Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
2.
Article in English | MEDLINE | ID: mdl-31001570

ABSTRACT

Transplantation of insulin-producing cells into immunodeficient mice represents an important tool for the assessment of viability and function of cellular products intended to restore beta cell function in the clinical and research applications. The in vivo bioassay allows for the assessment of the ability of the cellular product to restore and maintain euglycemia in vivo after transplantation into chemically-induced diabetic recipients. Herein we describe the protocol utilized for the in vivo potency assessment of human islet cell products. Modifications of the protocol enable the use of the bioassay for the potency assessment of human or animal insulin-producing cells from adult, neonatal or fetal sources or generated from stem cells.

3.
Am J Transplant ; 13(10): 2550-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24010969

ABSTRACT

Transplantation of mature islets into portal vein has been most effective thus far, although attrition of transplanted islets constitutes a major limitation, and alternative approaches are required. We analyzed the mechanisms by which islets engrafted, vascularized and functioned over the long term in the small intestinal submucosa. To determine engraftment, survival and function, 350 syngenic islets were transplanted into either intestinal segments or portal vein of diabetic rats. Islet reorganization, vascularization and function were analyzed by histological analysis, RT-PCR analysis as well as glycemic control over up to 1 year. Transplantation of syngeneic islets in marginal numbers successfully restored normoglycemia in diabetic rats. Transplantation of semi-pure islet preparation did not impair their engraftment, vascularization and function. Islets were morphologically intact and expressed insulin as well as glucagon over the year. Expression of angiogenic genes permitted revascularization of transplanted islets. We identified the expression of transcription factors required for maintenance of beta cells. These studies demonstrated that marginal mass of transplanted islets was sufficient to restore euglycemia in streptozotocin-treated rats. These superior results were obtained despite use of an impure preparation of islets in animals with small intestinal segment. Our findings will help advance new horizons for cell therapy in patients with diabetes.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Graft Survival/physiology , Intestine, Small/surgery , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Portal Vein/surgery , Animals , Diabetes Mellitus, Experimental/metabolism , Intestine, Small/blood supply , Intestine, Small/metabolism , Islets of Langerhans/blood supply , Islets of Langerhans/surgery , Portal Vein/metabolism , RNA, Messenger/genetics , Rats , Rats, Inbred Lew , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Isogeneic
4.
Cell Transplant ; 21(1): 39-47, 2012.
Article in English | MEDLINE | ID: mdl-21929867

ABSTRACT

The selection of enzyme blend is critical for the success of human islet isolations. Liberase HI collagenase (Roche) was introduced in the 1990s and had been widely used for clinical islet transplantation. More recently, a blend collagenase NB1 has been rendered available. The aim of this study was to evaluate the isolation outcomes and islet quality comparing human islet cells processed using NB1 and Liberase HI. A total of 90 isolations processed using NB1 (n = 40) or Liberase HI (n = 50) was retrospectively analyzed. Islet yield, function in vitro and in vivo, cellular (including ß-cell-specific) viability and content, as well as isolation-related factors were compared. No significant differences in donor-related factors were found between the groups. There were also no significant differences in islet yields (NB1 vs. Liberase: 263,389 ± 21,550 vs. 324,256 ± 27,192 IEQ; p = n.s., respectively). The pancreata processed with NB1 showed a significantly longer digestion time (18.6 ± 0.7 vs. 14.5 ± 0.5 min, p < 0.01), lower ß-cell viability (54.3 ± 3.4% vs. 72.0 ± 2.1%, p < 0.01), ß-cell mass (93,671 ± 11,150 vs. 148,961 ± 12,812 IEQ, p < 0.01), and viable ß-cell mass (47,317 ± 6,486 vs. 106,631 ± 10,228 VßIEQ, p < 0.01) than Liberase HI. In addition, islets obtained with Liberase showed significantly better graft function in in vivo assessment of islet potency. The utilization of collagenase NB1 in human islet isolation was associated with significantly lower ß-cell viability, mass, and islet potency in vivo in our series when compared to Liberase HI, even though there was no significant difference in islet yields between the groups. Evaluation of viable ß-cell mass contained in human islet preparations will be useful for selecting enzyme blends.


Subject(s)
Collagenases/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/transplantation , Islets of Langerhans Transplantation/methods , Thermolysin/metabolism , Adult , Cell Separation , Cell Survival , Female , Humans , Insulin-Secreting Cells/physiology , Male , Middle Aged
5.
Cell Transplant ; 21(4): 633-48, 2012.
Article in English | MEDLINE | ID: mdl-21669040

ABSTRACT

Exenatide is an analog of the incretin hormone glucagon-like peptide (GLP-1) that is used for the treatment of T2D for their metabolic effects. In addition to its insulinotropic effects, exenatide increases functional islet mass and improves their survival. Improved outcomes have been reported in recent clinical islet transplantation trials for the treatment of type 1 diabetes. The purpose of this study was to investigate whether exenatide has anti-inflammatory properties in human islets. Exenatide treatment improved islet function, significantly reduced content of inflammation-related molecules (tissue factor, IFN-γ, IL-17, IL-1ß, and IL-2) and caspase 3 activation, whereas increased phosphorylation of ERK1/2, STAT3, and Akt in vitro. Immunostaining showed expression of GLP-1R in ß-cells but not in α-cells. IL-1ß colocalized with GLP-1R in ß-cells. Induction of serine proteinase inhibitor 9 (PI-9) was detected after exposure of human islets to exenatide in vitro and after transplantation into immunodeficient mice. GLP-1 induced PI-9 expression in vitro but to a lower extent than exenatide. This effect was partially blocked by the antagonist exendin-9 in vitro. As assessed by immunostaining PI-9 is mostly expressed in ß-cells but not in α-cells. In conclusion, we describe anti-inflammatory and cytoprotective properties of exenatide in human islets. Exenatide-mediated PI-9 expression, the only known granzyme B inhibitor, unveils potential immunoregulatory properties.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Peptides/pharmacology , Venoms/pharmacology , Animals , Blotting, Western , Caspase 3/metabolism , Exenatide , Glucagon-Like Peptide 1/pharmacology , Humans , In Vitro Techniques , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-2/metabolism , Mice , Mice, Nude
6.
Cell Transplant ; 19(12): 1537-46, 2010.
Article in English | MEDLINE | ID: mdl-20719078

ABSTRACT

Islet isolation and purification using a continuous density gradient may reduce the volume of tissue necessary for implantation into patients, therefore minimizing the risks associated with intraportal infusion in islet transplantation. On the other hand, the purification procedure might result in a decreased number of islets recovered due to various stresses such as exposure to cytokine/chemokine. While a Ficoll-based density gradient has been widely used in purification for clinical trials, purification with iodixanol (OptiPrep) has been recently reported in islet transplant series with successful clinical outcomes. The aim of the current study was to compare the effects of the purification method using OptiPrep-based and Ficoll-based density gradients. Human islet isolations were performed using a modified automated method. After the digestion phase, pre-purification digests were divided into two groups and purified using a semiautomated cell processor with either a continuous Ficoll- or OptiPrep-based density gradient. The quantity, purity, viability, and cellular composition of islet preparations from each group were assessed. Cytokine/chemokine and tissue factor production from islet preparations after 48-h culture were also measured. Although islet purity, post-purification IEQ, islet recovery rate, FDA/PI, and fractional ß-cell viability were comparable, ß-cell mass after 48-h culture significantly improved in the OptiPrep group when compared to the Ficoll group. The production of cytokine/chemokine including IL-1ß, TNF-α, IFN-γ, IL-6, IL-8, MIP-1ß, MCP-1, and RANTES but not tissue factor from the OptiPrep group was significantly lower during 48-h culture after isolation. Each preparation contained the similar number of ductal cells and macrophages. Endotoxin level in both gradient medium was also comparable. The purification method using OptiPrep gradient media significantly reduced cytokine/chemokine production but not tissue factor from human islet preparations and improved ß-cell survival during pretransplant culture. Our results suggest that the purification method using OptiPrep gradient media may be of assistance in increasing successful islet transplantation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Separation/methods , Centrifugation, Density Gradient/methods , Insulin-Secreting Cells/drug effects , Islets of Langerhans/cytology , Triiodobenzoic Acids/pharmacology , Adolescent , Adult , Animals , Cell Separation/instrumentation , Cell Survival , Cells, Cultured , Centrifugation, Density Gradient/instrumentation , Chemokines/metabolism , Contrast Media , Cytokines/metabolism , Female , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/physiology , Islets of Langerhans Transplantation/methods , Male , Mice , Mice, Nude , Middle Aged , Thromboplastin/metabolism
7.
Pharmazie ; 65(6): 421-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20614690

ABSTRACT

Emerging biotechnologies, such as the use of biohybrid devices for cellular therapies, are showing increasing therapeutic promise for the treatment of various diseases, including type 1 diabetes mellitus. The functionality of such devices could be greatly enhanced if successful localized immunosuppression regimens could be established, since they would eliminate the many otherwise unavoidable side effects of currently used systemic immunosuppressive therapies. The existence of local immune privilege at some specialized tissues, such as the eye, CNS, or pregnant uterus, supports the feasibility of localized immunomodulation, and such an approach is particularly well-suited for cell transplant therapies where all transplanted tissue is localized within a device. Following the success of syngeneic transplantation in a subcutaneous prevascularized device as a bioartificial pancreas in a rodent model, we now report the first results of exploratory in vivo islet allograft studies in rats using locally delivered glucocorticoids (dexamethasone phosphate and the soft steroid loteprednol etabonate). Following in vitro assessments, in silico drug distribution models were used to establish tentative therapeutic dose ranges. Sustained local delivery was achieved via implantable osmotic mini-pumps through a central sprinkler, as well as with a sustained-delivery formulation for loteprednol etabonate using poly(D,L-lactic) acid (PLA) microspheres. Doses delivered locally were approximately hundred-fold smaller than those typically used in systemic treatments. While several solubility, stability, and implantation problems still remain to be addressed, both compounds showed promise in their ability to prolong graft survival after tapering of systemic immunosuppression, compared to control groups.


Subject(s)
Cell Transplantation/instrumentation , Glucocorticoids/pharmacology , Immunosuppressive Agents , Islets of Langerhans Transplantation/immunology , Algorithms , Androstadienes/administration & dosage , Animals , Biotechnology , Computer Simulation , Delayed-Action Preparations , Drug Delivery Systems , Drug Implants , Feasibility Studies , Female , Glucocorticoids/administration & dosage , Glucocorticoids/adverse effects , Humans , Immunosuppressive Agents/adverse effects , Kaplan-Meier Estimate , Lactic Acid , Loteprednol Etabonate , Microspheres , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Pregnancy , Rats , Tissue Distribution
8.
Transplant Proc ; 41(5): 1808-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19545733

ABSTRACT

We used a rat model of pancreas cold preservation to assess its effects on islets. Glands were surgically retrieved and stored in University of Wisconsin (UW) solution for 3 hours (Short) or 18 hours (Long) cold ischemia time (CIT). Islet yield was significantly lower in the Long-CIT than the Short-CIT group, as well as islet recovery after overnight culture (P < .01). Islet cell viability after isolation was significantly reduced in the Long-CIT group (P < .05). Reversal of diabetes following transplantation of suboptimal islet grafts occurred earlier in the Short-CIT group than the Long-CIT. All animals in the Short-CIT group and 80% in the Long-CIT group achieved euglycemia. Freshly isolated islets showed a significant increase of JNK and p38 (P < .05) phosphorylation in Long-CIT compared with Short-CIT. Histopathological assessment of the pancreas showed a significantly higher injury score. Proteomic analysis of pancreatic tissue led to identification of 5 proteins consistently differentially expressed between Short-CIT and Long-CIT. Better understanding of the molecular pathways involved in this phenomenon will be of assistance in defining targeted interventions to improve organ use in the clinical arena.


Subject(s)
Islets of Langerhans/cytology , Pancreas/cytology , Adenosine , Allopurinol , Animals , Cell Survival , Glutathione , Insulin , Ischemia , Islets of Langerhans/pathology , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Organ Preservation Solutions , Pancreas/blood supply , Pancreas/pathology , Phosphotransferases/metabolism , Raffinose , Rats , Rats, Inbred Lew , Tissue and Organ Harvesting/methods
9.
Transplant Proc ; 41(1): 352-3, 2009.
Article in English | MEDLINE | ID: mdl-19249555

ABSTRACT

The aim of this study was to explore the effect of sirolimus (Sir) and tacrolimus (Tac) on islets implanted into a subcutaneous (SC), prevascularized device in syngeneic rats. Animals received a 40-day treatment with Tac and Sir (alone or in combination) starting either on day 0 or 40 days after islet transplantation. Controls received no treatment. A 40-day washout period was performed after immunosuppression (IS). Glycemia and intravenous glucose tolerance tests (IVGTT) were assessed at follow-up. In the control group, 75% of recipients achieved stable normoglycemia after islet transplantation, while none reversed diabetes with any IS regimen started on day 0. Graft dysfunction was irreversible after IS withdrawal. Glucose clearance (IVGTT) was significantly impaired among Tac-treated compared with control groups (P < .05 with IS; P < .01 after washout). Among animals with established grafts, islet dysfunction which occurred under IS treatment persisted after washout in animals treated with Tac and Sir plus Tac. When compared with controls, glucose clearance was significantly impaired in the Tac and Tac plus Sir groups before and after IS (P < .01, Tac; P < 0.01, Tac plus Sir). Sir and Tac showed profound deleterious effects on islet cell engraftment and function, which may hinder the success of implantation into biohybrid devices. Nondiabetogenic IS protocols must be developed for clinical application of islet transplantation into biohybrid devices.


Subject(s)
Immunosuppression Therapy/methods , Immunosuppressive Agents/therapeutic use , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/methods , Animals , Blood Glucose/metabolism , Rats , Rats, Inbred Lew , Sirolimus/therapeutic use , Tacrolimus/therapeutic use , Transplantation, Isogeneic/immunology
10.
Transplant Proc ; 41(1): 354-5, 2009.
Article in English | MEDLINE | ID: mdl-19249556

ABSTRACT

Increasing evidence supports the beneficial effects of ischemic preconditioning (IPC) of organs on subsequent ischemia. The aim of this study was to assess the effects of IPC of the pancreas on islet cell recovery after cold preservation using a rat model. The pancreas was deprived of perfusion (celiac artery and superior mesenteric artery occlusion) for 10 minutes followed by 10 minutes of reperfusion. Islet isolation was performed after 18 hours of cold ischemia. Glands undergoing IPC yielded significantly greater numbers of islets than controls. Following overnight culture, a significantly greater proportion of islets was recovered from IPC-treated pancreata. Microarray genomic analysis of pancreatic tissue revealed a significant differential expression of approximately 600 unique mRNA strands within IPC pancreata compared to only <100 unique mRNA strands within non-IPC pancreata (>2-fold change; P < .05). Proteomic analysis revealed significant differential expression of at least 5 proteins >1.5-fold change; P < .05) within the IPC vs control group. Our data indicated that IPC of the pancreas prior to cold preservation was associated with improved islet cell recovery after cold ischemia. IPC of the pancreas may represent a viable therapeutic intervention to increase islet transplantation success from a single donor and to maximize organ utilization.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Ischemic Preconditioning/methods , Islets of Langerhans/physiology , Organ Preservation/methods , Pancreas/cytology , Animals , Gene Expression Regulation , Islets of Langerhans/cytology , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Rats , Transplantation, Heterologous/physiology
11.
Diabetologia ; 51(12): 2271-80, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18853132

ABSTRACT

AIMS/HYPOTHESIS: Inhibition of c-jun N-terminal kinase (JNK) favours pancreatic islet function and survival. Since two JNK isoforms are present in the pancreas (JNK1 and JNK2), we addressed their specific roles in experimental islet transplantation. METHODS: C57BL/6J (wild-type [WT]), Jnk1 (also known as Mapk8)(-/-) and Jnk2 (also known as Mapk9)(-/-) mice were used as donor/recipients in a syngeneic islet transplantation model. Islet cell composition, function, viability, production of cytokines and of vascular endothelial growth factor (VEGF) were also studied in vitro. RESULTS: Jnk1 ( -/- ) islets secreted more insulin in response to glucose and were more resistant to cytokine-induced cell death compared with WT and Jnk2 (-/-) islets (p < 0.01). Cytokines reduced VEGF production in WT and Jnk2 (-/-) but not Jnk1 ( -/- ) islets; VEGF blockade restored Jnk1 ( -/- ) islet susceptibility to cytokine-induced cell death. Transplantation of Jnk1 ( -/- ) or WT islets into WT recipients made diabetic had similar outcomes. However, Jnk1 ( -/- ) recipients of WT islets had shorter time to diabetes reversal (17 vs 55 days in WT, p = 0.033), while none of the Jnk2 (-/-) recipients had diabetes reversal (0% vs 71% in WT, p = 0.0003). Co-culture of WT islets with macrophages from each strain revealed a discordant cytokine production. CONCLUSIONS/INTERPRETATION: We have shown a deleterious effect of JNK2 deficiency on islet graft outcome, most likely related to JNK1 activation, suggesting that specific JNK1 blockade may be superior to general JNK inhibition, particularly when administered to transplant recipients.


Subject(s)
Islets of Langerhans/cytology , Islets of Langerhans/enzymology , Mitogen-Activated Protein Kinase 8/metabolism , Animals , Cell Survival/drug effects , Coculture Techniques , Cytokines/pharmacology , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/surgery , Graft Survival , Islets of Langerhans/surgery , Islets of Langerhans Transplantation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Phosphorylation
12.
Clin Exp Immunol ; 154(1): 15-21, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18759852

ABSTRACT

Previous studies have shown that human alpha-1 antitrypsin (hAAT) gene delivery prevents type 1 diabetes (T1D) in non-obese diabetic (NOD) mice. Furthermore, hAAT protein administration prolongs acceptance of islet allografts. Therefore, we evaluated the use of purified hAAT protein therapy to prevent T1D in NOD mice. Female NOD, non-obese resistant (NOR), Balb/c and C57BL/6 mice were injected intraperitoneally with vehicle alone or vehicle containing hAAT, human albumin or mouse albumin (or mg/injection/mouse; 2x/week). Preparations of clinical-grade hAAT included API(R), Aralast, Prolastin and Zemaira. Surprisingly, hAAT administration was associated with a high rate of fatal anaphylaxis. In studies seeking T1D prevention at 4 weeks of age, 100% mice died after six injections of hAAT. When administrated at 8-10 weeks of age, most (80-100%) NOD mice died following the fourth injection of hAAT, while 0% of Balb/c and C57BL/6 mice and 10% of NOR mice died. Interestingly, repeated injections of human albumin, but not mouse albumin, also induced sudden death in NOD mice. Antibodies to hAAT were induced 2-3 weeks after hAAT administration and death was prevented by treatment with anti-platelet-activating factor along with anti-histamine. In studies of disease reversal in NOD mice, using the four pharmaceutical grade formulations of hAAT, anaphylactic deaths were observed with all hAAT preparations. The propensity for fatal anaphylaxis following antigenic administration appears to be NOD- but not hAAT-specific. The susceptibility of NOD mice to hypersensitivity provides a significant limitation for testing of hAAT. Development of strategies to avoid this unwanted response is required to use this promising therapeutic agent for T1D.


Subject(s)
Anaphylaxis/immunology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , alpha 1-Antitrypsin/adverse effects , Albumins/adverse effects , Animals , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Drug Administration Schedule , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Species Specificity , alpha 1-Antitrypsin/therapeutic use
13.
Pharmazie ; 63(3): 226-32, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18444512

ABSTRACT

Transplantation of pancreatic islets into subcutaneous, neovascularized devices is one of the possibilities explored as part of our search for a cure of diabetes. We have recently reported that syngeneic transplantation in a subcutaneous prevascularized device can restore euglycemia and sustain long-term function in rats and that explanted grafts showed preserved islets and intense vascular networks. Because all of the transplanted tissue is localized within the device, if such a bioartificial pancreas approach is used, localized immunosuppression might provide sufficient protection against rejection to achieve long-term function, while also avoiding the serious systemic side effects and the susceptibility for opportunistic infections that are commonly associated with systemic immunosuppressive therapies as only much smaller and localized doses are needed. Soft steroids are obvious candidates because soft drugs are specifically designed to produce targeted local activity, but no systemic side effects due to prompt metabolic (preferably extrahepatic, e.g., hydrolytic) inactivation. However, local concentrations that are effective for immunosuppression, but non-toxic to insulin-producing beta-cells have to be found, and nontrivial difficulties related to long-term local deliverability have to be addressed. Here, we report preliminary results obtained using in vitro studies with human islets used to establish a tentative therapeutic concentration range together with fully scaled three-dimensional finite element method (FEM)-based Comsol multiphysics computational models that were used to explore various possibilities to achieve and maintain these concentration levels within the device.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Androstadienes/pharmacology , Anti-Inflammatory Agents/pharmacology , Immunosuppressive Agents/pharmacology , Islets of Langerhans Transplantation/immunology , Adrenal Cortex Hormones/pharmacokinetics , Algorithms , Androstadienes/pharmacokinetics , Animals , Anti-Inflammatory Agents/pharmacokinetics , Chromatography, High Pressure Liquid , Finite Element Analysis , Humans , Immunosuppressive Agents/pharmacokinetics , Islets of Langerhans/immunology , Loteprednol Etabonate , Models, Statistical , Perfusion , Rats , Tissue Distribution
14.
Transplant Proc ; 40(2): 382-3, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374075

ABSTRACT

INTRODUCTION: Many cytoprotective agents have been reported to improve islet isolation and transplantation outcomes. However, several of these agents improve all cell subsets within an islet preparation; selection of non-beta-cell components (eg, acinar cells) may have a negative effect on beta-cell function and survival. In this study, we examined the effect of prolactin (PRL) supplementation in the culture medium to determine whether it exerted beta-cell-selective cytoprotection on islet viability and function. MATERIALS AND METHODS: Human islets were precultured with or without recombinant human PRL (500 microg/L) for 48 hours. The fractional viability and cellular composition of non-beta-cell and beta-cell-specific components were assessed using FACS and Laser Scanning Cytometry (LSC). Islet potency was assessed in vivo by transplantation into chemically induced diabetic immunodeficient mice. RESULTS: The relative viable beta-cell mass and the relative islet beta-cell content in the PRL group were 28% higher (P = .018) and 19% higher (P = .029) than the control group, respectively. All transplanted mice achieved normoglycemia in both groups, indicating that PRL treatment did not alter islet function. CONCLUSION: PRL treatment improved beta-cell-specific viability and survival of human islets in vitro. The development of novel beta-cell-specific cytoprotective strategies may be of assistance in improving islet transplantation.


Subject(s)
Cell Survival/drug effects , Diabetes Mellitus, Experimental/surgery , Insulin-Secreting Cells/cytology , Prolactin/pharmacology , Animals , Cell Culture Techniques , Disease Models, Animal , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/transplantation , Islets of Langerhans Transplantation , Mice , Mice, Nude
15.
Transplant Proc ; 40(2): 436-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374093

ABSTRACT

During pregnancy a high rate of beta-cell proliferation occurs, making of this a useful model for the study of islet cell expansion in vivo. We used the murine pregnancy model to assess the effect of Rapamycin treatment on islet cell proliferation in vivo. Rapamycin is routinely used for the prevention of graft rejection in transplanted patients, including islet transplant recipients. As expected, pregnancy led to increased beta-cell proliferation, islet yield and skewing in size distribution after isolation and pancreatic insulin content, when compared to non-pregnant females. Rapamycin treatment resulted in reduced beta cell proliferation in pregnant mice, while minimal effects of Rapamycin treatment were observed on islet function both in vivo and in vitro. Rapamycin treatment of islets resulted in reduced phosphorylation of p70s6k, a downstream effector molecule of mTOR and increased ERK1/2 phosphorylation. In conclusion, beta-cell replication is reduced under Rapamycin treatment in vivo, suggesting that this mechanism may be operational and impair beta-cell renewal in transplanted patients.


Subject(s)
Cell Division/drug effects , Insulin-Secreting Cells/cytology , Sirolimus/pharmacology , Animals , Female , Insulin-Secreting Cells/drug effects , Mice , Mice, Inbred C57BL , Pregnancy , Pregnancy, Animal/drug effects , Pregnancy, Animal/physiology
16.
Transplant Proc ; 40(2): 455-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374099

ABSTRACT

Immunomodulatory properties have been recognized for human alpha-1 antitrypsin (hAAT). However, production of anti-hAAT antibodies in mice may inactivate the protein. In this study, we evaluated the effects of chronic hAAT administration on allogeneic islet graft survival. Chemically diabetic mice lacking an efficient humoral response due to the targeted disruption of the Ig mu-chain (muMT mice) or wild-type (WT) C57BL/6 mice received DBA/2 mouse islets under the kidney capsule. hAAT (Prolastin or Aralast) was given intraperitoneally on day 0 and every 3 days thereafter. Control animals received no treatment. hAAT administration in WT mice resulted in prolongation of islet allograft survival in a dose-dependent fashion in both hAAT-treated groups. Lack of Ig response (muMT mice) per se conferred a beneficial effect on graft survival that worsened in the Prolastin-treated groups but improved in the Aralast-treated group. Our data indicate that systemic administration of hAAT results in prolongation of islet allograft survival. Absence of mature B cells and Ig mu-chain resulted in improved graft survival, pointing to a role for B cells in the rejection process in this model. Treatment with Prolastin worsened graft survival in muMT mice, whereas Aralast did improve it, suggesting a different efficacy and possible actions of the two drug formulations.


Subject(s)
Antibody Formation/physiology , Diabetes Mellitus, Experimental/surgery , Graft Survival/drug effects , Islets of Langerhans Transplantation/physiology , alpha 1-Antitrypsin/therapeutic use , Animals , Antibody Formation/drug effects , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Subrenal Capsule Assay , Transplantation, Homologous
17.
Transplant Proc ; 40(2): 457-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374100

ABSTRACT

Alpha-1 antitrypsin (AAT) is a serine protease inhibitor able to prevent diabetes onset in nonobese diabetic (NOD) mice and to prolong islet allograft survival in a nonautoimmune murine model. In this study, we explored the effect of chronic administration of human AAT (hAAT) on allogeneic (C57BL/6) islet graft survival in spontaneously diabetic female NOD mice. Mice received intraperitoneal treatment with saline, Prolastin (1 or 2 mg/mouse) or Aralast (2 mg/mouse) on days -1, 0, 3, 6, and 9. Saline-treated mice rejected the grafts 10.0 +/- 2.5 days after transplantation (n = 9). Prolastin 1 mg (n = 9) and 2 mg (n = 3) resulted in rejection on 8.7 +/- 1.4 (not significant) and 13.0 +/- 4.3 days (P < .03), respectively. Aralast-treated mice showed prolongation of graft survival (13 +/- 5.9 days; n = 5; P < .03). Notably, repeated administrations of either hAAT formulation led to sudden death of a proportion of treated animals. Collectively, our preliminary data indicate that prolongation of islet allograft survival in the stringent autoimmune diabetic NOD mouse model can be achieved with hAAT monotherapy. The death of a proportion of treated animals may be consequent to immunization to hAAT and lethal hypersensitivity. Interestingly, this phenomenon was not observed in a non-autoimmune mouse strain (C57BL/6) despite extended hAAT treatment (>100 days).


Subject(s)
Graft Survival/drug effects , Islets of Langerhans Transplantation/physiology , Serine Proteinase Inhibitors/therapeutic use , alpha 1-Antitrypsin/therapeutic use , Animals , Female , Graft Rejection/prevention & control , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Transplantation, Homologous
18.
Diabetologia ; 51(2): 298-308, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18066521

ABSTRACT

AIMS/HYPOTHESIS: Activation of c-jun N-terminal kinase (JNK) has been described in islet isolation and engraftment, making JNK a key target in islet transplantation. The objective of this study was to investigate if JNK inhibition with a cell-permeable TAT peptide inhibitor (L-JNKI) protects functional beta cell mass in human islets and affects AKT and its substrates in islet cells. METHODS: The effect of L-JNKI (10 micromol/l) on islet count, mitochondrial membrane potential, glucose-stimulated insulin release and phosphorylation of both AKT and its substrates, as well as on reversal of diabetes in immunodeficient diabetic Nu/Nu mice was studied. RESULTS: In vitro, L-JNKI reduced the islet loss in culture and protected from cell death caused by acute cytokine exposure. In vivo, treatment of freshly isolated human islets and diabetic Nu/Nu mice recipients of such islets resulted in improved functional beta cell mass. We showed that L-JNKI activates AKT and downregulates glycogen synthase kinase-3 beta (GSK-3B) in human islets exposed to cytokines, while other AKT substrates were unaffected, suggesting that a specific AKT/GSK-3B regulation by L-JNKI may represent one of its mechanisms of cytoprotection. CONCLUSIONS/INTERPRETATION: In conclusion, we have demonstrated that targeting JNK in human pancreatic islets results in improved functional beta cell mass and in the regulation of AKT/GSK3B activity.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Insulin-Secreting Cells/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Peptides/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Combined Modality Therapy , Cytokines/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/surgery , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/methods , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Nude , Peptides/therapeutic use , Phosphorylation/drug effects , Transplantation, Heterologous , Treatment Outcome
19.
Diabetologia ; 48(2): 268-76, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15690148

ABSTRACT

AIMS/HYPOTHESIS: Despite differences in function and embryonic origin, pancreatic islet cells and neurons express proteins belonging to the tumour necrosis factor receptor superfamily. While neurons express the CD40 receptor, it is unknown whether islet cells also express it. We investigated CD40 expression in human and mouse pancreatic islets as well as in NIT-1 insulinoma cells. METHODS: CD40 expression was studied by reverse transcriptase polymerase chain reaction, flow cytometry, immunohistochemistry and western blot. Responses mediated by CD40 were assessed by a luciferase gene reporter assay following stimulation with a CD40 agonist antibody. RESULTS: We found that CD40 is expressed in mouse and human pancreatic islet cells. CD40 is expressed by beta cells, and its expression is upregulated by proinflammatory cytokines (IL-1beta, IFN-gamma and TNF-alpha). CD40 signalling in NIT-1 insulinoma cells activates nuclear factor kappa-B, demonstrating that CD40 is functional. CONCLUSIONS/INTERPRETATION: We present evidence that, in addition to immune cell types, mouse and human pancreatic beta cells express CD40. Its expression is upregulated by proinflammatory stimuli, and signalling through this receptor activates NF-kappaB. We suggest that the effects of inflammatory stimuli that affect beta cell function and survival may be also mediated by signalling through the CD40 receptor. Thus, CD40 may have a role in processes associated with islet autoimmunity and transplantation.


Subject(s)
CD40 Antigens/genetics , Islets of Langerhans/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Base Sequence , CD40 Antigens/immunology , DNA Primers , DNA, Complementary/genetics , Genes, Reporter , Humans , Luciferases/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , NF-kappa B/genetics , RNA, Messenger/genetics
20.
Cell Transplant ; 10(7): 633-7, 2001.
Article in English | MEDLINE | ID: mdl-11714198

ABSTRACT

A role of macrophage-mediated inflammatory events in early islet graft loss is increasingly acknowledged. Osteopetrotic mice (op/op) have a complete absence of CSF-1, and thus of most tissue macrophages. We have investigated whether the absence of CSF-1-dependent macrophages in the graft itself or at the transplant site could decrease the delay to function of a syngeneic marginal islet mass. Islets transplanted into op/op or control recipients reversed diabetes in 59 days vs. 10 days (p = 0.28, NS). Islets isolated from op/op or control mice reversed diabetes in 11 days vs. 10 days. IL-1 and TNF-alpha release by cultured islets was markedly decreased for op/op islets compared with control islets (IL-1: 0 vs. 4.2 pg/ml, p = 0.07; TNF-alpha: 67 vs. 311 pg/ml, p = 0.002). In contrast, IL-6 release by op/op islets was significantly increased (11.1 vs. 4.3 ng/ml, p = 0.006). CSF-1-dependent tissue macrophages may not be critical in the inflammatory insult to islet transplants. Alternate patterns of intraislet release of deleterious proinflammatory cytokines may exist.


Subject(s)
Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Macrophage Colony-Stimulating Factor/genetics , Macrophages/immunology , Osteopetrosis/genetics , Animals , Diabetes Mellitus, Type 1/therapy , Female , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Osteopetrosis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...