Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Nat Immunol ; 17(4): 387-96, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26878112

RESUMEN

Activation of natural killer (NK) cells by hematopoietic target cells is controlled by the SLAM family of receptors and by the associated SAP family of adaptors. Here we found that SLAM receptors also enhanced NK cell activation by nonhematopoietic target cells, which lack ligands for SLAM receptors. This function was mediated by SLAMF6, a homotypic SLAM receptor found on NK cells and other hematopoietic cells, and was regulated by SAP adaptors, which uncoupled SLAM receptors from phosphatase SHP-1 and diminished the effect of SLAMF6 on NK cell responsiveness toward nonhematopoietic cells. Thus, in addition to their role in NK cell activation by hematopoietic cells, the SLAM-SAP pathways influence responsiveness toward nonhematopoietic targets by a process akin to NK cell 'education'.


Asunto(s)
Antígenos CD/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Células Asesinas Naturales/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Receptores de Superficie Celular/inmunología , Traslado Adoptivo , Animales , Línea Celular Tumoral , Células HeLa , Humanos , Inmunidad Innata , Activación de Linfocitos , Melanoma Experimental , Ratones , Transducción de Señal , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria
2.
BMC Immunol ; 24(1): 45, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37974069

RESUMEN

BACKGROUND: SARS-CoV-2 remains a world-wide health issue. SARS-CoV-2-specific immunity is induced upon both infection and vaccination. However, defining the long-term immune trajectory, especially after infection, is limited. In this study, we aimed to further the understanding of long-term SARS-CoV-2-specific immune response after infection. RESULTS: We conducted a longitudinal cohort study among 93 SARS-CoV-2 recovered individuals. Immune responses were continuously monitored for up to 20 months after infection. The humoral responses were quantified by Spike- and Nucleocapsid-specific IgG levels. T cell responses to Spike- and non-Spike epitopes were examined using both intercellular cytokine staining (ICS) assay and Activation-Induced marker (AIM) assay with quantification of antigen-specific IFNγ production. During the 20 months follow-up period, Nucleocapsid-specific antibody levels and non-Spike-specific CD4 + and CD8 + T cell frequencies decreased in the blood. However, a majority of participants maintained a durable immune responses 20 months after infection: 59% of the participants were seropositive for Nucleocapsid-specific IgG, and more than 70% had persisting non-Spike-specific T cells. The Spike-specific response initially decreased but as participants were vaccinated against COVID-19, Spike-specific IgG levels and T cell frequencies were boosted reaching similar or higher levels compared to 1 month post-infection. The trajectory of infection-induced SARS-CoV-2-specific immunity decreases, but for the majority of participants it persists beyond 20 months. The T cell response displays a greater durability. Vaccination boosts Spike-specific immune responses to similar or higher levels as seen after primary infection. CONCLUSIONS: For most participants, the response persists 20 months after infection, and the cellular response appears to be more long-lived compared to the circulating antibody levels. Vaccination boosts the S-specific response but does not affect the non-S-specific response. Together, these findings support the understanding of immune contraction, and with studies showing the immune levels required for protection, adds to the knowledge of durability of protection against future SARS-CoV-2.


Asunto(s)
COVID-19 , Humanos , Estudios Longitudinales , SARS-CoV-2 , Inmunidad Celular , Inmunoglobulina G , Anticuerpos Antivirales , Inmunidad Humoral , Vacunación
3.
Nature ; 544(7651): 493-497, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28424516

RESUMEN

Cancer cells elude anti-tumour immunity through multiple mechanisms, including upregulated expression of ligands for inhibitory immune checkpoint receptors. Phagocytosis by macrophages plays a critical role in cancer control. Therapeutic blockade of signal regulatory protein (SIRP)-α, an inhibitory receptor on macrophages, or of its ligand CD47 expressed on tumour cells, improves tumour cell elimination in vitro and in vivo, suggesting that blockade of the SIRPα-CD47 checkpoint could be useful in treating human cancer. However, the pro-phagocytic receptor(s) responsible for tumour cell phagocytosis is(are) largely unknown. Here we find that macrophages are much more efficient at phagocytosis of haematopoietic tumour cells, compared with non-haematopoietic tumour cells, in response to SIRPα-CD47 blockade. Using a mouse lacking the signalling lymphocytic activation molecule (SLAM) family of homotypic haematopoietic cell-specific receptors, we determined that phagocytosis of haematopoietic tumour cells during SIRPα-CD47 blockade was strictly dependent on SLAM family receptors in vitro and in vivo. In both mouse and human cells, this function required a single SLAM family member, SLAMF7 (also known as CRACC, CS1, CD319), expressed on macrophages and tumour cell targets. In contrast to most SLAM receptor functions, SLAMF7-mediated phagocytosis was independent of signalling lymphocyte activation molecule-associated protein (SAP) adaptors. Instead, it depended on the ability of SLAMF7 to interact with integrin Mac-1 (refs 18, 19, 20) and utilize signals involving immunoreceptor tyrosine-based activation motifs. These findings elucidate the mechanism by which macrophages engulf and destroy haematopoietic tumour cells. They also reveal a novel SAP adaptor-independent function for a SLAM receptor. Lastly, they suggest that patients with tumours expressing SLAMF7 are more likely to respond to SIRPα-CD47 blockade therapy.


Asunto(s)
Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Antígeno de Macrófago-1/metabolismo , Macrófagos/inmunología , Fagocitosis/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Actinas/metabolismo , Animales , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/metabolismo , Antígeno CD47/inmunología , Antígeno CD47/metabolismo , Femenino , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/deficiencia
4.
J Biol Chem ; 289(48): 33311-9, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25315772

RESUMEN

Human Galectin-3 (Gal-3), a ß-galactoside-binding protein expressed by tumor cells, has been reported to act as an immune regulator in antitumor T cells. However, its effect on natural killer (NK) cells is elusive. Using a recombinant human NK cell-activating receptor, NKp30 fusion protein (NKp30-Fc), we found that soluble NKp30-Fc could immunoprecipitate Galectin-3. The direct interaction between NKp30 and Galectin-3 was further confirmed using surface plasmon resonance experiments. Because Galectin-3 was mainly released from tumor cells in a soluble form in our study, the binding assay was performed to show that soluble Galectin-3 specifically bound to NK cells and NKp30 on the surface of the NK cells. Functionally, when soluble Galectin-3 was added to the NK-tumor cell coculture system, the NKp30-mediated, but not NKG2D-mediated, cytolysis and CD107a expression in the NK cells were inhibited, and these phenotypes could be restored by preincubation of soluble Galectin-3 with NKp30-Fc fusion protein or the addition of anti-Gal-3 antibody alone. Moreover, genetic down-regulation of Galectin-3 (shGal-3) resulted in tumor cells being more sensitive to NK cell lysis, and, reversely, Galectin-3-overexpressing HeLa cells (exGal-3) became less sensitive to NK cell killing. The results of these in vitro experiments were supported by studies in shGal-3-HeLa or exGal-3-HeLa xenograft non-obese diabetic/severe combined immunodeficiency mice after NK cell adoptive immunotherapy, indicating that Galectin-3 strongly antagonizes human NK cell attack against tumors in vivo. These findings indicate that Galectin-3 may function as an immune regulator to inhibit NK cell function against tumors, therefore providing a new therapeutic target for tumor treatment.


Asunto(s)
Galectina 3/inmunología , Inmunidad Celular , Receptor 3 Gatillante de la Citotoxidad Natural/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Escape del Tumor , Animales , Proteínas Sanguíneas , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Galectina 3/genética , Galectinas , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Células HeLa , Xenoinjertos , Humanos , Proteína 1 de la Membrana Asociada a los Lisosomas/genética , Proteína 1 de la Membrana Asociada a los Lisosomas/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Receptor 3 Gatillante de la Citotoxidad Natural/genética , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Neoplasias/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
5.
PLoS Pathog ; 9(9): e1003637, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24068938

RESUMEN

Herpes simplex encephalitis (HSE) is a lethal neurological disease resulting from infection with Herpes Simplex Virus 1 (HSV-1). Loss-of-function mutations in the UNC93B1, TLR3, TRIF, TRAF3, and TBK1 genes have been associated with a human genetic predisposition to HSE, demonstrating the UNC93B-TLR3-type I IFN pathway as critical in protective immunity to HSV-1. However, the TLR3, UNC93B1, and TRIF mutations exhibit incomplete penetrance and represent only a minority of HSE cases, perhaps reflecting the effects of additional host genetic factors. In order to identify new host genes, proteins and signaling pathways involved in HSV-1 and HSE susceptibility, we have implemented the first genome-wide mutagenesis screen in an in vivo HSV-1 infectious model. One pedigree (named P43) segregated a susceptible trait with a fully penetrant phenotype. Genetic mapping and whole exome sequencing led to the identification of the causative nonsense mutation L3X in the Receptor-type tyrosine-protein phosphatase C gene (Ptprc(L3X)), which encodes for the tyrosine phosphatase CD45. Expression of MCP1, IL-6, MMP3, MMP8, and the ICP4 viral gene were significantly increased in the brain stems of infected Ptprc(L3X) mice accounting for hyper-inflammation and pathological damages caused by viral replication. Ptprc(L3X) mutation drastically affects the early stages of thymocytes development but also the final stage of B cell maturation. Transfer of total splenocytes from heterozygous littermates into Ptprc(L3X) mice resulted in a complete HSV-1 protective effect. Furthermore, T cells were the only cell population to fully restore resistance to HSV-1 in the mutants, an effect that required both the CD4⁺ and CD8⁺ T cells and could be attributed to function of CD4⁺ T helper 1 (Th1) cells in CD8⁺ T cell recruitment to the site of infection. Altogether, these results revealed the CD45-mediated T cell function as potentially critical for infection and viral spread to the brain, and also for subsequent HSE development.


Asunto(s)
Codón sin Sentido , Encefalitis por Herpes Simple/genética , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Inmunidad Celular , Antígenos Comunes de Leucocito/metabolismo , Células TH1/inmunología , Animales , Tronco Encefálico/inmunología , Tronco Encefálico/metabolismo , Tronco Encefálico/patología , Tronco Encefálico/virología , Células Cultivadas , Cruzamientos Genéticos , Susceptibilidad a Enfermedades , Encefalitis por Herpes Simple/etiología , Femenino , Estudio de Asociación del Genoma Completo , Herpes Simple/patología , Herpes Simple/fisiopatología , Herpes Simple/virología , Antígenos Comunes de Leucocito/genética , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutagénesis , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Neuronas/patología , Neuronas/virología , Análisis de Supervivencia , Células TH1/metabolismo , Células TH1/patología , Células TH1/virología
6.
Cell Rep ; 43(3): 113800, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38386559

RESUMEN

Infection of mice by mouse cytomegalovirus (MCMV) triggers activation and expansion of Ly49H+ natural killer (NK) cells, which are virus specific and considered to be "adaptive" or "memory" NK cells. Here, we find that signaling lymphocytic activation molecule family receptors (SFRs), a group of hematopoietic cell-restricted receptors, are essential for the expansion of Ly49H+ NK cells after MCMV infection. This activity is largely mediated by CD48, an SFR broadly expressed on NK cells and displaying augmented expression after MCMV infection. It is also dependent on the CD48 counter-receptor, 2B4, expressed on host macrophages. The 2B4-CD48 axis promotes expansion of Ly49H+ NK cells by repressing their phagocytosis by virus-activated macrophages through inhibition of the pro-phagocytic integrin lymphocyte function-associated antigen-1 (LFA-1) on macrophages. These data identify key roles of macrophages and the 2B4-CD48 pathway in controlling the expansion of adaptive NK cells following MCMV infection. Stimulation of the 2B4-CD48 axis may be helpful in enhancing adaptive NK cell responses for therapeutic purposes.


Asunto(s)
Infecciones por Citomegalovirus , Receptores Inmunológicos , Animales , Ratones , Receptores Inmunológicos/metabolismo , Antígeno CD48/metabolismo , Antígenos CD/metabolismo , Activación de Linfocitos , Células Asesinas Naturales , Receptores de Superficie Celular/metabolismo , Proteínas Portadoras/metabolismo , Macrófagos/metabolismo , Fagocitosis
7.
J Exp Med ; 213(10): 2187-207, 2016 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-27573813

RESUMEN

Signaling lymphocytic activation molecule (SLAM) family receptors (SFRs) can mediate either activating or inhibitory effects during natural killer cell (NK cell) activation. In this study, we addressed the global role, regulation, and mechanism of action of the SLAM family in NK cells by analyzing a mouse lacking the entire ∼400-kilobase Slam locus, which encodes all six SFRs and CD48, the ligand of SFR 2B4. This mouse displayed enhanced NK cell activation responses toward hematopoietic target cells. Analyses of mice lacking individual SFRs showed that the inhibitory function of the Slam locus was due solely to 2B4 and was not influenced positively or negatively by other SFRs. Differences in NK cell responses between recognition of targets expressing or lacking ligands for SFRs were enhanced by IL-12 but suppressed by type I interferon. Cytokines also changed the levels of SLAM-associated protein adaptors, which prevent the inhibitory function of SFRs. The enhanced activation responses of SFR-deficient NK cells were dependent on integrin LFA-1 but not on DNAM-1 or NKG2D. SFR-mediated inhibition prevented the generation of activated forms of LFA-1. Hence, the Slam locus has an overall inhibitory role during NK cell activation that is solely dependent on 2B4. This effect is influenced by cytokines and leads to suppression of LFA-1 activity.


Asunto(s)
Citocinas/metabolismo , Eliminación de Gen , Sitios Genéticos , Células Asesinas Naturales/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Animales , Citotoxicidad Inmunológica , Hematopoyesis , Humanos , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica , Ratones Endogámicos C57BL , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Linfocitos T/metabolismo
8.
Mol Cell Biol ; 35(1): 41-51, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25312647

RESUMEN

Signaling lymphocytic activation molecule F7 (SLAMF7) is a receptor present on immune cells, including natural killer (NK) cells. It is also expressed on multiple myeloma (MM) cells. This led to development of an anti-SLAMF7 antibody, elotuzumab, showing efficacy against MM. SLAMF7 mediates activating or inhibitory effects in NK cells, depending on whether cells express or do not express the adaptor EAT-2. Since MM cells lack EAT-2, we elucidated the inhibitory effectors of SLAMF7 in EAT-2-negative NK cells and tested whether these effectors were triggered in MM cells. SLAMF7-mediated inhibition in NK cells lacking EAT-2 was mediated by SH2 domain-containing inositol phosphatase 1 (SHIP-1), which was recruited via tyrosine 261 of SLAMF7. Coupling of SLAMF7 to SHIP-1 required Src kinases, which phosphorylated SLAMF7. Although MM cells lack EAT-2, elotuzumab did not induce inhibitory signals in these cells. This was at least partly due to a lack of CD45, a phosphatase required for Src kinase activation. A defect in SLAMF7 function was also observed in CD45-deficient NK cells. Hence, SLAMF7-triggered inhibition is mediated by a mechanism involving Src kinases, CD45, and SHIP-1 that is defective in MM cells. This defect might explain why elotuzumab eliminates MM cells by an indirect mechanism involving the activation of NK cells.


Asunto(s)
Células Asesinas Naturales/citología , Antígenos Comunes de Leucocito/metabolismo , Mieloma Múltiple/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Receptores Inmunológicos/metabolismo , Familia-src Quinasas/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Células COS , Chlorocebus aethiops , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Inositol Polifosfato 5-Fosfatasas , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Fosforilación , Estructura Terciaria de Proteína , Interferencia de ARN , Transducción de Señal , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Tirosina/química
9.
J Exp Med ; 211(4): 727-42, 2014 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-24687958

RESUMEN

Ewing's sarcoma-associated transcript 2 (EAT-2) is an Src homology 2 domain-containing intracellular adaptor related to signaling lymphocytic activation molecule (SLAM)-associated protein (SAP), the X-linked lymphoproliferative gene product. Both EAT-2 and SAP are expressed in natural killer (NK) cells, and their combined expression is essential for NK cells to kill abnormal hematopoietic cells. SAP mediates this function by coupling SLAM family receptors to the protein tyrosine kinase Fyn and the exchange factor Vav, thereby promoting conjugate formation between NK cells and target cells. We used a variety of genetic, biochemical, and imaging approaches to define the molecular and cellular mechanisms by which EAT-2 controls NK cell activation. We found that EAT-2 mediates its effects in NK cells by linking SLAM family receptors to phospholipase Cγ, calcium fluxes, and Erk kinase. These signals are triggered by one or two tyrosines located in the carboxyl-terminal tail of EAT-2 but not found in SAP. Unlike SAP, EAT-2 does not enhance conjugate formation. Rather, it accelerates polarization and exocytosis of cytotoxic granules toward hematopoietic target cells. Hence, EAT-2 promotes NK cell activation by molecular and cellular mechanisms distinct from those of SAP. These findings explain the cooperative and essential function of these two adaptors in NK cell activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Calcio/metabolismo , Gránulos Citoplasmáticos/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Fosfolipasa C gamma/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Activación Enzimática , Exocitosis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Datos de Secuencia Molecular , Fosfolipasa C gamma/química , Fosforilación , Estructura Terciaria de Proteína , Transducción de Señal , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Relación Estructura-Actividad , Factores de Transcripción/química , Tirosina/metabolismo
10.
Crit Rev Oncol Hematol ; 88(1): 168-77, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23731618

RESUMEN

Signaling lymphocytic activation molecule (SLAM) family receptors have been implicated in normal immunity, immunodeficiencies and autoimmunity. CS1 (also known as CRACC, CD319 and SLAMF7) is a member of the SLAM family expressed on several normal hematopoietic cell types. It is also highly and nearly universally expressed on multiple myeloma (MM) cells. This review focuses on the biology of CS1, both in normal hematopoietic cells and in MM cells. It also discusses the preclinical and clinical data on the use of a humanized anti-CS1 monoclonal antibody, elotuzumab, for the treatment of MM. Based on current knowledge, CS1 is a compelling new target for the treatment of MM.


Asunto(s)
Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA