Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
Sci Transl Med ; 15(711): eadf6376, 2023 08 30.
Article En | MEDLINE | ID: mdl-37647390

Prior studies of anti-CD40 ligand (CD40L)-based immunosuppression demonstrated effective prevention of islet and kidney allograft rejection in nonhuman primate models; however, clinical development was halted because of thromboembolic complications. An anti-CD40L-specific monoclonal antibody, AT-1501 (Tegoprubart), was engineered to minimize risk of thromboembolic complications by reducing binding to Fcγ receptors expressed on platelets while preserving binding to CD40L. AT-1501 was tested in both a cynomolgus macaque model of intrahepatic islet allotransplantation and a rhesus macaque model of kidney allotransplantation. AT-1501 monotherapy led to long-term graft survival in both islet and kidney transplant models, confirming its immunosuppressive potential. Furthermore, AT-1501-based regimens after islet transplant resulted in higher C-peptide, greater appetite leading to weight gain, and reduced occurrence of cytomegalovirus reactivation compared with conventional immunosuppression. These data support AT-1501 as a safe and effective agent to promote both islet and kidney allograft survival and function in nonhuman primate models, warranting further testing in clinical trials.


Antibodies, Monoclonal , Kidney , Animals , Ligands , Macaca mulatta , Antibodies, Monoclonal/pharmacology , CD40 Ligand , Macaca fascicularis , Allografts
2.
Nat Commun ; 13(1): 7951, 2022 12 26.
Article En | MEDLINE | ID: mdl-36572684

Pancreatic islet transplantation efficacy for type 1 diabetes (T1D) management is limited by hypoxia-related graft attrition and need for systemic immunosuppression. To overcome these challenges, we developed the Neovascularized Implantable Cell Homing and Encapsulation (NICHE) device, which integrates direct vascularization for facile mass transfer and localized immunosuppressant delivery for islet rejection prophylaxis. Here, we investigated NICHE efficacy for allogeneic islet transplantation and long-term diabetes reversal in an immunocompetent, male rat model. We demonstrated that allogeneic islets transplanted within pre-vascularized NICHE were engrafted, revascularized, and functional, reverting diabetes in rats for over 150 days. Notably, we confirmed that localized immunosuppression prevented islet rejection without inducing toxicity or systemic immunosuppression. Moreover, for translatability efforts, we showed NICHE biocompatibility and feasibility of deployment as well as short-term allogeneic islet engraftment in an MHC-mismatched nonhuman primate model. In sum, the NICHE holds promise as a viable approach for safe and effective islet transplantation and long-term T1D management.


Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Islets of Langerhans , Rats , Animals , Male , Diabetes Mellitus, Type 1/therapy , Immunosuppression Therapy , Immune Tolerance , Immunosuppressive Agents/pharmacology , Graft Survival
3.
Sci Adv ; 8(26): eabm3145, 2022 07.
Article En | MEDLINE | ID: mdl-35767620

Polyethylene glycol (PEG)-based conformal coating (CC) encapsulation of transplanted islets is a promising ß cell replacement therapy for the treatment of type 1 diabetes without chronic immunosuppression because it minimizes capsule thickness, graft volume, and insulin secretion delay. However, we show here that our original CC method, the direct method, requiring exposure of islets to low pH levels and inclusion of viscosity enhancers during coating, severely affected the viability, scalability, and biocompatibility of CC islets in nonhuman primate preclinical models of type 1 diabetes. We therefore developed and validated in vitro and in vivo, in several small- and large-animal models of type 1 diabetes, an augmented CC method-emulsion method-that achieves hydrogel CCs around islets at physiological pH for improved cytocompatibility, with PEG hydrogels for increased biocompatibility and with fivefold increase in encapsulation throughput for enhanced scalability.


Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Islets of Langerhans , Animals , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/therapy , Emulsions , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/methods , Primates , Rodentia
4.
Am J Transplant ; 21(11): 3524-3537, 2021 11.
Article En | MEDLINE | ID: mdl-34008325

Mesenchymal stem cells (MSC) have been shown to be immunomodulatory, tissue regenerative, and graft promoting; however, several questions remain with regard to ideal MSC source and timing of administration. In this study, we utilized a rigorous preclinical model of allogeneic islet cell transplantation, incorporating reduced immune suppression and near to complete mismatch of major histocompatibility antigens between the diabetic cynomolgus monkey recipient and the islet donor, to evaluate both the graft promoting impact of MSC source, that is, derived from the islet recipient, the islet donor or an unrelated third party as well as the impact of timing. Co-transplant of MSC and islets on post-operative day 0, followed by additional IV MSC infusions in the first posttransplant month, resulted in prolongation of rejection free and overall islet survival and superior metabolic control for animals treated with recipient as compared to donor or third-party MSC. Immunological analyses demonstrated that infusion of MSC from either source did not prevent alloantibody formation to the islet or MSC donor; however, treatment with recipient MSC resulted in significant downregulation of memory T cells, decreased anti-donor T cell proliferation, and a trend toward increased Tregulatory:Tconventional ratios.


Islets of Langerhans Transplantation , Mesenchymal Stem Cells , Allografts , Animals , Macaca fascicularis , Transplantation, Homologous
5.
Am J Transplant ; 20(3): 689-700, 2020 03.
Article En | MEDLINE | ID: mdl-31597005

Islet cell transplantation can lead to insulin independence, reduced hypoglycemia, and amelioration of diabetes complications in patients with type 1 diabetes. The systemic delivery of anti-inflammatory agents, while considered crucial to limit the early loss of islets associated with intrahepatic infusion, increases the burden of immunosuppression. In an effort to decrease the pharmaceutical load to the patient, we modified the pancreatic islet surface with long-chain poly(ethylene glycol) (PEG) to mitigate detrimental host-implant interactions. The effect of PEGylation on islet engraftment and long-term survival was examined in a robust nonhuman primate model via three paired transplants of dosages 4300, 8300, and 10 000 islet equivalents per kg body weight. A reduced immunosuppressive regimen of anti-thymocyte globulin induction plus tacrolimus in the first posttransplant month followed by maintenance with sirolimus monotherapy was employed. To limit transplant variability, two of the three pairs were closely MHC-matched recipients and received MHC-disparate PEGylated or untreated islets isolated from the same donors. Recipients of PEGylated islets exhibited significantly improved early c-peptide levels, reduced exogenous insulin requirements, and superior glycemic control, as compared to recipients of untreated islets. These results indicate that this simple islet modification procedure may improve islet engraftment and survival in the setting of reduced immunosuppression.


Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Islets of Langerhans , Animals , Graft Survival , Humans , Polyethylene Glycols , Primates , Transplantation, Homologous
6.
Diabetologia ; 62(5): 811-821, 2019 05.
Article En | MEDLINE | ID: mdl-30701283

AIMS/HYPOTHESIS: Patients with autoimmune type 1 diabetes transplanted with pancreatic islets to their liver experience significant improvement in quality of life through better control of blood sugar and enhanced awareness of hypoglycaemia. However, long-term survival and efficacy of the intrahepatic islet transplant are limited owing to liver-specific complications, such as immediate blood-mediated immune reaction, hypoxia, a highly enzymatic and inflammatory environment and locally elevated levels of drugs including immunosuppressive agents, all of which are injurious to islets. This has spurred a search for new islet transplant sites and for innovative ways to achieve long-term graft survival and efficacy without life-long systemic immunosuppression and its complications. METHODS: We used our previously established approach of islet transplant in the anterior chamber of the eye in allogeneic recipient mouse models and a baboon model of diabetes, which were treated transiently with anti-CD154/CD40L blocking antibody in the peri-transplant period. Survival of the intraocular islet allografts was assessed by direct visualisation in the eye and metabolic variables (blood glucose and C-peptide measurements). We evaluated longitudinally the cytokine profile in the local microenvironment of the intraocular islet allografts, represented in aqueous humour, under conditions of immune rejection vs tolerance. We also evaluated the recall response in the periphery of the baboon recipient using delayed-type hypersensitivity (DTH) assay, and in mice after repeat transplant in the kidney following initial transplant with allogeneic islets in the eye or kidney. RESULTS: Results in mice showed >300 days immunosuppression-free survival of allogeneic islets transplanted in the eye or kidney. Notably, >70% of tolerant mice, initially transplanted in the eye, exhibited >400 days of graft survival after re-transplant in the kidney without immunosuppression compared with ~30% in mice that were initially transplanted in the kidney. Cytokine and DTH data provided evidence of T helper 2-driven local and peripheral immune regulatory mechanisms in support of operational immune tolerance towards the islet allografts in both models. CONCLUSIONS/INTERPRETATION: We are currently evaluating the safety and efficacy of intraocular islet transplantation in a phase 1 clinical trial. In this study, we demonstrate immunosuppression-free long-term survival of intraocular islet allografts in mice and in a baboon using transient peri-transplant immune intervention. These results highlight the potential for inducing islet transplant immune tolerance through the intraocular route. Therefore, the current findings are conceptually significant and may impact markedly on clinical islet transplantation in the treatment of diabetes.


Diabetes Mellitus, Type 1/immunology , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Transplantation Tolerance , Animals , Cytokines/metabolism , Female , Graft Rejection/immunology , Graft Survival/immunology , Hypoglycemia/immunology , Hypoxia , Immunosuppression Therapy , Immunosuppressive Agents , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Papio/immunology , Transplantation, Homologous , Treatment Outcome
7.
Cell Transplant ; 28(3): 262-268, 2019 03.
Article En | MEDLINE | ID: mdl-30675800

The need for chronic immune suppression (IS) is one of the hurdles precluding widespread use of islet cell transplantation to restore glycemic control in patients with type 1 diabetes. We report the case of a healthy nonhuman primate (NHP) treated on and off for over 2.5 years with steroid-free IS, consisting of daclizumab induction and maintenance therapy with rapamycin and low dose tacrolimus. Treatment for 1 year resulted in a striking destabilization of glycemic control, with concomitant decreases in fasting c-peptide and insulin levels. Although these changes gradually reversed during a wash out period of 7 months, retreatment with the same therapy led to accelerated deterioration in glycemic control. Intravenous glucose tolerance and percentage of glycosylated hemoglobin testing further supported a dramatic effect on metabolic control. IS also led to decreases in weight during treatment. Histological evaluation of the pancreas revealed islet hyperplasia, with varying sizes and endocrine cell ratios that differed from normal islet composition, and parenchymal infiltration with adipose tissue. These deleterious effects of IS on glucose control and endocrine components in the native pancreas of a healthy NHP suggest that IS agents commonly utilized for islet transplantation may contribute to failure in islet allograft function in long-term transplant patients.


Blood Glucose , Daclizumab/pharmacology , Graft Survival , Immunosuppression Therapy , Islets of Langerhans Transplantation , Islets of Langerhans , Sirolimus/pharmacology , Tacrolimus/pharmacology , Animals , Blood Glucose/immunology , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/surgery , Glucose Tolerance Test , Graft Survival/drug effects , Graft Survival/immunology , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Macaca fascicularis , Male , Transplantation, Homologous
8.
Xenotransplantation ; 25(6): e12450, 2018 11.
Article En | MEDLINE | ID: mdl-30117193

BACKGROUND: Xenogeneic donors would provide an unlimited source of islets for the treatment of type 1 diabetes (T1D). The goal of this study was to assess the function of microencapsulated adult porcine islets (APIs) transplanted ip in streptozotocin (STZ)-diabetic non-human primates (NHPs) given targeted immunosuppression. METHODS: APIs were encapsulated in: (a) single barium-gelled alginate capsules or (b) double alginate capsules with an inner, islet-containing compartment and a durable, biocompatible outer alginate layer. Immunosuppressed, streptozotocin-diabetic NHPs were transplanted ip with encapsulated APIs, and graft function was monitored by measuring blood glucose, %HbA1c, and porcine C-peptide. At graft failure, explanted capsules were assessed for biocompatibility and durability plus islet viability and functionality. Host immune responses were evaluated by phenotyping peritoneal cell populations, quantitation of peritoneal cytokines and chemokines, and measurement of anti-porcine IgG and IgM plus anti-Gal IgG. RESULTS: NHP recipients had reduced hyperglycemia, decreased exogenous insulin requirements, and lower percent hemoglobin A1c (%HbA1c) levels. Porcine C-peptide was detected in plasma of all recipients, but these levels diminished with time. However, relatively high levels of porcine C-peptide were detected locally in the peritoneal graft site of some recipients at sacrifice. IV glucose tolerance tests demonstrated metabolic function, but the grafts eventually failed in all diabetic NHPs regardless of the type of encapsulation or the host immunosuppression regimen. Explanted microcapsules were intact, "clean," and free-floating without evidence of fibrosis at graft failure, and some reversed diabetes when re-implanted ip in diabetic immunoincompetent mice. Histology of explanted capsules showed scant evidence of a host cellular response, and viable islets could be found. Flow cytometric analyses of peritoneal cells and peripheral blood showed similarly minimal evidence of a host immune response. Preformed anti-porcine IgG and IgM antibodies were present in recipient plasma, but these levels did not rise post-transplant. Peritoneal graft site cytokine or chemokine levels were equivalent to normal controls, with the exception of minimal elevation observed for IL-6 or IL-1ß, GRO-α, I-309, IP-10, and MCP-1. However, we found central necrosis in many of the encapsulated islets after graft failure, and explanted islets expressed endogenous markers of hypoxia (HIF-1α, osteopontin, and GLUT-1), suggesting a role for non-immunologic factors, likely hypoxia, in graft failure. CONCLUSIONS: With donor xenoislet microencapsulation and host immunosuppression, APIs corrected hyperglycemia after ip transplantation in STZ-diabetic NHPs in the short term. The islet xenografts lost efficacy gradually, but at graft failure, some viable islets remained, substantial porcine C-peptide was detected in the peritoneal graft site, and there was very little evidence of a host immune response. We postulate that chronic effects of non-immunologic factors, such as in vivo hypoxic and hyperglycemic conditions, damaged the encapsulated islet xenografts. To achieve long-term function, new approaches must be developed to prevent this damage, for example, by increasing the oxygen supply to microencapsulated islets in the ip space.


Diabetes Mellitus, Experimental/chemically induced , Drug Compounding , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Transplantation, Heterologous , Animals , Drug Compounding/methods , Graft Rejection/immunology , Graft Survival/immunology , Heterografts/immunology , Immunosuppression Therapy/methods , Islets of Langerhans Transplantation/immunology , Primates , Streptozocin/pharmacology , Swine
9.
Cell Metab ; 27(3): 549-558.e4, 2018 03 06.
Article En | MEDLINE | ID: mdl-29514065

Every animal species has a signature blood glucose level or glycemic set point. These set points are different, and the normal glycemic levels (normoglycemia) of one species would be life threatening for other species. Mouse normoglycemia can be considered diabetic for humans. The biological determinants of the glycemic set point remain unclear. Here we show that the pancreatic islet imposes its glycemic set point on the organism, making it the bona fide glucostat in the body. Moreover, and in contrast to rodent islets, glucagon input from the alpha cell to the insulin-secreting beta cell is necessary to fine-tune the distinctive human set point. These findings affect transplantation and regenerative approaches to treat diabetes because restoring normoglycemia may require more than replacing only the beta cells. Furthermore, therapeutic strategies using glucagon receptor antagonists as hypoglycemic agents need to be reassessed, as they may reset the overall glucostat in the organism.


Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Animals , Diabetes Mellitus, Experimental , Humans , Hypoglycemic Agents/pharmacology , Islets of Langerhans Transplantation , Macaca fascicularis , Mice , Mice, Inbred C57BL , Mice, Nude , Paracrine Communication , Receptors, Glucagon/antagonists & inhibitors
11.
Adv Exp Med Biol ; 938: 57-66, 2016.
Article En | MEDLINE | ID: mdl-27586422

Nonhuman primates (NHP) constitute a highly relevant pre-clinical animal model to develop strategies for beta cell replacement. The close phylogenetic and immunologic relationship between NHP and humans results in cross-reactivity of various biological agents with NHP cells, as well as a very similar cytoarchitecture between islets from human and NHP that is strikingly different from that observed in rodent islets. The composition and location of endocrine cells in human or NHP islets, randomly distributed and associated with blood vessels, have functional consequences and a predisposition for paracrine interactions. Furthermore, translation of approaches that proved successful in rodent models to the clinic has been limited. Consequently, data collected from NHP studies can form the basis for an IND submission to the FDA. This chapter describes in detail the key aspects for isolation of islets from NHP, from organ procurement up to assessment of islet function, comparing and emphasizing the similarities between isolation procedures for human and NHP islets.


Islets of Langerhans/physiology , Tissue Culture Techniques/methods , Animals , Collagenases/metabolism , Humans , Macaca fascicularis , Tissue and Organ Procurement
12.
Diabetes ; 65(5): 1350-61, 2016 05.
Article En | MEDLINE | ID: mdl-26916086

Transplantation of pancreatic islets is a therapeutic option to preserve or restore ß-cell function. Our study was aimed at developing a clinically applicable protocol for extrahepatic transplantation of pancreatic islets. The potency of islets implanted onto the omentum, using an in situ-generated adherent, resorbable plasma-thrombin biologic scaffold, was evaluated in diabetic rat and nonhuman primate (NHP) models. Intraomental islet engraftment in the biologic scaffold was confirmed by achievement of improved metabolic function and preservation of islet cytoarchitecture, with reconstitution of rich intrainsular vascular networks in both species. Long-term nonfasting normoglycemia and adequate glucose clearance (tolerance tests) were achieved in both intrahepatic and intraomental sites in rats. Intraomental graft recipients displayed lower levels of serum biomarkers of islet distress (e.g., acute serum insulin) and inflammation (e.g., leptin and α2-macroglobulin). Importantly, low-purity (30:70% endocrine:exocrine) syngeneic rat islet preparations displayed function equivalent to that of pure (>95% endocrine) preparations after intraomental biologic scaffold implantation. Moreover, the biologic scaffold sustained allogeneic islet engraftment in immunosuppressed recipients. Collectively, our feasibility/efficacy data, along with the simplicity of the procedure and the safety of the biologic scaffold components, represented sufficient preclinical testing to proceed to a pilot phase I/II clinical trial.


Biocompatible Materials , Diabetes Mellitus, Experimental/surgery , Hyperglycemia/prevention & control , Islets of Langerhans Transplantation/methods , Pancreas, Artificial , Tissue Scaffolds , Animals , Biocompatible Materials/adverse effects , Biocompatible Materials/chemistry , Biomarkers/blood , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Feasibility Studies , Female , Immunosuppression Therapy/adverse effects , Islets of Langerhans/cytology , Islets of Langerhans/ultrastructure , Islets of Langerhans Transplantation/adverse effects , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/pathology , Macaca fascicularis , Male , Microscopy, Electron, Scanning , Omentum , Pancreas, Artificial/adverse effects , Plasma/chemistry , Plasma/metabolism , Rats, Inbred Lew , Rats, Inbred WF , Recombinant Proteins/adverse effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Surface Properties , Thrombin/adverse effects , Thrombin/chemistry , Thrombin/metabolism , Tissue Engineering , Tissue Scaffolds/adverse effects , Tissue Scaffolds/chemistry , Transplantation, Heterologous/adverse effects , Transplantation, Heterotopic/adverse effects , Transplantation, Isogeneic/adverse effects
13.
Cell Transplant ; 22(7): 1123-35, 2013.
Article En | MEDLINE | ID: mdl-23031502

Clinical islet transplantation has demonstrated success in treating type 1 diabetes. A current limitation is the intrahepatic portal vein transplant site, which is prone to mechanical stress and inflammation. Transplantation of pancreatic islets into alternative sites is preferable, but challenging, as it may require a three-dimensional vehicle to confer mechanical protection and to confine islets to a well-defined, retrievable space where islet neovascularization can occur. We have fabricated biostable, macroporous scaffolds from poly(dimethylsiloxane) (PDMS) and investigated islet retention and distribution, metabolic function, and glucose-dependent insulin secretion within these scaffolds. Islets from multiple sources, including rodents, nonhuman primates, and humans, were tested in vitro. We observed high islet retention and distribution within PDMS scaffolds, with retention of small islets (< 100 µm) improved through the postloading addition of fibrin gel. Islets loaded within PDMS scaffolds exhibited viability and function comparable to standard culture conditions when incubated under normal oxygen tensions, but displayed improved viability compared to standard two-dimensional culture controls under low oxygen tensions. In vivo efficacy of scaffolds to support islet grafts was evaluated after transplantation in the omental pouch of chemically induced diabetic syngeneic rats, which promptly achieved normoglycemia. Collectively, these results are promising in that they indicate the potential for transplanting islets into a clinically relevant, extrahepatic site that provides spatial distribution of islets as well as intradevice vascularization.


Dimethylpolysiloxanes/chemistry , Islets of Langerhans Transplantation , Adult , Animals , Cell Survival/drug effects , Child, Preschool , Female , Humans , Hydrogels/chemistry , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Middle Aged , Omentum/pathology , Porosity , Primates , Rats , Rats, Inbred Lew , Stress, Mechanical , Young Adult
14.
Diabetes ; 59(10): 2558-68, 2010 Oct.
Article En | MEDLINE | ID: mdl-20622174

OBJECTIVE: To test the graft-promoting effects of mesenchymal stem cells (MSCs) in a cynomolgus monkey model of islet/bone marrow transplantation. RESEARCH DESIGN AND METHODS: Cynomolgus MSCs were obtained from iliac crest aspirate and characterized through passage 11 for phenotype, gene expression, differentiation potential, and karyotype. Allogeneic donor MSCs were cotransplanted intraportally with islets on postoperative day (POD) 0 and intravenously with donor marrow on PODs 5 and 11. Recipients were followed for stabilization of blood glucose levels, reduction of exogenous insulin requirement (EIR), C-peptide levels, changes in peripheral blood T regulatory cells, and chimerism. Destabilization of glycemia and increases in EIR were used as signs of rejection; additional intravenous MSCs were administered to test the effect on reversal of rejection. RESULTS: MSC phenotype and a normal karyotype were observed through passage 11. IL-6, IL-10, vascular endothelial growth factor, TGF-ß, hepatocyte growth factor, and galectin-1 gene expression levels varied among donors. MSC treatment significantly enhanced islet engraftment and function at 1 month posttransplant (n = 8), as compared with animals that received islets without MSCs (n = 3). Additional infusions of donor or third-party MSCs resulted in reversal of rejection episodes and prolongation of islet function in two animals. Stable islet allograft function was associated with increased numbers of regulatory T-cells in peripheral blood. CONCLUSIONS: MSCs may provide an important approach for enhancement of islet engraftment, thereby decreasing the numbers of islets needed to achieve insulin independence. Furthermore, MSCs may serve as a new, safe, and effective antirejection therapy.


Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/physiology , Mesenchymal Stem Cell Transplantation/methods , Animals , Antigens, CD/analysis , Blood Glucose/metabolism , Cell Differentiation , Culture Media , Diabetes Mellitus, Experimental/blood , Epidermal Growth Factor/genetics , Forkhead Transcription Factors/analysis , Galectin 1/genetics , Hepatocyte Growth Factor/genetics , Histocompatibility Antigens Class II/analysis , Histocompatibility Testing , Interleukins/genetics , Karyotyping , Macaca fascicularis/immunology , Macaca fascicularis/physiology , Major Histocompatibility Complex , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Phenotype , RNA/genetics , RNA/isolation & purification , Transforming Growth Factor beta/genetics , Transplantation, Homologous
15.
Cell Transplant ; 19(12): 1547-61, 2010.
Article En | MEDLINE | ID: mdl-20587138

This retrospective study reviews the results of our experience with the occurrence of CMV DNAemia in islet cell transplanted cynomolgus monkeys subjected to different immunosuppressive protocols, including induction treatment with thymoglobulin (TMG), with a combination of thymoglobulin and fludarabine (FLUD), with cyclophosphamide, or with daclizumab. CMV DNA in the peripheral blood (CMV DNAemia) of 47 monkeys was quantified by real-time PCR on a weekly to biweekly basis. As compared to other immunosuppressive regimens, and in association with greater decreases in WBC, lymphocyte, CD3+CD4+, and CD3+CD8+ lymphocyte counts, frequent CMV DNAemia occurred earlier (within the first month posttransplant), and was of greater severity and duration in recipients of TMG ± FLUD. Treatment of recipients with alternative induction agents that resulted in less dramatic reductions in WBC and lymphocyte counts, however, resulted in occurrence of CMV DNAemia after postoperative day 60. The frequency, average intensity, duration, and area under the curve (AUC) for CMV DNAemia in animals receiving TMG ± FLUD were 75-100%, 4.02 ± 1.75 copies/ng DNA, 23.0 ± 5.3 days, and 367.0 ± 121.1 days × copies/ng DNA, respectively; corresponding values in animals receiving other treatments (0-44%, 0.19 ± 0.10 copies/ng DNA, 0.5 ± 0.3 days, and 75.4 ± 40.2 days × copies/ng DNA, respectively) were significantly different. The value of WBC, T and B cells at the nadir of cell depletion greatly affects the occurrence of CMV DNAemia. No animals developed CMV DNAemia within the next 3 weeks when the lowest value of WBC, lymphocyte, CD3+, CD3+CD4+, CD3+CD8+, or CD20+ cells was above 4500, 1800, 300, 200, 150, or 300 cells/µl, respectively. Oral valganciclovir prophylaxis did not completely prevent the appearance of CMV DNAemia.


Cytomegalovirus/drug effects , Cytomegalovirus/genetics , DNA, Viral/blood , Immunosuppressive Agents/pharmacology , Islets of Langerhans Transplantation/immunology , Transplantation, Homologous/immunology , Analgesics, Non-Narcotic/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antilymphocyte Serum/pharmacology , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Cytomegalovirus/immunology , Daclizumab , Ganciclovir/analogs & derivatives , Ganciclovir/pharmacology , Humans , Immunoglobulin G/pharmacology , Leukocytes/drug effects , Leukocytes/immunology , Macaca fascicularis , Organometallic Compounds/pharmacology , Organophosphorus Compounds/pharmacology , Retrospective Studies , Sirolimus/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tacrolimus/pharmacology , Valganciclovir , Vidarabine/analogs & derivatives , Vidarabine/pharmacology
16.
Proc Natl Acad Sci U S A ; 107(14): 6465-70, 2010 Apr 06.
Article En | MEDLINE | ID: mdl-20308565

Extracellular ATP has been proposed as a paracrine signal in rodent islets, but it is unclear what role ATP plays in human islets. We now show the presence of an ATP signaling pathway that enhances the human beta cell's sensitivity and responsiveness to glucose fluctuations. By using in situ hybridization, RT-PCR, immunohistochemistry, and Western blotting as well as recordings of cytoplasmic-free Ca(2+) concentration, [Ca(2+)](i), and hormone release in vitro, we show that human beta cells express ionotropic ATP receptors of the P2X(3) type and that activation of these receptors by ATP coreleased with insulin amplifies glucose-induced insulin secretion. Released ATP activates P2X(3) receptors in the beta-cell plasma membrane, resulting in increased [Ca(2+)](i) and enhanced insulin secretion. Therefore, in human islets, released ATP forms a positive autocrine feedback loop that sensitizes the beta cell's secretory machinery. This may explain how the human pancreatic beta cell can respond so effectively to relatively modest changes in glucose concentration under physiological conditions in vivo.


Adenosine Triphosphate/metabolism , Autocrine Communication , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, Purinergic P2/metabolism , Calcium/metabolism , Humans , Insulin Secretion , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X3 , Signal Transduction
17.
Cell Transplant ; 18(10): 1213-22, 2009.
Article En | MEDLINE | ID: mdl-19650973

Islet transplantation is a promising cellular therapy for the treatment of type 1 diabetes (T1D). The immunogenicity of isolated islets has been of interest to the transplant community for many years, as upon transplantation, islets are damaged or destroyed through specific and nonspecific inflammatory and immune events. Antigen presenting cells (APC) are crucial intermediates in the generation of both innate and specific immune responses, and it has long been understood that some APC are resident in islets in situ, as well as after isolation. Our aim was to identify and characterize intraislet resident populations of APC and other immune cells in islets from nonhuman primates (Macaca fascicularis) in situ (pancreas biopsies obtained prerecovery) and after isolation using immunohistochemistry, confocal microscopy, and flow cytometry. The numbers of cells obtained in situ are similar to those in islets postisolation. Each isolated islet equivalent contains an average of 21.8 immune cells, 14.7 (67%) of which are APC. Many of these APC are dentritic cells and, surprisingly, 50% are B lymphocytes. The number of islet-resident immune cells increases with islet size, with greater numbers in large versus small islets (p < 0.001). The APC were localized around the exterior or spread evenly throughout the islets, with no definitive orientation identified. This knowledge will be useful to develop tailored modulation strategies to decrease immunogenicity, enhance engraftment, and ultimately prevent islet rejection.


Islets of Langerhans/immunology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Flow Cytometry , Islets of Langerhans/anatomy & histology , Islets of Langerhans/cytology , Islets of Langerhans Transplantation , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Macaca fascicularis , Macrophages/cytology , Macrophages/immunology , Organ Size , T-Lymphocytes/cytology , T-Lymphocytes/immunology
18.
Diabetes ; 57(10): 2745-54, 2008 Oct.
Article En | MEDLINE | ID: mdl-18820214

OBJECTIVE: Adenoviral delivery of hepatocyte growth factor (HGF) to rodent islets improves islet graft survival and function, markedly reducing the number of islets required to achieve glucose control. Here, we asked whether these prior observations in rodent models extend to nonhuman primate (NHP) islets. RESEARCH DESIGN AND METHODS: NHP islets were transduced with murine (Ad.mHGF) or human (Ad.hHGF) adenoviral HGF (Ad.HGF) at low multiplicity of infection and studied in vitro. To study the function of Ad.HGF-transduced NHP islets in vivo, a renal subcapsular marginal mass islet transplant model was developed in streptozotocin-induced diabetic NOD-SCID mice. RESULTS: Baseline glucose values were 454.7 +/- 11.3 mg/dl (n = 7). Transplant of 500 NHP islet equivalents (IE) had only a marginal effect on blood glucose (369.1 +/- 9.7 mg/dl, n = 5). In striking contrast, 500 NHP IE transduced with Ad.mHGF promptly and continuously corrected blood glucose (142.0 +/- 6.2 mg/dl, n = 7) for the 6-week duration of the experiment. Unilateral nephrectomy resulted in an immediate return of glucose to baseline diabetic levels. Interestingly, adenoviral DNA, as well as mouse HGF (mHGF) mRNA derived from the adenovirus, were present for 42 days posttransplantation. Surprisingly, transplant of 500 IE with Ad.hHGF, as compared with Ad.mHGF, resulted in only marginal correction of blood glucose, suggesting that human HGF is less efficient than mHGF in this system. CONCLUSIONS: These studies demonstrate that mHGF markedly improves islet transplant outcomes in the highest preclinical species examined to date. HGF has promise as an agent that can improve islet mass and function in transplant models and likely in other models of types 1 and 2 diabetes.


Graft Survival/physiology , Hepatocyte Growth Factor/physiology , Islets of Langerhans Transplantation/methods , Islets of Langerhans/metabolism , Adenoviridae/genetics , Animals , Cell Proliferation , Genetic Vectors/genetics , Hepatocyte Growth Factor/genetics , Immunohistochemistry , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Macaca fascicularis , Mice , Mice, Inbred NOD , Mice, SCID , Mitogen-Activated Protein Kinases/metabolism
19.
Cell Metab ; 7(6): 545-54, 2008 Jun.
Article En | MEDLINE | ID: mdl-18522835

An important feature of glucose homeostasis is the effective release of glucagon from the pancreatic alpha cell. The molecular mechanisms regulating glucagon secretion are still poorly understood. We now demonstrate that human alpha cells express ionotropic glutamate receptors (iGluRs) that are essential for glucagon release. A lowering in glucose concentration results in the release of glutamate from the alpha cell. Glutamate then acts on iGluRs of the AMPA/kainate type, resulting in membrane depolarization, opening of voltage-gated Ca(2+) channels, increase in cytoplasmic free Ca(2+) concentration, and enhanced glucagon release. In vivo blockade of iGluRs reduces glucagon secretion and exacerbates insulin-induced hypoglycemia in mice. Hence, the glutamate autocrine feedback loop endows the alpha cell with the ability to effectively potentiate its own secretory activity. This is a prerequisite to guarantee adequate glucagon release despite relatively modest changes in blood glucose concentration under physiological conditions.


Autocrine Communication , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Glutamic Acid/physiology , Animals , Blood Glucose , Cells, Cultured , Glucagon-Secreting Cells/physiology , Haplorhini , Homeostasis , Humans , Mice , Receptors, AMPA/metabolism , Receptors, Kainic Acid , Receptors, Metabotropic Glutamate
20.
Cell Transplant ; 16(10): 1039-48, 2008.
Article En | MEDLINE | ID: mdl-18351020

An important challenge in pancreatic islet transplantation in association with type 1 diabetes is to define automatic high-throughput assays for evaluation of human islet function. The physiological techniques presently used are amenable to small-scale experimental samples and produce descriptive results. The postgenomic era provides an opportunity to analyze biological processes on a larger scale, but the transition to high-throughput technologies is still a challenge. As a first step to implement high-throughput assays for the study of human islet function, we have developed two methodologies: multiple automated perifusion to determine islet hormone secretion and high-throughput kinetic imaging to examine islet cellular responses. Both technologies use fully automated devices that allow performing simultaneous experiments on multiple islet preparations. Our results illustrate that these technologies can be applied to study the functional status and explore the pharmacological profiles of islet cells. These methodologies will enable functional characterization of human islet preparations before transplantation and thereby provide the basis for the establishment of predictive tests for beta-cell potency.


Islets of Langerhans/physiology , Animals , Autoanalysis , Calcium/analysis , Diabetes Mellitus, Type 2/physiopathology , Glucagon/metabolism , Glucose/analysis , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation , Living Donors , Macaca fascicularis , Pancreatic Hormones/metabolism , Perfusion , Species Specificity
...