Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Surgery ; 138(2): 342-51, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16153446

ABSTRACT

BACKGROUND: Evidence is emerging that the endoplasmic reticulum (ER) participates in initiation of apoptosis induced by the unfolded protein response and by aberrant Ca(++) signaling during cellular stress such as ischemia/reperfusion injury (I/R injury). ER-induced apoptosis involves the activation of caspase-12 and C/EBP homologous protein (CHOP), and the shutdown of translation initiated by phosphorylation of eIF2alpha. Sodium 4-phenylbutyrate (PBA) is a low molecular weight fatty acid that acts as a chemical chaperone reducing the load of mutant or unfolded proteins retained in the ER during cellular stress and also exerting anti-inflammatory activity. It has been used successfully for treatment of urea cycle disorders and sickle cell disease. Thus, we hypothesized that PBA may reduce ER-induced apoptosis triggered by I/R injury to the liver. METHODS: Groups of male C57BL/6 mice were subjected to warm ischemia (70% of the liver mass, 45 minutes). Serum aspartate aminotransferase was assessed 6 hours after reperfusion; apoptosis was evaluated by enzyme-linked immunosorbent assays of caspase-12 and plasma tumor necrosis factor alpha, Western blot analyses of eIF2alpha, and reverse transcriptase-polymerase chain reaction of CHOP expression. RESULTS: A dose-dependent decrease in aspartate aminotransferase was demonstrated in mice given intraperitoneal PBA (1 hour before and 12 hours after reperfusion), compared with vehicle-treated controls; this effect was associated with reduced pyknosis, parenchymal hemorrhages, and neutrophil infiltrates in PBA-treated mice, compared with controls. In a lethal model of total liver I/R injury, all vehicle-treated controls died within 3 days after reperfusion. In contrast, 50% survival (>30 days) was observed in animals given PBA. The beneficial effects of PBA were associated with a greater than 45% reduction in apoptosis, decreased ER-mediated apoptosis characterized by significant reduction in caspase-12 activation, and reduced levels of both phosphorylated eIF2alpha and CHOP. Significant reductions in plasma levels of tumor necrosis factor alpha and liver myeloperoxidase content were demonstrated after PBA treatment. CONCLUSIONS: Reduction in ER stress-induced hepatocellular injury was achieved by the administration of PBA. Targeting the ER-associated cell death pathway might offer a novel approach to reduce I/R injury to the liver.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum/drug effects , Phenylbutyrates/pharmacology , Reperfusion Injury/drug therapy , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Caspase 12 , Caspases/metabolism , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/metabolism , Hot Temperature , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Transcription Factor CHOP , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Biomaterials ; 26(4): 403-12, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15275814

ABSTRACT

Functional poly(ethylene glycol) (PEG) derivatives, including monosuccinimidyl PEG (MSPEG) with molecular weight (MW) of 2000 (2 kDa) as well as 5 kDa and disuccinimidyl PEG (DSPEG) with MW of 3 and 6 kDa, were synthesized and characterized. They were used to modify the surface of adult porcine islets for cytoprotection. The islets were isolated, purified and modified with functional PEG. Untreated porcine islets were used as control. An in vitro human antibody/complement-mediated cytotoxicity test based on the release of intracellular lactate dehydrogenase was used to evaluate cytotoxicity of human serum to the modified islets. In vitro cell viability was assessed using membrane-integrity straining and islet metabolism in culture. In vitro islet functionality was evaluated by glucose-stimulated insulin release of islets in static incubation with human serum. In vivo islet functionality was evaluated by monitoring non-fasting blood glucose level in streptozotocin-induced diabetic (SCID) immunocompromized mice after intraportal transplantation of porcine islets. Results show that all the PEG derivatives used in the study showed significant in vitro and in vivo cytoprotections against cytotoxic effects elicited by human serum and diabetic SCID mice, respectively, to porcine islets. DSPEG derivatives combined with human albumin exhibited a better cytoprotection, as compared to MSPEG ones, due to the capacity of the succinimidyl groups to selectively react with amino groups of the albumin under physiological conditions. The effects of both MW and concentration of the PEG derivatives on cytoprotection were significant. It appears that this novel biotechnology will be an attractive approach for improved xenotransplantation of islets.


Subject(s)
Cytoprotection/physiology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/methods , Islets of Langerhans/physiology , Pancreas, Artificial , Polyethylene Glycols/chemistry , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Blood Glucose/analysis , Cattle , Cells, Cultured , Diabetes Mellitus, Experimental/diagnosis , Female , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Male , Materials Testing , Mice , Mice, SCID , Transplantation, Heterologous
3.
Diabetes ; 53(11): 2804-14, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15504960

ABSTRACT

Clinical studies indicate that significant loss of functional islet mass occurs in the peritransplant period. Islets are injured as a result of detrimental effects of brain death, pancreas preservation, islet isolation, hypoxia, hyperglycemia, and immune-mediated events. In addition, recent studies demonstrated that islets are injured as a result of their exposure to blood and of activation of intrahepatic endothelial and Kupffer cells, resulting in inflammation and thrombosis. Activated protein C (APC) is an anticoagulant enzyme that also exerts anti-inflammatory and antiapoptotic activities by acting directly on cells. Here, we report that exogenous administration of recombinant murine APC (mAPC) significantly reduced loss of functional islet mass after intraportal transplantation in diabetic mice. Animals given mAPC exhibited better glucose control, higher glucose disposal rates, and higher arginine-stimulated acute insulin release. These effects were associated with reduced plasma proinsulin, intrahepatic fibrin deposition, and islet apoptosis early after the transplant. In vitro and in vivo data demonstrated that mAPC treatment was associated with a significant reduction of proinflammatory cytokine release after exposure of hepatic endothelial cells to islets. mAPC treatment also prevented endothelial cell activation and dysfunction elicited by intrahepatic embolization of isolated islets inherent to pancreatic islet transplantation (PIT). This study demonstrates multiple remarkable beneficial effects of mAPC for PIT and suggests that APC therapy may enhance the therapeutic efficacy of PIT in diabetic patients.


Subject(s)
Islets of Langerhans Transplantation/physiology , Islets of Langerhans/drug effects , Protein C/pharmacology , Animals , Arginine/pharmacology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/surgery , Enzyme Activation , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/anatomy & histology , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Transplantation, Isogeneic/physiology
4.
Surgery ; 136(3): 537-47, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15349100

ABSTRACT

BACKGROUND: Effective cytoprotection to xenoislets would circumvent the major tissue limitation for pancreatic islet transplantation (PIT). Cell-surface engineering with poly[ethylene glycol] (PEG) derivatives can successfully prevent antibody binding to the surface antigens. Gene transfer of the antiapoptotic Bcl-2 gene has been shown to decrease cytotoxicity mediated by xenoreactive natural antibodies and complement. In this study, we assessed survival and function of surface-engineered porcine islets genetically modified to overexpress Bcl-2. METHODS: Incorporation of PEG derivatives into the islet surface and adenovirus-mediated gene transfer of Bcl-2 (AdBcl-2) was accomplished within 24 hours post-isolation. Cytotoxicity induced by human xenoreactive natural antibodies was evaluated by islet intracellular lactate dehydrogenase release and microscopic analysis using membrane-integrity staining. Islet functionality was assessed by static incubation and after intraportal infusion (5000 IEQ) into diabetic NOD-SCID mice reconstituted with human lymphocytes (5 x 10 8 /intraperitoneally/15 days before PIT). RESULTS: No significant change in islet viability, morphology, and functionality was demonstrated after the incorporation of PEG-mono-succimidyl-succinate (MSPEG), or PEG-di-succimidyl-succinate "end"-capped with albumin (DSPEG) with or without gene transfer of Bcl-2. Islets treated with MSPEG presented a significant reduction in lactate dehydrogenase release compared with controls (41.2 +/- 3 vs 72.1 +/- 7, respectively, P <.05). Further protection was accomplished by DSPEG or AdBcl-2. The maximal cytoprotection was achieved by DSPEG +AdBcl-2 (15.5 +/- 4.9%, P <.001). Nonfasting glucose >200 mg/dL was found in 100% of the animals given control islets (n = 6) within 48 hours post-transplant. In contrast, euglycemia was achieved in 100% of the animals given islets modified with DSPEG + AdBcl-2 during the observation time. CONCLUSIONS: Surface-engineering with functionalized PEG derivatives in combination with genetic modification with Bcl-2 significantly reduced islet loss after PIT. Application of this novel technology may improve results in xenoislet transplantation.


Subject(s)
Gene Transfer Techniques , Genes, bcl-2/genetics , Islets of Langerhans Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Apoptosis/immunology , Cell Survival/immunology , Cells, Cultured , Gene Expression , Islets of Langerhans/immunology , Models, Animal , Polyethylene Glycols/pharmacology , Surface-Active Agents/pharmacology , Swine
5.
Surgery ; 136(2): 336-45, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15300200

ABSTRACT

BACKGROUND: Current isolation techniques recover only 20% to 50% of the pancreatic islets. Brain death (BD) is characterized by activation of proinflammatory cytokines (PICs) with reduced islet yields and functionality. We previously reported that 17beta-estradiol (E2) induces cytoprotection to human islets exposed to PICs. Furthermore, inhibition of PIC release has been demonstrated after E2 treatment. In the present study, we evaluated if E2 treatment to BD donors would improve pancreatic islet recovery and functionality. METHODS: BD was induced in male, 250- to 350-g Lewis rats by inflation of a Fogarty catheter placed intracranially. Rats were mechanically ventilated for 6 hours. Only rats with mean arterial blood pressure > 75 mm Hg were used. Animals (n = 6) received E2 (1 mg/kg/iv immediately after BD induction), vehicle (V), or the combination of 17beta-estradiol and a selective estrogen receptor antagonist ICI 182,780 (ICI, 3 mg/kg/ip/1 hour before BD induction). Islet viability was determined by ethidium bromide-acridine orange. PICs were assessed by ELISA. Islet functionality was determined by static incubation and glucose disposal rate (Kg) after intraportal transplantation (3000 islet equivalent[IEQ]/syngeneic streptozotocin-induced diabetic rat). RESULTS: A 2- to 3-fold reduction in TNF-alpha, IL-1beta, and IL-6 was demonstrated in BD donors given E2; this effect reversed by ICI 182,780. Pancreatic sections from control BD donors presented 26.5% +/- 4% TUNEL-positive beta-cells compared with 15.1% +/- 3% in 17beta-estradio-treated animals. Islet recovery was enhanced in E2-treated donors (1233.4 +/- 123 IEQ/pancreas) compared with controls (725 +/- 224 IEQ, P < .05). Islet viability was significantly enhanced by E2. Higher islet functionality was demonstrated in vitro and in vivo after transplantation in islets recovered from E2-treated BD donors. CONCLUSIONS: Islet recovery and functionality in vitro and in vivo were significantly improved by 17beta-estradiol treatment to BD donors. These observations may lead to strategies to reduce the effects of BD on isolated islets and improve the results in clinical islet transplantation.


Subject(s)
Brain Death , Estradiol/pharmacology , Islets of Langerhans/drug effects , Tissue Donors , Animals , Apoptosis/drug effects , Cytokines/biosynthesis , Islets of Langerhans/physiology , Islets of Langerhans Transplantation , Male , Rats , Rats, Inbred Lew
6.
ILAR J ; 45(3): 334-42, 2004.
Article in English | MEDLINE | ID: mdl-15229380

ABSTRACT

The recent success of "steroid-free" immunosuppressive protocols and improvements in islet preparation techniques have proven that pancreatic islet transplantation (PIT) is a valid therapeutic approach for patients with type 1 diabetes. However, there are major obstacles to overcome before PIT can become a routine therapeutic procedure, such as the need for chronic immunosuppression, the loss of functional islet mass after transplantation requiring multiple islet infusion to achieve euglycemia without exogenous administration of insulin, and the shortage of human tissue for transplantation. With reference to the first obstacle, stable islet allograft function without immunosuppressive therapy has been achieved after tolerance was induced in diabetic primates. With reference to the second obstacle, different strategies, including gene transfer of antiapoptotic genes, have been used to protect isolated islets before and after transplantation. With reference to the third obstacle, pigs are an attractive islet source because they breed rapidly, there is a long history of porcine insulin use in humans, and there is the potential for genetic engineering. To accomplish islet transplantation, experimental opportunities must be balanced by complementary characteristics of basic mouse and rat models and preclinical large animal models. Well-designed preclinical studies in primates can provide the quality of information required to translate islet transplant research safely into clinical transplantation.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Disease Models, Animal , Haplorhini , Islets of Langerhans Transplantation , Monkey Diseases/surgery , Animals , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/pathology , Monkey Diseases/etiology , Monkey Diseases/pathology , Pancreatectomy , Streptozocin
7.
Diabetes ; 52(12): 2935-42, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14633854

