timocitos/trasplante , Factor de Crecimiento Transformador beta/inmunología
19.
Science ; 384(6694): 428-437, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38662827

RESUMEN

A role for vitamin D in immune modulation and in cancer has been suggested. In this work, we report that mice with increased availability of vitamin D display greater immune-dependent resistance to transplantable cancers and augmented responses to checkpoint blockade immunotherapies. Similarly, in humans, vitamin D-induced genes correlate with improved responses to immune checkpoint inhibitor treatment as well as with immunity to cancer and increased overall survival. In mice, resistance is attributable to the activity of vitamin D on intestinal epithelial cells, which alters microbiome composition in favor of Bacteroides fragilis, which positively regulates cancer immunity. Our findings indicate a previously unappreciated connection between vitamin D, microbial commensal communities, and immune responses to cancer. Collectively, they highlight vitamin D levels as a potential determinant of cancer immunity and immunotherapy success.


Asunto(s)
Bacteroides fragilis , Microbioma Gastrointestinal , Inhibidores de Puntos de Control Inmunológico , Neoplasias , Vitamina D , Animales , Femenino , Humanos , Masculino , Ratones , Bacteroides fragilis/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Ratones Endogámicos C57BL , Neoplasias/inmunología , Neoplasias/microbiología , Neoplasias/terapia , Vitamina D/administración & dosificación , Vitamina D/metabolismo , Dieta , Línea Celular Tumoral , Calcifediol/administración & dosificación , Calcifediol/metabolismo , Proteína de Unión a Vitamina D/genética , Proteína de Unión a Vitamina D/metabolismo
20.
Cancers (Basel) ; 15(6)2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36980678

RESUMEN

Treating colorectal cancer (CRC) is a major challenge due to the heterogeneous immunological, clinical and pathological landscapes. Immunotherapy has so far only proven effective in a very limited subgroup of CRC patients. To better define the immune landscape, we examined the immune gene expression profile in various subsets of CRC patients and used a mouse model of intestinal tumors to dissect immune functions. We found that the NK cell receptor, natural-killer group 2 member D (NKG2D, encoded by KLRK1) and NKG2D ligand gene expression is elevated in the most immunogenic subset of CRC patients. High level of KLRK1 positively correlated with the mRNA expression of IFNG and associated with a poor survival of CRC patients. We further show that NKG2D deficiency in the Apcmin/+ mouse model of intestinal tumorigenesis led to reduced intratumoral IFNγ production, reduced tumorigenesis and enhanced survival, suggesting that the high levels of IFNγ observed in the tumors of CRC patients may be a consequence of NKG2D engagement. The mechanisms governing the contribution of NKG2D to CRC progression highlighted in this study will fuel discussions about (i) the benefit of targeting NKG2D in CRC patients and (ii) the need to define the predictive value of NKG2D and NKG2D ligand expression across tumor types.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA