Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Xenotransplantation ; 27(1): e12552, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31544995

RESUMEN

BACKGROUND: We have recently demonstrated that human-CD47 (hCD47) expressed on endothelial cells of porcine lung xenografts extended median graft survival from 3.5 days to 8.7 days in baboons. Intra-bone bone marrow transplantation (IBBMTx) in a pig-to-baboon model was previously shown to markedly prolong the duration of macrochimerism up to 21 days from 1 to 4 days by intravenous BMTx. We now examined whether the use of hCD47 transgenic (Tg) BM further prolonged the duration of chimerism following IBBMTx. We then tested if lung xenograft survival was prolonged following IBBMTx. METHODS: Baboons received GalTKO-hCD47/hCD55Tg (n = 5) or -hCD55Tg (n = 1) or -hCD46/HLA-E Tg (n = 1) pig IBBMTx. Macrochimerism, anti-pig T cells and antibody responses were assessed. Animals received lung xenografts from either hCD47+ or hCD47- porcine lungs 1-3 months later. RESULTS: All baboons that received hCD47Tg porcine IBBM maintained durable macrochimerism >30 days, and two maintained chimerism for >8 weeks. Notably, anti-pig antibody levels decreased over time and anti-pig cellular unresponsiveness developed following IBBMTx. Lungs from hCD47Tg IBBMTx matched pigs were transplanted at day 33 or day 49 after IBBMTx. These animals showed extended survival up to 13 and 14 days, while animals that received lungs from hCD47 negative pigs displayed no prolonged survival (1-4 days). CONCLUSION: This is the first report demonstrating durable macrochimerism beyond 8 weeks, as well as evidence for B cell tolerance in large animal xenotransplantation. Using hCD47Tg pigs as both IBBMTx and lung donors prolongs lung xenograft survival. However, additional strategies are required to control the acute loss of lung xenografts.


Asunto(s)
Linfocitos B/inmunología , Trasplante de Médula Ósea , Antígeno CD47/metabolismo , Trasplante de Pulmón , Animales , Animales Modificados Genéticamente , Médula Ósea/cirugía , Antígeno CD47/genética , Células Cultivadas , Quimerismo , Supervivencia de Injerto , Humanos , Tolerancia Inmunológica , Papio , Porcinos , Trasplante Heterólogo
2.
Curr Opin Organ Transplant ; 24(5): 517-521, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31385887

RESUMEN

PURPOSE OF REVIEW: Xenotransplantation offers the opportunity to alleviate the imbalance between the demand of patients with end stage organ failure and the supply of organs available for transplantation but remains aspirational. This review highlights how collaboration between academia and industry are essential for success. RECENT FINDINGS: The science of xenotransplantation has accelerated in recent years with key discoveries in genetic engineering, enabling disruption of genes facilitating rejection, and transgenic expression of desired human genes. Combined with similar progress directed toward induction of transplant tolerance, the stage has been set for meaningful progress. These advances are reviewed in detail elsewhere in this volume and argue that the breakthroughs needed to deliver substantial cross-species organ survival have largely been achieved, heralding a liminal stage of human xenotransplantation. However, xenotransplantation as a meaningful therapy for medically refractory end organ failure will not be realized through scientific innovation alone. The advent of broadly available, therapeutic xenogeneic tissues requires extensive development and regulatory expertise; the biotechnology/pharmaceutical industry can provide extensive resources and expertise in those essential areas. SUMMARY: Successful delivery of xenotransplantation as an available therapy for curing end stage organ failure is best accomplished through partnership and collaboration between academia and industry.


