Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Am Soc Nephrol ; 31(3): 517-531, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31988271

RESUMEN

BACKGROUND: In donor kidneys subjected to ischemia-reperfusion injury during kidney transplant, phagocytes coexpressing the F4/80 and CD11c molecules mediate proinflammatory responses and trigger adaptive immunity in transplantation through antigen presentation. After injury, however, resident renal macrophages coexpressing these surface markers acquire a proreparative phenotype, which is pivotal in controlling inflammation and fibrosis. No data are currently available regarding the effects of transplant-induced ischemia-reperfusion injury on the ability of donor-derived resident renal macrophages to act as professional antigen-presenting cells. METHODS: We evaluated the phenotype and function of intragraft CD11c+F4/80+ renal macrophages after cold ischemia. We also assessed the modifications of donor renal macrophages after reversible ischemia-reperfusion injury in a mouse model of congeneic renal transplantation. To investigate the role played by IL-1R8, we conducted in vitro and in vivo studies comparing cells and grafts from wild-type and IL-R8-deficient donors. RESULTS: Cold ischemia and reversible ischemia-reperfusion injury dampened antigen presentation by renal macrophages, skewed their polarization toward the M2 phenotype, and increased surface expression of IL-1R8, diminishing activation mediated by toll-like receptor 4. Ischemic IL-1R8-deficient donor renal macrophages acquired an M1 phenotype, effectively induced IFNγ and IL-17 responses, and failed to orchestrate tissue repair, resulting in severe graft fibrosis and aberrant humoral immune responses. CONCLUSIONS: IL-1R8 is a key regulator of donor renal macrophage functions after ischemia-reperfusion injury, crucial to guiding the phenotype and antigen-presenting role of these cells. It may therefore represent an intriguing pathway to explore with respect to modulating responses against autoantigens and alloantigens after kidney transplant.


Asunto(s)
Inmunidad Adaptativa/genética , Antígeno CD11c/inmunología , Trasplante de Riñón/efectos adversos , Receptores de Interleucina-1/genética , Daño por Reperfusión/genética , Animales , Presentación de Antígeno , Antígeno CD11c/metabolismo , Células Cultivadas , Isquemia Fría/métodos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Trasplante de Riñón/métodos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Receptores de Interleucina-1/inmunología , Daño por Reperfusión/prevención & control , Sensibilidad y Especificidad , Transducción de Señal/genética
2.
Nephron ; 144(1): 38-48, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31550726

RESUMEN

BACKGROUND: Tissue fibrosis is the final common phase of chronic allograft injury, the leading cause of late graft loss in kidney transplantation. Preclinical evidence points to the involvement of lysophosphatidic acid (LPA), a bioactive phospholipid, in the development of renal fibrosis. OBJECTIVES: We assessed whether treatment with an orally available inhibitor of autotaxin (ATXi), the main LPA-producing enzyme, could slow the progression of chronic allograft injury in a fully major histocompatibility complex-mismatched rat kidney transplant model and compared its effects with those of the angiotensin-converting enzyme inhibitor lisinopril. METHODS: Kidney allograft recipients were given ciclosporin for the first 15 postoperative days to prevent early acute rejection. Thereafter, they received either no treatment or ATXi or lisinopril and were followed for 180 days after transplantation. RESULTS: Renal LPA levels were increased in allograft rats, providing the rationale for using ATXi in this model. Chronic treatment with ATXi or lisinopril limited progressive proteinuria and ameliorated tubulointerstitial fibrosis compared with allograft rats, although the effects were more robust under ATX inhibition. The administration of ATXi, but not lisinopril, attenuated systemic hypertension, reduced intragraft T cell infiltration, and eventually improved renal graft survival. CONCLUSIONS: In summary, ATXi had protective effects on indices of chronic allograft injury and could be of therapeutic add-on value in the kidney transplant setting. Notably, an ATX inhibitor is currently being investigated in 2 large phase 3 studies in idiopathic pulmonary fibrosis, underscoring the clinical relevance of our findings.


