Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 173(24): 3443-3453, 2016 12.
Article in English | MEDLINE | ID: mdl-27515367

ABSTRACT

BACKGROUND AND PURPOSE: Liraglutide improves the metabolic control of diabetic animals after islet transplantation. However, the mechanisms underlying this effect remain unknown. The objective of this study was to evaluate the anti-inflammatory and anti-oxidative properties of liraglutide on rat pancreatic islets in vitro and in vivo. EXPERIMENTAL APPROACH: In vitro, rat islets were incubated with 10 µmol·L-1 liraglutide for 12 and 24 h. Islet viability functionality was assessed. The anti-inflammatory properties of liraglutide were evaluated by measuring CCL2, IL-6 and IL-10 secretion and macrophage chemotaxis. The anti-oxidative effect of liraglutide was evaluated by measuring intracellular ROS and the total anti-oxidative capacity. In vivo, 1000 islets were cultured for 24 h with or without liraglutide and then transplanted into the liver of streptozotocin-induced diabetic Lewis rats with or without injections of liraglutide. Effects of liraglutide on metabolic control were evaluated for 1 month. KEY RESULTS: Islet viability and function were preserved and enhanced with liraglutide treatment. Liraglutide decreased CCL2 and IL-6 secretion and macrophage activation after 12 h of culture, while IL-10 secretion was unchanged. However, intracellular levels of ROS were increased with liraglutide treatment at 12 h. This result was correlated with an increase of anti-oxidative capacity. In vivo, liraglutide decreased macrophage infiltration and reduced fasting blood glucose in transplanted rats. CONCLUSIONS AND IMPLICATIONS: The beneficial effects of liraglutide on pancreatic islets appear to be linked to its anti-inflammatory and anti-oxidative properties. These findings indicated that analogues of glucagon-like peptide-1 could be used to improve graft survival.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation/drug therapy , Islets of Langerhans Transplantation , Islets of Langerhans/drug effects , Liraglutide/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Liraglutide/administration & dosage , Male , Oxidative Stress/drug effects , Rats , Rats, Inbred Lew , Rats, Wistar , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
2.
J Diabetes Res ; 2016: 8310516, 2016.
Article in English | MEDLINE | ID: mdl-27504460

ABSTRACT

Exogenous insulin is the only treatment available for type 1 diabetic patients and is mostly administered by subcutaneous (SC) injection in a basal and bolus scheme using insulin pens (injection) or pumps (preimplanted SC catheter). Some divergence exists between these two modes of administration, since pumps provide better glycaemic control compared to injections in humans. The aim of this study was to compare the impacts of two modes of insulin administration (single injections of long-acting insulin or pump delivery of rapid-acting insulin) at the same dosage (4 IU/200 g/day) on rat metabolism and tissues. The rat weight and blood glucose levels were measured periodically after treatment. Immunostaining for signs of oxidative stress and for macrophages was performed on the liver and omental tissues. The continuous insulin delivery by pumps restored normoglycaemia, which induced the reduction of both reactive oxygen species and macrophage infiltration into the liver and omentum. Injections controlled the glucose levels for only a short period of time and therefore tissue stress and inflammation were elevated. In conclusion, the insulin administration mode has a crucial impact on rat metabolic parameters, which has to be taken into account when studies are designed.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/administration & dosage , Insulin Glargine/administration & dosage , Insulin/administration & dosage , Liver/drug effects , Macrophages/drug effects , Omentum/drug effects , Animals , Blood Glucose/metabolism , Body Weight , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin Glargine/pharmacology , Insulin Infusion Systems , Liver/cytology , Macrophages/cytology , Male , Omentum/cytology , Rats , Rats, Inbred Lew , Reactive Oxygen Species/metabolism
3.
J Diabetes Res ; 2016: 3615286, 2016.
Article in English | MEDLINE | ID: mdl-26824040

ABSTRACT

In bioartificial pancreases (BP), the number of islets needed to restore normoglycaemia in the diabetic patient is critical. However, the confinement of a high quantity of islets in a limited space may impact islet survival, particularly in regard to the low oxygen partial pressure (PO2) in such environments. The aim of the present study was to evaluate the impact of islet number in a confined space under hypoxia on cell survival. Rat islets were seeded at three different concentrations (150, 300, and 600 Islet Equivalents (IEQ)/cm(2)) and cultured in normal atmospheric pressure (160 mmHg) as well as hypoxic conditions (15 mmHg) for 24 hours. Cell viability, function, hypoxia-induced changes in gene expression, and cytokine secretion were then assessed. Notably, hypoxia appeared to induce a decrease in viability and increasing islet density exacerbated the observed increase in cellular apoptosis as well as the loss of function. These changes were also associated with an increase in inflammatory gene transcription. Taken together, these data indicate that when a high number of islets are confined to a small space under hypoxia, cell viability and function are significantly impacted. Thus, in order to improve islet survival in this environment during transplantation, oxygenation is of critical importance.


Subject(s)
Islets of Langerhans/metabolism , Oxygen/metabolism , Animals , Apoptosis , Atmospheric Pressure , Cell Hypoxia , Cell Survival , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation Mediators/metabolism , Islets of Langerhans/pathology , Male , Rats, Wistar , Time Factors , Tissue Culture Techniques
4.
Int J Pharm ; 491(1-2): 402-8, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26049075

ABSTRACT

Insulin delivery by oral route would be ideal, but has no effect, due to the harsh conditions of the gastrointestinal tract. Protection of insulin using encapsulation in self-assembled particles is a promising approach. However, the lack of stability of this kind of particles in biological environments induces a low bioavailability of encapsulated insulin after oral administration. The objective of this work was to evaluate the effect of two stabilisation strategies alone or combined, freeze-drying and cross-linking, on insulin-loaded chitosan NPs, and to determine their bioefficiency in vitro and in vivo. NPs were prepared by complex coacervation between insulin and chitosan, stabilised either by cross linking with sodium tripolyphosphate solution (TPP), by freeze-drying or both treatments. In vitro bioefficiency NP uptake was evaluated by flow cytometry on epithelial models (Caco-2/RevHT29MTX (mucus secreting cells)). In vivo, NPs were injected via catheter in the peritoneum or duodenum on insulinopenic rats. Freeze-drying increased in size and charge (+15% vs control 412 ± 7 nm; + 36 ± 0.3 mV) in comparison with cross linking which decreased NP size (-25%) without impacting the NP charge. When combined the consecutive treatments reduced NPs size and increased charges as compared to standard level. Freeze drying is necessary to prevent the destruction of NP in intestinal environment in comparison with no freeze dryed one where 60% of NP were destroyed after 2h. Additionally freeze drying combined with cross linking treatments improved bioefficiency of NP with uptake in cell increased when mucus is present. Combination of both treatment showed a protection of insulin in vivo, with a reduction of glycemia when NPs were administrated. This work showed that the combination of freeze drying and cross linking treatment is necessary to stabilize (freeze-drying) and increase bioefficiency (cross-linking) of self assembled NP in the delivery of insulin in vitro and in vivo.


Subject(s)
Chitosan/chemistry , Insulin/administration & dosage , Nanoparticles/chemistry , Animals , Blood Glucose/metabolism , Caco-2 Cells , Chemistry, Pharmaceutical , Cross-Linking Reagents , Drug Delivery Systems , Drug Design , Drug Stability , Excipients , Freeze Drying , Humans , Insulin/chemistry , Insulin/pharmacology , Male , Mucus/metabolism , Rats , Rats, Wistar
5.
Biomaterials ; 52: 180-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818424

ABSTRACT

Disruption of the pancreatic islet environment combined with the decrease in oxygen supply that occurs during isolation leads to poor islet survival. The aim of this study was to validate the benefit of using a plasma-based scaffold supplemented with perfluorodecalin to improve islet transplantation outcome. Rat islets were cultured in three conditions: i) control group, ii) plasma based-matrix (P-matrix), and iii) P-matrix supplemented with emulsified perfluorodecalin. After 24 h culture, matrix/cell contacts (Integrinß1, p-FAK/FAK, p-Akt/Akt), survival (caspase 3, TUNEL, FDA/PI), function, and HIF-1α translocation were assessed. Afterwards, P-matrices were dissolved and the islets were intraportally transplanted. Graft function was monitored for 31 days with glycaemia and C-peptide follow up. Inflammation was assessed by histology (macrophage and granulocyte staining) and thrombin/anti-thrombin complex measurement. Islet survival correlated with an increase in integrin, FAK, and Akt activation in P-matrices and function was maintained. Perfluorodecalin supplementation decreased translocation of HIF-1α in the nucleus and post-transplantation islet structure was better preserved in P-matrices, but a quicker activation of IBMIR resulted in early loss of graft function. "Oxygenating" P-matrices provided a real benefit to islet survival and resistance in vivo. However, intraportal transplantation is not suitable for this kind of culture due to IBMIR; thus, alternative sites must be explored.


Subject(s)
Cell Culture Techniques/methods , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Oxygen/metabolism , Animals , Cell Hypoxia , Cell Survival , Cells, Cultured , Fluorocarbons/metabolism , Graft Survival , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , Male , Rats , Rats, Inbred Lew , Rats, Wistar
6.
Cell Transplant ; 21(4): 657-69, 2012.
Article in English | MEDLINE | ID: mdl-21944582

ABSTRACT

As oxygen carriers, perfluorocarbon emulsions might be useful to decrease hypoxia of pancreatic islets before transplantation. However, their hydrophobicity prevents their homogenisation in culture medium. To increase the surface of contact between islets and Perfluorooctyl bromide (PFOB), and consequently oxygen delivery, we tested effect of a PFOB emulsion in culture medium on ß-cell lines and rat pancreatic islets. RINm5F ß-cell line or pancreatic rat islets were incubated for 3 days in the presence of PFOB emulsion in media (3.5% w/v). Preoxygenation of the medium was performed before culture. Cell viability was assessed by apoptotic markers (Bax and Bcl-2) and by staining (fluoresceine diacetate and propidium iodide). ß-Cell functionality was determined by insulin release during a glucose stimulation test and. Hypoxia markers, HIF-1α and VEGF, were studied at days 1 and 3 using RT-PCR, Western blotting, and ELISA. PFOB emulsions preserved viability and functionality of RINm5F cells with a decrease of HIF-1α and VEGF expression. Islets viability was preserved during 3 days of culture. Secretion of VEGF was higher in untreated control (0.09 ± 0.041 µg VEGF/mg total protein) than in PFOB emulsion incubated islets (0.02 ± 0.19 µg VEGF/mg total protein, n = 4, p < 0.05) at day 1. At day 3, VEGF secretion was increased as compared to day 1 in control (0.23 ± 0.04 µg VEGF/mg total protein) but it was imbalance by the presence of PFOB emulsion (0.09 ± 0.03 µg VEGF/mg total protein, n = 5, p < 0.05). While insulin secretion was maintained in response to a glucose stimulation test until day 3 when islets were incubated in the presence of PFOB emulsion preoxygenated (0.81 ± 0.16 at day 1 vs. 0.75 ± 0.24 at day 3), the ability to secrete insulin in the presence of high glucose concentration was lost in islets controls (0.51 ± 0.18 at day 1 vs. 0.21 ± 0.13 at day 3). Atmospheric oxygen delivery by PFOB emulsion might be sufficient to decrease islets hypoxia. However, to improve islets functionality, overoxygenation is needed. Finally, maintenance of islet viability and functionality for several days after isolation could improve the outcome of islets transplantation.


Subject(s)
Cell Hypoxia/drug effects , Fluorocarbons/pharmacology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Animals , Cell Survival/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Rats , Vascular Endothelial Growth Factor A/metabolism
7.
Transplant Proc ; 43(9): 3192-4, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22099754

ABSTRACT

The instant blood-mediated inflammatory reaction (IBMIR) leads to massive destruction of transplanted islets. Islet isolation and time of culture may elicit the release of potent activators of Toll-like receptors (TLRs) signaling pathways during IBMIR. This work sought to evaluate the role of TLR signaling pathways to mediate inflammatory reactions. Isolated rat pancreatic islets were cultured for 12, 24, or 48 hours. Their viability was assessed by fluorescein diacetate/propidium iodide and their functionality, by glucose stimulation tests. Endotoxin levels were quantified using the Limulus Amebocyte Lysate assays. After RNA extraction and reverse transcription, we performed polymerase chain reaction (PCR) arrays. Samples obtained immediately after isolation were defined as controls. Eighty-four genes belonging to the TLR signaling pathways, were compared with control samples. After culture, islets were viable and functional with low endotoxin levels (< 0.1 endotoxin units/mL) showed TLR activation not due to exogenous contamination. Analysis of PCR arrays highlighted significant up-regulation of TLR-2. After 24 hours of culture, TLR-2 was up-regulated to 6.8 ± 0.6-fold (P < .001) compared with controls but decreased to 4.3 ± 1.4-fold after 48 hours. In the same way, expression of myeloid differentiation primary response gene 88 (Myd88) was significantly up-regulated (3.2 ± 0.4-fold [P < .001]) compared with controls. After 12 hours of culture, interleukin-10 gene expression was significantly up-regulated at 11.6 ± 3.7- fold (P < .05), reaching 17.5 ± 8.3 after 24 hours. Finally, the cyclo-oxygenase-2 gene expression was up-regulated to 509 ± 67.1-fold (P < .05) after 12 hours of culture. These data confirmed the implication of TLR signaling pathways in early inflammatory events.


Subject(s)
Inflammation/pathology , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Toll-Like Receptors/metabolism , Animals , Cell Survival , Gene Expression Regulation , Insulin/metabolism , Male , Models, Biological , Rats , Rats, Wistar , Signal Transduction , Toll-Like Receptor 2/biosynthesis
8.
Transplant Proc ; 43(9): 3201-4, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22099757

ABSTRACT

Early events hampering islet engraftment may relate to instant blood-mediated inflammatory reaction (IBMIR) and to insufficient islet revascularization inducing ß-cell death. We evaluated the influence of time of culture on angiogenic and inflammatory cellular mechanisms in islet loss in vitro. Rat pancreatic islets cultured for 0, 12, 24, and 48 hours were assessed for functionality using glucose stimulation tests and identification of signaling pathways using polymerase chain reaction (PCR) arrays. Islet functionality decreased significantly immediately. Index of stimulation (IS) was decreased to 2.29 ± 1.05 after 48 hours of culture versus 18.47 ± 4.84 at 0 hours (P < .001). Gene expression studies at 12 hours of culture showed significant overexpression of proinflammatory cytokines and chemokines--interleukin (IL)-6 884.22 ± 282.58 (P < .001) and Cxcl-1 448.09 ± 196.05-fold change (P < .01). Moreover, islets exhibited significant under-expression after 48 hours of genes encoding angiogenic growth factors, such as epidermal growth factor, vascular endothelial growth factor, platelet endothelial cell adhesion molecule 1, a major protein involved in angiogenesis: 0.07 ± 0.02, 0.11 ± 0.08 (P < .001), and 0.17 ± 0.15-fold change (P < .01) respectively. Moreover, tissue inhibitor of metalloproteinases 1, an inhibitor of metallopeptidase, was significantly more over-expressed, namely 54.58 ± 18.08 at 12 hours of culture versus 0.93 ± 0.15/fold change at 0 hours. This study revealed current culture conditions to be deleterious for islet engraftment, possibly due to expression of angiogenic genes and proinflamatory genes during culture.


Subject(s)
Inflammation/pathology , Islets of Langerhans/cytology , Neovascularization, Pathologic , Animals , Cell Culture Techniques/methods , Chemokine CXCL1/biosynthesis , Gene Expression Profiling , Interleukin-6/biosynthesis , Islets of Langerhans Transplantation/methods , Polymerase Chain Reaction , Proteomics/methods , Rats , Rats, Inbred Lew , Rats, Wistar , Time Factors
9.
Cell Transplant ; 20(9): 1333-42, 2011.
Article in English | MEDLINE | ID: mdl-21294962

ABSTRACT

Delayed and insufficient revascularization during islet transplantation deprives islets of oxygen and nutrients, resulting in graft failure. Vascular endothelial growth factor (VEGF) could play a critical role in islet revascularization. We aimed to develop pharmacological strategies for VEGF overexpression in pancreatic islets using the iron chelator deferoxamine (DFO), thus avoiding obstacles or safety risks associated with gene therapy. Rat pancreatic islets were infected in vivo using an adenovirus (ADE) encoding human VEGF gene (4.10(8) pfu/pancreas) or were incubated in the presence of DFO (10 µmol/L). In vitro viability, functionality, and the secretion of VEGF were evaluated in islets 1 and 3 days after treatment. Infected islets or islets incubated with DFO were transplanted into the liver of syngenic diabetic rats and the graft efficiency was estimated in vivo by measuring body weight, glycemia, C-peptide secretion, and animal survival over a period of 2 months. DFO induced transient VEGF overexpression over 3 days, whereas infection with ADE resulted in prolonged VEGF overexpression lasting 14 days; however, this was toxic and decreased islet viability and functionality. The in vivo study showed a decrease in rat deaths after the transplantation of islets treated with DFO or ADE compared with the sham and control group. ADE treatment improved body weight and C-peptide levels. Gene therapy and DFO improved metabolic control in diabetic rats after transplantation, but this effect was limited in the presence of DFO. The pharmacological approach is an interesting strategy for improving graft efficiency during transplantation, but this approach needs to be improved with drugs that are more specific.


Subject(s)
Deferoxamine/pharmacology , Islets of Langerhans Transplantation , Tissue Survival/drug effects , Vascular Endothelial Growth Factor A/metabolism , Adenoviridae Infections/pathology , Animals , Body Weight/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans/virology , Male , Rats , Rats, Inbred Lew , Rats, Wistar , Reproducibility of Results
10.
Transplant Proc ; 40(2): 473-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374106

ABSTRACT

During pancreatic islet transplantation, delayed and insufficient revascularization can deprive islets of oxygen and nutrients, resulting in cell death and early graft failure. Deferoxamine (DFO), an iron chelator, increases vascular endothelial growth factor (VEGF) expression in cells. The aim of this work was to study the effect of DFO on beta-cell and pancreatic islet viability as well as VEGF expression. beta-cell lines from rat insulinoma (Rin m5f) and primary cultures of pancreatic islets from Wistar rats were incubated with DFO (10, 100, and 1000 micromol/L). The viability was evaluated using fluorescein diacetate/propidium iodide for dying pancreatic islets and using cell titers for Rin m5f. Expression of VEGF messenger RNA (mRNA) was quantified using reverse transcriptase polymerase chain reaction (RT-PCR). Finally, VEGF secretion was determined using enzyme-linked immunosorbent assays at 1 to 3 days after treatment. The addition of 10 micromol/L of DFO preserved Rin m5F viability at 24 hours after treatment (10 micromol/L; 101.33% +/- 5.66%; n = 7). However, 100 and 1000 micromol/L of DFO induced cell death (68.92% +/- 5.83% and 65.89% +/- 5.83%, respectively; n = 4). In the same way, viability of pancreatic islets in the presence of DFO was preserved. RT-PCR analysis showed stimulation of VEGF mRNA in the presence of 10 micromol/L of DFO in islets at 3 days after culture. Finally, 10 micromol/L of DFO stimulated secretion of VEGF 7.95 +/- 0.84 versus 1.80 +/- 1.10 pg/microg total protein with 10 micromol/L of DFO in rat islets at 3 days after culture, n = 3; P < .001). The use of DFO to stimulate VEGF expression and increase islet vascularization may be a realistic approach to improve islet viability during transplantation.


Subject(s)
Deferoxamine/therapeutic use , Insulin-Secreting Cells/physiology , Islets of Langerhans Transplantation/physiology , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/genetics , Animals , Cell Survival/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/blood supply , Neovascularization, Physiologic/physiology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...