Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Transplantation ; 96(7): 601-8, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23903009

RESUMEN

BACKGROUND: Alloantibody can contribute significantly to rejection of heart transplants by activation of complement and interactions with a variety of effector cells, including macrophages and monocytes through activating FcγRI, FcγRIII, FcγRIV, the inhibitory FcγRIIB and complement receptors. These receptors link cellular and humoral immunity by bridging the antibody specificity to effector cells. Activating FcγRs are also involved in serum amyloid P component (SAP)-mediated clearance of apoptotic bodies. METHODS: B10.A (H-2a) hearts were transplanted into wild-type (WT) or FcγRIII-knockout (KO) C57BL/6 (H-2b) mouse recipients. Levels of alloantibodies and SAP in the circulation were determined by flow cytometry and enzyme-linked immunosorbent assay, respectively. Intragraft cytokine mRNA expression was measured by real-time polymerase chain reaction. Intragraft deposition of C4d, von Willebrand factor, SAP, and activated caspase 3 was visualized by immunochemistry. RESULTS: B10.A hearts in C57BL/6 FcγRIII-KO recipients were rejected acutely within 6 to 8 days compared with 10 to 14 days in WT. The rejection in FcγRIII-KO was accompanied by higher levels of circulating IgM/IgG alloantibodies and SAP than in WT recipients. Histology in FcγRIII-KO cardiac allograft recipients indicated perivascular margination of monocytes and neutrophils, vascular endothelial cell injury, and intense vasculocentric infiltrates with extensive apoptosis. Higher numbers of apoptotic cells, stronger C4d and SAP deposition, and extensive activated caspase 3 were found in areas of dense pockets of apoptotic blebs in FcγRIII-KO. CONCLUSIONS: We propose that absence of FcγRIII is associated with the lack of efficient SAP-mediated clearance of apoptotic cells through FcγRs. Apoptotic cells become immunogenic and induce enhanced inflammation, alloantibody production, and complement activation leading to accelerated cardiac allograft rejection.


Asunto(s)
Rechazo de Injerto/inmunología , Trasplante de Corazón/efectos adversos , Mediadores de Inflamación/metabolismo , Inflamación/inmunología , Miocardio/inmunología , Receptores de IgG/deficiencia , Animales , Apoptosis , Activación de Complemento , Citocinas/metabolismo , Rechazo de Injerto/sangre , Rechazo de Injerto/patología , Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Inflamación/sangre , Inflamación/patología , Inflamación/prevención & control , Mediadores de Inflamación/sangre , Isoanticuerpos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Receptores de IgG/genética , Componente Amiloide P Sérico/metabolismo , Factores de Tiempo
4.
Transplantation ; 90(12): 1307-11, 2010 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-21048532

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs) are multipotent cells with immunoregulatory capacity that are present in most adult organs. We previously demonstrated that co-culture of C57BL/6 kidney-derived MSCs (KSCs) in syngeneic bone marrow-derived dendritic cell (DC) culture induced a DC phenotype (KSC-DC) with reduced major histocompatibility complex (MHC) class II/increased CD80 expression and ability to suppress T-cell responses. METHODS: To study their effects on allogeneic DCs, C57BL/6 KSCs were added to incipient BALB/c DC culture, with surface expression of MHC class II/CD80 measured by fluorescence-activated cell sorting. The ability to stimulate T-cell responses was then assessed in an allogeneic mixed leukocyte response. Next, we isolated either BALB/c (donor) or C57BL/6 (recipient) KSC-DCs from co-culture and measured the tempo of rejection after cotransplantation with islet grafts in a mouse model of islet transplantation. Finally, we measured the effects of KSC-DC stimulation on B-cell proliferation and IgM/IgG production in allogeneic cultures. RESULTS: C57BL/6 KSCs induced a BALB/c DC phenotype with significantly decreased MHC class II, increased CD80 expression, and decreased T-cell stimulatory capacity in the mixed leukocyte response (P<0.01 vs. control). Cotransplantation of donor (BALB/c) but not recipient (C57BL/6) KSC-DCs resulted in significant delay of rejection after islet transplantation (P<0.01 vs. control). Finally, stimulation by KSC-DCs resulted in significantly reduced B-cell proliferation and antibody production in allogeneic culture (P<0.01 vs. control). CONCLUSIONS: Our results highlight an important mechanism of MSC-based immunotherapy and its potential for use in clinical transplantation as prevention of rejection and possibly sensitization.


Asunto(s)
Rechazo de Injerto/inmunología , Células Madre Mesenquimatosas/inmunología , Trasplante Isogénico/inmunología , Animales , Formación de Anticuerpos , Linfocitos B/inmunología , Antígeno B7-1/genética , Antígeno B7-1/inmunología , Células de la Médula Ósea/inmunología , Técnicas de Cultivo de Célula , Células Dendríticas/citología , Células Dendríticas/inmunología , Citometría de Flujo/métodos , Rechazo de Injerto/prevención & control , Antígenos de Histocompatibilidad Clase II/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Activación de Linfocitos , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante Homólogo/inmunología
5.
Immunol Res ; 47(1-3): 25-44, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20135240

RESUMEN

Antibody-mediated rejection has become critical clinically because this form of rejection is usually unresponsive to conventional anti-rejection therapy, and therefore, it has been recognized as a major cause of allograft loss. Our group developed experimental animal models of vascularized organ transplantation to study pathogenesis of antibody- and complement-mediated endothelial cell injury leading to graft rejection. In this review, we discuss mechanisms of antibody-mediated graft rejection resulting from activation of complement by C1q- and MBL (mannose-binding lectin)-dependent pathways and interactions with a variety of effector cells, including macrophages and monocytes through Fcgamma receptors and complement receptors.


Asunto(s)
Activación de Complemento/inmunología , Rechazo de Injerto/patología , Isoanticuerpos/inmunología , Animales , Modelos Animales de Enfermedad , Células Endoteliales/patología , Rechazo de Injerto/inmunología , Trasplante de Corazón/inmunología , Humanos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología
6.
J Mol Med (Berl) ; 87(9): 859-64, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19562316

RESUMEN

Kidney ischemia-reperfusion injury (IRI) engages both the innate and adaptive immune responses. Cellular mediators of immunity, such as dendritic cells, neutrophils, macrophages, natural killer T, T, and B cells, contribute to the pathogenesis of renal injury after IRI. Postischemic kidneys express increased levels of adhesion molecules on endothelial cells and toll-like receptors on tubular epithelial cells. Soluble components of the immune system, such as complement activation proteins and cytokines, also participate in injury/repair of postischemic kidneys. Experimental studies on the immune response in kidney IRI have resulted in better understanding of the mechanisms underlying IRI and led to the discovery of novel therapeutic and diagnostic targets.


Asunto(s)
Enfermedades Renales/inmunología , Daño por Reperfusión/inmunología , Humanos , Fenómenos del Sistema Inmunológico , Inmunidad , Enfermedades Renales/patología
7.
Transplantation ; 84(10): 1324-34, 2007 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-18049118

RESUMEN

BACKGROUND: Antibody-mediated rejection is characterized by macrophage margination against vascular endothelium. The potential interactions triggered by antibodies between endothelial cells (EC) and macrophages have not been examined thoroughly in transplants. We used in vivo and in vitro models of antibody-mediated rejection. METHODS: Passive transfer of monoclonal alloantibodies (Allo-mAbs) to donor major histocompatibility complex-class I antigens was used to restore acute rejection of B10.A (H-2a) hearts to C57BL/6 (H-2b) immunoglobulin knockout (IgKO) recipients. Intragraft cytokine mRNA expression was measured by real-time polymerase chain reaction. In vitro, mouse EC were cultured in the presence of Allo-mAbs to donor major histocompatibility complex class I antigens and mononuclear cells. Levels of cytokines in culture supernatants were determined in enzyme-linked immunosorbent assay. RESULTS: Expression of MCP-1, IL-6 and IL-1alpha mRNA was higher in rejecting transplants from recipients treated with Allo-mAbs compared to non-rejecting transplants. EC sensitized with Allo-mAbs produced high levels of MCP-1 and KC. The addition of macrophages to sensitized EC stimulated high levels of IL-6 in addition to MCP-1, KC, Rantes, and TIMP-1. The levels of MCP-1 and IL-6 were significantly lower in co-cultures of EC sensitized with IgG1 Allo-mAbs in the presence of mononuclear cells from Fcgamma-Receptor III KO (FcgammaRIII-KO) graft recipients compared to co-cultures with wild-type cells. The levels of both cytokines were also lower in co-cultures of EC stimulated with F(ab')2 fragments of antibody. CONCLUSIONS: Our findings indicate that IgG1 Allo-mAbs to major histocompatibility complex class I antigens can augment graft injury by stimulating EC to produce MCP-1 and by activating mononuclear cells through their Fc receptors.


Asunto(s)
Rechazo de Injerto/inmunología , Trasplante de Corazón/inmunología , Inmunoglobulinas/deficiencia , Receptores Fc/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular Transformada , Inmunoglobulina G/inmunología , Isoanticuerpos/inmunología , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Bazo/inmunología
8.
Cell Immunol ; 248(1): 18-30, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17950717

RESUMEN

In transplantation, activation of complement has largely been equated to antibody-mediated rejection, but complement is also important in recognition of apoptotic and necrotic cells as well as in modifying antigen presentation to T cells and B cells. As a part of the innate immune system, complement is one of the first responses to injury, and it can determine the direction and magnitude of the subsequent responses. Consequently, the effects of complement in allorecognition and graft rejection are increased when organs are procured from cadaver donors because these organs sustain a series of stresses from brain death, prolonged life support, ischemia and finally reperfusion that initiate proinflammatory processes and tissue injury. In addition, these organs are transplanted to patients, who frequently have been sensitized to histocompatibility antigens as the result of transfusions, pregnancies or transplants. Complement activation generates a series of biologically active effector molecules that can modulate graft rejection by directly binding to the graft or by modifying the response of macrophages, T and B cells of the recipient. However, complement is regulated and the process of regulation produces split products that can decrease as well as increase immune responses. Small animal models have been developed to test these variables. The guide for evaluating results from these models remains clinical findings because there are significant differences between the rodent and human complement systems.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento , Rechazo de Injerto/inmunología , Embarazo/inmunología , Trasplante Homólogo/inmunología , Animales , Presentación de Antígeno/inmunología , Linfocitos B/inmunología , Cadáver , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Femenino , Trasplante de Corazón , Humanos , Isoantígenos/inmunología , Modelos Animales , Daño por Reperfusión/inmunología , Linfocitos T/inmunología , Reacción a la Transfusión , Trasplante Heterólogo/inmunología
9.
Proc Natl Acad Sci U S A ; 104(4): 1301-6, 2007 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-17229850

RESUMEN

Although antibodies to HLA play a role in the pathogenesis of diseases processes such as rejection of transplanted organs, the precise mechanisms by which antibodies cause tissue injury are not completely understood. We hypothesized that antibodies to host tissues cause inflammation in part by activating endothelial exocytosis of granules that contain prothrombotic mediators such as von Willebrand Factor (VWF) and proinflammatory mediators such as P-selectin. To test this hypothesis, we treated human endothelial cells with murine monoclonal antibody W6/32 to HLA class I and then measured exocytosis by the release of VWF and the externalization of P-selectin. Antibody to HLA activates endothelial exocytosis in a dose-dependent manner over time. The biologically active complement split product, C5a, adds a slight but significant increase to antibody induction of exocytosis. Antibody to HLA alone or with C5a did not damage the cells. Cross-linking of HLA appears to play a role in the ability of antibody to activate exocytosis, because the W6/32 monovalent Fab fragment did not activate VWF release, but the bivalent Fab'2 was effective in triggering exocytosis. To explore the in vivo effects of antibody upon graft injury, we infused W6/32 Fab'2 antibody to human HLA into severe combined immunodeficient/beige mice that had been transplanted with human skin grafts. Antibody to HLA activated exocytosis and inflammation in human skin grafts. Our data show that antibody to host antigens can activate human endothelial cell exocytosis and leukocyte trafficking. By triggering vascular inflammation, antibody activation of exocytosis may play a role in transplant rejection.


Asunto(s)
Autoanticuerpos/inmunología , Endotelio Vascular/citología , Exocitosis/inmunología , Antígenos HLA/inmunología , Células Cultivadas , Endotelio Vascular/inmunología , Humanos , Cuerpos de Weibel-Palade/inmunología
10.
Transplantation ; 82(7): 857-64, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17038898

RESUMEN

BACKGROUND: Many patients on the waiting list for transplants are sensitized from previous blood transfusions, pregnancy, or transplants. We investigated the role of complement in acute and chronic pathology in hearts transplanted to sensitized rats. METHODS: Blood was transfused from allogeneic PVG.R8 rats or control isogeneic PVG.1U rats to C6-sufficient and -deficient PVG.1U rats. Three weeks later hearts were transplanted from PVG.R8 donors and low-dose cyclosporin A was initiated. RESULTS: Allogeneic but not isogeneic blood transfusion elicited strong immunoglobulin (Ig) M, IgG1 and IgG2b alloantibody responses. Sensitization caused accelerated acute rejection of cardiac allografts by C6-sufficient recipients (4 days). In contrast, allografts functioned over 40 days in all C6-deficient recipients, but sensitization caused increased interstitial fibrosis and chronic vasculopathy. Circulating alloantibodies were associated with deposits of C4d on the vascular endothelium together with pericapillary accumulation of neutrophils and macrophages in the grafts. In contrast, T cells accumulated in periarterial lymphatics that did not have C4d deposits. CONCLUSIONS: Presensitization by allogeneic blood transfusion causes accelerated acute graft rejection in the presence of the complete complement cascade. In the absence of C6, macrophages colocalized with deposits of C4d and T cells accumulated in the periarterial lymphatics.


Asunto(s)
Formación de Anticuerpos , Complemento C6/deficiencia , Complemento C6/fisiología , Trasplante de Corazón/inmunología , Isoanticuerpos/sangre , Reacción a la Transfusión , Animales , Eliminación de Gen , Supervivencia de Injerto , Inmunoglobulina G/sangre , Inmunoglobulina G/clasificación , Inmunoglobulina M/sangre , Macrófagos/inmunología , Complejo Mayor de Histocompatibilidad , Modelos Animales , Ratas , Ratas Endogámicas , Trasplante Homólogo , Trasplante Isogénico
11.
Am J Transplant ; 5(4 Pt 1): 694-701, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15760392

RESUMEN

Chronic pathological changes in transplanted lungs are unique because they center on the airways. We examined the relative role of MHC class I and II antigens in causing bronchial pathology in orthotopic lung transplants to rats maintained on cyclosporin A (CsA). Transplants mismatched for MHC class II antigens had significantly more peri-bronchiolar infiltrates than MHC class I incompatible transplants. No significant increase in infiltrates was found in lung transplants incompatible for MHC class I plus II antigens compared to MHC class II antigens alone. Immunohistochemistry demonstrated that MHC class II antigen expression was confined to macrophages in MHC class I incompatible transplants, but was upregulated on bronchial epithelium in transplants with MHC class II incompatibilities. Vascular endothelium was notably devoid of MHC class II antigen expression in all transplants. However, both peri-bronchial and peri-vascular infiltrates were frequently cuffed by alveolar macrophages and type II pneumocytes that expressed MHC class II antigens. PCR analysis demonstrated that IFN-gamma and regulated on activation, normal T cells expressed and secreted (RANTES) were upregulated in MHC class II incompatible transplants. Thus, MHC class II incompatible orthotopic lung transplants in rats maintained on CsA immunosuppression undergo a bronchiolcentric upregulation of alloantigens.


Asunto(s)
Bronquios/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Histocompatibilidad/inmunología , Leucocitos Mononucleares/inmunología , Trasplante de Pulmón , Animales , Animales Congénicos , Bronquios/patología , Quimiocina CCL5/metabolismo , Femenino , Interferón gamma/metabolismo , Pulmón/metabolismo , Masculino , Ratas , Trasplante Homólogo
12.
Am J Transplant ; 4(3): 311-8, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14961982

RESUMEN

Complement is a multifunctional system of receptors and regulators as well as effector molecules. Both the pathogenic and diagnostic power of complement is based on the capacity of the complement system to amplify innate and adaptive immunity. This amplification is accomplished through two strategies: (1) enzymatic reactions in the complement cascade, and (2) stimulation of leukocytes, platelets and parenchymal cells through specific receptors or receptor-independent pore formation. The mechanisms by which complement mediates and modifies nonspecific inflammation, antibody-mediated injury and T-cell responses are of particular significance to the pathogenesis of transplant rejection. Understanding the mechanisms by which complement integrates the interactions of leukocytes, platelets and parenchymal cells offers opportunities to further refine the diagnosis of rejection.


Asunto(s)
Anticuerpos/inmunología , Proteínas del Sistema Complemento , Rechazo de Injerto/diagnóstico , Animales , Complemento C1/inmunología , Complemento C4/inmunología , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Humanos , Mediadores de Inflamación/inmunología , Ratones
13.
Am J Transplant ; 4(3): 326-34, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14961984

RESUMEN

Alloantibodies (AlloAbs) are a clinically significant component of the immune response to organ transplants. In our experimental model, B10.A (H-2a) cardiac transplants survived significantly longer in C57BL/6 (H-2b) immunoglobulin knock-out (IgKO) recipients than in their wild-type (WT) counterparts. Passive transfer of a single 50-200-microg dose of complement-activating IgG2b AlloAbs to IgKO recipients reconstituted acute rejection of cardiac allografts. Although passive transfer of a subthreshold dose of 25 microg of IgG2b or a single 100-200-microg dose of non-complement-activating IgG1 AlloAbs did not restore acute rejection to IgKO recipients, a combination of these AlloAbs did cause acute graft rejection. Histologically, rejection was accompanied by augmented release of von Willebrand factor from endothelial cells. IgG1 AlloAbs did not activate complement on their own and did not augment complement activation by IgG2b AlloAbs. However, IgG1 AlloAbs stimulated cultured mouse endothelial cells to produce monocyte chemotactic protein 1 (MCP-1) and neutrophil chemoattractant growth-related oncogene alpha (KC). TNF-alpha augmented IgG1 induced secretion of MCP-1 and KC. These findings indicate that non-complement-activating AlloAbs can augment injury to allografts by complement-activating AlloAbs. Non-complement-activating AlloAbs stimulate endothelial cells to produce chemokines and this effect is augmented in the milieu of proinflammatory cytokines.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Rechazo de Injerto/inmunología , Trasplante de Corazón/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Sinergismo Farmacológico , Rechazo de Injerto/inducido químicamente , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Isoanticuerpos/inmunología , Ratones
14.
J Immunol ; 169(8): 4620-7, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12370401

RESUMEN

The lung is known to be particularly susceptible to complement-mediated injury. Both C5a and the membrane attack complex (MAC), which is formed by the terminal components of complement (C5b-C9), can cause acute pulmonary distress in nontransplanted lungs. We used C6-deficient rats to investigate whether MAC causes injury to lung allografts. PVG.R8 lungs were transplanted orthotopically to MHC class I-incompatible PVG.1U recipients. Allografts from C6-sufficient (C6(+)) donors to C6(+) recipients were rejected with an intense vascular infiltration and diffuse alveolar hemorrhage 7 days after transplantation (n = 5). Ab and complement (C3d) deposition was accompanied by extensive vascular endothelial injury and intravascular release of von Willebrand factor. In contrast, lung allografts from C6-deficient (C6(-)) donors to C6(-) recipients survived 13-17 days (n = 5). In the absence of C6, perivascular mononuclear infiltrates of ED1(+) macrophages and CD8(+) T lymphocytes were present 7 days after transplantation, but vascular endothelial cells were quiescent, with minimal von Willebrand factor release and no evidence of alveolar hemorrhage or edema. Lung allografts were performed from C6(-) donors to C6(+) recipients (n = 5) and from C6(+) donors to C6(-) recipients (n = 5) to separate the effects of systemic and local C6 production. Lungs transplanted from C6(+) donors to C6(-) recipients had increased alveolar macrophages and capillary injury. C6 production by lung allografts was demonstrated at the mRNA and protein levels. These results demonstrate that MAC causes vascular injury in lung allografts and that the location of injury is dependent on the source of C6.


Asunto(s)
Complemento C6/fisiología , Complejo de Ataque a Membrana del Sistema Complemento/fisiología , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Trasplante de Pulmón/inmunología , Trasplante de Pulmón/patología , Enfermedad Aguda , Animales , Animales Congénicos , Movimiento Celular/genética , Movimiento Celular/inmunología , Complemento C3d/metabolismo , Complemento C6/biosíntesis , Complemento C6/deficiencia , Complemento C6/genética , Complejo de Ataque a Membrana del Sistema Complemento/deficiencia , Complejo de Ataque a Membrana del Sistema Complemento/genética , Femenino , Rechazo de Injerto/genética , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Antígenos de Histocompatibilidad/genética , Antígenos de Histocompatibilidad Clase I/genética , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Isoanticuerpos/biosíntesis , Masculino , Hemorragia Posoperatoria/genética , Hemorragia Posoperatoria/inmunología , Hemorragia Posoperatoria/patología , Hemorragia Posoperatoria/prevención & control , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/patología , Ratas , Ratas Endogámicas
15.
J Immunol ; 169(3): 1270-6, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12133948

RESUMEN

Although the CD154-CD40 T cell costimulation pathway has been shown to mediate alloimmune responses in normal recipients, little is known about its role in sensitized hosts. In this work, by using novel models of cardiac allograft rejection in skin-sensitized CD154- and CD40-deficient mice, we reaffirm the key role of CD154-CD40 signaling in host sensitization to alloantigen in vivo. First, we identified CD8(+) T cells as principal effectors in executing accelerated rejection in our model. Disruption of CD154-CD40 signaling in recipients at the T cell side (CD154-deficient) but not at the APC side (CD40-deficient) abrogated accelerated (<2 days) rejection and resulted in long-term (>100 days) graft survival. This suggests that the CD154-dependent mechanism in host CD8(+) T cell sensitization operates via the direct Ag presentation. Then, in comparative studies of alloimmune responses in CD154-deficient and wild-type recipients, we showed that, although alloreactive B cell responses were inhibited, alloreactive T cell responses were down-regulated selectively in the CD8(+) T cell compartment, leaving CD4(+) T cells largely unaffected. This unique alteration in host alloreactivity, seen not only in peripheral lymphocytes but also in allograft infiltrate, may represent the key mechanism by which disruption of CD154-CD40 signaling prevents sensitization to alloantigen in vivo and leads to long-term allograft survival.


Asunto(s)
Presentación de Antígeno , Antígenos CD40/fisiología , Ligando de CD40/fisiología , Linfocitos T CD8-positivos/inmunología , Trasplante de Piel/inmunología , Animales , Diferenciación Celular , Supervivencia de Injerto , Inmunoglobulina G/biosíntesis , Inmunoglobulina M/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA