Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Chem Biol ; 2023 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-37945898

RESUMEN

After the discovery of insulin, a century ago, extensive work has been done to unravel the molecular network regulating insulin secretion. Here we performed a chemical screen and identified AZD7762, a compound that potentiates glucose-stimulated insulin secretion (GSIS) of a human ß cell line, healthy and type 2 diabetic (T2D) human islets and primary cynomolgus macaque islets. In vivo studies in diabetic mouse models and cynomolgus macaques demonstrated that AZD7762 enhances GSIS and improves glucose tolerance. Furthermore, genetic manipulation confirmed that ablation of CHEK2 in human ß cells results in increased insulin secretion. Consistently, high-fat-diet-fed Chk2-/- mice show elevated insulin secretion and improved glucose clearance. Finally, untargeted metabolic profiling demonstrated the key role of the CHEK2-PP2A-PLK1-G6PD-PPP pathway in insulin secretion. This study successfully identifies a previously unknown insulin secretion regulating pathway that is conserved across rodents, cynomolgus macaques and human ß cells in both healthy and T2D conditions.

2.
J Vasc Interv Radiol ; 35(6): 809-817.e1, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38219903

RESUMEN

Traditionally, rodent cancer models have driven preclinical oncology research. However, they do not fully recapitulate characteristics of human cancers, and their size poses challenges when evaluating tools in the interventional oncologists' armamentarium. Pig models, however, have been the gold standard for validating surgical procedures. Their size enables the study of image-guided interventions using human ultrasound (US), computed tomography (CT), and magnetic resonance (MR) imaging platforms. Furthermore, pigs have immunologic features that are similar to those of humans, which can potentially be leveraged for studying immunotherapy. Novel pig models of cancer are being developed, but additional research is required to better understand both the pig immune system and malignancy to enhance the potential for pig models in interventional oncology research. This review aims to address the main advantages and disadvantages of using a pig model for interventional oncology and outline the specific characteristics of pig models that make them more suitable for investigation of locoregional therapies.


Asunto(s)
Modelos Animales de Enfermedad , Inmunoterapia , Neoplasias , Animales , Inmunoterapia/métodos , Neoplasias/terapia , Neoplasias/diagnóstico por imagen , Neoplasias/inmunología , Humanos , Porcinos , Radiografía Intervencional , Sus scrofa , Oncología Médica
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.
Biol Blood Marrow Transplant ; 22(11): 1953-1960, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27543159

RESUMEN

We previously described successful hematopoietic stem cell engraftment across MHC barriers in miniature swine without graft-versus-host disease (GVHD) using novel reduced-intensity conditioning regimens consisting of partial transient recipient T cell-depletion, thymic or low-dose total body irradiation, and a short course of cyclosporine A. Here we report that stable chimeric animals generated with these protocols are strongly resistant to donor leukocyte infusion (DLI)-mediated GVH effects. Of 33 total DLIs in tolerant chimeras at clinical doses, 21 failed to induce conversion to full donor hematopoietic chimerism or cause GVHD. We attempted to overcome this resistance to conversion through several mechanisms, including using sensitized donor lymphocytes, increasing the DLI dose, removing chimeric host peripheral blood cells through extensive recipient leukapheresis before DLI, and using fully mismatched lymphocytes. Despite our attempts, the resistance to conversion in our model was robust, and when conversion was achieved, it was associated with GVHD in most animals. Our studies suggest that delivery of unmodified hematopoietic stem cell doses under reduced-intensity conditioning can induce a potent, GVHD-free, immune tolerant state that is strongly resistant to DLI.


Asunto(s)
Donantes de Sangre , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/prevención & control , Transfusión de Linfocitos/efectos adversos , Trasplante Haploidéntico/métodos , Animales , Ciclosporina/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/métodos , Depleción Linfocítica , Porcinos , Quimera por Trasplante , Acondicionamiento Pretrasplante , Irradiación Corporal Total
5.
Biol Blood Marrow Transplant ; 21(10): 1732-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26210443

RESUMEN

Post-transplant lymphoproliferative disease (PTLD) is a major complication of clinical organ and cell transplantation. Conditioning and immunosuppressive regimens that significantly impair T cell immunity, including depleting antibodies and calcineurin inhibitors, increase the risk of PTLD after transplantation. Swine PTLD has been shown to closely resemble human PTLD in morphology, histology, and viral-driven reactivation of B cells. Previously, we reported high incidences of PTLD after hematopoietic cell transplantation (HCT) in miniature swine recipients conditioned with thymic irradiation (TI) in addition to T cell depletion and cyclosporine A monotherapy after transplantation. Replacement of TI with 100 cGy of total body irradiation resulted in similar numbers of B cells early post-transplantation, greater numbers of T cells at day 0, and markedly decreased incidence of PTLD, suggesting that a threshold number of T cells may be necessary to prevent subsequent B cell proliferation and development of overt PTLD. Results from this large cohort of animals provide insight into the important effect of irradiation and T cell immunity on the incidence of PTLD after HCT and reinforce the pig model as a valuable tool for the study of PTLD and HCT.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/efectos adversos , Inmunosupresores/efectos adversos , Irradiación Linfática/efectos adversos , Trastornos Linfoproliferativos/prevención & control , Porcinos Enanos , Timo/efectos de la radiación , Acondicionamiento Pretrasplante/efectos adversos , Irradiación Corporal Total , Animales , Inhibidores de la Calcineurina/efectos adversos , Inhibidores de la Calcineurina/uso terapéutico , Ciclosporina/efectos adversos , Ciclosporina/uso terapéutico , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/veterinaria , Herpesvirus Suido 1/patogenicidad , Histocompatibilidad , Humanos , Inmunosupresores/uso terapéutico , L-Lactato Deshidrogenasa/sangre , Irradiación Linfática/métodos , Depleción Linfocítica/efectos adversos , Trastornos Linfoproliferativos/etiología , Trastornos Linfoproliferativos/veterinaria , Porcinos , Enfermedades de los Porcinos/etiología , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/virología , Porcinos Enanos/inmunología , Porcinos Enanos/virología , Linfocitos T/efectos de la radiación , Acondicionamiento Pretrasplante/métodos , Infecciones Tumorales por Virus/veterinaria , Irradiación Corporal Total/efectos adversos
6.
Xenotransplantation ; 20(6): 458-68, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24289469

RESUMEN

BACKGROUND: The development of genetically modified pigs, which lack the expression of alpha 1-3 galactosyl transferase, (GalT-KO pigs) has facilitated the xenogeneic transplantation of porcine organs and tissues into primates by avoiding hyperacute rejection due to pre-existing antibodies against the Gal epitope. However, antibodies against other antigens (anti-non-Gal antibodies), are found at varying levels in the pre-transplant sera of most primates. We have previously found that baboons with high levels of pre-transplant anti-non-Gal IgG, conditioned with a non-myeloablative conditioning regimen, failed to engraft following pig-to-baboon bone marrow transplantation (Xenotransplantation, 17, 2010 and 300). Two baboons with low levels of pre-transplant anti-non-Gal IgG, conditioned with the same regimen, showed porcine bone marrow progenitors at 28 days following transplantation, suggesting engraftment. These baboons also showed evidence of donor-specific hyporesponsiveness. This observation led us to investigate the hypothesis that selecting for baboon recipients with low pre-transplant anti-non-Gal IgG levels might improve engraftment levels following GalT-KO pig-to-baboon bone marrow transplantation. METHODS: Five baboons, with low pre-transplant anti-non-Gal IgG levels, received transplantation of bone marrow cells (1-5 × 10(9) /kg of recipient weight) from GalT-KO pigs. They received a non-myeloablative conditioning regimen consisting of low-dose total body irradiation (TBI) (150 cGy), thymic irradiation (700 cGy), anti-thymocyte globulin (ATG), and tacrolimus. In addition, two baboons received Rituximab and Bortezomib (Velcade) treatment as well as extra-corporeal immunoadsorption using GalT-KO pig livers. Bone marrow engraftment was assessed by porcine-specific PCR on colony forming units (CFU) of day 28 bone marrow aspirates. Anti-non-Gal antibody levels were assessed by serum binding toward GalT-KO PBMC using flow cytometry (FACS). Peripheral macro-chimerism was measured by FACS using pig and baboon-specific antibodies and baboon anti-pig cellular responses were assessed by mixed lymphocyte reactions (MLR). RESULTS: As previously reported, two of five baboons demonstrated detectable bone marrow engraftment at 4 weeks after transplantation. Engraftment was associated with lack of an increase in anti-non-Gal IgG levels as well as cellular hyporesponsiveness toward pig. Three subsequent baboons with similarly low levels of pre-existing anti-non-Gal IgG showed no engraftment and an increase in anti-non-Gal IgG antibody levels following transplantation. Peripheral macrochimerism was only seen for a few days following transplantation regardless of antibody development. CONCLUSIONS: Selecting for baboon recipients with low levels of pre-transplant anti-non-Gal IgG did not ensure bone marrow engraftment. Failure to engraft was associated with an increase in anti-non-Gal IgG levels following transplantation. These results suggest that anti-non-Gal-IgG is likely involved in early bone marrow rejection and that successful strategies for combating anti-non-Gal IgG development may allow better engraftment. Since engraftment was only low and transient regardless of antibody development, innate immune, or species compatibility mechanisms will likely also need to be addressed to achieve long term engraftment.


Asunto(s)
Anticuerpos Heterófilos/sangre , Trasplante de Médula Ósea/efectos adversos , Xenoinjertos/inmunología , Inmunoglobulina G/sangre , Papio hamadryas/inmunología , Porcinos Enanos/inmunología , Animales , Animales Modificados Genéticamente , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Técnicas de Inactivación de Genes , Rechazo de Injerto/etiología , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Inmunidad Celular , Prueba de Cultivo Mixto de Linfocitos , Porcinos
7.
Cell Rep Med ; 4(10): 101241, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37852175

RESUMEN

Allogeneic invariant natural killer T cells (allo-iNKTs) induce clinical remission in patients with otherwise incurable cancers and COVID-19-related acute respiratory failure. However, their functionality is inconsistent among individuals, and they become rapidly undetectable after infusion, raising concerns over rejection and limited therapeutic potential. We validate a strategy to promote allo-iNKT persistence in dogs, an established large-animal model for novel cellular therapies. We identify donor-specific iNKT biomarkers of survival and sustained functionality, conserved in dogs and humans and retained upon chimeric antigen receptor engineering. We reason that infusing optimal allo-iNKTs enriched in these biomarkers will prolong their persistence without requiring MHC ablation, high-intensity chemotherapy, or cytokine supplementation. Optimal allo-iNKTs transferred into MHC-mismatched dogs remain detectable for at least 78 days, exhibiting sustained immunomodulatory effects. Our canine model will accelerate biomarker discovery of optimal allo-iNKT products, furthering application of MHC-unedited allo-iNKTs as a readily accessible universal platform to treat incurable conditions worldwide.


Asunto(s)
COVID-19 , Trasplante de Células Madre Hematopoyéticas , Células T Asesinas Naturales , Humanos , Perros , Animales , Trasplante Homólogo , Biomarcadores
8.
mSphere ; 8(4): e0017723, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37404023

RESUMEN

The microbiota mediate multiple aspects of skin barrier function, including colonization resistance to pathogens such as Staphylococcus aureus. The endogenous skin microbiota limits S. aureus colonization via competition and direct inhibition. Novel mechanisms of colonization resistance are promising therapeutic targets for drug-resistant infections, such as those caused by methicillin-resistant S. aureus (MRSA). Here, we developed and characterized a swine model of topical microbiome perturbation and MRSA colonization. As in other model systems, topical antimicrobial treatment had a little discernable effect on community diversity though the overall microbial load was sensitive to multiple types of intervention, including swabbing. In parallel, we established a porcine skin culture collection and screened 7,700 isolates for MRSA inhibition. Using genomic and phenotypic criteria, we curated three isolates to investigate whether prophylactic colonization would inhibit MRSA colonization in vivo. The three-member consortium together, but not individually, provided protection against MRSA colonization, suggesting cooperation and/or synergy among the strains. Inhibitory isolates were represented across all major phyla of the pig skin microbiota and did not have a strong preference for inhibiting closely related species, suggesting that relatedness is not a condition of antagonism. These findings reveal the porcine skin as an underexplored reservoir of skin commensal species with the potential to prevent MRSA colonization and infection. IMPORTANCE The skin microbiota is protective against pathogens or opportunists such as S. aureus, the most common cause of skin and soft tissue infections. S. aureus can colonize normal skin and nasal passages, and colonization is a risk factor for infection, especially on breach of the skin barrier. Here, we established a pig model to study the competitive mechanisms of the skin microbiota and their role in preventing colonization by MRSA. This drug-resistant strain is also a livestock pathogen, and swine herds can be reservoirs of MRSA carriage. From 7,700 cultured skin isolates, we identified 37 unique species across three phyla that inhibited MRSA. A synthetic community of three inhibitory isolates provided protection together, but not individually, in vivo in a murine model of MRSA colonization. These findings suggest that antagonism is widespread in the pig skin microbiota, and these competitive interactions may be exploited to prevent MRSA colonization.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Microbiota , Infecciones Estafilocócicas , Animales , Porcinos , Ratones , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus/genética , Cavidad Nasal , Infecciones Estafilocócicas/prevención & control , Infecciones Estafilocócicas/veterinaria
9.
Biol Blood Marrow Transplant ; 18(11): 1629-37, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22892552

RESUMEN

Loss of chimerism is an undesirable outcome of allogeneic hematopoietic cell transplantation (HCT) after reduced-intensity conditioning. Understanding the nature of cellular and humoral immune responses to HCT after graft loss could lead to improved retransplantation strategies. We investigated the immunologic responses after graft loss in miniature swine recipients of haploidentical HCT that received reduced-intensity conditioning. After the loss of peripheral blood chimerism, antidonor cellular responses were present without detectable antidonor antibody. Reexposure to donor hematopoietic cells after graft loss induced a sensitized antidonor cellular response. No induced antidonor antibody response could be detected despite evidence of cellular sensitization to donor cells. In contrast, unconditioned animals exposed repeatedly to similar doses of haploidentical donor cells developed antidonor antibody responses. These results could have important implications for the design of treatment strategies to overcome antidonor responses in HCT and improve the outcome of retransplantation after graft loss.


Asunto(s)
Rechazo de Injerto/inmunología , Trasplante de Células Madre Hematopoyéticas , Isoanticuerpos/sangre , Acondicionamiento Pretrasplante/métodos , Animales , Haplotipos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Prueba de Histocompatibilidad , Inmunidad Celular , Inmunidad Humoral , Inmunización , Porcinos , Porcinos Enanos , Quimera por Trasplante/inmunología , Trasplante Homólogo
10.
Protein Expr Purif ; 82(1): 70-4, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22154875

RESUMEN

Yeast Pichia pastoris has been widely utilized to express heterologous recombinant proteins. P. pastoris expressed recombinant porcine interleukin 3 (IL3) has been used for porcine stem cell mobilization in allo-hematopoietic cell transplantation models and pig-to-primate xeno-hematopoietic cell transplantation models in our lab for many years. Since the yeast glycosylation mechanism is not exactly the same as those of other mammalian cells, P. pastoris expressed high-mannose glycoprotein porcine IL3 has been shown to result in a decreased serum half-life. Previously this was avoided by separation of the non-glycosylated porcine IL3 from the mixture of expressed glycosylated and non-glycosylated porcine IL3. However, this process was very inefficient and lead to a poor yield following purification. To overcome this problem, we engineered a non-N-glycosylated version of porcine IL3 by replacing the four potential N-glycosylation sites with four alanines. The codon-optimized non-N-glycosylated porcine IL3 gene was synthesized and expressed in P. pastoris. The expressed non-N-glycosylated porcine IL3 was captured using Ni-Sepharose 6 fast flow resin and further purified using strong anion exchange resin Poros 50 HQ. In vivo mobilization studies performed in our research facility demonstrated that the non-N-glycosylated porcine IL3 still keeps the original stem cell mobilization function.


Asunto(s)
Interleucina-3/genética , Interleucina-3/aislamiento & purificación , Pichia/genética , Porcinos/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Expresión Génica , Glicosilación , Interleucina-3/química , Datos de Secuencia Molecular , Plásmidos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Porcinos/genética
11.
J Immunol ; 185(7): 3866-72, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20810991

RESUMEN

Allogeneic bone marrow transplantation is an effective treatment for a number of malignant and nonmalignant diseases (Applebaum. 2001. Nature. 411: 385-389 and Copelan. 2006. N Engl J Med. 354: 1813-1826). However, the application of this therapeutic modality has been impeded by a number of confounding side effects, the most frequent and severe of which is the development of graft-versus-host disease (GVHD) (Copelan. 2006. N Engl J Med. 354: 1813-1826 and Blazar and Murphy. 2005. Philos Trans R Soc Lond B Biol Sci. 360: 1747-1767). Alloreactive donor T cells are critical for causing GVHD (Fowler. 2006. Crit Rev Oncol Hematol. 57: 225-244 and Ferrara and Reddy. 2006. Semin Hematol. 43: 3-10), whereas recent data demonstrated a significant role for the naturally occurring thymic-derived donor CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) (Bluestone and Abbas. 2003. Nat Rev Immunol. 3: 253-257 and Shevach. 2006. Immunity. 25: 195-201) in suppressing experimental GVHD after bone marrow transplantation (Blazar and Taylor. 2005. Biol Blood Marrow Transpl. 11: 46-49 and Joffe and van Meerwijk. 2006. Semin Immunol. 18: 128-135) . Host APCs are required for induction of GVHD by the conventional donor T cells. However, it is not known whether they are also obligatory for donor Treg-mediated suppression of GVHD. Using multiple clinically relevant MHC-matched and -mismatched murine models of GVHD, we investigated the role of host APCs in the suppression of GVHD by donor Tregs. We found that alloantigen expression by the host APCs is necessary and sufficient for induction of GVHD protection by donor Tregs. This requirement was independent of their effect on the maintenance of Treg numbers and the production of IL-10 or IDO by the host APCs.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Enfermedad Injerto contra Huésped/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/metabolismo , Trasplante de Médula Ósea/efectos adversos , Trasplante de Médula Ósea/inmunología , Separación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo
12.
Transplantation ; 106(5): 928-937, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-34720103

RESUMEN

Over the last 40 y, a specialized herd of miniature swine has been intentionally bred to develop lines of animals homozygous for the swine major histocompatibility complex (MHC), which have facilitated transplantation studies across reproducible MHC and minor antigen mismatch barriers. These MHC-characterized miniature swine (Mc-MS) have been used for the study of novel surgical techniques, various approaches to tolerance induction of solid organ and vascularized composite allografts, as well as studies of the immunobiology of allografts and xenografts. Mc-MS possess characteristics that are highly advantageous to these studies, and their continued use will likely continue to play an important role in bridging "bench-to-cage-to bedside" therapies in the field of transplantation. In this review, we highlight the seminal contributions of the Mc-MS model to the field and analyze their role in the broader context of large animal models in transplantation research.


Asunto(s)
Aloinjertos Compuestos , Trasplante de Riñón , Animales , Aloinjertos Compuestos/trasplante , Rechazo de Injerto , Humanos , Tolerancia Inmunológica , Complejo Mayor de Histocompatibilidad/genética , Porcinos , Porcinos Enanos
13.
Cell Rep Med ; 3(5): 100614, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35551746

RESUMEN

Adoptive transfer of chimeric antigen receptor regulatory T cells (CAR Tregs) is a promising way to prevent allograft loss without the morbidity associated with current therapies. Non-human primates (NHPs) are a clinically relevant model to develop transplant regimens, but manufacturing and engraftment of NHP CAR Tregs have not been demonstrated yet. Here, we describe a culture system that massively expands CAR Tregs specific for the Bw6 alloantigen. In vitro, these Tregs suppress in an antigen-specific manner without pro-inflammatory cytokine secretion or cytotoxicity. In vivo, Bw6-specific CAR Tregs preferentially traffic to and persist in bone marrow for at least 1 month. Following transplant of allogeneic Bw6+ islets and autologous CAR Tregs into the bone marrow of diabetic recipients, CAR Tregs traffic to the site of islet transplantation and maintain a phenotype of suppressive Tregs. Our results establish a framework for the optimization of CAR Treg therapy in NHP disease models.


Asunto(s)
Isoantígenos , Receptores Quiméricos de Antígenos , Traslado Adoptivo , Animales , Macaca , Receptores Quiméricos de Antígenos/genética , Linfocitos T Reguladores
14.
STAR Protoc ; 3(4): 101784, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36386869

RESUMEN

It is technically challenging to generate large doses of regulatory T cells (Tregs) engineered to express a chimeric antigen receptor (CAR) in non-human primates (NHP). Here, we have optimized the manufacturing of CAR Tregs by stringent sorting of Tregs, stimulation by artificial antigen-presenting cells, transduction by simian tropic lentiviral vectors, and antigen-specific expansion. The result of this method is highly suppressive CAR Tregs for use in a pre-clinical, large animal model of transplant tolerance. For complete details on the use and execution of this protocol, please refer to Ellis et al. (2022).


Asunto(s)
Receptores Quiméricos de Antígenos , Animales , Receptores Quiméricos de Antígenos/genética , Células Presentadoras de Antígenos , Linfocitos T Reguladores , Primates , Tolerancia al Trasplante
15.
J Clin Invest ; 118(7): 2562-73, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18568076

RESUMEN

Histone deacetylase (HDAC) inhibitors are antitumor agents that also have antiinflammatory properties. However, the mechanisms of their immunomodulatory functions are not known. We investigated the mechanisms of action of 2 HDAC inhibitors, suberoylanilide hydroxamic acid (SAHA) and ITF 2357, on mouse DC responses. Pretreatment of DCs with HDAC inhibitors significantly reduced TLR-induced secretion of proinflammatory cytokines, suppressed the expression of CD40 and CD80, and reduced the in vitro and in vivo allostimulatory responses induced by the DCs. In addition, injection of DCs treated ex vivo with HDAC inhibitors reduced experimental graft-versus-host disease (GVHD) in a murine allogeneic BM transplantation model. Exposure of DCs to HDAC inhibitors increased expression of indoleamine 2,3-dioxygenase (IDO), a suppressor of DC function. Blockade of IDO in WT DCs with siRNA and with DCs from IDO-deficient animals caused substantial reversal of HDAC inhibition-induced in vitro suppression of DC-stimulated responses. Direct injection of HDAC inhibitors early after allogeneic BM transplantation to chimeric animals whose BM-derived cells lacked IDO failed to protect from GVHD, demonstrating an in vivo functional role for IDO. Together, these data show that HDAC inhibitors regulate multiple DC functions through the induction of IDO and suggest that they may represent a novel class of agents to treat immune-mediated diseases.


Asunto(s)
Células Dendríticas/fisiología , Inhibidores Enzimáticos/farmacología , Enfermedad Injerto contra Huésped/prevención & control , Inhibidores de Histona Desacetilasas , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Animales , Antígenos CD/metabolismo , Trasplante de Médula Ósea , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/trasplante , Femenino , Expresión Génica/efectos de los fármacos , Enfermedad Injerto contra Huésped/patología , Humanos , Ácidos Hidroxámicos/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , ARN Interferente Pequeño/genética , Análisis de Supervivencia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Vorinostat
16.
Bioconjug Chem ; 22(10): 2014-20, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-21866954

RESUMEN

Anti-CD3 immunotoxins, which induce profound but transient T-cell depletion in vivo by inhibiting eukaryotic protein synthesis in CD3+ cells, are effective reagents in large animal models of transplantation tolerance and autoimmune disease therapy. A diphtheria toxin based antiporcine CD3 recombinant immunotoxin was constructed by fusing the truncated diphtheria toxin DT390 with two identical tandem single chain variable fragments (scFv) derived from the antiporcine CD3 monoclonal antibody 898H2-6-15. The recombinant immunotoxin was expressed in a diphtheria-toxin resistant yeast Pichia pastoris strain under the control of the alcohol oxidase promoter. The secreted recombinant immunotoxin was purified sequentially with hydrophobic interaction chromatography (Butyl 650 M) followed by strong anion exchange (Poros 50 HQ). The purified antiporcine CD3 immunotoxin was tested in vivo in four animals; peripheral blood CD3+ T-cell numbers were reduced by 80% and lymph node T-cells decreased from 74% CD3+ cells pretreatment to 24% CD3+ cells remaining in the lymph node following 4 days of immunotoxin treatment. No clinical toxicity was observed in any of the experimental swine. We anticipate that this conjugate will provide an important tool for in vivo depletion of T-cells in swine transplantation models.


Asunto(s)
Complejo CD3/inmunología , Toxina Diftérica/inmunología , Inmunotoxinas/inmunología , Proteínas Recombinantes de Fusión/inmunología , Porcinos/inmunología , Secuencia de Aminoácidos , Animales , Complejo CD3/genética , Toxina Diftérica/genética , Expresión Génica , Inmunotoxinas/genética , Modelos Moleculares , Datos de Secuencia Molecular , Pichia/genética , Plásmidos/genética , Proteínas Recombinantes de Fusión/genética , Porcinos/genética , Linfocitos T/inmunología
17.
Transplant Cell Ther ; 27(1): 36-44, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33017660

RESUMEN

Hematopoietic stem cell (HSC) transplantation and solid organ transplantation remain the only curative options for many hematologic malignancies and end-stage organ diseases. Unfortunately, the sequelae of long-term immunosuppression, as well as acute and chronic rejection, carry significant morbidities, including infection, malignancy, and graft loss. Numerous murine models have demonstrated the efficacy of adjunctive cellular therapies using HSCs, regulatory T cells, mesenchymal stem cells, and regulatory dendritic cells in modulating the alloimmune response in favor of graft tolerance; however, translation of such murine approaches to other preclinical models and in the clinic has yielded mixed results. Large animals, including nonhuman primates, swine, and canines, provide a more immunologically rigorous model in which to test the clinical translatability of these cellular therapies. Here, we highlight the contributions of large animal models to the development and optimization of HSCs and additional cellular therapies to improve organ transplantation outcomes.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Animales , Perros , Inmunoterapia , Ratones , Modelos Animales , Porcinos , Tolerancia al Trasplante , Trasplante Homólogo
18.
Transpl Immunol ; 68: 101431, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34157374

RESUMEN

Acute rejection is a leading cause of organ transplant failure and the most common indication for re-transplantation. Clinically, suspicion of acute rejection is often dependent upon serum laboratory values which may only manifest after organ injury. The gold standard for diagnosis requires an invasive biopsy which can carry serious clinical risks including bleeding and graft loss as well as the possibility of sampling error. The use of noninvasive imaging modalities to monitor transplanted organs is of great clinical value, particularly as a tool for early detection of graft dysfunction or acute rejection. Herein, we provide an overview of the existing literature evaluating noninvasive imaging modalities of solid organ and cellular allografts after transplantation, including both preclinical and clinical studies.


Asunto(s)
Trasplante de Corazón , Trasplante de Riñón , Biopsia , Rechazo de Injerto/diagnóstico , Trasplante Homólogo
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA