Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Transplant ; 12(1): 102-14, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21966953

RESUMO

Autophagy is a lysosomal degradation process of redundant or faulty cell components in normal cells. However, certain diseases are associated with dysfunctional autophagy. Rapamycin, a major immunosuppressant used in islet transplantation, is an inhibitor of mammalian target of rapamycin and is known to cause induction of autophagy. The objective of this study was to evaluate the in vitro and in vivo effects of rapamycin on pancreatic ß cells. Rapamycin induced upregulation of autophagy in both cultured isolated islets and pancreatic ß cells of green fluorescent protein-microtubule-associated protein 1 light chain 3 transgenic mice. Rapamycin reduced the viability of isolated ß cells and down-regulated their insulin function, both in vitro and in vivo. In addition, rapamycin increased the percentages of apoptotic ß cells and dead cells in both isolated and in vivo intact islets. Treatment with 3-methyladenine, an inhibitor of autophagy, abrogated the effects of rapamycin and restored ß-cell function in both in vitro experiments and animal experiments. We conclude that rapamycin-induced islet dysfunction is mediated through upregulation of autophagy, with associated downregulation of insulin production and apoptosis of ß cells. The results also showed that the use of an autophagy inhibitor abrogated these effects and promoted islet function and survival. The study findings suggest that targeting the autophagy pathway could be beneficial in promoting islet graft survival after transplantation.


Assuntos
Autofagia/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Sirolimo/farmacologia , Regulação para Cima/efeitos dos fármacos , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Células Cultivadas , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência
2.
J Nanosci Nanotechnol ; 12(8): 6526-30, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22962778

RESUMO

Aluminum nitride containing diamond-like carbon was fabricated with pulsed laser deposition without post processing. The compositions of the targets used were varied at 1, 5, 10, 15 at.% and pure carbon was used as a reference. The films were comprehensively characterized with Atomic force microscope (AFM), X-ray photoelectron spectroscopy (XPS) and Transmission electron microscopy (TEM). Roughness analysis using AFM showed an increasing root-mean-square (RMS) roughness with increasing AIN content in target, while XPS analysis showed that the aluminum-nitrogen bonding was still present in the films after the fabrication process. Microstructural studies and selected area electron diffraction (SAED) pattern confirmed the presence of AIN crystals in DLC matrix. This nanostructured composite material is useful for luminescence applications.

3.
J Nanosci Nanotechnol ; 11(12): 11011-4, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22409045

RESUMO

This paper presents direct growth of horizontally-aligned carbon nanotubes (CNTs) between two predefined various inter-spacing up to tens of microns of electrodes (pads) and its use as CNT field-effect transistors (CNT-FETs). Using the conventional photolithography technique followed by thin film evaporation and lift off, the catalytic electrodes (pads) were prepared, consisting of Pt, Al and Fe triple layers on SiO2/Si substrate. The grown CNTs were horizontally-aligned across the catalytic electrodes on the modified gold image furnace hot stage (thermal CVD) at 800 degrees C by using an alcohol vapor as the carbon source. Scanning and transmission electron microcopies (SEM/TEM) were used to observe the structure, growth direction and density of CNTs, while Raman spectrum analysis was used to indicate the degree of amorphous impurity and diameter of CNTs. Both single- and multi-wall CNTs with diameters of 1.1-2.2 nm were obtained and the CNT density was controlled by thickness of Fe catalytic layer. Following horizontally-aligned growth of CNTs, the electrical properties of back-gate CNT-FETs were measured and showd p-type conduction behaviors of FET.

4.
Br J Cancer ; 102(10): 1483-90, 2010 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-20407444

RESUMO

BACKGROUND: A striking efficiency of interferon (IFN)-based anticancer therapy for advanced hepatocellular carcinoma (HCC) has been reported. Because its clinical efficiency greatly depends on each patient's local response, prediction of local response is crucial. METHODS: Continuous exposure of IFN-alpha to parental PLC/PRF/5 cells (PLC-P) and a limiting dilution method resulted in the establishment of IFN-resistant cell clones (PLC-Rs). Microarray analyses of PLC-P and PLC-Rs identified insulin-like growth factor-binding protein 7 (IGFBP7) as one of the most significantly downregulated genes in PLC-Rs. Changes in anticancer effects of IFN-alpha were examined in HCC cells after genetic manipulation of IGFBP7 expression. The correlation between immunohistochemically determined IGFBP7 expression and the response to IFN-alpha/5-fluorouracil (5-FU) therapy was investigated in surgically resected HCC specimens. RESULTS: PLC-R cells showed a remarkable downregulation of IGFBP7 and resistance to IFN-alpha, compared with PLC-P. Parental PLC/PRF/5 cells transfected with short hairpin RNA against IGFBP7 showed a significant resistance to IFN-alpha relative to control cells (IC(50) fold increase=14.38 times). Insulin-like growth factor-binding protein 7 transfection into PLC-R restored sensitivity to IFN-alpha. In resected specimens, IGFBP7 expression significantly correlated with the response to IFN-alpha/5-FU therapy. CONCLUSION: IGFBP7 could be a useful predictor of the response to IFN-based therapy in advanced HCC.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Hepatocelular/genética , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Interferon-alfa/farmacologia , Neoplasias Renais/genética , Biomarcadores Tumorais/análise , Western Blotting , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/mortalidade , Linhagem Celular Tumoral , Fluoruracila/uso terapêutico , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Imunoprecipitação , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Estimativa de Kaplan-Meier , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/mortalidade , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Br J Cancer ; 103(10): 1617-26, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20978511

RESUMO

BACKGROUND: We reported recently the clinical efficiency of interferon (IFN)-α/5-fluorouracil (5-FU) combination therapy in advanced hepatocellular carcinoma (HCC). However, prediction of the response to the combination therapy remains unsatisfactory. The aim of this study was to investigate the anti-tumour effects of microRNA (miR)-21 on the sensitivity of HCC cells to IFN-α/5-FU and whether miR-21 can be used as a predictor of the response to such therapy in HCC. METHODS: Changes in the sensitivity of HCC cells (PLC/PRF/5 and HepG2) to IFN-α/5-FU were examined after transfection with pre-miR-21 or anti-miR-21. The correlation between miR-21 expression level, evaluated by qRT-PCR, and response to the therapy was also investigated in clinical HCC specimens. RESULTS: Hepatocellular carcinoma cells transfected with pre-miR-21 were significantly resistant to IFN-α/5-FU. Annexin V assay showed that the percentage of apoptotic cells was significantly lower in cells transfected with pre-miR-21 than control cells. Transfection of anti-miR-21 rendered HCC cells sensitive to IFN-α/5-FU, and such sensitivity was weakened by transfection of siRNAs of target molecules, PETN and PDCD4. miR-21 expression in clinical HCC specimens was significantly associated with the clinical response to the IFN-α/5-FU combination therapy and survival rate. CONCLUSIONS: The miR-21 in HCC cell lines and clinical HCC samples is a significant modulator of the anti-tumour effect of IFN-α and 5-FU. This suggests that miR-21 is a potentially suitable marker for the prediction of the clinical response to the IFN-α/5-FU combination therapy.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , MicroRNAs/genética , Antineoplásicos/antagonistas & inibidores , Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Divisão Celular/efeitos dos fármacos , Primers do DNA , Avaliação Pré-Clínica de Medicamentos , Resistência a Medicamentos , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/antagonistas & inibidores , Fluoruracila/uso terapêutico , Humanos , Imuno-Histoquímica , Interferon-alfa/antagonistas & inibidores , Interferon-alfa/uso terapêutico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , MicroRNAs/farmacologia , RNA Mensageiro/genética , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Transfecção
6.
Transplant Proc ; 41(1): 319-22, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19249545

RESUMO

Cell-mediated immunity, especially of human CD8+ cytotoxic T lymphocytes (CTLs) is believed to have an important role in the long-term survival of pig islet xenografts. Protection against human CD8+ CTL cytotoxicity may reduce the direct damage to pig islets and enable long-term xenograft survival in pig-to-human islet xenotransplantation. We have previously reported that c-FLIP(S/L) genes, which are potent inhibitors of death receptor-mediated proapoptotic signals through binding competition with caspase-8 for recruitment to the Fas-associated via death domain (FADD), markedly suppress human CD8+ CTL-mediated xenocytotoxicity. In addition, the cytoprotective effects of c-FLIP(L) seem to be significantly stronger than those of c-FLIP(S). Accordingly, in the present study, expression of c-FLIP(L) was induced in intact pig islets by adenoviral transduction. Consequently, the cytoprotective capacity of the transgene in pig islets was examined in in vitro and in vivo exposure to human CD8+ CTLs. Cells from untransduced islets or mock islets were sensitive to CD8+ CTL-mediated lysis (59.3% +/- 15.9% and 64.0% +/- 8.9% cytotoxicity, respectively). In contrast, cells from pig islets transduced with the c-FLIP(L) gene were markedly protected from lysis (30.5% +/- 3.5%). Furthermore, prolonged xenograft survival was elicited from pig islets transduced with this molecule as assessed using an islet transplant model using the rat kidney capsule. Thus, these data indicate that intact pig islets can be transduced to express c-FLIP(L) with adenovirus. Pig islets expressing c-FLIP(L) are significantly resistant to human CTL killing and further exhibit beneficial effects to prolong xenograft survival.


Assuntos
Adenoviridae/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Regulação da Expressão Gênica , Ilhotas Pancreáticas/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Sobrevivência Celular , Humanos , Imuno-Histoquímica , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/imunologia , Ratos , Ratos Endogâmicos Lew , Suínos , Transfecção , Transplante Heterólogo/imunologia
7.
Transplant Proc ; 41(1): 331-3, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19249549

RESUMO

The critical problem with clinical islet transplantation for patients with type 1 diabetes is the severe shortage of human donors. Pig islet xenotransplantation has the potential to provide a virtually unlimited source of donor pancreata. However, our previous studies demonstrated that cell-mediated rejection, especially human CD8(+) cytotoxic T lymphocyte (CTL)-mediated cytotoxicity, remains a major obstacle for long-term islet xenograft survival. Moreover, we have demonstrated that the overexpression of either membrane-bound human FasL (mFasL) or human decoy Fas antigen (decoy Fas) in pig islets not only prevented CTL xenocytotoxicity in vitro, but also prolonged histological survival of pig islet xenografts in vivo. Therefore, the aim of the present study was to determine whether adenoviral transfer of these genes into pig islets ex vivo prior to transplantation had a beneficial effect on posttransplantation glycemic control of diabetic recipients. Isolated pig islets were transfected with adenovirus vector carrying complementary DNA (cDNA) of either mFasL or decoy Fas. The transfected islets were transplanted under the kidney capsule of diabetic recipient rats. Rats transplanted with either mFasL- or decoy Fas-transfected pig islet grafts showed significantly suppressed blood glucose levels from 12 hours to 18 hours posttransplantation compared with control groups transplanted with empty vector-transfected pig islets. Unfortunately, blood glucose levels of these groups were increased, with no significant difference observed at 24 hours posttransplantation. However, transgenic expression of these molecules with clinically tolerable amount of immunosuppressants may be more effective to achieve islet xenograft survival in the future.


Assuntos
Adenoviridae/genética , Proteína Ligante Fas/genética , Sobrevivência de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Linhagem Celular , Células Endoteliais/fisiologia , Vetores Genéticos , Humanos , Imuno-Histoquímica , Ratos , Ratos Endogâmicos Lew , Suínos , Transfecção , Transplante Heterólogo
8.
Transplant Proc ; 41(1): 334-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19249550

RESUMO

Islet transplantation can provide insulin independence in patients with type 1 diabetes mellitus. However, islet allograft recipients exhibit a gradual decline in insulin independence, and only 10% do not require insulin at 5 years. This decline may reflect drug toxicity to islet beta cells. Rapamycin, a central immunosuppressant in islet transplantation, is a mammalian target of rampamycin inhibitor that induces autophagy. The relative contributions of autophagy in transplanted islets are poorly understood. Therefore, in the present study we sought to evaluate the effects of rapamycin on islet beta cells. Rapamycin treatment of islets resulted in accumulation of membrane-bound light chain 3 (LC3-II) protein, an early marker of autophagy. In addition, rapamycin treatment of isolated islets elicited not only reduction of viability but also downregulation of in vitro potency. To further examine the occurrence of autophagy in rapamycin-treated islets, we used GFP (green fluorescent protein)-LC3 transgenic mice that express a fluorescent autophagosome marker. The GFP-LC3 signals were markedly increased in rapamycin treated islets compared with control islets. In addition, to show improvement by blockade of autophagic signaling, islets were treated with rapamycin in the presence of 3-methyladenine, which inhibits autophagy. Thereafter, both islet viability and islet potency were dramatically improved. The number of GFP-LC3 dots clearly increased after 3-MA treatment. Thus, rapamycin treatment of islets induces autophagy in vitro. This phenomenon may contribute to the progressive graft dysfunction of transplanted islets. Therapeutically targeting this novel signaling may yield significant benefits for long-term islet survival.


Assuntos
Autofagia/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Sirolimo/farmacologia , Animais , Autofagia/fisiologia , Genes Reporter , Glucose/farmacologia , Cadeias Leves de Imunoglobulina/genética , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Transfecção
9.
Transplant Proc ; 41(1): 391-4, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19249564

RESUMO

Human CD8(+) cytotoxic T lymphocyte (CTL)-mediated cytotoxicity, which participates in xenograft rejection, is mediated mainly by the Fas/FasL apoptotic pathway. We previously developed methods to inhibit human CTL xenocytotoxicity by extracellular remodeling using overexpression of membrane-bound human FasL on pig xenograft cells, and by intracellular blockade of death receptor-mediated apoptotic signals, such as the Fas/FasL pathway using the pig c-FLIP(L) molecule. To investigate the cooperative effects of both membrane-bound FasL and pig c-FLIP(L), we cotransfected both genes into pig endothelial cells (PEC). The double remodeling with these molecules effectively prevented CD8(+) CTL killing. Although double transfectants and single high transfectants of either membrane-bound FasL or c-FLIP(L) gene displayed similar inhibition of CTL cytotoxicity, the expression levels of these 2 molecules in double transfectants were almost half the expression levels of single transfectants. Furthermore, to show in vivo prolongation of xenograft survival, we transplanted PEC transfectants under the rat kidney capsule. Prolonged survival was displayed by PEC double transfectant xenografts whereas those from either parental PEC or MOCK (vehicle control) were completely rejected by day 5 posttransplantation. These data suggested that intracellular and extracellular remodeling by coexpression of membrane-bound FasL and pig c-FLIP(L) in xenograft cells may prevent an innate cellular response to xenografts. The gene compatibility of these molecules to generate transgenic pigs may be sufficient to create a window of opportunity to facilitate long-term xenograft survival.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Proteína Ligante Fas/genética , Linfócitos T Citotóxicos/imunologia , Transplante Heterólogo/imunologia , Animais , Técnicas de Cultura de Células , Sobrevivência Celular/imunologia , Citotoxicidade Imunológica , DNA Complementar/genética , Humanos , Imuno-Histoquímica , Plasmídeos/genética , Suínos , Linfócitos T Citotóxicos/citologia , Transfecção
10.
Am J Transplant ; 8(2): 288-97, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18211505

RESUMO

Overcoming cell-mediated immunity, especially of human CD8(+) CTLs, is important for the success of xenotransplantation. Our group has previously reported that the cytotoxicity of human CD8(+) CTLs against pig endothelial cells (PEC) is highly detrimental and mediated in major part by the Fas/FasL apoptotic pathway. Cellular FLICE inhibitory protein (c-FLIP) was originally identified as an inhibitor of death-receptor signaling through binding competition with caspase-8 for recruitment to Fas-associated via death domain (FADD). Two major c-FLIP variants result from alternative mRNA splicing: a short, 26-KDa protein (c-FLIP(S)) and a long, 55-KDa form (c-FLIP(L)). The cytoprotective effects of c-FLIP(S/L) in xenograft cells remain controversial. This study demonstrates that the overexpression of c-FLIP(S/L) genes markedly suppress human CD8(+) CTL-mediated xenocytotoxicity and, in addition, the cytoprotective effects of c-FLIP(L) appear to be significantly stronger than those of c-FLIP(S). Furthermore, to prove the prolonged effects of xenograft survival, PEC transfectants with c-FLIP(S/L) genes were transplanted under rat kidney capsules. Prolonged survival was elicited from FLIP(S/L) transfectants, whereas parental PEC was completely rejected through day 5, posttransplant. Thus, intracellular remodeling with the overexpression of c-FLIP(S/L) in xenograft cells may avoid innate cellular attacks against xenografts and facilitate long-term xenograft survival.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Transfusão de Linfócitos , Linfócitos T Citotóxicos/imunologia , Transplante Heterólogo/imunologia , Animais , Caspases/metabolismo , Linhagem Celular , Citotoxicidade Imunológica , Fragmentação do DNA , Endotélio Vascular , Vetores Genéticos , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos
11.
Transplant Proc ; 40(2): 559-63, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18374129

RESUMO

Although the use of organs from alpha1,3-galactosyltransferase gene knockout pigs may prolong xenograft survival, resulting in overcoming antibody-mediated hyperacute rejection, pig xenografts will be destroyed directly by cell-mediated immunity, such as NK cells, macrophages, and CD8+ cytotoxic T lymphocytes (CTLs). Therefore, conquering cell-mediated immunity, especially of human CD8+ CTLs, is of particular importance to the success of long-term xenograft survival. We have previously reported that the cytotoxicity of human CD8+ CTLs is strong against pig endothelial cells (PEC) and mediated in major part by the Fas/FasL apoptotic pathway. Cellular FLICE inhibitory protein (c-FLIP) was originally identified as a potent inhibitor of death-receptor signaling through binding competition with caspase-8 for recruitment to Fas-associated via death domain (FADD). Two major c-FLIP variants result from alternative mRNA splicing: a short, 26-kDa protein (c-FLIP S) and a long, 55-kDa form (c-FLIP L). The present study demonstrated that overexpression of c-FLIP S/L genes in PEC markedly suppressed human CD8+ CTL-mediated xenocytotoxicity; moreover, the cytoprotective effects of c-FLIP L appeared to be significantly stronger than those of c-FLIP S. Furthermore, to prove the in vivo prolongation effects of xenograft survival, we transplanted PEC transfectants with c-FLIP(S/L) genes under the rat kidney capsule. Prolonged survival was displayed by xenografts of FLIP S/L PEC transfectants, whereas xenografts of parental PEC were completely rejected by day 5 posttransplantation. Thus, intracellular blocking of death receptor-mediated apoptotic signals by overexpression of c-FLIP S/L in xenograft cells may prevent innate cellular attacks against xenografts opening the window of opportunity for long-term xenograft survival.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T Citotóxicos/imunologia , Transplante Heterólogo/imunologia , Animais , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Linhagem Celular , Sobrevivência Celular/imunologia , Fragmentação do DNA , Endotélio Vascular , Expressão Gênica , Humanos , Plasmídeos , Suínos
12.
Transplant Proc ; 40(2): 438-40, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18374094

RESUMO

BACKGROUND: We previously demonstrated that syngeneic pancreas transplantation has a potential to reverse diabetes even in a rat model of type 2 diabetes mellitus, namely Spontaneously Diabetic Torii (SDT; RT1a). The onset of diabetes was significantly delayed in the pancreas transplant recipients. We speculated that perfect diabetic control achieved by pancreas transplantation showed a beneficial effect on the native pancreata & recipients. MATERIALS AND METHODS: Twenty-five-week-old diabetic SDT rats were divided into 3 groups: untreated controls and syngeneic and allogeneic transplant recipients. We transplanted pancreaticoduodenal grafts from nondiabetic 10-week-old SDT rats and from 10-week-old allogeneic Dark Agouti (DA; RT1a) rats using daily administration of FK506. RESULTS: Untreated SDT rats showed disappearance of pancreatic and duodenal homeobox-1 (PDX-1) expression in the pancreas and a marked decrease in beta-cell mass. Among syngeneic and allogeneic pancreas transplant recipients, islet-like cell clusters were found in the native pancreata. The beta-cell mass at 40 weeks of age was significantly increased in the native pancreata of recipients compared with age-matched controls. Interestingly, we observed the reexpression of PDX-1 in the nuclei of islet-like cell clusters. CONCLUSIONS: Our results indicated the benefits of avoiding glucose toxicity by pancreas transplantation which induced PDX-1 expression in the native pancreata of type 2 diabetic recipients, resulting in regeneration of beta cells in the native pancreata.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Experimental/cirurgia , Diabetes Mellitus Tipo 2/cirurgia , Células Secretoras de Insulina/fisiologia , Transplante de Pâncreas , Animais , Peso Corporal , Proteínas de Homeodomínio/genética , Células Secretoras de Insulina/citologia , Masculino , Ratos , Ratos Endogâmicos , Regeneração , Transativadores/genética , Transplante Homólogo , Transplante Isogênico
13.
Transplant Proc ; 40(2): 477-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18374107

RESUMO

The success of pancreatic islet transplantation is limited because of the severe shortage of allogeneic pancreas donors. Accordingly, pig islets are considered to be an attractive, promising alternative. However, cell-mediated immunity, especially CD8+ cytotoxic T lymphocyte (CTL)-mediated cytotoxicity, remains a formidable barrier to prevent long-term islet survival in xenograft recipients. Therefore, it is particularly important to explore methods to specifically prevent cell-mediated immunity against pig islets. Our group previously demonstrated that the overexpression of either membrane-bound human FasL or human decoy Fas antigen in pig endothelial cells prevented CTL xenocytotoxicity. In this study, we assessed the cytoprotective effects of adenoviral-mediated overexpression of either membrane-bound human FasL or human decoy Fas antigen in pig islets to inhibit CTL xenocytotoxicity. The CTL-mediated killing of pig islets infected with an adenoviral vector carrying either membrane-bound human FasL or human decoy Fas was significantly reduced compares with that of control pig islets transfected with adenoviral vector encoding enhanced green fluorescent protein (EGFP). Moreover, we transfected pig islets with these molecules to confirm their cytoprotective effects in in vivo studies. The significant long-term survival of pig islets expressing these molecules was elicited through days 3 to 5 posttransplantation. Thus, these results demonstrated that the remodeling of either death receptor or death ligand on pig islets by adenoviral gene transfer prevented innate cellular immunity against xeno-islet grafts facilitating long-term xenograft survival.


Assuntos
Adenoviridae/genética , Proteína Ligante Fas/genética , Sobrevivência de Enxerto/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Transplante Heterólogo/fisiologia , Animais , Linhagem Celular , Clonagem Molecular , Endotélio Vascular , Regulação da Expressão Gênica , Vetores Genéticos , Humanos , Ratos , Suínos , Receptor fas/genética
15.
J Clin Invest ; 105(3): 301-10, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10675356

RESUMO

Xenograft recipients produce large amounts of high-affinity anti-Gal IgG in response to Galalpha1-3Galbeta1- 4GlcNAc-R (alpha-gal) epitopes on the graft. In contrast, ABO-mismatched allograft recipients undergo "accommodation," a state of very weak immune response to ABO antigens. These differences in anti-carbohydrate immune response were studied in alpha1,3galactosyltransferase knock-out mice. Pig kidney membranes administered to these mice elicited extensive production of anti-Gal IgG, whereas allogeneic kidney membranes expressing alpha-gal epitopes elicited only a weak anti-Gal IgM response. Anti-Gal IgG response to xenograft membranes depended on helper T cell activation and was inhibited by anti-CD40L antibody. These T cells were activated by xenopeptides and not by alpha-gal epitopes. Moreover, allogeneic cell membranes manipulated to express xenoproteins also induced anti-Gal IgG response. Xenoglycoproteins with alpha-gal epitopes are processed by anti-Gal B cells. Xenopeptides presented by these cells activate a large repertoire of helper T cells required for the differentiation of anti-Gal B cells into cells secreting anti-Gal IgG. Alloglycoproteins with alpha- gal epitopes have very few immunogenic peptides and fail to activate helper T cells. Similarly, ineffective helper T-cell activation prevents a strong immune response to blood group antigens in ABO-mismatched allograft recipients, thus enabling the development of accommodation.


Assuntos
Imunologia de Transplantes , Trissacarídeos/imunologia , Animais , Tipagem e Reações Cruzadas Sanguíneas , Membrana Celular/imunologia , Membrana Celular/transplante , Epitopos/imunologia , Imunidade , Transplante de Rim , Camundongos , Camundongos Knockout , Suínos , Transplante Heterólogo/imunologia , Transplante Homólogo/imunologia
16.
Transplant Proc ; 38(10): 3181-3, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17175216

RESUMO

AIMS: The potency of immunosuppression is a critical factor in small bowel transplantation (SBTx). FTY720 altered lymphocyte trafficking and prevented the donor T cells from migrating into target organs, resulting in the prolongation of recipient survival in acute graft-versus-host disease (GVHD) of SBTx. However, the effect of FTY720 on donor T cells in the chronic phase of GVHD following SBTx remains unclear. METHODS: Heterotopic SBTx was performed in a WF-to-F1 (WF x ACI) rat combination. Recipients were given FTY720 for 14 days after SBTx. The subpopulations of donor-derived T cells and the cytokine production in the target tissues were evaluated on postoperative day 150. RESULTS: FTY720 treatment significantly prolonged recipient survival over 150 days without any clinical signs of GVHD. The numbers of donor-derived CD4+ and CD8+ T cells in the peripheral blood, mesenteric lymph nodes, and Peyer's patches of recipients were maintained at low levels on postoperative 150, which were almost similar to the levels on postoperative day 14. In the host lamina propria, however, a significant higher number of donor T cells (CD4+, 18.4 +/- 4.3 x 10(4); CD8+, 13.9 +/- 3.6 x 10(4)) were still observed on postoperative day 150. Production of interferon-gamma was significantly reduced in target tissues by FTY720 treatment both in the acute and chronic phase. However, interleukin-4 and interleukin-10 production, which was significantly higher on day 14, returned to the level of naive rats in the chronic phase. CONCLUSIONS: A 14-day treatment of FTY720 induced tolerance in our SBTx model. Down-regulation of both Th1 and Th2 immune response was observed in the chronic phase.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Tolerância Imunológica , Imunossupressores/uso terapêutico , Intestino Delgado/transplante , Propilenoglicóis/uso terapêutico , Esfingosina/análogos & derivados , Linfócitos T/imunologia , Animais , Citocinas/biossíntese , Cloridrato de Fingolimode , Doença Enxerto-Hospedeiro/prevenção & controle , Masculino , Ratos , Ratos Endogâmicos ACI , Ratos Endogâmicos WF , Esfingosina/uso terapêutico , Transplante Homólogo , Transplante Isogênico
17.
Transplant Proc ; 38(10): 3286-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17175251

RESUMO

Pig islets are considered to be most suitable source of islets for xenotransplantation into patients with type 1 diabetes mellitus. However, cellular rejection, especially CD8+ CTL-mediated cytotoxicity, remains a formidable barrier preventing long-term xenograft survival. Our previous study demonstrated that human CD8+ CTLs were highly detrimental to xenograft cells and that this strong cytotoxicity of human CTLs was mediated mainly by the Fas/FasL apoptotic pathway. Furthermore, we exploited novel methods for inhibiting human CD8+ CTL-mediated xenocytotoxicity with overexpression of membrane-bound human FasL and human decoy Fas antigen in xenografted cells. In the present study, we assessed the cytoprotective effects of these novel inhibitory molecules overexpressed by an adenoviral-mediated system in pig islets. Isolated pig islets were transfected with adenovirus vector encoding either human decoy Fas or membrane-bound human FasL genes. Thirty percent to 60% of transfected pig islets expressed these molecules producing 60% to 88% suppression of CTL killing compared with parental pig islets. These data indicated that pig islet grafts isolated from transgenic pigs with either membrane-bound human FasL or human decoy Fas antigen genes may control the innate cellular response to xenografts, and creating a window of opportunity to facilitate xenograft survival.


Assuntos
Adenoviridae/genética , Citotoxicidade Imunológica , Proteína Ligante Fas/genética , Ilhotas Pancreáticas/imunologia , Transfusão de Linfócitos , Linfócitos T Citotóxicos/transplante , Receptor fas/genética , Animais , Humanos , Suínos , Transfecção , Transplante Heterólogo
18.
Transplant Proc ; 38(10): 3315-7, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17175259

RESUMO

Human CD8+ cytotoxic T lymphocyte (CTL)-mediated cytotoxicity in xenograft recipients is an important obstacle for successful xenotransplantation of pig organs into humans. In our previous study, we demonstrated that xenocytotoxicity of human CD8+ CTL detrimental to pig endothelial cells (PEC) is mediated mainly by the Fas/FasL apoptotic pathway. Furthermore, we developed new methods to prevent this CTL killing by extracellular remodeling using overexpression of human decoy Fas antigen and membrane-bound human FasL on pig xenograft cells. The cellular FLICE-inhibitory protein (c-FLIP), a caspase-8 inhibitor that lacks the cysteine domain, is a negative regulator of death receptor-mediated apoptosis. c-FLIP proteins exist as long (c-FLIP(L)) and short (c-FLIPs) splice variants, both capable of protecting cells from death receptor-mediated apoptosis. In this report, we have demonstrated that both pig c-FLIPs and pig c-FLIP(L) significantly inhibit human CD8+ CTL-mediated xenocytotoxicity toward stably transfected PEC, although the expression level of pig Fas antigen on cell surface was not changed. These data suggested that intracellular remodeling with overexpression of pig c-FLIP in xenograft cells may decrease the innate cellular responses against xenografts, facilitating long-term xenograft survival.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Citotoxicidade Imunológica , Endotélio Vascular/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Linhagem Celular , Primers do DNA , DNA Complementar , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Regulação da Expressão Gênica/imunologia , Sobrevivência de Enxerto , Humanos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos , Transfecção , Transplante Heterólogo/imunologia
19.
Transplant Proc ; 37(10): 4607-9, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16387181

RESUMO

The principal barrier to the use of pigs as donors to humans is hyperacute rejection mediated by the interaction of alpha-gal abundantly expressed on pig cells and the natural anti-Gal antibody, abundantly produced in humans. This antibody-mediated hyperacute rejection may be overcome by an alpha1, 3 galactosyltransferase gene-knockout pig. However, xenograft cells could be rejected by T cells, especially CD8+ cytotoxic T lymphocytes (CTL)-mediated response, because these elements show great cytotoxicity against xenografted cells. We previously demonstrated that the Fas/FasL pathway is a major contributor to CD8+ CTL function. Furthermore, we sought to prevent this cytotoxicity by overexpression of membrane-bound FasL carrying the deletion at the metalloproteinase cleavage site or by decoy Fas antigen that does not contain the death domain in its cytoplasmic region. To investigate the effects of coexpression of these molecules, we cotransfected both genes into swine endothelial cells (SEC). The double-overexpression effectively prevented CD8+ CTL-mediated killing. Although cotransfectants and single transfectants of either membrane-bound FasL or decoy Fas gene showed similar inhibition of cytotoxicity, the expression levels of decoy Fas in SEC cotransfectants were much lower than those of decoy Fas single transfectants. These data suggest that beneficial effects for prevention of CTL-mediated xenocytotoxicity may be produced by the double expression of these molecules. The overexpression of both molecules on xenografted cells may decrease the innate cellular response to xenografts creating a window of opportunity to facilitate xenograft survival.


Assuntos
Rejeição de Enxerto/imunologia , Glicoproteínas de Membrana/genética , Linfócitos T Citotóxicos/imunologia , Transplante Heterólogo/imunologia , Fatores de Necrose Tumoral/genética , Receptor fas/genética , Doença Aguda , Animais , Sobrevivência Celular/imunologia , Citotoxicidade Imunológica , DNA Complementar/genética , Proteína Ligante Fas , Humanos , Glicoproteínas de Membrana/imunologia , Plasmídeos , Suínos , Transfecção , Fatores de Necrose Tumoral/imunologia , Receptor fas/imunologia
20.
Transplant Proc ; 37(1): 226-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15808602

RESUMO

AIMS: Type 2 diabetes is characterized by a combination of insulin resistance and pancreatic beta-cell dysfunction. Although pancreas transplantation (PTx) is mainly performed in patients with type 1 disease, both clinical and experimental data have demonstrated that PTx improves insulin sensitivity in type 2 diabetic recipients. However, it remains unclear whether PTx has the potential to induce islet neogenesis in a recipient's native pancreas. METHODS: Nondiabetic 10-week-old and diabetic (defined as blood glucose level >250 mg/dL) 25-week-old (average onset age of diabetes) male spontaneously diabetic Torii (SDT; RT1(a)) rats served as donors and recipients, respectively. RESULTS: In nontreated control SDT rats, beta-cell mass gradually decreased and blood glucose levels progressively increased (>600 mg/dL after 40 weeks of age). In PTx rats, however, the onset of diabetes was significantly delayed (>47.5 +/- 18.2 [graft age] versus 25.2 +/- 3.9 weeks in control rats). On immunohistochemical staining, insulin-secreting islets were observed in the naive pancreata of 40-week-old recipients with PTx (PTx40w), whereas no islets were found in 40-week-old control SDT rats. Moreover, the islets in the native pancreata of PTx40w recipients were located close to ductal structures, and PDX-1 (pancreatic duodenal homeobox-1)-positive cells were more clearly visible. These results indicate the possibility of beta-cell regeneration in the recipient native pancreas by avoiding glucose toxicity under normoglycemic condition achieved by PTx. CONCLUSIONS: Pancreas transplantation has beneficial effects on impaired islet, inducing regeneration in the spontaneously diabetic Torii rat.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Ilhotas Pancreáticas/fisiologia , Transplante de Pâncreas/fisiologia , Regeneração/fisiologia , Envelhecimento/fisiologia , Animais , Biomarcadores , Diabetes Mellitus Tipo 1/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Ilhotas Pancreáticas/citologia , Masculino , Transplante de Pâncreas/métodos , Ratos , Ratos Mutantes , Transativadores/genética , Transativadores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA