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











Base de dados
Intervalo de ano de publicação
1.
Clin Exp Immunol ; 202(1): 119-135, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32562271

RESUMO

Heparin is a widely used anti-coagulant that enhances anti-thrombin (AT) activity. However, heparin also suppresses immune and inflammatory responses in various rodent models and clinical trials, respectively. The mechanism by which heparin suppresses immune responses is unclear. The effect of heparin on regulatory T cells (Tregs ) in allogeneic immune responses was analysed using an acute graft-versus-host disease (aGVHD) mouse model and mixed lymphocyte reactions (MLRs). In-vitro culture systems were utilized to study the effects of heparin on Tregs . Heparin administration reduced mortality rates and increased the proportion of Tregs in the early post-transplantation period of aGVHD mice. In both murine and human MLRs, heparin increased Tregs and inhibited responder T cell proliferation. Heparin promoted functional CD4+ CD25+ forkhead box protein 3 (FoxP3)+ Treg generation from naive CD4+ T cells, increased interleukin (IL)-2 production and enhanced the activation of pre-existing Tregs with IL-2. Heparin-induced Treg increases were not associated with anti-coagulant activity through AT, but required negatively charged sulphation of heparin. Importantly, N-acetyl heparin, a chemically modified heparin without anti-coagulant activity, induced Tregs and decreased mortality in aGVHD mice. Our results indicate that heparin contributes to Treg -mediated immunosuppression through IL-2 production and suggest that heparin derivatives may be useful for immunopathological control by efficient Treg induction.


Assuntos
Anticoagulantes/farmacologia , Coagulação Sanguínea , Fibrinolíticos/farmacologia , Doença Enxerto-Hospedeiro/imunologia , Linfócitos T Reguladores/imunologia , Animais , Anticoagulantes/efeitos adversos , Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/imunologia , Modelos Animais de Doenças , Fibrinolíticos/efeitos adversos , Heparina , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Reguladores/patologia
2.
Haemophilia ; 20(1): e40-4, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24354485

RESUMO

The use of induced pluripotent stem cells (iPSCs) as an autologous cell source has shed new light on cell replacement therapy with respect to the treatment of numerous hereditary disorders. We focused on the use of iPSCs for cell-based therapy of haemophilia. We generated iPSCs from mesenchymal stem cells that had been isolated from C57BL/6 mice. The mouse iPSCs were generated through the induction of four transcription factor genes Oct3/4, Klf-4, Sox-2 and c-Myc. The derived iPSCs released functional coagulation factor VIII (FVIII) following transduction with a simian immunodeficiency virus vector. The subcutaneous transplantation of iPSCs expressing FVIII into nude mice resulted in teratoma formation, and significantly increased plasma levels of FVIII. The plasma concentration of FVIII was at levels appropriate for human therapy at 2-4 weeks post transplantation. Our data suggest that iPSCs could be an attractive and prospective autologous cell source for the production of coagulation factor, and that engineered iPSCs expressing coagulation factor might provide a cell-based therapeutic strategy appropriate for haemophilia.


Assuntos
Fator VIII/biossíntese , Fator VIII/genética , Vetores Genéticos/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Vírus da Imunodeficiência Símia/genética , Animais , Diferenciação Celular , Células Cultivadas , Expressão Gênica , Ordem dos Genes , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/citologia , Camundongos , Fatores de Tempo , Transdução Genética
3.
J Thromb Haemost ; 10(9): 1802-13, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22784361

RESUMO

BACKGROUND: Transplantation of cells overexpressing a target protein represents a viable gene therapeutic approach for treating hemophilia. Here, we focused on the use of autologous mesenchymal stem cells (MSCs) expressing coagulation factor for the treatment of coagulation factor VIII (FVIII) deficiency in mice. METHODS AND RESULTS: Analysis of luciferase gene constructs driven by different promoters revealed that the plasminogen activator inhibitor-1 (PAI-1) gene promoter coupled with the cytomegalovirus promoter enhancer region was one of the most effective promoters for producing the target protein. MSCs transduced with the simian immunodeficiency virus (SIV) vector containing the FVIII gene driven by the PAI-1 promoter expressed FVIII for several months, and this expression was maintained after multiple mesenchymal lineage differentiation. Although intravenous injection of cell supernatant derived from MSCs transduced with an SIV vector containing the FVIII gene driven by the PAI-1 promoter significantly increased plasma FVIII levels, subcutaneous implantation of the MSCs resulted in a transient and weak increase in plasma FVIII levels in FVIII-deficient mice. Interestingly, intra-articular injection of the transduced MSCs significantly ameliorated the hemarthrosis and hemophilic arthropathy induced by knee joint needle puncture in FVIII-deficient mice. The therapeutic effects of a single intra-articular injection of transduced MSCs to inhibit joint bleeding persisted for at least 8 weeks after administration. CONCLUSIONS: MSCs provide a promising autologous cell source for the production of coagulation factor. Intra-articular injection of MSCs expressing coagulation factor may offer an attractive treatment approach for hemophilic arthropathy.


Assuntos
Fatores de Coagulação Sanguínea/metabolismo , Transplante de Células , Fator VIII/genética , Hemofilia A/terapia , Artropatias/terapia , Células-Tronco Mesenquimais/citologia , Animais , Hemofilia A/complicações , Injeções Intra-Articulares , Artropatias/complicações , Células-Tronco Mesenquimais/metabolismo , Camundongos , Inibidor 1 de Ativador de Plasminogênio/genética , Regiões Promotoras Genéticas
4.
Haemophilia ; 18(3): e323-30, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22044430

RESUMO

Haemophilia A is a life long bleeding disorder caused by an inherited deficiency of factor VIII (FVIII). About 30% of haemophilia A patients develop neutralizing antibodies as a consequence of treatment with FVIII concentrates. Immune tolerance protocols for the eradication of inhibitors require daily delivery of intravenous FVIII. We evaluated the immune responses to serial intravenous administration of FVIII in preimmunized haemophilia A mice. We introduced an implantable venous-access device (iVAD) system into haemophilia A mice to facilitate sequential infusion of FVIII. After preimmunization with FVIII, the haemophilia A mice were subjected to serial intravenous administration of FVIII through the iVAD system. In all mice with serial infusion of FVIII, high titers of anti-FVIII inhibitory antibodies developed at 10 exposure days (EDs). However, the anti-FVIII IgG titers were decreased after 150 EDs of sequential low-dose infusion of FVIII [0.05 U g(-1) body weight (BW) five times per week]. Proliferative response to ex vivo FVIII stimulation was significantly suppressed in splenic CD4(+) T cells from mice with serial low-dose FVIII infusion compared with those from mice with high-dose FVIII infusion (0.5 U g(-1) BW five times per week) or preimmunized mice. Moreover, splenic CD4(+) T cells from mice with serial low-dose infusion of FVIII failed to produce interleukin-2 and interferon-γ. These data suggest that serial infusion of FVIII could induce T-cell anergy in haemophilia A mice with inhibitor antibodies.


Assuntos
Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Coagulantes/imunologia , Fator VIII/imunologia , Hemofilia A/imunologia , Tolerância Imunológica/efeitos dos fármacos , Animais , Inibidores dos Fatores de Coagulação Sanguínea/sangue , Cateterismo Venoso Central , Cateteres de Demora , Proliferação de Células/efeitos dos fármacos , Coagulantes/administração & dosagem , Citocinas/metabolismo , Modelos Animais de Doenças , Fator VIII/administração & dosagem , Hemofilia A/tratamento farmacológico , Hemofilia A/metabolismo , Imunoglobulina G/sangue , Infusões Intravenosas , Isoanticorpos/sangue , Camundongos
5.
Cancer Gene Ther ; 17(7): 484-91, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20168351

RESUMO

OBP-301 (a telomerase-specific, replication-competent adenovirus with hTERT promoter) was constructed in a previous study and it showed a strong anticancer effect by inducing cell lysis in human lung and prostate cancer cells. This study investigated the effectiveness of a combination therapy of OBP-301 and interleukin-2 (IL-2) in a mouse model of renal cell carcinoma (RCC). The cell-killing effect of OBP-301 was confirmed in vitro in the RENCA cancer cells. In in vivo experiment, luciferase-expressing RENCA cells were implanted in the left kidney and lung of BALB/c mice to prepare the RCC metastatic model. The animals were randomly divided into four treatment groups: PBS, IL-2 alone, OBP-301 alone and the combination. The analyses of orthotopic tumor weight, lung metastasis and luciferin-stained tumor images 14 days after each treatment showed significant tumor growth inhibition in the combination group in comparison with that in the OBP-301- or IL-2-treated groups. In addition, the percentage of regulatory T-cells (Tregs) in the combination group was significantly suppressed in comparison with that in the PBS and single-agent treatment groups. The outcomes of this study suggest that tumor-specific oncolytic immunovirotherapy may become an attractive strategy for the treatment of human RCC.


Assuntos
Carcinoma de Células Renais/terapia , Terapia Genética/métodos , Interleucina-2/administração & dosagem , Neoplasias Renais/terapia , Terapia Viral Oncolítica/métodos , Telomerase/metabolismo , Adenoviridae/enzimologia , Adenoviridae/genética , Adenoviridae/fisiologia , Animais , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/patologia , Carcinoma de Células Renais/virologia , Linhagem Celular Tumoral , Terapia Combinada , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/patologia , Neoplasias Renais/virologia , Camundongos , Camundongos Endogâmicos BALB C , Replicação Viral
6.
J Thromb Haemost ; 7(5): 811-24, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19220731

RESUMO

SUMMARY BACKGROUND: Hemophilia A is a congenital bleeding disorder caused by a deficiency of coagulation factor VIII. Approximately 30% of hemophilia A patients develop inhibitors against FVIII following replacement therapy. We have reported that neonatal exposure of FVIII antigen can induce antigen-specific immune tolerance by interferon-gamma (IFN-gamma)-dependent T-cell anergy in hemophilia A mice. OBJECTIVE: The thymus plays crucial roles in self-tolerance, with negative selection of self-reactive effector T cells and positive selection of self-reactive regulatory T cells. We investigated the possibility of the induction of antigen-specific immune tolerance by intrathymic injection of FVIII in hemophilia A mice. METHODS: Hemophilia A mice were injected with recombinant FVIII into the thymus under real-time high-resolution image guidance. RESULTS: Anti-FVIII inhibitory antibody titers in mice challenged with intravenous administration of FVIII were significantly lower in mice (n = 22) that had received thymic FVIII injection than in mice (n = 18) without thymic injection (9.4 +/- 2.3 vs. 122.5 +/- 27.6 BU mL(-1), respectively, P = 0.00078). The CD4(+) T cells from thymic-injected mice could not proliferate or produce interleukin (IL)-2, IL-12 and IFN-gamma in response to FVIII. The CD4(+)CD25(+) T cells generated from thymic-treated mice but not from naïve mice efficiently suppressed the in vitro proliferative response of CD4(+) T cells and blocked the in vivo development of anti-FVIII antibodies in the adoptive transfer. CONCLUSION: These data suggest that intrathymic administration of FVIII could result in immune tolerance by induction of FVIII-specific regulatory T cells.


Assuntos
Fator VIII/imunologia , Hemofilia A/imunologia , Timo/metabolismo , Animais , Autoanticorpos/biossíntese , Autoanticorpos/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Fator VIII/administração & dosagem , Citometria de Fluxo , Camundongos
7.
Cancer Gene Ther ; 16(1): 65-72, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18654608

RESUMO

The overexpression of reduced expression in immortalized cells (REIC)/Dickkopf-3 (Dkk-3), a tumor suppressor gene, induced apoptosis in human prostatic and testicular cancer cells. The aim of this study is to examine the potential of REIC/Dkk-3 as a therapeutic target against breast cancer. First, the in vitro apoptotic effect of Ad-REIC treatment was investigated in breast cancer cell lines and the adenovirus-mediated overexpression of REIC/Dkk-3 was thus found to lead to apoptotic cell death in a c-Jun-NH(2)-kinase (JNK) phosphorylaion-dependent manner. Moreover, an in vivo apoptotic effect and MCF/Wt tumor growth inhibition were observed in the mouse model after intratumoral Ad-REIC injection. As multidrug resistance (MDR) is a major problem in the chemotherapy of progressive breast cancer, the in vitro effects of Ad-REIC treatment were investigated in terms of the sensitivity of multidrug-resistant MCF7/ADR cells to doxorubicin and of the P-glycoprotein expression. Ad-REIC treatment in MCF7/ADR cells also downregulated P-glycoprotein expresssion through JNK activation, and sensitized its drug resistance against doxorubicin. Therefore, not only apoptosis induction but also the reversal of anticancer drug resistance was achieved using Ad-REIC. We suggest that REIC/Dkk-3 is a novel target for breast cancer treatment and that Ad-REIC might be an attractive agent against drug-resistant cancer in combination with conventional antineoplastic agents.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Adenoviridae , Antibióticos Antineoplásicos/farmacologia , Apoptose , Neoplasias da Mama/terapia , Regulação para Baixo , Doxorrubicina/farmacologia , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Terapia Genética , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antibióticos Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Quimiocinas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Doxorrubicina/uso terapêutico , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Feminino , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias
8.
Cancer Gene Ther ; 15(5): 315-22, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18274558

RESUMO

We previously constructed OBP-301 (Telomelysin, a telomerase-specific replication-competent adenovirus with human telomerase reverse transcriptase (hTERT) promoter), which showed a strong anticancer effect by inducing cell lysis of human non-small cell lung cancer and colorectal cancer cells. To investigate the utility of OBP-301 for prostate cancer treatment, we herein evaluate the cell killing and antitumor effects. First, in vitro hTERT-specific adenovirus transduction in human prostate cancer cells (LNCaP, PC3, DU145) was confirmed using OBP-401 (Telomelysin-green fluorescent protein (GFP)). There was no detectable GFP transduction in the human prostate normal cells (PrEC, PrSC). Consistently, the cell-killing effect of OBP-301 was observed only in the cancer cells. Second, using an in vivo subcutaneous LNCaP tumor model in nude mice, we demonstrated that three intratumoral OBP-301 injections (10(7) PFU per tumor x 3 days) were sufficient to eradicate the detectable LNCaP prostate tumor. We also demonstrated that the ispilateral treatment with OBP-301 significantly suppressed contralateral LNCaP tumor growth in both sides of the tumor model. Histological and immunohistochemical analyses revealed diffuse oncolytic degeneration and adenoviral E1A protein expression in both sides of the tumors. Therefore, in situ OBP-301 administration could be a promising therapeutic strategy against prostate cancer and its metastatic lesions.


Assuntos
Adenoviridae/genética , Antineoplásicos/uso terapêutico , Antivirais/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Taxoides/uso terapêutico , Telomerase/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos Fitogênicos/uso terapêutico , Modelos Animais de Doenças , Docetaxel , Terapia Genética/métodos , Proteínas de Fluorescência Verde/uso terapêutico , Humanos , Masculino , Camundongos
9.
Cancer Gene Ther ; 14(9): 765-72, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17599093

RESUMO

We had previously reported that REIC/Dkk-3, a member of the Dickkopf (Dkk) gene family, works as a tumor suppressor. In this study, we evaluated the therapeutic effects of an intratumoral injection with adenoviral vector encoding REIC/Dkk-3 gene (Ad-REIC) using an orthotopic mouse prostate cancer model of RM-9 cells. We also investigated the in vivo anti-metastatic effect and in vitro anti-invasion effect of Ad-REIC gene delivery. We demonstrated that the Ad-REIC treatment inhibited prostate cancer growth and lymph node metastasis, and prolonged mice survival in the model. These therapeutic responses were consistent with the intratumoral apoptosis induction and in vitro suppression of cell invasion/migration with reduced matrix metalloprotease-2 activity. We thus concluded that in situ Ad-REIC/Dkk-3 gene transfer may be a promising therapeutic intervention modality for the treatment of prostate cancer.


Assuntos
Adenoviridae/genética , Divisão Celular/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Modelos Biológicos , Metástase Neoplásica/genética , Neoplasias da Próstata/patologia , Transfecção , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose , Linhagem Celular Tumoral , Quimiocinas , Injeções Intralesionais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias da Próstata/genética
10.
Gene Ther ; 12(14): 1126-32, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15920464

RESUMO

Testicular germ cell-induced autoimmune orchitis is characterized by inflammatory cell infiltration followed by disturbance of spermatogenesis. Experimental autoimmune orchitis (EAO) is an animal model for human immunological male infertility; delayed-type hypersensitivity (DTH) response plays a key role in its induction. Interleukin-10 (IL-10) is a regulatory cytokine that is critical in preventing organ-specific autoimmune inflammation. To determine the effects on EAO of human IL-10 (hIL-10) gene transfer, C3H/He mice immunized by unilateral testicular injury were administered intramuscular (i.m.) injections of adeno-associated viral (AAV) vector-encoding hIL-10 on the day of immunization. Serum hIL-10 was detected beginning at 1 week postinjection, and peaked at 3 weeks. Histological examinations showed a significantly low incidence of orchitis and disturbance of spermatogenesis in AAV hIL-10-treated mice, and the DTH response to autologous testicular cells was significantly suppressed. Immunohistochemical analysis of IFN- and IL-2, T-cell-associated cytokines, in the spleen and testes revealed significantly fewer cytokine-expressing cells after treatment. We conclude that a single i.m. administration of AAV hIL-10 significantly suppresses EAO and hypospermatogenesis by regulating cell-mediated immunity in the testes.


Assuntos
Doenças Autoimunes/prevenção & controle , Terapia Genética/métodos , Interleucina-10/genética , Orquite/prevenção & controle , Adenoviridae/genética , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/fisiopatologia , Citocinas/metabolismo , Vetores Genéticos/genética , Humanos , Hipersensibilidade Tardia/prevenção & controle , Interleucina-10/sangue , Masculino , Camundongos , Camundongos Endogâmicos C3H , Orquite/imunologia , Orquite/fisiopatologia , Espermatogênese , Baço/imunologia , Testículo/imunologia
11.
Adv Myocardiol ; 4: 163-70, 1983.
Artigo em Inglês | MEDLINE | ID: mdl-6682992

RESUMO

This investigation was designed to evaluate the difference in DNA activity between biopsy specimens obtained from right and left ventricles. Nucleic DNA in the myocardial cells of hypertrophied and congestive forms of cardiomyopathy was analyzed to investigate the relationship between cell function and clinical manifestations. Endomyocardial biopsy specimens were obtained simultaneously from right ventricular septal wall and left ventricular inferolateral wall by a transcatheter biotome. Measurement of DNA was based on the Feulgen reaction and dual wavelength cytophotometry. In this series, 12 patients with hypertrophic cardiomyopathy and four patients with congestive cardiomyopathy were studied. In the normal heart, the DNA value (arbitrary units) of the right ventricle was 138.6, whereas that of the left ventricle was 144.4. In the hypertrophic group, the mean DNA value in the right ventricle was 279.9, whereas that of the left ventricle was 317.5. In the congestive group, the DNA mean value in the right ventricle was 108.8, whereas that of left ventricle was 144.8. The linear relationship (r = 0.67) between right and left ventricular DNA values suggests that cellular function of one ventricle is affected by that of the other side. Higher DNA values of the left ventricle may indicate the difference in work load between the ventricles. The relationships among DNA values, LV wall thickness, LV mass, and parameters of contractility were statistically high.


Assuntos
Cardiomiopatia Hipertrófica/metabolismo , DNA/biossíntese , Insuficiência Cardíaca/metabolismo , Adolescente , Adulto , Idoso , Biópsia , Cardiomiopatia Hipertrófica/patologia , DNA/análise , Feminino , Insuficiência Cardíaca/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Ploidias
17.
Artigo em Inglês | MEDLINE | ID: mdl-1031995

RESUMO

Myocardial infarction was produced in dogs, and the changes in nucleic acid synthetic activity were investigated quantitatively by microspectrophotometer in the myocardial cells as time progressed. DNA value, immediately after infarction, was greatly increased in comparison to that of the control group. At two weeks after infarction the value had increased to the highest level. After this point the value decreased, and, in 12 weeks, the mean value was back to the control level. Changes in RNA followed a pattern similar to DNA changes. The mechanism of the repair process of myocardial infarction was investigated.


Assuntos
DNA/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , RNA/metabolismo , Animais , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Cães
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA