Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 34(13): 1650-7, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24793791

RESUMO

The signal transducer and activator of transcription 3 (STAT3) can be activated by the tyrosine kinase domain of the chimeric protein nucleophosmin/anaplastic lymphoma kinase (NPM/ALK), and has a pivotal role in mediating NPM/ALK-related malignant cell transformation. Although the role of STAT3 and wild-type NPM in oncogenesis has been extensively investigated, the relationship between both molecules in cancer remains poorly understood. In the present study, we first demonstrate that STAT3 phosphorylation at tyrosine 705 is accompanied by a concomitant increase in the expression level of NPM. Nuclear co-translocation of phosphorylated STAT3 with NPM can be triggered by interferon-alpha (IFN-α) stimulation of Jurkat cells and phosphorylated STAT3 co-localizes with NPM in cancer cells showing constitutive STAT3 activation. We further demonstrate that STAT3 phosphorylation can transcriptionally mediate NPM upregulation in IFN-α-stimulated Jurkat cells and is responsible for maintaining its expression in cancer cells showing constitutive STAT3 activation. Inhibition of STAT3 phosphorylation or knockdown of NPM expression abrogates their simultaneous transnuclear movements. Finally, we found evidence for a physical interaction between NPM and STAT3 in conditions of STAT3 activation. In conclusion, NPM is a downstream effector of the STAT3 signaling, and can facilitate the nuclear entry of phosphorylated STAT3. These observations might open novel opportunities for targeting the STAT3 pathway in cancer.


Assuntos
Neoplasias/metabolismo , Proteínas Nucleares/fisiologia , Fator de Transcrição STAT3/metabolismo , Transcrição Gênica , Linhagem Celular Tumoral , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Interferon-alfa/farmacologia , Proteínas Nucleares/genética , Nucleofosmina , Fosforilação , Transporte Proteico , Transdução de Sinais/fisiologia
2.
Gene Ther ; 21(3): 262-71, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24401835

RESUMO

The release of cytokines by T cells strongly defines their functional activity in vivo. The ability to produce multiple cytokines has been associated with beneficial immune responses in cancer and infectious diseases, while their progressive loss is associated with T-cell exhaustion, senescence and anergy. Consequently, strategies that enhance the multifunctional status of T cells are a key for immunotherapy. Dendritic cells (DCs) are professional antigen presenting cells that regulate T-cell functions by providing positive and negative co-stimulatory signals. A key negative regulator of T-cell activity is provided by binding of programmed death-1 (PD-1) receptor on activated T cells, to its ligand PD-L1, expressed on DCs. We investigated the impact of interfering with PD-L1/PD-1 co-stimulation on the multifunctionality of T cells, by expression of the soluble extracellular part of PD-1 (sPD-1) or PD-L1 (sPD-L1) in human monocyte-derived DCs during antigen presentation. Expression, secretion and binding of these soluble molecules after mRNA electroporation were demonstrated. Modification of DCs with sPD-1 or sPD-L1 mRNA resulted in increased levels of the co-stimulatory molecule CD80 and a distinct cytokine profile, characterized by the secretion of IL-10 and TNF-α, respectively. Co-expression in DCs of sPD-1 and sPD-L1 with influenza virus nuclear protein 1 (Flu NP1) stimulated Flu NP1 memory T cells, with a significantly higher number of multifunctional T cells and increased cytokine secretion, while it did not induce regulatory T cells. These data provide a rationale for the inclusion of interfering sPD-1 or sPD-L1 in DC-based immunotherapeutic strategies.


Assuntos
Apresentação de Antígeno , Antígenos B7/imunologia , Células Dendríticas/imunologia , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T/imunologia , Antígenos B7/genética , Antígenos B7/metabolismo , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Células Dendríticas/metabolismo , Células HEK293 , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Proteínas do Nucleocapsídeo , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
3.
Mol Hum Reprod ; 20(2): 168-77, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24217388

RESUMO

Gain of 20q11.21 is a chromosomal abnormality that is recurrently found in human pluripotent stem cells and cancers, strongly suggesting that this mutation confers a proliferative or survival advantage to these cells. In this work we studied three human embryonic stem cell (hESC) lines that acquired a gain of 20q11.21 during in vitro culture. The study of the mRNA gene expression levels of the loci located in the common region of duplication showed that HM13, ID1, BCL2L1, KIF3B and the immature form of the micro-RNA miR-1825 were up-regulated in mutant cells. ID1 and BCL2L1 were further studied as potential drivers of the phenotype of hESC with a 20q11.21 gain. We found no increase in the protein levels of ID1, nor the downstream effects expected from over-expression of this gene. On the other hand, hESC with a gain of 20q11.21 had on average a 3-fold increase of Bcl-xL (the anti-apoptotic isoform of BCL2L1) protein levels. The mutant hESC underwent 2- to 3-fold less apoptosis upon loss of cell-to-cell contact and were ∼2-fold more efficient in forming colonies from a single cell. The key role of BCL2L1 in this mutation was further confirmed by transgenic over-expression of BCL2L1 in the wild-type cells, leading to apoptosis-resistant cells, and BCL2L1-knock-down in the mutant hESC, resulting in a restoration of the wild-type phenotype. This resistance to apoptosis supposes a significant advantage for the mutant cells, explaining the high frequency of gains of 20q11.21 in human pluripotent stem cells.


Assuntos
Duplicação Cromossômica , Cromossomos Humanos Par 20 , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteína bcl-X/genética , Apoptose , Linhagem Celular , Sobrevivência Celular , Mapeamento Cromossômico , Células-Tronco Embrionárias/patologia , Expressão Gênica , Técnicas de Inativação de Genes , Loci Gênicos , Humanos , Células-Tronco Pluripotentes/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína bcl-X/antagonistas & inibidores , Proteína bcl-X/metabolismo
4.
Leukemia ; 28(4): 830-41, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24091848

RESUMO

Peripheral blood T cells transduced with a tumor-specific T-cell receptor (TCR) face problems of auto-reactivity and lack of efficacy caused by cross-pairing of exogenous and endogenous TCR chains, as well as short term in vivo survival due to activation and growth factor-induced differentiation. We here studied an alternative strategy for the efficient generation of naive CD8(+) T cells with a single TCR. TCR-transduced human postnatal thymus-derived and adult mobilized blood-derived hematopoietic progenitor cells (HPCs) were differentiated to CD4(+)CD8(+) double-positive T cells using OP9-Delta-like 1 (OP9-DL1) cultures. Addition of the agonist peptide induced double positive cells to cross-present the peptide, leading, in the absence of co-stimulation, to cell cycle arrest and differentiation into mature CD8(+) T cells. Comprehensive phenotypic, molecular and functional analysis revealed the generation of naive and resting CD8(+) T cells through a process similar to thymic positive selection. These mature T cells show a near complete inhibition of endogenous TCRA and TCRB rearrangements and express high levels of the introduced multimer-reactive TCR. Upon activation, specific cytokine production and efficient killing of tumor cells were induced. Using this strategy, large numbers of high-avidity tumor-specific naive T cells can be generated from readily available HPCs without TCR chain cross-pairing.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Receptores de Antígenos de Linfócitos T/fisiologia , Adulto , Diferenciação Celular , Linhagem Celular Tumoral , Criança , Pré-Escolar , Rearranjo Gênico do Linfócito T , Humanos , Imunoterapia Adotiva , Lactente , Recém-Nascido , Receptores de Antígenos de Linfócitos T/agonistas
5.
Ann Oncol ; 24(10): 2686-2693, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23904461

RESUMO

BACKGROUND: Autologous monocyte-derived dendritic cells (DCs) electroporated with synthetic messenger RNA (mRNA) encoding a CD40 ligand, a constitutively active Toll-like receptor 4 and CD70, together with mRNA encoding fusion proteins of a human leukocyte antigen (HLA)-class II targeting signal (DC-LAMP) and a melanoma-associated antigen (MAA); either MAGE-A3, MAGE-C2, tyrosinase or gp100) (TriMixDC-MEL) are superiorly immunogenic. PATIENTS AND METHODS: In this phase IB clinical trial, 24 million viable DCs were administered by four biweekly combined intradermal (id) and intravenous (iv) administrations, and a fifth administration on week 16. The number of iv-administered DCs was escalated in four sequentially treated cohorts. Immune responses were assessed by analysis of antigen specificity of blood-derived T-cells and skin infiltrating lymphocytes (SKILs). RESULTS: Fifteen patients with pretreated advanced melanoma tolerated administration of TriMixDC-MEL well. Two patients achieved a complete response and two patients a partial response. All objective responders are progression-free after a follow-up of, respectively, 24+, 28+, 33+, and 34+ months. Post-therapy antigen-specific SKILs were documented in 6 of 12 patients, and antigen-specific CD8(+) T-cells were detected in the blood of 4 of 5 patients. CONCLUSIONS: Cellular immunotherapy with TriMixDC-MEL is safe and immunogenic. Antitumor activity with durable disease control is observed across the investigated iv-dose levels. CLINICALTRIALSGOV IDENTIFIER: NCT01066390.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Dendríticas/imunologia , Imunoterapia/métodos , Melanoma/terapia , Neoplasias Cutâneas/terapia , Adulto , Idoso , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Ligante CD27/genética , Ligante CD27/metabolismo , Ligante de CD40/genética , Ligante de CD40/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/citologia , Intervalo Livre de Doença , Eletroporação , Feminino , Humanos , Proteína 3 de Membrana Associada ao Lisossomo/genética , Proteína 3 de Membrana Associada ao Lisossomo/metabolismo , Masculino , Pessoa de Meia-Idade , Monofenol Mono-Oxigenase/genética , Monofenol Mono-Oxigenase/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
6.
Gene Ther ; 20(11): 1085-92, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23804077

RESUMO

Persistent activation of the transcription factor, signal transducer and activator of transcription 3 (Stat3) has been shown to mediate several oncogenic features in many types of cancers, including melanoma. In this study, we investigated whether lentiviral (LV) delivery of Stat3-targeting short hairpin RNA (shRNA; LV-shStat3) to K1735-C4 melanoma cells modulates antitumor immunity. Three shStat3 sequences, starting at the position 446, 830 and 1412, were cloned into a mir30 cassette. A shRNA with scrambled sequence served as a control. Transduction with LV-shStat3 resulted in downregulation of Stat3 in vitro. The latter coincided with low cell viability, a reduced expression of survivin and matrix metalloproteinase (MMP)-2. A single injection of LV-shStat3 in K1735-C4 tumors efficiently downregulated Stat3 in vivo and resulted in reduction of both vascular endothelial growth factor secretion and in myeloid-derived suppressor cell (MDSC) numbers. In contrast, we observed an increase in interleukin-6 and interferon-γ secretion, mature dendritic cells (DCs) and CD8(+) T cells. Both DCs and CD8(+) T cells displayed enhanced activity, whereas granulocytic MDSCs lost their suppressive capacity upon Stat3 downregulation. Importantly, a single injection of LV-shStat3 was sufficient to reduce tumor growth, hence prolong survival of tumor-bearing mice. These data demonstrate that Stat3 downregulation in melanoma reinvigorates existing antitumor immunity.


Assuntos
Melanoma Experimental/genética , Melanoma/genética , Fator de Transcrição STAT3/genética , Microambiente Tumoral/imunologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos , Células HEK293 , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Lentivirus/genética , Metaloproteinase 2 da Matriz/metabolismo , Melanoma/imunologia , Melanoma/metabolismo , Melanoma/terapia , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/administração & dosagem , Proteínas Repressoras/metabolismo , Fator de Transcrição STAT3/metabolismo , Survivina , Transdução Genética , Células Tumorais Cultivadas
7.
Tissue Antigens ; 79(3): 174-85, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22309258

RESUMO

Antigen-specific immunity is crucially important for containing viral replication in human immunodeficiency virus (HIV)-1-infected hosts. Several epitopes have been predicted for the early expressed HIV-1 proteins Tat and Rev, but few have been studied in detail. We characterized the human leukocyte antigen (HLA)-B44-restricted Rev epitope EELLKTVRL (EL9) in an HIV-1-infected subject treated with antiretroviral therapy. Interestingly, a high sequence similarity was found between the EL9 epitope and the human nucleolar protein 6 (NOL6). However, this similarity does not seem to impede immunogenicity as CD8(+) T-cells, previously stimulated with EL9-pulsed dendritic cells, were able to specifically recognize the HIV-1 Rev epitope without cross-recognizing the human self-antigen NOL6. After the subject interrupted antiretroviral therapy and virus rebounded, mutations within the EL9 epitope were identified. Although the emerging mutations resulted in decreased or abolished T-cell recognition, they did not impair Rev protein function. Mutations leading to escape from T-cell recognition persisted for up to 124 weeks following treatment interruption. This study shows that the HLA-B44-restricted Rev CD8(+) T-cell epitope EL9 is immunogenic notwithstanding its close resemblance to a human peptide. The epitope mutates as a consequence of dynamic interaction between T-cells and HIV-1. Clinical status, CD4(+) T-cell count and viral load remained stable despite escape from T-cell recognition.


Assuntos
Evolução Molecular , Infecções por HIV/imunologia , HIV-1 , Proteínas Nucleares/genética , Produtos do Gene rev do Vírus da Imunodeficiência Humana/genética , Produtos do Gene rev do Vírus da Imunodeficiência Humana/imunologia , Sequência de Aminoácidos , Animais , Antirretrovirais/administração & dosagem , Sequência de Bases , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Infecções por HIV/tratamento farmacológico , HIV-1/genética , Antígenos HLA/genética , Células HeLa , Humanos , Ativação Linfocitária/imunologia , Mimetismo Molecular , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/química , Linfócitos T/imunologia , Linfócitos T/virologia
8.
Gene Ther ; 19(12): 1133-40, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22241177

RESUMO

Lentiviral vectors (LVs) provide unique opportunities for the development of immunotherapeutic strategies, as they transduce a variety of cells in situ, including antigen-presenting cells (APCs). Engineering LVs to specifically transduce APCs is required to promote their translation towards the clinic. We report on the Nanobody (Nb) display technology to target LVs to dendritic cells (DCs) and macrophages. This innovative approach exploits the budding mechanism of LVs to incorporate an APC-specific Nb and a binding-defective, fusion-competent form of VSV.G in the viral envelope. In addition to production of high titer LVs, we demonstrated selective, Nb-dependent transduction of mouse DCs and macrophages both in vitro and in situ. Moreover, this strategy was translated to a human model in which selective transduction of in vitro generated or lymph node (LN)-derived DCs and macrophages, was demonstrated. In conclusion, the Nb display technology is an attractive approach to generate LVs targeted to specific cell types.


Assuntos
Células Apresentadoras de Antígenos , Marcação de Genes/métodos , Vetores Genéticos , Lentivirus/genética , Macrófagos , Anticorpos de Domínio Único , Animais , Técnicas de Transferência de Genes , Humanos , Camundongos , Transdução Genética
9.
Gene Ther ; 13(13): 1027-36, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16511519

RESUMO

The maturation state of dendritic cells (DCs) is an important determinant for the initiation and regulation of adaptive immune responses. In this study, we wanted to assess whether functional activation of human monocyte-derived DCs can be achieved by electroporation of an activation signal in the form of double-stranded (ds) RNA and whether simultaneous electroporation of the dsRNA with tumor antigen encoding mRNA can lead to the induction of a cytotoxic T-lymphocyte (CTL) response. Electroporation of immature DCs with poly(I:C(12)U), a dsRNA analogue, resulted in phenotypic as well as functional changes, indicative of DC maturation. Co-electroporation of DCs with both poly(I:C(12)U) and Melan-A/MART-1 encoding mRNA induced strong anti-Melan-A/MART-1 CD8(+) T-cell responses in vitro. Higher numbers of Melan-A/MART-1-specific CTLs were consistently obtained with poly(I:C(12)U)-activated DCs compared to DCs matured in the presence of an inflammatory cytokine cocktail. These results indicate that DC co-electroporation with both dsRNA and tumor antigen encoding mRNA induces fully activated and antigen-loaded DCs that promote antigen-specific CTL responses and may provide the basis for future immunotherapeutic strategies.


Assuntos
Antígenos de Neoplasias/genética , Linfócitos T CD8-Positivos/imunologia , Terapia Genética/métodos , Imunoterapia/métodos , Melanoma Experimental/terapia , Neoplasias Cutâneas/terapia , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Testes Imunológicos de Citotoxicidade , Células Dendríticas/imunologia , Eletroporação/métodos , Citometria de Fluxo , Engenharia Genética , Humanos , Interferon gama/imunologia , Ativação Linfocitária , Antígeno MART-1 , Melanoma Experimental/imunologia , Proteínas de Neoplasias/genética , RNA/administração & dosagem , RNA de Cadeia Dupla/administração & dosagem , Neoplasias Cutâneas/imunologia , Células Tumorais Cultivadas
10.
Gene Ther ; 13(7): 630-40, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16355115

RESUMO

Ex vivo lentivirally transduced dendritic cells (DC) have been described to induce CD8+ and CD4+ T-cell responses against various tumor-associated antigens (TAAs) in vitro and in vivo. We report here that direct administration of ovalbumin (OVA) encoding lentiviral vectors caused in vivo transduction of cells that were found in draining lymph nodes (LNs) and induced potent anti-OVA cytotoxic T cells similar to those elicited by ex vivo transduced DC. The cytotoxic T-lymphocyte (CTL) response following direct injection of lentiviral vectors was highly effective in eliminating target cells in vivo up to 30 days after immunization and was efficiently recalled after a boost immunization. Injection of lentiviral vectors furthermore activated OVA-specific CD4+ T cells and this CD4 help was shown to be necessary for an adequate primary and memory CTL response. When tested in therapeutic tumor experiments with OVA+ melanoma cells, direct administration of lentiviral vectors slowed down tumor growth to a comparable extent with the highest dose of ex vivo transduced DC. Taken together, these data indicate that direct in vivo administration of lentiviral vectors encoding TAAs has strong potential for anticancer vaccination.


Assuntos
Células Dendríticas/imunologia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Imunoterapia/métodos , Lentivirus/genética , Neoplasias/terapia , Animais , Antígenos Glicosídicos Associados a Tumores/imunologia , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular Tumoral , Células Dendríticas/virologia , Feminino , Memória Imunológica , Interferon gama/imunologia , Linfonodos/imunologia , Linfonodos/virologia , Ativação Linfocitária , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Ovalbumina/genética , Ovalbumina/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Citotóxicos/imunologia , Transdução Genética/métodos
11.
Gene Ther ; 12(9): 772-82, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15750615

RESUMO

Until now, studies utilizing mRNA electroporation as a tool for the delivery of tumor antigens to human monocyte-derived dendritic cells (DC) have focused on DC electroporated in an immature state. Immature DC are considered to be specialized in antigen capture and processing, whereas mature DC present antigen and have an increased T-cell stimulatory capacity. Therefore, the consensus has been to electroporate DC before maturation. We show that the transfection efficiency of DC electroporated either before or after maturation was similarly high. Both immature and mature electroporated DC, matured in the presence of an inflammatory cytokine cocktail, expressed mature DC surface markers and preserved their capacity to secrete cytokines and chemokines upon CD40 ligation. In addition, both immature and mature DC can be efficiently cryopreserved before or after electroporation without deleterious effects on viability, phenotype or T-cell stimulatory capacity including in vitro antigen-specific T-cell activation. However, DC electroporated after maturation are more efficient in in vitro migration assays and at least as effective in antigen presentation as DC electroporated before maturation. These results are important for vaccination strategies where an optimal antigen presentation by DC after migration to the lymphoid organs is crucial.


Assuntos
Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Eletroporação , Apresentação de Antígeno , Diferenciação Celular/imunologia , Sobrevivência Celular , Criopreservação , Citocinas/imunologia , Humanos , Imunofenotipagem , Ativação Linfocitária/imunologia , Teste de Cultura Mista de Linfócitos , RNA Mensageiro/metabolismo , Linfócitos T/imunologia , Transfecção
12.
Cancer Res ; 60(22): 6272-5, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11103782

RESUMO

Antigens encoded by MAGE-A3 and recognized by T cells are interesting targets for tumor immunotherapy because they are strictly tumor specific and shared by many tumors of various histological types. A number of MAGE-A3 antigenic peptides presented by HLA class I molecules have been used in clinical trials, and regressions of melanoma metastasis have been observed. We report here the identification of a MAGE-A3 epitope, TQHFVQENYLEY, presented to CD4+ T lymphocytes by HLA-DP4 molecules, which are expressed in approximately 76% of Caucasians. This new epitope may be useful both for therapeutic vaccination and for the evaluation of the immune response in cancer patients. Interest ingly, the CD4+ T cells lysed HLA-DP4 tumor cells expressing MAGE-A3, indicating that this epitope, in contrast to other class-II MAGE-A3 epitopes, is presented at the surface of tumor cells. The study of this disparity in the presentation of two epitopes from the same protein may lead to a better understanding of the endogenous class II presentation pathway.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Antígenos HLA-DP/imunologia , Proteínas de Neoplasias , Linfócitos T Citotóxicos/imunologia , Animais , Baculoviridae/genética , Células Clonais , Células Dendríticas/imunologia , Escherichia coli/genética , Escherichia coli/metabolismo , Cadeias beta de HLA-DP , Humanos , Melanoma/imunologia , Spodoptera/metabolismo , Spodoptera/virologia , Células Tumorais Cultivadas
13.
Br J Haematol ; 107(3): 625-40, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10583269

RESUMO

Follicular lymphoma (FL) cells have inherited an activated hypermutation mechanism from their origin of germinal centre B cells. Based on today's knowledge of the intrinsic properties related to this mechanism and the VH base composition, reconsideration of previous reports should be made on a broader range of samples. The present study examined the mutation pattern of the VH genes expressed by 55 cases of FL. FL VH genes showed evidence of antigenic selection in 30% of cases with 88% carrying a functional sIg and 78.2% showing intraclonal variation. VH family and gene segment utilization was found to be roughly similar to that of normal B lymphocytes. FL VH genes revealed extensive variations. 17% of the VH exons harboured a total of five deletions, three duplications and two insertions as compared to the most homologous germline counterpart. The VH genes of one tumour displayed three populations with varying CDR3 length at diagnosis. At relapse, emergence of a differently mutated gene, additional mutations reminiscent of ongoing mutations or no variation was prominent. From this study the heterogeneity of FLs is well established and ongoing mutations are seen in the scope of the activated status of the hypermutation mechanism rather than antigen-stimulated tumour growth.


Assuntos
Genes de Imunoglobulinas/genética , Linfoma Folicular/genética , Mutação/genética , Substituição de Aminoácidos , Elementos de DNA Transponíveis/genética , Deleção de Genes , Humanos
14.
Br J Haematol ; 106(4): 1033-6, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10520008

RESUMO

Recently it was reported that Kaposi's sarcoma-associated herpesvirus (KSHV/HHV-8) infects bone marrow (BM) dendritic cells (DC) in multiple myeloma (MM) patients and therefore might play a role in MM development. Because of the use of myeloid growth factors like GM-CSF and G-CSF for the mobilization of peripheral blood progenitor cells (PBPC), the subsequent increase of DC precursors might imply a risk for KSHV contamination in PBPC grafts. Therefore, in this study leukapheresis products and ex vivo cultured CD34+ cell suspensions were analysed. KSHV DNA could not be amplified in any of them.


Assuntos
DNA Viral/análise , Herpesvirus Humano 8/genética , Mieloma Múltiplo/virologia , Antígenos CD34 , Genoma Viral , Mobilização de Células-Tronco Hematopoéticas , Humanos , Leucaférese , Reação em Cadeia da Polimerase/métodos , Sensibilidade e Especificidade
15.
J Leukoc Biol ; 66(2): 242-6, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10449161

RESUMO

Data from adoptive transfer of mature dendritic cells (DC) indicate that they are responsible for the induction of primary immunity. Two subclasses of DC have been recently identified in spleen that differ in their phenotype and in certain regulatory features. In vitro, both subsets have the capacity to activate naive T cells, although CD8a+ DC have been shown to induce T cell apoptosis and to stimulate lower levels of cytokines compared with CD8alpha- DC. The objective of this study was to analyze the function of these distinct DC types in vivo. Our results show that both subsets, pulsed extracorporeally with antigen and injected in the footpads of syngeneic mice, sensitize an antigen-specific T cell primary response. However, CD8alpha+ cells trigger the development of Thl-type cells, whereas CD8alpha- DC induce a Th2-type response. These observations suggest that the Th1/Th2 balance in vivo is regulated by the antigen-presenting-cells of the primary immune responses.


Assuntos
Antígenos CD8/imunologia , Células Dendríticas/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Diferenciação Celular , Humanos , Imunofenotipagem , Interleucina-12/imunologia , Ligantes , Proteínas de Membrana/imunologia , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Th1/citologia , Células Th1/imunologia , Células Th2/citologia , Células Th2/imunologia
16.
J Exp Med ; 189(3): 587-92, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9927520

RESUMO

Cells of the dendritic family display some unique properties that confer to them the capacity to sensitize naive T cells in vitro and in vivo. In the mouse, two subclasses of dendritic cells (DCs) have been described that differ by their CD8alpha expression and their localization in lymphoid organs. The physiologic function of both cell populations remains obscure. Studies conducted in vitro have suggested that CD8alpha+ DCs could play a role in the regulation of immune responses, whereas conventional CD8alpha- DCs would be more stimulatory. We report here that both subclasses of DCs efficiently prime antigen-specific T cells in vivo, and direct the development of distinct T helper (Th) populations. Antigen-pulsed CD8alpha+ and CD8alpha- DCs are separated after overnight culture in recombinant granulocyte/macrophage colony-stimulating factor and injected into the footpads of syngeneic mice. Administration of CD8alpha- DCs induces a Th2-type response, whereas injection of CD8alpha+ DCs leads to Th1 differentiation. We further show that interleukin 12 plays a critical role in Th1 development by CD8alpha+ DCs. These findings suggest that the nature of the DC that presents the antigen to naive T cells may dictate the class selection of the adaptative immune response.


Assuntos
Antígenos CD8/imunologia , Células Dendríticas/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Apresentação de Antígeno , Células Dendríticas/classificação , Tolerância Imunológica , Memória Imunológica , Interferon gama/biossíntese , Interleucinas/biossíntese , Células de Langerhans/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Fenótipo , Células Th1/imunologia , Células Th2/imunologia
17.
J Immunol ; 162(1): 144-51, 1999 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9886380

RESUMO

It has been extensively documented that murine dendritic cells loaded with tumor-associated Ag (TAA)-derived peptides or protein can prime Ag-specific CD8+ cytotoxic T cells in vivo and can elicit Ag-specific immunity. Optimal presentation of TAA might be achieved by retroviral transduction of DCs allowing long term and stable expression of the TAA-peptides as well as the presentation of multiple epitopes in the context of MHC class I and/or class II molecules. Here we show that retroviral transduction of bone marrow-derived dendritic cells (DCs) with chicken OVA cDNA or the reporter gene green fluorescent protein retained their potent stimulatory capacity and that the transduced DCs could process and present the endogenously expressed OVA protein. The DCs transduced with cDNA encoding native OVA protein presented OVA-derived peptides in the context of MHC class I as well as MHC class II and induced a strong Ag-specific CTL response. DCs expressing a cytosolic form of OVA presented OVA peptides only in the context of MHC class I and failed to induce an OVA-specific CTL response in vivo when they had been cultured in the absence of exogenous protein. Immunization with retrovirally transduced DCs resulted in an Ag-specific immunity and rejection of a tumor cell challenge and a significant survival advantage in tumor-bearing mice. These results obtained in this rapidly lethal tumor model suggest that DCs transduced with TAA may be useful for tumor immunotherapy and underscore the importance of the simultaneous delivery of T cell help in the development of Ag-specific cytotoxic T-cells.


Assuntos
Células da Medula Óssea/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Vetores Genéticos/imunologia , Imunoterapia Adotiva/métodos , Vírus da Leucemia Murina de Moloney/imunologia , Animais , Apresentação de Antígeno/genética , Células da Medula Óssea/virologia , Transplante de Medula Óssea , Separação Celular , Células Cultivadas , Células Dendríticas/transplante , Células Dendríticas/virologia , Epitopos de Linfócito T/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe II/genética , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vírus da Leucemia Murina de Moloney/genética , Ovalbumina/imunologia , Ovalbumina/metabolismo , Peptídeos/imunologia , Peptídeos/metabolismo , Linfócitos T Citotóxicos/imunologia
18.
J Immunol ; 161(3): 1454-61, 1998 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9686611

RESUMO

As demonstrated in several preclinical models, bispecific Abs are attractive immunotherapeutic agents for tumor treatment. We have previously reported that a bacterially produced anti-CD3 x antitumor bispecific single chain variable fragment of Ab fragment (BsscFv), which is capable of retargeting CTLs toward BCL1 tumor cells, exhibits antitumor activity in vitro. To further facilitate BsscFv production, the coding sequence was subcloned in a eukaryotic expression vector and introduced into Chinese hamster ovary cells for large-scale production. In this report, we have determined the serum stability and the clearance rate from the circulation of BsscFv. Most important, we prove here the therapeutic value of BsscFv in the treatment of BCL1 lymphoma, a murine model for human non-Hodgkin's lymphoma. Tumor-bearing mice that were treated with rscFv in combination with staphylococcal enterotoxin B superantigen, human rIL-2, or murine rIL-12 showed long-term survival, whereas untreated mice all died. This is the first report of the successful in vivo use of BsscFv as an immunotherapeutic agent. Furthermore, long-term survival was the result of complete tumor removal and was not due to the induction of dormancy.


Assuntos
Anticorpos Anti-Idiotípicos/uso terapêutico , Anticorpos Biespecíficos/uso terapêutico , Complexo CD3/imunologia , Fragmentos de Imunoglobulinas/genética , Linfoma de Células B/imunologia , Linfoma de Células B/terapia , Proteínas Recombinantes/imunologia , Animais , Anticorpos Anti-Idiotípicos/biossíntese , Anticorpos Anti-Idiotípicos/genética , Anticorpos Biespecíficos/biossíntese , Anticorpos Biespecíficos/genética , Modelos Animais de Doenças , Intervalo Livre de Doença , Enterotoxinas/administração & dosagem , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Fragmentos de Imunoglobulinas/biossíntese , Fragmentos de Imunoglobulinas/uso terapêutico , Região Variável de Imunoglobulina/biossíntese , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/uso terapêutico , Injeções Intravenosas , Ativação Linfocitária , Linfoma de Células B/mortalidade , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/uso terapêutico , Linfócitos T/imunologia , Células Tumorais Cultivadas
19.
Infect Immun ; 66(6): 2722-7, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9596739

RESUMO

We have previously shown that the addition of exogenous granulocyte-macrophage colony-stimulating factor (GM-CSF) to nonactivated mouse peritoneal macrophages (MPM) limits Trypanosoma cruzi infections in vitro (E. Olivares Fontt and B. Vray, Parasite Immunol. 17:135-141, 1995). Lower levels of infection were correlated with a higher level of production of tumor necrosis factor alpha (TNF-alpha) in the absence of nitric oxide (NO) release. These data suggested that GM-CSF and/or TNF-alpha might have a direct parasitocidal effect on T. cruzi trypomastigotes, independently of NO release. To address this question, T. cruzi trypomastigotes were treated with recombinant murine GM-CSF (rmGM-CSF), recombinant murine TNF-alpha (rmTNF-alpha), or both cytokines in a cell-free system. Treatment with rmGM-CSF but not rmTNF-alpha caused morphological changes in the parasites, and most became spherical after 7 h of incubation. Both cytokines exerted a cytolytic activity on the trypomastigotes, yet the trypanolytic activity of rmTNF-alpha was more effective than that of rmGM-CSF. Viable rmGM-CSF- and rmTNF-alpha-treated parasites were less able to infect MPM than untreated parasites, and this reduction in infectivity was greatest for rmGM-CSF. Treatments with both cytokines resulted in more lysis and almost complete inhibition of infection. The direct parasitocidal activity of rmTNF-alpha was inhibited by carbohydrates and monoclonal antibodies specific for the lectin-like domain of TNF-alpha. Collectively, these results suggest that cytokines such as GM-CSF and TNF-alpha may directly control the level of T. cruzi trypomastigotes at least in vitro and so could determine the outcome of infection in vivo.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Tripanossomicidas/farmacologia , Trypanosoma cruzi/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Relação Dose-Resposta a Droga , Interações Medicamentosas , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Camundongos , Proteínas Recombinantes/farmacologia , Trypanosoma cruzi/citologia , Trypanosoma cruzi/patogenicidade , Fator de Necrose Tumoral alfa/genética , Virulência/efeitos dos fármacos
20.
Clin Cancer Res ; 3(12 Pt 1): 2501-6, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9815653

RESUMO

Multiple myeloma (MM) is a B-cell malignancy characterized by the expansion of malignant plasma cells within the bone marrow. Previous studies that have examined the Ig VH genes of IgG and IgA MMs have shown the presence of somatic mutations, suggesting that in these cases, the myeloma precursor cell passed through the phase of antigenic selection within the germinal center but is no longer exposed to the somatic mutation process. However, no information about this matter is available in the rare IgD and IgM MM variants. Therefore, we have analyzed the Ig VH genes of three IgD, one IgM, and one biclonal (IgG and IgM) MM for the presence of somatic mutations. Our study demonstrates that all of these myeloma clones have accumulated a high number of somatic mutations within their Ig VH genes but show no intraclonal variation. Moreover, proof that the clone sustained a strong antigenic selection pressure could be provided in three cases (one IgD and two IgMs). Therefore, this study strongly implies that IgD and IgM MMs emerge from a postgerminal center preswitched B cell that is no longer exposed to the somatic mutation process or able to undergo further isotype switching in vivo.


Assuntos
Genes de Imunoglobulinas , Imunoglobulina D/genética , Cadeias Pesadas de Imunoglobulinas/genética , Imunoglobulina M/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/imunologia , Mutação Puntual , Sequência de Aminoácidos , Clonagem Molecular , Amplificação de Genes , Humanos , Região Variável de Imunoglobulina/genética , Dados de Sequência Molecular , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...