Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
Am J Transplant ; 17(11): 2810-2819, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28444847

RESUMEN

Building on studies showing that ischemia-reperfusion-(I/R)-injury is complement dependent, we tested links among complement activation, transplantation-associated I/R injury, and murine cardiac allograft rejection. We transplanted BALB/c hearts subjected to 8-h cold ischemic storage (CIS) into cytotoxic T-lymphocyte associated protein 4 (CTLA4)Ig-treated wild-type (WT) or c3-/- B6 recipients. Whereas allografts subjected to 8-h CIS rejected in WT recipients with a median survival time (MST) of 37 days, identically treated hearts survived >60 days in c3-/- mice (p < 0.05, n = 4-6/group). Mechanistic studies showed recipient C3 deficiency prevented induction of intragraft and serum chemokines/cytokines and blunted the priming, expansion, and graft infiltration of interferon-γ-producing, donor-reactive T cells. MST of hearts subjected to 8-h CIS was >60 days in mannose binding lectin (mbl1-/- mbl2-/- ) recipients and 42 days in factor B (cfb-/- ) recipients (n = 4-6/group, p < 0.05, mbl1-/- mbl2-/- vs. cfb-/- ), implicating the MBL (not alternative) pathway. To pharmacologically target MBL-initiated complement activation, we transplanted BALB/c hearts subjected to 8-h CIS into CTLA4Ig-treated WT B6 recipients with or without C1 inhibitor (C1-INH). Remarkably, peritransplantation administration of C1-INH prolonged graft survival (MST >60 days, p < 0.05 vs. controls, n = 6) and prevented CI-induced increases in donor-reactive, IFNγ-producing spleen cells (p < 0.05). These new findings link donor I/R injury to T cell-mediated rejection through MBL-initiated, complement activation and support testing C1-INH administration to prevent CTLA4Ig-resistant rejection of deceased donor allografts in human transplant patients.


Asunto(s)
Abatacept/farmacología , Antígeno CTLA-4/inmunología , Proteínas del Sistema Complemento/inmunología , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/inmunología , Trasplante de Corazón/efectos adversos , Linfocitos T/inmunología , Aloinjertos , Animales , Rechazo de Injerto/etiología , Supervivencia de Injerto/efectos de los fármacos , Inmunosupresores/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Donantes de Tejidos
3.
Mucosal Immunol ; 9(1): 137-45, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26013006

RESUMEN

Foxp3-expressing regulatory T cells (Tregs) are central regulators of immune homeostasis and tolerance. As it has been suggested that proper Treg function is compromised under inflammatory conditions, seeking for a pathway that enhances or stabilizes Treg function is a subject of considerable interest. We report that interleukin (IL)-27, an IL-12 family cytokine known to have both pro- and anti-inflammatory roles in T cells, plays a pivotal role in enhancing Treg function to control T cell-induced colitis, a model for inflammatory bowel disease (IBD) in humans. Unlike wild-type (WT) Tregs capable of inhibiting colitogenic T-cell expansion and inflammatory cytokine expression, IL-27R-deficient Tregs were unable to downregulate inflammatory T-cell responses. Tregs stimulated with IL-27 expressed substantially improved suppressive function in vitro and in vivo. IL-27 stimulation of Tregs induced expression of Lag3, a surface molecule implicated in negatively regulating immune responses. Lag3 expression in Tregs was critical to mediate Treg function in suppressing colitogenic responses. Human Tregs also displayed enhanced suppressive function and Lag3 expression following IL-27 stimulation. Collectively, these results highlight a novel function for the IL-27/Lag3 axis in modulating Treg regulation of inflammatory responses in the intestine.


Asunto(s)
Antígenos CD/inmunología , Colitis/inmunología , Factores de Transcripción Forkhead/inmunología , Interleucinas/inmunología , Receptores de Interleucina/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD/genética , Proliferación Celular , Colitis/genética , Colitis/patología , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Humanos , Interleucinas/genética , Interleucinas/farmacología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , Receptores de Interleucina/deficiencia , Receptores de Interleucina/genética , Transducción de Señal , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/patología , Proteína del Gen 3 de Activación de Linfocitos
4.
Am J Transplant ; 15(5): 1192-204, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25731734

RESUMEN

We have reported that B6.CCR5(-/-) mice reject renal allografts with high serum donor-specific antibody (DSA) titers and marked C4d deposition in grafts, features consistent with antibody-mediated rejection (AMR). B6.huCD20/CCR5(-/-) mice, where human CD20 expression is restricted to B cells, rejected A/J renal allografts by day 26 posttransplant with DSA first detected in serum on day 5 posttransplant and increased thereafter. Recipient treatment with anti-huCD20 mAb prior to the transplant and weekly up to 7 weeks posttransplant promoted long-term allograft survival (>100 days) with low DSA titers. To investigate the effect of B cell depletion at the time serum DSA was first detected, recipients were treated with anti-huCD20 mAb on days 5, 8, and 12 posttransplant. This regimen significantly reduced DSA titers and graft inflammation on day 15 posttransplant and prolonged allograft survival >60 days. However, DSA returned to the titers observed in control treated recipients by day 30 posttransplant and histological analyses on day 60 posttransplant indicated severe interstitial fibrosis. These results indicate that anti-huCD20 mAb had the greatest effect as a prophylactic treatment and that the distinct kinetics of DSA responses accounts for acute renal allograft failure versus the development of fibrosis.


Asunto(s)
Anticuerpos/inmunología , Antígenos CD20/química , Rechazo de Injerto/prevención & control , Trasplante de Riñón , Insuficiencia Renal/inmunología , Insuficiencia Renal/cirugía , Aloinjertos , Animales , Formación de Anticuerpos/inmunología , Creatinina/sangre , Modelos Animales de Enfermedad , Fibrosis/fisiopatología , Citometría de Flujo , Rechazo de Injerto/inmunología , Supervivencia de Injerto , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Receptores CCR5/genética , Factores de Tiempo , Trasplante Homólogo
5.
Am J Transplant ; 15(2): 333-45, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25582188

RESUMEN

Acute and chronic rejection impact distinct compartments of cardiac allografts. Intramyocardial mononuclear cell infiltrates define acute rejection, whereas chronic rejection affects large arteries. Hearts transplanted from male to female C57BL/6 mice undergo acute rejection with interstitial infiltrates at 2 weeks that resolve by 6 weeks when large arteries develop arteriopathy. These processes are dependent on T cells because no infiltrates developed in T cell-deficient mice and transfer of CD4 T cells restored T cell as well as macrophage infiltrates and ultimately neointima formation. Markers of inflammatory macrophages were up-regulated in the interstitium acutely and decreased as markers of wound healing macrophages increased chronically. Programmed cell death protein, a negative costimulator, and its ligand PDL1 were up-regulated in the interstitium during resolution of acute rejection. Blocking PDL1:PD1 interactions in the acute phase increased interstitial T cell infiltrates. Toll-like receptor (TLR) 4 and its endogenous ligand hyaluronan were increased in arteries with neointimal expansion. Injection of hyaluronan fragments increased intragraft production of chemokines. Our data indicate that negative costimulatory pathways are critical for the resolution of acute interstitial infiltrates. In the arterial compartment recognition of endogenous ligands including hyaluronan by the innate TLRs may support the progression of arteriopathy.


Asunto(s)
Rechazo de Injerto/fisiopatología , Trasplante de Corazón , Miocardio/metabolismo , Miocardio/patología , Transducción de Señal/fisiología , Animales , Antígeno B7-H1/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Quimiocina CCL2/metabolismo , Quimiocina CXCL9/metabolismo , Femenino , Ácido Hialurónico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Receptor Toll-Like 4/metabolismo
6.
Am J Transplant ; 15(2): 346-57, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25496308

RESUMEN

Donor-reactive memory T cells undermine organ transplant survival and are poorly controlled by immunosuppression or costimulatory blockade. Memory CD4 T cells provide CD40-independent help for the generation of donor-reactive effector CD8 T cells and alloantibodies (alloAbs) that rapidly mediate allograft rejection. The goal of this study was to investigate the role of B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) in alloresponses driven by memory CD4 T cells. The short-term neutralization of BAFF alone or BAFF plus APRIL synergized with anti-CD154 mAb to prolong heart allograft survival in recipients containing donor-reactive memory CD4 T cells. The prolongation was associated with reduction in antidonor alloAb responses and with inhibited reactivation and helper functions of memory CD4 T cells. Additional depletion of CD8 T cells did not enhance the prolonged allograft survival suggesting that donor-reactive alloAbs mediate late graft rejection in these recipients. This is the first report that targeting the BAFF cytokine network inhibits both humoral and cellular immune responses induced by memory CD4 T cells. Our results suggest that reagents neutralizing BAFF and APRIL may be used to enhance the efficacy of CD40/CD154 costimulatory blockade and improve allograft survival in T cell-sensitized recipients.


Asunto(s)
Factor Activador de Células B/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/fisiología , Antígenos CD40/fisiología , Trasplante de Corazón , Memoria Inmunológica/fisiología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Antígenos CD40/inmunología , Ligando de CD40/inmunología , Ligando de CD40/fisiología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Inmunidad Celular/fisiología , Inmunidad Humoral/fisiología , Isoanticuerpos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales
7.
Am J Transplant ; 14(8): 1753-64, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25040187

RESUMEN

The pathogenic role of macrophages in antibody-mediated rejection (AMR) remains unclear. Monocyte chemoattractant protein-1 (MCP-1/CCL2) is a potent chemotactic factor for monocytes and macrophages. The current studies used a murine model of AMR to investigate the role of graft-derived CCL2 in AMR and how macrophages may participate in antibody-mediated allograft injury. B6.CCR5−/−/CD8−/− recipients rejected MHC-mismatched WT A/J allografts with high donor-reactive antibody titers and diffuse C4d deposition in the large vessels and myocardial capillaries, features consistent with AMR. In contrast, A/J.CCL2−/− allografts induced low donor-reactive antibody titers and C4d deposition at Day 7 posttransplant. Decreased donor-reactive CD4 T cells producing interferon gamma were induced in response to A/J.CCL2−/− versus WT allografts. Consequently, A/J.CCL2−/− allograft survival was modestly but significantly longer than A/J allografts. Macrophages purified from WT allografts expressed high levels of IL-1ß and IL-12p40 and this expression and the numbers of classically activated macrophages were markedly reduced in CCL2-deficient allografts on Day 7. The results indicate that allograft-derived CCL2 plays an important role in directing classically activated macrophages into allografts during AMR and that macrophages are important contributors to the inflammatory environment mediating graft tissue injury in this pathology, suggesting CCL2 as a therapeutic target for AMR.


Asunto(s)
Anticuerpos/sangre , Quimiocina CCL2/metabolismo , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Trasplante de Corazón , Animales , Linfocitos T CD4-Positivos/citología , Proliferación Celular , Supervivencia Celular , Quimiocina CCL2/genética , Quimiotaxis , Citocinas/metabolismo , Citometría de Flujo , Supervivencia de Injerto , Interferón gamma/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Factores de Tiempo , Trasplante Homólogo
8.
Am J Transplant ; 14(6): 1277-89, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24842641

RESUMEN

The presence of CD28(-) memory CD8 T cells in the peripheral blood of renal transplant patients is a risk factor for graft rejection and resistance to CTLA-4Ig induction therapy. In vitro analyses have indicated poor alloantigen-induced CD28(-) memory CD8 T cell proliferation, raising questions about mechanisms mediating their clonal expansion in kidney grafts to mediate injury. Candidate proliferative cytokines were tested for synergy with alloantigen in stimulating CD28(-) memory CD8 T cell proliferation. Addition of IL-15, but not IL-2 or IL-7, to co-cultures of CD28(-) or CD28(+) memory CD8 T cells and allogeneic B cells rescued proliferation of the CD28(-) and enhanced CD28(+) memory T cell proliferation. Proliferating CD28(-) memory CD8 T cells produced high amounts of interferon gamma and tumor necrosis factor alpha and expressed higher levels of the cytolytic marker CD107a than CD28(+) memory CD8 T cells. CTLA-4Ig inhibited alloantigen-induced proliferation of CD28(+) memory CD8 T cell proliferation but had no effect on alloantigen plus IL-15-induced proliferation of either CD28(-) or CD28(+) memory CD8 T cells. These results indicate the ability of IL-15, a cytokine produced by renal epithelial during inflammation, to provoke CD28(-) memory CD8 T cell proliferation and to confer memory CD8 T cell resistance to CTLA-4Ig-mediated costimulation blockade.


Asunto(s)
Antígenos CD28/inmunología , Linfocitos T CD8-positivos/citología , Antígeno CTLA-4/inmunología , Memoria Inmunológica , Interleucina-15/fisiología , Activación de Linfocitos , Linfocitos T CD8-positivos/inmunología , Humanos , Prueba de Cultivo Mixto de Linfocitos
9.
Am J Transplant ; 13(9): 2293-307, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23914930

RESUMEN

Endogenous memory CD8 T cells infiltrate MHC-mismatched cardiac allografts within 12-24 h posttransplant in mice and are activated to proliferate and produce IFN-γ. To more accurately assess the graft injury directly imposed by these endogenous memory CD8 T cells, we took advantage of the ability of anti-LFA-1 mAb given to allograft recipients on days 3 and 4 posttransplant to inhibit the generation of primary effector T cells. When compared to grafts from IgG-treated recipients on day 7 posttransplant, allografts from anti-LFA-1 mAb-treated recipients had increased numbers of CD8 T cells but these grafts had marked decreases in expression levels of mRNA encoding effector mediators associated with graft injury and decreases in donor-reactive CD8 T cells producing IFN-γ. Despite this decreased activity within the allograft, CD8 T cells in allografts from recipients treated with anti-LFA-1 mAb continued to proliferate up to day 7 posttransplant and did not upregulate expression of the exhaustion marker LAG-3 but did have decreased expression of ICOS. These results indicate that endogenous memory CD8 T cells infiltrate and proliferate in cardiac allografts in mice but do not express sufficient levels of functions to mediate overt graft injury and acute rejection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Trasplante de Corazón , Inmunología del Trasplante , Aloinjertos , Animales , Anticuerpos Monoclonales/farmacología , Antígenos CD/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Ligando de CD40/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Proteína Coestimuladora de Linfocitos T Inducibles/biosíntesis , Activación de Linfocitos , Ratones , Proteína del Gen 3 de Activación de Linfocitos
10.
Am J Transplant ; 12(8): 2017-28, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22578247

RESUMEN

Murine CCR5(-/-) recipients produce high titers of antibody to complete MHC-mismatched heart and renal allografts. To study mechanisms of class I MHC antibody-mediated allograft injury, we tested the rejection of heart allografts transgenically expressing a single class I MHC disparity in wild-type C57BL/6 (H-2(b)) and B6.CCR5(-/-) recipients. Donor-specific antibody titers in CCR5(-/-) recipients were 30-fold higher than in wild-type recipients. B6.K(d) allografts survived longer than 60 days in wild-type recipients whereas CCR5(-/-) recipients rejected all allografts within 14 days. Rejection was accompanied by infiltration of CD8 T cells, neutrophils and macrophages, and C4d deposition in the graft capillaries. B6.K(d) allografts were rejected by CD8(-/-)/CCR5(-/-), but not µMT(-/-)/CCR5(-/-), recipients indicating the need for antibody but not CD8 T cells. Grafts recovered at day 10 from CCR5(-/-) and CD8(-/-)/CCR5(-/-) recipients and from RAG-1(-/-) allograft recipients injected with anti-K(d) antibodies expressed high levels of perforin, myeloperoxidase and CCL5 mRNA. These studies indicate that the continual production of antidonor class I MHC antibody can mediate allograft rejection, that donor-reactive CD8 T cells synergize with the antibody to contribute to rejection, and that expression of three biomarkers during rejection can occur in the absence of this CD8 T cell activity.


Asunto(s)
Rechazo de Injerto/inmunología , Trasplante de Corazón , Antígenos de Histocompatibilidad Clase I/inmunología , Animales , Formación de Anticuerpos , Linfocitos T CD8-positivos/inmunología , Quimiocina CCL5/genética , Citometría de Flujo , Inmunoglobulina G/inmunología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Perforina/genética , Peroxidasa/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CCR5/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Am J Transplant ; 12(7): 1708-19, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22420367

RESUMEN

Antibody-mediated allograft rejection is an increasingly recognized problem in clinical transplantation. However, the primary location of donor-specific alloantibody (DSA)-producing cells after transplantation have not been identified. The purpose of this study was to test the contribution of allospecific antibody-secreting cells (ASCs) from different anatomical compartments in a mouse transplantation model. Fully MHC-mismatched heart allografts were transplanted into three groups of recipients: nonsensitized wild type, alloantigen-sensitized wild-type and CCR5(-/-) mice that have exaggerated alloantibody responses. We found that previous sensitization to donor alloantigens resulted in the development of antidonor alloantibody (alloAb) with accelerated kinetics. Nevertheless, the numbers of alloantibody-secreting cells and the serum titers of antidonor IgG alloantibody were equivalent in sensitized and nonsensitized recipients 6 weeks after transplantation. Regardless of recipient sensitization status, the spleen contained higher numbers of donor-reactive ASCs than bone marrow at days 7-21 after transplantation. Furthermore, individual spleen ASCs produced more antidonor IgG alloantibody than bone marrow ASCs. Taken together, our results indicate that the spleen rather than bone marrow is the major source of donor-reactive alloAb early after transplantation in both sensitized and nonsensitized recipients.


Asunto(s)
Formación de Anticuerpos , Trasplante de Corazón , Modelos Animales , Bazo/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Donantes de Tejidos , Trasplante Homólogo
14.
Am J Transplant ; 11(6): 1131-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21645250

RESUMEN

The presence of preformed, donor-specific alloantibodies inpatients undergoing renal transplantation is associated with a high risk of hyperacute and acute antibody-mediated rejection (ABMR), and often limits potential recipients' access to organs from living and deceased donors. Over the last decade, understanding of ABMR has improved markedly and given rise to numerous, diverse strategies for the transplantation of allosensitized recipients. Antibody desensitization programs have been developed to allow renal transplant recipients with a willing but antibody-incompatible living donor to undergo successful transplantation, whereas kidney paired exchange schemes circumvent the antibody incompatibility altogether by finding suitable pairs to donors and recipients. Recognizing the complexity of ABMR and the recent developments that have occurred in this important clinical research field, the Roche Organ Transplantation Research Foundation (ROTRF) organized a symposium during the XXIII Congress of The Transplantation Society in Vancouver, Canada, to discuss current understanding in ABMR and ways to prevent it. This Meeting Report summarizes the presentations of the symposium, which addressed key areas that included the interactions between alloantibodies and the complement system in mediating graft injury, technological advancements for assessing antibody-mediated immune responses to HLA antigens, and the potential benefits and challenges of desensitization and kidney paired donation schemes.


Asunto(s)
Rechazo de Injerto/inmunología , Isoanticuerpos/inmunología , Trasplante de Riñón/inmunología , Formación de Anticuerpos , Rechazo de Injerto/prevención & control , Humanos , Inmunidad Celular
15.
Am J Transplant ; 11(5): 923-35, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21466654

RESUMEN

Alloreactive memory T cells are present in virtually all transplant recipients due to prior sensitization or heterologous immunity and mediate injury undermining graft outcome. In mouse models, endogenous memory CD8 T cells infiltrate MHC-mismatched cardiac allografts and produce IFN-γ in response to donor class I MHC within 24 h posttransplant. The current studies analyzed the efficacy of anti-LFA-1 mAb to inhibit early CD8 T cell cardiac allograft infiltration and activation. Anti-LFA-1 mAb given to C57BL/6 6 (H-2(b)) recipients of A/J (H-2(a)) heart grafts on days -1 and 0 completely inhibited CD8 T cell allograft infiltration, markedly decreased neutrophil infiltration and significantly reduced intragraft expression levels of IFN-γ-induced genes. Donor-specific T cells producing IFN-γ were at low/undetectable numbers in spleens of anti-LFA-1 mAb treated recipients until day 21. These effects combined to promote substantial prolongation (from day 8 to 27) in allograft survival. Delaying anti-LFA-1 mAb treatment until days 3 and 4 posttransplant did not inhibit early memory CD8 T cell infiltration and proliferation within the allograft. These data indicate that peritransplant anti-LFA-1 mAb inhibits early donor-reactive memory CD8 T cell allograft infiltration and inflammation suggesting an effective strategy to attenuate the negative effects of heterologous immunity in transplant recipients.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Trasplante de Corazón/métodos , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Linfocitos T/citología , Animales , Anticuerpos Monoclonales/metabolismo , Linfocitos T CD8-positivos/citología , Citometría de Flujo/métodos , Inmunohistoquímica/métodos , Memoria Inmunológica , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Homólogo/métodos
16.
Am J Transplant ; 10(5): 1117-25, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20353479

RESUMEN

Studies in the past decade advanced our understanding of the development, execution and regulation of T-cell-mediated allograft rejection. This review outlines recent progress and focuses on three major areas of investigation that are likely to guide the development of graft-prolonging therapies in the future. The discussed topics include the contribution of recently discovered molecules to the activation and functions of alloreactive T cells, the emerging problem of alloreactive memory T cells and recently gained insights into the old question of transplantation tolerance.


Asunto(s)
Linfocitos T/inmunología , Tolerancia al Trasplante/inmunología , Animales , Ratones
17.
Am J Transplant ; 10(3): 464-71, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20121738

RESUMEN

The 10th Banff Conference on Allograft Pathology was held in Banff, Canada from August 9 to 14, 2009. A total of 263 transplant clinicians, pathologists, surgeons, immunologists and researchers discussed several aspects of solid organ transplants with a special focus on antibody mediated graft injury. The willingness of the Banff process to adapt continuously in response to new research and improve potential weaknesses, led to the implementation of six working groups on the following areas: isolated v-lesion, fibrosis scoring, glomerular lesions, molecular pathology, polyomavirus nephropathy and quality assurance. Banff working groups will conduct multicenter trials to evaluate the clinical relevance, practical feasibility and reproducibility of potential changes to the Banff classification. There were also sessions on quality improvement in biopsy reading and utilization of virtual microscopy for maintaining competence in transplant biopsy interpretation. In addition, compelling molecular research data led to the discussion of incorporation of omics-technologies and discovery of new tissue markers with the goal of combining histopathology and molecular parameters within the Banff working classification in the near future.


Asunto(s)
Anticuerpos/química , Trasplante de Órganos/métodos , Biopsia , Canadá , Complemento C4b/metabolismo , Fibrosis/patología , Humanos , Enfermedades Renales/diagnóstico , Enfermedades Renales/patología , Enfermedades Renales/virología , Trasplante de Riñón , Estudios Multicéntricos como Asunto , Fragmentos de Péptidos/metabolismo , Fenotipo , Infecciones por Polyomavirus/diagnóstico , Control de Calidad
18.
Am J Transplant ; 9(1): 14-22, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19067663

RESUMEN

This review relates the basic functions of platelets to specific aspects of organ allograft rejection. Platelet activation can occur in the donor or recipient before transplantation as well as during antibody- and cell-mediated rejection. Biopsies taken during organ procurement from cadaver donors have documented that activated platelets are attached to vascular endothelial cells or leukocytes. In addition, many patients waiting for transplants have activated platelets due to the diseases that lead to organ failure or as a result of interventions used to support patients before and during transplantation. The contribution of platelets to hyperacute rejection of both allografts and xenografts is well recognized. Intravascular aggregates of platelets can also be prominent in experimental and clinical transplants that undergo acute antibody or cell-mediated rejection. In acute rejection, platelets can recruit mononuclear cells by secretion of chemokines. After contact, monocytes, macrophages and T cells interact with platelets through receptor/ligand pairs, including P-selectin/PSGL-1 and CD40/CD154. There is a potential for therapy to inhibit platelet mediated immune stimulation, but it is counterbalanced by the need to maintain coagulation in the perioperative period.


Asunto(s)
Plaquetas/inmunología , Trasplante de Órganos , Rechazo de Injerto/inmunología , Humanos
19.
Am J Transplant ; 7(11): 2447-54, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17927608

RESUMEN

Platelet interactions with dendritic cells, T cells and B cells have been best studied in vasculitis and atherosclerosis, but similar mechanisms may contribute to acute and chronic vascular lesions in transplants. In acute inflammation, platelets adhere to vessels and release mediators that increase endothelial cell activation and leukocyte recruitment. Adherent platelets can also augment antibody and cellular immune responses. Activated platelets recruit T cells and initiate a feedback loop. In this loop, platelets secrete chemokines to recruit T cells, and then activated T cells stimulate platelets through CD40-CD154 interactions to secrete more chemokines thereby recruiting more T cells. The interaction of platelets and T cells is enhanced by P-selectin/PSGL-1 stimulation. Both helper and cytotoxic T cells are stimulated by platelets. Antibody production that is stimulated through increased helper T-cell function can activate complement. This sets up another activation loop because platelets express receptors for antibodies and complement. In addition to inflammation, platelets stimulate repair by releasing growth factors and chemokines to recruit circulating vascular progenitor cells. These repair mechanisms could promote the replacement of donor parenchmal cells with recipient cells and contribute to vascuplopathy. This review discusses the interplay of platelets and the immune system in relation to transplantation.


Asunto(s)
Plaquetas/fisiología , Inflamación/inmunología , Inflamación/fisiopatología , Inmunología del Trasplante , Trasplante/efectos adversos , Linfocitos B/inmunología , Plaquetas/inmunología , Proteínas del Sistema Complemento/inmunología , Células Dendríticas , Humanos , Inflamación/sangre , Células Madre/fisiología , Linfocitos T/inmunología
20.
Am J Transplant ; 7(11): 2605-14, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17868071

RESUMEN

The role of non-complement-activating alloantibodies in humoral graft rejection is unclear. We hypothesized that the non-complement-activating alloantibodies synergistically activate complement in combination with complement-activating antibodies. B10.A hearts were transplanted into immunoglobulin knock out (Ig-KO) mice reconstituted with monoclonal antibodies to MHC class I antigens. In allografts of unreconstituted Ig-KO recipients, no C4d was detected. Similarly, reconstitution with IgG1 or low dose IgG2b alloantibodies did not induce C4d deposition. However, mice administered with a low dose of IgG2b combined with IgG1 had heavy linear deposits of C4d on vascular endothelium. C4d deposits correlated with decreased graft survival. To replicate this synergy in vitro, mononuclear cells from B10.A mice were incubated with antibodies to MHC class I antigens followed by incubation in normal mouse serum. Flow cytometry revealed that both IgG2a and IgG2b synergized with IgG1 to deposit C4d. This synergy was significantly decreased in mouse serum deficient in mannose binding lectin (MBL) and in serum deficient in C1q. Reconstitution of MBL-A/C knock out (MBL-KO) serum with C1q-knock out (C1q-KO) serum reestablished the synergistic activity. This suggests a novel role for non-complement-activating alloantibodies and MBL in humoral rejection.


Asunto(s)
Complemento C4b/inmunología , Proteínas del Sistema Complemento/inmunología , Trasplante de Corazón/inmunología , Isoanticuerpos/sangre , Fragmentos de Péptidos/inmunología , Animales , Activación de Complemento/inmunología , Complemento C4b/análisis , Citometría de Flujo , Trasplante de Corazón/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Modelos Animales , Fragmentos de Péptidos/análisis , Conejos , Trasplante Homólogo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...