Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Cell Biochem ; 120(5): 8534-8546, 2019 May.
Article in English | MEDLINE | ID: mdl-30474134

ABSTRACT

OBJECTIVE: We investigated the effects of sodium-glucose cotransporter 2 inhibitor, empagliflozin, and α-glucosidase inhibitor, voglibose, on hepatic steatosis in an animal model of type 2 diabetes (T2DM). METHODS: Empagliflozin (OLETF-EMPA) or voglibose (OLETF-VOG) was administered to Otsuka Long-Evans Tokushima fatty (OLETF) rats once daily for 12 weeks. Control Long-Evans Tokushima Otsuka (LETO) and OLETF (OLETF-C) rats received saline. RESULTS: Blood glucose levels were significantly suppressed in OLETF-EMPA and OLETF-VOG compared with the OLETF-C group. The liver fat content was significantly higher in the OLETF-C group than in the OLETF-EMPA and OLETF-VOG. Hepatic gene expressions involved in gluconeogenesis (glucose 6-phosphatase [G6Pase], fructose-1,6-bisphosphatase [FBP1], and phosphoenolpyruvate carboxykinase [PEPCK]) and lipogenesis (acetyl-CoA carboxylase [ACC], fatty acid synthase [FAS], and sterol regulatory element-binding transcription factor 1c [SREBP-1c]) were significantly decreased in the OLETF-EMPA group compared with other OLETF groups (OLETF-C and OLETF-VOG). Sirtuin 1 (SIRT1) expression level and SIRT1 activity were markedly reduced in OLETF-C rats; however, its expression increased in the OLETF-EMPA and OLETF-VOG. AMP-activated protein kinase (AMPK) phosphorylation level was remarkably increased by empagliflozin treatment in OLETF rats compared with other OLETF groups. Long-term empagliflozin and voglibose treatment reduced hepatic steatosis with suppression of gluconeogenesis and lipogenesis pathway in OLETF rats. CONCLUSION: We suggest that this metabolic improvement might be related to SIRT1 and AMPK pathway in T2DM. But empagliflozin is thought to have more advantage to prevent hepatic steatosis than voglibose in T2DM.

2.
J Cell Biochem ; 117(1): 239-46, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26105159

ABSTRACT

The activation of AMP-activated protein kinase (AMPK) is known to repress the expression of the insulin gene and glucose-stimulated insulin secretion (GSIS). However, the mechanisms by which this occurs, as well as the effects of AMPK activation on glucolipotoxicity-induced ß-cell dysfunction, have not been elucidated. To investigate the effects of 5-amino-4-imidazolecarboxamide ribonucleotide (AICAR) and peroxisome proliferator-activated receptorγ-coactivator-1α (PGC-1α) on ß-cell-specific genes under glucolipotoxic conditions, we performed real-time PCR and measured insulin secretion by primary islets. To study these effects in vivo, we administered AICAR for 10 days (1 mg/g body weight) to 90% pancreatectomized hyperglycemic mice. The exposure of isolated rat and human islets to glucolipotoxic conditions and the overexpression of PGC-1α suppressed insulin and NEUROD1 mRNA expression. However, the expression of these genes was preserved by AICAR treatment and by PGC-1α inhibition. Exposure of isolated islets to glucolipotoxic conditions for 3 days decreased GSIS, which was also well maintained by AICAR treatment and by PGC-1α inhibition. The administration of AICAR to 90% pancreatectomized hyperglycemic mice improved glucose tolerance and insulin secretion. These results indicate that treatment of islets with an AMPK agonist under glucolipotoxic conditions protects against glucolipotoxicity-induced ß-cell dysfunction. A better understanding of the functions of molecules such as PGC-1α and AMPK, which play key roles in intracellular fuel regulation, could herald a new era for the treatment of patients with type 2 diabetes mellitus by providing protection against glucolipotoxicity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/pharmacology , Insulin/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology
3.
J Korean Med Sci ; 30(7): 991-4, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26130966

ABSTRACT

Pancreatic islet transplantation is a physiologically advantageous and minimally invasive procedure for the treatment of type 1 diabetes mellitus. Here, we describe the first reported case of successful allogeneic islet transplantation alone, using single-donor, marginal-dose islets in a Korean patient. A 59-yr-old patient with type 1 diabetes mellitus, who suffered from recurrent severe hypoglycemia, received 4,163 islet equivalents/kg from a single brain-death donor. Isolated islets were infused intraportally without any complications. The immunosuppressive regimen was based on the Edmonton protocol, but the maintenance dosage was reduced because of mucositis and leukopenia. Although insulin independence was not achieved, the patient showed stabilized blood glucose concentration, reduced insulin dosage and reversal of hypoglycemic unawareness, even with marginal dose of islets and reduced immunosuppressant. Islet transplantation may successfully improve endogenous insulin production and glycemic stability in subjects with type 1 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Hypoglycemia/surgery , Islets of Langerhans Transplantation/methods , Islets of Langerhans/surgery , Blood Glucose/analysis , Female , Humans , Immunosuppression Therapy/methods , Immunosuppressive Agents/therapeutic use , Islets of Langerhans/physiology , Middle Aged , Republic of Korea , Tissue Donors
4.
Metabolism ; 153: 155795, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38253121

ABSTRACT

BACKGROUND AND AIMS: The incidence of statin-induced new-onset diabetes (NOD) is increasing but its underlying mechanisms remain unclear. We aimed to investigate the effects of various doses of atorvastatin (ATO)-induced autophagy on the development of NOD. METHODS AND RESULTS: The isolated rat islets and MIN6 cells-treated with ATO, exhibited impaired glucose-stimulated insulin secretion, reduced insulin content, and induced apoptosis. Additionally, autophagy was induced at all doses (in vitro: 5, 10, 20 µM; in vivo: 10, 15, 20 mg/kg) in ATO-treated MIN6 cells or western diet (WD)-fed mice. In contrast to normal glucose-tolerant mice administered a low-dose (10 mg/kg) ATO, those treated with high-doses (15 or 20 mg/kg) exhibited impaired glucose tolerance. Furthermore, high-dose ATO-treated mice showed decreased ß-cell mass and increased apoptosis compared to that of vehicle-treated mice. We also observed that the number of vesicophagous cells in the pancreas of 20 mg/kg ATO-treated WD-fed mice was higher than in vehicle-treated WD-fed mice. Inhibiting autophagy using 3-methyladenine (3-MA) and siAtg5 improved glucose tolerance in vivo and in vitro by preventing apoptotic ß-cell death and restoring insulin granules. CONCLUSION: These results indicate that high doses of ATO induced hyperactivated autophagy in pancreatic cells, leading to impaired insulin storage, decreased cell viability, and reduced functional cell mass, ultimately resulting in NOD development.


Subject(s)
Diabetes Mellitus , Diet, Western , Mice , Rats , Animals , Atorvastatin/pharmacology , Diet, Western/adverse effects , Glucose/pharmacology , Insulin/pharmacology , Autophagy
5.
J Diabetes Investig ; 15(6): 693-703, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38634411

ABSTRACT

AIMS/INTRODUCTION: To investigate the long-term efficacy of various encapsulated xenogeneic islet transplantation, and to explore the impact of different donor porcine genetic traits on islet transplantation outcomes. MATERIALS AND METHODS: Donor porcine islets were obtained from wild-type, α1,3-galactosyltransferase knockout (GTKO) and GTKO with overexpression of membrane cofactor protein genotype. Naked, alginate, alginate-chitosan (AC), alginate-perfluorodecalin (A-PFD) and AC-perfluorodecalin (AC-PFD) encapsulated porcine islets were transplanted into diabetic mice. RESULTS: In vitro assessments showed no differences in the viability and function of islets across encapsulation types and donor porcine islet genotypes. Xenogeneic encapsulated islet transplantation with AC-PFD capsules showed the most favorable long-term outcomes, maintaining normal blood glucose levels for 180 days. A-PFD capsules showed comparable results to AC-PFD capsules, followed by AC capsules and alginate capsules. Conversely, blood glucose levels in naked islet transplantation increased to >300 mg/dL within a week after transplantation. Naked islet transplantation outcomes showed no improvement based on donor islet genotype. However, alginate or AC capsules showed delayed increases in blood glucose levels for GTKO and GTKO with overexpression of membrane cofactor protein porcine islets compared with wild-type porcine islets. CONCLUSION: The AC-PFD capsule, designed to ameliorate both hypoxia and inflammation, showed the highest long-term efficacy in xenogeneic islet transplantation. Genetic modifications of porcine islets with GTKO or GTKO with overexpression of membrane cofactor protein did not influence naked islet transplantation outcomes, but did delay graft failure when encapsulated.


Subject(s)
Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Transplantation, Heterologous , Islets of Langerhans Transplantation/methods , Animals , Swine , Mice , Transplantation, Heterologous/methods , Diabetes Mellitus, Experimental/therapy , Alginates , Galactosyltransferases/genetics , Graft Survival , Islets of Langerhans , Blood Glucose/analysis , Male , Genotype , Tissue Donors
6.
Diabetes Metab J ; 46(5): 677-688, 2022 09.
Article in English | MEDLINE | ID: mdl-35124687

ABSTRACT

BACKGROUND: Neonatal porcine pancreatic cell clusters (NPCCs) have been proposed as an alternative source of ß cells for islet transplantation because of their low cost and growth potential after transplantation. However, the delayed glucose lowering effect due to the immaturity of NPCCs and immunologic rejection remain as a barrier to NPCC's clinical application. Here, we demonstrate accelerated differentiation and immune-tolerant NPCCs by in vitro chemical treatment and microencapsulation. METHODS: NPCCs isolated from 3-day-old piglets were cultured in F-10 media and then microencapsulated with alginate on day 5. Differentiation of NPCCs is facilitated by media supplemented with activin receptor-like kinase 5 inhibitor II, triiodothyronine and exendin-4 for 2 weeks. Marginal number of microencapsulated NPCCs to cure diabetes with and without differentiation were transplanted into diabetic mice and observed for 8 weeks. RESULTS: The proportion of insulin-positive cells and insulin mRNA levels of NPCCs were significantly increased in vitro in the differentiated group compared with the undifferentiated group. Blood glucose levels decreased eventually after transplantation of microencapsulated NPCCs in diabetic mice and normalized after 7 weeks in the differentiated group. In addition, the differentiated group showed nearly normal glucose tolerance at 8 weeks after transplantation. In contrast, neither blood glucose levels nor glucose tolerance were improved in the undifferentiated group. Retrieved graft in the differentiated group showed greater insulin response to high glucose compared with the undifferentiated group. CONCLUSION: in vitro differentiation of microencapsulated immature NPCCs increased the proportion of insulin-positive cells and improved transplant efficacy in diabetic mice without immune rejection.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Islets of Langerhans , Alginates/metabolism , Alginates/pharmacology , Animals , Animals, Newborn , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/surgery , Exenatide/pharmacology , Insulin/metabolism , Mice , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Swine , Transplantation, Heterologous , Triiodothyronine/metabolism , Triiodothyronine/pharmacology
7.
J Drug Target ; 29(1): 88-98, 2021 01.
Article in English | MEDLINE | ID: mdl-32749162

ABSTRACT

Islet transplantation (ITx) is being developed as a treatment for type 1 diabetes mellitus, but hypoxic damage to transplanted islet grafts is an important factor affecting successful transplantation. To investigate the role of sirtuin-1 (SIRT1) under hypoxic injury in INS-1 cells, one type of pancreatic ß-cell lines, we used SRT1720 and GW4064 for SIRT1 activation. The small interfering RNA SIRT1 (si-SIRT1) was used to suppress SIRT1 gene expression. We measured cell viability, apoptosis, and the levels of inflammatory cytokines, including tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6), and reactive oxygen species (ROS), under hypoxic conditions. Real-time PCR and Western blot analysis were performed. Cell viability was significantly reduced to 71% and 40% after 4 and 6 h of hypoxic conditions, respectively. Apoptosis increased significantly 2.8-fold and 5.3-fold after 4 and 6 h of hypoxia, respectively. SIRT1 expression was significantly reduced at the mRNA and protein levels during hypoxia. Hypoxic damage significantly increased the TNF-α, IL-6 and ROS levels in INS-1 cells. However, the reduced cell viability and increased inflammatory cytokines from hypoxic damage were ameliorated by SIRT1 activation in INS-1 cells. These results suggest that SIRT1 is a potential target for the protection of pancreatic ß-cells against hypoxic damage during ITx.


Subject(s)
Cytokines/metabolism , Insulin-Secreting Cells/metabolism , Oxidative Stress/physiology , Sirtuin 1/metabolism , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cytokines/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Insulin-Secreting Cells/drug effects , Oxidative Stress/drug effects , Rats
8.
Endocrinol Metab (Seoul) ; 36(1): 146-156, 2021 02.
Article in English | MEDLINE | ID: mdl-33677936

ABSTRACT

BACKGROUND: The microencapsulation is an ideal solution to overcome immune rejection without immunosuppressive treatment. Poor biocompatibility and small molecular antigens secreted from encapsulated islets induce fibrosis infiltration. Therefore, the aims of this study were to improve the biocompatibility of microcapsules by dexamethasone coating and to verify its effect after xenogeneic transplantation in a streptozotocin-induced diabetes mice. METHODS: Dexamethasone 21-phosphate (Dexa) was dissolved in 1% chitosan and was cross-linked with the alginate microcapsule surface. Insulin secretion and viability assays were performed 14 days after microencapsulation. Dexa-containing chitosan-coated alginate (Dexa-chitosan) or alginate microencapsulated porcine islets were transplanted into diabetic mice. The fibrosis infiltration score was calculated from the harvested microcapsules. The harvested microcapsules were stained with trichrome and for insulin and macrophages. RESULTS: No significant differences in glucose-stimulated insulin secretion and islet viability were noted among naked, alginate, and Dexa-chitosan microencapsulated islets. After transplantation of microencapsulated porcine islets, nonfasting blood glucose were normalized in both the Dexa-chitosan and alginate groups until 231 days. The average glucose after transplantation were lower in the Dexa-chitosan group than the alginate group. Pericapsular fibrosis and inflammatory cell infiltration of microcapsules were significantly reduced in Dexa-chitosan compared with alginate microcapsules. Dithizone and insulin were positive in Dexa-chitosan capsules. Although fibrosis and macrophage infiltration was noted on the surface, some alginate microcapsules were stained with insulin. CONCLUSION: Dexa coating on microcapsules significantly suppressed the fibrotic reaction on the capsule surface after transplantation of xenogenic islets containing microcapsules without any harmful effects on the function and survival of the islets.


Subject(s)
Chitosan , Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Islets of Langerhans , Alginates/metabolism , Alginates/pharmacology , Animals , Capsules/metabolism , Capsules/pharmacology , Chitosan/metabolism , Chitosan/pharmacology , Dexamethasone/metabolism , Dexamethasone/pharmacology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/surgery , Fibrosis , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Swine
9.
Stem Cell Res ; 48: 101958, 2020 10.
Article in English | MEDLINE | ID: mdl-32882526

ABSTRACT

For alternative sources of ß cells, patient-specific induced pluripotent stem cells (iPSCs) could be promising, as cells derived from the "self" allow autologous transplantation. However, only a few studies have investigated insulin-producing cells (IPCs) using iPSCs of patients with type 1 diabetes (T1D). In this study, we generated IPCs using iPSCs derived from patients with T1D and type 2 diabetes (T2D) and compared them with IPCs from a non-diabetic (ND) individual. To facilitate differentiation of human iPSCs into IPCs, we induced PDX-1 gene expression using Ad-PDX-1/VP16. IPCs derived from T1D- and T2D-specific iPSCs expressed islet-specific markers such as Pdx-1, MafA, Beta2/NeuroD, and insulin, similar to IPCs derived from ND-specific iPSCs. In addition, IPCs derived from T1D- and T2D-specific iPSCs showed comparable glucose-stimulated insulin secretion as IPCs derived from ND-specific iPSCs. These results suggest the potential for autologous transplantation using patient-specific iPSCs in patients with T1D and T2D. This study was clinically significant because the majority of people with diabetes have T2D and insulin secretion declines over time in T2D. To the best of our knowledge, this is the first study to generate and simultaneously compare IPCs from ND-, T1D-, and T2D-specific iPSCs.


Subject(s)
Diabetes Mellitus, Type 2 , Induced Pluripotent Stem Cells , Insulin-Secreting Cells , Cell Differentiation , Humans , Insulin
10.
Islets ; 10(1): 25-39, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29333922

ABSTRACT

We evaluated the effect of resveratrol (RSV) on graft survival after islet transplantation (ITx) in diabetic mice. Isolated islets from Balb/c mice (200 IEQ) were transplanted under the kidney capsule of diabetic Balb/c mice. Vehicle or RSV (200 mg/kg/day, orally) was given for 14 days after ITx. Two more control groups [STZ-treated (No-ITx-Control) and STZ+RSV-treated (No-ITx-RSV) mice without ITx] were added. Glucose tolerance tests (GTT) was performed at 14 days after ITx. In vitro, isolated islets pretreated with vehicle or RSV (1 µM) were incubated in a hypoxic chamber (O2 1%, 1hr). Some of the ITx was performed in mouse insulin 1 gene promoter-green fluorescent protein (MIP-GFP) transgenic mice and analyzed using an in vivo imaging system. After 14 days of ITx, 2-hr glucose levels on GTT in the RSV-treated group were significantly lower than those of other control groups. But the glucose status was not improved in No-ITx mice with RSV. At day 3, the percentage of Ki-67/insulin co-stained cells in islet graft was significantly increased in the RSV-ITx group. Immunostaining with anti-insulin and anti-BS-1 antibodies revealed significantly higher insulin-stained area and vascular density in RSV-treated islet grafts. The mean vessel volume per islet graft measured by in vivo imaging was significantly higher in the RSV-treated group at day 3. In isolated islets cultured in hypoxic conditions, the cell death rate and oxidative stress were significantly attenuated with RSV pretreatment. Hypoxic treatment for isolated islets decreased the expression of SIRT-1 mRNA, and this attenuation was recovered by RSV pretreatment. Our data suggest that RSV treatment improved glycemic control, beta-cell proliferation, reduced oxidative stress, and enhanced islet revascularization and the outcome of ITx in diabetic mice.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Graft Survival/drug effects , Islets of Langerhans Transplantation , Islets of Langerhans/blood supply , Islets of Langerhans/drug effects , Resveratrol/therapeutic use , Animals , Cells, Cultured , Chemotherapy, Adjuvant , Combined Modality Therapy , Diabetes Mellitus, Experimental/chemically induced , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neovascularization, Physiologic , Streptozocin
11.
PLoS One ; 11(12): e0163187, 2016.
Article in English | MEDLINE | ID: mdl-27977690

ABSTRACT

Pancreatic stellate cells (PSCs) play a major role to fibrotic islet destruction observed in diabetic patients and animal model of diabetes. Exendin-4 (Ex-4) is a potent insulinotropic agent and has been approved for the treatment of type 2 diabetes. However, there have been no reports demonstrating the effects of Ex-4 on pancreatic islet fibrosis. In this study, Ex-4 treatment clearly attenuated fibrotic islet destruction and improved glucose tolerance and islet survival. GLP-1 receptor expression was upregulated during activation and proliferation of PSCs by hyperglycemia. The activation of PKA pathway by Ex-4 plays a role in ROS production and angiotensin II (Ang II) production. Exposure to high glucose stimulated ERK activation and Ang II-TGF- ß1 production in PSCs. Interestingly, Ex-4 significantly reduced Ang II and TGF-ß1 production by inhibition of ROS production but not ERK phosphorylation. Ex-4 may be useful not only as an anti-diabetic agent but also as an anti-fibrotic agent in type 2 diabetes due to its ability to inhibit PSC activation and proliferation and improve islet fibrosis in OLETF rats.


Subject(s)
Cell Survival/drug effects , Glucose/pharmacology , Pancreatic Stellate Cells/drug effects , Peptides/pharmacology , Reactive Oxygen Species/metabolism , Venoms/pharmacology , Angiotensin II/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Exenatide , Fibrosis/chemically induced , Fibrosis/metabolism , Fibrosis/pathology , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Rats , Rats, Inbred OLETF , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
12.
Transplantation ; 100(2): 334-43, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26479281

ABSTRACT

BACKGROUND: Clinical application of encapsulated islet transplantation is hindered by low biocompatibility of capsules leading to pericapsular fibrosis and decreased islet viability. To improve biocompatibility, we designed a novel chitosan-coated alginate capsules and compared them to uncoated alginate capsules. METHODS: Alginate capsules were formed by crosslinking with BaCl2, then they were suspended in chitosan solution for 10 minutes at pH 4.5. Xenogeneic islet transplantation, using encapsulated porcine islets in 1,3-galactosyltransferase knockout mice, and allogeneic islet transplantation, using encapsulated canine islets in beagles, were performed without immunosuppressants. RESULTS: The chitosan-alginate capsules showed similar pore size, islet viability, and insulin secretory function compared to alginate capsules, in vitro. Xenogeneic transplantation of chitosan-alginate capsules demonstrated a trend toward superior graft survival (P = 0.07) with significantly less pericapsular fibrosis (cell adhesion score: 3.77 ± 0.41 vs 8.08 ± 0.05; P < 0.001) compared to that of alginate capsules up to 1 year after transplantation. Allogeneic transplantation of chitosan-alginate capsules normalized the blood glucose level up to 1 year with little evidence of pericapsular fibrotic overgrowth on graft explantation. CONCLUSIONS: The efficacy and biocompatibility of chitosan-alginate capsules were demonstrated in xenogeneic and allogeneic islet transplantations using small and large animal models of diabetes. This capsule might be a potential candidate applicable in the treatment of type 1 diabetes mellitus patients, and further studies in nonhuman primates are required.


Subject(s)
Alginates/chemistry , Chitosan/chemistry , Coated Materials, Biocompatible , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation/methods , Islets of Langerhans/surgery , Animals , Barium Compounds/chemistry , Blood Glucose/metabolism , Cell Adhesion , Chlorides/chemistry , Cross-Linking Reagents/chemistry , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/pathology , Dogs , Female , Fibrosis , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Glucuronic Acid/chemistry , Graft Survival , Heterografts , Hexuronic Acids/chemistry , Insulin/blood , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/adverse effects , Male , Mice , Mice, Knockout , Porosity , RAW 264.7 Cells , Swine , Swine, Miniature , Time Factors , Transplantation, Homologous
13.
Transplantation ; 97(3): 273-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24448589

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor gamma-coactivator-1α (PGC-1α) has recently been implicated as a crucial factor in the glucocorticoid-suppressed expansion and transdifferentiation of porcine neonatal pancreatic cell clusters (NPCCs). However, the molecular mechanism has not been clarified. METHODS: We investigated whether the suppression of PGC-1α expression protects against ß-cell dysfunction induced by dexamethasone (Dx) treatment in vitro and in vivo and determined the mechanism of action of PGC-1α in porcine NPCCs. RESULTS: The reduction in Pdx-1 gene expression caused by either Dx treatment or PGC-1α overexpression was normalized by siPGC-1α. Nuclear FOXO1 and cytoplasmic Pdx-1 were detected after Dx treatment. However, FOXO1 was observed in the cytoplasm, and Pdx-1 was observed in the nucleus after siPGC-1α. Suppression of PGC-1α by siPGC-1α improved the Dx-induced repression of insulin secretion and insulin content. In vivo studies showed that the glucose level in the Ad-siPGC-1α-infected group was significantly lower than that in the Dx-treated group. Insulin expression in the graft tissue disappeared in the Dx-injected group. However, the siPGC-1α- and Dx-treated group showed increased insulin expression and an increase in graft mass, ß-cell mass, and ß-cell % in the graft. Conversely, the Dx-induced ductal cystic area was markedly reduced in the siPGC-1α- and Dx-treated group. CONCLUSIONS: Our results suggest that the transdifferentiation of porcine NPCCs into ß cells is influenced by the duration of the Dx treatment, which might result from the suppression of key pancreatic transcription factors. PGC-1α is an attractive target for modulating the deleterious effects of glucocorticoids on pancreatic stem cells.


Subject(s)
Glucocorticoids/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Pancreas Transplantation , Pancreas/cytology , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Cell Culture Techniques , Cell Differentiation , Cell Nucleus/metabolism , Cell Transdifferentiation , Cells, Cultured , Dexamethasone/chemistry , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreas/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Stem Cells/cytology , Swine
14.
Diabetes Metab J ; 37(6): 475-83, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24404519

ABSTRACT

BACKGROUND: We aimed to quantify stress-induced hyperglycemia and differentiate the glucose response between normal animals and those with diabetes. We also examined the pattern in glucose fluctuation induced by stress according to type of diabetes. METHODS: To load psychological stress on animal models, we used a predator stress model by exposing rats to a cat for 60 minutes and measured glucose level from the beginning to the end of the test to monitor glucose fluctuation. We induced type 1 diabetes model (T1D) for ten Sprague-Dawley rats using streptozotocin and used five Otsuka Long-Evans Tokushima Fatty rats as obese type 2 diabetes model (OT2D) and 10 Goto-Kakizaki rats as nonobese type 2 diabetes model (NOT2D). We performed the stress loading test in both the normal and diabetic states and compared patterns of glucose fluctuation among the three models. We classified the pattern of glucose fluctuation into A, B, and C types according to speed of change in glucose level. RESULTS: Increase in glucose, total amount of hyperglycemic exposure, time of stress-induced hyperglycemia, and speed of glucose increase were significantly increased in all models compared to the normal state. While the early increase in glucose after exposure to stress was higher in T1D and NOT2D, it was slower in OT2D. The rate of speed of the decrease in glucose level was highest in NOT2D and lowest in OT2D. CONCLUSION: The diabetic state was more vulnerable to stress compared to the normal state in all models, and the pattern of glucose fluctuation differed among the three types of diabetes. The study provides basic evidence for stress-induced hyperglycemia patterns and characteristics used for the management of diabetes patients.

15.
Endocrinology ; 154(8): 2626-39, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23698718

ABSTRACT

Growing evidence suggests that advanced glycation end-products (AGEs) are cytotoxic to pancreatic ß-cells. The aims of this study were to investigate whether glycated albumin (GA), an early precursor of AGEs, would induce dysfunction in pancreatic ß-cells and to determine which kinds of cellular mechanisms are activated in GA-induced ß-cell apoptosis. Decreased viability and increased apoptosis were induced in INS-1 cells treated with 2.5 mg/mL GA under 16.7mM high-glucose conditions. Insulin content and glucose-stimulated secretion from isolated rat islets were reduced in 2.5 mg/mL GA-treated cells. In response to 2.5 mg/mL GA in INS-1 cells, autophagy induction and flux decreased as assessed by green fluorescent protein-microtubule-associated protein 1 light chain 3 dots, microtubule-associated protein 1 light chain 3-II conversion, and SQSTM1/p62 in the presence and absence of bafilomycin A1. Accumulated SQSTM1/p62 through deficient autophagy activated the nuclear factor-κB (p65)-inducible nitric oxide synthase-caspase-3 cascade, which was restored by treatment with small interfering RNA against p62. Small interfering RNA treatment against autophagy-related protein 5 significantly inhibited the autophagy machinery resulting in a significant increase in iNOS-cleaved caspase-3 expression. Treatment with 500µM 4-phenyl butyric acid significantly alleviated the expression of endoplasmic reticulum stress markers and iNOS in parallel with upregulated autophagy induction. However, in the presence of bafilomycin A1, the decreased viability of INS-1 cells was not recovered. Glycated albumin, an early precursor of AGE, caused pancreatic ß-cell death by inhibiting autophagy induction and flux, resulting in nuclear factor-κB (p65)-iNOS-caspase-3 cascade activation as well as by increasing susceptibility to endoplasmic reticulum stress and oxidative stress.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Insulin-Secreting Cells/drug effects , Serum Albumin/pharmacology , Animals , Autophagy-Related Protein 5 , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Glucose/pharmacology , Glycation End Products, Advanced , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulinoma/metabolism , Insulinoma/pathology , Insulinoma/physiopathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/physiopathology , Male , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Proteins/genetics , Proteins/metabolism , RNA Interference , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sequestosome-1 Protein , Transcription Factor RelA/metabolism , Glycated Serum Albumin
16.
BMB Rep ; 45(1): 51-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22281014

ABSTRACT

The purpose of this study was to determine the effects of duration and timing of glucocorticoid treatment on the expansion and differentiation of porcine neonatal pancreas cell clusters (NPCCs) into ß-cells. After transplantation of NPCCs, the ductal cyst area and ß-cell mass in the grafts both showed positive and negative correlations with duration of dexamethasone (Dx) treatment. Pdx-1 and HNF-3ß gene expression was significantly downregulated following Dx treatment, whereas PGC-1α expression increased. Pancreatic duct cell apoptosis significantly increased following Dx treatment, whereas proliferation did not change. Altogether, transdifferentiation of porcine NPCCs into ß-cells was influenced by the duration of Dx treatment, which might have been due to the suppression of key pancreatic transcription factors. PGC-1α plays an important role in the expansion and transdifferentiation of porcine NPCCs, and the initial 2 weeks following transplantation of porcine NPCCs is a critical period in determining the final ß-cell mass in grafts.


Subject(s)
Cell Proliferation/drug effects , Cell Transdifferentiation/drug effects , Dexamethasone/pharmacology , Pancreas/cytology , Pancreas/drug effects , Animals , Animals, Newborn , Apoptosis/drug effects , Cells, Cultured , Insulin/analysis , Structure-Activity Relationship , Swine
17.
Diabetes Metab J ; 35(2): 119-29, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21738894

ABSTRACT

BACKGROUND: A limitation in the number of insulin-producing pancreatic beta-cells is a special feature of diabetes. The identification of alternative sources for the induction of insulin-producing surrogate beta-cells is a matter of profound importance. PDX-1/VP16, BETA2/NeuroD, and MafA overexpression have been shown to influence the differentiation and proliferation of pancreatic stem cells. However, few studies have been conducted using adult animal pancreatic stem cells. METHODS: Adult pig pancreatic cells were prepared from the non-endocrine fraction of adult pig pancreata. Porcine neonatal pancreas cell clusters (NPCCs) were prepared from neonatal pigs aged 1-2 days. The dispersed pancreatic cells were infected with PDX-1/VP16, BETA2/NeuroD, and MafA adenoviruses. After infection, these cells were transplanted under the kidney capsules of normoglycemic nude mice. RESULTS: The adenovirus-mediated overexpression of PDX-1, BETA2/NeuroD and MafA induced insulin gene expression in NPCCs, but not in adult pig pancreatic cells. Immunocytochemistry revealed that the number of insulin-positive cells in NPCCs and adult pig pancreatic cells was approximately 2.6- and 1.1-fold greater than those in the green fluorescent protein control group, respectively. At four weeks after transplantation, the relative volume of insulin-positive cells in the grafts increased in the NPCCs, but not in the adult porcine pancreatic cells. CONCLUSION: These data indicate that PDX-1, BETA2/NeuroD, and MafA facilitate the beta-cell differentiation of NPCCs, but not adult pig pancreatic cells. Therefore PDX-1, BETA2/NeuroD, and MafA-induced NPCCs can be considered good sources for the induction of pancreatic beta-cells, and may also have some utility in the treatment of diabetes.

18.
Endocrinology ; 150(9): 4074-83, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19520786

ABSTRACT

Peroxisome proliferator-activated receptor gamma-coactivator-1alpha (PGC-1alpha) is significantly elevated in the islets of animal models of diabetes. However, the molecular mechanism has not been clarified. We investigated whether the suppression of PGC-1alpha expression protects against beta-cell dysfunction in vivo and determined the mechanism of action of PGC-1alpha in beta-cells. The studies were performed in glucolipotixicity-induced primary rat islets and INS-1 cells. In vitro and in vivo approaches using adenoviruses were used to evaluate the role of PGC-1alpha in glucolipotoxicity-associated beta-cell dysfunction. The expression of PGC-1alpha in cultured beta-cells increased gradually with glucolipotoxicity. The overexpression of PGC-1alpha also suppressed the expression of the insulin and beta-cell E-box transcription factor (BETA2/NeuroD) genes, which was reversed by PGC-1alpha small interfering RNA (siRNA). BETA2/NeuroD, p300-enhanced BETA2/NeuroD, and insulin transcriptional activities were significantly suppressed by Ad-PGC-1alpha but were rescued by Ad-siPGC-1alpha. PGC-1alpha binding at the glucocorticoid receptor site on the BETA2/NeuroD promoter increased in the presence of PGC-1alpha. Ad-siPGC-1alpha injection through the celiac arteries of 90% pancreatectomized diabetic rats improved their glucose tolerance and maintained their fasting insulin levels. The suppression of PGC-1alpha expression protects the glucolipotoxicity-induced beta-cell dysfunction in vivo and in vitro. A better understanding of the functions of molecules such as PGC-1alpha, which play key roles in intracellular fuel regulation, could herald a new era of the treatment of patients with type 2 diabetes mellitus by providing protection from glucolipotoxicity, which is an important cause of the development and progression of the disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/drug effects , RNA-Binding Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Disease Models, Animal , Fatty Acids/metabolism , Glucose/metabolism , Insulin-Secreting Cells/physiology , Pancreatectomy , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Promoter Regions, Genetic/drug effects , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL