Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Transplant ; 16(12): 3416-3429, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27172087

RESUMO

Pretransplant autoantibodies to LG3 and angiotensin II type 1 receptors (AT1R) are associated with acute rejection in kidney transplant recipients, whereas antivimentin autoantibodies participate in heart transplant rejection. Ischemia-reperfusion injury (IRI) can modify self-antigenic targets. We hypothesized that ischemia-reperfusion creates permissive conditions for autoantibodies to interact with their antigenic targets and leads to enhanced renal damage and dysfunction. In 172 kidney transplant recipients, we found that pretransplant anti-LG3 antibodies were associated with an increased risk of delayed graft function (DGF). Pretransplant anti-LG3 antibodies are inversely associated with graft function at 1 year after transplantation in patients who experienced DGF, independent of rejection. Pretransplant anti-AT1R and antivimentin were not associated with DGF or its functional outcome. In a model of renal IRI in mice, passive transfer of anti-LG3 IgG led to enhanced dysfunction and microvascular injury compared with passive transfer with control IgG. Passive transfer of anti-LG3 antibodies also favored intrarenal microvascular complement activation, microvascular rarefaction and fibrosis after IRI. Our results suggest that anti-LG3 antibodies are novel aggravating factors for renal IRI. These results provide novel insights into the pathways that modulate the severity of renal injury at the time of transplantation and their impact on long-term outcomes.


Assuntos
Autoanticorpos/sangue , Função Retardada do Enxerto/etiologia , Sobrevivência de Enxerto/imunologia , Proteoglicanas de Heparan Sulfato/imunologia , Transplante de Rim/efeitos adversos , Traumatismo por Reperfusão/etiologia , Animais , Autoanticorpos/imunologia , Função Retardada do Enxerto/sangue , Função Retardada do Enxerto/patologia , Feminino , Seguimentos , Taxa de Filtração Glomerular , Humanos , Falência Renal Crônica/cirurgia , Testes de Função Renal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Prognóstico , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/patologia , Estudos Retrospectivos , Fatores de Risco
2.
Am J Transplant ; 15(5): 1205-18, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25808553

RESUMO

Transplant vasculopathy is associated with neointimal accumulation of recipient-derived mesenchymal stem cells. Increased circulating levels of LG3, a C-terminal fragment of perlecan, were found in renal transplant patients with vascular rejection. Here, we evaluated whether LG3 regulates the migration and homing of mesenchymal stem cells and the accumulation of recipient-derived neointimal cells. Mice were transplanted with a fully-MHC mismatched aortic graft followed by intravenous injection of recombinant LG3. LG3 injections increased neointimal accumulation of α-smooth muscle actin positive cells. When green fluorescent protein (GFP)-transgenic mice were used as recipients, LG3 injection favored accumulation of GFP+ cells to sites of neointima formation. LG3 increased horizontal migration and transmigration of mouse and human MSC in vitro and led to increased ERK1/2 phosphorylation. Neutralizing ß1 integrin antibodies or use of mesenchymal stem cells from α2 integrin-/- mice decreased migration in response to recombinant LG3. Reduced intima-media ratios and decreased numbers of neointimal cells showing ERK1/2 phosphorylation were found in α2-/- recipients injected with recombinant LG3. Collectively, our results suggest that LG3, through interactions with α2ß1 integrins on recipient-derived cells leading to activation of ERK1/2 and increased migration, favors myointimal thickening.