Asunto(s)
Colaboración Intersectorial , Trasplante de Órganos , Donantes de Tejidos/provisión & distribución , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente , Rechazo de Injerto , Humanos , Obtención de Tejidos y Órganos , Tolerancia al Trasplante
3.
Xenotransplantation ; 25(5): e12391, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29527745

RESUMEN

BACKGROUND: Despite recent progress in survival times of xenografts in non-human primates, there are no reports of survival beyond 5 days of histologically well-aerated porcine lung grafts in baboons. Here, we report our initial results of pig-to-baboon xeno-lung transplantation (XLTx). METHODS: Eleven baboons received genetically modified porcine left lungs from either GalT-KO alone (n = 3), GalT-KO/humanCD47(hCD47)/hCD55 (n = 3), GalT-KO/hD47/hCD46 (n = 4), or GalT-KO/hCD39/hCD46/hCD55/TBM/EPCR (n = 1) swine. The first 2 XLTx procedures were performed under a non-survival protocol that allowed a 72-hour follow-up of the recipients with general anesthesia, while the remaining 9 underwent a survival protocol with the intention of weaning from ventilation. RESULTS: Lung graft survivals in the 2 non-survival animals were 48 and >72 hours, while survivals in the other 9 were 25 and 28 hours, at 5, 5, 6, 7, >7, 9, and 10 days. One baboon with graft survival >7 days, whose entire lung graft remained well aerated, was euthanized on POD 7 due to malfunction of femoral catheters. hCD47 expression of donor lungs was detected in both alveoli and vessels only in the 3 grafts surviving >7, 9, and 10 days. All other grafts lacked hCD47 expression in endothelial cells and were completely rejected with diffuse hemorrhagic changes and antibody/complement deposition detected in association with early graft loss. CONCLUSIONS: To our knowledge, this is the first evidence of histologically viable porcine lung grafts beyond 7 days in baboons. Our results indicate that GalT-KO pig lungs are highly susceptible to acute humoral rejection and that this may be mitigated by transgenic expression of hCD47.


Asunto(s)
Animales Modificados Genéticamente/inmunología , Antígeno CD47/inmunología , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Papio/inmunología , Animales , Rechazo de Injerto/patología , Xenoinjertos/inmunología , Humanos , Pulmón/inmunología , Trasplante de Pulmón/métodos , Porcinos , Trasplante Heterólogo/métodos , Trasplantes/inmunología
4.
Circulation ; 124(2): 196-205, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21690493

RESUMEN

BACKGROUND: Ligands activating the transcription factor peroxisome proliferator-activated receptor-γ (PPARγ) have antiinflammatory effects. Vascular rejection induced by allogeneic T cells can be responsible for acute and chronic graft loss. Studies in rodents suggest that PPARγ agonists may inhibit graft vascular rejection, but human T-cell responses to allogeneic vascular cells differ from those in rodents, and the effects of PPARγ in human transplantation are unknown. METHODS AND RESULTS: We tested the effects of PPARγ agonists on human vascular graft rejection using a model in which human artery is interposed into the abdominal aorta of immunodeficient mice, followed by adoptive transfer of allogeneic (to the artery donor) human peripheral blood mononuclear cells. Interferon-γ-dependent rejection ensues within 4 weeks, characterized by intimal thickening, T-cell infiltrates, and vascular cell activation, a response resembling clinical intimal arteritis. The PPARγ agonists 15-deoxy-prostaglandin-J(2), ciglitazone, and pioglitazone reduced intimal expansion, intimal infiltration of CD45RO(+) memory T cells, and plasma levels of inflammatory cytokines. The PPARγ antagonist GW9662 reversed the protective effects of PPARγ agonists, confirming the involvement of PPARγ-mediated pathways. In vitro, pioglitazone inhibited both alloantigen-induced proliferation and superantigen-induced transendothelial migration of memory T cells, indicating the potential mechanisms of PPARγ effects. CONCLUSION: Our results suggest that PPARγ agonists inhibit allogeneic human memory T cell responses and may be useful for the treatment of vascular graft rejection.


Asunto(s)
Arterias/inmunología , Arterias/trasplante , Rechazo de Injerto/inmunología , Hipoglucemiantes/farmacología , PPAR gamma/agonistas , Prostaglandina D2/análogos & derivados , Linfocitos T/inmunología , Tiazolidinedionas/farmacología , Traslado Adoptivo , Anilidas/farmacología , Animales , Arterias/patología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Proliferación Celular/efectos de los fármacos , Citocinas/inmunología , Rechazo de Injerto/tratamiento farmacológico , Rechazo de Injerto/patología , Humanos , Memoria Inmunológica/efectos de los fármacos , Memoria Inmunológica/inmunología , Isoantígenos/inmunología , Ratones , Ratones SCID , PPAR gamma/antagonistas & inhibidores , PPAR gamma/inmunología , Pioglitazona , Prostaglandina D2/farmacología , Superantígenos/farmacología , Linfocitos T/patología , Linfocitos T/trasplante , Trasplante Heterólogo , Trasplante Homólogo
5.
J Clin Invest ; 114(6): 846-56, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15372109

RESUMEN

Allograft vascular dysfunction predisposes to arteriosclerosis and graft loss. We examined how dysfunction develops in transplanted human arteries in response to circulating allogeneic T cells in vivo using immunodeficient murine hosts. Within 7-9 days, transplanted arteries developed endothelial cell (EC) dysfunction but remained sensitive to exogenous NO. By 2 weeks, the grafts developed impaired contractility and desensitization to NO, both signs of VSMC dysfunction. These T cell-dependent changes correlated with loss of eNOS and expression of iNOS--the latter predominantly within infiltrating T cells. Neutralizing IFN-gamma completely prevented both vascular dysfunction and changes in NOS expression; neutralizing TNF reduced IFN-gamma production and partially prevented dysfunction. Inhibiting iNOS partially preserved responses to NO at 2 weeks and reduced graft intimal expansion after 4 weeks in vivo. In vitro, memory CD4+ T cells acted on allogeneic cultured ECs to reduce eNOS activity and expression of protein and mRNA. These effects required T cell activation by class II MHC antigens and costimulators (principally lymphocyte function-associated antigen-3, or LFA-3) on the ECs and were mediated by production of soluble mediators including IFN-gamma and TNF. We conclude that IFN-gamma is a central mediator of vascular dysfunction and, through dysregulation of NOS expression, links early dysfunction with late arteriosclerosis.


Asunto(s)
Arterias/trasplante , Endotelio Vascular/patología , Interferón gamma/fisiología , Óxido Nítrico Sintasa/genética , Linfocitos T/inmunología , Trasplante Homólogo/fisiología , Animales , Arterias/inmunología , Arteriosclerosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Endotelio Vascular/inmunología , Humanos , Ratones , Ratones SCID , Óxido Nítrico Sintasa de Tipo II , Trasplante Heterólogo/inmunología , Trasplante Heterólogo/patología , Trasplante Homólogo/inmunología
6.
Transplantation ; 83(11): 1501-5, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17565324

RESUMEN

Vascular remodeling rather than intimal thickening is the most important determinant of luminal loss in cardiac graft arteriosclerosis. The impact of donor-transmitted atherosclerotic lesions on alloimmune-mediated arterial injury in an experimental setting is not known. We investigated this issue in a chimeric model of human coronary artery grafts to immunodeficient mouse recipients reconstituted with allogeneic human peripheral blood mononuclear cells. Rejecting grafts demonstrated robust intimal expansion, outward vascular remodeling, and variable lumen loss. There was no significant relationship between preexistent atherosclerosis, gender, and age of the artery donors vs. the degree of alloimmune-induced changes in vessel morphology. Our experimental findings, in a system without the potentially confounding variable of immunosuppressive drugs, are in agreement with the majority of clinical studies that alloimmune-mediated intimal injury and vascular remodeling is independent of preexisting coronary atherosclerosis. Our results support the concept of extending the criteria for organ donors to include modest coronary atherosclerosis.


Asunto(s)
Enfermedad de la Arteria Coronaria/fisiopatología , Vasos Coronarios/fisiopatología , Vasos Coronarios/trasplante , Ratones SCID , Inmunología del Trasplante , Anciano , Animales , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/inmunología , Vasos Coronarios/patología , Femenino , Rechazo de Injerto/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Quimera por Trasplante , Trasplante Heterólogo
7.
Transplantation ; 81(4): 559-66, 2006 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-16495804

RESUMEN

BACKGROUND: Chronic allograft dysfunction may result from arterial injury, manifest as transplant arteriosclerosis (TA). This represents an important factor limiting long-term outcomes after heart and kidney transplantation; a relationship between acute allograft arterial injury and TA has been suggested. We have used SCID/bg mice bearing transplanted human artery, inoculated with allogeneic human PBMC to study arteriopathy in human vessels. Earlier work demonstrated arteriopathy similar to that observed clinically, and identified interferon-gamma as a mediator of the process. This study evaluated whether sirolimus (SRL), with cyclosporine A (CsA) or alone, affects TA, and examined possible mechanisms of action. METHODS: CB17/SCID/bg mice were transplanted with human arteries replacing the abdominal aorta; reconstituted with allogeneic human PBMC. Controls received vehicle alone for comparison with mice given CsA (5 mg/kg/d), SRL (0.1 or 0.5 mg/kg/d), or CsA (5 mg/kg/d) plus SRL (0.1 mg/kg/d). Transplant arteries were examined 28 days later by histology and immunohistochemistry; circulating human interferon-gamma was evaluated by ELISA, and intragraft interferon-gamma mRNA by qRT-PCR. RESULTS: The characteristic TA was modestly reduced by CsA or low-dose SRL, but eliminated by combination CsA plus SRL or higher dose SRL alone. Circulating interferon-gamma was reduced by CsA, but inhibition was dramatic with SRL alone or combined with CsA. Intragraft interferon-gamma and HLA-DR expression were moderately reduced by CsA or SRL, and eliminated with combined CsA plus SRL. CONCLUSIONS: SRL plus CsA prevented allograft arteriopathy, correlating with suppression of intragraft interferon-gamma, suggesting that SRL effects may result from anti-inflammatory consequences from inhibiting interferon-gamma.


Asunto(s)
Arterias/trasplante , Ciclosporina/uso terapéutico , Interferón gamma/fisiología , Sirolimus/uso terapéutico , Trasplante Homólogo/patología , Animales , Arterias/patología , Humanos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Interferón gamma/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/fisiología , Ratones , Ratones SCID , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Trasplante Heterólogo/patología
8.
Transplantation ; 80(2): 244-52, 2005 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-16041270

RESUMEN

BACKGROUND: This 36-month, randomized, parallel-group study compared safety and efficacy of two doses of everolimus with mycophenolate mofetil (MMF) in de novo renal-transplant recipients. METHODS: Renal-allograft recipients received 1.5 mg/day or 3 mg/day of everolimus or 2 g/day of MMF, plus full-dose cyclosporine (CsA) and corticosteroids after randomization. For at least their first year, patients received study medication according to a double-blinded, double-dummy design. Concerns over nephrotoxicity led to a protocol amendment to an open-label design with reduced CsA troughs. RESULTS: Incidences of primary efficacy failure at 36 months (biopsy-proven acute rejection, graft loss, death, or loss to follow-up) were everolimus 1.5 mg/day, 33.7% (65/193); everolimus 3 mg/day, 34.0% (66/194); and MMF, 31.1% (61/196) (P=0.810). Antibody-treated acute rejection at 36 months was significantly lower with everolimus 1.5 mg (9.8%) than MMF (18.4%, P=0.014). Discontinuation for adverse events was more frequent with everolimus and hemolytic uremic syndrome, lymphoproliferative disease, and proteinuria, and higher serum creatinine occurred at increased frequency relative to the MMF arm. Creatinine levels in the everolimus arms were stable in follow-up: the mean rise in creatinine over the first 6 months of the open-label phase was 3 micromol/L or greater with everolimus and 7 micromol/L with MMF. However, serum creatinine levels were lower in the MMF group throughout. Death and graft loss were higher in the everolimus arms (not significant). CONCLUSIONS: As part of triple-drug immunosuppression, everolimus (1.5 or 3 mg/day) was as efficacious as MMF, although the side-effect profile featured increased adverse events. Nephrotoxicity/calcineurin-inhibitor-related adverse events will require judicious lowering of CsA exposure with monitoring of everolimus troughs.


Asunto(s)
Rechazo de Injerto/prevención & control , Inmunosupresores/uso terapéutico , Trasplante de Riñón/inmunología , Ácido Micofenólico/análogos & derivados , Sirolimus/análogos & derivados , Adolescente , Adulto , Anciano , Quimioterapia Combinada , Everolimus , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ácido Micofenólico/uso terapéutico , Seguridad , Sirolimus/uso terapéutico , Donantes de Tejidos/estadística & datos numéricos
9.
FASEB J ; 17(15): 2250-6, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14656987

RESUMEN

Clinical performance of currently available human skin equivalents is limited by failure to develop perfusion. To address this problem we have developed a method of endothelial cell transplantation that promotes vascularization of human skin equivalents in vivo. Enhancement of vascularization by Bcl-2 overexpression was demonstrated by seeding human acellular dermis grafts with human umbilical vein endothelial cells (HUVEC) transduced with the survival gene Bcl-2 or an EGFP control transgene, and subcutaneous implantation in immunodeficient mice (n=18). After 1 month the grafts with Bcl-2-transduced cells contained a significantly greater density of perfused HUVEC-lined microvessels (55.0/mm3) than controls (25.4/mm3,P=0.026). Vascularized skin equivalents were then constructed by sequentially seeding the apical and basal surfaces of acellular dermis with cultured human keratinocytes and Bcl-2-transduced HUVEC, respectively. Two weeks after orthotopic implantation onto mice, 75% of grafts (n=16) displayed both a differentiated human epidermis and perfusion through HUVEC-lined microvessels. These vessels, which showed evidence of progressive maturation, accelerated the rate of graft vascularization. Successful transplantation of such vascularized human skin equivalents should enhance clinical utility, especially in recipients with impaired angiogenesis.


Asunto(s)
Trasplante de Piel , Piel/irrigación sanguínea , Animales , Trasplante de Células , Células Cultivadas , Dermis/anatomía & histología , Dermis/irrigación sanguínea , Endotelio Vascular/citología , Humanos , Ratones , Ratones SCID , Proteínas Proto-Oncogénicas c-bcl-2/genética , Piel/anatomía & histología , Transducción Genética
10.
FASEB J ; 18(3): 606-8, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14734640

RESUMEN

Vascular remodeling (change in vessel diameter) rather than intimal hyperplasia is the most important predictor of luminal loss in immune-mediated arterial injury, yet little is known about its mechanisms. Here, we show that outward vascular remodeling and intimal thickening, two manifestations of arteriosclerosis with opposing effects on luminal size, result from immune effector mechanisms that are T-cell dependent and interferon (IFN)-gamma mediated. In our in vivo model of human coronary artery injury by allogeneic peripheral blood mononuclear cells, both processes occur concurrently and are characterized by T-cell infiltrates with a predominantly IFN-gamma-producing cytokine profile. Neutralization of IFN-gamma inhibits the arterial and intimal expansion, whereas administration of IFN-gamma enhances these effects. The nonredundant role of IFN-gamma in T-cell-dependent remodeling of human coronary arteries demonstrated here presents a new therapeutic target for preservation of vessel lumen in arteriosclerosis.


Asunto(s)
Vasos Coronarios/inmunología , Rechazo de Injerto/inmunología , Interferón gamma/fisiología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Aorta Abdominal/cirugía , Vasos Coronarios/patología , Vasos Coronarios/trasplante , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Interferón gamma/antagonistas & inhibidores , Interferón gamma/farmacología , Ratones , Ratones Mutantes , Ratones SCID , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/trasplante , Quimera por Trasplante , Trasplante Heterólogo , Túnica Íntima/patología
11.
Int Urol Nephrol ; 36(3): 451-6, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15783123

RESUMEN

Strict consideration of the renal transplant candidate's chronologic age is generally supplanted by more subjective reflection on his (her) physiologic state. In the US, patients over 64 years old represented 9.0% of renal transplant recipients in the year 2000, yet little prior experience is available with which to guide the management of geriatric patients. Two hundred and forty six consecutive recipients of primary kidney transplants at the Yale-New Haven Organ Transplant Center between 1990 and 1995 were included in an outcome analysis. Age at transplantation ranged from 2 to 68 years; the study group consisted of the 16 (6.5%) over age 60. The immunosuppressive protocol was uniform for all patients. There was a disproportionately high use of cadaveric organs by older patients; only 1/16 (6.3%) received a living donor kidney. The overall rate of rejection within the first 90 days was 6.7% of cadaveric recipients over 60 versus 37.6% of younger recipients, P=0.001. Actual patient survival rates at 6 years were 100% of patients younger than 11 years versus 69% (11/16) of those older than 60 years. Death censored 5 year graft survival was 100% in older patients versus 85% among the younger patients. The older and younger patients received quantitatively equivalent immunosuppression, but acute rejection was uncommon in the former (6%) versus the younger cohort (34%). It seems logical to consider whether older renal transplant recipients may benefit from a less aggressive immunosuppression strategy.


Asunto(s)
Rechazo de Injerto/inmunología , Terapia de Inmunosupresión , Trasplante de Riñón/inmunología , Adolescente , Adulto , Factores de Edad , Anciano , Niño , Preescolar , Humanos , Persona de Mediana Edad , Factores de Riesgo
14.
Transplantation ; 89(11): 1347-53, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20216481

RESUMEN

BACKGROUND: The reasons for kidney allograft failure subsequent to pancreas after kidney (PAK) are multifactorial; therefore, we examined these factors to identify a meaningful risk assessment that could assist in patient selection. METHODS: Five transplant centers in New England collaborated for this multiinstitutional retrospective study of 126 PAK transplantation recipients who had a functioning pancreas allograft 7 days after transplantation. Host factors (age at pancreas transplant, gender, body weight, glomerular filtration rate at 3 months pre-PAK and at 3-, 6-, 9-, and 12-month post-PAK, presence of proteinuria, pre- or post-PAK kidney rejection, pancreas rejection, cytomegalovirus disease, and HbA1C at 6-month post-PAK) and transplant factors (time to PAK, use of induction antibody therapy, and combinations of immunosuppressive medications) were assessed in both univariate and multivariate analyses for the primary outcome of kidney allograft failure. RESULTS: Of the variables assessed, factors associated with kidney allograft loss after PAK include impaired renal function in the 3 months before PAK, proteinuria, the occurrence of a post-PAK kidney rejection episode, and interval between kidney and pancreas transplantation more than 1 year. CONCLUSIONS: In our analysis, post-PAK kidney allograft loss was strongly associated with glomerular filtration rate less than 45 mL/min pre-PAK, K to P interval of over 1 year, pre-PAK kidney rejection episode, and pre-PAK proteinuria. Diabetic candidates for PAK with any of these conditions should be counseled regarding the risk of post-PAK renal transplant failure.


Asunto(s)
Rechazo de Injerto/epidemiología , Trasplante de Riñón/efectos adversos , Trasplante de Páncreas/fisiología , Insuficiencia del Tratamiento , Adulto , Suero Antilinfocítico/uso terapéutico , Peso Corporal , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/epidemiología , Femenino , Tasa de Filtración Glomerular , Hemoglobina Glucada/metabolismo , Humanos , Inmunosupresores/uso terapéutico , Trasplante de Riñón/mortalidad , Trasplante de Riñón/fisiología , Masculino , Persona de Mediana Edad , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapéutico , Valor Predictivo de las Pruebas , Proteinuria/diagnóstico , Proteinuria/epidemiología , Terapia de Reemplazo Renal , Medición de Riesgo , Factores de Tiempo , Resultado del Tratamiento
18.
Transplantation ; 87(2): 189-97, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19155972

RESUMEN

BACKGROUND: Nearly half of all infiltrating leukocytes in rejecting human allografts are macrophages, yet, in comparison with T cells, much less is known about the contribution of this cell type to rejection. Our laboratory has previously described models of rejection of human skin or artery grafts in immunodeficient mouse hosts mediated by adoptively transferred allogeneic T cells. However, mature human monocyte/macrophages have consistently failed to engraft in these animals. Here, we describe the introduction of human CD68+ macrophages into irradiated immunodeficient mice by transplantation of enriched CD34+ hematopoietic stem-cells isolated from peripheral blood of G-colony-stimulating factor pretreated adults. METHODS: We investigated strains of immunodeficient mice bearing human tissue grafts (skin and artery) inoculated with 1 x 10(6) human CD34+ adult hematopoietic stem cells, peripheral blood monuclear cells autologous to the CD34 donor, or both for human cell engraftment. RESULTS: In the absence of T cells, CD68+ CD14+ macrophages infiltrate allogeneic human skin but produce little injury or thrombosis. Both responses are enhanced when combined with adoptive transfer of T cells autologous to the hematopoietic stem cells as exemplified by the induction of the macrophage activation marker CD163. CD68+ macrophages also infiltrate allogeneic arterial interposition grafts, producing intimal expansion and calcification in the absence of T cells. CONCLUSIONS: These new models may be used to study the role of human macrophages in transplant rejection and other pathologies in vivo.


Asunto(s)
Células Madre Adultas/inmunología , Arterias/inmunología , Arterias/trasplante , Rechazo de Injerto/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Macrófagos/inmunología , Trasplante de Piel/inmunología , Piel/inmunología , Traslado Adoptivo , Animales , Antígenos CD/análisis , Antígenos CD34/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Arterias/patología , Modelos Animales de Enfermedad , Rechazo de Injerto/patología , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/inmunología , Receptores de Lipopolisacáridos/análisis , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Receptores de Superficie Celular/análisis , Piel/patología , Linfocitos T/inmunología , Linfocitos T/trasplante , Trasplante Homólogo , Irradiación Corporal Total
19.
Transplantation ; 88(8): 1010-8, 2009 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19855247

RESUMEN

BACKGROUND.: Sirolimus (SRL) is an important component of clinical immunosuppression in renal transplantation, but few international studies have examined how this agent is used in routine practice. METHODS.: Within a large prospective pharmacoepidemiological study, 718 de novo renal graft recipients treated with SRL in 65 centers in 10 countries were monitored for up to 5 years posttransplant to compare the principal outcomes and adverse effects by treatment regimen. RESULTS.: Principal treatment regimens were SRL without a calcineurin inhibitor (33%), SRL+cyclosporine A (CsA) (33%), and SRL+tacrolimus (TAC) (34%); 18% of subjects discontinued SRL, 124/718 (17%) developed biopsy-confirmed acute rejection (BCAR), 64/718 (9%) lost their graft, and 50/718 (7%) died during follow-up. Calculated creatinine clearance was 66+/-26 mL/min at 2 years. The most common adverse events were hypertension, hyperlipidemia, anemia, urinary tract infections, and diabetes. BCAR was significantly lower in subjects receiving SRL+TAC (hazard ratio [HR] 0.46, P=0.009) but not significantly lower in those receiving SRL+CsA (HR 0.62, P=0.102) compared with SRL without a calcineurin inhibitor. Graft loss or death did not significantly differ between treatment groups but were associated, respectively, with deceased donor grafts (HR 3.33, P<0.001) and increased age (HR 1.04, P<0.001). No improvement was observed in patients receiving mycophenolate mofetil in any treatment combination (HR 0.80, P=0.438 for BCAR; HR 0.93, P=0.849 for graft loss; and HR 0.75, P=0.531 for death). CONCLUSIONS.: SRL is most commonly used in combination with mycophenolate mofetil, CsA, or TAC. BCAR was least common in subjects receiving SRL+TAC, but other outcomes seemed comparable between the treatment regimens in routine practice.


Asunto(s)
Trasplante de Riñón/inmunología , Sirolimus/uso terapéutico , Tacrolimus/uso terapéutico , Algoritmos , Anemia/inducido químicamente , Estudios de Cohortes , Creatinina/metabolismo , Quimioterapia Combinada , Rechazo de Injerto/epidemiología , Humanos , Hiperlipidemias/inducido químicamente , Hipertensión/inducido químicamente , Inmunosupresores/efectos adversos , Inmunosupresores/uso terapéutico , Trasplante de Riñón/mortalidad , Trasplante de Riñón/fisiología , Estudios Longitudinales , Variaciones Dependientes del Observador , Estudios Prospectivos , Sirolimus/efectos adversos , Infecciones Urinarias/inducido químicamente
20.
Transplantation ; 86(5): 719-27, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18791454

RESUMEN

BACKGROUND: Graft arteriosclerosis (GA) is an important factor limiting long-term outcomes after organ transplantation. We have used a chimeric humanized mouse system to model this arteriopathy in human vessels, and found that the morphologic and functional changes of experimental GA are interferon (IFN)-gamma dependent. This study evaluated whether 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, described as inhibitors of IFN-gamma production, affect GA in our model. METHODS: C.B.-17 severe combined immunodeficiency-beige mice were transplanted with human artery segments as aortic interposition grafts and inoculated with allogeneic human peripheral blood mononuclear cells (PBMCs) or replication-deficient adenovirus encoding human IFN-gamma. Transplant arteries were analyzed from recipients treated with vehicle vs. atorvastatin or simvastatin at different doses. The effects of statins on T-cell alloresponses to vascular endothelial cells were also investigated in vitro. RESULTS: Graft arteriosclerosis-like arteriopathy induced by PBMCs was reduced by atorvastatin at 30 mg/kg/day or simvastatin at 100 mg/kg/day that correlated with decreased graft-infiltrating CD3+ T cells. Circulating IFN-gamma was also reduced, as were graft IFN-gamma and IFN-gamma-inducible chemokine transcripts and graft human leukocyte antigen-DR expression. Graft arteriosclerosis directly induced by human IFN-gamma in the absence of human PBMCs was also reduced by atorvastatin, but only at the highest dose of 100 mg/kg/day. Finally, atorvastatin decreased the clonal expansion and production of interleukin-2, but not IFN-gamma, by human CD4+ T cells in response to allogeneic endothelial cells in coculture. CONCLUSIONS: Our results suggest that a benefit of statin administration in transplantation may include amelioration of GA primarily by inhibiting alloreactive T-cell accumulation and consequent IFN-gamma production and secondarily through suppression of the arterial response to IFN-gamma.


Asunto(s)
Ácidos Heptanoicos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Interferón gamma/biosíntesis , Leucocitos Mononucleares/inmunología , Pirroles/uso terapéutico , Simvastatina/uso terapéutico , Linfocitos T/inmunología , Adulto , Animales , Arterias/trasplante , Atorvastatina , Citometría de Flujo , Humanos , Interferón gamma/antagonistas & inhibidores , Transfusión de Linfocitos , Ratones , Ratones SCID , Linfocitos T/efectos de los fármacos , Trasplante Heterólogo/inmunología , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA