Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
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
2.
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.

3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Front Oncol ; 9: 418, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31293961

RESUMEN

The absence of clinically relevant large animal tumor models has historically forced experimental cellular therapies for hematological malignancies to translate directly from murine models to clinical trials. However, recent advances highlight swine as an ideal large animal model to demonstrate the safety of murine proof of concept studies prior to their implementation clinically. The availability of the MHC defined MGH miniature swine herd has been key for the development of novel approaches for hematopoietic cell and solid organ transplantation. New spontaneously arising hematological malignancies in these swine, specifically myeloid leukemias and B cell lymphomas, resemble human malignancies, which has allowed for development of immortalized tumor cell lines and has implications for the development of a large animal transplantable tumor model. The novel development of a SCID swine model has further advanced the field of large animal cancer models, allowing for engraftment of human tumor cells in a large animal model. Here, we will highlight the advantages of the swine pre-clinical model for the study of hematological malignancies. Further, we will discuss our experience utilizing spontaneously arising tumors in MGH swine to create a transplantable tumor model, describe the potential of the immunodeficient swine model, and highlight several novel cellular and biological therapies for the treatment of hematological malignancies in swine as a large animal pre-clinical bridge.

13.
Front Oncol ; 9: 209, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31001475

RESUMEN

The lack of a reliable and reproducible large animal tumor model for the study of hemolymphatic malignancies limits the ability to explore the underlying pathophysiology and testing of novel therapies. The goal of this study was to develop an aggressive, trackable swine tumor cell line in mice for adoptive transfer into MHC matched swine. Two tumor cell lines, post-transplant lymphoproliferative disease (PTLD) 13271 and chronic myelogenous leukemia (CML) 14736, were previously established from the Massachusetts General Hospital (MGH) miniature swine herd. PTLD 13271 is a swine B-cell lymphoma line originating from an animal that developed PTLD following hematopoietic cell transplantation (HCT), while CML 14736 was generated from a swine that spontaneously developed CML. In order to select for aggressive tumor variants, both lines were passage into NOD/SCID IL-2 receptor γ-/- (NSG) mice. Tumor induced mortality in mice injected with CML14736 was 68% while 100% of mice injected with PTLD 13271 succumbed to PTLD by day 70. Based on aggressiveness, PTLD 13271 was selected for further development and re-passage into NSG mice resulting in increased tumor burden and metastasis. Transduction of the PTLD 13271 cell line with a green fluorescent protein (GFP)-expressing lentivirus facilitated tumor tracking when re-passaged in mice. Utilizing a tolerance induction strategy, GFP+ tumors were injected into an MHC matched miniature swine and successfully followed via flow cytometry for 48 h in circulation, although tumor engraftment was not observed. In summary, we report the development of an aggressive GFP+B-cell lymphoma cell line which has the potential for facilitating development of a large animal tumor model.

14.
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
15.
J Immunol Regen Med ; 1: 45-56, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30364570

RESUMEN

The immunosuppressive capacity of human mesenchymal stromal cells (MSCs) renders them promising candidates for treating diverse immune disorders. However, after hundreds of clinical trials, there are still no MSC therapies approved in the United States. MSCs require specific cues to adopt their immunosuppressive phenotype, and yet most clinical trials use cells expanded in basic culture medium and growth conditions. We propose that priming MSCs prior to administration will improve their therapeutic efficacy. Interferon-gamma (IFN-γ) priming are cues common to situations of immune escape that have individually shown promise as MSC priming cues but have not been systematically compared. Using mixed lymphocyte reactions, we show that priming MSCs with either cue alone improves T-cell inhibition. However, combining the two cues results in additive effects and markedly enhances the immunosuppressive phenotype of MSCs. We demonstrate that IFN-γ induces expression of numerous immunosuppressive proteins (IDO, PD-L1, HLA-E, HLA-G), whereas hypoxia switches MSCs to glycolysis, causing rapid glucose consumption and production of T-cell inhibitory lactate levels. Dual IFN-γ/hypoxia primed MSCs display both attributes and have even higher induction of immunosuppressive proteins over IFN-γ priming alone (IDO and HLA-G), which may reflect another benefit of metabolic reconfiguration.

16.
Biomaterials ; 167: 226-234, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29574308

RESUMEN

Over the past 15 years, mesenchymal stem cells (MSCs) have been assessed for their capacity to suppress inflammation and promote tissue repair. Regardless of whether the cells are primed (exposed to instructive cues) before administration, their phenotype will respond to environmental signals present in the pathophysiological setting being treated. Since hypoxia and inflammation coexist in the settings of acute injury and chronic disease we sought to explore how the proteome and metabolome of MSCs changes when cells were exposed to 48 h of 1% oxygen, interferon gamma (IFN-γ), or both cues together. We specifically focused on changes in cell metabolism, immune modulation, extracellular matrix secretion and modification, and survival capacity. IFN-γ promoted expression of anti-pathogenic proteins and induced MSCs to limit inflammation and fibrosis while promoting their own survival. Hypoxia instead led to cell adaptation to low oxygen, including upregulation of proteins involved in anaerobic metabolism, autophagy, angiogenesis, and cell migration. While dual priming resulted in additive effects, we also found many instances of synergy. These data lend insight to how MSCs may behave after administration to a patient and suggest how priming cells beforehand could improve their therapeutic capacity.


Asunto(s)
Hipoxia/inmunología , Interferón gamma/inmunología , Células Madre Mesenquimatosas/inmunología , Metaboloma , Proteoma/inmunología , Hipoxia de la Célula , Supervivencia Celular , Células Cultivadas , Humanos , Hipoxia/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo
17.
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
18.
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
19.
Comp Med ; 66(4): 308-23, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27538862

RESUMEN

Cynomolgus macaques (CYNO; Macaca fascicularis) are a well-established NHP model used for studies in immunology. To provide reference values on the baseline cell distributions in the hematopoietic and lymphoid organs (HLO) of these animals, we used flow cytometry to analyze the peripheral blood, bone marrow, mesenteric lymph nodes, spleen, and thymus of a cohort of male, adult, research-naïve, Mauritian CYNO. Our findings demonstrate that several cell distribution patterns differ between CYNO and humans. First, the CD4(+):CD8(+) T-cell ratio is lower in CYNO compared with humans. Second, the peripheral blood of CYNO contains a population of CD4(+)CD8(+) T cells. Third, the CD31 level was elevated in all organs studied, suggesting that CD31 may not be an accurate marker of recent thymic emigrants within the CD4(+) T cells of CYNO. Finally the B-cell population is lower in CYNO compared with humans. In summary, although the majority of immune cell populations are similar between cynomolgus macaques and humans, several important differences should be considered when using CYNO in immunologic studies. Our current findings provide valuable information to not only researchers but also veterinarians working with CYNO at research centers, in zoos, or in the wild.


Asunto(s)
Leucocitos/clasificación , Macaca fascicularis/anatomía & histología , Macaca fascicularis/inmunología , Animales , Antígenos CD34/metabolismo , Linfocitos B/citología , Relación CD4-CD8 , Antígeno CD56/metabolismo , Factores de Transcripción Forkhead/metabolismo , Sistema Hematopoyético/citología , Sistema Hematopoyético/inmunología , Humanos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Leucocitos/citología , Leucocitos/inmunología , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Macaca fascicularis/sangre , Masculino , Monocitos/citología , Especificidad de Órganos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Especificidad de la Especie , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...