Assuntos
Rejeição de Enxerto/patologia , Proteoglicanas de Heparan Sulfato/química , Integrina alfa2beta1/metabolismo , Células-Tronco Mesenquimais/citologia , Neointima/patologia , Enxerto Vascular , Animais , Aorta/patologia , Aorta/transplante , Prótese Vascular , Espessura Intima-Media Carotídea , Movimento Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Integrina beta1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Miócitos de Músculo Liso/citologia , Fenótipo , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/metabolismo
3.
Am J Transplant ; 13(4): 861-874, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23432943

RESUMO

Acute vascular rejection (AVR) is characterized by immune-mediated vascular injury and heightened endothelial cell (EC) apoptosis. We reported previously that apoptotic ECs release a bioactive C-terminal fragment of perlecan referred to as LG3. Here, we tested the possibility that LG3 behaves as a neoantigen, fuelling the production of anti-LG3 antibodies of potential importance in regulating allograft vascular injury. We performed a case-control study in which we compared anti-LG3 IgG titers in kidney transplant recipients with AVR (n=15) versus those with acute tubulo-interstitial rejection (ATIR) (n=15) or stable graft function (n=30). Patients who experienced AVR had elevated anti-LG3 titers pre and posttransplantation compared to subjects with ATIR or stable graft function (p<0.05 for both mediators). Elevated pretransplant anti-LG3 titers (OR: 4.62, 95% CI: 1.08-19.72) and pretransplant donor-specific antibodies (DSA) (OR 4.79, 95% CI: 1.03-22.19) were both independently associated with AVR. To address the functional role of anti-LG3 antibodies in AVR, we turned to passive transfer of anti-LG3 antibodies in an animal model of vascular rejection based on orthotopic aortic transplantation between fully MHC-mismatched mice. Neointima formation, C4d deposition and allograft inflammation were significantly increased in recipients of an ischemic aortic allograft passively transferred with anti-LG3 antibodies. Collectively, these data identify anti-LG3 antibodies as novel accelerators of immune-mediated vascular injury and obliterative remodeling.


Assuntos
Rejeição de Enxerto/imunologia , Proteoglicanas de Heparan Sulfato/imunologia , Imunoglobulina G/sangue , Doenças Vasculares/imunologia , Adulto , Animais , Antígenos/imunologia , Aorta/patologia , Apoptose , Estudos de Casos e Controles , Células Endoteliais/patologia , Feminino , Rejeição de Enxerto/sangue , Humanos , Imunização Passiva , Imunoglobulina G/imunologia , Inflamação/patologia , Rim/irrigação sanguínea , Rim/patologia , Transplante de Rim/efeitos adversos , Transplante de Rim/métodos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteínas Recombinantes/imunologia , Estudos Retrospectivos , Doenças Vasculares/sangue
4.
Am J Transplant ; 12(6): 1378-84, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22420581

RESUMO

Transplanted organs have to cope with diverse immunologic and metabolic stressors that augment the percentage of stressed and dying cells. Cell death, whether apoptotic or necrotic, is crucial in various transplantation-associated conditions. Necrosis, a proinflammatory type of cell death classically considered as accidental, is increasingly recognized as a highly controlled death program. Apoptosis, the classical programmed cell death mode program, is tightly orchestrated and culminates in the activation of caspases. Apoptosis was classically regarded as a silent form of cell death, but mounting evidence indicates that apoptotic cells "don't go silently" and leave a heritage to the local microenvironment. This apoptotic legacy, embedded within the effector phase of apoptosis, is aimed, at least in part, at controlling leukocyte trafficking and fostering tissue remodeling at sites of apoptotic cell deletion and can promote maladaptive remodeling pathways of importance for obliterative vascular remodeling. Moreover, apoptotic cells can transfer bioactive molecules by the release of apoptotic membrane vesicles that, in turn, shapes the phenotype and functions of immune cells. In this review, we summarize recent data highlighting the importance of apoptosis-associated intercellular communication networks in the regulation of allograft remodeling and immune responses in transplantation.


Assuntos
Apoptose , Transplante , Humanos , Proteólise
5.
Lupus ; 19(4): 347-53, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20353968

RESUMO

The antiphospholipid syndrome (APS), as both a primary syndrome and a syndrome in association with systemic lupus erythematosus (SLE), can be a devastating disease. It is unclear what factors (genetic and/or environmental) lead to the generation of antiphospholipid antibodies (aPL). It is equally unclear why only certain individuals with aPL develop clinical events. We hypothesize that innate immune activation plays a critical role at two distinct stages of APS, namely, the initiation phase, in which aPL first appear, and the effector phase, in which aPL precipitate a thrombotic event. According to the model we propose, aPL alone are insufficient to cause thrombosis and a concomitant trigger of innate immunity, e.g. a toll-like receptor (TLR) ligand, must be present for thrombosis to occur. Here, we discuss our findings that mice immunized with beta(2)-glycoprotein I (beta(2)GPI) and lipopolysaccharide (LPS), a TLR ligand, produce high levels of aPL and other SLE-associated autoantibodies, and develop lupus-like glomerulonephritis. We also discuss our data showing that autoantibodies to heat shock protein 60 (HSP60), an 'endogenous TLR ligand', promote thrombus generation in a murine model of arterial injury. Thus, both pathogen-derived TLR ligands (e.g. LPS) and endogenous TLR ligands (e.g. HSP60) may contribute to the pathogenesis of APS. This putative dual role of innate immunity provides new insight into the generation of aPL as well as the enigma of why some individuals with aPL develop APS, while others do not.


Assuntos
Anticorpos Antifosfolipídeos/imunologia , Síndrome Antifosfolipídica/imunologia , Imunidade Inata , Animais , Síndrome Antifosfolipídica/complicações , Modelos Animais de Doenças , Humanos , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Trombose/etiologia , Trombose/imunologia , Receptores Toll-Like/metabolismo
6.
J Thromb Haemost ; 7(4): 710-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19192108

RESUMO

BACKGROUND AND OBJECTIVES: Anti-heat shock protein (HSP)60 autoantibodies are associated with atherosclerosis and are known to affect endothelial cells in vitro. However, their role in thrombus formation remains unclear. We hypothesized that anti-HSP60 autoantibodies could potentiate thrombosis, and evaluated the effect of anti-murine HSP60 antibodies in a ferric chloride (FeCl3)-induced murine model of carotid artery injury. METHODS: Anti-HSP60, or control, IgG was administered to BALB/c mice 48 h prior to inducing carotid artery injury, and blood flow was monitored using an ultrasound probe. RESULTS: Thrombus formation was more rapid and stable in anti-HSP60 IGG-treated mice than in controls (blood flow=1.7%+/-0.6% vs. 34%+/-12.6%, P=0.0157). Occlusion was complete in all anti-HSP60 IgG-treated mice (13/13), with no reperfusion being observed. In contrast, 64% (9/14) of control mice had complete occlusion, with reperfusion occurring in 6/9 mice. Thrombi were significantly larger in anti-HSP60 IgG-treated mice (P=0.0001), and contained four-fold more inflammatory cells (P=0.0281) than in controls. Non-injured contralateral arteries of anti-HSP60 IgG-treated mice were also affected, exhibiting abnormal endothelial cell morphology and significantly greater von Willebrand factor (VWF) and P-selectin expression than control mice (P=0.0024 and P=0.001, respectively). CONCLUSIONS: In summary, the presence of circulating anti-HSP60 autoantibodies resulted in increased P-selectin and VWF expression and altered cell morphology in endothelial cells lining uninjured carotid arteries, and promoted thrombosis and inflammatory cell recruitment in FeCl3-injured carotid arteries. These findings suggest that anti-HSP60 autoantibodies may constitute an important prothrombotic risk factor in cardiovascular disease in human vascular disease.


Assuntos
Autoanticorpos/farmacologia , Chaperonina 60/imunologia , Trombose/imunologia , Animais , Arteriopatias Oclusivas/etiologia , Arteriopatias Oclusivas/imunologia , Autoanticorpos/administração & dosagem , Doenças das Artérias Carótidas/etiologia , Doenças das Artérias Carótidas/imunologia , Cloretos , Modelos Animais de Doenças , Compostos Férricos , Camundongos , Selectina-P/análise , Fluxo Sanguíneo Regional , Reperfusão , Trombose/etiologia , Fator de von Willebrand/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA