Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.624
Filtrar
1.
Transplantation ; 108(5): 1115-1126, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38192025

RESUMEN

BACKGROUND: The utilization of islet-like cells derived from pluripotent stem cells may resolve the scarcity of islet transplantation donors. The subcutaneous space is a promising transplantation site because of its capacity for graft observation and removal, thereby ensuring safety. To guarantee subcutaneous islet transplantation, physicians should ensure ample blood supply. Numerous methodologies, including prevascularization, have been investigated to augment blood flow, but the optimal approach remains undetermined. METHODS: From C57BL/6 mice, 500 syngeneic islets were transplanted into the prevascularized subcutaneous site of recipient mice by implanting agarose rods with basic fibroblast growth factor at 1 and 2 wk. Before transplantation, the blood glucose levels, cell infiltration, and cytokine levels at the transplant site were evaluated. Furthermore, we examined the impact of the extracellular matrix capsule on graft function and the inflammatory response. RESULTS: Compared with the 1-wk group, the 2-wk group exhibited improved glycemic control, indicating that longer prevascularization enhanced transplant success. Flow cytometry analysis detected immune cells, such as neutrophils and macrophages, in the extracellular matrix capsules, whereas cytometric bead array analysis indicated the release of inflammatory and proinflammatory cytokines. Treatment with antitumor necrosis factor and anti-interleukin-6R antibodies in the 1-wk group improved graft survival, similar to the 2-wk group. CONCLUSIONS: In early prevascularization before subcutaneous transplantation, neutrophil and macrophage accumulation prevented early engraftment owing to inflammatory cytokine production.


Asunto(s)
Glucemia , Citocinas , Supervivencia de Injerto , Trasplante de Islotes Pancreáticos , Ratones Endogámicos C57BL , Trasplante de Islotes Pancreáticos/métodos , Trasplante de Islotes Pancreáticos/inmunología , Animales , Glucemia/metabolismo , Citocinas/metabolismo , Ratones , Masculino , Factores de Tiempo , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/cirugía , Tejido Subcutáneo/irrigación sanguínea , Tejido Subcutáneo/inmunología , Matriz Extracelular/metabolismo , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/irrigación sanguínea , Neovascularización Fisiológica
2.
Biomed Pharmacother ; 142: 112042, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34403963

RESUMEN

Mesenchymal stem cells (MSCs) therapy has brought a great enthusiasm to the treatment of various immune disorders, tissue regeneration and transplantation therapy. MSCs are being extensively investigated for their immunomodulatory actions. MSCs can deliver immunomodulatory signals to inhibit allogeneic T cell immune responses by downregulating pro-inflammatory cytokines and increasing regulatory cytokines and growth factors. Islet transplantation is a therapeutic alternative to the insulin therapy for the treatment of type 1 diabetes mellitus (T1DM). However, the acute loss of islets due to the lack of vasculature and hypoxic milieu in the immediate post-transplantation period may lead to treatment failure. Moreover, despite the use of potent immunosuppressive drugs, graft failure persists because of immunological rejection. Many in vitro and in vivo researches have demonstrated the multipotency of MSCs as a mediator of immunomodulation and a great approach for enhancement of islet engraftment. MSCs can interact with immune cells of the innate and adaptive immune systems via direct cell-cell contact or through secretomes containing numerous soluble growth and immunomodulatory factors or mitochondrial transfer. This review highlights the interactions between MSCs and different immune cells to mediate immunomodulatory functions along with the importance of MSCs therapy for the successful islet transplantation.


Asunto(s)
Inmunomodulación/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/inmunología , Animales , Exosomas/inmunología , Humanos , Linfocitos/inmunología , Esferoides Celulares/inmunología
3.
PLoS One ; 16(6): e0253029, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34138941

RESUMEN

Understanding the anti-carbohydrate antibody response toward epitopes expressed on porcine cells, tissues, and organs is critical to advancing xenotransplantation toward clinical application. In this study, we determined IgM and IgG antibody specificities and relative concentrations in five cynomolgus monkeys at baseline and at intervals following intraportal xenotransplantation of adult porcine islets. This study utilized a carbohydrate antigen microarray that comprised more than 400 glycoconjugates, including historically reported α-Gal and non-α-Gal carbohydrate antigens with various modifications. The elicited anti-carbohydrate antibody responses were predominantly IgM compared to IgG in 4 out of 5 monkeys. Patterns of elicited antibody responses greater than 1.5 difference (log2 base units; 2.8-fold on a linear scale) from pre-serum to post-serum sampling specific for carbohydrate antigens were heterogeneous and recipient-specific. Increases in the elicited antibody response to α-Gal, Sda, GM2 antigens, or Lexis X antigen were found in individual monkeys. The novel carbohydrate structures Galß1-4GlcNAcß1-3Galß1 and N-linked glycans with Manα1-6(GlcNAcß1-2Manα1-3)Manß1-4GlcNAcß structure were common targets of elicited IgM antibodies. These results provide important insights into the carbohydrate epitopes that elicit antibodies following pig-to-monkey islet xenotransplantation and reveal possible targets for gene editing.


Asunto(s)
Carbohidratos/análisis , Rechazo de Injerto/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Trasplante de Islotes Pancreáticos/inmunología , Animales , Secuencia de Carbohidratos , Carbohidratos/genética , Carbohidratos/inmunología , Rechazo de Injerto/sangre , Macaca fascicularis , Masculino , Análisis por Micromatrices , Porcinos , Trasplante Heterólogo
4.
J Mater Chem B ; 9(11): 2631-2640, 2021 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-33683280

RESUMEN

Islet cell transplantation has been an effective method for the treatment of type 1 diabetes. The transplanted islets release insulin in response to changes in blood glucose levels. The clinical application of islet transplantation, however, has been hindered because of some critical problems including immune responses to grafted islets and side effects caused by overdosed immunosuppressive drugs. Herein, surface modification technology using poly(ethylene glycol) (PEG)-dendron was proposed to safeguard islets from the host immune system. PEG-dendron was synthesized by a divergent polymerization method and utilized to cover the islet antigen surface. Successful conjugation of PEG-dendron on the islet surface was achieved without affecting islet morphology, viability, and functionality at a concentration of 1.00%. Surface modification using PEG-dendron effectively prevented protein absorption and immune activation. Foremost, it improved the survival rate of islet grafts in vivo when combined with a low dose of immunosuppressive drugs. In conclusion, PEG-dendron is a potential candidate for the surface modification of pancreatic islets to mitigate immune responses after transplantation.


Asunto(s)
Dendrímeros/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Inmunosupresores/farmacología , Islotes Pancreáticos/efectos de los fármacos , Polietilenglicoles/farmacología , Animales , Dendrímeros/química , Diabetes Mellitus Experimental/inmunología , Inmunosupresores/química , Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Polietilenglicoles/química , Ratas , Ratas Sprague-Dawley
5.
Diabetes ; 70(1): 171-181, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33122392

RESUMEN

The induction of antigen (Ag)-specific tolerance represents a therapeutic option for autoimmune diabetes. We demonstrated that administration of a lentiviral vector enabling expression of insulin B chain 9-23 (InsB9-23) (LV.InsB) in hepatocytes arrests ß-cell destruction in prediabetic NOD mice by generating InsB9-23-specific FoxP3+ T regulatory cells (Tregs). LV.InsB in combination with a suboptimal dose of anti-CD3 monoclonal antibody (combined therapy [CT], 1 × 5 µg [CT5]) reverts diabetes and prevents recurrence of autoimmunity after islet transplantation in ∼50% of NOD mice. We investigated whether CT optimization could lead to abrogation of recurrence of autoimmunity. Therefore, alloislets were transplanted after optimized CT tolerogenic conditioning (1 × 25 µg [CT25]). Diabetic NOD mice conditioned with CT25 when glycemia was <500 mg/dL remained normoglycemic for 100 days after alloislet transplantation and displayed reduced insulitis, but independently from the graft. Accordingly, cured mice showed T-cell unresponsiveness to InsB9-23 stimulation and increased Treg frequency in islet infiltration and pancreatic lymph nodes. Additional studies revealed a complex mechanism of Ag-specific immune regulation driven by CT25, in which both Tregs and PDL1 costimulation cooperate to control diabetogenic cells, while transplanted islets play a crucial role, although transient, recruiting diabetogenic cells. Therefore, CT25 before alloislet transplantation represents an Ag-specific immunotherapy to resolve autoimmune diabetes in the presence of residual endogenous ß-cell mass.


Asunto(s)
Diabetes Mellitus Tipo 1/cirugía , Hepatocitos/metabolismo , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Péptidos/uso terapéutico , Animales , Autoinmunidad/efectos de los fármacos , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Técnicas de Transferencia de Gen , Supervivencia de Injerto/inmunología , Hepatocitos/inmunología , Células Secretoras de Insulina/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Recurrencia , Prevención Secundaria , Linfocitos T Reguladores/inmunología
6.
Int J Mol Sci ; 21(13)2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32629968

RESUMEN

Cytotoxic T lymphocytes (CTL) are an essential part of our immune system by killing infected and malignant cells. To fully understand this process, it is necessary to study CTL function in the physiological setting of a living organism to account for their interplay with other immune cells like CD4+ T helper cells and macrophages. The anterior chamber of the eye (ACE), originally developed for diabetes research, is ideally suited for non-invasive and longitudinal in vivo imaging. We take advantage of the ACE window to observe immune responses, particularly allorejection of islets of Langerhans cells by CTLs. We follow the onset of the rejection after vascularization on islets until the end of the rejection process for about a month by repetitive two-photon microscopy. We find that CTLs show reduced migration on allogeneic islets in vivo compared to in vitro data, indicating CTL activation. Interestingly, the temporal infiltration pattern of T cells during rejection is precisely regulated, showing enrichment of CD4+ T helper cells on the islets before arrival of CD8+ CTLs. The adaptation of the ACE to immune responses enables the examination of the mechanism and regulation of CTL-mediated killing in vivo and to further investigate the killing in gene-deficient mice that resemble severe human immune diseases.


Asunto(s)
Cámara Anterior/inmunología , Rechazo de Injerto/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Linfocitos T Citotóxicos/fisiología , Animales , Ratones Endogámicos DBA
7.
Cell Transplant ; 29: 963689720913876, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32216448

RESUMEN

Anti-CD154 blockade-based regimens remain unequaled in prolonging graft survival in various organ transplantation models. Several studies have focused on transplantation tolerance with the anti-CD154 blockade, but none of these studies has investigated the mechanisms associated with its use as the sole treatment in animal models, delaying our understanding of anti-CD154 blockade-mediated immune tolerance. The purpose of this study was to investigate the mechanism underlying the anti-CD154 monoclonal antibody (mAb) blockade in inducing immune tolerance using an intrahepatic murine allogeneic islet transplantation model. Allogeneic BALB/c AnHsd (BALB/c) islets were infused into the liver of diabetic C57BL/6 (B6) mice via the cecal vein. Anti-CD154 mAb (MR1) was administered on -1, 0, 1, 3, 5, and 7 d posttransplantation at 0.5 mg per mouse. We showed that short-term MR1 monotherapy could prolong the allogeneic islet grafts to more than 250 d in the murine intrahepatic islet transplantation model. The second islet grafts transplanted under the kidney capsule of the recipients were protected from rejection. We also found that rejection of same-donor skin grafts transplanted to the tolerant mice was modestly delayed. Using a DEREG mouse model, FoxP3+ regulatory T (Treg) cells were shown to play important roles in transplantation tolerance. In mixed lymphocyte reactions, Treg cells from the tolerant mice showed more potency in suppressing BALB/c splenocyte-stimulated Teff cell proliferation than those from naïve mice. In this study, we demonstrated for the first time that a short-term anti-CD154 mAb single treatment could induce FoxP3+ Treg cell-mediated immune tolerance in the intrahepatic murine allogeneic islet transplantation model.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Rechazo de Injerto/tratamiento farmacológico , Tolerancia Inmunológica/efectos de los fármacos , Trasplante de Islotes Pancreáticos/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Animales , Rechazo de Injerto/inmunología , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante de Piel/métodos , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante , Trasplante Homólogo/métodos
8.
J Diabetes Sci Technol ; 14(2): 212-225, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32116026

RESUMEN

Islet transplantation is a promising curative treatment option for type 1 diabetes (T1D) as it can provide physiological blood glucose control. The widespread utilization of islet transplantation is limited due to systemic immunosuppression requirements, persisting graft immunodestruction, and poor islet engraftment. Traditional macro- and micropolymeric encapsulation strategies can alleviate the need for antirejection immunosuppression, yet the increased graft volume and diffusional distances imparted by these coatings can be detrimental to graft viability and glucose control. Additionally, systemic administration of pro-engraftment and antirejection therapeutics leaves patients vulnerable to adverse off-target side effects. Nanoscale engineering techniques can be used to immunocamouflage islets, modulate the transplant microenvironment, and provide localized pro-engraftment cues. In this review, we discuss the applications of nanotechnology to advance the clinical potential of islet transplantation, with a focus on cell surface engineering, bioactive functionalization, and use of nanoparticles in T1D cell-based treatments.


Asunto(s)
Trasplante de Células , Diabetes Mellitus Tipo 1/terapia , Inmunidad , Nanotecnología/métodos , Animales , Trasplante de Células/efectos adversos , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/inmunología , Supervivencia de Injerto/inmunología , Humanos , Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Nanotecnología/tendencias , Inmunología del Trasplante
9.
Transplant Proc ; 52(3): 982-986, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32113692

RESUMEN

Immunologic and nonimmunologic mechanisms contribute to islet loss on transplantation in the liver. A site outside of the vascular bed may act as a sanctuary site avoiding immediate inflammatory response. We developed a unique approach by using a human amniotic membrane (HAM) for islet transplant onto the liver surface by combining it with human amniotic epithelial cells (HAECs). Decellularized 1.0 × 1.0-cm HAM was placed on the surface of the liver; 2000 islet equivalent of human islets mixed with 0.4 × 106 HAECs were infused into the space between the membrane and surface. Two pieces of the decellularized HAM were placed under the subcutaneous space to study the angiogenic potential. After cotransplant 57.1% of recipient mice became normoglycemic by the third day after transplant, and 100% attained euglycemia 2 weeks post-transplant. Bodyweight of 85.7% of mice gradually increased over time. The HAM and islets were identified in histologic section in all. Neovascularization was observed to be dramatically increased. In conclusion, HAM is a very versatile biological membrane; HAECs help angiogenesis and better engraftment of islets on the liver surface, eliminating the need for vascular deployment of islet cells and avoiding the risk of portal vein thrombosis and instant blood-mediated inflammatory reaction-related islet loss in the liver.


Asunto(s)
Amnios , Células Epiteliales , Trasplante de Islotes Pancreáticos/métodos , Animales , Supervivencia de Injerto , Humanos , Trasplante de Islotes Pancreáticos/inmunología , Hígado , Ratones
10.
Front Immunol ; 11: 612737, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33658995

RESUMEN

Pancreatic islet transplantation is a promising method for the treatment of type 1 and type 3 diabetes whereby replacement of islets may be curative. However, long-term treatment with immunosuppressive drugs (ISDs) remains essential for islet graft survival. Current ISD regimens carry significant side-effects for transplant recipients, and are also toxic to the transplanted islets. Pre-clinical efforts to induce immune tolerance to islet allografts identify ways in which the recipient immune system may be reeducated to induce a sustained transplant tolerance and even overcome autoimmune islet destruction. The goal of these efforts is to induce tolerance to transplanted islets with minimal to no long-term immunosuppression. Two most promising cell-based therapeutic strategies for inducing immune tolerance include T regulatory cells (Tregs) and donor and recipient hematopoietic mixed chimerism. Here, we review preclinical studies which utilize Tregs for tolerance induction in islet transplantation. We also review myeloablative and non-myeloablative hematopoietic stem cell transplantation (HSCT) strategies in preclinical and clinical studies to induce sustained mixed chimerism and allograft tolerance, in particular in islet transplantation. Since Tregs play a critical role in the establishment of mixed chimerism, it follows that the combination of Treg and HSCT may be synergistic. Since the success of the Edmonton protocol, the feasibility of clinical islet transplantation has been established and nascent clinical trials testing immune tolerance strategies using Tregs and/or hematopoietic mixed chimerism are underway or being formulated.


Asunto(s)
Tolerancia Inmunológica/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante/inmunología , Animales , Quimerismo , Supervivencia de Injerto/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Inmunosupresores/inmunología
11.
J Clin Invest ; 130(1): 287-294, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31763998

RESUMEN

Activation of host T cells that mediate allograft rejection is a 2-step process. The first occurs in secondary lymphoid organs where T cells encounter alloantigens presented by host DCs and differentiate to effectors. Antigen presentation at these sites occurs principally via transfer of intact, donor MHC-peptide complexes from graft cells to host DCs (cross-dressing) or by uptake and processing of donor antigens into allopeptides bound to self-MHC molecules (indirect presentation). The second step takes place in the graft, where effector T cells reengage with host DCs before causing rejection. How host DCs present alloantigens to T cells in the graft is not known. Using mouse islet and kidney transplantation models, imaging cytometry, and 2-photon intravital microscopy, we demonstrate extensive cross-dressing of intragraft host DCs with donor MHC-peptide complexes that occurred early after transplantation, whereas host DCs presenting donor antigen via the indirect pathway were rare. Cross-dressed DCs stably engaged TCR-transgenic effector CD8+ T cells that recognized donor antigen and were sufficient for sustaining acute rejection. In the chronic kidney rejection model, cross-dressing declined over time but was still conspicuous 8 weeks after transplantation. We conclude that cross-dressing of host DCs with donor MHC molecules is a major antigen presentation pathway driving effector T cell responses within allografts.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Rechazo de Injerto/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Riñón , Activación de Linfocitos , Aloinjertos , Animales , Linfocitos T CD8-positivos/patología , Células Dendríticas/patología , Rechazo de Injerto/patología , Ratones , Ratones Noqueados , Inmunología del Trasplante
12.
Islets ; 11(5): 119-128, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31483188

RESUMEN

Islet transplantation is efficacious to prevent severe hypoglycemia and glycemic liability of selected patients of type 1 diabetes. However, since calcineurin inhibitor (CNI) causes ß-cell and nephrotoxicity, alternative drug(s) with similar potency and safety profile to CNI will be highly desirable. Here we tested whether JAK3 inhibitor, tofacitinib could be used instead of tacrolimus in CIT07 immunosuppression regimen in cynomolgus nonhuman primate (NHP) model. Five independent streptozotocin (STZ)-induced diabetic monkeys were transplanted with MHC-mismatched allogeneic islets and three animals were further re-transplanted upon insufficient glycemic control or early islet graft rejection. After islet transplantation, blood glucose levels were quickly stabilized and maximal islet graft survival as measured by serum C-peptide concentration was >330, 98, >134, 31, or 22 days, respectively, after transplantation (median survival day; 98 days). Cellular and humoral immune responses were efficiently suppressed by JAK3 inhibitor-based immunosuppression during the follow-up periods. Although intermittent increases of the genome copy number of cynomolgus cytomegalovirus (CMV) were detected by quantitative real-time PCR analyses, serious infections or posttransplant lymphoproliferative disease (PTLD) was not found in all animals. Taken together, we have shown that JAK3 inhibitor could be used in replacement of tacrolimus in a highly translatable NHP islet transplantation model and these results suggest that JAK3 inhibitor will be potentially incorporated in human allogeneic islet transplantation.


Asunto(s)
Terapia de Inmunosupresión/métodos , Inmunosupresores/uso terapéutico , Trasplante de Islotes Pancreáticos , Janus Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Evaluación Preclínica de Medicamentos , Femenino , Rechazo de Injerto/inmunología , Supervivencia de Injerto/efectos de los fármacos , Terapia de Inmunosupresión/veterinaria , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/métodos , Macaca fascicularis , Masculino , Acondicionamiento Pretrasplante/métodos , Acondicionamiento Pretrasplante/veterinaria , Inmunología del Trasplante/efectos de los fármacos , Trasplante Heterólogo
14.
J Control Release ; 305: 176-193, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31029742

RESUMEN

Islet transplantation is an alternative method of replacing exogenous insulin to treat type 1 diabetes. However, transplantation of allo- or xenograft islets causes the activation of host's immune reaction, which leads to the failure of the transplanted grafts. Immunosuppressive-sparing strategies have been introduced to avoid adverse effects associated with a long-term use of the immunosuppressive drugs. In this regard, macro/microencapsulation, surface camouflage, and surface modification with immune-privileged cells have been performed to protect the transplanted islets against instant blood-mediated inflammatory reactions or immune reactions. However, the increased size of the encapsulated islets after transplantation leads to insufficient oxygen and nutrients for the islets, causing most of them to undergo apoptosis. Therefore, recent studies have aimed at reducing the capsule thickness while maintaining immunoprotective ability of encapsulated islets. In this review, we discuss several techniques of thin-layer surface coating of pancreatic islets using a variety of polymers, therapeutic agents (TA), TA-loaded nano or microparticles, and living cells.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Rechazo de Injerto/prevención & control , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/inmunología , Animales , Anticoagulantes/uso terapéutico , Células Inmovilizadas/citología , Células Inmovilizadas/inmunología , Células Inmovilizadas/trasplante , Diabetes Mellitus Tipo 1/inmunología , Rechazo de Injerto/inmunología , Humanos , Islotes Pancreáticos/citología , Trasplante de Islotes Pancreáticos/inmunología , Polietilenglicoles/uso terapéutico
15.
Front Immunol ; 10: 742, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024566

RESUMEN

The IL-7/IL-7R pathway is essential for lymphocyte development and disturbances in the pathway can lead to immune deficiency or T cell mediated destruction. Here, the effect of transient hyperexpression of IL-7 was investigated on immune regulation and allograft rejection under immunosuppression. An experimental in vivo immunosuppressive mouse model of IL-7 hyperexpression was developed using transgenic mice (C57BL/6 background) carrying a tetracycline inducible IL-7 expression cassette, which allowed the temporally controlled induction of IL-7 hyperexpression by Dexamethasone and Doxycycline treatment. Upon induction of IL-7, the B220+ c-kit+ Pro/Pre-B I compartment in the bone marrow increased as compared to control mice in a serum IL-7 concentration-correlated manner. IL-7 hyperexpression also preferentially increased the population size of memory CD8+ T cells in secondary lymphoid organs, and reduced the proportion of CD4+Foxp3+ T regulatory cells. Of relevance to disease, conventional CD4+ T cells from an IL-7-rich milieu escaped T regulatory cell-mediated suppression in vitro and in a model of autoimmune diabetes in vivo. These findings were validated using an IL-7/anti-IL7 complex treatment mouse model to create an IL-7 rich environment. To study the effect of IL-7 on islet graft survival in a mismatched allograft model, BALB/c mice were rendered diabetic by streptozotocin und transplanted with IL-7-inducible or control islets from C57BL/6 mice. As expected, Dexamethasone and Doxycycline treatment prolonged graft median survival as compared to the untreated control group in this transplantation mouse model. However, upon induction of local IL-7 hyperexpression in the transplanted islets, graft survival time was decreased and this was accompanied by an increased CD4+ and CD8+ T cell infiltration in the islets. Altogether, the findings show that transient elevations of IL-7 can impair immune regulation and lead to graft loss also under immune suppression.


Asunto(s)
Rechazo de Injerto/inmunología , Interleucina-7/biosíntesis , Linfocitos T/inmunología , Animales , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/terapia , Femenino , Rechazo de Injerto/etiología , Rechazo de Injerto/genética , Supervivencia de Injerto/inmunología , Homeostasis/inmunología , Memoria Inmunológica , Inmunosupresores/farmacología , Interleucina-7/genética , Trasplante de Islotes Pancreáticos/efectos adversos , Trasplante de Islotes Pancreáticos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Inmunológicos , Células Precursoras de Linfocitos B/inmunología , Subgrupos de Linfocitos T/inmunología , Tolerancia al Trasplante/inmunología , Trasplante Homólogo , Regulación hacia Arriba
16.
Transpl Int ; 32(9): 903-912, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31033036

RESUMEN

Pancreatic islet allotransplantation is a treatment for patients with severe forms of type 1 diabetes. As long-term graft function and survival are not yet optimal, additional studies are warranted in order to continue improving transplant outcomes. The mechanisms of islet graft loss and tolerance induction are often studied in murine diabetes models. Despite numerous islet transplantation studies successfully performed over recent years, translation from experimental mouse models to human clinical application remains elusive. This review aims at critically discussing the strengths and limitations of current mouse models of diabetes and experimental islet transplantation. In particular, we will analyze the causes leading to diabetes and compare the immunological mechanisms responsible for rejection between mouse and human. A better understanding of the experimental mouse models should facilitate translation to human clinical application.


Asunto(s)
Trasplante de Islotes Pancreáticos/inmunología , Animales , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Humanos , Ratones , Trasplante Homólogo
17.
Sci Rep ; 9(1): 3918, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30850640

RESUMEN

The anterior chamber of the eye (ACE) has emerged as a promising clinical islet transplantation site because of its multiple advantages over the conventional intra-hepatic portal site. This includes reduced surgical invasiveness and increased islet graft survival rate. It also allows for enhanced accessibility and monitoring of the islets. Although the ACE is initially an immuno-privileged site, this privilege is disrupted once the islet grafts are re-vascularized. Given that the ACE is a confined space, achieving graft immune tolerance through local immunosuppressive drug delivery is therefore feasible. Here, we show that islet rejection in the ACE of mice can be significantly suppressed through local delivery of rapamycin by carefully designed sustained-release microparticles. In this 30-day study, allogeneic islet grafts with blank microparticles were completely rejected 18 days post-transplantation into mice. Importantly, allogeneic islet grafts co-injected with rapamycin releasing microparticles into a different eye of the same recipient were preserved much longer, with some grafts surviving for more than 30 days. Hence, islet allograft survival was enhanced by a localized and prolonged delivery of an immunosuppressive drug. We envisage that this procedure will relieve diabetic transplant recipients from harsh systemic immune suppression, while achieving improved glycemic control and reduced insulin dependence.


Asunto(s)
Cámara Anterior , Rechazo de Injerto/prevención & control , Inmunosupresores/administración & dosificación , Trasplante de Islotes Pancreáticos/métodos , Sirolimus/administración & dosificación , Aloinjertos , Animales , Cámara Anterior/efectos de los fármacos , Cámara Anterior/inmunología , Cámara Anterior/cirugía , Preparaciones de Acción Retardada , Diabetes Mellitus/inmunología , Diabetes Mellitus/cirugía , Sistemas de Liberación de Medicamentos , Supervivencia de Injerto/efectos de los fármacos , Humanos , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Modelos Animales , Tolerancia al Trasplante/efectos de los fármacos
18.
Biomaterials ; 192: 271-281, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30458362

RESUMEN

Intraportal allogeneic islet transplantation has been demonstrated as a potential therapy for type 1 diabetes (T1D). The placement of islets into the liver and chronic immunosuppression to control rejection are two major limitations of islet transplantation. We hypothesize that localized immunomodulation with a novel form of FasL chimeric with streptavidin, SA-FasL, can provide protection and long-term function of islets at an extrahepatic site in the absence of chronic immunosuppression. Allogeneic islets modified with biotin and engineered to transiently display SA-FasL on their surface showed sustained survival following transplantation on microporous scaffolds into the peritoneal fat in combination with a short course (15 days) of rapamycin treatment. The challenges with modifying islets for clinical translation motivated the modification of scaffolds with SA-FasL as an off-the-shelf product. Poly (lactide-co-glycolide) (PLG) was conjugated with biotin and fabricated into particles and subsequently formed into microporous scaffolds to allow for rapid and efficient conjugation with SA-FasL. Biotinylated particles and scaffolds efficiently bound SA-FasL and induced apoptosis in cells expressing Fas receptor (FasR). Scaffolds functionalized with SA-FasL were subsequently seeded with allogeneic islets and transplanted into the peritoneal fat under the short-course of rapamycin treatment. Scaffolds modified with SA-FasL had robust engraftment of the transplanted islets that restored normoglycemia for 200 days. Transplantation without rapamycin or without SA-FasL did not support long-term survival and function. This work demonstrates that scaffolds functionalized with SA-FasL support allogeneic islet engraftment and long-term survival and function in an extrahepatic site in the absence of chronic immunosuppression with significant potential for clinical translation.


Asunto(s)
Proteína Ligando Fas/inmunología , Proteínas Inmovilizadas/inmunología , Tolerancia Inmunológica , Trasplante de Islotes Pancreáticos/métodos , Andamios del Tejido , Animales , Supervivencia de Injerto , Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Proteínas Recombinantes/inmunología , Estreptavidina/inmunología , Andamios del Tejido/química
19.
FASEB J ; 33(3): 3137-3151, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30383447

RESUMEN

Immunodeficient mice engrafted with human peripheral blood mononuclear cells (PBMCs) support preclinical studies of human pathogens, allograft rejection, and human T-cell function. However, a major limitation of PBMC engraftment is development of acute xenogeneic graft- versus-host disease (GVHD) due to human T-cell recognition of murine major histocompatibility complex (MHC). To address this, we created 2 NOD- scid IL-2 receptor subunit γ ( IL2rg) null (NSG) strains that lack murine MHC class I and II [NSG-ß-2-microglobulin ( B2M) null ( IA IE)null and NSG -( Kb Db) null ( IAnull)]. We observed rapid human IgG clearance in NSG- B2Mnull ( IA IE) null mice whereas clearance in NSG -( Kb Db) null ( IAnull) mice and NSG mice was comparable. Injection of human PBMCs into both strains enabled long-term engraftment of human CD4+ and CD8+ T cells without acute GVHD. Engrafted human T-cell function was documented by rejection of human islet allografts. Administration of human IL-2 to NSG -( Kb Db) null ( IAnull) mice via adeno-associated virus vector increased human CD45+ cell engraftment, including an increase in human regulatory T cells. However, high IL-2 levels also induced the development of GVHD. These data document that NSG mice deficient in murine MHC support studies of human immunity in the absence of acute GVHD and enable evaluation of human antibody therapeutics targeting human T cells.-Brehm, M. A., Kenney, L. L., Wiles, M. V., Low, B. E., Tisch, R. M., Burzenski, L., Mueller, C., Greiner, D. L., Shultz, L. D. Lack of acute xenogeneic graft- versus-host disease, but retention of T-cell function following engraftment of human peripheral blood mononuclear cells in NSG mice deficient in MHC class I and II expression.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/trasplante , Linfocitos T/inmunología , Animales , Femenino , Genes MHC Clase I , Genes MHC Clase II , Supervivencia de Injerto/inmunología , Xenoinjertos , Humanos , Trasplante de Islotes Pancreáticos/inmunología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Fenotipo
20.
Xenotransplantation ; 26(2): e12474, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30461074

RESUMEN

BACKGROUND: Aberrant microRNA (miRNA) expression levels are associated with various graft rejections. We used our humanized mouse model with transplanted human islets to identify miRNAs in islet grafts related to xenograft rejection and circulating miRNAs associated with xenograft rejection-mediated ß-cell loss. METHODS: Diabetic immunodeficient NOD.scid mice were transplanted with human islets and subsequently achieved stable normoglycemia. Lymphocytes from NOD mice were then adoptively transferred to the humanized mice to induce human ß-cell destruction. Islet graft and plasma were collected immediately once blood glucose reached >200 mg/dL. miRNAs in the islet grafts and in the plasma with or without adoptive lymphocyte transfer (ALT) were measured using NanoString nCounter® miRNA Expression Assay and qPCR. RESULTS: A set of immune-related miRNAs was significantly increased in human islet grafts of ALT-treated mice compared to control mice. Of these miRNAs, miR-150-5p was significantly increased in the circulation of ALT-treated mice at tissue collection and the increase was a result of immune activation rather than simply the presence of lymphocytes in circulation. Furthermore, miR-150-5p was significantly increased in human islet graft and circulation prior to the development of hyperglycemia in the ALT-treated mice. CONCLUSIONS: Our data demonstrated that immune-related miRNAs are associated with human islet xenograft rejection in mice. miR-150-5p is increased in human islet graft and in the circulation during islet xenograft rejection and ß-cell destruction prior to hyperglycemia and may be an early biomarker for islet xenograft rejection.


Asunto(s)
Trasplante de Islotes Pancreáticos/inmunología , Linfocitos/inmunología , MicroARNs/genética , Trasplante Heterólogo , Animales , Modelos Animales de Enfermedad , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Xenoinjertos/inmunología , Humanos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos NOD , MicroARNs/inmunología , Trasplante Heterólogo/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...