Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 31: 413-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23298206

RESUMEN

NKG2D is an activating receptor expressed by all NK cells and subsets of T cells. It serves as a major recognition receptor for detection and elimination of transformed and infected cells and participates in the genesis of several inflammatory diseases. The ligands for NKG2D are self-proteins that are induced by pathways that are active in certain pathophysiological states. NKG2D ligands are regulated transcriptionally, at the level of mRNA and protein stability, and by cleavage from the cell surface. In some cases, ligand induction can be attributed to pathways that are activated specifically in cancer cells or infected cells. We review the numerous pathways that have been implicated in the regulation of NKG2D ligands, discuss the pathologic states in which those pathways are likely to act, and attempt to synthesize the findings into general schemes of NKG2D ligand regulation in NK cell responses to cancer and infection.


Asunto(s)
Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Animales , Humanos , Células Asesinas Naturales/patología , Ligandos , Ratones , Subfamilia K de Receptores Similares a Lectina de Células NK/biosíntesis , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Células T Asesinas Naturales/patología
2.
Immunity ; 49(4): 754-763.e4, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30332631

RESUMEN

Detection of cytosolic DNA by the enzyme cGAS triggers the production of cGAMP, a second messenger that binds and activates the adaptor protein STING, which leads to interferon (IFN) production. Here, we found that in vivo natural killer (NK) cell killing of tumor cells, but not of normal cells, depends on STING expression in non-tumor cells. Experiments using transplantable tumor models in STING- and cGAS-deficient mice revealed that cGAS expression by tumor cells was critical for tumor rejection by NK cells. In contrast, cGAS expression by host cells was dispensable, suggesting that tumor-derived cGAMP is transferred to non-tumor cells, where it activates STING. cGAMP administration triggered STING activation and IFN-ß production in myeloid cells and B cells but not NK cells. Our results reveal that the anti-tumor response of NK cells critically depends on the cytosolic DNA sensing pathway, similar to its role in defense against pathogens, and identify tumor-derived cGAMP as a major determinant of tumor immunogenicity with implications for cancer immunotherapy.


Asunto(s)
Interferones/inmunología , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/inmunología , Neoplasias/inmunología , Nucleótidos Cíclicos/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Regulación de la Expresión Génica/inmunología , Humanos , Interferones/metabolismo , Células Asesinas Naturales/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/genética , Neoplasias/metabolismo , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/inmunología , Nucleotidiltransferasas/metabolismo , Transducción de Señal/inmunología
3.
Nature ; 579(7800): E12, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32144410

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Nature ; 573(7774): 434-438, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31511694

RESUMEN

The accumulation of DNA in the cytosol serves as a key immunostimulatory signal associated with infections, cancer and genomic damage1,2. Cytosolic DNA triggers immune responses by activating the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway3. The binding of DNA to cGAS activates its enzymatic activity, leading to the synthesis of a second messenger, cyclic guanosine monophosphate-adenosine monophosphate (2'3'-cGAMP)4-7. This cyclic dinucleotide (CDN) activates STING8, which in turn activates the transcription factors interferon regulatory factor 3 (IRF3) and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), promoting the transcription of genes encoding type I interferons and other cytokines and mediators that stimulate a broader immune response. Exogenous 2'3'-cGAMP produced by malignant cells9 and other CDNs, including those produced by bacteria10-12 and synthetic CDNs used in cancer immunotherapy13,14, must traverse the cell membrane to activate STING in target cells. How these charged CDNs pass through the lipid bilayer is unknown. Here we used a genome-wide CRISPR-interference screen to identify the reduced folate carrier SLC19A1, a folate-organic phosphate antiporter, as the major transporter of CDNs. Depleting SLC19A1 in human cells inhibits CDN uptake and functional responses, and overexpressing SLC19A1 increases both uptake and functional responses. In human cell lines and primary cells ex vivo, CDN uptake is inhibited by folates as well as two medications approved for treatment of inflammatory diseases, sulfasalazine and the antifolate methotrexate. The identification of SLC19A1 as the major transporter of CDNs into cells has implications for the immunotherapeutic treatment of cancer13, host responsiveness to CDN-producing pathogenic microorganisms11 and-potentially-for some inflammatory diseases.


Asunto(s)
ADN/metabolismo , Nucleótidos Cíclicos/metabolismo , Proteína Portadora de Folato Reducido/metabolismo , Animales , Citosol , ADN/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Nucleótidos Cíclicos/inmunología , Nucleotidiltransferasas/metabolismo , Proteína Portadora de Folato Reducido/inmunología
5.
Proc Natl Acad Sci U S A ; 119(22): e2200568119, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35588144

RESUMEN

Cyclic dinucleotides (CDN) and Toll-like receptor (TLR) ligands mobilize antitumor responses by natural killer (NK) cells and T cells, potentially serving as complementary therapies to immune checkpoint therapy. In the clinic thus far, however, CDN therapy targeting stimulator of interferon genes (STING) protein has yielded mixed results, perhaps because it initiates responses potently but does not provide signals to sustain activation and proliferation of activated cytotoxic lymphocytes. To improve efficacy, we combined CDN with a half life-extended interleukin-2 (IL-2) superkine, H9-MSA (mouse serum albumin). CDN/H9-MSA therapy induced dramatic long-term remissions of the most difficult to treat major histocompatibility complex class I (MHC I)­deficient and MHC I+ tumor transplant models. H9-MSA combined with CpG oligonucleotide also induced potent responses. Mechanistically, tumor elimination required CD8 T cells and not NK cells in the case of MHC I+ tumors and NK cells but not CD8 T cells in the case of MHC-deficient tumors. Furthermore, combination therapy resulted in more prolonged and more intense NK cell activation, cytotoxicity, and expression of cytotoxic effector molecules in comparison with monotherapy. Remarkably, in a primary autochthonous sarcoma model that is refractory to PD-1 checkpoint therapy, the combination of CDN/H9-MSA with checkpoint therapy yielded long-term remissions in the majority of the animals, mediated by T cells and NK cells. This combination therapy has the potential to activate responses in tumors resistant to current therapies and prevent MHC I loss accompanying acquired resistance of tumors to checkpoint therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Antígenos de Histocompatibilidad Clase I , Inmunoterapia , Interleucina-2 , Proteínas de la Membrana , Neoplasias , Nucleótidos Cíclicos , Oligodesoxirribonucleótidos , Albúmina Sérica , Animales , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Inmunoterapia/métodos , Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/agonistas , Ratones , Neoplasias/genética , Neoplasias/terapia , Nucleótidos Cíclicos/uso terapéutico , Oligodesoxirribonucleótidos/uso terapéutico , Albúmina Sérica/uso terapéutico
7.
Immunol Rev ; 280(1): 93-101, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29027233

RESUMEN

Natural killer (NK) cells recognize and kill cancer cells and infected cells by engaging cell surface ligands that are induced preferentially or exclusively on these cells. These ligands are recognized by activating receptors on NK cells, such as NKG2D. In addition to activation by cell surface ligands, the acquisition of optimal effector activity by NK cells is driven in vivo by cytokines and other signals. This review addresses a developing theme in NK cell biology: that NK-activating ligands on cells, and the provision of cytokines and other signals that drive high effector function in NK cells, are driven by abnormalities that arise from transformation or the infected state. The pathways include genomic damage, which causes self DNA to be exposed in the cytosol of affected cells, where it activates the DNA sensor cGAS. The resulting signaling induces NKG2D ligands and also mobilizes NK cell activation. Other key pathways that regulate NKG2D ligands include PI-3 kinase activation, histone acetylation, and the integrated stress response. This review summarizes the roles of these pathways and their relevance in both viral infections and cancer.


Asunto(s)
Infecciones/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Animales , Citotoxicidad Inmunológica , Daño del ADN , Humanos , Vigilancia Inmunológica , Activación de Linfocitos , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Nucleotidiltransferasas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal
8.
PLoS Pathog ; 12(6): e1005708, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27295349

RESUMEN

Natural killer (NK) cells produce interferon (IFN)-γ and thus have been suggested to promote type I immunity during bacterial infections. Yet, Listeria monocytogenes (Lm) and some other pathogens encode proteins that cause increased NK cell activation. Here, we show that stimulation of NK cell activation increases susceptibility during Lm infection despite and independent from robust NK cell production of IFNγ. The increased susceptibility correlated with IL-10 production by responding NK cells. NK cells produced IL-10 as their IFNγ production waned and the Lm virulence protein p60 promoted induction of IL-10 production by mouse and human NK cells. NK cells consequently exerted regulatory effects to suppress accumulation and activation of inflammatory myeloid cells. Our results reveal new dimensions of the role played by NK cells during Lm infection and demonstrate the ability of this bacterial pathogen to exploit the induction of regulatory NK cell activity to increase host susceptibility.


Asunto(s)
Interleucina-10/inmunología , Células Asesinas Naturales/inmunología , Listeriosis/inmunología , Traslado Adoptivo , Animales , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Interleucina-10/biosíntesis , Listeria monocytogenes/inmunología , Listeriosis/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
J Immunol ; 197(10): 4127-4136, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798146

RESUMEN

Many NK cells express inhibitory receptors that bind self-MHC class I (MHC I) molecules and prevent killing of self-cells, while enabling killing of MHC I-deficient cells. But tolerance also occurs for NK cells that lack inhibitory receptors for self-MHC I, and for all NK cells in MHC I-deficient animals. In both cases, NK cells are unresponsive to MHC I-deficient cells and hyporesponsive when stimulated through activating receptors, suggesting that hyporesponsiveness is responsible for self-tolerance. We generated irradiation chimeras, or carried out adoptive transfers, with wild-type (WT) and/or MHC I-deficient hematopoietic cells in WT or MHC I-deficient C57BL/6 host mice. Unexpectedly, in WT hosts, donor MHC I-deficient hematopoietic cells failed to induce hyporesponsiveness to activating receptor stimulation, but did induce tolerance to MHC I-deficient grafts. Therefore, these two properties of NK cells are separable. Both tolerance and hyporesponsiveness occurred when the host was MHC I deficient. Interestingly, infections of mice or exposure to inflammatory cytokines reversed the tolerance of NK cells that was induced by MHC I-deficient hematopoietic cells, but not the tolerance induced by MHC I-deficient nonhematopoietic cells. These data have implications for successful bone marrow transplantation, and suggest that tolerance induced by hematopoietic cells versus nonhematopoietic cells may be imposed by distinct mechanisms.


Asunto(s)
Tolerancia Inmunológica , Células Asesinas Naturales/inmunología , Autotolerancia , Traslado Adoptivo , Animales , Trasplante de Médula Ósea , Citocinas , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Asesinas Naturales/clasificación , Células Asesinas Naturales/fisiología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Quimera por Radiación
10.
Semin Immunol ; 26(2): 138-44, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24629893

RESUMEN

Natural killer (NK) cells represent a first line of defense against pathogens and tumor cells. The activation of NK cells is regulated by the integration of signals deriving from activating and inhibitory receptors expressed on their surface. However, different NK cells respond differently to the same stimulus, be it target cells or agents that crosslink activating receptors. The processes that determine the level of NK cell responsiveness have been referred to collectively as NK cell education. NK cell education plays an important role in steady state conditions, where potentially auto-reactive NK cells are rendered tolerant to the surrounding environment. According to the "tuning" concept, the responsiveness of each NK cell is quantitatively adjusted to ensure self tolerance while at the same time ensuring useful reactivity against potential threats. MHC-specific inhibitory receptors displayed by NK cells play a major role in tuning NK cell responsiveness, but recent studies indicate that signaling from activating receptors is also important, suggesting that the critical determinant is an integrated signal from both types of receptors. An important and still unresolved question is whether NK cell education involves interactions with a specific cell population in the environment. Whether hematopoietic and/or non-hematopoietic cells play a role is still under debate. Recent results demonstrated that NK cell tuning exhibits plasticity in steady state conditions, meaning that it can be re-set if the MHC environment changes. Other evidence suggests, however, that inflammatory conditions accompanying infections may favor high responsiveness, indicating that inflammatory agents can over-ride the natural tendency of NK cells to adjust to the steady state environment. These findings raise many questions such as whether viruses and tumor cells manipulate NK cell responsiveness to evade immune-recognition. As knowledge of the underlying processes grows, the possibility of modulating NK cell responsiveness for therapeutic purposes is becoming increasingly attractive, and is now under serious investigation in clinical studies.


Asunto(s)
Células Asesinas Naturales/inmunología , Autotolerancia/inmunología , Animales , Citotoxicidad Inmunológica , Antígenos de Histocompatibilidad/inmunología , Antígenos de Histocompatibilidad/metabolismo , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo
11.
Blood ; 125(23): 3655-63, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25788701

RESUMEN

In allogeneic hematopoietic stem cell transplantation (HSCT), controlling graft-versus-host disease (GVHD) while maintaining graft-versus-tumor (GVT) responses is of critical importance. Using a mouse model of allogeneic HSCT, we hereby demonstrate that NKG2D expression by CD8(+) T cells plays a major role in mediating GVHD and GVT effects by promoting the survival and cytotoxic function of CD8(+) T cells. The expression of NKG2D ligands was not induced persistently on normal tissues of allogeneic HSCT-recipient mice treated with anti-NKG2D antibody, suggesting that transient NKG2D blockade might be sufficient to attenuate GVHD and allow CD8(+) T cells to regain their GVT function. Indeed, short-term treatment with anti-NKG2D antibody restored GVT effects while maintaining an attenuated GVHD state. NKG2D expression was also detected on CD8(+) T cells from allogeneic HSCT patients and trended to be higher in those with active GVHD. Together, these data support a novel role for NKG2D expression by CD8(+) T cells during allogeneic HSCT, which could be potentially therapeutically exploited to separate GVHD from GVT effects.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Regulación de la Expresión Génica/inmunología , Enfermedad Injerto contra Huésped/inmunología , Efecto Injerto vs Tumor/inmunología , Trasplante de Células Madre Hematopoyéticas , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Aloinjertos , Animales , Anticuerpos Neutralizantes/farmacología , Linfocitos T CD8-positivos/patología , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Efecto Injerto vs Tumor/efectos de los fármacos , Efecto Injerto vs Tumor/genética , Ratones , Ratones Noqueados , Subfamilia K de Receptores Similares a Lectina de Células NK/antagonistas & inhibidores , Subfamilia K de Receptores Similares a Lectina de Células NK/genética
12.
Immunity ; 28(4): 571-80, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18394936

RESUMEN

Ligands for the NKG2D stimulatory receptor are frequently upregulated on tumor lines, rendering them sensitive to natural killer (NK) cells, but the role of NKG2D in tumor surveillance has not been addressed in spontaneous cancer models. Here, we provided the first characterization of NKG2D-deficient mice, including evidence that NKG2D was not necessary for NK cell development but was critical for immunosurveillance of epithelial and lymphoid malignancies in two transgenic models of de novo tumorigenesis. In both models, we detected NKG2D ligands on the tumor cell surface ex vivo, providing needed evidence for ligand expression by primary tumors. In a prostate cancer model, aggressive tumors arising in NKG2D-deficient mice expressed higher amounts of NKG2D ligands than did similar tumors in wild-type mice, suggesting an NKG2D-dependent immunoediting of tumors in this model. These findings provide important genetic evidence for surveillance of primary tumors by an NK receptor.


Asunto(s)
Adenocarcinoma/inmunología , Fibrosarcoma/inmunología , Síndromes de Inmunodeficiencia/inmunología , Vigilancia Inmunológica , Linfoma de Células B/inmunología , Neoplasias de la Próstata/inmunología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Adenocarcinoma/genética , Animales , Benzo(a)Antracenos/toxicidad , Modelos Animales de Enfermedad , Femenino , Fibrosarcoma/inducido químicamente , Fibrosarcoma/genética , Síndromes de Inmunodeficiencia/genética , Vigilancia Inmunológica/genética , Linfoma de Células B/genética , Masculino , Metilcolantreno , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Subfamilia K de Receptores Similares a Lectina de Células NK , Neoplasias de la Próstata/genética , Receptores Inmunológicos/fisiología , Receptores de Células Asesinas Naturales
13.
Nat Rev Immunol ; 6(7): 520-31, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16799471

RESUMEN

Natural killer (NK) cells, similar to other lymphocytes, acquire tolerance to self. This means that NK cells have the potential to attack normal self cells but that there are mechanisms to ensure that this does not usually occur. Self-tolerance is acquired by NK cells during their development, but the underlying molecular and cellular mechanisms remain poorly understood. Recent studies have produced important new information about NK-cell self-tolerance. Here, we review the evidence for and against possible mechanisms of NK-cell self-tolerance, with an emphasis on the role of MHC-specific receptors.


Asunto(s)
Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Modelos Inmunológicos , Autotolerancia/inmunología , Animales , Antígenos de Histocompatibilidad/inmunología , Humanos , Activación de Linfocitos/inmunología , Receptores Inmunológicos/inmunología
14.
Blood ; 121(25): 5025-33, 2013 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-23649470

RESUMEN

The immunoreceptors NKG2D and NKp46 are known for their capacity to activate natural killer (NK) cell cytotoxicity and secretory responses in the contexts of tumors and infections, yet their roles in NK cell education remain unclear. Here, we provide the first characterization of mice deficient for both NKG2D and NKp46 receptors to address the relevance of their concomitant absence during NK cell development and function. Our findings reveal that NK cells develop normally in double-mutant (DKO) mice. Mice lacking NKG2D but not NKp46 showed subtle differences in the percentages of NK cells expressing inhibitory Ly49 receptors and the adhesion molecule DNAM-1. A slightly increased percentage of terminally differentiated NK cells and functional response to in vitro stimuli was observed in some experiments. These alterations were modest and did not affect NK cell function in vivo in response to mouse cytomegalovirus infection. NKp46 deficiency alone, or in combination with NKG2D deficiency, had no effect on frequency or function of NK cells.


Asunto(s)
Antígenos Ly/inmunología , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Subfamilia K de Receptores Similares a Lectina de Células NK/deficiencia , Receptor 1 Gatillante de la Citotoxidad Natural/deficiencia
15.
Blood ; 121(13): 2512-21, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23349395

RESUMEN

Mechanisms of spontaneous tumor regression have been difficult to characterize in a systematic manner due to their rare occurrence and the lack of model systems. Here, we provide evidence that early-stage B cells in Eµ-myc mice are tumorigenic and sharply regress in the periphery between 41 and 65 days of age. Regression depended on CD4(+), CD8(+), NK1.1(+) cells and the activation of the DNA damage response, which has been shown to provide an early barrier against cancer. The DNA damage response can induce ligands that enhance immune recognition. Blockade of DNAM-1, a receptor for one such ligand, impaired tumor regression. Hence, Eµ-myc mice provide a model to study spontaneous regression and possible mechanisms of immune evasion or suppression by cancer cells.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/fisiología , Células Asesinas Naturales/fisiología , Leucemia de Células B/inmunología , Regresión Neoplásica Espontánea/genética , Regresión Neoplásica Espontánea/inmunología , Proteínas Serina-Treonina Quinasas/fisiología , Linfocitos T/fisiología , Proteínas Supresoras de Tumor/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Apoptosis/inmunología , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos/genética , Genes myc/fisiología , Cadenas mu de Inmunoglobulina/genética , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucemia de Células B/genética , Leucemia de Células B/patología , Ratones , Ratones SCID , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
16.
J Immunol ; 190(6): 2880-5, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23390293

RESUMEN

NK cells use NK cell receptors to be able to recognize and eliminate infected, transformed, and allogeneic cells. Human NK cells are prevented from killing autologous healthy cells by virtue of inhibitory NKRs, primarily killer cell Ig-like receptors (KIR) that bind "self" HLA class I molecules. Individual NK cells stably express a selected set of KIR, but it is currently disputed whether the fraction of NK cells expressing a particular inhibitory KIR is influenced by the presence of the corresponding HLA ligand. The extreme polymorphism of the KIR and HLA loci, with wide-ranging affinities for individual KIR and HLA allele combinations, has made this issue particularly hard to tackle. In this study, we used a transgenic mouse model to investigate the effect of HLA on KIR repertoire and function in the absence of genetic variation inside and outside the KIR locus. These H-2K(b-/-) and H-2D(b-/-) mice lacked ligands for inhibitory Ly49 receptors and were transgenic for HLA-Cw3 and a KIR B haplotype. In this reductionist system, the presence of HLA-Cw3 reduced the frequency of KIR2DL2(+) cells, as well as the surface expression levels of KIR2DL2. In addition, in the presence of HLA-Cw3, the frequency of NKG2A(+) cells and the surface expression levels of NKG2A were reduced. In line with these findings, both transgene-encoded KIR and endogenous NKG2A contributed to the rejection of cells lacking HLA-Cw3. These findings support the idea that HLA influences the human KIR repertoire.


Asunto(s)
Antígenos HLA-C/fisiología , Modelos Inmunológicos , Receptores KIR2DL2/antagonistas & inhibidores , Receptores KIR/antagonistas & inhibidores , Animales , Antígenos H-2/genética , Antígenos HLA-C/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Subfamília C de Receptores Similares a Lectina de Células NK/antagonistas & inhibidores , Subfamília C de Receptores Similares a Lectina de Células NK/biosíntesis , Subfamília C de Receptores Similares a Lectina de Células NK/genética , Receptores KIR/biosíntesis , Receptores KIR/genética , Receptores KIR2DL2/genética , Receptores KIR2DL2/metabolismo
17.
Nat Rev Immunol ; 3(10): 781-90, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14523385

RESUMEN

According to present concepts, innate immunity is regulated by receptors that determine danger levels by responding to molecules that are associated with infection or cellular distress. NKG2D is, perhaps, the best characterized receptor that is associated with responses to cellular distress, defined as transformation, infection or cell stress. This review summarizes recent findings that concern NKG2D, its ligands, its signalling properties and its role in disease, and provides a framework for considering how the induction of immune responses can be regulated by cellular responses to injury.


Asunto(s)
Receptores Inmunológicos/inmunología , Animales , Humanos , Inmunidad Innata/inmunología , Células Asesinas Naturales/inmunología , Ligandos , Glicoproteínas de Membrana/inmunología , Ratones , Subfamilia K de Receptores Similares a Lectina de Células NK , Neoplasias/inmunología , Receptores de Superficie Celular/inmunología , Receptores de Células Asesinas Naturales , Transducción de Señal/inmunología , Receptores Toll-Like
18.
Blood ; 120(3): 592-602, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22661698

RESUMEN

Ly49-mediated recognition of MHC-I molecules on host cells is considered vital for natural killer (NK)-cell regulation and education; however, gene-deficient animal models are lacking because of the difficulty in deleting this large multigene family. Here, we describe NK gene complex knockdown (NKC(KD)) mice that lack expression of Ly49 and related MHC-I receptors on most NK cells. NKC(KD) NK cells exhibit defective killing of MHC-I-deficient, but otherwise normal, target cells, resulting in defective rejection by NKC(KD) mice of transplants from various types of MHC-I-deficient mice. Self-MHC-I immunosurveillance by NK cells in NKC(KD) mice can be rescued by self-MHC-I-specific Ly49 transgenes. Although NKC(KD) mice display defective recognition of MHC-I-deficient tumor cells, resulting in decreased in vivo tumor cell clearance, NKG2D- or antibody-dependent cell-mediated cytotoxicity-induced tumor cell cytotoxicity and cytokine production induced by activation receptors was efficient in Ly49-deficient NK cells, suggesting MHC-I education of NK cells is a single facet regulating their total potential. These results provide direct genetic evidence that Ly49 expression is necessary for NK-cell education to self-MHC-I molecules and that the absence of these receptors leads to loss of MHC-I-dependent "missing-self" immunosurveillance by NK cells.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Células Asesinas Naturales/inmunología , Subfamilia A de Receptores Similares a Lectina de Células NK/genética , Subfamilia A de Receptores Similares a Lectina de Células NK/inmunología , Animales , Antígenos Ly/genética , Antígenos Ly/inmunología , Degranulación de la Célula/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular Tumoral , Silenciador del Gen/inmunología , Células Asesinas Naturales/citología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Subfamília D de Receptores Similares a Lectina de las Células NK/genética , Subfamília D de Receptores Similares a Lectina de las Células NK/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Neoplasias/genética , Neoplasias/inmunología , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Transexualidad/genética
19.
J Immunol ; 188(9): 4468-75, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22467655

RESUMEN

Chronic obstructive pulmonary disease (COPD) is characterized by peribronchial and perivascular inflammation and largely irreversible airflow obstruction. Acute disease exacerbations, due frequently to viral infections, lead to enhanced disease symptoms and contribute to long-term progression of COPD pathology. Previously, we demonstrated that NK cells from cigarette smoke (CS)-exposed mice exhibit enhanced effector functions in response to stimulating cytokines or TLR ligands. In this article, we show that the activating receptor NKG2D is a key mediator for CS-stimulated NK cell hyperresponsiveness, because CS-exposed NKG2D-deficient mice (Klrk1(-/-)) did not exhibit enhanced effector functions as assessed by cytokine responsiveness. NK cell cytotoxicity against MHC class I-deficient targets was not affected in a COPD model. However, NK cells from CS-exposed mice exhibit greater cytotoxic activity toward cells that express the NKG2D ligand RAET1ε. We also demonstrate that NKG2D-deficient mice exhibit diminished airway damage and reduced inflammation in a model of viral COPD exacerbation, which do not affect viral clearance. Furthermore, adoptive transfer of NKG2D(+) NK cells into CS-exposed, influenza-infected NKG2D-deficient mice recapitulated the phenotypes observed in CS-exposed, influenza-infected wild-type mice. Our findings indicate that NKG2D stimulation during long-term CS exposure is a central pathway in the development of NK cell hyperresponsiveness and influenza-mediated exacerbations of COPD.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A/inmunología , Células Asesinas Naturales/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Infecciones por Orthomyxoviridae/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Contaminación por Humo de Tabaco/efectos adversos , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Células Asesinas Naturales/patología , Ratones , Ratones Noqueados , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/virología
20.
Sci Signal ; 17(827): eade3643, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38470955

RESUMEN

Activation of the endoplasmic reticulum (ER)-resident adaptor protein STING, a component of a cytosolic DNA-sensing pathway, induces the transcription of genes encoding type I interferons (IFNs) and other proinflammatory factors. Because STING is activated at the Golgi apparatus, control of the localization and activation of STING is important in stimulating antiviral and antitumor immune responses. Through a genome-wide CRISPR interference screen, we found that STING activation required the Golgi-resident protein ACBD3, which promotes the generation of phosphatidylinositol 4-phosphate (PI4P) at the trans-Golgi network, as well as other PI4P-associated proteins. Appropriate localization and activation of STING at the Golgi apparatus required ACBD3 and the PI4P-generating kinase PI4KB. In contrast, STING activation was enhanced when the lipid-shuttling protein OSBP, which removes PI4P from the Golgi apparatus, was inhibited by the US Food and Drug Administration-approved antifungal itraconazole. The increase in the abundance of STING-activating phospholipids at the trans-Golgi network resulted in the increased production of IFN-ß and other cytokines in THP-1 cells. Furthermore, a mutant STING that could not bind to PI4P failed to traffic from the ER to the Golgi apparatus in response to a STING agonist, whereas forced relocalization of STING to PI4P-enriched areas elicited STING activation in the absence of stimulation with a STING agonist. Thus, PI4P is critical for STING activation, and manipulating PI4P abundance may therapeutically modulate STING-dependent immune responses.


Asunto(s)
Aparato de Golgi , Fosfolípidos , Fosfolípidos/metabolismo , Aparato de Golgi/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA