Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 201(5): 1460-1467, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30068594

RESUMEN

Four killer cell Ig-like receptor (KIR) genes, collectively referred to as framework genes, characterize almost all KIR haplotypes. In particular, KIR3DL3 and KIR3DL2 mark the ends of the locus, whereas KIR3DP1 and KIR2DL4 are located in the central part. A recombination hot spot, mapped between KIR3DP1 and KIR2DL4, splits the haplotypes into two regions: a centromeric (Cen) region (spanning from KIR3DL3 to KIR3DP1) and a telomeric region (from KIR2DL4 to KIR3DL2), both varying in KIR gene content. In this study, we analyzed KIR3DP1 polymorphism in a cohort of 316 healthy, unrelated individuals. To this aim, we divided KIR3DP1 alleles into two groups by the use of a sequence-specific primer- PCR approach. Our data clearly indicated that KIR3DP1 alleles present on haplotypes carrying Cen-A or Cen-B1 regions differ from those having Cen-B2 motifs. Few donors (∼3%) made exceptions, and they were all, except one, characterized by uncommon haplotypes, including either KIR deletions or KIR duplications. Consequently, as KIR2DL1 is present in Cen-A and Cen-B1 regions but absent in Cen-B2 regions, we demonstrated that KIR3DP1 polymorphism might represent a suitable marker for KIR2DL1 gene copy number analysis. Moreover, because Cen-B1 and Cen-B2 regions are characterized by different KIR3DP1 alleles, we showed that KIR3DP1 polymorphism analysis also provides information to dissect between Cen-B1/Cen-B1 and Cen-B1/Cen-B2 donors. Taken together, our data suggest that the analysis of KIR3DP1 polymorphism should be included in KIR repertoire evaluation.


Asunto(s)
Alelos , Centrómero/genética , Haplotipos , Polimorfismo Genético , Receptores KIR2DL4/genética , Receptores KIR3DS1/genética , Centrómero/inmunología , Femenino , Eliminación de Gen , Duplicación de Gen , Humanos , Masculino , Receptores KIR2DL4/inmunología , Receptores KIR3DS1/inmunología
2.
Clin Immunol ; 204: 31-36, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30391652

RESUMEN

X-linked lymphoproliferative disease 1 (XLP1) is a monogenic disorder caused by mutations in SH2D1A, resulting in the absence/dysfunction of the signaling lymphocyte activation molecule (SLAM)-associated protein (SAP). Consequently, SLAM receptors as 2B4 (CD244) and NTB-A (SLAMF6), upon ligand engagement, exert inhibitory instead of activating function. This causes an immune dysfunction that is worsened by the selective inability of NK and T cells to kill EBV-infected B cells with dramatic clinical sequelae (e.g. fulminant mononucleosis, hyperinflammation, lymphoma). Here we outline recent findings on the interplay between inhibitory 2B4 and the various activating receptors in NK cells. 2B4 engagement selectively blocks ITAM-dependent activating receptors as NCR and CD16, while it does not affect NKG2D and DNAM-1. Furthermore, inhibitory 2B4 participates to NK cell education, as highlighted by the existence in XLP1 patients of a large subset of fully functional NK cells that lack self-HLA specific inhibitory receptors and exert autoreactivity against mature dendritic cells.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Síndromes de Inmunodeficiencia/inmunología , Células Asesinas Naturales/inmunología , Trastornos Linfoproliferativos/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Animales , Humanos , Síndromes de Inmunodeficiencia/metabolismo , Células Asesinas Naturales/metabolismo , Trastornos Linfoproliferativos/metabolismo , Masculino , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo
3.
J Immunol ; 199(4): 1516-1525, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28701512

RESUMEN

Tyrosine kinase inhibitors (TKIs) are used in the clinical management of hematological neoplasms. Moreover, in solid tumors such as stage 4 neuroblastomas (NB), imatinib showed benefits that might depend on both on-target and immunological off-target effects. We investigated the effects of imatinib and nilotinib on human NK cells, monocytes, and macrophages. High numbers of monocytes died upon exposure to TKI concentrations similar to those achieved in patients. Conversely, NK cells were highly resistant to the TKI cytotoxic effect, were properly activated by immunostimulatory cytokines, and degranulated in the presence of NB cells. In NB, neither drug reduced the expression of ligands for activating NK receptors or upregulated that of HLA class I, B7-H3, PD-L1, and PD-L2, molecules that might limit NK cell function. Interestingly, TKIs modulated the chemokine receptor repertoire of immune cells. Acting at the transcriptional level, they increased the surface expression of CXCR4, an effect observed also in NK cells and monocytes of patients receiving imatinib for chronic myeloid leukemia. Moreover, TKIs reduced the expression of CXCR3 (in NK cells) and CCR1 (in monocytes). Monocytes also decreased the expression of M-CSFR, and low numbers of cells underwent differentiation toward macrophages. M0 and M2 macrophages were highly resistant to TKIs and maintained their phenotypic and functional characteristics. Importantly, also in the presence of TKIs, the M2 immunosuppressive polarization was reverted by TLR engagement, and M1-oriented macrophages fully activated autologous NK cells. Our results contribute to better interpreting the off-target efficacy of TKIs in tumors and to envisaging strategies aimed at facilitating antitumor immune responses.


Asunto(s)
Antineoplásicos/farmacología , Mesilato de Imatinib/farmacología , Células Asesinas Naturales/efectos de los fármacos , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Pirimidinas/farmacología , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Diferenciación Celular/efectos de los fármacos , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/fisiología , Activación de Linfocitos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/fisiología , Monocitos/inmunología , Monocitos/fisiología , Neuroblastoma/inmunología , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Receptores CCR1/genética , Receptores CCR1/inmunología , Receptores CCR1/metabolismo , Receptores CXCR3/genética , Receptores CXCR3/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
4.
Eur J Immunol ; 47(6): 1051-1061, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28386908

RESUMEN

X-linked lymphoproliferative disease 1 (XLP1) is an inherited immunodeficiency, caused by mutations in SH2D1A encoding Signaling Lymphocyte Activation Molecule (SLAM)-associated protein (SAP). In XLP1, 2B4, upon engagement with CD48, has inhibitory instead of activating function. This causes a selective inability of cytotoxic effectors to kill EBV-infected cells, with dramatic clinical sequelae. Here, we investigated the NK cell education in XLP1, upon characterization of killer Ig-like receptor (KIR)/KIR-L genotype and phenotypic repertoire of self-HLA class I specific inhibitory NK receptors (self-iNKRs). We also analyzed NK-cell cytotoxicity against CD48+ or CD48- KIR-ligand matched or autologous hematopoietic cells in XLP1 patients and healthy controls. XLP1 NK cells may show a defective phenotypic repertoire with substantial proportion of cells lacking self-iNKR. These NK cells are cytotoxic and the inhibitory 2B4/CD48 pathway plays a major role to prevent killing of CD48+ EBV-transformed B cells and M1 macrophages. Importantly, self-iNKR defective NK cells kill CD48- targets, such as mature DCs. Self-iNKR- NK cells in XLP1 patients are functional even in resting conditions, suggesting a role of the inhibitory 2B4/CD48 pathway in the education process during NK-cell maturation. Killing of autologous mature DC by self-iNKR defective XLP1 NK cells may impair adaptive responses, further exacerbating the patients' immune defect.


Asunto(s)
Células Asesinas Naturales/inmunología , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/fisiopatología , Receptores de Células Asesinas Naturales/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Antígeno CD48/inmunología , Antígeno CD48/metabolismo , Genes MHC Clase I , Humanos , Células Asesinas Naturales/metabolismo , Activación de Linfocitos , Canales de Potasio de Rectificación Interna/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología
5.
Eur J Immunol ; 46(6): 1511-7, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26990677

RESUMEN

KIR3DL1 is a natural killer (NK) cell receptor that recognizes the Bw4 epitope of human leukocyte antigen (HLA) class I molecules. Following hematopoietic stem cell transplantation for patients lacking Bw4, KIR3DL1-expressing NK cells from Bw4-positive donors can be alloreactive and eliminate tumor cells. However, KIR3DL1 alleles having T instead of C at nucleotide 320 (encoding leucine 86 instead of serine 86) are not expressed on the cell surface. Thus, not all individuals testing positive for KIR3DL1 are optimal donors for Bw4-negative recipients. Therefore, we developed a method for genotyping codon 86, which was validated by its perfect correlation with NK cell phenotype for 100 donors of diverse KIR3DL1/S1 genotype. We typed 600 donors and found that ∼12.2% had the KIR3DL1 gene, but did not express cell-surface KIR3DL1. By contrast, high-expressing allotypes were identified when haplotypes from four families with duplicated KIR3DL1/S1 genes were characterized at high resolution. Identifying donors who have KIR3DL1 but lack cell-surface KIR3DL1 would refine donor selection. With this technique, the number of individuals identified who may not be optimal donors for Bw4-negative patients increases by threefold, when compared with standard methods. Taken together, we propose that allele typing of killer cell Ig-like receptor (KIR) polymorphisms should become a standard practice when selecting donors.


Asunto(s)
Codón , Selección de Donante , Antígenos HLA-B/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Receptores KIR3DL1/genética , Receptores KIR3DS1/genética , Alelos , Membrana Celular/metabolismo , Expresión Génica , Genotipo , Haplotipos , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Modelos Biológicos , Polimorfismo de Nucleótido Simple , Receptores KIR3DL1/metabolismo , Receptores KIR3DS1/metabolismo , Trasplante Homólogo
6.
Stem Cells ; 34(7): 1909-21, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27015881

RESUMEN

Mesenchymal stromal cells (MSCs) support hematopoiesis and exert immunoregulatory activities. Here, we analyzed the functional outcome of the interactions between MSCs and monocytes/macrophages. We showed that MSCs supported the survival of monocytes that underwent differentiation into macrophages, in the presence of macrophage colony-stimulating factor. However, MSCs skewed their polarization toward a peculiar M2-like functional phenotype (M(MSC) ), through a prostaglandin E2-dependent mechanism. M(MSC) were characterized by high expression of scavenger receptors, increased phagocytic capacity, and high production of interleukin (IL)-10 and transforming growth factor-ß. These cytokines contributed to the immunoregulatory properties of M(MSC) , which differed from those of typical IL-4-induced macrophages (M2). In particular, interacting with activated natural killer (NK) cells, M(MSC) inhibited both the expression of activating molecules such as NKp44, CD69, and CD25 and the production of IFNγ, while M2 affected only IFNγ production. Moreover, M(MSC) inhibited the proliferation of CD8(+) T cells in response to allogeneic stimuli and induced the expansion of regulatory T cells (Tregs). Toll-like receptor engagement reverted the phenotypic and functional features of M(MSC) to those of M1 immunostimulatory/proinflammatory macrophages. Overall our data show that MSCs induce the generation of a novel type of alternatively activated macrophages capable of suppressing both innate and adaptive immune responses. These findings may help to better understand the role of MSCs in healthy tissues and inflammatory diseases including cancer, and provide clues for novel therapeutic approaches. Stem Cells 2016;34:1909-1921.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Activación de Macrófagos , Macrófagos/citología , Células Madre Mesenquimatosas/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Línea Celular , Proliferación Celular , Supervivencia Celular , Niño , Humanos , Inmunomodulación , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Macrófagos/metabolismo , Monocitos/citología , Linfocitos T Reguladores
7.
Eur J Immunol ; 44(6): 1814-22, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24510590

RESUMEN

We analyzed the functional outcome of the interaction between tumor-associated macrophages (TAMs) and natural killer (NK) cells. TAMs from ascites of ovarian cancer patients displayed an alternatively activated functional phenotype (M2) characterized by a remarkably high frequency and surface density of membrane-bound IL-18. Upon TLR engagement, TAMs acquired a classically activated functional phenotype (M1), released immunostimulatory cytokines (IL-12, soluble IL-18), and efficiently triggered the cytolytic activity of NK cells. TAMs also induced the release of IFN-γ from NK cells, which however was significantly lower compared with that induced by in vitro-polarized M2 cells. Most tumor-associated NK cells displayed a CD56(bright) , CD16(neg) or CD56(bright) , CD16(dim) phenotype, and very poor cytolytic activities, despite an increased expression of the activation marker CD69. They also showed downregulation of DNAM-1, 2B4, and NTB-A activating receptors, and an altered chemokine receptor repertoire. Importantly however, when appropriately stimulated, NK cells from the patients, including those cells isolated from ascites, efficiently killed autologous TAMs that expressed low, "nonprotective" levels of HLA class I molecules. Overall, our data show the existence of a complex tumor microenvironment in which poorly cytolytic/immature NK cells deal with immunosuppressive tumor-educated macrophages.


Asunto(s)
Tolerancia Inmunológica , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias Ováricas/inmunología , Receptores Toll-Like/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/inmunología , Citocinas/inmunología , Femenino , Humanos , Inmunidad Celular , Células Asesinas Naturales/patología , Macrófagos/patología , Persona de Mediana Edad , Neoplasias Ováricas/patología
8.
Eur J Immunol ; 44(5): 1526-34, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24496997

RESUMEN

X-linked lymphoproliferative disease 1 (XLP1) is a rare congenital immunodeficiency caused by SH2D1A (Xq25) mutations resulting in lack or dysfunction of SLAM-associated protein adaptor molecule. In XLP1 patients, upon ligand (CD48) engagement, 2B4 delivers inhibitory signals that impair the cytolytic activity of NK (and T) cells. This causes the selective inability to control EBV infections and the occurrence of B-cell lymphomas. Here, we show that in the absence of SLAM-associated protein, co-engagement of 2B4 with different activating receptors, either by antibodies or specific ligands on target cells, inhibits different ITAM-dependent signaling pathways including activating killer Ig-like receptors. In XLP1 NK cells, 2B4 affected both the cytolytic and IFN-γ production capabilities, functions that were restored upon disruption of the 2B4/CD48 interactions. Notably, we provide evidence that 2B4 dysfunction does not affect the activity of DNAM-1 and NKG2D triggering receptors. Thus, while CD48(+) B-EBV and lymphoma B cells devoid of NKG2D and DNAM-1 ligands were resistant to lysis, the preferential usage of these receptors allowed XLP1 NK cells to kill lymphomas that expressed sufficient amounts of the specific ligands. The study sheds new light on the XLP1 immunological defect and on the cross-talk of inhibitory 2B4 with triggering NK (and T) receptors.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Células Asesinas Naturales/inmunología , Trastornos Linfoproliferativos/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Receptores Inmunológicos/inmunología , Transducción de Señal/inmunología , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/genética , Antígeno CD48 , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/patología , Femenino , Herpesvirus Humano 4/inmunología , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Células Asesinas Naturales/patología , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/patología , Masculino , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Receptores Inmunológicos/genética , Transducción de Señal/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria
9.
J Immunol ; 190(10): 5321-8, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23576682

RESUMEN

In this study, we show that neuroblastoma (NB) cell conditioning affects the chemokine receptor repertoire of human resting NK cells. In particular, NB cells upregulated the expression of CXCR4 and CXCR3 in all NK cells and downregulated CX3CR1 in the CD56(dim) subset. On the contrary, the expression of CXCR1 and CCR7 remained unaltered. The phenomenon was dependent on the release by NB cells of TGF-ß1, and rTGF-ß1 induced a chemokine receptor repertoire identical to that of NB-conditioned NK cells. The immune modulatory role of TGF-ß1 appears to be dose dependent because low amounts of the cytokine were sufficient to modulate CXCR4 and CX3CR1 expression, intermediate amounts modified that of CXCR3, and high amounts were necessary to downregulate the expression of the NKp30 activating receptor. Notably, a similar receptor modulation was observed in rTGF-ß2-conditioned NK cells. Finally, the analysis of NK cells from patients with stage 4 NB suggests that NB conditioning could exert in vivo an immune modulatory effect resembling that emerged from in vitro experiments. Altogether our data propose a novel tumor escape-mechanism based on the modulation of chemokine receptors that play pivotal roles in NK cells bone marrow homing, egress, or recruitment into peripheral tissues.


Asunto(s)
Células Asesinas Naturales/metabolismo , Neuroblastoma/inmunología , Neuroblastoma/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Escape del Tumor , Antígeno CD56 , Receptor 1 de Quimiocinas CX3C , Línea Celular Tumoral , Niño , Humanos , Receptor 3 Gatillante de la Citotoxidad Natural/biosíntesis , Receptores CCR7/biosíntesis , Receptores CXCR3/biosíntesis , Receptores CXCR4/biosíntesis , Receptores de Quimiocina/biosíntesis , Receptores de Interleucina-8A/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes/metabolismo , Regulación hacia Arriba
10.
J Allergy Clin Immunol ; 134(6): 1381-1387.e7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24985396

RESUMEN

BACKGROUND: Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening, heterogeneous, hyperinflammmatory disorder. Prompt identification of inherited forms resulting from mutation in genes involved in cellular cytotoxicity can be crucial. X-linked lymphoproliferative disease 1 (XLP1), due to mutations in SH2D1A (Xq25) encoding signaling lymphocyte activation molecule-associated protein (SAP), may present with HLH. Defective SAP induces paradoxical inhibitory function of the 2B4 coreceptor and impaired natural killer (NK) (and T) cell response against EBV-infected cells. OBJECTIVE: To characterize a cohort of patients with HLH and XLP1 for SAP expression and 2B4 function in lymphocytes, proposing a rapid diagnostic screening to direct mutation analysis. METHODS: We set up rapid assays for 2B4 function (degranulation or (51)Cr-release) to be combined with intracellular SAP expression in peripheral blood NK cells. We studied 12 patients with confirmed mutation in SH2D1A and some family members. RESULTS: The combined phenotypic/functional assays allowed efficient and complete diagnostic evaluation of all patients with XLP1, thus directing mutation analysis and treatment. Nine cases were SAP(-), 2 expressed SAP with mean relative fluorescence intensity values below the range of healthy controls (SAP(dull)), and 1, carrying the R55L mutation, was SAP(+). NK cells from all patients showed inhibitory 2B4 function and defective killing of B-EBV cells. Carriers with SH2D1A mutations abolishing SAP expression and low percentage of SAP(+) cells showed neutral 2B4 function at the polyclonal NK cell level. Three novel SH2D1A mutations have been identified. CONCLUSIONS: Study of SAP expression is specific but may have insufficient sensitivity for screening XLP1 as a single tool. Combination with 2B4 functional assay allows identification of all cases.


Asunto(s)
Antígenos CD/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Linfohistiocitosis Hemofagocítica/diagnóstico , Trastornos Linfoproliferativos/diagnóstico , Receptores Inmunológicos/inmunología , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Péptidos y Proteínas de Señalización Intracelular/genética , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/inmunología , Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/inmunología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/inmunología , Masculino , Mutación , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Adulto Joven
11.
J Allergy Clin Immunol ; 143(6): 2296-2299, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30771411
12.
Haematologica ; 99(3): 448-57, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24179150

RESUMEN

Decidual natural killer cells accumulate at the fetal-maternal interface and play a key role in a successful pregnancy. However, their origin is still unknown. Do they derive from peripheral natural killer cells recruited in decidua or do they represent a distinct population that originates in situ? Here, we identified natural killer precursors in decidua and uterus of pregnant mice. These precursors underwent rapid in situ differentiation and large proportions of proliferating immature natural killer cells were present in decidua and uterus as early as gestation day 4.5. Here, we investigated the origin of decidua- and uterus-natural killer cells by performing transfer experiments of peripheral mature natural killer cells or precursors from EGFP(+) mice. Results showed that mature natural killer cells did not migrate into decidua and uterus, while precursors were recruited in these organs and differentiated towards natural killer cells. Moreover, decidua- and uterus-natural killer cells displayed unique phenotypic and functional features. They expressed high levels of the activating Ly49D receptor in spite of their immature phenotype. In addition, decidua- and uterus-natural killer cells were poorly cytolytic and produced low amounts of IFN-γ, while they released factors (GM-CSF, VEGF, IP-10) involved in neo-angiogenesis and tissue remodeling. Our data reveal in situ generation of decidual natural killer cells and provide an important correlation between mouse and human decidual natural killer cells, allowing further studies to be carried out on their role in pregnancy-related diseases.


Asunto(s)
Diferenciación Celular , Decidua/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Animales , Antígenos de Superficie/metabolismo , Proliferación Celular , Citotoxicidad Inmunológica , Decidua/inmunología , Decidua/metabolismo , Femenino , Hematopoyesis , Inmunomodulación , Inmunofenotipificación , Células Asesinas Naturales/inmunología , Ratones , Ratones Transgénicos , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Embarazo , Útero/citología , Útero/inmunología
13.
Eur J Immunol ; 42(6): 1618-26, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22678914

RESUMEN

IL-18 is a proinflammatory cytokine belonging to the "IL-1 family" that has been shown to play a prominent role in the induction of type 1 immune responses. Here, we show that M-CSF induces the expression of a membrane-bound form of IL-18 (mIL-18) in a subset of human blood monocytes differentiating toward macrophages. While monocytes, DC, and GM-CSF-treated monocytes did not express mIL-18, its expression was detected in approximately 30-40% of M-CSF-primed macrophages differentiating from both CD16(-) and CD16(+) monocytes. Treatment with the caspase-1 inhibitor significantly reduced mIL-18 expression suggesting the requirement of an assembled inflammasome for IL-18 surface expression. Polarization toward M2 did not modify mIL-18 expression. On the contrary, LPS stimulation of both M0 and M2 (mIL-18(+) ) macrophages induced shedding of mIL-18, which was likely mediated by the activation of cellular protease(s). Importantly, the soluble form IL-18 (sIL-18) induced in autologous resting NK cells both the expression of CCR7 and the production of high amounts of IFN-γ, which was virtually abrogated by Ab-mediated neutralization of sIL-18. Overall our data shed new light on the cells and mechanisms leading to the release of sIL-18, the major IFN-γ-inducing factor in both physiological and pathological immune responses.


Asunto(s)
Diferenciación Celular , Interleucina-18/fisiología , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Monocitos/efectos de los fármacos , Caspasa 1/fisiología , Células Cultivadas , Proteínas Ligadas a GPI/análisis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interferón gamma/biosíntesis , Interleucina-18/análisis , Macrófagos/química , Monocitos/química , Monocitos/citología , Receptores de IgG/análisis
14.
Blood ; 118(22): 5840-50, 2011 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21989990

RESUMEN

Soluble HLA-G (sHLA-G) inhibits natural killer (NK) cell functions. Here, we investigated sHLA-G-mediated modulation of (1) chemokine receptor and NK receptor expression and function and (2) cytokine and chemokine secretion in CD56bright and CD56dim NK cells. sHLA-G-treated or untreated peripheral blood (PB) and tonsil NK cells were analyzed for chemokine receptor and NK receptor expression by flow cytometry. sHLA-G down-modulated (1) CXCR3 on PB and tonsil CD56bright and CD56dim, (2) CCR2 on PB and tonsil CD56bright, (3) CX3CR1 on PB CD56dim, (4) CXCR5 on tonsil CD56dim, and (5) CD94/NKG2A on PB and tonsil CD56brigh) and CD56dim NK cells. Such sHLA-G-mediated down-modulations were reverted by adding anti-HLA-G or anti-ILT2 mAbs. sHLA-G inhibited chemotaxis of (1) PB NK cells toward CXCL10, CXCL11, and CX3CL1 and (2) PB CD56bright NK cells toward CCL2 and CXCL10. IFN-γ secretion induced by NKp46 engagement was inhibited by NKG2A engagement in untreated but not in sHLA-G-treated NK cells. sHLA-G up-regulated secretion of (1) CCL22 in CD56bright and CD56dim and (2) CCL2, CCL8, and CXCL2-CXCL3 in CD56dim PB NK cells. Signal transduction experiments showed sHLA-G-mediated down-modulation of Stat5 phosphorylation in PB NK cells. In conclusion, our data delineated novel mechanisms of sHLA-G-mediated inhibition of NK-cell functions.


Asunto(s)
Antígeno CD56/metabolismo , Quimiocinas/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Citocinas/metabolismo , Antígenos HLA-G/farmacología , Células Asesinas Naturales/efectos de los fármacos , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Subfamília D de Receptores Similares a Lectina de las Células NK/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Citometría de Flujo , Antígenos HLA-G/química , Humanos , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/fisiología , Subfamília C de Receptores Similares a Lectina de Células NK/fisiología , Subfamília D de Receptores Similares a Lectina de las Células NK/fisiología , Concentración Osmolar , Isoformas de Proteínas/farmacología , Solubilidad
15.
Proc Natl Acad Sci U S A ; 107(50): 21659-64, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21118979

RESUMEN

The cross-talk among cells of the innate immunity can greatly affect both innate and adaptive responses. Here we analyzed the molecular interactions between human natural killer (NK) cells and autologous macrophages. Activated NK cells killed M0 and M2, whereas M1 macrophages were more resistant to lysis because of their higher expression of HLA class I molecules. Following exposure to LPS or bacillus Calmette-Guérin, M0 and M2, but not polarized (endotoxin tolerant) M1 macrophages, induced strong activation of resting NK cells. The expression of CD69 and CD25 activation markers and the acquisition of cytotoxicity against tumor cells and immature dendritic cells required soluble factors being mostly contact independent. On the contrary, IFN-γ production was contact dependent and required the interaction of DNAM-1 and 2B4 (on NK) with their ligands on macrophages as well as IL-18. IL-18 was involved also in the acquisition of CCR7 by NK cells. Interestingly, M0 and M2 cells expressed a membrane-bound form of IL-18, which was released in small amounts after LPS treatment. Our data indicate that, upon interaction with M0 macrophages exposed to microbial products, NK cells may amplify classical type 1 immune responses. In addition, M1-polarizing stimuli can rescue M2 macrophages from their immunomodulatory state and shape their functional behavior toward NK stimulatory capability.


Asunto(s)
Polaridad Celular , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Células Cultivadas , Técnicas de Cocultivo , Citocinas/inmunología , Genes MHC Clase I , Humanos , Subunidad alfa del Receptor de Interleucina-2/inmunología , Células K562 , Células Asesinas Naturales/citología , Lectinas Tipo C/inmunología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Activación de Linfocitos/inmunología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Receptores CCR7/inmunología
16.
Cells ; 12(6)2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36980226

RESUMEN

Over the past decade, immunotherapy has represented an enormous step forward in the fight against cancer. Immunotherapeutic approaches have increasingly become a fundamental part of the combined therapies currently adopted in the treatment of patients with high-risk (HR) neuroblastoma (NB). An increasing number of studies focus on the understanding of the immune landscape in NB and, since this tumor expresses low or null levels of MHC class I, on the development of new strategies aimed at enhancing innate immunity, especially Natural Killer (NK) cells and macrophages. There is growing evidence that, within the NB tumor microenvironment (TME), tumor-associated macrophages (TAMs), which mainly present an M2-like phenotype, have a crucial role in mediating NB development and immune evasion, and they have been correlated to poor clinical outcomes. Importantly, TAM can also impair the antibody-dependent cellular cytotoxicity (ADCC) mediated by NK cells upon the administration of anti-GD2 monoclonal antibodies (mAbs), the current standard immunotherapy for HR-NB patients. This review deals with the main mechanisms regulating the crosstalk among NB cells and TAMs or other cellular components of the TME, which support tumor development and induce drug resistance. Furthermore, we will address the most recent strategies aimed at limiting the number of pro-tumoral macrophages within the TME, reprogramming the TAMs functional state, thus enhancing NK cell functions. We also prospectively discuss new or unexplored aspects of human macrophage heterogeneity.


Asunto(s)
Antineoplásicos , Neuroblastoma , Humanos , Monocitos , Neuroblastoma/patología , Células Asesinas Naturales , Antineoplásicos/farmacología , Macrófagos/patología , Microambiente Tumoral
17.
Cancers (Basel) ; 15(13)2023 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-37444389

RESUMEN

B7-H3 is a 4Ig transmembrane protein that emerged as a tumor-associated antigen in neuroblastoma. It belongs to the B7 family, shows an immunoregulatory role toward NK and T cells, and, therefore, has been included in the growing family of immune checkpoints. Besides neuroblastoma, B7-H3 is expressed by many pediatric cancers including tumors of the central nervous system, sarcomas, and acute myeloid leukemia. In children, particularly those affected by solid tumors, the therapeutic protocols are aggressive and cause important life-threatening side effects. Moreover, despite the improved survival observed in the last decade, a relevant number of patients show therapy resistance and fatal relapses. Immunotherapy represents a new frontier in the cure of cancer patients and the targeting of tumor antigens or immune checkpoints blockade showed exciting results in adults. In this encouraging scenario, researchers and clinicians are exploring the possibility to use immunotherapeutics targeting B7-H3; these include mAbs and chimeric antigen receptor T-cells (CAR-T). These tools are rapidly evolving to improve the efficacy and decrease the unwanted side effects; drug-conjugated mAbs, bi-tri-specific mAbs or CAR-T, and, very recently, NK cell engagers (NKCE), tetra-specific molecules engaging a tumor-associated antigen and NK cells, have been generated. Preclinical data are promising, and clinical trials are ongoing. Hopefully, the B7-H3 targeting will provide important benefits to cancer patients.

18.
J Immunol ; 185(1): 433-41, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20525888

RESUMEN

In humans, recent clinical and experimental data from hematopoietic stem cell transplantation revealed that donor-derived alloreactive NK cells exert a beneficial graft versus leukemia effect. The existence of donor-derived alloreactive NK cells can be predicted on the basis of donor killer cell Ig-like receptor (KIR) gene profile and HLA class I typing of both donor and recipient. Moreover, the size of the alloreactive NK cell population can be directly assessed by the combined use of anti-KIR-specific mAb. In this study, in an attempt to improve the definition of alloreactive NK cell subsets, we assessed the KIR genotype and phenotype in a cohort of 44 donors. This approach allowed the identification of two different KIR2DL3 alleles (KIR2DL3*005 and the novel allele KIR2DL3*015) that did not react with the anti-KIR2DL3-specific ECM41 mAb. In contrast, both alleles were recognized at the cell surface by several mAb reacting with KIR2DL2/L3/S2. Notably, KIR2DL3*005 was also stained by the anti-KIR2DL1/S1-specific EB6B and 11PB6 mAb. Functional analysis revealed that, despite its particular mAb reactivity, the specificity of KIR2DL3*005 for HLA-C molecules did not differ from that of other KIR2DL2/L3 alleles. Finally, site-directed mutagenesis demonstrated that glutamine at position 35 is required for ECM41 staining, whereas glutamic acid 35 and arginine 50 are relevant for staining with EB6B or 11PB6 mAb. Our present data represent a substantial progress in the characterization of the NK cell repertoire and an improved phenotypic/functional definition of given KIR(+) subsets.


Asunto(s)
Alelos , Anticuerpos Monoclonales/metabolismo , Inmunofenotipificación , Receptores KIR2DL3/genética , Receptores KIR2DL3/inmunología , Análisis de Secuencia de Proteína , Aminoácidos/metabolismo , Reacciones Antígeno-Anticuerpo/genética , Línea Celular , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/inmunología , Citotoxicidad Inmunológica/genética , Perfilación de la Expresión Génica/métodos , Genotipo , Antígenos HLA-C/genética , Antígenos HLA-C/metabolismo , Humanos , Inmunofenotipificación/métodos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Mutagénesis Sitio-Dirigida , Receptores KIR2DL3/metabolismo , Análisis de Secuencia de Proteína/métodos , Coloración y Etiquetado/métodos
19.
Cell Signal ; 98: 110415, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35870695

RESUMEN

MicroRNAs are involved in the regulation of different functions in immune and non-immune cells. Here we show that miR-24-3p functionally interacts with FASLG mRNA and down-regulates its expression. This interaction occurs in human natural killer cells (NK), leading to the modulation of FasL surface expression. Moreover, miR-24-3p also modulates the mRNA and protein expression of BIM in NK cells. Thus, it likely contributes to the control of both the extrinsic and intrinsic apoptotic pathways. In line with this hypothesis, inhibition of miR-24-3p improves both initiator caspase-8 and effector caspase-3 and -7 activities, increases cell apoptosis, and reduces cell viability. Our data suggest that miR-24-3p can act as a survival factor in NK cells, affecting the FasL-mediated killing of Fas expressing cells and the BIM-dependent cell death. More generally, miR-24-3p may condition the level of cell apoptosis, which increases at the contraction phase of the immune response when the clearance of various expanded effector cells is needed.


Asunto(s)
MicroARNs , Apoptosis/genética , Humanos , Células Asesinas Naturales/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal
20.
Cancers (Basel) ; 14(4)2022 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-35205760

RESUMEN

In recent years, immunotherapy has emerged as a promising novel therapeutic strategy for cancer treatment. In a relevant percentage of patients, however, clinical benefits are lower than expected, pushing researchers to deeply analyze the immune responses against tumors and find more reliable and efficient tools to predict the individual response to therapy. Novel tissue engineering strategies can be adopted to realize in vitro fully humanized matrix-based models, as a compromise between standard two-dimensional (2D) cell cultures and animal tests, which are costly and hardly usable in personalized medicine. In this review, we describe the main mechanisms allowing cancer cells to escape the immune surveillance, which may play a significant role in the failure of immunotherapies. In particular, we discuss the role of the tumor microenvironment (TME) in the establishment of a milieu that greatly favors cancer malignant progression and impact on the interactions with immune cells. Then, we present an overview of the recent in vitro engineered preclinical three-dimensional (3D) models that have been adopted to resemble the interplays between cancer and immune cells and for testing current therapies and immunotherapeutic approaches. Specifically, we focus on 3D hydrogel-based tools based on different types of polymers, discussing the suitability of each of them in reproducing the TME key features based on their intrinsic or tunable characteristics. Finally, we introduce the possibility to combine the 3D models with technological fluid dynamics platforms, reproducing the dynamic complex interactions between tumor cells and immune effectors migrated in situ via the systemic circulation, pointing out the challenges that still have to be overcome for setting more predictive preclinical assays.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA