Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Neuro Oncol ; 26(2): 335-347, 2024 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-37758193

RESUMEN

BACKGROUND: Central nervous system (CNS) WHO grade 2 low-grade glioma (LGG) patients are at high risk for recurrence and with unfavorable long-term prognosis due to the treatment resistance and malignant transformation to high-grade glioma. Considering the relatively intact systemic immunity and slow-growing nature, immunotherapy may offer an effective treatment option for LGG patients. METHODS: We conducted a prospective, randomized pilot study to evaluate the safety and immunological response of the multipeptide IMA950 vaccine with agonistic anti-CD27 antibody, varlilumab, in CNS WHO grade 2 LGG patients. Patients were randomized to receive combination therapy with IMA950 + poly-ICLC and varlilumab (Arm 1) or IMA950 + poly-ICLC (Arm 2) before surgery, followed by adjuvant vaccines. RESULTS: A total of 14 eligible patients were enrolled in the study. Four patients received pre-surgery vaccines but were excluded from postsurgery vaccines due to the high-grade diagnosis of the resected tumor. No regimen-limiting toxicity was observed. All patients demonstrated a significant increase of anti-IMA950 CD8+ T-cell response postvaccine in the peripheral blood, but no IMA950-reactive CD8+ T cells were detected in the resected tumor. Mass cytometry analyses revealed that adding varlilumab promoted T helper type 1 effector memory CD4+ and effector memory CD8+ T-cell differentiation in the PBMC but not in the tumor microenvironment. CONCLUSION: The combinational immunotherapy, including varlilumab, was well-tolerated and induced vaccine-reactive T-cell expansion in the peripheral blood but without a detectable response in the tumor. Further developments of strategies to overcome the blood-tumor barrier are warranted to improve the efficacy of immunotherapy for LGG patients.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Vacunas contra el Cáncer , Glioma , Péptidos , Humanos , Proyectos Piloto , Leucocitos Mononucleares , Estudios Prospectivos , Glioma/tratamiento farmacológico , Diferenciación Celular , Microambiente Tumoral
2.
Cancer Immunol Res ; 11(7): 925-945, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37172100

RESUMEN

IMA101 is an actively personalized, multi-targeted adoptive cell therapy (ACT), whereby autologous T cells are directed against multiple novel defined peptide-HLA (pHLA) cancer targets. HLA-A*02:01-positive patients with relapsed/refractory solid tumors expressing ≥1 of 8 predefined targets underwent leukapheresis. Endogenous T cells specific for up to 4 targets were primed and expanded in vitro. Patients received lymphodepletion (fludarabine, cyclophosphamide), followed by T-cell infusion and low-dose IL2 (Cohort 1). Patients in Cohort 2 received atezolizumab for up to 1 year (NCT02876510). Overall, 214 patients were screened, 15 received lymphodepletion (13 women, 2 men; median age, 44 years), and 14 were treated with T-cell products. IMA101 treatment was feasible and well tolerated. The most common adverse events were cytokine release syndrome (Grade 1, n = 6; Grade 2, n = 4) and expected cytopenias. No patient died during the first 100 days after T-cell therapy. No neurotoxicity was observed. No objective responses were noted. Prolonged disease stabilization was noted in three patients lasting for 13.7, 12.9, and 7.3 months. High frequencies of target-specific T cells (up to 78.7% of CD8+ cells) were detected in the blood of treated patients, persisted for >1 year, and were detectable in posttreatment tumor tissue. Individual T-cell receptors (TCR) contained in T-cell products exhibited broad variation in TCR avidity, with the majority being low avidity. High-avidity TCRs were identified in some patients' products. This study demonstrates the feasibility and tolerability of an actively personalized ACT directed to multiple defined pHLA cancer targets. Results warrant further evaluation of multi-target ACT approaches using potent high-avidity TCRs. See related Spotlight by Uslu and June, p. 865.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias , Adulto , Femenino , Humanos , Masculino , Linfocitos T CD8-positivos , Estudios de Factibilidad , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Neoplasias/etiología , Receptores de Antígenos de Linfocitos T/genética
3.
Mol Cell Proteomics ; 18(6): 1255-1268, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31154438

RESUMEN

Glioblastoma multiforme (GBM) is the most aggressive brain tumor with poor prognosis to most patients. Immunotherapy of GBM is a potentially beneficial treatment option, whose optimal implementation may depend on familiarity with tumor specific antigens, presented as HLA peptides by the GBM cells. Further, early detection of GBM, such as by a routine blood test, may improve survival, even with the current treatment modalities. This study includes large-scale analyses of the HLA peptidome (immunopeptidome) of the plasma-soluble HLA molecules (sHLA) of 142 plasma samples, and the membranal HLA of GBM tumors of 10 of these patients' tumor samples. Tumor samples were fresh-frozen immediately after surgery and the plasma samples were collected before, and at multiple visits after surgery. In total, this HLA peptidome analysis involved 52 different HLA allotypes and resulted in the identification of more than 35,000 different HLA peptides. Strong correlations were observed in the signal intensities and in the repertoires of identified peptides between the tumors and plasma-soluble HLA peptidomes of the individual patients, whereas low correlations were observed between these HLA peptidomes and the tumors' proteomes. HLA peptides derived from Cancer/Testis Antigens (CTAs) were selected based on their presence among the HLA peptidomes of the patients and absence of expression of their source genes from any healthy and essential human tissues, except from immune-privileged sites. Additionally, peptides were selected as potential biomarkers if their levels in the plasma-sHLA peptidome were significantly reduced after the removal of tumor mass. The CTAs identified among the analyzed HLA peptidomes provide new opportunities for personalized immunotherapy and for early diagnosis of GBM.


Asunto(s)
Antígenos de Neoplasias/sangre , Neoplasias Encefálicas/sangre , Glioblastoma/sangre , Antígenos de Histocompatibilidad Clase I/sangre , Péptidos/sangre , Proteoma/metabolismo , Alelos , Biomarcadores de Tumor/sangre , Neoplasias Encefálicas/cirugía , Glioblastoma/cirugía , Humanos
6.
Nature ; 565(7738): 240-245, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30568303

RESUMEN

Patients with glioblastoma currently do not sufficiently benefit from recent breakthroughs in cancer treatment that use checkpoint inhibitors1,2. For treatments using checkpoint inhibitors to be successful, a high mutational load and responses to neoepitopes are thought to be essential3. There is limited intratumoural infiltration of immune cells4 in glioblastoma and these tumours contain only 30-50 non-synonymous mutations5. Exploitation of the full repertoire of tumour antigens-that is, both unmutated antigens and neoepitopes-may offer more effective immunotherapies, especially for tumours with a low mutational load. Here, in the phase I trial GAPVAC-101 of the Glioma Actively Personalized Vaccine Consortium (GAPVAC), we integrated highly individualized vaccinations with both types of tumour antigens into standard care to optimally exploit the limited target space for patients with newly diagnosed glioblastoma. Fifteen patients with glioblastomas positive for human leukocyte antigen (HLA)-A*02:01 or HLA-A*24:02 were treated with a vaccine (APVAC1) derived from a premanufactured library of unmutated antigens followed by treatment with APVAC2, which preferentially targeted neoepitopes. Personalization was based on mutations and analyses of the transcriptomes and immunopeptidomes of the individual tumours. The GAPVAC approach was feasible and vaccines that had poly-ICLC (polyriboinosinic-polyribocytidylic acid-poly-L-lysine carboxymethylcellulose) and granulocyte-macrophage colony-stimulating factor as adjuvants displayed favourable safety and strong immunogenicity. Unmutated APVAC1 antigens elicited sustained responses of central memory CD8+ T cells. APVAC2 induced predominantly CD4+ T cell responses of T helper 1 type against predicted neoepitopes.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Glioblastoma/diagnóstico , Glioblastoma/terapia , Medicina de Precisión/métodos , Adulto , Anciano , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Femenino , Glioblastoma/inmunología , Antígenos HLA-A/inmunología , Humanos , Memoria Inmunológica/inmunología , Masculino , Persona de Mediana Edad , Linfocitos T Colaboradores-Inductores/inmunología , Resultado del Tratamiento
7.
Mol Cell Proteomics ; 17(11): 2132-2145, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30072578

RESUMEN

Glioblastoma multiforme (GBM) is the most aggressive brain tumor with poor prognosis to most patients. Immunotherapy of GBM is a potentially beneficial treatment option, whose optimal implementation may depend on familiarity with tumor specific antigens, presented as HLA peptides by the GBM cells. Furthermore, early detection of GBM, such as by a routine blood test, may improve survival, even with the current treatment modalities. This study includes large-scale analyses of the HLA peptidome (immunopeptidome) of the plasma-soluble HLA molecules (sHLA) of 142 plasma samples, and the membranal HLA of GBM tumors of 10 of these patients' tumor samples. Tumor samples were fresh-frozen immediately after surgery and the plasma samples were collected before, and at multiple visits after surgery. In total, this HLA peptidome analysis involved 52 different HLA allotypes and resulted in the identification of more than 35,000 different HLA peptides. Strong correlations were observed in the signal intensities and in the repertoires of identified peptides between the tumors and plasma-soluble HLA peptidomes of the individual patients, whereas low correlations were observed between these HLA peptidomes and the tumors' proteomes. HLA peptides derived from Cancer/Testis Antigens (CTAs) were selected based on their presence among the HLA peptidomes of the patients and absence of expression of their source genes from any healthy and essential human tissues, except from immune-privileged sites. Additionally, peptides were selected as potential biomarkers if their levels in the plasma-sHLA peptidome were significantly reduced after the removal of tumor mass. The CTAs identified among the analyzed HLA peptidomes provide new opportunities for personalized immunotherapy and for early diagnosis of GBM.


Asunto(s)
Antígenos de Neoplasias/sangre , Glioblastoma/sangre , Antígenos HLA/metabolismo , Péptidos/metabolismo , Proteoma/metabolismo , Alelos , Secuencia de Aminoácidos , Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/sangre , Membrana Celular/metabolismo , Glioblastoma/cirugía , Humanos , Péptidos/sangre , Péptidos/química , Solubilidad
8.
Clin Cancer Res ; 22(19): 4776-4785, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27225692

RESUMEN

PURPOSE: To perform a two-cohort, phase I safety and immunogenicity study of IMA950 in addition to standard chemoradiotherapy and adjuvant temozolomide in patients with newly diagnosed glioblastoma. IMA950 is a novel glioblastoma-specific therapeutic vaccine containing 11 tumor-associated peptides (TUMAP), identified on human leukocyte antigen (HLA) surface receptors in primary human glioblastoma tissue. EXPERIMENTAL DESIGN: Patients were HLA-A*02-positive and had undergone tumor resection. Vaccination comprised 11 intradermal injections with IMA950 plus granulocyte macrophage colony-stimulating factor (GM-CSF) over a 24-week period, beginning 7 to 14 days prior to initiation of chemoradiotherapy (Cohort 1) or 7 days after chemoradiotherapy (Cohort 2). Safety was assessed according to NCI CTCAE Version 4.0 and TUMAP-specific T-cell immune responses determined. Secondary observations included progression-free survival (PFS), pretreatment regulatory T cell (Treg) levels, and the effect of steroids on T-cell responses. RESULTS: Forty-five patients were recruited. Related adverse events included minor injection site reactions, rash, pruritus, fatigue, neutropenia and single cases of allergic reaction, anemia and anaphylaxis. Two patients experienced grade 3 dose-limiting toxicity of fatigue and anaphylaxis. Of 40 evaluable patients, 36 were TUMAP responders and 20 were multi-TUMAP responders, with no important differences between cohorts. No effect of pretreatment Treg levels on IMA950 immunogenicity was observed, and steroids did not affect TUMAP responses. PFS rates were 74% at 6 months and 31% at 9 months. CONCLUSIONS: IMA950 plus GM-CSF was well-tolerated with the primary immunogenicity endpoint of observing multi-TUMAP responses in at least 30% of patients exceeded. Further development of IMA950 is encouraged. Clin Cancer Res; 22(19); 4776-85. ©2016 AACRSee related commentary by Lowenstein and Castro, p. 4760.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Vacunas contra el Cáncer/uso terapéutico , Glioblastoma/tratamiento farmacológico , Péptidos/uso terapéutico , Adulto , Anciano , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/mortalidad , Quimioradioterapia/métodos , Supervivencia sin Enfermedad , Femenino , Glioblastoma/mortalidad , Humanos , Estimación de Kaplan-Meier , Activación de Linfocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Linfocitos T/efectos de los fármacos , Reino Unido , Adulto Joven
9.
Nat Med ; 18(8): 1254-61, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22842478

RESUMEN

IMA901 is the first therapeutic vaccine for renal cell cancer (RCC) consisting of multiple tumor-associated peptides (TUMAPs) confirmed to be naturally presented in human cancer tissue. We treated a total of 96 human leukocyte antigen A (HLA-A)*02(+) subjects with advanced RCC with IMA901 in two consecutive studies. In the phase 1 study, the T cell responses of the patients to multiple TUMAPs were associated with better disease control and lower numbers of prevaccine forkhead box P3 (FOXP3)(+) regulatory T (T(reg)) cells. The randomized phase 2 trial showed that a single dose of cyclophosphamide reduced the number of T(reg) cells and confirmed that immune responses to multiple TUMAPs were associated with longer overall survival. Furthermore, among six predefined populations of myeloid-derived suppressor cells, two were prognostic for overall survival, and among over 300 serum biomarkers, we identified apolipoprotein A-I (APOA1) and chemokine (C-C motif) ligand 17 (CCL17) as being predictive for both immune response to IMA901 and overall survival. A randomized phase 3 study to determine the clinical benefit of treatment with IMA901 is ongoing.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Células Renales/terapia , Ciclofosfamida/uso terapéutico , Inmunosupresores/uso terapéutico , Inmunoterapia Activa , Neoplasias Renales/terapia , Linfocitos T Reguladores/inmunología , Vacunas de Subunidad/uso terapéutico , Antígenos de Neoplasias/inmunología , Apolipoproteína A-I/sangre , Biomarcadores , Linfocitos T CD8-positivos/inmunología , Carcinoma de Células Renales/sangre , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/inmunología , Quimiocina CCL17/sangre , Terapia Combinada , Ciclofosfamida/administración & dosificación , Ciclofosfamida/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Antígeno HLA-A2/inmunología , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacología , Estimación de Kaplan-Meier , Neoplasias Renales/sangre , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/inmunología , Depleción Linfocítica , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Linfocitos T Reguladores/efectos de los fármacos , Resultado del Tratamiento
10.
Brain ; 135(Pt 4): 1042-54, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22418738

RESUMEN

Peptides presented at the cell surface reflect the protein content of the cell; those on HLA class I molecules comprise the critical peptidome elements interacting with CD8 T lymphocytes. We hypothesize that peptidomes from ex vivo tumour samples encompass immunogenic tumour antigens. Here, we uncover >6000 HLA-bound peptides from HLA-A*02(+) glioblastoma, of which over 3000 were restricted by HLA-A*02. We prioritized in-depth investigation of 10 glioblastoma-associated antigens based on high expression in tumours, very low or absent expression in healthy tissues, implication in gliomagenesis and immunogenicity. Patients with glioblastoma showed no T cell tolerance to these peptides. Moreover, we demonstrated specific lysis of tumour cells by patients' CD8(+) T cells in vitro. In vivo, glioblastoma-specific CD8(+) T cells were present at the tumour site. Overall, our data show the physiological relevance of the peptidome approach and provide a critical advance for designing a rational glioblastoma immunotherapy. The peptides identified in our study are currently being tested as a multipeptide vaccine (IMA950) in patients with glioblastoma.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Péptidos/inmunología , Presentación de Antígeno/fisiología , Antígenos CD/metabolismo , Antígenos de Neoplasias/química , Antígenos de Neoplasias/uso terapéutico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Linfocitos T CD8-positivos/inmunología , Cromatografía Liquida , Citocinas/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/metabolismo , Glioblastoma/patología , Glioblastoma/terapia , Antígenos HLA-A/análisis , Antígenos HLA-A/química , Antígenos HLA-A/inmunología , Humanos , Espectrometría de Masas , Análisis de Secuencia por Matrices de Oligonucleótidos , Péptidos/análisis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Análisis de Secuencia de Proteína
11.
Blood ; 111(12): 5610-20, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18310500

RESUMEN

The tyrosine kinase inhibitors sorafenib and sunitinib are approved for the treatment of patients with malignant diseases. To analyze the possible use of these compounds in combination with immunotherapeutic approaches, we analyzed the effects of both inhibitors on the immunostimulatory capacity of human dendritic cells (DCs) and the induction of primary immune responses in vivo. Sorafenib, but not sunitinib, inhibits function of DCs, characterized by reduced secretion of cytokines and expression of CD1a, major histocompatibility complex, and costimulatory molecules in response to TLR ligands as well as by their impaired ability to migrate and stimulate T-cell responses. These inhibitory effects are mediated by inhibition of PI3 and MAP kinases and NFkappaB signaling. In contrast, sorafenib had no influence on the phenotype and proliferation of T cells. To analyze the effects of both TKIs on cytotoxic T-cell induction in vivo, C57BL/6 mice were pretreated with sorafenib or sunitinib and immunized with OVA(257-264) peptide. Sorafenib, but not sunitinib, application significantly reduced the induction of antigen-specific T cells. Numbers of regulatory T cells were reduced in peripheral blood mononuclear cells from mice treated with sunitinib. These results indicate that sunitinib, but not sorafenib, is suitable for combination with immunotherapeutic approaches for treatment of cancer patients.


Asunto(s)
Antineoplásicos/farmacología , Bencenosulfonatos/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Indoles/farmacología , Piridinas/farmacología , Pirroles/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/inmunología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/citología , Dextranos/farmacocinética , Endocitosis/efectos de los fármacos , Endocitosis/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interleucina-4/farmacología , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos C57BL , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Sorafenib , Sunitinib , Linfocitos T Reguladores/efectos de los fármacos , Receptor Toll-Like 4/metabolismo
12.
Cell Stress Chaperones ; 10(3): 221-9, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16184767

RESUMEN

Heat shock proteins (Hsps) are able to induce protective immune responses against pathogens and tumors after injection into immunocompetent hosts. The activation of components of the adaptive immune system, including cytotoxic T lymphocytes specific for pathogen- or tumor-derived peptides, is crucial for the establishment of immunoprotection. Hsps acquire these peptides during intracellular protein degradation and when released during necrotic cell death, facilitate their uptake and Minor Histocompatibility Complex (MHC)-restricted representation by professional antigen-presenting cells (APCs). In addition, the interaction of Hsps with APCs, including the Endoplasmatic Reticulum (ER)-resident chaperone glycoprotein 96 (Gp96), induces the maturation of these cells by Toll-like receptor (TLR)-mediated signaling events. We now provide evidence that in contrast to lipopolysaccharides (LPS)-mediated dendritic cell (DC) maturation, the interaction of Gp96 with DCs leads to the preferential expansion of antigen-specific CD8-positive T cells in vitro and in vivo. This CD8 preference induced by mouse and human DCs did not correlate with enhanced levels of interleukin-12 secretion. Thus, despite the fact that both LPS and Gp96 activate DCs in a TLR4-dependent manner, the experiments of this study clearly demonstrate qualitative differences in the outcome of this maturation process, which preferentially favors the expansion of CD8-positive T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , Subgrupos Linfocitarios/inmunología , Glicoproteínas de Membrana/farmacología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Humanos , Mediadores de Inflamación/inmunología , Lipopolisacáridos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4 , Receptores Toll-Like
13.
J Immunol ; 175(2): 891-8, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16002687

RESUMEN

Influenza virus infection induces maturation of murine dendritic cells (DCs), which is most important for the initiation of an immune response. However, in contrast to EL-4 and MC57 cells, DCs present viral CTL epitopes with a delay of up to 10 h. This delay in Ag presentation coincides with the up-regulation of MHC class I molecules as well as costimulatory molecules on the cell surface and the accumulation of newly synthesized ubiquitinated proteins in large cytosolic structures, called DC aggresome-like-induced structures (DALIS). These structures were observed previously after LPS-induced maturation of DCs, and it was speculated that they play a role in the regulation of MHC class I Ag presentation. Our findings provide the first evidence for a connection between DC maturation, MHC class I-restricted Ag presentation, and DALIS formation, which is further supported by the observation that DALIS contain ubiquitinated influenza nucleoprotein.


Asunto(s)
Presentación de Antígeno/inmunología , Estructuras Citoplasmáticas/inmunología , Estructuras Citoplasmáticas/virología , Células Dendríticas/citología , Células Dendríticas/inmunología , Virus de la Influenza A/inmunología , Animales , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/virología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/virología , Diferenciación Celular/inmunología , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Estructuras Citoplasmáticas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/virología , Epítopos de Linfocito T/biosíntesis , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/metabolismo , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Proteínas de la Nucleocápside , Nucleoproteínas/biosíntesis , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores Inmunológicos/fisiología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/virología , Factores de Tiempo , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Ubiquitina/metabolismo , Proteínas del Núcleo Viral/biosíntesis , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo
14.
J Endotoxin Res ; 11(2): 89-96, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15949135

RESUMEN

In this study, we analyzed the activation of bone-marrow derived dendritic cells (BMDCs) from mice lacking the cd14-gene with purified Legionella pneumophila lipopolysaccharide and with viable or formalin-killed L. pneumophila. We found that low concentrations of LPS and doses of L. pneumophila that are relevant to infection are dependent on CD14 to activate BMDCs. Higher concentrations of LPS are able to overcome the lack of CD14 indicating that other receptors areinvolved. We, therefore, included studies using BMDCs from mice lacking functional TLR2 and/or TLR4 molecules. We found that purified L. pneumophila LPS as well as L. pneumophila either viable or formalin-killed are able to activate BMDCs from TLR4-deficient C3H/HeJ mice but fail to activate BMDCs from TLR2-knockout mice. Our data show that not only purified LPS from L. pneumophila but also the microorganism itself stimulate BMDCs via TLR2 and that this stimulation is dependent on CD14 in this mouse model.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Legionella pneumophila/química , Receptores de Lipopolisacáridos/fisiología , Lipopolisacáridos/farmacología , Receptores Inmunológicos/fisiología , Animales , Células de la Médula Ósea/fisiología , Citocinas/metabolismo , Indicadores y Reactivos , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Legionella pneumophila/inmunología , Receptores de Lipopolisacáridos/genética , Lipopolisacáridos/química , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Noqueados , Receptores Inmunológicos/genética , Salmonella typhimurium/química , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2
15.
Blood ; 101(7): 2810-5, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12446445

RESUMEN

The endoplasmic reticulum (ER)-resident heat shock protein Gp96 is involved in protein folding and is released into the extracellular space after necrotic cell death. In this context, Gp96 has immunostimulatory properties: it activates dendritic cells or macrophages and delivers associated peptides into the antigen presentation pathway, resulting in the induction of specific T-cell responses. The inflammatory response after necrotic tissue damage leads to the recruitment of polymorphonuclear neutrophils (PMNs) and monocytes, allowing them to make their first encounter with Gp96. We therefore investigated whether PMNs and monocytes interact with Gp96. We were able to show that PMNs and monocytes specifically bind fluorescein isothiocyanate (FITC)-conjugated Gp96. The binding of Gp96-FITC was competed by lipopolysaccharide (LPS) or fucoidan, a known inhibitor of scavenger receptors. Interestingly, the binding of LPS-FITC was also competed not only by fucoidan, but by Gp96, suggesting that LPS and Gp96 share a common receptor on PMNs. One important effector function of PMNs is the clearance of an inflammatory site by phagocytosis. We therefore assessed the influence of Gp96 on phagocytic activity using fluorochrome-labeled polystyrene beads. We found a marked enhancement of phagocytosis in the presence of Gp96 and concluded that PMNs not only bind Gp96, but are also activated by it. Additionally, Gp96-stimulated PMNs and especially monocytes release large amounts of interleukin-8, a potent neutrophil-attracting chemokine. In conclusion, we demonstrate that Gp96 specifically binds to and activates PMNs and monocytes, extending the function of Gp96 as a danger signal to additional members of the innate immune system.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Monocitos/metabolismo , Neutrófilos/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/farmacología , Citometría de Flujo , Fluoresceína-5-Isotiocianato , Humanos , Interleucina-8/biosíntesis , Ratones , Monocitos/efectos de los fármacos , Monocitos/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Fagocitosis/efectos de los fármacos , Unión Proteica/inmunología
16.
Blood ; 99(10): 3676-82, 2002 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11986223

RESUMEN

Immunizations using the endoplasmic reticulum-resident heat shock protein Gp96 induce specific immune responses. Specificity is based on the major histocompatibility complex class I-restricted cross-presentation of Gp96-associated peptides derived from endogenous proteins. Initiation of the immune response depends on the ability of Gp96 to induce the production of proinflammatory cytokines by macrophages and dendritic cells (DCs) and of their maturation in a fashion presumably independent of associated peptide. Both events are mediated by Gp96 receptors on antigen-presenting cells. It is known that Gp96 is released from cells at necrosis induced, for example, by virus infection. Although this event supports the efficient induction of immune responses, it might also interfere with processes that are susceptible to chronic inflammation, such as wound healing after tissue damage. Therefore, Gp96-mediated stimulation of the immune system requires tight regulation. Here we show that human thrombocytes specifically interact with Gp96 and that binding of Gp96 to platelets is enhanced more than 10-fold on activation by thrombin. Gp96 interferes with neither thrombin-induced platelet activation nor platelet aggregation. However, the presence of platelets during Gp96-mediated DC activation reduces the secretion of proinflammatory cytokines and the activation of DCs. This effect is independent of soluble platelet factors and cell-to-cell contact between DCs and thrombocytes. Thus, we provide evidence for a regulatory mechanism that neutralizes Gp96 molecules systemically, especially in the blood. This effect might be of significance in wounds in which chronic inflammation and immune responses against autoantigens have to be prevented.


Asunto(s)
Antígenos de Neoplasias/farmacología , Plaquetas/metabolismo , Plaquetas/fisiología , Células Dendríticas/inmunología , Antígenos de Neoplasias/metabolismo , Unión Competitiva , Diferenciación Celular , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Fluoresceína-5-Isotiocianato/química , Colorantes Fluorescentes/química , Humanos , Cinética , Activación Plaquetaria , Agregación Plaquetaria , Receptores de Superficie Celular/metabolismo
17.
J Biol Chem ; 277(23): 20847-53, 2002 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-11912201

RESUMEN

The heat shock protein Gp96 has been shown to induce specific immune responses. On one hand, this phenomenon is based on the specific interaction with CD91 that mediates endocytosis and results in major histocompatibility complex class I-restricted representation of the Gp96-associated peptides. On the other hand, Gp96 induces activation of professional antigen-presenting cells, resulting in the production of pro-inflammatory cytokines and up-regulation of costimulatory molecules by unknown mechanisms. In this study, we have analyzed the consequences of Gp96 interaction with cells expressing different Toll-like receptors (TLRs) and with bone marrow-derived dendritic cells from mice lacking functional TLR2 and/or TLR4 molecules. We find that the Gp96-TLR2/4 interaction results in activation of nuclear factor kappaB-driven reporter genes and mitogen- and stress-activated protein kinases and induces IkappaBalpha degradation. Bone marrow-derived dendritic cells of C3H/HeJ and more pronounced C3H/HeJ/TLR2(-/-) mice fail to respond to Gp96. Interestingly, activation of bone marrow-derived dendritic cells depends on endocytosis of Gp96 molecules. Our results provide, for the first time, the molecular basis for understanding the Gp96-mediated activation of antigen-presenting cells by describing the simultaneous stimulation of the innate and adaptive immune system. This feature explains the remarkable ability of Gp96 to induce specific immune responses against tumors and pathogens.


Asunto(s)
Células Dendríticas/inmunología , Proteínas de Drosophila , Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Receptores de Superficie Celular/genética , Transducción de Señal , Receptor Toll-Like 2 , Receptor Toll-Like 4 , Receptores Toll-Like
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA