Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Immunol ; 199(4): 1516-1525, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28701512

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Mesilato de Imatinib/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Pirimidinas/farmacologia , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Diferenciação Celular/efeitos dos fármacos , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/fisiologia , Ativação Linfocitária/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/fisiologia , Monócitos/imunologia , Monócitos/fisiologia , Neuroblastoma/imunologia , Proteína 2 Ligante de Morte Celular Programada 1/genética , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Receptores CCR1/genética , Receptores CCR1/imunologia , Receptores CCR1/metabolismo , Receptores CXCR3/genética , Receptores CXCR3/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
2.
Eur J Immunol ; 47(6): 1051-1061, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28386908

RESUMO

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.


Assuntos
Células Matadoras Naturais/imunologia , Transtornos Linfoproliferativos/imunologia , Transtornos Linfoproliferativos/fisiopatologia , Receptores de Células Matadoras Naturais/imunologia , Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Antígeno CD48/imunologia , Antígeno CD48/metabolismo , Genes MHC Classe I , Humanos , Células Matadoras Naturais/metabolismo , Ativação Linfocitária , Canais de Potássio Corretores do Fluxo de Internalização/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária/metabolismo , Família de Moléculas de Sinalização da Ativação Linfocitária/imunologia
3.
Stem Cells ; 34(7): 1909-21, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27015881

RESUMO

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.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Ativação de Macrófagos , Macrófagos/citologia , Células-Tronco Mesenquimais/citologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Criança , Humanos , Imunomodulação , Interferon gama/metabolismo , Células Matadoras Naturais/metabolismo , Macrófagos/metabolismo , Monócitos/citologia , Linfócitos T Reguladores
4.
Eur J Immunol ; 44(6): 1814-22, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24510590

RESUMO

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.


Assuntos
Tolerância Imunológica , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Proteínas de Neoplasias/imunologia , Neoplasias Ovarianas/imunologia , Receptores Toll-Like/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/imunologia , Citocinas/imunologia , Feminino , Humanos , Imunidade Celular , Células Matadoras Naturais/patologia , Macrófagos/patologia , Pessoa de Meia-Idade , Neoplasias Ovarianas/patologia
5.
J Immunol ; 190(10): 5321-8, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23576682

RESUMO

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.


Assuntos
Células Matadoras Naturais/metabolismo , Neuroblastoma/imunologia , Neuroblastoma/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Evasão Tumoral , Antígeno CD56 , Receptor 1 de Quimiocina CX3C , Linhagem Celular Tumoral , Criança , Humanos , Receptor 3 Desencadeador da Citotoxicidade Natural/biossíntese , Receptores CCR7/biossíntese , Receptores CXCR3/biossíntese , Receptores CXCR4/biossíntese , Receptores de Quimiocinas/biossíntese , Receptores de Interleucina-8A/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Recombinantes/metabolismo , Regulação para Cima
6.
Eur J Immunol ; 42(6): 1618-26, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22678914

RESUMO

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.


Assuntos
Diferenciação Celular , Interleucina-18/fisiologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/citologia , Monócitos/efeitos dos fármacos , Caspase 1/fisiologia , Células Cultivadas , Proteínas Ligadas por GPI/análise , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interferon gama/biossíntese , Interleucina-18/análise , Macrófagos/química , Monócitos/química , Monócitos/citologia , Receptores de IgG/análise
7.
Proc Natl Acad Sci U S A ; 107(50): 21659-64, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21118979

RESUMO

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.


Assuntos
Polaridade Celular , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Células Cultivadas , Técnicas de Cocultura , Citocinas/imunologia , Genes MHC Classe I , Humanos , Subunidade alfa de Receptor de Interleucina-2/imunologia , Células K562 , Células Matadoras Naturais/citologia , Lectinas Tipo C/imunologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/imunologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Receptores CCR7/imunologia
8.
Int Immunol ; 22(2): 91-100, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20008459

RESUMO

To define novel human NK cell markers, we generated two mAbs specific for G-protein-coupled receptor 56 (GPR56), a surface glycoprotein that appears to be involved in cell-to-cell and cell-to-matrix interactions. GPR56 has been described in selected normal tissues, and in certain tumors, while, as yet, its expression on leukocytes is unknown. In this study, we show that anti-GPR56 mAbs, among leukocytes, prevalently recognize NK cells. In particular, these mAbs brightly stain CD56(dull) CD16(+) NK cells while react poorly with CD56(bright) CD16(+/-) NK cells. Consistently, we found that GPR56 was expressed on NK cells populating inflamed peripheral tissues while it was absent in lymph node-derived NK cells. We also show that activating stimuli, such as cytokines or exposure to monocyte-derived dendritic cell, down-regulate NK cell expression of GPR56 both at the protein and at the transcriptional level. Interestingly, IL-18, known to induce de novo expression of CCR7 on CD56(dull) CD16(+) NK cells, displayed the highest capability of modulating GPR56. Thus, together with the identification of GPR56 as a novel marker capable of discriminating different NK cells subsets, our data suggest that GPR56 may take part to the mechanisms regulating NK cell migration through the blood stream, peripheral tissues and lymph nodes.


Assuntos
Antígeno CD56/análise , Inflamação/metabolismo , Células Matadoras Naturais/metabolismo , Subpopulações de Linfócitos/imunologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de IgG/análise , Anticorpos Monoclonais , Biomarcadores/metabolismo , Células Cultivadas , Células Dendríticas/imunologia , Proteínas Ligadas por GPI , Humanos , Inflamação/imunologia , Interleucinas/metabolismo , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Subpopulações de Linfócitos/metabolismo , Receptores Acoplados a Proteínas G/sangue , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/imunologia , Transfecção
9.
J Immunol ; 182(6): 3530-9, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19265131

RESUMO

In this study, cancer cells were isolated from tumor specimens of nine glioblastoma patients. Glioblastoma cells, cultured under suitable culture conditions, displayed markers typical of neural stem cells, were capable of partial multilineage differentiation in vitro, and gave origin to infiltrating tumors when orthotopically injected in NOD/SCID mice. These cells, although resistant to freshly isolated NK cells, were highly susceptible to lysis mediated by both allogeneic and autologous IL-2 (or IL-15)-activated NK cells. Indeed, all stem cell-cultured glioblastoma cells analyzed did not express protective amounts of HLA class I molecules, while expressing various ligands of activating NK receptors that triggered optimal NK cell cytotoxicity. Importantly, glioblastoma stem cells expressed high levels of PVR and Nectin-2, the ligands of DNAM-1-activating NK receptor.


Assuntos
Citotoxicidade Imunológica , Glioblastoma/imunologia , Glioblastoma/patologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , Animais , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Glioblastoma/metabolismo , Humanos , Imunidade Inata , Células Matadoras Naturais/metabolismo , Ligantes , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Linfócitos do Interstício Tumoral/transplante , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Receptores de Células Matadoras Naturais/biossíntese , Receptores de Células Matadoras Naturais/genética , Receptores de Células Matadoras Naturais/fisiologia , Células Tumorais Cultivadas
10.
Cancers (Basel) ; 11(1)2019 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-30641867

RESUMO

TGF-ß1 is a pleiotropic factor exerting a strong regulatory role in several cell types, including immune cells. In NK cells it profoundly alters the surface expression of crucial activating and chemokine receptors. To understand which soluble signals might better contrast these effects, we cultured human NK cells in the presence of TGF-ß1 and different innate and adaptive cytokines, generally referred as "immunostimulatory". These included IL-2, IL-15, IL-21, IL-27, and IL-18. Unexpectedly, IL-18 strengthened rather than contrasting important TGF-ß1-mediated functions. In particular, IL-18 further reduced the expression of CX3CR1 and NKp30, leading to the virtual abrogation of the triggering capability of this activating receptor. Moreover, IL-18 further increased the expression of CXCR4. The IL-18-mediated additive effect on NKp30 and CXCR4 expression involved transcriptional regulation and activation of MEK/ERK and/or p38MAPK. A proteomic approach quantified both surface and intracellular proteins significantly modified in cytokine-treated NK cells, thus giving global information on the biological processes involving TGF-ß1 and IL-18. Our data support the concept that IL-18 may have a different behavior depending on the type of soluble factors characterizing the microenvironment. In a TGF-ß1 rich milieu such as tumors, it may contribute to the impairment of both NK cells recruitment and killing capability.

11.
Front Immunol ; 9: 2324, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30364222

RESUMO

A large body of data shows that Natural Killer (NK) cells are immune effectors exerting a potent cytolytic activity against tumors and virus infected cells. The discovery and characterization of several inhibitory and activating receptors unveiled most of the mechanisms allowing NK cells to spare healthy cells while selectively attacking abnormal tissues. Nevertheless, the mechanisms ruling NK cell subset recirculation among the different compartments of human body have only lately started to be investigated. This is particularly true for pathological settings such as tumors or infected tissues but also for para-physiological condition like pregnant human uterine mucosa. It is becoming evident that the microenvironment associated to a particular clinical condition can deeply influence the migratory capabilities of NK cells. In this review we describe the main mechanisms and stimuli known to regulate the expression of chemokine receptors and other molecules involved in NK cell homing to either normal or pathological/inflamed tissues, including tumors or organs such as lung and liver. We will also discuss the role played by the chemokine/chemokine receptor axes in the orchestration of physiological events such as NK cell differentiation, lymphoid organ retention/egress and recruitment to decidua during pregnancy.


Assuntos
Movimento Celular/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Animais , Movimento Celular/genética , Citocinas/metabolismo , Citotoxicidade Imunológica , Feminino , Humanos , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Especificidade de Órgãos/imunologia , Gravidez
12.
Oncotarget ; 8(21): 35088-35102, 2017 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-28456791

RESUMO

Angiogenesis represents a hallmark of tumor progression in Multiple Myeloma (MM), a still incurable malignancy. Here we analyzed the activity of cytokine-stimulated NK cells against tumor-associated endothelial cells isolated from bone marrow aspirates of MM patients with active disease (MMECs). We show that NK cells activated with optimal doses of IL-15 killed MMECs thanks to the concerted action of multiple activating receptors. In particular, according to the high expression of PVR and Nectin-2 on MMECs, DNAM-1 actively participated in target recognition. Interestingly, in MMECs the surface density of PVR was significantly higher than that detected in endothelium from patients with MM in complete remission or with monoclonal gammopathy of undetermined significance (MGUS). Importantly, IL-27, which unlike IL-15 does not display pro-angiogenic properties, maintained or increased the NK cell functions induced by suboptimal concentrations of IL-15. NK cell properties included killing of MMECs, IFN-γ production as well as a peculiar increase of NKp46 expression on NK cell surface. Finally, IL-27 showed a striking capability of up-regulating the expression of PD-L2 and HLA-I on tumor endothelium, whereas it did not modify that of PD-L1 and HLA-II.Our results suggest that cytokine-activated endogenous or adoptively transferred NK cells might support conventional therapies improving the outcome of MM patients.


Assuntos
Células Endoteliais/efeitos dos fármacos , Interleucinas/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Mieloma Múltiplo/imunologia , Idoso , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Humanos , Interleucina-15/farmacologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/metabolismo , Neovascularização Patológica , Receptores Virais/metabolismo
13.
Oncoimmunology ; 5(1): e1064578, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26942080

RESUMO

The prognosis of high-risk neuroblastoma (NB) remains poor, although immunotherapies with anti-GD2 antibodies have been reported to provide some benefit. Immunotherapies can be associated with an IFNγ storm that induces in tumor cells the "adaptive immune resistance" characterized by the de-novo expression of Programmed Death Ligands (PD-Ls). Tumor cells can also constitutively express PD-Ls in response to oncogenic signaling. Here, we analyze the constitutive and the inducible surface expression of PD-Ls in NB cells. We show that virtually all HLA class Ipos NB cell lines constitutively express PD-L1, whereas PD-L2 is rarely detected. IFNγ upregulates or induces PD-L1 both in NB cell lines in vitro and in NB engrafted nude/nude mice. Importantly, after IFNγ stimulation PD-L1 can be acquired by NB cell lines, as well as by metastatic neuroblasts isolated from bone marrow aspirates of high-risk NB patients, characterized by different MYCN amplification status. Interestingly, in one patient NB cells were poorly responsive to IFNγ stimulation, pointing out that responsiveness to IFNγ might represent a further element of heterogeneity in metastatic neuroblasts. Finally, we document the presence of lymphocytes expressing the PD-1 receptor in NB-infiltrated bone marrow of patients. PD-1pos cells are mainly represented by αß T cells, but also include small populations of γδ T cells and NK cells. Moreover, PD-1pos T cells have a higher expression of activation markers. Overall, our data show that a PD-L1-mediated immune resistance mechanism occurs in metastatic neuroblasts and provide a biological rationale for blocking the PD-1/PD-Ls axis in future combined immunotherapeutic approaches.

14.
Biofactors ; 42(1): 80-92, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26669587

RESUMO

Macrophages play a crucial role in atherosclerosis progression. Classically activated M1 macrophages have been found in rupture-prone atherosclerotic plaques whereas alternatively activated macrophages, M2, localize in stable plaque. Macrophage accumulation of cholesterol and of its oxidized derivatives (oxysterols) leads to the formation of foam cells, a hallmark of atherosclerotic lesions. In this study, the effects of oxysterols in determining the functional polarization of human macrophages were investigated. Monocytes, purified from peripheral blood mononuclear cells of healthy donors, were differentiated into macrophages (M0) and treated with an oxysterol mixture, cholesterol, or ethanol, every 4 H for a total of 4, 8, and 12 H. The administration of the compounds was repeated in order to maintain the levels of oxysterols constant throughout the treatment. Compared with ethanol treatment, the oxysterol mixture decreased the surface expression of CD36 and CD204 scavenger receptors and reduced the amount of reactive oxygen species whereas it did not affect either cell viability or matrix metalloprotease-9 activity. Moreover, the oxysterol mixture increased the expression of both liver X receptor α and ATP-binding cassette transporter 1. An enhanced secretion of the immunoregulatory cytokine IL-10 accompanied these events. The results supported the hypothesis that the constant levels of oxysterols and, in particular, of 27-hydroxycholesterol stimulate macrophage polarization toward the M2 immunomodulatory functional phenotype, contributing to the stabilization of atherosclerotic plaques.


Assuntos
Aterosclerose/tratamento farmacológico , Hidroxicolesteróis/administração & dosagem , Macrófagos/efeitos dos fármacos , Placa Aterosclerótica/tratamento farmacológico , Transportadores de Cassetes de Ligação de ATP/biossíntese , Aterosclerose/metabolismo , Aterosclerose/patologia , Antígenos CD36/metabolismo , Polaridade Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colesterol/administração & dosagem , Colesterol/metabolismo , Etanol/administração & dosagem , Humanos , Interleucina-10/metabolismo , Receptores X do Fígado , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/biossíntese , Monócitos/efeitos dos fármacos , Receptores Nucleares Órfãos/biossíntese , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Espécies Reativas de Oxigênio/metabolismo , Receptores Depuradores Classe A/metabolismo
15.
Front Immunol ; 5: 56, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24575100

RESUMO

Neuroblastoma (NB) is the most common extra-cranial solid tumor of childhood and arises from developing sympathetic nervous system. Most primary tumors localize in the abdomen, the adrenal gland, or lumbar sympathetic ganglia. Amplification in tumor cells of MYCN, the major oncogenic driver, patients' age over 18 months, and the presence at diagnosis of a metastatic disease (stage IV, M) identify NB at high risk of treatment failure. Conventional therapies did not significantly improve the overall survival of these patients. Moreover, the limited landscape of somatic mutations detected in NB is hampering the development of novel pharmacological approaches. Major efforts aim to identify novel NB-associated surface molecules that activate immune responses and/or direct drugs to tumor cells and tumor-associated vessels. PVR (Poliovirus Receptor) and B7-H3 are promising targets, since they are expressed by most high-risk NB, are upregulated in tumor vasculature and are essential for tumor survival/invasiveness. PVR is a ligand of DNAM-1 activating receptor that triggers the cytolytic activity of natural killer (NK) cells against NB. In animal models, targeting of PVR with an attenuated oncolytic poliovirus induced tumor regression and elimination. Also B7-H3 was successfully targeted in preclinical studies and is now being tested in phase I/II clinical trials. B7-H3 down-regulates NK cytotoxicity, providing NB with a mechanism of escape from immune response. The immunosuppressive potential of NB can be enhanced by the release of soluble factors that impair NK cell function and/or recruitment. Among these, TGF-ß1 modulates the cytotoxicity receptors and the chemokine receptor repertoire of NK cells. Here, we summarize the current knowledge on the main cell surface molecules and soluble mediators that modulate the function of NK cells in NB, considering the pros and cons that must be taken into account in the design of novel NK cell-based immunotherapeutic approaches.

16.
Immunol Lett ; 161(2): 168-73, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24361820

RESUMO

In early seventies "natural killer (NK) cells", a third lymphocyte subset was discovered that revealed an unexpected ability to kill syngeneic and allogeneic tumor targets, thus emerging as the most potent non-specific cytotoxic cells in both human and mouse. Decades of research revealed the multifaceted nature of these cells. Now we know that NK cells are highly specific cells able to discriminate between self (which is spared) and non-self (which is attacked). Most of the specific and non HLA-specific surface receptors involved in NK cell recognition and function have been identified and, to date, only few of them still remain orphans. We also know that NK cells contribute to both innate and adaptive immune responses, interact with other immune cell types and release type 1 cytokines and chemokines. Moreover, fundamental data are accumulating on NK cell development and migration under both physiological and pathological conditions. The time is arrived to exploit these cells in the cure of cancer patients. While encouraging results emerged in hematological malignances, the road to treat solid tumors using NK cells is still covered by obstacles that hamper their function and that just begin to be unveiled.


Assuntos
Células Matadoras Naturais/fisiologia , Receptores de Células Matadoras Naturais/metabolismo , Imunidade Adaptativa , Animais , Comunicação Celular , Citotoxicidade Imunológica , Humanos , Imunidade Inata , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , Microambiente Tumoral/imunologia
17.
Eur J Immunol ; 37(11): 3190-6, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17918205

RESUMO

Adoptive cellular immunotherapy has been proposed as an additional treatment of medulloblastoma, an intracranial tumor characterized by a particularly poor prognosis. However, little is known on the ability of the immune system to effectively attack this tumor. In this study, we show that activated human NK cells efficiently kill medulloblastoma cell lines in vitro. NK-mediated killing involved different activating receptors (including NKp46, NKp30, DNAM-1 and NKG2D) and correlated with the presence of their specific ligands on tumor cells. In contrast, the absence of major adhesion interactions, such as LFA-1/ICAM did not impair the NK-mediated cytotoxicity. Medulloblastoma expressed a number of tumor-associated molecules including CD146 and CD133, considered a marker for cancer stem cells. Remarkably, both CD133-positive and CD133-negative cell lines were susceptible to lysis. Tumor cells also expressed molecules that are currently used as diagnostic tools for neuroblastoma cell identification. In particular, B7 homolog 3 (B7-H3) was expressed by all the medulloblastoma cell lines analyzed, while the presence of GD(2) and NB84 was restricted to given cell lines and/or marked a defined tumor cell subset.


Assuntos
Antígenos CD/metabolismo , Citotoxicidade Imunológica , Glicoproteínas/metabolismo , Células Matadoras Naturais/metabolismo , Meduloblastoma/imunologia , Meduloblastoma/metabolismo , Peptídeos/metabolismo , Receptores Imunológicos/metabolismo , Antígeno AC133 , Biomarcadores Tumorais , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Células Matadoras Naturais/imunologia , Ligantes , Receptores Imunológicos/imunologia
18.
Blood ; 108(13): 4078-85, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16931625

RESUMO

While during the first trimester of pregnancy natural killer (NK) cells represent the most abundant lymphocyte population in the decidua, their actual function at this site is still debated. In this study we analyzed NK cells isolated from decidual tissue for their surface phenotype and functional capability. We show that decidual NK (dNK) cells express normal surface levels of certain activating receptors, including NKp46, NKG2D, and 2B4, as well as of killer cell immunoglobulin-like receptors (KIRs) and CD94/NKG2A inhibitory receptor. In addition, they are characterized by high levels of cytoplasmic granules despite their CD56(bright) CD16- surface phenotype. Moreover, we provide evidence that in dNK cells, activating NK receptors display normal triggering capability whereas 2B4 functions as an inhibitory receptor. Thus, cross-linking of 2B4 resulted in inhibition of both cytolytic activity and interferon-gamma (IFN-gamma) production. Clonal analysis revealed that, in the majority of dNK cell clones, the 2B4 inhibitory function is related to the deficient expression of signaling lymphocyte activation molecule (SLAM)-associated protein (SAP) mRNA. Moreover, biochemical analysis revealed low levels of SAP in the dNK polyclonal population. This might suggest that dNK cells, although potentially capable of killing, are inhibited in their function when interacting with cells expressing CD48.


Assuntos
Antígenos CD/imunologia , Antígenos de Diferenciação/imunologia , Decídua/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Gravidez/imunologia , Receptores Imunológicos/imunologia , Antígenos CD/biossíntese , Antígenos de Diferenciação/biossíntese , Comunicação Celular/imunologia , Células Cultivadas , Decídua/citologia , Decídua/embriologia , Feminino , Feto/embriologia , Feto/imunologia , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Humanos , Imunidade Celular/fisiologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Gravidez/metabolismo , Receptores Imunológicos/biossíntese , Vesículas Secretórias/imunologia , Vesículas Secretórias/metabolismo , Família de Moléculas de Sinalização da Ativação Linfocitária
19.
J Immunol ; 174(7): 3992-8, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15778356

RESUMO

In the course of inflammatory responses in peripheral tissues, NK cells may be exposed to cytokines such as IL-12 and IL-4 released by other cell types that may influence their functional activities. In the present study we comparatively analyzed purified human peripheral blood NK cells that had been exposed to either IL-12 or IL-4 during short (overnight) incubation. We show that although IL-12-cultured NK cells produced abundant IFN-gamma, TNF-alpha, and GM-CSF in response to stimuli acting on the NKp46-activating receptor, IL-4-cultured NK cells did not release detectable levels of these cytokines. In contrast, IL-4-cultured NK cells produced significant levels of TNF-alpha and GM-CSF only when stimulated with PMA and ionomycin. In no instance could the production of IL-5 and IL-13 be detected. Importantly, IL-12-cultured, but not IL-4-cultured, NK cells displayed strong cytolytic activity against various tumor cells or immature dendritic cells (DCs). Moreover, only NK cells that had been cultured in IL-12 were able to induce substantial DC maturation. Our data suggest that NK cells exposed to IL-12 for a time interval compatible with in vivo responses may favor the selection of appropriate mature DCs for subsequent Th1 cell priming in secondary lymphoid organs. On the contrary, NK cells exposed to IL-4 do not exert DC selection, may impair efficient Th1 priming, and favor either tolerogenic or Th2-type responses.


Assuntos
Comunicação Celular/imunologia , Células Dendríticas/fisiologia , Interleucina-12/imunologia , Interleucina-4/imunologia , Células Matadoras Naturais/fisiologia , Neoplasias/patologia , Células Cultivadas , Citocinas/biossíntese , Citotoxicidade Imunológica , Humanos , Inflamação , Interleucina-12/fisiologia , Interleucina-4/fisiologia , Células K562 , Ativação Linfocitária , Células Th1 , Células Th2
20.
Blood ; 100(10): 3782-9, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12393650

RESUMO

Human transferrin receptor-2 (TFR-2) is a protein highly homologous to TFR-1/CD71 and is endowed with the ability to bind transferrin (TF) with low affinity. High levels of TFR-2 mRNA were found in the liver and in erythroid precursors. Mutations affecting the TFR-2 gene led to hemochromatosis type 3, a form of inherited iron overload. Several issues on distribution and function of the receptor were answered by raising a panel of 9 monoclonal antibodies specific for TFR-2 by immunizing mice with murine fibroblasts transfected with the human TFR-2 cDNA. A polyclonal antiserum was also produced in mice immunized with 3 peptides derived from the TFR-2 sequence, exploiting an innovative technique. The specificity of all the reagents produced was confirmed by reactivity with TFR-2(+) target cells and simultaneous negativity with TFR-1(+) cells. Western blot analyses showed a dominant chain of approximately 90 kDa in TFR-2 transfectants and HepG2 cell line. Analysis of distribution in normal tissues and in representative cell lines revealed that TFR-2 displays a restricted expression pattern--it is present at high levels in hepatocytes and in the epithelial cells of the small intestine, including the duodenal crypts. Exposure of human TFR-2(+) cells to TF-bound iron is followed by a significant up-regulation and relocalization of membrane TFR-2. The tissue distribution pattern, the behavior following exposure to iron-loaded TF, and the features of the disease resulting from TFR-2 inactivation support the hypothesis that TFR-2 contributes to body iron sensing.


Assuntos
Anticorpos Monoclonais , Receptores da Transferrina/imunologia , Receptores da Transferrina/metabolismo , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Endotélio/química , Hepatócitos/química , Humanos , Soros Imunes/imunologia , Imuno-Histoquímica , Intestinos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Distribuição Tecidual , Transferrina/farmacologia , Células Tumorais Cultivadas , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA