Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transplantation ; 104(8): 1580-1590, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32732835

RESUMEN

BACKGROUND: Although short-term outcomes for liver transplantation have improved, patient and graft survival are limited by infection, cancer, and other complications of immunosuppression. Rapid induction of tolerance after liver transplantation would decrease these complications, improving survival and quality of life. Tolerance to kidneys, but not thoracic organs or islets, has been achieved in nonhuman primates and humans through the induction of transient donor chimerism. Since the liver is considered to be tolerogenic, we tested the hypothesis that the renal transplant transient chimerism protocol would induce liver tolerance. METHODS: Seven cynomolgus macaques received immune conditioning followed by simultaneous donor bone marrow and liver transplantation. The more extensive liver surgery required minor adaptations of the kidney protocol to decrease complications. All immunosuppression was discontinued on postoperative day (POD) 28. Peripheral blood chimerism, recipient immune reconstitution, liver function tests, and graft survival were determined. RESULTS: The level and duration of chimerism in liver recipients were comparable to those previously reported in renal transplant recipients. However, unlike in the kidney model, the liver was rejected soon after immunosuppression withdrawal. Rejection was associated with proliferation of recipient CD8 T effector cells in the periphery and liver, increased serum interleukin (IL)-6 and IL-2, but peripheral regulatory T cell (Treg) numbers did not increase. Antidonor antibody was also detected. CONCLUSIONS: These data show the transient chimerism protocol does not induce tolerance to livers, likely due to greater CD8 T cell responses than in the kidney model. Successful tolerance induction may depend on greater control or deletion of CD8 T cells in this model.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Rechazo de Injerto/prevención & control , Trasplante de Hígado/efectos adversos , Quimera por Trasplante/inmunología , Acondicionamiento Pretrasplante/métodos , Aloinjertos/inmunología , Animales , Médula Ósea/inmunología , Trasplante de Médula Ósea/métodos , Modelos Animales de Enfermedad , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Humanos , Hígado/inmunología , Trasplante de Hígado/métodos , Macaca fascicularis , Linfocitos T Citotóxicos/inmunología , Tolerancia al Trasplante , Trasplante Homólogo/efectos adversos
2.
Transplantation ; 104(2): 270-279, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31385931

RESUMEN

BACKGROUND: Cytomegalovirus (CMV) infection is a serious complication in immunosuppressed patients, specifically transplant recipients. Here, we describe the development and use of an assay to monitor the incidence and treatment of CMV viremia in a Cynomolgus macaque model of bone marrow transplantation (BMT) for tolerance induction. We address the correlation between the course of viremia and immune reconstitution. METHODS: Twenty-one animals received a nonmyeloablative conditioning regimen. Seven received cyclosporine A for 28 days and 14 received rapamycin. A CMV polymerase chain reaction assay was developed and run twice per week to monitor viremia. Nineteen recipients were CMV seropositive before BMT. Immune reconstitution was monitored through flow cytometry and CMV viremia was tracked via quantitative polymerase chain reaction. RESULTS: Recipients developed CMV viremia during the first month post-BMT. Two animals developed uncontrollable CMV disease. CMV reactivation occurred earlier in cyclosporine A-treated animals compared with those receiving rapamycin. Post-BMT, T-cell counts remained significantly lower compared with pretransplant levels until CMV reactivation, at which point they increased during the viremic phase and approached pretransplant levels 3 months post-BMT. Management of CMV required treatment before viremia reached 10 000 copies/mL; otherwise clinical symptoms were observed. High doses of ganciclovir resolved the viremia, which could subsequently be controlled with valganciclovir. CONCLUSIONS: We developed an assay to monitor CMV in Cynomolgus macaques. CMV reactivation occurred in 100% of seropositive animals in this model. Rapamycin delayed CMV reactivation and ganciclovir treatment was effective at high doses. As in humans, CD8 T cells proliferated during CMV viremia.


Asunto(s)
Trasplante de Médula Ósea/métodos , Infecciones por Citomegalovirus/terapia , Rechazo de Injerto/inmunología , Reconstitución Inmune/fisiología , Tolerancia Inmunológica , Sirolimus/farmacología , Activación Viral , Animales , Antifúngicos/farmacología , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Modelos Animales de Enfermedad , Rechazo de Injerto/prevención & control , Macaca fascicularis , Receptores de Trasplantes
3.
Am J Transplant ; 19(8): 2186-2198, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30768842

RESUMEN

Reliable in vitro expansion protocols of regulatory T cells (Tregs) are needed for clinical use. We studied the biology of Mauritian Cynomolgus macaque (MCM) Tregs and developed four in vitro Treg expansion protocols for translational studies. Tregs expanded 3000-fold when artificial antigen presenting cells (aAPCs) expressing human CD80, CD58 and CD32 were used throughout the culture. When donor peripheral blood mononuclear cells (PBMCs) were used as the single source of APCs followed by aAPCs, Tregs expanded 2000-fold. Tregs from all protocols suppressed the proliferation of anti-CD2CD3CD28 bead-stimulated autologous PBMCs albeit with different potencies, varying from 1:2-1:4 Treg:PBMC ratios, up to >1:32. Reculture of cryopreserved Tregs permitted reexpansion with improved suppressive activity. Occasionally, CD8 contamination was observed and resolved by resorting. Specificity studies showed greater suppression of stimulation by anti-CD2CD3CD28 beads of PBMCs from the same donor used for stimulation during the Treg cultures and of autologous cells than of third-party PBMC responders. Similar to humans, the Treg-specific demethylated region (TSDR) within the Foxp3 locus correlated with suppressive activity and expression of Foxp3. Contrary to humans, FoxP3 expression did not correlate with CD45RA or CD127 expression. In summary, we have characterized MCM Tregs and developed four Treg expansion protocols that can be used for preclinical applications.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Metilación de ADN , Factores de Transcripción Forkhead/metabolismo , Leucocitos Mononucleares/citología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Animales , Células Cultivadas , Factores de Transcripción Forkhead/genética , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Macaca fascicularis , Linfocitos T Reguladores/metabolismo
4.
Tissue Eng Part A ; 24(9-10): 740-751, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28938862

RESUMEN

Cell implantation for tissue repair is a promising new therapeutic strategy. Although direct injection of cells into tissue is appealing, cell viability and retention are not very good. Cell engraftment and survival following implantation are dependent on a sufficient supply of oxygen and nutrients through functional microcirculation as well as a suitable local microenvironment for implanted cells. In this study, we describe the development of a porous, biocompatible, three-dimensional (3D) alginate scaffold covalently modified with the synthetic cyclic RGDfK (Arg-Gly-Asp-D-Phe-Lys) peptide. Cyclic RGDfK peptide is protease resistant, highly stable in aqueous solutions, and has high affinity for cellular integrins. Cyclic RGDfK-modified alginate scaffolds were generated using a novel silicone sheet sandwich technique in combination with freeze-gelation, resulting in highly porous nonimmunogenic scaffolds that promoted both human and rodent cell survival in vitro, and neoangiogenesis in vivo. Two months following implantation in abdominal rectus muscles in rats, cyclic RGDfK-modified scaffolds were fully populated by host cells, especially microvasculature without an overt immune response or fibrosis, whereas unmodified control scaffolds did not show cell ingrowth. Importantly, modified scaffolds that were seeded with human mesenchymal precursor cells and were patched to the epicardial surface of infarcted myocardium induced myocardial neoangiogenesis and significantly improved cardiac function. In summary, purified cyclic RGDfK peptide-modified 3D alginate scaffolds are biocompatible and nonimmunogenic, enhance cell viability, promote angiogenesis, and may be used as a means to deliver cells to myocardial infarct areas to improve neovascularization and cardiac function.


Asunto(s)
Oligopéptidos/química , Andamios del Tejido/química , Animales , Materiales Biocompatibles/química , Adhesión Celular/fisiología , Línea Celular , Trasplante de Células/métodos , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Microscopía Electrónica de Rastreo , Miocardio/citología , Neovascularización Fisiológica/fisiología , Ratas , Ingeniería de Tejidos/métodos
5.
Comp Med ; 67(5): 436-441, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28935006

RESUMEN

Cynomolgus monkeys are often used in preclinical transplantation research. Performing liver transplantation in cynomolgus monkeys is challenging because they poorly tolerate portal vein clamping during the anhepatic phase. Finding an alternative to portal vein clamping is necessary before preclinical liver transplant models can be performed with reliable outcomes. We used 3 different techniques to perform 5 liver transplants in male cynomolgus macaques (weight, 7.4-10.8 kg; mismatched for MHC I and II; matched for ABO). In procedure A, we clamped the portal vein briefly, as in human transplants, as well as the superior mesentery artery to minimize congestion at the expense of temporary ischemia (n = 2). In procedure B, we performed a temporary portocaval shunt with extracorporeal venovenous bypass (n = 1). For procedure C, we developed an H-shunt system (modified portocaval shunt) with extracorporeal bypass (n = 2). Postoperative immunosuppression comprised cyclosporine A, mycophenolate mofetil, and steroids. Recipients in procedure A developed hemodynamic instability and were euthanized within 2 d. The recipient that underwent procedure B was euthanized within 11 d due to inferior vena caval thrombosis. The H-shunt in procedure C led to minimal PV congestion during the anhepatic phase, and both recipients reached the 21-d survival endpoint with good graft function. Our novel H-shunt bypass system resulted in successful liver transplantation in cynomolgus macaques, with long-term posttransplant survival possible. This technical innovation makes possible the use of cynomolgus monkeys for preclinical liver transplant tolerance models.


Asunto(s)
Ciclosporina/uso terapéutico , Inmunosupresores/uso terapéutico , Trasplante de Hígado/veterinaria , Macaca fascicularis/cirugía , Derivación Portocava Quirúrgica/veterinaria , Anastomosis Quirúrgica/métodos , Anastomosis Quirúrgica/veterinaria , Animales , Femenino , Humanos , Pruebas de Función Hepática/veterinaria , Trasplante de Hígado/efectos adversos , Trasplante de Hígado/métodos , Masculino , Arterias Mesentéricas/cirugía , Modelos Animales , Vena Porta
6.
Transplantation ; 101(2): 274-283, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27846155

RESUMEN

BACKGROUND: Infusion of recipient regulatory T (Treg) cells promotes durable mixed hematopoietic chimerism and allograft tolerance in mice receiving allogeneic bone marrow transplant (BMT) with minimal conditioning. We applied this strategy in a Cynomolgus macaque model. METHODS: CD4 CD25 Treg cells that were polyclonally expanded in culture were highly suppressive in vitro and maintained high expression of FoxP3. Eight monkeys underwent nonmyeloablative conditioning and major histocompatibility complex mismatched BMT with or without Treg cell infusion. Renal transplantation (from the same BMT donor) was performed 4 months post-BMT without immunosuppression to assess for robust donor-specific tolerance. RESULTS: Transient mixed chimerism, without significant T cell chimerism, was achieved in the animals that received BMT without Treg cells (N = 3). In contrast, 2 of 5 recipients of Treg cell BMT that were evaluable displayed chimerism in all lineages, including T cells, for up to 335 days post-BMT. Importantly, in the animal that survived long-term, greater than 90% of donor T cells were CD45RA CD31, suggesting they were new thymic emigrants. In this animal, the delayed (to 4 months) donor kidney graft was accepted more than 294 days without immunosuppression, whereas non-Treg cell BMT recipients rejected delayed donor kidneys within 3 to 4 weeks. Early CMV reactivation and treatment was associated with early failure of chimerism, regardless of Treg cell administration. CONCLUSIONS: Our studies provide proof-of-principle that, in the absence of early CMV reactivation (and BM-toxic antiviral therapy), cotransplantation of host Treg cell can promote prolonged and high levels of multilineage allogeneic chimerism and robust tolerance to the donor.


Asunto(s)
Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Antígenos de Histocompatibilidad/inmunología , Histocompatibilidad , Trasplante de Riñón/métodos , Linfocitos T Reguladores/trasplante , Quimera por Trasplante/inmunología , Acondicionamiento Pretrasplante/métodos , Tolerancia al Trasplante , Aloinjertos , Animales , Antivirales/uso terapéutico , Biomarcadores/metabolismo , Trasplante de Médula Ósea , Proliferación Celular , Células Cultivadas , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/inmunología , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Antígenos de Histocompatibilidad/metabolismo , Trasplante de Riñón/efectos adversos , Macaca fascicularis , Masculino , Modelos Animales , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factores de Tiempo , Acondicionamiento Pretrasplante/efectos adversos
7.
J Biomater Appl ; 31(4): 510-520, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27114440

RESUMEN

Sodium alginate is an effective biomaterial for tissue engineering applications. Non-purified alginate is contaminated with protein, lipopolysaccharide, DNA, and RNA, which could elicit adverse immunological reactions. We developed a purification protocol to generate biocompatible alginate based on (a) activated charcoal treatment, (b) use of hydrophobic membrane filtration (we used hydrophobic polyvinylidene difluoride membranes to remove organic contaminants), (c) dialysis, and finally (d) ethanol precipitation. Using this approach, we could omit pre-treatment with chloroform and significantly reduce the quantities of reagents used. Purification resulted in reduction of residual protein by 70% down to 0.315 mg/g, DNA by 62% down to 1.28 µg/g, and RNA by 61% down to less than 10 µg/g, respectively. Lipopolysaccharide levels were reduced by >90% to less than 125 EU/g. Purified alginate did not induce splenocyte proliferation in vitro. Three-dimensional scaffolds generated from purified alginate did not elicit a significant foreign body reaction, fibrotic overgrowth, or macrophage infiltration 4 weeks after implantation. This study describes a simplified and economical alginate purification method that results in alginate purity, which meets clinically useful criteria.


Asunto(s)
Compuestos de Aluminio/efectos adversos , Compuestos de Aluminio/aislamiento & purificación , Reacción a Cuerpo Extraño/inmunología , Polivinilos/química , Compuestos de Sodio/efectos adversos , Compuestos de Sodio/aislamiento & purificación , Andamios del Tejido/efectos adversos , Ultrafiltración/métodos , Absorción Fisicoquímica , Compuestos de Aluminio/química , Animales , Materiales Biocompatibles/efectos adversos , Materiales Biocompatibles/química , Materiales Biocompatibles/aislamiento & purificación , Carbón Orgánico/química , Precipitación Química , Etanol/química , Reacción a Cuerpo Extraño/prevención & control , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Masculino , Ensayo de Materiales , Membranas Artificiales , Ratas , Ratas Endogámicas Lew , Compuestos de Sodio/química
8.
J Cell Mol Med ; 15(10): 2117-29, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21155976

RESUMEN

Stromal precursor antigen (STRO)-3 has previously been shown to identify a subset of adult human bone marrow (BM)-derived mesenchymal lineage precursors, which may have cardioprotective potential. We sought to characterize STRO-3-immunoselected and culture-expanded mesenchymal precursor cells (MPCs) with respect to their biology and therapeutic potential in myocardial ischemia. Immunoselection of STRO-3(+) MPCs enriched for fibroblastic colony forming units from unfractionated BM mononuclear cells (MNCs). Compared to mesenchymal stem cells conventionally isolated by plastic adherence, MPCs demonstrated increased proliferative capacity during culture expansion, expressed higher levels of early 'stem cell' markers and various pro-angiogenic and cardioprotective cytokines, and exhibited greater trilineage developmental efficiency. Intramyocardial injection of MPCs into a rat model of myocardial infarction (MI) promoted left ventricular recovery and inhibited left ventricular dilatation. These beneficial effects were associated with cardioprotective and pro-angiogenic effects at the tissue level, despite poor engraftment of cells. Treatment of MI rats with MPC-conditioned medium (CM) preserved left ventricular function and dimensions, reduced myocyte apoptosis and fibrosis, and augmented neovascularization, involving both resident vascular cells and circulating endothelial progenitor cells (EPCs). Profiling of CM revealed various cardioprotective and pro-angiogenic factors, which had biological activity in cultures of myocytes, tissue-resident vascular cells and EPCs. Prospective immunoselection of STRO-3(+) MPCs from BM MNCs conferred advantage in maintaining a population of immature MPCs during ex vivo expansion. Transplantation of culture-expanded MPCs into the post-MI heart resulted in therapeutic benefit, attributable at least in part to paracrine mechanisms of action. Thus, MPCs represent a promising therapy for myocardial ischemia.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Isquemia Miocárdica/terapia , Animales , Antígenos/análisis , Médula Ósea/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratas Desnudas , Células del Estroma/metabolismo
9.
Eur J Intern Med ; 17(6): 441-3, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16962956

RESUMEN

We describe the case of a 45-year-old man with mitral and aortic prosthetic valve replacement who presented with symptoms of subacute bacterial endocarditis. Bartonella quintana was grown from blood after prolonged culture. The course of the disease was complicated by splenic infarction, glomerulonephritis resulting in progressive renal insufficiency, and cerebroventricular hemorrhage. Notably, cardiac ultrasonography showed no extensive vegetations but a strand-like lesion. Culture-positive B. quintana prosthetic valve endocarditis in a formerly healthy subject represents a newly observed entity. It should be added to the differential diagnosis of prosthetic valve endocarditis, especially when it presents with features suggesting subacute bacterial endocarditis.

10.
Nat Clin Pract Cardiovasc Med ; 3 Suppl 1: S18-22, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16501624

RESUMEN

Mesenchymal lineage precursors can be reproducibly isolated from adult mammalian bone marrow and grown in culture. Immunoselection with monoclonal antibodies against STRO-1 and vascular-cell-adhesion molecule 1 (VCAM1/CD106) prior to expansion results in a 1,000-fold enrichment of mesenchymal precursors compared to standard isolation techniques. Intramyocardial injection of human STRO-1-selected precursors in an athymic rat model of acute myocardial infarction results in induction of vascular network formation and arteriogenesis coupled with global functional cardiac recovery.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Isquemia Miocárdica/terapia , ADP-Ribosil Ciclasa/análisis , Animales , Antígenos CD/análisis , Células de la Médula Ósea/inmunología , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Circulación Coronaria , Modelos Animales de Enfermedad , Proteínas Ligadas a GPI , Hemodinámica , Humanos , Separación Inmunomagnética , Células Madre Mesenquimatosas/inmunología , Isquemia Miocárdica/fisiopatología , Neovascularización Fisiológica , Ratas , Ratas Desnudas , Molécula 1 de Adhesión Celular Vascular/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...