ABSTRACT

Although approximately 1 million islets exist in the adult human pancreas, current pancreas preservation and islet isolation techniques recover <50%. Presently, cadaveric donors remain the sole source of pancreatic tissue for transplantation. Brain death is characterized by activation of proinflammatory cytokines and organ injury during preservation and reperfusion. In this study, we assessed the effects of brain death on islet isolation yields and functionality. Brain death was induced in male 250- to 350-g Lewis rats by inflation of a Fogarty catheter placed intracranially. The rats were mechanically ventilated for 2, 4, and 6 h before removal of the pancreas (n = 6). In controls, the catheter was not inflated (n = 6). Shortly after brain death induction, a significant increase in serum tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-1beta, and IL-6 was demonstrated in a time-dependent manner. Upregulation of TNF-alpha, IL-1beta, and IL-6 mRNA was noted in the pancreas. Brain death donors presented lower insulin release after glucose stimulation assessed by in situ perfusion of the pancreas. Islet recovery was reduced in brain death donors compared with controls (at 6 h 602.3 +/- 233.4 vs. 1,792.5 +/- 325.4 islet equivalents, respectively; P < 0.05). Islet viability assessed in dissociated islet cells and in intact cultured islets was reduced in islets recovered from brain death donors, an effect associated with higher nuclear activities of NF-kappaB p50, c-Jun, and ATF-2. Islet functionality evaluated in vitro by static incubation and in vivo after intraportal transplantation in syngeneic streptozotocin-induced diabetic rats was significantly reduced in preparations obtained from brain death donors. In conclusion, brain death significantly reduced islet yields and functionality. These observations may lead to strategies to reduce the effects of brain death on pancreatic islets and improve the results in clinical transplantation.


Subject(s)
Brain Death/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/surgery , Insulin/metabolism , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Animals , Apoptosis , Cell Nucleus/metabolism , Cytokines/metabolism , Graft Survival , In Vitro Techniques , Inflammation Mediators/metabolism , Insulin Secretion , Male , Rats , Rats, Inbred Lew , Recovery of Function , Tissue Donors , Tissue Survival , Transcription Factors/metabolism
8.
Surgery ; 134(2): 169-79, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12947315

ABSTRACT

BACKGROUND: The c-Jun N-terminal kinase (JNK) activation occurs after islet isolation, oxidative stress, and proinflammatory cytokine (PIC) exposure to beta-cells. Previous studies demonstrated that 17beta-estradiol modulates the activity of JNK; therefore we assessed the effects of 17beta-estradiol on JNK activation on islet survival and function after transplantation. METHODS: Isolated human pancreatic islets were incubated with PIC and 17beta-estradiol. Viability was analyzed by a colorimetric assay, islet mass by DNA content, JNK activity by Western blots, AP-1 nuclear activity with a promoter-Luciferase AP-1 responsive construct, and c-Fos, Jun-D, and ATF-2 nuclear activities by an enzyme-linked immunosorbent assay. Islet functionality was evaluated after transplantation in streptozotocin-induced diabetic NOD-SCID mice. RESULTS: The 17beta-estradiol enhanced islet viability and islet mass after exposure to PIC. A significant reduction in JNK activation occurred in islets treated with 17beta-estradiol, compared with controls, an effect partially dependent on estrogen receptors. The 17beta-estradiol induced a significant reduction in nuclear AP-1, c-fos, Jun-D, and ATF-2 activities. Animals that received 17beta-estradiol-treated islets had better islet functionality compared with saline solution-treated controls. CONCLUSIONS: The 17beta-estradiol improved isolated human pancreatic islets survival after PIC exposure by inhibition of JNK. These effects were associated with reduction in JNK targets, including the nuclear activities of transcription factors AP-1, c-Jun, c-Fos, Jun-D and ATF-2, involved in apoptosis in beta-cells. The 17beta-estradiol therapy may improve the results in clinical transplantation.


Subject(s)
Cytokines/pharmacology , Estradiol/pharmacology , Inflammation Mediators/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Activating Transcription Factor 2 , Animals , Blood Glucose/analysis , Cell Death/physiology , Cell Nucleus/metabolism , Culture Techniques , Cyclic AMP Response Element-Binding Protein/metabolism , Diabetes Mellitus/blood , Diabetes Mellitus/surgery , Down-Regulation , Enzyme Activation/drug effects , Humans , Islets of Langerhans Transplantation , JNK Mitogen-Activated Protein Kinases , Male , Mice , Mice, SCID , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Tissue Survival/drug effects , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism
9.
Surgery ; 134(2): 390-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12947346

ABSTRACT

BACKGROUND: Pancreatic islet transplantation has been validated as a treatment for type 1 diabetes. However, a high number of islets is required to establish euglycemia. Transplantation of islets leads to loss of islet vasculature, which requires revascularization to ensure adequate survival. Islet vascular density in transplanted islets is markedly decreased compared with endogenous islets. The feasibility of revascularization of ischemic tissues by mobilizing endothelial progenitor cells or angioblasts has been demonstrated. Therefore, we investigated the therapeutic potential of angioblast mobilization for stimulation of islet revascularization and therefore engraftment after transplantation. METHODS: FVB/NJ mice underwent bone marrow transplantation from transgenic mice constitutively expressing beta-galactosidase encoded by LacZ under regulation of the endothelial cell-specific promoter TIE-2 (FEV/NJ-TIE-2-LacZ). Three weeks after reconstitution, animals received an intrahepatic islet syngeneic infusion (FVB/NJ donors). The contribution of angioblasts into sites of islet revascularization was analyzed by reverse transcriptase-polymerase chain reaction (RT-PCR), beta-galactosidase (beta-gal) activity, and immunohistochemistry. Islet vascular density was assessed morphometrically followed by in situ BS-1 lectin staining and functional islet mass after transplantation by metabolic studies. Angioblasts were mobilized with murine granulocyte-macrophage colony-stimulating factor (GM-CSF) (0.5 microg/day/7 days). RESULTS: An islet dose-dependent increase in beta-gal was demonstrated after transplantation. These results were confirmed by RT-PCR and immunohistochemistry. GM-CSF increased the number of peripheral angioblasts and their localization into sites of islet revascularization. A significant increase in islet vascular density was observed in animals treated with GM-CSF versus controls. Higher functional islet mass was demonstrated in animals treated with GM-CSF. CONCLUSIONS: Augmentation of angioblasts in the peripheral circulation resulted in higher islet vascular density and engraftment. This novel strategy may improve the results in clinical islet transplantation.


Subject(s)
Endothelium, Vascular/physiopathology , Hematopoietic Stem Cell Mobilization , Islets of Langerhans Transplantation , Islets of Langerhans/blood supply , Portal Vein , Animals , Endothelium, Vascular/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunohistochemistry , Injections, Intravenous , Islets of Langerhans/enzymology , Islets of Langerhans/physiopathology , Islets of Langerhans Transplantation/methods , Mice , Mice, Inbred Strains , Mice, Transgenic , Neovascularization, Physiologic/drug effects , Reverse Transcriptase Polymerase Chain Reaction , beta-Galactosidase/metabolism
10.
Transplantation ; 76(1): 252-61, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12865820

ABSTRACT

BACKGROUND: New strategies for improving durable functional islet mass will be instrumental in facilitating islet transplantation as a cure for type 1 diabetes mellitus. The ability to transfer immunoregulatory or cytoprotective genes into pancreatic islets may enhance survival. Adenoviral vectors (Ad5) have been used widely to deliver therapeutic genes to different tissues. Limitations associated with the use of Ad5 for gene therapy are related to the reliance of the virus on the presence of its primary receptor, the transient nature of the transgene expression, and the immediate inflammatory and immune response elicited by the infection. Because the arginine-glycine-aspartame (RGD) and polylysine (pK7) motifs have been shown to enhance Ad5 infection through an Ad5 receptor-independent pathway, we hypothesized that they could act additively to improve infectivity and reduce toxicity to isolated human pancreatic islets (IHPI). METHODS: Hand-picked IHPI were infected with nonmodified Ad5, single-modified Ad5 with RGD (Ad5RGD) or pK7 (ad5pK7), and Ad5RGDpK7. Transfection efficiency was evaluated by green fluorescent protein and luciferase expression. Apoptosis was assessed using a quantitative assay, activation of caspase 3 by a colorimetric assay, nuclear factor (NF)-kappaB nuclear translocation using a promoter-luciferase NF-kappaB responsive construct, regulated on activation normal T-cell expressed and secreted (RANTES) by enzyme-linked immunosorbent assay. In vivo functionality was evaluated after transplantation into diabetic nonobese diabetic severe combined immunodeficiency mice. RESULTS: Compared with unmodified and singly-modified Ad5 vectors, Ad5RGDpK7 demonstrated the highest infectivity. After the infection of IHPI with adenoviral vectors using the minimal dose required to infect greater than 80% of the islet cells (Ad5, 500 viral particles [VP]/cell; Ad5RGD and Ad5pK7, 10 VP/cell; Ad5RGDpK7, 0.1 VP/cell), islets infected with Ad5RGDpK7 presented a significant reduction in apoptosis, NF-kappaB nuclear translocation, RANTES expression, and higher glucose disposal rate; reduced Ad5-driven specific Th1 and antibody response were also observed. CONCLUSIONS: Ad5RGDpK7 exhibited higher transfection efficiency, allowing a significant reduction in the viral dose required to infect greater than 80% of the islet cells. The reduction in the viral dose was associated with reduced toxicity, inflammation, and immune responses related to Ad5 infection. This strategy may thus be used to successfully modify isolated pancreatic islets.


Subject(s)
Adenoviridae/physiology , Genetic Vectors , Islets of Langerhans Transplantation/methods , Islets of Langerhans/physiology , Oligopeptides/pharmacology , Polylysine/pharmacology , Transplantation, Heterologous/methods , Adenoviridae/genetics , Animals , Cadaver , Diabetes Mellitus, Experimental/surgery , Gene Transfer Techniques , Humans , Islets of Langerhans/virology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Tissue Donors
11.
Transpl Int ; 16(7): 537-42, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12819863

ABSTRACT

A variety of toxic insults can result in endoplasmic reticulum (ER)-stress that ultimately leads to apoptosis. beta-cells have a highly developed ER due to a great commitment to insulin production. The present study was carried out to determine the role of ER-stress in isolated human pancreatic islet apoptosis, and the potential protective effects of Bcl-2. Isolated human islets were infected with an adenoviral vector encoding Bcl-2 and then exposed to brefeldin-A, tunicamycin, A23187 and pro-inflammatory cytokines. Activation of caspase-12 was analyzed by means of Western blots. Apoptosis was evaluated using a commercial quantitative assay. ER-stress-inducers promoted caspase-12 activation and apoptosis, effect reversed by overexpression of Bcl-2. Co-localization of caspase-12 and Bcl-2 in the microsomal islet fractions were demonstrated by means of Western blots. We can conclude that the current studies highlight the importance of Bcl-2 as an anti-apoptotic protein, and shed new light on the mechanisms underlying its cytoprotective effects on pancreatic islets.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum/physiology , Gene Transfer Techniques , Genes, bcl-2 , Islets of Langerhans/physiology , Apoptosis/drug effects , Blotting, Western , Brefeldin A/pharmacology , Calcimycin/pharmacology , Caspase 12 , Caspases/metabolism , Cytokines/pharmacology , Endoplasmic Reticulum/drug effects , Enzyme Activation/drug effects , Humans , Inflammation Mediators/pharmacology , Islets of Langerhans/drug effects , Proto-Oncogene Proteins c-bcl-2/pharmacology , Proto-Oncogene Proteins c-bcl-2/physiology , Tunicamycin/pharmacology
12.
Transplantation ; 74(9): 1252-9, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12451262

ABSTRACT

INTRODUCTION: Proinflammatory cytokines (PIC) (interleukin-1beta, interferon-gamma, and tumor necrosis factor alpha) are released after intraportal islet transplantation lead to functional suppression and islet apoptosis. Estradiol has been shown to promote survival of cells undergoing PIC-induced apoptosis. In this study, we evaluated the effects of estradiol on isolated human pancreatic islet (IHPI) survival after exposure to PIC and analyzed potential mechanisms of action. METHODS: Hand-picked, freshly isolated IHPI were incubated with PIC and estradiol. Viability was analyzed from single islet cells stained with ethidium bromide and acridine orange, apoptosis using a quantitative kit, NF-kappaB nuclear translocation using a promoter-Luciferase NF-kappaB responsive construct, mitochondrial permeability transition using the ApoAlert Mitochondrial kit, and caspase 9 by a fluorometric assay. In vitro functionality was examined by static incubation, and a limited number of islets were transplanted in nonobese diabetic, severe combined immunodeficient mice. RESULTS: 17beta-Estradiol induced a dose-dependent increase in islet viability, an effect partially reversed by the estrogen receptor antagonist ICI 182,780. In vitro, islets treated with estradiol presented higher stimulation index. Euglycemia was achieved in 6 of 12 animals that received estradiol-treated islets compared with 1 of 12 control animals. Lower NF-kappaB nuclear translocation, cytochrome release, and caspase 9 activation occurred in islets treated with estradiol. CONCLUSIONS: Estradiol promoted IHPI survival and improved functionality after PIC exposure in vitro and in vivo after transplantation. The molecular mechanisms involved included a decrease in NF-kappaB nuclear translocation, decrease in mitochondrial cytochrome release, and caspase 9 activation. The use of estradiol might be beneficial in clinical islet transplantation.


Subject(s)
Cytokines/pharmacology , Estradiol/pharmacology , Inflammation Mediators/pharmacology , Islets of Langerhans/drug effects , Animals , Biological Transport , Blood Glucose/analysis , Cell Death/drug effects , Cell Nucleus/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/surgery , Glucose/pharmacology , Humans , In Vitro Techniques , Insulin/metabolism , Islets of Langerhans Transplantation , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Mitochondria/physiology , NF-kappa B/metabolism
13.
Transplantation ; 74(8): 1063-9, 2002 Oct 27.
Article in English | MEDLINE | ID: mdl-12438947

ABSTRACT

BACKGROUND: Pancreatic islets are susceptible to myriad insults that occur during islet isolation and transplantation. Studies demonstrated the role of Akt in regulating pancreatic beta-cell growth and survival. Activation of Akt maintains Bad phosphorylation and prevents its binding to mitochondrial targets, decreases caspase-9 activity, and prevents the translocation of forkhead transcription factors (FKHR). Simvastatin activates Akt in mammalian cells; therefore, we investigated the role of simvastatin on human pancreatic islets (HPI) survival. METHODS: HPI were treated with simvastatin, with and without LY294002, an inhibitor of phosphoinositide 3-kinase. PI viability was examined with ethidium bromide-acridine orange, and apoptosis was examined using a quantitative assay. Akt, Bad, FKHR phosphorylation, and mitochondrial cytochrome release were analyzed by Western blots. Caspase-9 activity was assessed by a fluorometric assay. A limited number of HPI were transplanted after simvastatin treatment in diabetic NOD-SCID mice. RESULTS: Low levels of Akt phosphorylation (activation) were demonstrated early after islet isolation. Akt activation; increase in islet viability; and decrease in Bad phosphorylation, cytochrome release, caspase-9 activation, and translocation of FKHR were observed after simvastatin treatment, effects reversed by LY294002. Among recipients of islets without simvastatin, none demonstrated reversal of diabetes after the transplant. In contrast, 58% of the recipients given islets treated with simvastatin remained euglycemic 30 days after the transplant. CONCLUSIONS: Targeting the survival pathway with simvastatin exerts a cytoprotective effect on isolated PI. Activation of the Akt pathway is a potential new therapeutic approach to reduce loss of functional islet mass to bolster success in clinical islet transplantation.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Simvastatin/pharmacology , Animals , Carrier Proteins/metabolism , Caspase 9 , Caspases/metabolism , Cell Survival , Cells, Cultured , Cytochrome c Group/metabolism , Diabetes Mellitus, Type 1/therapy , Fas Ligand Protein , Forkhead Transcription Factors , Gene Expression , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/metabolism , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt , Transcription Factors/metabolism , bcl-Associated Death Protein
14.
Am J Transplant ; 2(3): 237-43, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12096786

ABSTRACT

The ability to transfer immunoregulatory, cytoprotective, or antiapoptotic genes into pancreatic islets (PIs) may allow enhanced post-transplantation survival. The available gene transfer vectors differ greatly in their ability to infect and express genes in different cell types. One limitation associated with the use of viral vectors is related to the virus reliance on the presence of its primary binding site. Tropism of the viral vectors can be altered using retargeting strategies. Results on phage biopanning proved that the RGD motif has in vivo targeting capabilities. This motif interacts especially with cellular integrins of the alphavbeta3 and alphavbeta5 types, highly expressed on pancreatic islets. In this report, we have explored the utility of a retargeted adenovirus vector (Ad) containing an RGD motif in the HI loop of the fiber knob in order to improve the infection efficiency to intact isolated nonhuman primate PIs and reduce toxicity after the genetic modification. Nonhuman primate Pis were isolated by a semi-automated technique. Steptozotocin-induced diabetic mice with severe combined immunodeficiency disease (SCID) were used as recipients. A recombinant Ad containing a heterologous RGD peptide and expressing luciferase (AdRGDLuc) or green fluorescent protein (AdRGDGFP) were generated in our laboratory. Similar Ads without the RGD peptide were used as a control (AdLuc and AdGFP). Higher transfection efficiency was demonstrated using AdRGDGFP compared with AdGFP (>80% of the islet cells were infected at 10 particle-forming units (pfu)/cell using AdRGDGFP vs. 7% after infection with AdGFP).More than 90% of the infected cells were insulin-producing cells. Significantly higher transgene expression was demonstrated after infection with AdRGDLuc compared with AdLuc at different titers. Analysis of the glucose-stimulated insulin response demonstrated better performance of PI transfected with AdRGDLuc at low titers (10 pfu/cell in order to achieve > 80% transfection efficiency) compared with AdLuc at high titers. Finally, long-term euglycemia (>250d) was observed in 89% of the animals that received PI infected with AdRGDLuc compared with none of the animals that received PI infected with AdLuc. The present study provides new information about the possibility of tropism modification of Ad vectors to increase the transfection efficiency and transgene expression to isolated PI. Incorporation of the RGD sequence in the HI loop of the fiber knob allows highly efficient transfection efficiency to nonhuman primate insulin-producing cells and adequate long-term function of the p-cell after transplantation.


Subject(s)
Adenoviridae/genetics , Diabetes Mellitus, Experimental/surgery , Genetic Vectors , Islets of Langerhans Transplantation/physiology , Islets of Langerhans/physiology , Oligopeptides , Transplantation, Heterologous/physiology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 1/surgery , Genes, Reporter , Glucose/pharmacology , Green Fluorescent Proteins , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Luciferases/genetics , Luminescent Proteins/genetics , Macaca mulatta , Male , Mice , Mice, SCID , Primates , Transfection/methods
15.
Kidney Int ; 61(1 Suppl): S79-84, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11841618

ABSTRACT

Pancreatic islet transplantation (PIT) is an attractive alternative to insulin-dependent diabetes treatment but is not yet a clinical reality. The first few days after PIT are characterized by substantial pancreatic islet dysfunction and death. Apoptosis has been documented in PI after extracellular matrix removal, during culture time, after exposure to proinflammatory cytokines, hypoxic conditions before islet revascularization, and rejection. Targeting the apoptosis pathway by adenoviral-mediated gene transfer of the anti-apoptotic Bcl-2 gene exerts a major cytoprotective effect on isolated macaque pancreatic islets. Bcl-2 transfection ex vivo protects islets from apoptosis induced by disruption of the islet extracellular matrix during pancreatic digestion. Additionally, over-expression of Bcl-2 confers long-term, stable protection and maintenance of functional islet mass after transplantation into diabetic SCID mice. Genetic modification of PI also reduced the islet mass required to achieve stable euglycemia. Ex vivo gene transfer of anti-apoptotic genes has potential as a therapeutic approach to both minimize loss of functional islet mass post-transplant and reduce the high islet requirement currently needed for successful stable reversal of insulin-dependent diabetes [1, 2].


Subject(s)
Cytoprotection , Genetic Therapy , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Animals , Gene Transfer Techniques , Macaca mulatta , Male , Postoperative Care , Preoperative Care , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...