Asunto(s)
Trasplante de Riñón/efectos adversos , Hidrolasas Diéster Fosfóricas/efectos de los fármacos , Trasplante Homólogo/efectos adversos , Animales , Enfermedad Crónica , Rechazo de Injerto , Riñón/metabolismo , Lisofosfolípidos/metabolismo , Masculino , Ratas , Ratas Endogámicas
3.
Transplantation ; 103(6): 1121-1130, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30801518

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs) have protolerogenic effects in renal transplantation, but they induce long-term regulatory T cells (Treg)-dependent graft acceptance only when infused before transplantation. When given posttransplant, MSCs home to the graft where they promote engraftment syndrome and do not induce Treg. Unfortunately, pretransplant MSC administration is unfeasible in deceased-donor kidney transplantation. METHODS: To make MSCs a therapeutic option also for deceased organ recipients, we tested whether MSC infusion at the time of transplant (day 0) or posttransplant (day 2) together with inhibition of complement receptors prevents engraftment syndrome and allows their homing to secondary lymphoid organs for promoting tolerance. We analyzed intragraft and splenic MSC localization, graft survival, and alloimmune response in mice recipients of kidney allografts and syngeneic MSCs given on day 0 or on posttransplant day 2. C3a receptor (C3aR) or C5a receptor (C5aR) antagonists were administered to mice in combination with the cells or were used together to treat MSCs before infusion. RESULTS: Syngeneic MSCs given at day 0 homed to the spleen increased Treg numbers and induced long-term graft acceptance. Posttransplant MSC infusion, combined with a short course of C3aR or C5aR antagonist or administration of MSCs pretreated with C3aR and C5aR antagonists, prevented intragraft recruitment of MSCs and graft inflammation, inhibited antidonor T-cell reactivity, but failed to induce Treg, resulting in mild prolongation of graft survival. CONCLUSIONS: These data support testing the safety/efficacy profile of administering MSCs on the day of transplant in deceased-donor transplant recipients and indicate that complement is crucial for MSC recruitment into the kidney allograft.


Asunto(s)
Inactivadores del Complemento/administración & dosificación , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Riñón , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Receptores de Complemento/antagonistas & inhibidores , Tolerancia al Trasplante/efectos de los fármacos , Animales , Esquema de Medicación , Femenino , Rechazo de Injerto/inmunología , Células Madre Mesenquimatosas/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores de Complemento/inmunología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/inmunología , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Factores de Tiempo , Trasplante Homólogo , Trasplante Isogénico
4.
Kidney Int ; 81(9): 903-18, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22318420

RESUMEN

Anemia can contribute to chronic allograft injury by limiting oxygen delivery to tissues, particularly in the tubulointerstitium. To determine mechanisms by which erythropoietin (EPO) prevents chronic allograft injury we utilized a rat model of full MHC-mismatched kidney transplantation (Wistar Furth donor and Lewis recipients) with removal of the native kidneys. EPO treatment entirely corrected post-transplant anemia. Control rats developed progressive proteinuria and graft dysfunction, tubulointerstitial damage, inflammatory cell infiltration, and glomerulosclerosis, all prevented by EPO. Normalization of post-transplant hemoglobin levels by blood transfusions, however, had no impact on chronic allograft injury, indicating that EPO-mediated graft protection went beyond the correction of anemia. Compared to syngeneic grafts, control allografts had loss of peritubular capillaries, higher tubular apoptosis, tubular and glomerular oxidative injury, and reduced expression of podocyte nephrin; all prevented by EPO treatment. The effects of EPO were associated with preservation of intragraft expression of angiogenic factors, upregulation of the anti-apoptotic factor p-Akt in tubuli, and increased expression of Bcl-2. Inhibition of p-Akt by Wortmannin partially antagonized the effect of EPO on allograft injury and tubular apoptosis, and prevented EPO-induced Bcl-2 upregulation. Thus non-erythropoietic derivatives of EPO may be useful to prevent chronic renal allograft injury.


Asunto(s)
Anemia/tratamiento farmacológico , Eritropoyetina/farmacología , Hematínicos/farmacología , Enfermedades Renales/prevención & control , Trasplante de Riñón/efectos adversos , Riñón/efectos de los fármacos , Anemia/sangre , Anemia/etiología , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/sangre , Transfusión Sanguínea , Enfermedad Crónica , Glomerulonefritis/inmunología , Glomerulonefritis/prevención & control , Hemoglobinas/metabolismo , Histocompatibilidad , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/inmunología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Trasplante de Riñón/inmunología , Ratones , Disfunción Primaria del Injerto/inmunología , Disfunción Primaria del Injerto/prevención & control , Proteinuria/inmunología , Proteinuria/prevención & control , Ratas , Ratas Endogámicas Lew , Ratas Endogámicas WF , Factores de Tiempo
5.
Transpl Int ; 25(3): 347-56, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22239163

RESUMEN

One of the leading causes of long-term kidney graft loss is chronic allograft injury (CAI), a pathological process triggered by alloantigen-dependent and alloantigen-independent factors. Alloantigen-independent factors, such as cold ischemia (CI) may amplify the recipient immune response against the graft. We investigated the impact of prolonged cold ischemia and the subsequent delayed graft function on CAI in a fully MHC-mismatched rat model of kidney allotransplantation. Prolonged CI was associated with anticipation of proteinuria onset and graft function deterioration (ischemia: 90d; no ischemia: 150d), more severe tubular atrophy, interstitial fibrosis, and glomerulosclerosis, and increased mortality rate (180d survival, ischemia: 0%; no ischemia: 67%). In ischemic allografts, T and B cells were detected very early and were organized in inflammatory clusters. Higher expression of BAFF-R and TACI within the ischemic allografts indicates that B cells are mature and activated. As a consequence of B cell activity, anti-donor antibodies, glomerular C4d and IgG deposition, important features of chronic humoral rejection, appeared earlier in ischemic than in non-ischemic allograft recipients. Thus, prolonged CI time plays a main role in CAI development by triggering acceleration of cellular and humoral reactions of chronic rejection. Limiting CI time should be considered as a main target in kidney transplantation.


Asunto(s)
Isquemia Fría/efectos adversos , Funcionamiento Retardado del Injerto/etiología , Rechazo de Injerto/etiología , Trasplante de Riñón/inmunología , Animales , Apoptosis , Funcionamiento Retardado del Injerto/inmunología , Funcionamiento Retardado del Injerto/patología , Citometría de Flujo , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Etiquetado Corte-Fin in Situ , Riñón/inmunología , Riñón/patología , Trasplante de Riñón/métodos , Trasplante de Riñón/mortalidad , Masculino , Ratas , Ratas Endogámicas WF , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Tiempo , Trasplante Homólogo/inmunología , Trasplante Homólogo/métodos , Trasplante Homólogo/mortalidad
6.
Transplantation ; 92(3): 271-9, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21705971

RESUMEN

BACKGROUND: Ischemia/reperfusion (I/R) injury is an important cause of renal graft dysfunction. Increases in cold and warm ischemia times lead to a higher risk of early posttransplant complications including delayed graft function and acute rejection. Moreover, prolonged cold ischemia is a predictor of long-term graft loss in kidney transplant patients. METHODS: Darbepoetin alfa (DA) and carbamylated nonerythropoietic derivative of erythropoietin (CEPO) protective effects were evaluated in a model of I/R injury after kidney transplantation in both syngeneic and allogeneic combinations. The effects of wortmannin (phosphorylated Akt [p-Akt] inhibitor) administration were also investigated. Serum creatinine was evaluated at 16, 24, 48 hr and at 4 and 7 days posttransplant. Animals were killed 24 hr or 7 days after transplant and kidneys were processed for histological analysis, immunohistochemistry assessment of erythropoietin receptor (EPOR) and ß-common chain receptor expression, granulocyte infiltration, nitrotyrosine staining, p-Akt expression, peritubular capillary (PTC) density, apoptosis, antioxidant, and antiapoptotic gene expression. RESULTS: DA and CEPO significantly reduced serum creatinine, tubular injury, tubular nitrotyrosine staining, and prevented I/R-induced tubular apoptosis, but only when given both to the donor and to the recipient. DA and CEPO cytoprotection was associated with prevention of I/R-induced drop of p-Akt expression in tubuli, and almost complete preservation of capillary density in the tubulointerstitium of the graft. CEPO was more effective than DA in reducing tubular oxidative stress and preserving PTCs. CONCLUSION: DA and CEPO when given both to the donor and to the recipient, prevented renal graft dysfunction, tubular oxidative stress, and apoptosis after I/R injury in kidney transplantation. Their cytoprotection was mediated by tubular p-Akt activation and PTC density preservation.


Asunto(s)
Eritropoyetina/análogos & derivados , Hematínicos/farmacología , Trasplante de Riñón , Disfunción Primaria del Injerto/prevención & control , Daño por Reperfusión/tratamiento farmacológico , Androstadienos/farmacología , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Frío , Darbepoetina alfa , Eritropoyetina/farmacología , Supervivencia de Injerto/efectos de los fármacos , Inmunosupresores/farmacología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Estrés Oxidativo/efectos de los fármacos , Disfunción Primaria del Injerto/metabolismo , Disfunción Primaria del Injerto/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Endogámicas Lew , Donantes de Tejidos , Trasplante Homólogo , Wortmanina
7.
Transpl Int ; 24(8): 829-38, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21545548

RESUMEN

Ischemia/reperfusion (I/R) injury is an important cause of renal graft dysfunction in humans. Increases in cold and warm ischemia times lead to a higher risk of early post-transplant complications including delayed graft function and acute rejection. Moreover, prolonged cold ischemia is a predictor of long-term kidney graft loss. The protective effect of rabbit anti-rat thymocyte immunoglobulin (rATG) was evaluated in a rat model of I/R injury following syngeneic kidney transplantation. Serum creatinine concentration was evaluated at 16 h and 24 h post-transplant. Animals were sacrificed 24 h post-transplant for evaluation of histology, infiltrating leukocytes, nitrotyrosine staining, and apoptosis. rATG was effective in preventing renal function impairment, tissue damage and tubular apoptosis associated with I/R only when was given 2 h before transplantation but not at the time of reperfusion. Pretransplant rATG treatment of recipient animals effectively reduced the amount of macrophages, CD4(+), CD8(+) T cells and LFA-1(+) cells infiltrating renal graft subjected to cold ischemia as well as granzyme-B expression within ischemic kidney. On the other hand, granulocyte infiltration and oxidative stress were not modified by rATG. If these results will be translated into the clinical setting, pretransplant administration of Thymoglobuline(®) could offer the additional advantage over peri-transplant administration of limiting I/R-mediated kidney graft damage.


Asunto(s)
Inmunoglobulinas/metabolismo , Trasplante de Riñón/métodos , Riñón/metabolismo , Daño por Reperfusión/metabolismo , Timocitos/citología , Animales , Apoptosis , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Granzimas/farmacología , Isquemia/patología , Masculino , Microscopía Fluorescente/métodos , Conejos , Ratas , Ratas Endogámicas Lew
8.
J Immunol ; 186(7): 4164-74, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21389254

RESUMEN

Tolerance induction toward allogeneic organ grafts represents one of the major aims of transplantation medicine. Stem cells are promising candidates for promoting donor-specific tolerance. In this study, we investigated the immunomodulatory properties of murine embryonic stem cells (ESCs), obtained either by in vitro fertilization (IVF-ESCs) or by nuclear transfer (NT-ESCs), in heart transplant mouse models. IVF-ESCs did not prolong the survival of fully allogeneic cardiac transplants but significantly prolonged the survival of semiallogeneic hearts from the same ESC donor strain for >100 d in 44% of the animals. However, 28% of transplanted animals infused with IVF-ESCs experienced development of a teratoma. NT-ESCs similarly prolonged semiallogeneic heart graft survival (>100 d in 40% of the animals) but were less teratogenic. By in vitro studies, IVF-ESC and NT-ESC immunoregulation was mediated both by cell contact-dependent mechanisms and by the release of soluble factors. By adding specific inhibitors, we identified PGE(2) as a soluble mediator of ESC immunoregulation. Expansion of regulatory T cells was found in lymphoid organs and in the grafts of IVF-ESC- and NT-ESC-tolerized mice. Our study demonstrates that both IVF-ESCs and NT-ESCs modulate recipient immune response toward tolerance to solid organ transplantation, and that NT-ESCs exhibit a lower tendency for teratoma formation. Because NT-ESCs are obtained by NT of a somatic cell from living individuals into an enucleated oocyte, they could represent a source of donor-derived stem cells to induce tolerance to solid organ allograft.


Asunto(s)
Células Madre Embrionarias/inmunología , Células Madre Embrionarias/trasplante , Fertilización In Vitro , Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Proteínas Nucleares/administración & dosificación , Traslado Adoptivo , Animales , Línea Celular , Femenino , Rechazo de Injerto/prevención & control , Trasplante de Corazón/métodos , Trasplante de Corazón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Distribución Aleatoria , Trasplante Homólogo/inmunología , Trasplante Homólogo/patología
9.
J Immunol ; 183(7): 4249-60, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19734209

RESUMEN

Members of the TLR/IL-1R superfamily mediate ischemia/reperfusion injury and initiate immune response in transplanted organs. In this study, we tested the hypothesis that Toll-IL-1R8 (TIR8), a negative regulator of TLR/IL-1R highly expressed in the kidney, modulates immune cell activation underlying kidney rejection. In a mouse model of fully mismatched kidney allotransplantation in which the graft is spontaneously accepted, intragraft Tir8 expression was enhanced compared with naive kidneys. Targeted deletion of Tir8 in the graft exerted a powerful antitolerogenic action leading to acute rejection. Similarly, in a mouse model of kidney graft acceptance induced by costimulation blockade, most Tir8(-/-) grafts were acutely rejected. Despite similar levels of TLR4, IL-1R, and their ligands, the posttransplant ischemia/reperfusion-induced inflammatory response was more severe in Tir8(-/-) than in Tir8(+/+) grafts and was followed by expansion and maturation of resident dendritic cell precursors. In vitro, Tir8(-/-) dendritic cell precursors acquired higher allostimulatory activity and released more IL-6 upon stimulation with a TLR4 ligand and TNF-alpha than Tir8(+/+) cells, which may explain the increased frequency of antidonor-reactive T cells and the block of regulatory T cell formation in recipients of a Tir8(-/-) kidney. Thus, TIR8 acts locally as a key regulator of allogeneic immune response in the kidney. Tir8 expression and/or signaling in donor tissue are envisaged as a novel target for control of innate immunity and amelioration of graft survival.


Asunto(s)
Regulación hacia Abajo/inmunología , Rechazo de Injerto/inmunología , Trasplante de Riñón/inmunología , Glicoproteínas de Membrana/fisiología , Receptores de Interleucina-1/fisiología , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Rechazo de Injerto/genética , Rechazo de Injerto/prevención & control , Tolerancia Inmunológica/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Ratones Noqueados , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Daño por Reperfusión/genética , Daño por Reperfusión/inmunología
10.
J Immunol ; 181(6): 3933-46, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18768848

RESUMEN

In this study, we investigated whether mesenchymal stem cells (MSC) had immunomodulatory properties in solid organ allotransplantation, using a semiallogeneic heart transplant mouse model, and studied the mechanism(s) underlying MSC tolerogenic effects. Either single (portal vein, day -7) or double (portal vein, day -7 and tail vein, day -1) pretransplant infusions of donor-derived B6C3 MSC in B6 recipients induced a profound T cell hyporesponsiveness and prolonged B6C3 cardiac allograft survival. The protolerogenic effect was abrogated when donor-derived MSC were injected together with B6C3 hematopoietic stem cells (HSC), suggesting that HSC negatively impact MSC immunomodulatory properties. Both the induction (pretransplant) and the maintenance phase (>100 days posttransplant) of donor-derived MSC-induced tolerance were associated with CD4(+)CD25(+)Foxp3(+) Treg expansion and impaired anti-donor Th1 activity. MSC-induced regulatory T cells (Treg) were donor-specific since adoptive transfer of splenocytes from tolerant mice prevented the rejection of fully MHC-mismatched donor-specific secondary allografts but not of third-party grafts. In addition, infusion of recipient-derived B6 MSC tolerized a semiallogeneic B6C3 cardiac allograft, but not a fully MHC-mismatched BALB/c graft, and expanded Treg. A double i.v. pretransplant infusion of recipient-derived MSC had the same tolerogenic effect as the combined intraportal/i.v. MSC infusions, which makes the tolerogenic protocol applicable in a clinical setting. In contrast, single MSC infusions given either peritransplant or 1 day after transplant were less effective. Altogether these findings indicate that MSC immunomodulatory properties require HSC removal, partial sharing of MHC Ags between the donor and the recipient and pretransplant infusion, and are associated with expansion of donor-specific Treg.


Asunto(s)
Diferenciación Celular/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Trasplante de Células Madre Mesenquimatosas , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Acondicionamiento Pretrasplante , Tolerancia al Trasplante/inmunología , Animales , Células de la Médula Ósea/inmunología , Femenino , Trasplante de Corazón/métodos , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Acondicionamiento Pretrasplante/métodos , Trasplante Heterotópico , Trasplante Homólogo , Trasplante Isogénico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...