Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Transplant ; 12(7): 1918-23, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22458552

RESUMO

Islet transplantation to treat type 1 diabetes has been limited in part by toxicities of current immunosuppression and recipient humoral sensitization. Blockade of the CD28/CD80/86 and CD40/CD154 pathways has shown promise to remedy both these limitations, but translation has been hampered by difficulties in translating CD154-directed therapies. Prior CD40-directed regimens have led to prolonged islet survival, but fail to prevent humoral allosensitization. We therefore evaluated the addition of CTLA4Ig to a CD40 blockade-based regimen in nonhuman primate (NHP) alloislet transplantation. Diabetic rhesus macaques were transplanted allogeneic islets using the CD40-specific antibody 3A8, basiliximab induction, and sirolimus with or without CTLA4Ig maintenance therapy. Allograft survival was determined by fasting blood glucose levels and flow cytometric techniques were used to test for donor-specific antibody (DSA) formation. CTLA4Ig plus 3A8, basiliximab and sirolimus was well tolerated and induced long-term islet allograft survival. The addition of CTLA4Ig prevented DSA formation, but did not facilitate withdrawal of the 3A8-based regimen. Thus, CTLA4Ig combines with a CD40-specific regimen to prevent DSA formation in NHPs, and offers a potentially translatable calcineurin inhibitor-free protocol inclusive of a single investigational agent for use in clinical islet transplantation without relying upon CD154 blockade.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos CD40/imunologia , Imunoconjugados/imunologia , Transplante das Ilhotas Pancreáticas , Isoanticorpos/biossíntese , Abatacepte , Animais , Sobrevivência de Enxerto/imunologia , Macaca mulatta
2.
Am J Transplant ; 11(5): 947-57, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21521467

RESUMO

The widespread clinical implementation of alloislet transplantation as therapy for type 1 diabetes has been hindered by the lack of suitable islet donors. Pig-to-human islet xenotransplantation is one strategy with potential to alleviate this shortage. Long-term survival of porcine islets has been achieved using CD154-specific antibodies to interrupt the CD40/CD154 costimulation pathway; however, CD154-specific antibodies seem unlikely candidates for clinical translation. An alternative strategy for CD40/CD154 pathway interruption is use of CD40-specific antibodies. Herein, we evaluate the ability of a chimeric CD40-specific monoclonal antibody (Chi220) to protect islet xenografts. Neonatal porcine islets (~50,000 IEQ/kg) were transplanted intraportally into pancreatectomized diabetic macaques. Immunosuppression consisted of induction therapy with Chi220 and the IL-2 receptor-specific antibody basiliximab, and maintenance therapy with sirolimus and the B7-specific fusion protein belatacept. Chi220 effectively promoted xenoislet engraftment and survival, with five of six treated recipients achieving insulin-independent normoglycemia (median rejection-free survival 59 days; mean 90.8 days, maximum 203 days). No thromboembolic phenomena were observed. CD40 represents a promising alternative to CD154 as a therapeutic target, and the efficacy of CD40-specific antibodies in islet xenotransplantation warrants further investigation.


Assuntos
Antígenos CD40/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Transplante Heterólogo/métodos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Basiliximab , Antígenos CD40/imunologia , Ligante de CD40/metabolismo , Feminino , Sobrevivência de Enxerto , Imuno-Histoquímica/métodos , Imunossupressores/uso terapêutico , Macaca mulatta , Masculino , Primatas , Receptores de Interleucina-2/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Sirolimo/uso terapêutico , Suínos
3.
Artigo em Inglês | MEDLINE | ID: mdl-21096605

RESUMO

Long QT syndrome is a repolarization disorder characterized by marked prolongation of QT interval. A clear consequence of long QT syndrome is the occurrence of a polymorphic ventricular tachycardia called Torsade de Pointes, which has been related to early after depolarizations (EADs) formation. This repolarizing disorder has been observed under pathological situations, such as heart failure, oxidative stress, ventricular hypertrophy and/or in the presence of pure class III antiarrhythmics. Under such pathologies electrophysiological changes affect the electrical activity of the cell. Lately, the enhancement of late sodium current (I(NaL)) and its role has become a source of interest. In this work, a mathematical model of I(NaL) has been proposed and incorporated to the ten Tussher model of the human ventricular action potential (AP), specifically in M cells. We simulated and analyzed the effects of I(NaL) enhancement in combination with LQT-related pathologies and administration of I(Kr) blockers, on the AP. This study demonstrates that I(NaL) prolongs AP duration (APD) in a rate-dependent manner. Indeed, a 10-fold increase of I(NaL) prolongs APD in 80% for a stimulation rate of 1 Hz and 100% for 0.25 Hz. Also, intracellular sodium concentration [Na(+)](i) significantly increases in the presence of enhanced I(NaL), increasing the probability of EADs formation through calcium overload in cells prone to develop EADs.


Assuntos
Sistema de Condução Cardíaco/fisiopatologia , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/fisiopatologia , Modelos Cardiovasculares , Bloqueadores dos Canais de Sódio/administração & dosagem , Canais de Sódio/efeitos dos fármacos , Sódio/metabolismo , Animais , Simulação por Computador , Sistema de Condução Cardíaco/efeitos dos fármacos , Humanos , Ativação do Canal Iônico/efeitos dos fármacos
4.
Am J Transplant ; 7(10): 2260-8, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17845561

RESUMO

Recent advances in human allogeneic islet transplantation have established beta-cell replacement therapy as a potentially viable treatment option for individuals afflicted with Type 1 diabetes. Two recent successes, one involving neonatal porcine islet xenografts transplanted into diabetic rhesus macaques treated with a costimulation blockade-based regimen and the other involving diabetic cynomolgus monkeys transplanted with adult porcine islet xenografts treated with an alternative multidrug immunosuppressive regimen have demonstrated the feasibility of porcine islet xenotransplantation in nonhuman primate models. In the current study, we assessed whether transplantation of adult porcine islet xenografts into pancreatectomized macaques, under the cover of a costimulation blockade-based immunosuppressive regimen (CD28 and CD154 blockade), could correct hyperglycemia. Our findings suggest that the adult porcine islets transplanted into rhesus macaques receiving a costimulation blockade-based regimen are not uniformly subject to hyperacute rejection, can engraft (2/5 recipients), and have the potential to provide sustained normoglycemia. These results provide further evidence to suggest that porcine islet xenotransplantation may be an attainable strategy to alleviate the islet supply crisis that is one of the principal obstacles to large-scale application of islet replacement therapy in the treatment of Type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Transplante das Ilhotas Pancreáticas/fisiologia , Transplante Heterólogo , Animais , Etanercepte , Heparina/farmacologia , Imunoglobulina G/farmacologia , Imunossupressores/uso terapêutico , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas/imunologia , Macaca mulatta , Monitorização Imunológica , Pancreatectomia , Primatas , Receptores do Fator de Necrose Tumoral , Proteínas Recombinantes de Fusão/farmacologia , Suínos , Transplante Heterólogo/imunologia
5.
Am J Transplant ; 7(2): 320-35, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17241112

RESUMO

A strategy for producing high-level hematopoietic chimerism after non-myeloablative conditioning has been established in the rhesus macaque. This strategy relies on hematopoietic stem cell transplantation after induction with a non-myeloablative dose of busulfan and blockade of the IL2-receptor in the setting of mTOR inhibition with sirolimus and combined CD28/CD154 costimulation blockade. Hematopoietic stem cells derived from bone marrow and leukopheresis products both were found to be successful in inducing high-level chimerism. Mean peripheral blood peak donor chimerism was 81% with a median chimerism duration of 145 days. Additional immune modulation strategies, such as pre-transplant CD8 depletion, donor-specific transfusion, recipient thymectomy or peritransplant deoxyspergualin treatment did not improve the level or durability of chimerism. Recipient immunologic assessment suggested that chimerism occurred amidst donor-specific down-regulation of alloreactive T cells, and the reappearance of vigorous T-mediated alloreactivity accompanied rejection of the transplants. Furthermore, viral reactivation constituted a significant transplant-related toxicity and may have negatively impacted the ability to achieve indefinite survival of transplanted stem cells. Nevertheless, this chimerism-induction regimen induced amongst the longest-lived stem cell chimerism reported to date for non-human primates and thus represents a platform upon which to evaluate emerging tolerance-induction strategies.


Assuntos
Quimerismo , Transplante de Células-Tronco Hematopoéticas/métodos , Terapia de Imunossupressão/métodos , Macaca mulatta/imunologia , Animais , Transplante de Medula Óssea/métodos , Bussulfano/farmacologia , Infecções por Citomegalovirus/complicações , Imunossupressores/farmacologia , Leucaférese/métodos , Macaca mulatta/genética , Receptores de Interleucina-2/antagonistas & inibidores , Sirolimo/farmacologia , Linfócitos T/imunologia , Tolerância ao Transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...