Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Am J Transplant ; 17(3): 682-691, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27501352

RESUMO

We hypothesized that de novo donor-specific antibody (DSA) causes complement-dependent endothelial cell injury in kidney transplants, as assessed by expression of endothelial cell-associated transcripts (ENDATs), that may be attenuated through complement inhibition. In total, 15 participants (five control, 10 treatment) with DSA and deteriorating renal function were enrolled. The treatment group received 6 mo of eculizumab followed by 6 mo of observation, whereas controls were observed. The primary end point was percentage change in estimated GFR (eGFR) trajectory over the treatment period. The treatment group had an improved eGFR trajectory versus control, based on our predetermined two-sided 0.10 significance level (p = 0.09). Within-subject analysis of treated participants at 6-mo intervals did not show significant change (p = 0.60). Modeling C1q status showed that C1q-positive patients had significantly higher mean eGFR than patients with negative C1q (p = 0.04). Biopsies revealed elevated renal ENDATs in most participants, but ENDATs were not reduced with complement inhibition. Our data suggest that eculizumab treatment may stabilize kidney function in patients with chronic persistent DSA based on our pilot a priori significance threshold. ENDAT expression predicative of acute humoral injury is not reduced with complement inhibition in this chronic setting. Further studies will be necessary to determine which patients may benefit from eculizumab.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Rejeição de Enxerto/tratamento farmacológico , Sobrevivência de Enxerto/efeitos dos fármacos , Isoanticorpos/sangue , Falência Renal Crônica/cirurgia , Transplante de Rim/efeitos adversos , Adolescente , Adulto , Idoso , Doença Crônica , Complemento C5/antagonistas & inibidores , Inativadores do Complemento/uso terapêutico , Intervenção Educacional Precoce , Feminino , Seguimentos , Taxa de Filtração Glomerular , Rejeição de Enxerto/etiologia , Sobrevivência de Enxerto/imunologia , Humanos , Testes de Função Renal , Doadores Vivos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Prognóstico , Fatores de Risco , Doadores de Tecidos , Transplantados , Adulto Jovem
2.
Am J Transplant ; 16(10): 2865-2876, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27104811

RESUMO

Allograft vasculopathy (AV) is characterized by diffuse stenoses in the vasculature of solid organ transplants. Previously, we developed two humanized models showing that alloantibody and ischemia reperfusion injury (IRI) exacerbated T cell-mediated AV in human arterial xenografts in vivo. Herein we examined a causal role for terminal complement activation in both settings. IRI, in contrast to alloantibody, elicited widespread membrane attack complex (MAC) assembly throughout the vessel wall. Both alloantibody and IRI caused early (24 h) and robust endothelial cell (EC) activation localized to regions of intimal MAC deposition, indicated by increases in nuclear factor kappa B (NF-κB)-inducing kinase, an MAC-dependent activator of noncanonical NF-kB, VCAM-1 expression and Gr-1+ neutrophil infiltration. Endothelial cell activation by alloantibody was inhibited by antimouse C5 mAb, but not by anti-C5a mAb or by control mAb, implicating MAC as the primary target of anti-C5 mAb. Antimouse C5 mAb significantly reduced alloantibody- and IRI-enhanced T cell infiltration and AV-like changes, including neointimal hyperplasia as well as intraluminal thrombosis in a subset of IRI-treated arterial grafts. These results indicate that increased AV lesion formation in response to either alloantibody or IRI is dependent on complement C5 activation and, accordingly, inhibition of this pathway may attenuate AV.


Assuntos
Complemento C5/antagonistas & inibidores , Isoanticorpos/imunologia , Traumatismo por Reperfusão/complicações , Linfócitos T/imunologia , Doenças Vasculares/prevenção & controle , Aloenxertos , Animais , Anticorpos Monoclonais/farmacologia , Células Cultivadas , Ativação do Complemento , Humanos , Camundongos , Camundongos SCID , NF-kappa B/metabolismo , Doenças Vasculares/etiologia
3.
Transplant Proc ; 36(5): 1585-91, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15251390

RESUMO

The hallmark of endothelial activation, an early and critical step in many alloimmune and inflammatory responses, is the transcriptional induction and expression of endothelial adhesion molecules (eg, vascular cell adhesion molecule-1 [VCAM-1]). We assessed the feasibility of VCAM-1-targeted in vivo detection of endothelial activation using I-125-labeled-F(ab')2 fragments of E1/6, a monoclonal antibody against human but not murine VCAM-1. The Kd and Bmax, determined by saturation binding in tumor necrosis factor (TNF)-activated human endothelial cells (ECs), were 3.2 +/- 0.6 nmol/L and 5600 +/- 300 binding sites per EC, respectively. Biodistribution and in vivo binding characteristics of I-125-E1/6 F(ab')2 were assessed in a novel chimeric human/mouse model, in which human skin (as a source of human microvasculature) is grafted onto SCID/beige mice. I-125-E1/6 F(ab')2 localized to TNF-activated human skin grafts as detected by autoradiography and gamma well-counting. Relative uptakes (uptake in human skin graft/uptake in the surrounding mouse skin) were, respectively, 2.6 +/- 0.8 (n = 14) and 1.6 +/- 0.3 (n = 12) for E1/6 and MOPC-21, an isotype-matched control antibody (P < .01). The preferential uptake in human skin graft was not due to differences in tissue vascularity assessed by Tc-99m-labeled murine red blood cells. In conclusion, the chimeric human/mouse model is a novel experimental tool for in vivo evaluation of human endothelial cell-specific radiopharmaceuticals. Although I-125-E1/6 F(ab')2 localized to human skin grafts, the limited number of VCAM-1 molecules/endothelial cell adversely affects its suitability as a target for in vivo imaging of endothelial activation.


Assuntos
Endotélio Vascular/imunologia , Microcirculação/imunologia , Molécula 1 de Adesão de Célula Vascular/genética , Animais , Afinidade de Anticorpos , Células Cultivadas , Eritrócitos/imunologia , Citometria de Fluxo , Humanos , Fragmentos Fab das Imunoglobulinas/análise , Imuno-Histoquímica , Camundongos , Modelos Animais , Pele/imunologia , Transplante de Pele/imunologia , Veias Umbilicais/imunologia
4.
Transplantation ; 71(11): 1622-30, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11435975

RESUMO

BACKGROUND: We have previously demonstrated that human artery grafts transplanted to immunodeficient mice are infiltrated and injured by unsensitized allogeneic human T cells. We extended our investigations to human anti-porcine xenoresponses in this model. METHODS: Pig coronary artery segments were interposed into the infrarenal aorta of severe combined immunodeficiency/beige mice. After 7 days, certain recipients were reconstituted with human leukocytes and/or treated with proinflammatory cytokines. The grafts were harvested after 1-70 days and examined by histology, immunohistochemistry, and morphometry. RESULTS: Pig artery grafts from untreated mice had no evidence of injury, leukocytic infiltrate, or endothelial cell activation up to 70 days postoperatively, despite deposition of murine complement. Host reconstitution with human peripheral blood mononuclear cells resulted in a discrete population of circulating T cells that did not infiltrate or injure the grafts up to 28 days after adoptive transfer. Administration of porcine interferon-gamma for up to 28 days sustained the expression of graft vascular cell adhesion molecule-1 and major histocompatibility complex antigens, but did not initiate recruitment of human leukocytes. In contrast, treatment with human tumor necrosis factor for 7 days induced the de novo expression of porcine E-selectin by graft endothelial cells and elicited human T cell infiltration and human peripheral blood mononuclear cell-dependent vascular injury. CONCLUSIONS: The human peripheral blood mononuclear cell-severe combined immunodeficiency/beige mouse model identifies a significant difference between human T cell allogeneic and xenogeneic responses in vivo. Xenografts with quiescent endothelium are not infiltrated or injured by T cells under the same conditions in which allografts are rejected. Activation of pig coronary artery endothelial cells by human tumor necrosis factor, but not porcine interferon-gamma, elicits cellular xenoresponses.


Assuntos
Vasos Coronários/transplante , Endotélio Vascular/fisiologia , Endotélio Vascular/transplante , Imunodeficiência Combinada Severa/cirurgia , Animais , Artérias/efeitos dos fármacos , Artérias/patologia , Artérias/transplante , Células Sanguíneas/patologia , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/patologia , Rejeição de Enxerto/induzido quimicamente , Humanos , Tolerância Imunológica , Interferon gama/farmacologia , Camundongos , Camundongos SCID , Imunodeficiência Combinada Severa/sangue , Suínos , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Imunologia de Transplantes , Fator de Necrose Tumoral alfa/farmacologia
5.
Lab Invest ; 81(3): 327-34, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11310826

RESUMO

Interleukin-11 (IL-11) reduces injury both in vivo and in vitro, but the mechanisms are unknown. Stimulation of serum- and growth factor-deprived HUVEC with IL-11 increased survivin mRNA and protein expression levels in a dose-dependent manner, with maximal induction at 50 to 100 ng/ml of IL-11. Survivin mRNA expression peaked after 3 to 6 hours of IL-11 treatment and decreased by 24 hours. Survivin protein expression was maximal at 6 hours of treatment and remained elevated through 24 hours. Survivin induction may be mediated by activation of protein kinase B/Akt, but IL-11 failed to activate this pathway in HUVEC. IL-11 did activate signal transducer and activator of transcription (STAT)-3 and IL-11 failed to induce survivin expression in HUVEC transduced with a dominant-negative STAT3 mutant, whereas control-transduced HUVEC responded normally. An IL-11 transgene caused increased survivin mRNA expression in mice compared with control littermates. Intradermal injection of IL-11 (500 ng) into human skin xenografts on immunodeficient mice up-regulated survivin protein in microvascular endothelium and epithelial keratinocytes. We conclude that IL-11 induces expression of survivin, an antiapoptotic protein, in vitro and in vivo, and identify STAT3 as a critical mediator of this response.


Assuntos
Endotélio Vascular/metabolismo , Interleucina-11/metabolismo , Proteínas Associadas aos Microtúbulos , Proteínas/genética , Transdução de Sinais/imunologia , Transcrição Gênica/imunologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Expressão Gênica/imunologia , Humanos , Proteínas Inibidoras de Apoptose , Interleucina-11/imunologia , Proteínas de Neoplasias , Fosforilação , Proteínas/imunologia , Proteínas/metabolismo , RNA Mensageiro/análise , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Serina/metabolismo , Survivina , Transativadores/genética , Transativadores/metabolismo , Transgenes , Veias Umbilicais/citologia
8.
J Immunol ; 164(12): 6601-9, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10843720

RESUMO

TNF activates endothelial cells to express cell surface molecules that are necessary to recruit a local infiltrate of leukocytes. Because the actions of this proinflammatory cytokine are not species restricted, we investigated whether human TNF can up-regulate porcine endothelial adhesion molecules to elicit human T cell infiltration and damage of pig skin xenografts in a chimeric immunodeficient mouse model. We have previously demonstrated the vigorous rejection of human skin allografts and the absence of injury to porcine skin xenografts in human PBMC-SCID/beige mice. Intradermal administration of human TNF at high doses (600 or 2000 ng) caused nonspecific inflammatory damage of pig skin grafts, whereas low concentrations of TNF (60 or 200 ng) resulted in human PBMC-dependent injury of porcine endothelial cells. There was a strong correlation among pig skin xenograft damage, human T cell infiltration, and the TNF-induced up-regulation of swine MHC class I and class II molecules, VCAM-1, and, in particular, the de novo expression of porcine E-selectin. The microvascular damage and leukocytic infiltration elicited by TNF were enhanced by porcine IFN-gamma, suggesting that xenografts may be less prone to cytokine-mediated injury due to the species-restricted effects of recipient IFN-gamma. Our results indicate that maintenance of a quiescent endothelium, which does not express E-selectin or other activation-dependent adhesion molecules, is important in preventing human anti-porcine T cell xenoresponses in vivo and that TNF signaling molecules and TNF-responsive gene products are appropriate therapeutic targets to protect against human T cell-mediated rejection of pig xenografts.


Assuntos
Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Transplante de Pele/imunologia , Transplante de Pele/patologia , Transplante Heterólogo , Fator de Necrose Tumoral alfa/toxicidade , Transferência Adotiva , Adulto , Animais , Moléculas de Adesão Celular/biossíntese , Relação Dose-Resposta Imunológica , Sinergismo Farmacológico , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Rejeição de Enxerto/fisiopatologia , Antígenos de Histocompatibilidade/biossíntese , Humanos , Interferon gama/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Microcirculação/imunologia , Microcirculação/patologia , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/fisiopatologia , Transplante de Pele/efeitos adversos , Suínos , Linfócitos T/transplante , Transplante Heterólogo/efeitos adversos , Regulação para Cima/imunologia
9.
Nature ; 403(6766): 207-11, 2000 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-10646607

RESUMO

Atherosclerosis and post-transplant graft arteriosclerosis are both characterized by expansion of the arterial intima as a result of the infiltration of mononuclear leukocytes, the proliferation of vascular smooth muscle cells (VSMCs) and the accumulation of extracellular matrix. They are also associated with the presence of the immunomodulatory cytokine interferon-gamma (IFN-gamma). Moreover, in mouse models of atheroma formation or allogeneic transplantation, the serological neutralization or genetic absence of IFN-gamma markedly reduces the extent of intimal expansion. However, other studies have found that exogenous IFN-gamma inhibits cultured VSMC proliferation and matrix synthesis, and reduces intimal expansion in response to mechanical injury. This discrepancy is generally explained by the idea that IFN-gamma either directly activates macrophages, or, by increasing antigen presentation, indirectly activates T cells within the lesions of atherosclerosis and graft arteriosclerosis. These activated leukocytes are thought to express the VSMC-activating cytokines and cell-surface molecules that cause the observed arteriosclerotic responses. Here we have inserted pig and human arteries into the aorta of immunodeficient mice, and we show that IFN-gamma can induce arteriosclerotic changes in the absence of detectable immunocytes by acting on VSMCs to potentiate growth-factor-induced mitogenesis.


Assuntos
Arteriosclerose/etiologia , Interferon gama/fisiologia , Leucócitos/fisiologia , Adulto , Animais , Arteriosclerose/imunologia , Divisão Celular/fisiologia , Células Cultivadas , Vasos Coronários/transplante , Antígenos de Histocompatibilidade/biossíntese , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Leucócitos/imunologia , Camundongos , Camundongos SCID , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Fator de Crescimento Derivado de Plaquetas/biossíntese , Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/biossíntese , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Suínos , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA