Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Crit Rev Immunol ; 41(2): 45-76, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34348002

RESUMEN

Melanoma is the most aggressive and deadliest form of skin cancer, and its prognosis is very poor. Although the early detection is responsive to many treatments, metastatic melanoma is refractory to most of them. In the United States, skin melanoma is the fifth most common type of cancer in men and the sixth in women. Current treatment modalities, depending on the cancer stage, consist primarily of surgical excision, chemotherapy, adjuvant therapy, targeted therapies, and immunotherapy. Despite the wide range of therapeutic options and the steadily increasing response rates, a large subset of the treated patients relapse and develop resistance to further treatments. One novel approach in preclinical and clinical trials in immunotherapy is the adaptation of natural killer (NK) cells against resistant cancer cells. NK cells can kill a variety of cancer cell types, as well as the cancer stem cells, while leaving normal cells intact. In skin melanoma, as in most cancers, NK cells in the tumor microenvironment (TME) are functionally impaired. Several factors underlie the defective cause of NK cells, one of which is the dysregulation of the activating receptor NKG2D. This is the dominant receptor in regulating the cytotoxic activity, cytokine production, and regulation of other receptors expressed on NK cells and other lymphocytes. The defective NK cells in cancer models were associated with tumor growth and metastasis. In this review, we discuss the role of NK cells and their phenotypic variants in skin melanoma. Using bioinformatics, we have further analyzed the expression of NKG2D, confirming its low transcript levels in patients with skin melanoma. Furthermore, we show that the CD133 subset of cancer stem cells expresses low levels of NKG2D. Based on these findings we discuss the potential therapeutic approaches that can be exploited to upregulate NKG2D in patients' NK cells and restore their anti-melanoma effects, resulting in tumor regression and prolonged survival.


Asunto(s)
Inmunoterapia , Melanoma , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Neoplasias Cutáneas , Humanos , Células Asesinas Naturales , Melanoma/terapia , Neoplasias Cutáneas/terapia , Microambiente Tumoral
2.
Nat Immunol ; 10(9): 973-80, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19648922

RESUMEN

The adaptors SAP, EAT-2 and ERT are specific to cells of the immune system and belong to the SAP family. All three are expressed in natural killer (NK) cells. Here we examined the global function of the SAP family using mice lacking SAP, EAT-2 and ERT. These adaptors acted together in a mechanism that was essential for the elimination of hematopoietic but not nonhematopoietic cells by NK cells. This function was mediated by many receptors of the SLAM family on NK cells that were engaged by ligands found solely on hematopoietic cells. In the absence of SAP-related adaptors, SLAM receptors lost their activating function and became inhibitory receptors that repressed other activating receptors, such as NKG2D. Hence, the SAP family is essential for the elimination of unwanted hematopoietic cells by NK cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Sistema Hematopoyético/citología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Células Asesinas Naturales/fisiología , Animales , Antígenos CD/fisiología , Antígenos Ly/fisiología , Antígeno CD48 , Células CHO , Cricetinae , Cricetulus , Antígenos de Histocompatibilidad Clase I/fisiología , Interferón gamma/biosíntesis , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Ratas , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Factores de Transcripción/fisiología
3.
Immunity ; 33(4): 632-44, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20951068

RESUMEN

Recognition of NKG2D ligands by natural killer (NK) cells plays an important role during antitumoral responses. To address how NKG2D engagement affects intratumoral NK cell dynamics, we performed intravital microscopy in a Rae-1ß-expressing solid tumor. This NKG2D ligand drove NK cell accumulation, activation, and motility within the tumor. NK cells established mainly dynamic contacts with their targets during tumor regression. In sharp contrast, cytotoxic T lymphocytes (CTLs) formed stable contacts in tumors expressing their cognate antigen. Similar behaviors were observed during effector functions in lymph nodes. In vitro, contacts between NK cells and their targets were cytotoxic but did not elicit sustained calcium influx nor adhesion, whereas CTL contact stability was critically dependent on extracellular calcium entry. Altogether, our results offer mechanistic insight into how NK cells and CTLs can exert cytotoxic activity with remarkably different contact dynamics.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Animales , Calcio/metabolismo , Línea Celular Tumoral , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología
4.
J Immunol ; 198(3): 1172-1182, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28031333

RESUMEN

NK cells, which are highly enriched in the liver, are potent regulators of antiviral T cells and immunopathology in persistent viral infection. We investigated the role of the NKG2D axis in T cell/NK cell interactions in hepatitis B. Activated and hepatitis B virus (HBV)-specific T cells, particularly the CD4 fraction, expressed NKG2D ligands (NKG2DL), which were not found on T cells from healthy controls (p < 0.001). NKG2DL-expressing T cells were strikingly enriched within HBV-infected livers compared with the periphery or to healthy livers (p < 0.001). NKG2D+NK cells were also increased and preferentially activated in the HBV-infected liver (p < 0.001), in direct proportion to the percentage of MICA/B-expressing CD4 T cells colocated within freshly isolated liver tissue (p < 0.001). This suggests that NKG2DL induced on T cells within a diseased organ can calibrate NKG2D-dependent activation of local NK cells; furthermore, NKG2D blockade could rescue HBV-specific and MICA/B-expressing T cells from HBV-infected livers. To our knowledge, this is the first ex vivo demonstration that non-virally infected human T cells can express NKG2DL, with implications for stress surveillance by the large number of NKG2D-expressing NK cells sequestered in the liver.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Hepatitis B Crónica/inmunología , Células Asesinas Naturales/inmunología , Hígado/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Adulto , Comunicación Celular , Células Cultivadas , Femenino , Humanos , Ligandos , Hígado/virología , Activación de Linfocitos , Masculino , Subfamilia K de Receptores Similares a Lectina de Células NK/antagonistas & inhibidores
5.
Am J Transplant ; 17(12): 3199-3209, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28805342

RESUMEN

It has already been shown that neutralization of the activating NK cell receptor NKG2D in combination with co-stimulation blockade prolongs graft survival of vascularized transplants. In order to clarify the underlying cellular mechanisms, we transplanted complete MHC-disparate BALB/c-derived cardiac grafts into C57BL/6 wildtypes or mice deficient for NKG2D (Klrk1-/- ). Although median survival was 8 days for both recipient groups, we detected already at day 5 posttransplantation significantly greater intragraft frequencies of NKp46+ NK cells in Klrk1-/- recipients than in wildtypes. This was followed by a significantly greater infiltration of CD4+ , but a lesser infiltration of CD8+ T cell frequencies. Contrary to published observations, co-stimulation blockade with CTLA4-Ig resulted in a significant acceleration of cardiac rejection by Klrk1-/- recipients, and this result was confirmed by applying a neutralizing antibody against NKG2D to wildtypes. In both experimental setups, grafts derived from Klrk1-/- recipients were characterized by significantly higher levels of interferon-γ mRNA, and both CD4+ and CD8+ T cells displayed a greater capacity for degranulation and interferon-γ production. In summary, our results clearly illustrate that NKG2D expression in the recipient is important for cardiac allograft survival, thus supporting the hypothesis that impairment of NK cells prevents the establishment of graft acceptance.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Rechazo de Injerto/etiología , Supervivencia de Injerto/inmunología , Trasplante de Corazón/efectos adversos , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Animales , Rechazo de Injerto/metabolismo , Rechazo de Injerto/patología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Complicaciones Posoperatorias , Tasa de Supervivencia , Trasplante Homólogo
6.
Cancer Immunol Immunother ; 65(3): 355-66, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26883876

RESUMEN

Natural killer (NK) cells are the primary effectors of the innate immune response against virus-infected cells or cells that have undergone malignant transformation. NK cells recognize their targets through a complex array of activating and inhibitory receptors, which regulate the intensity of the effector response against individual target cells. However, many studies have shown that tumor cells can escape immune cell recognition through a variety of mechanisms, developing resistance to NK cell killing. Using a lentiviral shRNA library, we previously demonstrated that several common signaling pathways modulate susceptibility of tumor cells to NK cell activity. In this study, we focused on one of the genes (PI3KCB), identified in this genetic screen. The PI3KCB gene encodes an isoform of the catalytic subunit of PI3K called P110ß. The PI3K pathway has been linked to diverse cellular functions, but has never been associated with susceptibility to NK cell activity. Gene silencing of PI3KCB resulted in increased susceptibility of several tumor cell lines to NK cell lytic activity and induced increased IFN-γ secretion by NK cells. Treatment of primary tumor cells with two different PI3K inhibitors also increased target cell susceptibility to NK cell activity. These effects are due, at least in part, to modulation of several activating and inhibitory ligands and appear to be correlated with PI3K signaling pathway inhibition. These findings identify a new and important role of PI3KCB in modulating tumor cell susceptibility to NK cells and open the way to future combined target immunotherapies.


Asunto(s)
Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal/fisiología , Antígenos de Diferenciación de Linfocitos T/fisiología , Línea Celular Tumoral , Humanos , Inmunoterapia , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Neoplasias/inmunología , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3
7.
PLoS Pathog ; 10(5): e1004058, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24787765

RESUMEN

NKG2D plays a major role in controlling immune responses through the regulation of natural killer (NK) cells, αß and γδ T-cell function. This activating receptor recognizes eight distinct ligands (the MHC Class I polypeptide-related sequences (MIC) A andB, and UL16-binding proteins (ULBP)1-6) induced by cellular stress to promote recognition cells perturbed by malignant transformation or microbial infection. Studies into human cytomegalovirus (HCMV) have aided both the identification and characterization of NKG2D ligands (NKG2DLs). HCMV immediate early (IE) gene up regulates NKGDLs, and we now describe the differential activation of ULBP2 and MICA/B by IE1 and IE2 respectively. Despite activation by IE functions, HCMV effectively suppressed cell surface expression of NKGDLs through both the early and late phases of infection. The immune evasion functions UL16, UL142, and microRNA(miR)-UL112 are known to target NKG2DLs. While infection with a UL16 deletion mutant caused the expected increase in MICB and ULBP2 cell surface expression, deletion of UL142 did not have a similar impact on its target, MICA. We therefore performed a systematic screen of the viral genome to search of addition functions that targeted MICA. US18 and US20 were identified as novel NK cell evasion functions capable of acting independently to promote MICA degradation by lysosomal degradation. The most dramatic effect on MICA expression was achieved when US18 and US20 acted in concert. US18 and US20 are the first members of the US12 gene family to have been assigned a function. The US12 family has 10 members encoded sequentially through US12-US21; a genetic arrangement, which is suggestive of an 'accordion' expansion of an ancestral gene in response to a selective pressure. This expansion must have be an ancient event as the whole family is conserved across simian cytomegaloviruses from old world monkeys. The evolutionary benefit bestowed by the combinatorial effect of US18 and US20 on MICA may have contributed to sustaining the US12 gene family.


Asunto(s)
Citomegalovirus , Antígenos de Histocompatibilidad Clase I/metabolismo , Evasión Inmune , Células Asesinas Naturales/inmunología , Lisosomas/metabolismo , Proteolisis , Proteínas Virales/fisiología , Adulto , Proteínas Bacterianas/metabolismo , Células Cultivadas , Citomegalovirus/inmunología , Citomegalovirus/patogenicidad , Inhibidores Enzimáticos/farmacología , Humanos , Evasión Inmune/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Leupeptinas/farmacología , Proteínas Luminiscentes/metabolismo , Lisosomas/efectos de los fármacos , Macrólidos/farmacología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Proteolisis/efectos de los fármacos , Proteínas Recombinantes/metabolismo
8.
Pediatr Blood Cancer ; 63(12): 2230-2239, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27433920

RESUMEN

BACKGROUND: Neuroblastoma (NB) is the most common solid extracranial tumor in childhood. Despite advances in therapy, the prognosis is poor and optimized therapies are urgently needed. Therefore, we investigated the antitumor potential of interleukin-15 (IL-15)-activated cytokine-induced killer (CIK) cells against different NB cell lines. PROCEDURE: CIK cells were generated from peripheral blood mononuclear cells by the stimulation with interferon-γ (IFN-γ), IL-2, OKT-3 and IL-15 over a period of 10-12 days. The cytotoxic activity against NB cells was analyzed by nonradioactive Europium release assay before and after blocking of different receptor-ligand interactions relevant in CIK cell-mediated cytotoxicity. RESULTS: The final CIK cell products consisted in median of 83% (range: 75.9-91.9%) CD3+ CD56- T cells, 14% (range: 5.2-20.7%) CD3+ CD56+ NK-like T cells and 2% (range: 0.9-4.8%) CD3- CD56+ NK cells. CIK cells expanded significantly upon ex vivo stimulation with median rates of 22.3-fold for T cells, 58.3-fold for NK-like T cells and 2.5-fold for NK cells. Interestingly, CD25 surface expression increased from less than equal to 1% up to median 79.7%. Cytotoxic activity of CIK cells against NB cells was in median 34.7, 25.9 and 34.8% against the cell lines UKF-NB-3, UKF-NB-4 and SK-N-SH, respectively. In comparison with IL-2-stimulated NK cells, CIK cells showed a significantly higher cytotoxicity. Antibody-mediated blocking of the receptors NKG2D, TRAIL, FasL, DNAM-1, NKp30 and lymphocyte function-associated antigen-1 (LFA-1) significantly reduced lytic activity, indicating that diverse cytotoxic mechanisms might be involved in CIK cell-mediated NB killing. CONCLUSIONS: Unlike the mechanism reported in other malignancies, NKG2D-mediated cytotoxicity does not constitute the major killing mechanism of CIK cells against NB.


Asunto(s)
Células Asesinas Inducidas por Citocinas/inmunología , Interleucina-15/farmacología , Neuroblastoma/terapia , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Antígeno-1 Asociado a Función de Linfocito/fisiología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Neuroblastoma/patología
9.
Eur J Immunol ; 44(7): 2074-84, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24752800

RESUMEN

NK cells are the main cells of the innate immune system that produce IFN-γ, and they express this cytokine at early stages of maturation in response to cytokine stimulation. Conversely, acquisition of IFN-γ-competence in CD4(+) T helper cells requires a differentiation process from naïve toward type 1 (Th1) cells, which is associated with epigenetic remodeling at the IFNG locus. In the present study, we show that the ability of NK cells to produce IFN-γ in response to activating receptor (actR) engagement is gradually acquired during terminal differentiation and is accompanied by progressively higher NF-κB activation in response to actR triggering. Moreover, during the differentiation process NK cells gradually display increasing expression of IFNG and TBX21 (encoding T-bet) transcripts and demethylation at the IFNG promoter. This study provides new insights in the molecular mechanisms underlying NK-cell ability to express IFN-γ upon actR engagement. Thus, we propose that in order to efficiently produce IFN-γ in response to infected or transformed cells, NK cells gain Th1-like features, such as higher IFN-γ competence and epigenetic remodeling of the IFNG promoter, during their terminal differentiation.


Asunto(s)
Diferenciación Celular , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Islas de CpG , Metilación de ADN , Humanos , Interferón gamma/genética , Células Asesinas Naturales/citología , FN-kappa B/fisiología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Receptor 1 Gatillante de la Citotoxidad Natural/fisiología , Receptor 3 Gatillante de la Citotoxidad Natural/fisiología , Regiones Promotoras Genéticas , Proteínas de Dominio T Box/fisiología
10.
J Immunol ; 190(8): 4408-19, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23509364

RESUMEN

Epithelial-mesenchymal transition (EMT) is a morphogenetic process characterized by the acquisition of mesenchymal properties linked with an invasive phenotype and metastasis of tumor cells. NK group 2, member D (NKG2D) is an NK cell-activating receptor crucially involved in cancer immunosurveillance. In this study, we show that induction of EMT by TGF-ß stimulation of human keratinocytes, by glycogen synthase kinase-3ß inhibition in several epithelial tumor cell lines, and by Snail1 overexpression in colorectal cancer cells strongly upregulated the expression of NKG2D ligands (NKG2DLs), MHC class I chain-related molecules A and B (MICA/B) and ULBP1-3. Overexpression of Snail1 and inhibition of glycogen synthase kinase-3ß in colorectal tumor cells markedly induced the activity of Sp1 transcription factor, which plays a key role in the upregulation of NKG2DL expression during EMT. The stimulation of MICA/B expression by TGF-ß treatment was independent of Sp1, but it involved posttranslational mechanisms mediated by mammalian target of rapamycin pathway. Accordingly, with the increased expression of NKG2DLs, triggering of EMT rendered cancer cells more susceptible to NKG2D-mediated killing by NK cells. In agreement, MICA/B were expressed in vivo in well-differentiated colorectal tumors with retained epithelial characteristics, whereas no expression of MICA/B was detected in poorly differentiated and invasive colorectal tumors that have lost epithelial characteristics. This decrease of MICA/B expression was associated with a dramatic increase of NKG2D(+)-tumor infiltrating lymphocytes. Overall, our findings indicate that EMT is a relevant checkpoint in the control of tumor progression through NKG2D-mediated immune responses.


Asunto(s)
Neoplasias Colorrectales/inmunología , Epitelio/inmunología , Mesodermo/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Animales , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Epitelio/metabolismo , Epitelio/patología , Células HT29 , Células HeLa , Células Hep G2 , Humanos , Inmunofenotipificación , Mesodermo/metabolismo , Mesodermo/patología , Ratones , Subfamilia K de Receptores Similares a Lectina de Células NK/biosíntesis , Invasividad Neoplásica/inmunología , Invasividad Neoplásica/patología
11.
J Allergy Clin Immunol ; 133(3): 827-35.e3, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24290277

RESUMEN

BACKGROUND: The diverse roles of innate immune cells in the pathogenesis of asthma remain to be fully defined. Natural killer (NK) cells are innate lymphocytes that can regulate adaptive immune responses. NK cells are activated in asthma; however, their role in allergic airway inflammation is not fully understood. OBJECTIVE: We investigated the importance of NK cells in house dust mite (HDM)-triggered allergic pulmonary inflammation. Specifically, we aimed to determine the role of the major NK-cell activating receptor NKG2D and NK-cell effector functions mediated by granzyme B. METHODS: Allergic airway inflammation was induced in the airways of mice by repeated intranasal HDM extract administration and responses in wild-type and NKG2D-deficient mice were compared. Adoptive transfer studies were used to identify the cells and mechanisms involved. RESULTS: Mice that lacked NKG2D were resistant to the induction of allergic inflammation and showed little pulmonary eosinophilia, few airway TH2 cells, and no rise in serum IgE after multiple HDM-allergen exposures. However, NKG2D was not required for pulmonary inflammation after a single inoculation of allergen. NKG2D-deficient mice showed no alteration in responses to respiratory virus infection. Transfer of wild-type NK cells (but not CD3(+) cells) into NKG2D-deficient mice restored allergic inflammatory responses only if the NK cells expressed granzyme B. CONCLUSIONS: These studies established a pivotal role for NK-cell NKG2D and granzyme B in the pathogenesis of HDM-induced allergic lung disease, and identified novel therapeutic targets for the prevention and treatment of asthma.


Asunto(s)
Asma/etiología , Granzimas/fisiología , Células Asesinas Naturales/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Neumonía/etiología , Animales , Asma/inmunología , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neumonía/inmunología , Pyroglyphidae/inmunología
12.
Clin Exp Immunol ; 178(3): 516-24, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25070361

RESUMEN

Bone marrow mesenchymal stem cells (BMSCs) inhibit immune cell responsiveness, and especially of T lymphocytes. We showed that BMSCs markedly inhibited the proliferation and cytokine production by CD8(+) T cells by a cell-to-cell contact phenomenon and secretion of soluble factors. BMSCs down-regulate the expression of natural killer group 2, member D protein (NKG2D) receptors on CD8(+) T cells when co-cultured with them. Moreover, CD8(+) T cells that express low levels of NKG2D had impaired proliferation after triggering by a mitogen. The major histocompatibility complex (MHC) class I chain-related (MIC) A/B molecule, which is a typical ligand for NKG2D, was expressed on BMSCs, and caused dampening of cell proliferation. Monoclonal antibody blocking experiments targeted to MIC A/B impaired CD8(+) T cell function, as evaluated by proliferation and cytokine production. In addition, the production of prostaglandin E2 (PGE2 ), indoleamine 2, 3-dioxygenase (IDO) and transforming growth factor (TGF)-ß1 were increased when BMSCs were co-cultured with CD8(+) T cells. The addition of specific inhibitors against PGE2 , IDO and TGF-ß partially restored the proliferation of CD8(+) T cells. Our results suggest that BMSCs suppress CD8(+) T cell-mediated activation by suppressing NKG2D expression and secretion of PGE2, IDO and TGF-ß. Our observations further confirm the feasibility of BMSCs as a potential adoptive cellular therapy in immune-mediated diseases such as graft-versus-host disease (GVHD).


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Dinoprostona/biosíntesis , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Activación de Linfocitos , Células Madre Mesenquimatosas/fisiología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Factor de Crecimiento Transformador beta/biosíntesis , Comunicación Celular , Células Cultivadas , Granzimas/biosíntesis , Antígenos de Histocompatibilidad Clase I/fisiología , Humanos , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis
13.
J Immunol ; 189(12): 5493-7, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23183896

RESUMEN

Monoclonal antibodies represent a promising approach to fight a variety of tumors, but their mode of action remains to be fully understood. NK cells can recognize Ab-coated targets, as well as stress ligands, on tumor cells. In this study, we investigated how NK cells integrate both kinds of activating signals. NK cell-mediated killing was maximal with the combined recognition of NKG2D ligands and Ab; surprisingly, only NKG2D engagement substantially enhanced degranulation. Conversely, Ab recognition by NK cells uniquely increased contact stability with tumor cells. Furthermore, using intravital imaging of solid tumors, we showed that Ab recognition favored prolonged interactions between NK cells and targets. Altogether, our results demonstrate that NK cell-mediated killing can be differentially regulated at the level of degranulation and contact stability by distinct activating receptors. Thus, complementary signals mediated by recognition of stress ligands and tumor-specific Abs may contribute to the efficacy of NK cells during mAb therapy.


Asunto(s)
Anticuerpos Antineoplásicos/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Comunicación Celular/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Regulación hacia Arriba/inmunología , Animales , Degranulación de la Célula/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Técnicas de Sustitución del Gen , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Linfoma/inmunología , Linfoma/metabolismo , Linfoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Receptores de IgG/metabolismo , Timoma/inmunología , Timoma/metabolismo , Timoma/patología
14.
Proc Natl Acad Sci U S A ; 108(10): 4081-6, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21321202

RESUMEN

The stimulatory natural killer group 2 member D (NKG2D) lymphocyte receptor and its tumor-associated ligands are important mediators in the immune surveillance of cancer. With advanced human tumors, however, persistent NKG2D ligand expression may favor tumor progression. We have found that cancer cells themselves express NKG2D in complex with the DNAX-activating protein 10 (DAP10) signaling adaptor. Triggering of NKG2D on ex vivo cancer cells or on tumor lines which express only few receptor complexes activates the oncogenic PI3K-protein kinase B (PKB/AKT)-mammalian target of rapamycin (mTOR) signaling axis and downstream effectors, the ribosomal protein S6 kinase 1 (S6K1) and the translation initiation factor 4E-binding protein 1 (4E-BP1). In addition, as in lymphocytes, NKG2D ligand engagement stimulates phosphorylation of JNK and ERK in MAP kinase cascades. Consistent with these signaling activities, above-threshold expression of NKG2D-DAP10 in a ligand-bearing tumor line increases its bioenergetic metabolism and proliferation, thus suggesting functional similarity between this immunoreceptor and tumor growth factor receptors. This relationship is supported by significant correlations between percentages of cancer cells that are positive for surface NKG2D and criteria of tumor progression. Hence, in a conceptual twist, these results suggest that tumor co-option of NKG2D immunoreceptor expression may complement the presence of its ligands for stimulation of tumor growth.


Asunto(s)
Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Neoplasias/fisiopatología , Transducción de Señal , Línea Celular Tumoral , Progresión de la Enfermedad , Activación Enzimática , Femenino , Humanos , Masculino , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
15.
Blood ; 117(24): 6571-81, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21518928

RESUMEN

Although innate immune signals shape the activation of naive T cells, it is unclear how innate signals influence effector T-cell function. This study determined the effects of stimulating the NKG2D receptor in conjunction with the TCR on human effector CD8(+) T cells. Stimulation of CD8(+) T cells through CD3 and NKG2D simultaneously or through a chimeric NKG2D receptor, which consists of NKG2D fused to the intracellular region of CD3ζ, activated ß-catenin and increased expression of ß-catenin-induced genes, whereas T cells stimulated through the TCR or a combination of the TCR and CD28 did not. Activation by TCR and NKG2D prevented expression and production of anti-inflammatory cytokines IL-10, IL-9, IL-13, and VEGF-α in a ß-catenin- and PPARγ- dependent manner. NKG2D stimulation also modulated the cytokine secretion of T cells activated simultaneously through CD3 and CD28. These data indicate that activating CD8(+) T cells through the NKG2D receptor along with the TCR modulates signal transduction and the production of anti-inflammatory cytokines. Thus, human effector T cells alter their function depending on which innate receptors are engaged in conjunction with the TCR complex.


Asunto(s)
Citocinas/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Antiinflamatorios/metabolismo , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/fisiología , Células Cultivadas , Citocinas/genética , Regulación de la Expresión Génica/fisiología , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/fisiología , Ratones , Subfamilia K de Receptores Similares a Lectina de Células NK/agonistas , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Proteína Oncogénica v-akt/metabolismo , Proteína Oncogénica v-akt/fisiología , PPAR gamma/metabolismo , PPAR gamma/fisiología , Células U937 , beta Catenina/metabolismo , beta Catenina/fisiología
16.
J Immunol ; 186(3): 1538-45, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21191066

RESUMEN

NK cell activation is negatively regulated by the expression of target cell MHC class I molecules. We show that this relationship is nonlinear due to an NK cell activation/inhibition threshold. Ewing's sarcoma family tumor cell monolayers, which were highly susceptible to NK cells in vitro, developed a highly resistant phenotype when cultured as three-dimensional multicellular tumor spheroid structures. This suggested that tumor architecture is likely to influence the susceptibility to NK cells in vivo. Resistance of the multicellular tumor spheroid was associated with the increased expression of MHC class I molecules and greatly reduced NK cell activation, implying that a threshold of NK cell activation/inhibition had been crossed. Reducing MHC class I expression on Ewing's sarcoma family tumor monolayers did not alter their susceptibility to NK cells, whereas increased expression of MHC class I rendered them resistant and allowed the threshold point to be identified. This threshold, as defined by MHC class I expression, was predictive of the number of NK-resistant target cells within a population. A threshold permits modest changes in the target cell surface phenotype to profoundly alter the susceptibility to NK cells. Whereas this allows for the efficient detection of target cells, it also provides a route for pathogens and tumors to evade NK cell attack.


Asunto(s)
Membrana Celular/inmunología , Predisposición Genética a la Enfermedad/genética , Células Asesinas Naturales/inmunología , Sarcoma de Ewing/inmunología , Sarcoma de Ewing/metabolismo , Escape del Tumor/inmunología , Línea Celular Tumoral , Membrana Celular/genética , Membrana Celular/patología , Pruebas Inmunológicas de Citotoxicidad/métodos , Células HeLa , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inmunofenotipificación/métodos , Células K562 , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Activación de Linfocitos/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Sarcoma de Ewing/patología , Esferoides Celulares/inmunología , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Translocación Genética/inmunología , Células Tumorales Cultivadas , Escape del Tumor/genética
17.
J Immunol ; 186(6): 3304-8, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21307295

RESUMEN

It is uncertain whether NK cells modulate T cell memory differentiation. By using a genetic model that allows the selective depletion of NK cells, we show in this study that NK cells shape CD8(+) T cell fate by killing recently activated CD8(+) T cells in an NKG2D- and perforin-dependent manner. In the absence of NK cells, the differentiation of CD8(+) T cells is strongly biased toward a central memory T cell phenotype. Although, on a per-cell basis, memory CD8(+) T cells generated in the presence or the absence of NK cells have similar functional features and recall capabilities, NK cell deletion resulted in a significantly higher number of memory Ag-specific CD8(+) T cells, leading to more effective control of tumors carrying model Ags. The enhanced memory responses induced by the transient deletion of NK cells may provide a rational basis for the design of new vaccination strategies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Depleción Linfocítica/métodos , Regulación hacia Arriba/inmunología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Femenino , Humanos , Memoria Inmunológica/genética , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Activación de Linfocitos/genética , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Subfamilia K de Receptores Similares a Lectina de Células NK/deficiencia , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Perforina , Proteínas Citotóxicas Formadoras de Poros/biosíntesis , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/fisiología , Regulación hacia Arriba/genética
18.
Mediators Inflamm ; 2013: 713859, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24453427

RESUMEN

In order to understand how tumor cells can escape immune surveillance mechanisms and thus develop antitumor therapies, it is critically important to investigate the mechanisms by which the immune system interacts with the tumor microenvironment. In our current study, IL-17 deficiency results in reduced melanoma tumor size, diminished numbers of proliferating cells and blood vessels, and decreased percentage of CD11b(+)Gr-1(+) MDSCs in tumor tissues. IL-17 promotes IL-6 induction and Stat3 activation. Treatment of Stat3 inhibitor WP1066 in B16-F10 tumor cells inoculated wild-type mice inhibits tumor growth. Additional administration of recombinant IL-6 into B16-F10 tumor-bearing IL-17(-/-) mice results in markedly increased tumor size and p-Stat3 expression, whereas additional recombinant IL-17 administration into B16-F10 tumor-bearing wild-type mice treated with anti-IL-6 mAb does not significantly alter the tumor growth and p-Stat3 expression. In our further study, blockade of Hmgb1-RAGE pathway inhibits melanoma tumor growth and reduces production of IL-23 and IL-17. All these data suggest that Hmgb1-IL-23-IL-17-IL-6-Stat3 axis plays a pivotal role in tumor development in murine models of melanoma, and blocking any portion of this axis will attenuate melanoma tumor growth.


Asunto(s)
Proliferación Celular , Proteína HMGB1/fisiología , Interleucina-17/fisiología , Interleucina-23/fisiología , Interleucina-6/fisiología , Melanoma Experimental/etiología , Factor de Transcripción STAT3/fisiología , Animales , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Receptor para Productos Finales de Glicación Avanzada , Receptores de Antígenos de Linfocitos T gamma-delta/fisiología , Receptores Inmunológicos/fisiología
19.
Lab Invest ; 92(7): 967-77, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22449797

RESUMEN

In mouse models it has been shown that natural killer (NK) cells can attenuate liver fibrosis via killing of activated hepatic stellate cells (HSCs) in a NKG2D- and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-dependent manner. However, only little data exist regarding interactions of human NK cells with HSCs and their potential role in hepatitis C virus (HCV)-associated fibrogenesis. Therefore, purified NK cells from untreated HCV RNA(+) patients (n=33), interferon-α (IFN-α)-treated patients (n=17) and healthy controls (n=18) were coincubated with activated primary HSCs, and were tested for degranulation (CD107a expression) and secretion of IFN-γ and TNF-α, respectively. Induction of HSC apoptosis was analyzed using an active caspase-3 assay. We found that following coincubation with HSCs a significant increase in CD107a expression could be observed in both NK cells from HCV(+) patients and healthy controls, whereas only negligible secretion of IFN-γ and TNF-α could be detected. More importantly, NK cells from untreated HCV RNA(+) patients were significantly more effective in induction of HSC apoptosis (17.8 ± 9.2%) than NK cells from healthy controls (6.2 ± 2.1%; P<0.0001). Additionally, we observed an inverse correlation of liver fibrosis stage and the ability of NK cells to induce HSC apoptosis. Induction of HSC apoptosis was contact dependent and could partly be blocked by antibodies specific for TRAIL, NKG2D and FasL, respectively. It is noteworthy that NK cells from IFN-α-treated HCV(+) patients displayed the highest capability to kill HSCs (27.6 ± 10.5%). Accordingly, pre-stimulation of NK cells with recombinant IFN-α significantly increased the ability of NK cells to induce cell death in primary HSCs and was dependent on upregulated expression of TRAIL. Here we demonstrate that NK cells from HCV-infected patients are highly efficient in inducing apoptosis of activated HSCs. Thus, NK cells may have an important anti-fibrotic role in chronic hepatitis C.


Asunto(s)
Proteína Ligando Fas/fisiología , Células Estrelladas Hepáticas/patología , Células Estrelladas Hepáticas/fisiología , Hepatitis C Crónica/patología , Hepatitis C Crónica/fisiopatología , Células Asesinas Naturales/fisiología , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Antivirales/administración & dosificación , Apoptosis/fisiología , Estudios de Casos y Controles , Femenino , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/inmunología , Humanos , Técnicas In Vitro , Interferón-alfa/administración & dosificación , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Masculino , Persona de Mediana Edad , Polietilenglicoles/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Ribavirina/provisión & distribución , Factor de Necrosis Tumoral alfa/biosíntesis
20.
RNA Biol ; 9(6): 792-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22617882

RESUMEN

Natural killer (NK) cells play an important role in the direct killing of cancerous and virus-infected cells. One of the important activating receptors which mediates this killing is NKG2D. This receptor recognizes various stress-induced ligands including the major histocompatibility complex class I-related chain A and B (MICA and MICB respectively). The mechanisms controlling the expression of the NKG2D ligands are not completely understood, yet various studies have demonstrated that the expression of the NKG2D ligands is manipulated by viruses and by tumor cells in order to escape the NKG2D detection. Cumulative data have emphasized that various microRNAs (miRNAs) of both human and viral origin control the expression of NKG2D ligands, particularly MICB. Herein we review recent findings regarding the miRNA regulation of the NKG2D ligands. We propose that these miRNAs generate a complex network of interactions that control the expression of the NKG2D ligands under normal conditions and during disease development.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/genética , Péptidos y Proteínas de Señalización Intercelular/genética , MicroARNs/fisiología , Interferencia de ARN , Secuencia de Bases , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Redes Reguladoras de Genes , Antígenos de Histocompatibilidad Clase I/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , ARN Viral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA