Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 15(6): 1214-27, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27134179

RESUMEN

Chronic rejection of solid organ allografts remains the major cause of transplant failure. Donor-derived tissue-resident lymphocytes are transferred to the recipient during transplantation, but their impact on alloimmunity is unknown. Using mouse cardiac transplant models, we show that graft-versus-host recognition by passenger donor CD4 T cells markedly augments recipient cellular and humoral alloimmunity, resulting in more severe allograft vasculopathy and early graft failure. This augmentation is enhanced when donors were pre-sensitized to the recipient, is dependent upon avoidance of host NK cell recognition, and is partly due to provision of cognate help for allo-specific B cells from donor CD4 T cells recognizing B cell MHC class II in a peptide-degenerate manner. Passenger donor lymphocytes may therefore influence recipient alloimmune responses and represent a therapeutic target in solid organ transplantation.


Asunto(s)
Inmunidad Adaptativa , Aloinjertos/inmunología , Linfocitos T CD4-Positivos/inmunología , Donantes de Tejidos , Animales , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Diferenciación Celular , Rechazo de Injerto/inmunología , Enfermedad Injerto contra Huésped/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Inmunidad Humoral/inmunología , Células Asesinas Naturales/inmunología , Ratones Endogámicos BALB C , Modelos Inmunológicos , Péptidos/metabolismo , Células Plasmáticas/patología , Receptores de Antígenos de Linfocitos B/metabolismo , Trasplante Homólogo
2.
BMJ Open ; 6(2): e010377, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26916695

RESUMEN

INTRODUCTION: There is significant intercentre variability in access to renal transplantation in the UK due to poorly understood factors. The overarching aims of this study are to improve equity of access to kidney and kidney-pancreas transplantation across the UK and to optimise organ allocation to maximise the benefit and cost-effectiveness of transplantation. METHODS AND ANALYSIS: 6844 patients aged 18-75 years starting dialysis and/or receiving a transplant together with matched patients active on the transplant list from all 72 UK renal units were recruited between November 2011 and March 2013 and will be followed for at least 3 years. The outcomes of interest include patient survival, access to the transplant list, receipt of a transplant, patient-reported outcome measures (PROMs) including quality of life, treatment satisfaction, well-being and health status on different forms of renal replacement therapy. Sociodemographic and clinical data were prospectively collected from case notes and from interviews with patients and local clinical teams. Qualitative process exploration with clinical staff will help identify unit-specific factors that influence access to renal transplantation. A health economic analysis will explore costs and outcomes associated with alternative approaches to organ allocation. The study will deliver: (1) an understanding of patient and unit-specific factors influencing access to renal transplantation in the UK, informing potential changes to practices and policies to optimise outcomes and reduce intercentre variability; (2) a patient-survival probability model to standardise access to the renal transplant list and (3) an understanding of PROMs and health economic impact of kidney and kidney-pancreas transplantation to inform the development of a more sophisticated and fairer organ allocation algorithm. ETHICS AND DISSEMINATION: The protocol has been independently peer reviewed by National Institute for Health Research (NIHR) and approved by the East of England Research Ethics Committee. The results will be published in peer-reviewed journals and presented at conferences.


Asunto(s)
Accesibilidad a los Servicios de Salud/estadística & datos numéricos , Trasplante de Riñón/estadística & datos numéricos , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Proyectos de Investigación , Adolescente , Adulto , Anciano , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Reino Unido , Listas de Espera , Adulto Joven
3.
Cell Rep ; 14(5): 1232-1245, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26804905

RESUMEN

MHC alloantigen is recognized by two pathways: "directly," intact on donor cells, or "indirectly," as self-restricted allopeptide. The duration of each pathway, and its relative contribution to allograft vasculopathy, remain unclear. Using a murine model of chronic allograft rejection, we report that direct-pathway CD4 T cell alloresponses, as well as indirect-pathway responses against MHC class II alloantigen, are curtailed by rapid elimination of donor hematopoietic antigen-presenting cells. In contrast, persistent presentation of epitope resulted in continual division and less-profound contraction of the class I allopeptide-specific CD4 T cell population, with approximately 10,000-fold more cells persisting than following acute allograft rejection. This expanded population nevertheless displayed sub-optimal anamnestic responses and was unable to provide co-stimulation-independent help for generating alloantibody. Indirect-pathway CD4 T cell responses are heterogeneous. Appreciation that responses against particular alloantigens dominate at late time points will likely inform development of strategies aimed at improving transplant outcomes.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Isoantígenos/inmunología , Inmunidad Adaptativa , Animales , Presentación de Antígeno/inmunología , Trasplante de Corazón , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunidad Innata , Activación de Linfocitos/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Péptidos/inmunología , Trasplante Homólogo
4.
Proc Natl Acad Sci U S A ; 112(41): 12788-93, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26420874

RESUMEN

Adaptive CD8 T-cell immunity is the principal arm of the cellular alloimmune response, but its development requires help. This can be provided by CD4 T cells that recognize alloantigen "indirectly," as self-restricted allopeptide, but this process remains unexplained, because the target epitopes for CD4 and CD8 T-cell recognition are "unlinked" on different cells (recipient and donor antigen presenting cells (APCs), respectively). Here, we test the hypothesis that the presentation of intact and processed MHC class I alloantigen by recipient dendritic cells (DCs) (the "semidirect" pathway) allows linked help to be delivered by indirect-pathway CD4 T cells for generating destructive cytotoxic CD8 T-cell alloresponses. We show that CD8 T-cell-mediated rejection of murine heart allografts that lack hematopoietic APCs requires host secondary lymphoid tissue (SLT). SLT is necessary because within it, recipient dendritic cells can acquire MHC from graft parenchymal cells and simultaneously present it as intact protein to alloreactive CD8 T cells and as processed peptide alloantigen for recognition by indirect-pathway CD4 T cells. This enables delivery of essential help for generating cytotoxic CD8 T-cell responses that cause rapid allograft rejection. In demonstrating the functional relevance of the semidirect pathway to transplant rejection, our findings provide a solution to a long-standing conundrum as to why SLT is required for CD8 T-cell allorecognition of graft parenchymal cells and suggest a mechanism by which indirect-pathway CD4 T cells provide help for generating effector cytotoxic CD8 T-cell alloresponses at late time points after transplantation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Rechazo de Injerto/inmunología , Trasplante de Corazón , Isoantígenos/inmunología , Aloinjertos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Células Dendríticas/inmunología , Células Dendríticas/patología , Rechazo de Injerto/patología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
5.
Lancet ; 385 Suppl 1: S17, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26312839

RESUMEN

BACKGROUND: Uniquely, alloantigen is recognised by two pathways: as intact antigen on the surface of donor antigen-presenting cells (direct) and as self-restricted processed allopeptide (indirect). The indirect pathway is believed to be longlasting, and is generally considered to be a single entity. Here we address how indirect responses against different alloantigens differ in their strength and longevity, and how this knowledge could be used to direct immunoregulatory therapy with antigen-specific regulatory T cells (Tregs). METHODS: A murine model of cardiac transplantation was used (bm12.Kd.IE to C57BL/6). Indirect CD4 T-cell allorecognition of mismatched donor MHC class I and II, and of H-Y minor histocompatibility antigen was assessed by quantifying proliferation of adoptively transferred monoclonal T-cell receptor transgenic T cells (TCR75, Tea, Mar). Antigen presentation by dendritic cells and B cells was assessed by selective depletion with diphtheria toxin or depleting anti-CD20 monoclonal antibody. Tregs were generated by in-vitro culture. FINDINGS: Indirect pathway responses were heterogeneous. Whereas the indirect response against class I alloantigen was longlasting and persistently strong, the response against class II alloantigen decayed within 2 weeks. Leucocyte depletion studies confirmed that this difference was due to rapid destruction of MHC class II expressing donor B cells and dendritic cells in the recipient, whereas anti-class I responses were generated by continual processing of graft parenchymal cells; recognition of donor haemopoietic fraction was not required. Notably, transfer of MHC class I specific Tregs at transplant or 3 weeks later abrogated germinal centre alloantibody responses and blocked development of allograft vasculopathy, whereas class II specific Tregs were ineffective when transferred at the late timepoint. INTERPRETATION: Although indirect allorecognition is considered to be a single entity, our results show that it consists of a number of responses that vary in duration and strength according to target alloantigen. The ability of class I allopeptide specific Tregs, but not class II specific Tregs, to prevent rejection when transferred at a late timepoint suggests that antigen-specific targeting of dominant and longlasting pathways might be particularly effective at preventing chronic rejection. FUNDING: Wellcome Trust Clinical Research Training Fellowship.

6.
Lancet ; 385 Suppl 1: S18, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26312840

RESUMEN

BACKGROUND: Memory T cells are known to reside in peripheral non-lymphoid tissue, but how their presence within solid organ allografts affects transplant outcomes is not known. We have previously described how graft-versus-host (GVH) allorecognition by passenger CD4 T cells within MHC class II-mismatched bm12 heart grafts provokes antinuclear humoral autoimmunity in C57BL/6 recipient mice. Here we aimed to examine how such GVH recognition affects the alloresponse to allografts with greater mismatching. METHODS: A MHC class I and II mismatched murine model of cardiac transplantation was developed (bm12.Kd.IE to C57BL/6). After transplantation, cellular and humoral responses against mismatched antigens were measured with ELISPOT and ELISA, and the effect of GVH recognition assessed by depletion of donor CD4 T cells before graft procurement. Antinuclear autoantibody development was assessedwith HeP-2 indirect immunofluorescence. The role of recipient natural killer (NK) cells was examined by depletion with anti-NK1.1 antibody. FINDINGS: Bm12.Kd.IE heart grafts provoked strong germinal centre alloantibody and autoantibody responses in C57BL/6 recipients and developed allograft vasculopathy. By contrast, heart grafts from CD4 T-cell-depleted donors developed only minimal vasculopathy, and the alloantibody responses were weaker, without observable autoantibody. Bm12.Kd.IE CD4 T cells survived long term when transferred to RAG hosts suggesting that avoidance of killing by host NK cells might be essential for autoantibody development. In support, in a model of alloantibody-mediated vasculopathy, depletion of NK cells from a C57BL/6 recipient of a BALB/c heart graft resulted in the development of autoantibody, amplification of the alloantibody response, and rapid allograft rejection. This amplification was abrogated by depletion of donor CD4 T cells. INTERPRETATION: Although host adaptive immunity is expected to bring about destruction of passenger lymphocytes within heart allografts, this process occurs too slowly to prevent GVH-mediated augmentation of the alloresponse to the graft. Rather, rapid killing of donor lymphocytes by host alloreactive NK cells is essential. Passenger CD4 lymphocytes might therefore contribute to chronic rejection in recipients receiving an allograft that does not prompt innate NK cell recognition. FUNDING: Wellcome Trust Clinical Research Training Fellowship.

7.
J Heart Lung Transplant ; 34(11): 1471-80, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26140808

RESUMEN

BACKGROUND: Free radical production and mitochondrial dysfunction during cardiac graft reperfusion is a major factor in post-transplant ischemia-reperfusion (IR) injury, an important underlying cause of primary graft dysfunction. We therefore assessed the efficacy of the mitochondria-targeted anti-oxidant MitoQ in reducing IR injury in a murine heterotopic cardiac transplant model. METHODS: Hearts from C57BL/6 donor mice were flushed with storage solution alone, solution containing the anti-oxidant MitoQ, or solution containing the non-anti-oxidant decyltriphenylphosphonium control and exposed to short (30 minutes) or prolonged (4 hour) cold preservation before transplantation. Grafts were transplanted into C57BL/6 recipients and analyzed for mitochondrial reactive oxygen species production, oxidative damage, serum troponin, beating score, and inflammatory markers 120 minutes or 24 hours post-transplant. RESULTS: MitoQ was taken up by the heart during cold storage. Prolonged cold preservation of donor hearts before IR increased IR injury (troponin I, beating score) and mitochondrial reactive oxygen species, mitochondrial DNA damage, protein carbonyls, and pro-inflammatory cytokine release 24 hours after transplant. Administration of MitoQ to the donor heart in the storage solution protected against this IR injury by blocking graft oxidative damage and dampening the early pro-inflammatory response in the recipient. CONCLUSIONS: IR after heart transplantation results in mitochondrial oxidative damage that is potentiated by cold ischemia. Supplementing donor graft perfusion with the anti-oxidant MitoQ before transplantation should be studied further to reduce IR-related free radical production, the innate immune response to IR injury, and subsequent donor cardiac injury.


Asunto(s)
Antioxidantes/uso terapéutico , Trasplante de Corazón , Mitocondrias Cardíacas/metabolismo , Compuestos Organofosforados/uso terapéutico , Disfunción Primaria del Injerto/etiología , Daño por Reperfusión/prevención & control , Ubiquinona/análogos & derivados , Animales , Modelos Animales de Enfermedad , Femenino , Depuradores de Radicales Libres/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Micronutrientes/uso terapéutico , Mitocondrias Cardíacas/patología , Preservación de Órganos , Estrés Oxidativo , Disfunción Primaria del Injerto/metabolismo , Disfunción Primaria del Injerto/patología , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Ubiquinona/uso terapéutico
8.
Redox Biol ; 5: 163-168, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25965144

RESUMEN

Ischemia-reperfusion (IR) injury to the kidney occurs in a range of clinically important scenarios including hypotension, sepsis and in surgical procedures such as cardiac bypass surgery and kidney transplantation, leading to acute kidney injury (AKI). Mitochondrial oxidative damage is a significant contributor to the early phases of IR injury and may initiate a damaging inflammatory response. Here we assessed whether the mitochondria targeted antioxidant MitoQ could decrease oxidative damage during IR injury and thereby protect kidney function. To do this we exposed kidneys in mice to in vivo ischemia by bilaterally occluding the renal vessels followed by reperfusion for up to 24h. This caused renal dysfunction, measured by decreased creatinine clearance, and increased markers of oxidative damage. Administering MitoQ to the mice intravenously 15 min prior to ischemia protected the kidney from damage and dysfunction. These data indicate that mitochondrial oxidative damage contributes to kidney IR injury and that mitochondria targeted antioxidants such as MitoQ are potential therapies for renal dysfunction due to IR injury.


Asunto(s)
Antioxidantes/farmacología , Mitocondrias/metabolismo , Compuestos Organofosforados/farmacología , Estrés Oxidativo/efectos de los fármacos , Daño por Reperfusión/prevención & control , Ubiquinona/análogos & derivados , Animales , Antioxidantes/uso terapéutico , Creatinina/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos Organofosforados/uso terapéutico , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Ubiquinona/farmacología , Ubiquinona/uso terapéutico
9.
Regen Med ; 10(3): 287-304, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25933238

RESUMEN

One of the major goals of regenerative medicine is repair or replacement of diseased and damaged tissues by transfer of differentiated stem cells or stem cell-derived tissues. The possibility that these tissues will be destroyed by immunological rejection remains a challenge that can only be overcome through a better understanding of the nature and expression of potentially immunogenic molecules associated with cell replacement therapy and the mechanisms and pathways resulting in their immunologic rejection. This review draws on clinical experience of organ and tissue transplantation, and on transplantation immunology research to consider practical approaches for avoiding and overcoming the possibility of rejection of stem cell-derived tissues.


Asunto(s)
Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Medicina Regenerativa/métodos , Células Madre/inmunología , Trasplante de Tejidos/métodos , Humanos
10.
Kidney Int ; 86(5): 1039-48, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24717292

RESUMEN

We have analyzed the relationship between donor mismatches at each HLA locus and development of HLA locus-specific antibodies in patients listed for repeat transplantation. HLA antibody screening was undertaken using single-antigen beads in 131 kidney transplant recipients returning to the transplant waiting list following first graft failure. The number of HLA mismatches and the calculated reaction frequency of antibody reactivity against 10,000 consecutive deceased organ donors were determined for each HLA locus. Two-thirds of patients awaiting repeat transplantation were sensitized (calculated reaction frequency over 15%) and half were highly sensitized (calculated reaction frequency of 85% and greater). Antibody levels peaked after re-listing for repeat transplantation, were independent of graft nephrectomy and were associated with length of time on the waiting list (odds ratio 8.4) and with maintenance on dual immunosuppression (odds ratio 0.2). Sensitization was independently associated with increasing number of donor HLA mismatches (odds ratio 1.4). All mismatched HLA loci contributed to the development of HLA locus-specific antibodies (HLA-A: odds ratio 3.2, HLA-B: odds ratio 3.4, HLA-C: odds ratio 2.5, HLA-DRB1: odds ratio 3.5, HLA-DRB3/4/5: odds ratio 3.9, and HLA-DQ: odds ratio 3.0 (all significant)). Thus, the risk of allosensitization following failure of a first renal transplant increases incrementally with the number of mismatches at all HLA loci assessed. Maintenance of re-listed patients on dual immunosuppression was associated with a reduced risk of sensitization.


Asunto(s)
Antígenos HLA-A/inmunología , Antígenos HLA-B/inmunología , Antígenos HLA-C/inmunología , Antígenos HLA-DQ/inmunología , Antígenos HLA-DR/inmunología , Histocompatibilidad , Isoanticuerpos/sangre , Trasplante de Riñón/efectos adversos , Donantes de Tejidos , Adolescente , Adulto , Biomarcadores/sangre , Niño , Preescolar , Femenino , Prueba de Histocompatibilidad , Humanos , Inmunosupresores/efectos adversos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Recurrencia , Factores de Riesgo , Factores de Tiempo , Insuficiencia del Tratamiento , Listas de Espera , Adulto Joven
11.
Transpl Int ; 27(5): 437-51, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24471420

RESUMEN

Renal transplantation is potentially curative in renal failure, but long-term efficacy is limited by untreatable chronic rejection. Endothelial damage contributes to chronic rejection and is potentially repairable by circulating endothelial progenitor cells (EPC). The frequency and function of EPC are variably influenced by end-stage renal failure (ESRF). Here, we isolated and functionally characterized the late outgrowth EPC (LO-EPC) from ESRF patients to investigate their potential for endothelial repair. Patients with ESRF generated more LO-EPC colonies than healthy controls and had higher plasma levels of IL-1rα, IL-16, IL-6, MIF, VEGF, Prolactin, and PLGF. Patients' LO-EPC displayed normal endothelial cell morphology, increased secretion of PLGF, MCP-1, and IL-1ß, and normal network formation in vitro and in vivo. They demonstrated decreased adhesion to extracellular matrix. Integrin gene profiles and protein expression were comparable in patients and healthy volunteers. In some patients, mesenchymal stem cells (MSC) were co-isolated and could be differentiated into adipocytes and osteocytes in vitro. This is the first study to characterize LO-EPC from patients with ESRF. Their behavior in vitro reflects the presence of elevated trophic factors; their ability to proliferate in vitro and angiogenic function makes them candidates for prevention of chronic rejection. Their impaired adhesion and the presence of MSC are areas for potential therapeutic intervention.


Asunto(s)
Células Progenitoras Endoteliales/fisiología , Fallo Renal Crónico/patología , Adulto , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Citocinas/fisiología , Células Progenitoras Endoteliales/citología , Femenino , Humanos , Integrinas/genética , Masculino , Neovascularización Fisiológica
12.
J Immunol ; 190(11): 5829-38, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23630361

RESUMEN

In transplantation, direct-pathway CD8 T cells that recognize alloantigen on donor cells require CD4 help for activation and cytolytic function. The ability of indirect-pathway CD4 T cells to provide this help remains unexplained, because a fundamental requirement for epitope linkage is seemingly broken. The simultaneous presentation, by host dendritic cells (DCs), of both intact MHC class I alloantigen and processed alloantigen would deliver linked help, but has not been demonstrated definitively. In this study, we report that following in vitro coculture with BALB/c DCs, small numbers (~1.5%) of C57BL/6 (B6) DCs presented acquired H-2(d) alloantigen both as processed allopeptide and as unprocessed Ag. This represented class I alloantigen provides a conformational epitope for direct-pathway allorecognition, because B6 DCs isolated from cocultures and transferred to naive B6 mice provoked cytotoxic CD8 T cell alloimmunity. Crucially, this response was dependent upon simultaneous presentation of class II-restricted allopeptide, because despite acquiring similar amounts of H-2(d) alloantigen upon coculture, MHC class II-deficient B6 DCs failed to elicit cytotoxic alloimmunity. The relevance of this pathway to solid-organ transplantation was then confirmed by the demonstration that CD8 T cell cytotoxicity was provoked in secondary recipients by transfer of DCs purified from wild-type, but not from MHC class II-deficient, C57BL/6 recipients of BALB/c heart transplants. These experiments demonstrate that representation of conformationally intact MHC alloantigen by recipient APC can induce cytotoxic alloimmunity, but simultaneous copresentation of processed allopeptide is essential, presumably because this facilitates linked recognition by indirect-pathway CD4 Th cells.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad/inmunología , Isoantígenos/inmunología , Animales , Trasplante de Corazón/inmunología , Inmunidad Celular , Inmunidad Humoral , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones
13.
Transplantation ; 96(8): 681-8, 2013 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-23715047

RESUMEN

With the advent of cellular therapies, it has become clear that the success of future therapies in prolonging allograft survival will require an intimate understanding of the allorecognition pathways and effector mechanisms that are responsible for chronic rejection and late graft loss.Here, we consider current understanding of T-cell allorecognition pathways and discuss the most likely mechanisms by which these pathways collaborate with other effector mechanisms to cause allograft rejection. We also consider how this knowledge may inform development of future strategies to prevent allograft rejection.Although both direct and indirect pathway CD4 T cells appear active immediately after transplantation, it has emerged that indirect pathway CD4 T cells are likely to be the dominant alloreactive T-cell population late after transplantation. Their ability to provide help for generating long-lived alloantibody is likely one of the main mechanisms responsible for the progression of allograft vasculopathy and chronic rejection.Recent work has suggested that regulatory T cells may be an effective cellular therapy in transplantation. Given the above, adoptive therapy with CD4 regulatory T cells with indirect allospecificity is a rational first choice in attempting to attenuate the development and progression of chronic rejection; those with additional properties that enable inhibition of germinal center alloantibody responses hold particular appeal.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Rechazo de Injerto/inmunología , Tolerancia Inmunológica/inmunología , Isoanticuerpos/inmunología , Trasplante de Órganos , Humanos , Trasplante Homólogo
14.
J Immunol ; 189(12): 5694-702, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23150718

RESUMEN

Fcγ receptors (FcγR) provide important immunoregulation. Targeting inhibitory FcγRIIb may therefore prolong allograft survival, but its role in transplantation has not been addressed. FcγRIIb signaling was examined in murine models of acute or chronic cardiac allograft rejection by transplanting recipients that either lacked FcγRIIb expression (FcγRIIb(-/-)) or overexpressed FcγRIIb on B cells (B cell transgenic [BTG]). Acute heart allograft rejection occurred at the same tempo in FcγRIIb(-/-) C57BL/6 (B6) recipients as wild type recipients, with similar IgG alloantibody responses. In contrast, chronic rejection of MHC class II-mismatched bm12 cardiac allografts was accelerated in FcγRIIb(-/-) mice, with development of more severe transplant arteriopathy and markedly augmented effector autoantibody production. Autoantibody production was inhibited and rejection was delayed in BTG recipients. Similarly, whereas MHC class I-mismatched B6.K(d) hearts survived indefinitely and remained disease free in B6 mice, much stronger alloantibody responses and progressive graft arteriopathy developed in FcγRIIb(-/-) recipients. Notably, FcγRIIb-mediated inhibition of B6.K(d) heart graft rejection was abrogated by increasing T cell help through transfer of additional H2.K(d)-specific CD4 T cells. Thus, inhibitory FcγRIIb signaling regulates chronic but not acute rejection, most likely because the supra-optimal helper CD4 T cell response in acute rejection overcomes FcγRIIb-mediated inhibition of the effector B cell population. Immunomodulation of FcγRIIb in clinical transplantation may hold potential for inhibiting progression of transplant arteriopathy and prolonging transplant survival.


Asunto(s)
Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Inmunoglobulina G/fisiología , Isoanticuerpos/biosíntesis , Receptores de IgG/antagonistas & inhibidores , Receptores de IgG/fisiología , Transducción de Señal/inmunología , Enfermedad Aguda , Animales , Enfermedad Crónica , Rechazo de Injerto/metabolismo , Trasplante de Corazón/inmunología , Células Hep G2 , Humanos , Inmunoglobulina G/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Receptores de IgG/deficiencia
15.
J Immunol ; 189(12): 5703-12, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23162131

RESUMEN

Essential help for long-lived alloantibody responses is theoretically provided only by CD4 T cells that recognize target alloantigen, processed and presented by the allospecific B cell. We demonstrate that in an alloresponse to multiple MHC disparities, cognate help for class-switched alloantibody may also be provided by CD4 T cells specific for a second "helper" alloantigen. This response was much shorter-lived than when help was provided conventionally, by Th cell recognition of target alloantigen. Nevertheless, long-lasting humoral alloimmunity developed when T cell memory against the helper alloantigen was first generated. Costimulatory blockade abrogated alloantibody produced through naive Th cell recognition of target alloantigen but, crucially, blockade was ineffective when help was provided by memory responses to the accessory helper alloantigen. These results suggest that memory Th cell responses against previously encountered graft alloantigen may be the dominant mechanism for providing help to generate new specificities of alloantibody in transplant patients receiving immunosuppression.


Asunto(s)
Memoria Inmunológica/inmunología , Isoanticuerpos/biosíntesis , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Femenino , Trasplante de Corazón/inmunología , Trasplante de Corazón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Quimera por Radiación/inmunología , Trasplante de Piel/inmunología , Trasplante de Piel/patología , Linfocitos T Colaboradores-Inductores/metabolismo , Factores de Tiempo
16.
Cell Stem Cell ; 11(2): 147-52, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22862941

RESUMEN

The likelihood for immunological rejection of Human Leukocyte Antigens (HLA)-mismatched induced pluripotent stem cells (iPSCs) limits their therapeutic potential. Here we show how a tissue bank from 150 selected homozygous HLA-typed volunteers could match 93% of the UK population with a minimal requirement for immunosuppression. Our model provides a practical approach for using existing HLA-typed samples to generate an iPSC stem cell bank that circumvents prospective typing of a large number of individuals.


Asunto(s)
Antígenos HLA/inmunología , Prueba de Histocompatibilidad , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/inmunología , Trasplante de Células Madre/métodos , Donantes de Tejidos , Humanos , Trasplante Homólogo
17.
J Immunol ; 188(6): 2643-52, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22323543

RESUMEN

The durable alloantibody responses that develop in organ transplant patients indicate long-lived plasma cell output from T-dependent germinal centers (GCs), but which of the two pathways of CD4 T cell allorecognition is responsible for generating allospecific T follicular helper cells remains unclear. This was addressed by reconstituting T cell-deficient mice with monoclonal populations of TCR-transgenic CD4 T cells that recognized alloantigen only as conformationally intact protein (direct pathway) or only as self-restricted allopeptide (indirect pathway) and then assessing the alloantibody response to a heart graft. Recipients reconstituted with indirect-pathway CD4 T cells developed long-lasting IgG alloantibody responses, with splenic GCs and allospecific bone marrow plasma cells readily detectable 50 d after heart transplantation. Differentiation of the transferred CD4 T cells into T follicular helper cells was confirmed by follicular localization and by acquisition of signature phenotype. In contrast, IgG alloantibody was not detectable in recipient mice reconstituted with direct-pathway CD4 T cells. Neither prolongation of the response by preventing NK cell killing of donor dendritic cells nor prior immunization to develop CD4 T cell memory altered the inability of the direct pathway to provide allospecific B cell help. CD4 T cell help for GC alloantibody responses is provided exclusively via the indirect-allorecognition pathway.


Asunto(s)
Centro Germinal/inmunología , Isoanticuerpos/inmunología , Activación de Linfocitos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Inmunología del Trasplante/inmunología , Animales , Diferenciación Celular/inmunología , Separación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Centro Germinal/citología , Inmunohistoquímica , Isoanticuerpos/biosíntesis , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Colaboradores-Inductores/citología , Trasplante Homólogo/inmunología
18.
Transpl Int ; 25(2): 229-41, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22117534

RESUMEN

Chronic allograft vasculopathy (CAV) is a major cause of organ transplant failure that responds poorly to treatment. Endothelial activation, dysfunction and apoptosis contribute to CAV, whereas strategies for protecting endothelium and maximizing endothelial repair may diminish it. Late outgrowth endothelial progenitor cells (LO-EPC) can home to areas of injury and integrate into damaged vessels, implying a role in vascular repair; however, in an allograft, LO-EPC would be exposed to the hazardous microenvironment associated with transplant-related ischaemia reperfusion (I/R) injury and persistent inflammation. We evaluated the in vitro effect of I/R injury and the proinflammatory cytokine tumour necrosis factor (TNF)-α on LO-EPC phenotype and function. We show that LO-EPC are intrinsically more tolerant than mature EC to I/R injury induced apoptosis, maintaining their proliferative, migratory and network formation capacity. Under inflammatory conditions, LO-EPC were activated and released higher levels of inflammatory cytokines, upregulated adhesion molecule expression, and were more susceptible to apoptosis. Lentiviral vector-mediated overexpression of the protective gene A20 in LO-EPC maintained their angiogenic phenotype and function, and protected them against TNF-α-mediated apoptosis, reducing ICAM-1 expression and inflammatory cytokine secretion. Administration of ex vivo modified LO-EPC overexpressing A20 might effect vascular repair of damaged allografts and protect from CAV.


Asunto(s)
Células Endoteliales/fisiología , Rechazo de Injerto/terapia , Trasplante de Órganos/efectos adversos , Daño por Reperfusión/prevención & control , Células Madre/fisiología , Enfermedades Vasculares/prevención & control , Apoptosis/efectos de los fármacos , Células Cultivadas , Humanos , Trasplante Homólogo , Factor de Necrosis Tumoral alfa/farmacología
19.
FASEB J ; 26(1): 51-62, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21926237

RESUMEN

Tertiary lymphoid organs (TLOs) may develop within allografts, but their contribution to graft rejection remains unclear. Here, we study a mouse model of autoantibody-mediated cardiac allograft vasculopathy to clarify the alloimmune responses mediated by intragraft TLOs and whether blocking lymphotoxin-ß-receptor (LTßR) signaling, a pathway essential for lymphoid organogenesis, abrogates TLO development. TLOs (defined as discrete lymphoid aggregates associated with high endothelial venules) were detectable in 9 of 13 heart allografts studied and were predominantly B cell in composition, harboring germinal-center activity. These are most likely manifestations of the humoral autoimmunity triggered in this model after transplantation; TLOs did not develop if autoantibody production was prevented. Treatment with inhibitory LTßR-Ig fusion protein virtually abolished allograft TLO formation (mean TLOs/heart: 0.2 vs. 2.2 in control recipients; P=0.02), with marked attenuation of the autoantibody response. Recipients primed for autoantibody before transplantation rejected grafts rapidly, but this accelerated rejection was prevented by postoperative administration of LTßR-Ig (median survival time: 18 vs. >50 d, respectively, P=0.003). Our results provide the first demonstration that TLOs develop within chronically rejecting heart allografts, are predominantly B cell in origin, and can be targeted pharmacologically to inhibit effector humoral responses.


Asunto(s)
Coristoma/prevención & control , Trasplante de Corazón/inmunología , Tejido Linfoide/patología , Receptor beta de Linfotoxina/metabolismo , Linfotoxina beta/metabolismo , Transducción de Señal/inmunología , Animales , Linfocitos B/inmunología , Médula Ósea/inmunología , Médula Ósea/patología , Linfocitos T CD4-Positivos/inmunología , Coristoma/inmunología , Coristoma/patología , Enfermedad Crónica , Rechazo de Injerto/inmunología , Isoanticuerpos/inmunología , Tejido Linfoide/irrigación sanguínea , Tejido Linfoide/inmunología , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/inmunología , Linfotoxina beta/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/inmunología , Miocardio/patología , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Bazo/inmunología , Bazo/patología , Trasplante Homólogo
20.
Philos Trans R Soc Lond B Biol Sci ; 366(1575): 2312-22, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21727137

RESUMEN

Recent advances in stem cell technology have generated enthusiasm for their potential to study and treat a diverse range of human disease. Pluripotent human stem cells for therapeutic use may, in principle, be obtained from two sources: embryonic stem cells (hESCs), which are capable of extensive self-renewal and expansion and have the potential to differentiate into any somatic tissue, and induced pluripotent stem cells (iPSCs), which are derived from differentiated tissue such as adult skin fibroblasts and appear to have the same properties and potential, but their generation is not dependent upon a source of embryos. The likelihood that clinical transplantation of hESC- or iPSC-derived tissues from an unrelated (allogeneic) donor that express foreign human leucocyte antigens (HLA) may undergo immunological rejection requires the formulation of strategies to attenuate the host immune response to transplanted tissue. In clinical practice, individualized iPSC tissue derived from the intended recipient offers the possibility of personalized stem cell therapy in which graft rejection would not occur, but the logistics of achieving this on a large scale are problematic owing to relatively inefficient reprogramming techniques and high costs. The creation of stem cell banks comprising HLA-typed hESCs and iPSCs is a strategy that is proposed to overcome the immunological barrier by providing HLA-matched (histocompatible) tissue for the target population. Estimates have shown that a stem cell bank containing around 10 highly selected cell lines with conserved homozygous HLA haplotypes would provide matched tissue for the majority of the UK population. These simulations have practical, financial, political and ethical implications for the establishment and design of stem cell banks incorporating cell lines with HLA types that are compatible with different ethnic populations throughout the world.


Asunto(s)
Bancos de Muestras Biológicas , Células Madre Embrionarias/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Histocompatibilidad , Humanos , Manejo de Especímenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...