Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pediatr Res ; 73(4 Pt 1): 402-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23269121

RESUMO

BACKGROUND: The propensity for sustained inflammation after bacterial infection in neonates, resulting in inflammatory sequelae such as bronchopulmonary dysplasia and periventricular leucomalacia, is well known, but its molecular mechanisms remain elusive. Termination of inflammatory reactions physiologically occurs early after removal of bacteria by phagocytosis-induced cell death (PICD) of immune effector cells such as monocytes. PICD from cord blood monocytes (CBMOs) was shown to be reduced as compared with that of peripheral blood monocytes (PBMOs) from adult donors in vitro. METHODS: PBMOs, CBMOs, and Fas (CD95)-deficient (lpr) mouse monocytes were analyzed in an in vitro infection model using green fluorescence protein-labeled Escherichia coli (E. coli-GFP). Phagocytosis and apoptosis were quantified by flow cytometry and CD95L secretion was quantified by enzyme-linked immunosorbent assay. RESULTS: We demonstrate the involvement of the CD95/CD95 ligand pathway (CD95/CD95L) in PICD and provide evidence that diminished CD95L secretion by CBMOs may result in prolonged activation of neonatal immune effector cells. CONCLUSION: These in vitro results offer for the first time a molecular mechanism accounting for sustained inflammation seen in neonates.


Assuntos
Apoptose , Proteína Ligante Fas/metabolismo , Sangue Fetal/imunologia , Inflamação/imunologia , Monócitos/imunologia , Fagocitose , Receptor fas/metabolismo , Adulto , Animais , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Ensaio de Imunoadsorção Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Citometria de Fluxo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Recém-Nascido , Inflamação/sangue , Inflamação/microbiologia , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/microbiologia , Monócitos/patologia , Transdução de Sinais , Receptor fas/deficiência , Receptor fas/genética
2.
Int J Cancer ; 128(12): 2911-22, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20960460

RESUMO

Azacytidine and decitabine are approved for treatment of acute myeloid leukemias and myelodysplastic syndromes. While clinical responses are attributed to epigenetic effects and induction of apoptosis in malignant cells, these azanucleosides also affect antitumor immune responses. NK cells as components of innate immunity may confine development and progression of cancer. Numerous therapeutic strategies presently aim to reinforce NK reactivity against hematopoietic malignancies. We here comparatively analyzed the effect of the two clinically available azanucleosides and report that NK cytotoxicity and IFN-γ production are significantly impaired by pharmacological concentrations of azacytidine but enhanced by decitabine. This was not due to alterations in the target cells but caused by direct effects on NK cells depending on the chemical modifications by which azanucleosides differ from their physiological analogues. Although azacytidine impaired mRNA synthesis and induced apoptosis in NK cells, decitabine did not per se alter NK cell viability or reactivity but enhanced responsiveness to activating stimuli by inducing transcription of genes involved in NK reactivity. Tantalizingly, these effects were independent of incorporation of the azanucleosides into DNA during cell division. While azacytidine impairs NK antitumor immunity, decitabine augments NK reactivity by yet unidentified mechanisms and may thus serve well in therapeutic strategies combining its effects on malignant cells with its ability to enhance NK functions.


Assuntos
Azacitidina/análogos & derivados , Azacitidina/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Citotoxicidade Imunológica , Decitabina , Humanos , Imunidade Inata , Interferon gama/biossíntese , Células Matadoras Naturais/imunologia
3.
Int J Cancer ; 127(9): 2119-28, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20143399

RESUMO

In chronic myeloid leukemia (CML), BCR/ABL-mediated oncogenic signaling can be targeted with the BCR/ABL-inhibitors Imatinib, Nilotinib and Dasatinib. However, these agents may also affect anti-tumor immunity. Here, we analyzed the effects of the 3 BCR/ABL-inhibitors on natural killer (NK) cell reactivity. Exposure of CML cells (K562, Meg-01) to pharmacological concentrations of Imatinib, Nilotinib and Dasatinib diminished expression of ligands for the activating immunoreceptor NKG2D to a similar extent. This resulted in comparably reduced NK cell cytotoxicity and IFN-gamma production. When direct effects on NK cell responses to K562 and primary CML cells as well as activating cytokines were studied, Dasatinib was found to abrogate NK cytotoxicity and cytokine production. Nilotinib did not alter cytotoxicity but, at high levels, impaired NK cytokine production, while Imatinib had no direct influence on NK cell reactivity. Of note, Nilotinib, but not the other BCR/ABL-inhibitors increased cell death within the preferentially cytokine-secreting CD56(bright)CD16(-) NK cell subset, which may, at least in part, serve to explain the effect of Nilotinib on NK cytokine production. Analysis of NK cell signaling revealed that Dasatinib inhibited proximal signaling events leading to decreased phosphorylation of PI3K and ERK that are crucial for NK cell reactivity. Imatinib and Nilotinib, in contrast, showed no relevant effect on NK cell PI3K or ERK activity. In light of the potential role of NK cells in the immunesurveillance of residual leukemia and for future combinatory immunotherapeutic approaches, our data indicate that choice and dosing of the most suitable BCR/ABL-inhibitor for a given patient require careful consideration.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Células Matadoras Naturais/efeitos dos fármacos , Piperazinas/farmacologia , Pirimidinas/farmacologia , Tiazóis/farmacologia , Silicatos de Alumínio/metabolismo , Benzamidas , Linhagem Celular Tumoral , Citotoxicidade Imunológica/efeitos dos fármacos , Dasatinibe , Regulação para Baixo , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Mesilato de Imatinib , Interferon gama/metabolismo , Células K562 , Células Matadoras Naturais/imunologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/embriologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia
4.
Blood ; 115(15): 3058-69, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20008791

RESUMO

Natural killer (NK) cells play an important role in the immunosurveillance of leukemia. Their reactivity is governed by a balance of activating and inhibitory receptors including various members of the tumor necrosis factor receptor (TNFR) family. Here we report that human NK cells acquire expression of the TNFR family member CD137 upon activation, and NK cells of acute myeloid leukemia (AML) patients display an activated phenotype with substantial CD137 expression. CD137 ligand (CD137L) was detectable on leukemic cells in 35% of 65 investigated AML patients, but not on healthy CD34(+) cells, and expression was associated with monocytic differentiation. Bidirectional signaling following CD137-CD137L interaction induced the release of the immunomodulatory cytokines interleukin-10 and TNF by AML cells and directly diminished granule mobilization, cytotoxicity, and interferon-gamma production of human NK cells, which was restored by blocking CD137. Cocultures of NK cells with CD137L transfectants confirmed that human CD137 inhibits NK-cell reactivity, while activating signals were transduced by its counterpart on NK cells in mice. Our data underline the necessity to study the function of seemingly analog immunoregulatory molecules in mice compared with men and demonstrate that CD137-CD137L interaction enables immune evasion of AML cells by impairing NK-cell tumor surveillance in humans.


Assuntos
Ligante 4-1BB/imunologia , Células Matadoras Naturais/imunologia , Leucemia Mieloide Aguda/imunologia , Ligante 4-1BB/genética , Idoso , Idoso de 80 Anos ou mais , Animais , Linhagem Celular Tumoral , Citocinas/metabolismo , Feminino , Regulação Leucêmica da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Tolerância Imunológica/imunologia , Células Matadoras Naturais/patologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Ligação Proteica , Transdução de Sinais/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Regulação para Cima/genética
5.
J Immunol ; 183(12): 8286-94, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20007592

RESUMO

Sunitinib and Sorafenib are protein kinase inhibitors (PKI) approved for treatment of patients with advanced renal cell cancer (RCC). However, long-term remissions of advanced RCC have only been observed after IL-2 treatment, which underlines the importance of antitumor immune responses in RCC patients. Because PKI, besides affecting tumor cells, also may inhibit signaling in immune effector cells, we determined how Sunitinib and Sorafenib influence antitumor immunity. We found that cytotoxicity and cytokine production of resting and IL-2-activated PBMC are inhibited by pharmacological concentrations of Sorafenib but not Sunitinib. Analysis of granule-mobilization within PBMC revealed that this was due to impaired reactivity of NK cells, which substantially contribute to antitumor immunity by directly killing target cells and shaping adaptive immune responses by secreting cytokines like IFN-gamma. Analyses with resting and IL-2-activated NK cells revealed that both PKI concentration dependently inhibit cytotoxicity and IFN-gamma production of NK cells in response to tumor targets. This was due to impaired PI3K and ERK phosphorylation which directly controls NK cell reactivity. However, while Sorafenib inhibited NK cell effector functions and signaling at levels achieved upon recommended dosing, pharmacological concentrations of Sunitinib had no effect, and this was observed upon stimulation of NK cell reactivity by tumor target cells and upon IL-2 treatment. In light of the important role of NK cells in antitumor immunity, and because multiple approaches presently aim to combine PKI treatment with immunotherapeutic strategies, our data demonstrate that choice and dosing of the most suitable PKI in cancer treatment requires careful consideration.


Assuntos
Antineoplásicos/farmacologia , Benzenossulfonatos/farmacologia , Citotoxicidade Imunológica/efeitos dos fármacos , Indóis/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Pirróis/farmacologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Citotoxicidade Imunológica/imunologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Células K562 , Camundongos , Niacinamida/análogos & derivados , Compostos de Fenilureia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Sorafenibe , Sunitinibe
6.
Cancer Res ; 69(3): 1037-45, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19155305

RESUMO

The reciprocal interaction of tumor cells with the immune system is influenced by various members of the tumor necrosis factor (TNF)/TNF receptor (TNFR) family, and recently, glucocorticoid-induced TNFR-related protein (GITR) was shown to stimulate antitumor immunity in mice. However, GITR may mediate different effects in mice and men and impairs the reactivity of human natural killer (NK) cells. Here, we studied the role of GITR and its ligand (GITRL) in human acute myeloid leukemia (AML). Surface expression of GITRL was observed on AML cells in six of seven investigated cell lines, and 34 of 60 investigated AML patients whereas healthy CD34(+) cells did not express GITRL. Furthermore, soluble GITRL (sGITRL) was detectable in AML patient sera in 18 of 55 investigated cases. While the presence of GITRL was not restricted to a specific AML subtype, surface expression was significantly associated with monocytic differentiation. Signaling via GITRL into patient AML cells induced the release of TNF and interleukin-10 (IL-10), and this was blocked by the inhibition of mitogen-activated protein kinases extracellular signal-regulated kinase 1/2. Furthermore, triggering GITR by surface-expressed and sGITRL impaired NK cell cytotoxicity and IFN-gamma production in cocultures with leukemia cells, and NK cell reactivity could be restored by blocking GITR and neutralization of sGITRL and IL-10. Thus, whereas a stimulatory role of the GITR-GITRL system in mouse antitumor immunity has been reported, our data show that in humans GITRL expression subverts NK cell immunosurveillance of AML. Our results provide useful information for therapeutic approaches in AML, which, like haploidentical stem cell transplantation, rely on a sufficient NK cell response.


Assuntos
Leucemia Mieloide/imunologia , Fatores de Necrose Tumoral/imunologia , Doença Aguda , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Citocinas/imunologia , Feminino , Humanos , Vigilância Imunológica/imunologia , Interferon gama/biossíntese , Células Matadoras Naturais/imunologia , Leucemia Mieloide/sangue , Masculino , Pessoa de Meia-Idade , Monócitos/imunologia , Fatores de Necrose Tumoral/biossíntese , Adulto Jovem
7.
J Immunol ; 181(10): 6711-9, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18981088

RESUMO

Reciprocal interactions between NK cells and dendritic cells have been shown to influence activation of NK cells, maturation, or lysis of dendritic cells and subsequent adaptive immune responses. However, little is known about the crosstalk between monocytes and NK cells and the receptors involved in this interaction. We report in this study that human monocytes, upon TLR triggering, up-regulate MHC class I-Related Chain (MIC) A, but not other ligands for the activating immunoreceptor NKG2D like MICB or UL-16 binding proteins 1-3. MICA expression was associated with CD80, MHC class I and MHC class II up-regulation, secretion of proinflammatory cytokines, and apoptosis inhibition, but was not accompanied by release of MIC molecules in soluble form. TLR-induced MICA on the monocyte cell surface was detected by autologous NK cells as revealed by NKG2D down-regulation. Although MICA expression did not render monocytes susceptible for NK cell cytotoxicity, LPS-treated monocytes stimulated IFN-gamma production of activated NK cells which was substantially dependent on MICA-NKG2D interaction. No enhanced NK cell proliferation or cytotoxicity against third-party target cells was observed after stimulation of NK cells with LPS-activated monocytes. Our data indicate that MICA-NKG2D interaction constitutes a mechanism by which monocytes and NK cells as an early source of IFN-gamma may communicate directly during an innate immune response to infections in humans.


Assuntos
Antígenos de Histocompatibilidade Classe I/imunologia , Células Matadoras Naturais/imunologia , Monócitos/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Receptor Cross-Talk/imunologia , Receptores Toll-Like/imunologia , Antígeno B7-1/imunologia , Antígeno B7-1/metabolismo , Citocinas/metabolismo , Citotoxicidade Imunológica , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Interferon gama/metabolismo , Células Matadoras Naturais/metabolismo , Ligantes , Monócitos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Toll-Like/metabolismo
8.
Blood ; 112(9): 3735-43, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18689545

RESUMO

NK cell anti-tumor reactivity is governed by a balance of activating and inhibitory receptors including various TNF receptor (TNFR) family members. Here we report that human tumor cells release a soluble form of the TNF family member Glucocorticoid-Induced TNFR-Related Protein (GITR) ligand (sGITRL), which can be detected in cell culture supernatants. Tumor-derived sGITRL concentration-dependently reduced NK cell cytotoxicity and IFN-gamma production, which could be overcome by neutralization of sGITRL using a GITR-Ig fusion protein. Although sGITRL did not induce apoptosis in NK cells, it diminished nuclear localized RelB, indicating that sGITRL negatively modulates NK cell NF-kappaB activity. Furthermore, we detected substantial levels of sGITRL in sera of patients with various malignancies, but not in healthy controls. Presence of sGITRL-containing patient serum in cocultures with tumor cells significantly reduced NK cell cytotoxicity and IFN-gamma production, which could again be restored by neutralization of sGITRL. The strong correlation of tumor incidence and elevated sGITRL levels indicates that sGITRL is released from cancers in vivo, leading to impaired NK cell immunosurveillance of human tumors. Our data suggest that determination of sGITRL levels might be implemented as a tumor marker in patients, and GITRL neutralization may be used to improve immunotherapeutic strategies relying on NK cell reactivity.


Assuntos
Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Inibidores do Fator de Necrose Tumoral , Fatores de Necrose Tumoral/metabolismo , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Técnicas de Cocultura , Citotoxicidade Imunológica , Neoplasias Hematológicas/sangue , Neoplasias Hematológicas/imunologia , Humanos , Interferon gama/biossíntese , Células Matadoras Naturais/metabolismo , NF-kappa B/metabolismo , Neoplasias/metabolismo , Testes de Neutralização , Solubilidade , Fatores de Necrose Tumoral/sangue
9.
Clin Cancer Res ; 14(11): 3520-8, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18519785

RESUMO

PURPOSE: Hepatocellular carcinoma (HCC) displays particular resistance to conventional cytostatic agents. Alternative treatment strategies focus on novel substances exhibiting antineoplastic and/or immunomodulatory activity enhancing for example natural killer (NK) cell antitumor reactivity. However, tumor-associated ligands engaging activating NK cell receptors are largely unknown. Exceptions are NKG2D ligands (NKG2DL) of the MHC class I-related chain and UL16-binding protein families, which potently stimulate NK cell responses. We studied the consequences of proteasome inhibition with regard to direct and NK cell-mediated effects against HCC. EXPERIMENTAL DESIGN: Primary human hepatocytes (PHH) from different donors, hepatoma cell lines, and NK cells were exposed to Bortezomib. Growth and viability of the different cells, and immunomodulatory effects including alterations of NKG2DL expression on hepatoma cells, specific induction of NK cell cytotoxicity and IFN-gamma production were investigated. RESULTS: Bortezomib treatment inhibited hepatoma cell growth with IC(50) values between 2.4 and 7.7 nmol/L. These low doses increased MICA/B mRNA levels, resulting in an increase of total and cell surface protein expression in hepatoma cells, thus stimulating cytotoxicity and IFN-gamma production of cocultured NK cells. Importantly, although NK cell IFN-gamma production was concentration-dependently reduced, low-dose Bortezomib neither induced NKG2DL expression or cell death in PHH nor altered NK cell cytotoxicity. CONCLUSIONS: Low-dose Bortezomib mediates a specific dual antitumor effect in HCC by inhibiting tumor cell proliferation and priming hepatoma cells for NK cell antitumor reactivity. Our data suggest that patients with HCC may benefit from Bortezomib treatment combined with immunotherapeutic approaches such as adoptive NK cell transfer taking advantage of enhanced NKG2D-mediated antitumor immunity.


Assuntos
Antineoplásicos/administração & dosagem , Ácidos Borônicos/administração & dosagem , Carcinoma Hepatocelular/tratamento farmacológico , Células Matadoras Naturais/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Pirazinas/administração & dosagem , Bortezomib , Carcinoma Hepatocelular/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Imunofluorescência , Proteínas Ligadas por GPI , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interferon gama/biossíntese , Células Matadoras Naturais/imunologia , Neoplasias Hepáticas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Blood ; 109(11): 4816-24, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17341666

RESUMO

Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis with potential for progression to acute myeloid leukemia (AML). We compared natural killer (NK) cytolytic function in 48 MDS patients with 37 healthy donors and found reduced activity in the patient population (K562 cytolysis, 19% +/- 21% SD versus 40% +/- 17%) (P < .001). NK cytotoxicity in MDS patients was reduced against 3 disparate tumor targets with differential activating receptor requirement, suggesting global defects in NK function. Reduced NK function in MDS was significantly associated with higher International Prognostic Score (P = .01), abnormal karyotype (P = .05), the presence of excess blasts (P = .01), and age-adjusted bone marrow hypercellularity (P = .04). MDS patients had a display of the activating receptor NKp30, and NKG2D down-regulation closely correlated with impaired NK function (P = .001). NKG2D ligands (MICA and MICB) were expressed on CD34(+) cells from bone marrow of 30% of MDS patients and a leukemic cell line derived from an MDS patient (MDS1). Collectively, these findings suggest that impairment of NK cytolytic function derives in part from reduced activating NK receptors such as NKG2D in association with disease progression. Evasion of NK immunosurveillance may have importance for MDS disease progression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Células Matadoras Naturais/citologia , Síndromes Mielodisplásicas/sangue , Síndromes Mielodisplásicas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Antígenos CD34/biossíntese , Células da Medula Óssea/citologia , Progressão da Doença , Feminino , Humanos , Células K562 , Células Matadoras Naturais/metabolismo , Leucemia Mieloide Aguda/metabolismo , Masculino , Pessoa de Meia-Idade , Lesões Pré-Cancerosas
11.
FASEB J ; 21(10): 2442-54, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17360848

RESUMO

Glucocorticoid-induced TNF-related protein (GITR) has been shown to stimulate T cell-mediated antitumor immunity in mice. However, the functional relevance of GITR and its ligand (GITRL) for non-T cells has yet to be fully explored. In addition, recent evidence suggests that GITR plays different roles in mice and humans. We studied the role of GITR-GITRL interaction in human tumor immunology and report for the first time that primary gastrointestinal cancers and tumor cell lines of different histological origin express substantial levels of GITRL. Signaling through GITRL down-regulated the expression of the immunostimulatory molecules CD40 and CD54 and the adhesion molecule EpCAM, and induced production of the immunosuppressive cytokine TGF-beta by tumor cells. On NK cells, GITR is constitutively expressed and up-regulated following activation. Blocking GITR-GITRL interaction in cocultures of tumor cells and NK cells substantially increased cytotoxicity and IFN-gamma production of NK cells demonstrating that constitutive expression of GITRL by tumor cells diminishes NK cell antitumor immunity. GITRL-Ig fusion protein or cell surface-expressed GITRL did not induce apoptosis in NK cells, but diminished nuclear localized c-Rel and RelB, indicating that GITR might negatively modulate NK cell NF-kappaB activity. Taken together, our data indicate that tumor-expressed GITRL mediates immunosubversion in humans.


Assuntos
Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Receptores de Fator de Crescimento Neural/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Fatores de Necrose Tumoral/fisiologia , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Clonagem Molecular , Proteína Relacionada a TNFR Induzida por Glucocorticoide , Humanos , Ligantes , Receptores de Fator de Crescimento Neural/genética , Receptores do Fator de Necrose Tumoral/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Fatores de Necrose Tumoral/genética
12.
Exp Hematol ; 34(7): 888-94, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16797416

RESUMO

OBJECTIVE: Expression of the B7 homolog B7-H1 (PD1-Ligand) has been proposed to enable tumor cells to evade immune surveillance. Recently, B7-H1 on murine leukemia cells was reported to mediate resistance to cytolytic T-cell destruction. We here investigate the expression and function of the B7 homolog B7-H1 in human leukemia. PATIENTS AND METHODS: Leukemia cells from 30 patients and 9 human leukemia cell lines were investigated for B7-H1 expression by flow cytometry. Functional relevance of B7-H1 for tumor-immune interactions was assessed by coculture experiments using purified, alloreactive CD4 and CD8 T cells in the presence of a neutralizing anti-B7-H1 antibody. RESULTS: Significant B7-H1 expression levels on leukemia cells were detected in 17 of 30 patients and in eight of nine cell lines. In contrast to various other tumor entities and the data reported from a murine leukemia system, no significant inhibitory effect of leukemia-derived B7-H1 on CD4 and CD8 cytokine production (IFN-gamma, IL-2), proliferation or expression of T-cell activation markers (ICOS, CD69) was observed. Furthermore, in the presence of neutralizing B7-H1 antibody (mAb 5H1) occurred no significant changes in T cell IFN-gamma or IL-2 production or proliferation. CONCLUSIONS: Our data demonstrate that leukemia-derived B7-H1 seems to have no direct influence on T-cell activation, proliferation, and cytokine production in humans. Further experiments are warranted to delineate factors and characterize yet-unidentified B7-H1 receptor(s) that determine inhibitory and stimulatory functions of B7-H1 in human leukemia.


Assuntos
Antígenos CD/imunologia , Leucemia/imunologia , Linfócitos T/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígeno B7-H1 , Western Blotting , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Citometria de Fluxo , Humanos , Leucemia/classificação , Masculino , Pessoa de Meia-Idade
13.
Cancer Res ; 65(14): 6321-9, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16024634

RESUMO

Natural killer (NK) cells as components of the innate immunity substantially contribute to antitumor immune responses. However, the tumor-associated ligands engaging activating NK cell receptors are largely unknown. An exception are the MHC class I chain-related molecules MICA and MICB and the UL16-binding proteins (ULBP) which bind to the activating immunoreceptor NKG2D expressed on cytotoxic lymphocytes. A therapeutic induction of NKG2D ligands that primes cancer cells for NK cell lysis has not yet been achieved. By microarray studies, we found evidence that treatment of human hepatocellular carcinoma cells with the histone deacetylase inhibitor (HDAC-I) sodium valproate (VPA) mediates recognition of cancer cells by cytotoxic lymphocytes via NKG2D. VPA induced transcription of MICA and MICB in hepatocellular carcinoma cells, leading to increased cell surface, soluble and total MIC protein expression. No significant changes in the expression of the NKG2D ligands ULBP1-3 were observed. The induction of MIC molecules increased lysis of hepatocellular carcinoma cells by NK cells which was abolished by addition of a blocking NKG2D antibody. Importantly, in primary human hepatocytes, VPA treatment did not induce MIC protein expression. Taken together, our data show that the HDAC-I VPA mediates specific priming of malignant cells for innate immune effector mechanisms. These results suggest the clinical evaluation of HDAC-I in solid tumors such as hepatocellular carcinoma, especially in combination with immunotherapy approaches employing adoptive NK cell transfer.


Assuntos
Carcinoma Hepatocelular/imunologia , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Células Matadoras Naturais/imunologia , Neoplasias Hepáticas/imunologia , Receptores Imunológicos/metabolismo , Ácido Valproico/farmacologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Ligantes , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Imunológicos/biossíntese , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Receptores de Células Matadoras Naturais , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...