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.
Auris Nasus Larynx ; 46(1): 50-57, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29935892

RESUMO

OBJECTIVE: Current spread by electrical stimulation via inserted cochlear implant (CI) electrodes and the consequential increase in sound input can affect the equilibrium of patients. The aim of the present study was to clarify the effect of CIs on the equilibrium of patients through cervical vestibular-evoked myogenic potential (cVEMP) testing and static stabilometry performed with the CIs turned on (CI-on) and off (CI-off). METHODS: This prospective study included nine adult patients who underwent unilateral cochlear implantation surgery at our institution. cVEMP testing and stabilometry were performed before surgery and repeated after surgery in the CI-on and CI-off modes. RESULTS: Before surgery, cVEMP responses were diminished in five of the nine patients (55.6%), while the results of stabilometry were poor in six patients (66.7%). After surgery, both cVEMP responses and stabilometry findings in the CI-off mode exhibited significant deterioration relative to the preoperative results (cVEMP: 7/9, 77.8%; stabilometry: 7/9, 77.8%). However, in the CI-on mode, there were significant improvements in both test results relative to the findings in the CI-off mode for all patients. CONCLUSION: CIs compensated for the surgical trauma-induced deterioration in static postural stability when turned on, resulting in a considerable improvement. Our findings suggest that postoperative cVEMP testing in the CI-on and CI-off modes will enable more accurate assessment of the saccule-inferior vestibular nerve system function after cochlear implant surgery.


Assuntos
Implante Coclear , Implantes Cocleares , Complicações Pós-Operatórias/fisiopatologia , Equilíbrio Postural/fisiologia , Doenças Vestibulares/fisiopatologia , Potenciais Evocados Miogênicos Vestibulares/fisiologia , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Nervo Vestibular/fisiopatologia
2.
Semin Immunopathol ; 33(6): 603-12, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21461992

RESUMO

In vivo transfer of dendritic cells (DC) has proven efficient in the priming of T cells and is regarded as a powerful means of providing anti-cancer immunotherapy. Clinical trials of anti-cancer therapy with DC pulsed with peptide antigens have been carried out in many institutions, although dramatic therapeutic effect has not been observed in most of the trials. Negative regulation of the immune response by DC might be applicable to treatment of autoimmune diseases and transplantation medicine. Currently, the DC used for anti-cancer vaccine therapy are generated from the peripheral blood monocytes of the patients. However, there is a limitation in the number of available monocytes and the potential of monocytes to differentiate into DC varies depending on the individual blood donors. To resolve the issue of the cell source for DC therapy, several groups have developed methods to generate DC from pluripotent stem cells. This review introduces methods to generate functional DC from pluripotent stem cells of mouse and human.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/transplante , Células-Tronco Pluripotentes/citologia , Animais , Autoimunidade , Diferenciação Celular , Células Dendríticas/metabolismo , Células-Tronco Embrionárias/metabolismo , Humanos , Imunomodulação , Terapia de Imunossupressão , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Linfócitos T Citotóxicos/imunologia
3.
Front Biosci (Elite Ed) ; 2(4): 1520-7, 2010 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-20515823

RESUMO

Dendritic cell (DC) is regarded as a powerful means for anti-cancer immunotherapy. Clinical trials of cancer therapy with DC loaded with cancer antigens, such as tumor cell-lysates or HLA class I-binding antigenic peptides, have been conducted. Antigen-specific negative manipulation of the immune response by DC is a potential treatment for autoimmune diseases and also for control of allo-reactive immune responses in transplantation medicine. Currently, DC for clinical use are generated from peripheral blood monocytes of the patients. However, the number of monocytes obtained from the patients is limited and the potential of monocytes to differentiate into DC varies depending on the blood donor. Thus, the issue of limited cells is a serious obstacle for DC therapy. ES cells and iPS cells have pluripotency and unlimited propagation capacity and may be an ideal cell source for DC-therapy. Several groups, including us, have developed methods to generate DC from ES cells or iPS cells. This review introduces the studies on generation, characterization, and genetic modification of DC derived from ES cells or iPS cells.


Assuntos
Células Dendríticas/citologia , Imunoterapia , Células-Tronco Pluripotentes/citologia , Animais , Transplante de Células , Encefalomielite Autoimune Experimental/terapia , Engenharia Genética , Humanos , Camundongos , Neoplasias/terapia
4.
Int J Hematol ; 91(3): 392-400, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20155337

RESUMO

Dendritic cells (DC) are the most potent antigen-presenting cells. In vivo transfer of antigen-bearing DC has proven efficient in priming T cell responses specific to the antigen. DC-based cellular vaccination is now regarded as a powerful means for immunotherapy, especially for anti-cancer immunotherapy. Clinical trials of therapy with DC pulsed with peptide antigens or genetically modified to present antigens are currently carried out in many institutions. In addition, antigen-specific negative regulation of immune response by DC is considered to be a promising approach for treatments of autoimmune diseases and also for regulation of allo-reactive immune response causing graft rejection and GVHD in transplantation medicine. DC for transfer therapy are now generated by in vitro differentiation of peripheral blood monocytes of the patients. However, there is a limitation in the number of available monocytes, and the DC-differentiation potential of monocytes varies depending on the blood donor. Embryonic stem (ES) cells possess both pluripotency and infinite propagation capacity. We consider ES cells to be an ideal source for DC to be used in immunotherapy. Several groups, including us, have developed methods to generate DC from ES cells. This review introduces the studies on generation, characterization, and genetic modification of DC derived from ES cells or induced pluripotent stem (iPS) cells. The issues to be resolved before clinical application of pluripotent stem cell-derived DC will also be discussed.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/imunologia , Imunoterapia/métodos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/imunologia , Animais , Linhagem da Célula/imunologia , Humanos
5.
BMJ Case Rep ; 20092009.
Artigo em Inglês | MEDLINE | ID: mdl-21686450

RESUMO

Primary squamous cell cancer of the thyroid gland (SCT) is a rare malignant tumour and is associated with a high mortality. A female patient who suffered from primary SCT is described in this report. The cancer was identified with acute painful swelling of the thyroid gland, when the patient was under periodical observation for her chronic thyroiditis at our outpatient's clinic. In spite of the highly malignant potential of the cancer as indicated by histological examinations, including p53 and MIB-1 index analyses, the patient has been successfully treated so far with surgery and radiation therapy, surviving for more than 34 months with no sign of recurrence or metastasis. Early diagnosis and prompt treatment might have been essential for the successful management of this patient. Further observations and investigations are necessary to clarify the mechanisms of the long survival and to find a better treatment for the disease.

6.
J Immunol ; 178(2): 918-25, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17202353

RESUMO

We previously reported the protection from myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) by the adoptive transfer of genetically modified embryonic stem cell-derived dendritic cells (ES-DC) presenting MOG peptide in the context of MHC class II molecules and simultaneously expressing TRAIL (ES-DC-TRAIL/MOG). In the present study, we found the severity of EAE induced by another myelin autoantigen, myelin basic protein, was also decreased after treatment with ES-DC-TRAIL/MOG. This preventive effect diminished, if the function of CD4(+)CD25(+) regulatory T cells (Treg) was abrogated by the injection of anti-CD25 mAb into mice before treatment with ES-DC-TRAIL/MOG. The adoptive transfer of CD4(+)CD25(+) T cells from ES-DC-TRAIL/MOG-treated mice protected the recipient mice from MOG- or myelin basic protein-induced EAE. The number of Foxp3(+) cells increased in the spinal cords of mice treated with ES-DC-TRAIL/MOG. In vitro experiments showed that TRAIL expressed in genetically modified ES-DC and also in LPS-stimulated splenic macrophages had a capacity to augment the proliferation of CD4(+)CD25(+) T cells. These results suggest that the prevention of EAE by treatment with ES-DC-TRAIL/MOG is mediated, at least in part, by MOG-reactive CD4(+)CD25(+) Treg propagated by ES-DC-TRAIL/MOG. For the treatment of organ-specific autoimmune diseases, induction of Treg reactive to the organ-specific autoantigens by the transfer of DC-presenting Ags and simultaneously overexpressing TRAIL therefore appears to be a promising strategy.


Assuntos
Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Glicoproteína Associada a Mielina/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transferência Adotiva , Animais , Anticorpos Monoclonais/imunologia , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Fatores de Transcrição Forkhead/metabolismo , Engenharia Genética , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Proteína Básica da Mielina/farmacologia , Proteínas da Mielina , Glicoproteína Associada a Mielina/genética , Glicoproteína Associada a Mielina/uso terapêutico , Glicoproteína Mielina-Oligodendrócito , Medula Espinal/patologia , Linfócitos T Reguladores/citologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico
7.
Cancer Res ; 66(4): 2414-22, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16489048

RESUMO

We have recently established a method to generate dendritic cells from mouse embryonic stem cells. By introducing exogenous genes into embryonic stem cells and subsequently inducing differentiation to dendritic cells (ES-DC), we can now readily generate transfectant ES-DC expressing the transgenes. A previous study revealed that the transfer of genetically modified ES-DC expressing a model antigen, ovalbumin, protected the recipient mice from a challenge with an ovalbumin-expressing tumor. In the present study, we examined the capacity of ES-DC expressing mouse homologue of human glypican-3, a recently identified oncofetal antigen expressed in human melanoma and hepatocellular carcinoma, to elicit protective immunity against glypican-3-expressing mouse tumors. CTLs specific to multiple glypican-3 epitopes were primed by the in vivo transfer of glypican-3-transfectant ES-DC (ES-DC-GPC3). The transfer of ES-DC-GPC3 protected the recipient mice from subsequent challenge with B16-F10 melanoma, naturally expressing glypican-3, and with glypican-3-transfectant MCA205 sarcoma. The treatment with ES-DC-GPC3 was also highly effective against i.v. injected B16-F10. No harmful side effects, such as autoimmunity, were observed for these treatments. The depletion experiments and immunohistochemical analyses suggest that both CD8+ and CD4+ T cells contributed to the observed antitumor effect. In conclusion, the usefulness of glypican-3 as a target antigen for antimelanoma immunotherapy was thus shown in the mouse model using the ES-DC system. Human dendritic cells expressing glypican-3 would be a promising means for therapy of melanoma and hepatocellular carcinoma.


Assuntos
Células Dendríticas/imunologia , Proteoglicanas de Heparan Sulfato/imunologia , Imunoterapia Adotiva/métodos , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Células-Tronco/imunologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Células Dendríticas/citologia , Epitopos de Linfócito T/imunologia , Feminino , Glipicanas , Proteoglicanas de Heparan Sulfato/biossíntese , Proteoglicanas de Heparan Sulfato/genética , Células Matadoras Naturais/imunologia , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Células-Tronco/citologia , Linfócitos T Citotóxicos/imunologia
8.
Cancer Sci ; 96(12): 889-96, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16367909

RESUMO

The close cooperation of both innate and acquired immunity is essential for the induction of truly effective antitumor immunity. We tested a strategy to enhance the cross-talk between NKT cells and conventional antigen-specific T cells with the use of alpha GalCer-loaded dendritic cells genetically engineered to express antigen plus chemokine, attracting both conventional T cells and NKT cells. DC genetically engineered to express a model antigen, OVA, along with SLC/CCL21 or monokine induced by IFN-gamma/CXCL9, had been generated using a method based on in vitro differentiation of DC from mouse ES cells. The ES-DC were loaded with alpha-GalCer and transferred to mice bearing MO4, an OVA-expressing melanoma, and their capacity to evoke antitumor immunity was evaluated. In vivo transfer of either OVA-expressing ES-DC, stimulating OVA-reactive T cells, or alpha-GalCer-loaded non-transfectant ES-DC, stimulating NKT cells, elicited a significant but limited degree of protection against the i.p. disseminated MO4. A more potent antitumor effect was observed when alpha-GalCer was loaded to ES-DC expressing OVA before in vivo transfer, and the effect was abrogated by the administration of anti-CD8, anti-NK1.1 or anti-asialo GM1 antibody. alpha-GalCer-loaded double transfectant ES-DC expressing SLC along with OVA induced the most potent antitumor immunity. Thus, alpha-GalCer-loaded ES-DC expressing tumor-associated antigen along with SLC can stimulate multiple subsets of effector cells to induce a potent therapeutic effect against peritoneally disseminated tumor cells. The present study suggests a novel way to use alpha-GalCer in immunotherapy for peritoneally


Assuntos
Antineoplásicos/uso terapêutico , Quimiocinas CC/genética , Células Dendríticas/imunologia , Galactosilceramidas/uso terapêutico , Neoplasias/imunologia , Animais , Antígenos de Neoplasias/imunologia , Comunicação Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Quimiocina CCL21 , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/fisiologia , Engenharia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Neoplasias/patologia , Ovalbumina/imunologia , Proteínas Recombinantes , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
9.
Biochem Biophys Res Commun ; 335(1): 5-13, 2005 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-16026756

RESUMO

Dendritic cells (DC) genetically modified to present tumor-associated antigen are a promising means for anti-cancer immunotherapy. By introducing expression vectors into ES cells and subsequently inducing differentiation to DC (ES-DC), we can generate transfectant DC expressing the transgenes. In the future clinical application of this technology, the unavailability of human ES cells genetically identical to the patients will be a problem. However, in most cases, semi-allogeneic ES cells sharing some of HLA alleles with recipients are expected to be available. In the present study, we observed that model tumor antigen (OVA)-expressing mouse ES-DC transferred into semi-allogeneic mice potently primed OVA-reactive CTL and elicited a significant protection against challenge with OVA-expressing tumor. Genetic modification of ES-DC to overexpress SPI-6, the specific inhibitor of granzyme B, further enhanced their capacity to prime antigen-specific CTL in semi-allogeneic recipient mice. These results suggest the potential of ES-DC as a novel means for anti-cancer immunotherapy.


Assuntos
Diferenciação Celular , Células Dendríticas/citologia , Células Dendríticas/imunologia , Neoplasias/prevenção & controle , Células-Tronco/citologia , Animais , Antígenos/genética , Antígenos/imunologia , Linhagem Celular , Células Dendríticas/transplante , Imunoterapia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Ovalbumina/genética , Ovalbumina/imunologia , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Serpinas/genética , Serpinas/metabolismo , Transplante de Células-Tronco , Taxa de Sobrevida , Linfócitos T Citotóxicos/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Immunol ; 174(4): 1888-97, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15699115

RESUMO

Experimental autoimmune encephalomyelitis (EAE) is caused by activation of myelin Ag-reactive CD4+ T cells. In the current study, we tested a strategy to prevent EAE by pretreatment of mice with genetically modified dendritic cells (DC) presenting myelin oligodendrocyte glycoprotein (MOG) peptide in the context of MHC class II molecules and simultaneously expressing TRAIL or Programmed Death-1 ligand (PD-L1). For genetic modification of DC, we used a recently established method to generate DC from mouse embryonic stem cells (ES cells) in vitro (ES-DC). ES cells were sequentially transfected with an expression vector for TRAIL or PD-L1 and an MHC class II-associated invariant chain-based MOG epitope-presenting vector. Subsequently, double-transfectant ES cell clones were induced to differentiate to ES-DC, which expressed the products of introduced genes. Treatment of mice with either of the double-transfectant ES-DC significantly reduced T cell response to MOG, cell infiltration into spinal cord, and the severity of MOG peptide-induced EAE. In contrast, treatment with ES-DC expressing MOG alone, irrelevant Ag (OVA) plus TRAIL, or OVA plus PD-L1, or coinjection with ES-DC expressing MOG plus ES-DC-expressing TRAIL or PD-L1 had no effect in reducing the disease severity. In contrast, immune response to irrelevant exogenous Ag (keyhole limpet hemocyanin) was not impaired by treatment with any of the genetically modified ES-DC. The double-transfectant ES-DC presenting Ag and simultaneously expressing immune-suppressive molecules may well prove to be an effective therapy for autoimmune diseases without inhibition of the immune response to irrelevant Ag.


Assuntos
Antígeno B7-1/administração & dosagem , Células Dendríticas/transplante , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Glicoproteínas/administração & dosagem , Glicoproteínas de Membrana/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Peptídeos/administração & dosagem , Transplante de Células-Tronco/métodos , Fator de Necrose Tumoral alfa/administração & dosagem , Transferência Adotiva/métodos , Sequência de Aminoácidos , Animais , Apoptose/genética , Apoptose/imunologia , Proteínas Reguladoras de Apoptose , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Antígeno B7-H1 , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linhagem Celular , Cruzamentos Genéticos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Glicoproteínas/biossíntese , Glicoproteínas/genética , Glicoproteínas/imunologia , Contagem de Linfócitos , Masculino , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Dados de Sequência Molecular , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Peptídeos/genética , Engenharia de Proteínas , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/transplante , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Transgenes/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
11.
Clin Cancer Res ; 10(19): 6437-48, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15475430

RESUMO

PURPOSE: To establish effective antitumor immunotherapy for esophageal cancer, we tried to identify an useful target antigen of esophageal cancer. EXPERIMENTAL DESIGN: We did cDNA microarray analysis to find a novel candidate antigen, proliferation potential-related protein (PP-RP). We examined cytotoxicity against tumor cells in vitro and in vivo of CTLs specific to PP-RP established from esophageal cancer patients. RESULTS: In 26 esophageal cancer tissues, an average of relative ratio of the expression of the PP-RP mRNA in cancer cells versus adjacent normal esophageal tissues was 396.2. Immunohistochemical analysis revealed that, in 20 of the 22 esophageal cancer tissues, PP-RP protein was strongly expressed only in the cancer cells and not so in normal esophageal epithelial cells. PP-RP protein contains 10 epitopes recognized by HLA-A24-restricted CTLs. These CTLs, generated from HLA-A24-positive esophageal cancer patients, had cytotoxic activity against cancer cell lines positive for both PP-RP and HLA-A24. Furthermore, adoptive transfer of the PP-RP-specific CTL line inhibited the growth of a human esophageal cancer cell line engrafted in nude mice. CONCLUSIONS: The expression of PP-RP in esophageal cancer cells was significantly higher than in normal cells, and the CTLs recognizing PP-RP killed tumor cells in vitro and also showed tumor rejection effects in a xenograft model. Therefore, PP-RP may prove to be an ideal tumor antigen useful for diagnosis and immunotherapy for patients with esophageal cancer. cDNA microarray analysis is a useful method to identify ideal tumor-associated antigens.


Assuntos
Proteínas de Transporte/genética , Proteínas de Ligação a DNA/genética , Neoplasias Esofágicas/terapia , Imunoterapia/métodos , Adulto , Idoso , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/farmacologia , Proteínas de Transporte/imunologia , Proteínas de Transporte/farmacologia , Linhagem Celular Tumoral , Testes Imunológicos de Citotoxicidade/métodos , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/farmacologia , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Antígenos HLA-A/imunologia , Antígeno HLA-A24 , Humanos , Imunoterapia Adotiva/métodos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Neoplasia Residual/genética , Neoplasia Residual/patologia , Placenta/metabolismo , Prognóstico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sobrevida , Linfócitos T Citotóxicos/imunologia , Testículo/metabolismo , Ubiquitina-Proteína Ligases , Ensaios Antitumorais Modelo de Xenoenxerto
12.
J Immunol ; 172(2): 776-86, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14707047

RESUMO

Dendritic cell (DC)-based immunotherapy is regarded as a promising means for anti-cancer therapy. The efficiency of T cell-priming in vivo by transferred DCs should depend on their encounter with T cells. In the present study, we attempted to improve the capacity of DCs to prime T cells in vivo by genetic modification to express chemokine with a T cell-attracting property. For genetic modification of DCs, we used a recently established method to generate DCs from mouse embryonic stem cells. We generated double-transfectant DCs expressing a chemokine along with a model Ag (OVA) by sequential transfection of embryonic stem cells, and then induced differentiation to DCs. We comparatively evaluated the effect of three kinds of chemokines; secondary lymphoid tissue chemokine (SLC), monokine induced by IFN-gamma (Mig), and lymphotactin (Lptn). All three types of double transfectant DCs primed OVA-specific CTLs in vivo more efficiently than did DCs expressing only OVA, and the coexpression of SLC or Lptn was more effective than that of Mig. Immunization with DCs expressing OVA plus SLC or Mig provided protection from OVA-expressing tumor cells more potently than did immunization with OVA alone, and SLC was more effective than Mig. In contrast, coexpression of Lptn gave no additive effect on protection from the tumor. Collectively, among the three chemokines, expression of SLC was the most effective in enhancing antitumor immunity by transferred DCs in vivo. The findings provide useful information for the development of a potent DC-based cellular immunotherapy.


Assuntos
Quimiocinas/biossíntese , Células Dendríticas/imunologia , Células Dendríticas/transplante , Proteínas do Ovo/biossíntese , Epitopos de Linfócito T/administração & dosagem , Melanoma Experimental/prevenção & controle , Ovalbumina/biossíntese , Transplante de Células-Tronco , Linfócitos T Citotóxicos/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem Celular , Movimento Celular/genética , Movimento Celular/imunologia , Sobrevivência Celular/imunologia , Células Cultivadas , Quimiocina CCL21 , Quimiocinas/genética , Quimiocinas CC/administração & dosagem , Quimiocinas CC/biossíntese , Quimiocinas CC/fisiologia , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Proteínas do Ovo/administração & dosagem , Proteínas do Ovo/imunologia , Embrião de Mamíferos , Epitopos de Linfócito T/biossíntese , Epitopos de Linfócito T/imunologia , Feminino , Perfilação da Expressão Gênica , Hibridomas , Imunoterapia Ativa/métodos , Injeções Intraperitoneais , Injeções Intravenosas , Masculino , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Transplante de Neoplasias , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Fragmentos de Peptídeos , Transplante de Células-Tronco/métodos , Transfecção
13.
Blood ; 101(9): 3501-8, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12406878

RESUMO

We developed a method to generate dendritic cells (DCs) from mouse embryonic stem (ES) cells. We cultured ES cells for 10 days on feeder cell layers of OP9, in the presence of granulocyte-macrophage colony-stimulating factor in the latter 5 days. The resultant ES cell-derived cells were transferred to bacteriologic Petri dishes without feeder cells and further cultured. In about 7 days, irregularly shaped floating cells with protrusions appeared and these expressed major histocompatibility complex class II, CD11c, CD80, and CD86, with the capacity to stimulate primary mixed lymphocyte reaction (MLR) and to process and present protein antigen to T cells. We designated them ES-DCs (ES cell-derived dendritic cells), and the functions of ES-DCs were comparable with those of DCs generated from bone marrow cells. Upon transfer to new dishes and stimulation with interleukin-4 plus tumor necrosis factor alpha, combined with anti-CD40 monoclonal antibody or lipopolysaccharide, ES-DCs completely became mature DCs, characterized by a typical morphology and higher capacity to stimulate MLR. Using an expression vector containing the internal ribosomal entry site-puromycin N-acetyltransferase gene or a Cre-lox-mediated exchangeable gene-trap system, we could efficiently generate ES cell transfectants expressing the products of introduced genes after their differentiation to DCs. ES-DCs expressing invariant chain fused to a pigeon cytochrome C epitope presented the epitope efficiently in the context of E(k). We primed ovalbumin (OVA)-specific cytotoxic T lymphocytes in vivo by injecting mice with ES-DCs expressing OVA, thus demonstrating immunization with ES-DCs genetically engineered to express antigenic protein. The methods may be applicable to immunomodulation therapy and gene-trap investigations of DCs.


Assuntos
Células Dendríticas/citologia , Células-Tronco Totipotentes/citologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Apresentação de Antígeno , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Antígenos CD40/imunologia , Diferenciação Celular , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Técnicas de Cocultura , Cruzamentos Genéticos , Grupo dos Citocromos c/imunologia , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Células Dendríticas/transplante , Embrião de Mamíferos/citologia , Epitopos/imunologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Antígenos de Histocompatibilidade Classe II/imunologia , Imunofenotipagem , Interleucina-4/farmacologia , Lipopolissacarídeos/farmacologia , Teste de Cultura Mista de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Ovalbumina , Fragmentos de Peptídeos , Linfócitos T Citotóxicos/imunologia , Células-Tronco Totipotentes/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA