51
|
Suzuki K, Bonner-Weir S, Hollister-Lock J, Colton CK, Weir GC. Number and Volume of Islets Transplanted in Immunobarrier Devices. Cell Transplant 2017; 7:47-52. [PMID: 9489762 DOI: 10.1177/096368979800700107] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Immunobarrier devices may prevent immune destruction of transplanted islets, but there are concerns about survival within such devices. Islets were transplanted in diffusion chambers that employed two laminated polytetrafluoroethylene membranes held together with titanium rings. Five hundred syngeneic mouse islets placed in devices were transplanted into the epididymal fat pads of streptozotocin (STZ) diabetic mice (B6AF1). After 2 wk the devices were removed. Sections were made parallel to the membrane surface. Eight to 13 systematically selected sections of each device were analyzed by planimetry to determine the area of the device space and of the islets within that space. From these data we estimated total volume of the device, volume of islets, and number of islets in a device. The data were segregated into two groups: group I (blood glucose less than 100 mg/dL 2 wk after implantation), and group II (over 150 mg/dL). The volume (mean + SE) of devices implanted for 2 wk was 2.1 + 0.4 μL in group I and 2.2 + 0.2 μL in group II. The islet volume and number within devices were 0.30 + 0.06 and 0.17 + 0.01 μL, or 340 + 50 and 230 + 20 islets in group I and group II, respectively. The volume of fibrous tissue in devices was about 0.50 μL. About 10% of the islet tissue had central necrosis. The beta cell volume in a membrane device needed for cure is comparable to that required with islets under the kidney capsule (0.25-0.80 μL). The mass of islets contained within membrane devices needed to cure diabetes is equivalent to that of a graft in an optimal transplant site such as under the kidney capsule.
Collapse
Affiliation(s)
- K Suzuki
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
52
|
Suzuki K, Ueda H, Yokono K, Taniguchi H. Normalization of Blood Glucose after Islet Cografting with Placental Tissues in Diabetic Mice. Cell Transplant 2017. [DOI: 10.3727/000000002783985701] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
A fetus in the uterus is not rejected at any time during the entire gestational period, even without the administration of immunosuppressive agents, though fetus is a kind of allograft. This prevention of rejection is considered to be associated with the presence of placental tissues. This hypothesis was tested by the allografting of islets together with placental tissues (trophoblasts) in streptozotocin (STZ)-induced diabetic mice. Placentae were harvested from the mice at the 14th postgestation day by being peeled off carefully and with the maternal decidua left behind, and cut into small pieces. Five hundred freshly isolated islets together with placental tissues obtained from ICR mice were placed under the left kidney capsule of STZ-induced diabetic C57BL/6J mice. The nonfasting blood glucose level was reduced from 477 ± 41 mg/dl at the time of pretransplant to that of the intact normal mice (161 ± 18 mg/dl) soon after the cografting, and did not return to the pretransplant level before the 14th posttransplant day. The grafting of the same number of islets alone and/or liver tissues dropped the blood glucose level, but not to that of the intact normal mice. It returned to the pretransplant level within 1 week. This is the first successful prolongation of survival of allografted islets without immunosuppressive agents through cotransplantation of allogenic placental tissues. The underlying mechanism remains to be clarified.
Collapse
Affiliation(s)
- Kazuhisa Suzuki
- Department of Metabolism, Kobe Mahoshi Hospital, Kobe 651-1242, Japan
- Department of Geriatric Medicine, Kobe University School of Medicine, Kobe 650-0017, Japan
| | - Hiroo Ueda
- Department of Geriatric Medicine, Kobe University School of Medicine, Kobe 650-0017, Japan
| | - Koichi Yokono
- Department of Geriatric Medicine, Kobe University School of Medicine, Kobe 650-0017, Japan
| | - Hiroshi Taniguchi
- Department of Metabolism and Community Health Science, Kobe University School of Medicine, Kobe 654-0142, Japan
| |
Collapse
|
53
|
Suzuki K, Bonner-Weir S, Hollister J, Weir GC. A Method for Estimating Number and Mass of Islets Transplanted within a Membrane Device. Cell Transplant 2017; 5:613-25. [PMID: 8951219 DOI: 10.1177/096368979600500604] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Immunobarrier devices may prevent the immune destruction of pancreatic islets transplanted into diabetic recipients, but there are concerns about the survival of islets within such devices. In this manuscript we described a method for estimating islet mass and number within a membrane device. Five hundred syngeneic mouse islets were placed in a membrane device, which was then transplanted into the epididymal space of streptozotocin diabetic mouse. After 14 days the device was removed from the recipient, fixed, and embedded in paraffin. Sections were made and then stained with hematoxylin. From a total of 58 sections, 12 sample sections were selected for analysis by computerized planimetry to determine area of the device chamber and the islet area for each of these sections. By making certain assumptions, it was possible to estimate the total volume of the device chamber, the number of islets contained within the chamber, and the mass of islet tissue. Two weeks after implantation of the chamber, into which approximately 500 microencapsulated islets were loaded, the blood glucose level of the recipient fell to 73 mg/dL. The volume of the device chamber, the total volume of islets in the device, and the total islet number within the device were 1.78 μL, 0.57 μL, and 277 islets, respectively. The detailed methodology, assumptions, and calculations for this approach are described in this manuscript. This new method makes it possible to determine islet mass within a membrane device by analyzing a relatively small number of selected sections. This approach should make it possible to carry out comprehensive studies on the fate of transplanted islets contained in such immunobarrier membrane devices.
Collapse
Affiliation(s)
- K Suzuki
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
54
|
Grochowiecki T, Gotoh M, Dono K, Takeda Y, Nishihara M, Ohta Y, Kimura F, Ohzato H, Umeshita K, Sakon M, Monden M. Pretreatment of Crude Pancreatic Islets with Mitomycin C (Mmc) Prolongs Islet Graft Survival in a Xenogeneic Rat-To-Mouse Model. Cell Transplant 2017; 7:411-2. [PMID: 9710312 DOI: 10.1177/096368979800700411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In this study, we examined the effect of mitomycin C (MMC) treatment on graft survival and evaluated its efficacy in immunomodulation of islet graft for transplantation. Male WS rats were used as islet donors and streptozotocin-induced diabetic C57BL/6 mice as recipients. The isolated islets were treated with MMC at concentrations of 0, 0.1, 1, 3.2, 10, 32, 100, 320, and 1000 μg/mL for 30 min, and were cultured for 20 h. Then, 300–400 islets were transplanted into the renal subcapsular space of diabetic mice. Significant prolongation of graft survival was obtained when the islets were treated with MMC at a concentration of 10, 32, or 100 μg/mL (MST 23 ± 7.4, 17.5 ± 5.4, 29.6 ± 9.7 days: p < 0.003, p < 0.012, p < 0.001, respectively, vs. 12.3 ± 2.7 days for culturing alone). Islets treated with MMC at a concentration of 320 μg/mL or more failed to restore normoglycemia in the diabetic recipient mice after transplantation. Viability of islets incubated with doses up to 100 μg/mL, assessed under the confocal microscope after propidium iodide and Hoechst 33342 staining, was maintained well comparable to that of freshly isolated islets, while those treated at 320 μg/mL was significantly decreased. Thus, a therapeutic window for MMC efficacy was found at concentrations from 10 μg/mL to 100 μg/mL. This modality is simple and effective and underlying molecular mechanisms need to be determined in the future.
Collapse
Affiliation(s)
- T Grochowiecki
- Department of Surgery II, Osaka University Medical School, Yamadaoka, Suita, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Suarez-Pinzon WL, Marcoux Y, Ghahary A, Rabinovitch A. Gene Transfection and Expression of Transforming Growth Factor-β1 in Nonobese Diabetic Mouse Islets Protects β-Cells in Syngeneic Islet Grafts from Autoimmune Destruction. Cell Transplant 2017. [DOI: 10.3727/000000002783985503] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Nonobese diabetic (NOD) mice develop diabetes and destroy syngeneic islet grafts through an autoimmune response. Because transforming growth factor (TGF)-β1 downregulates immune responses, we tested whether overexpression of TGF-β1 by gene transfection of NOD mouse islets could protect β-cells in islet grafts from autoimmune destruction. NOD mouse islet cells were transfected with an adenoviral DNA expression vector encoding porcine latent TGF-β1 (Ad TGF- β1) or the adenoviral vector alone (control Ad vector). The frequency of total islet cells expressing TGF-1 protein was increased from 12±1% in control Ad vector-transfected cells to 89 ± 4% in Ad TGF-β1-transfected islet cells, and the frequency of β-cells that expressed TGF-β1 was increased from 12 ± 1% to 60 ± 7%. Also, secretion of TGF-β1 was significantly increased in islets that overexpressed TGF-β1. Ad TGF-β1-transfected NOD mouse islets that overexpressed TGF-β1 prevented diabetes recurrence after transplantation into diabetic NOD mice for a median of 22 days compared with only 7 days for control Ad vector-transfected islets (p = 0.001). Immunohistochemical examination of the islet grafts revealed significantly more TGF-β1+ cells and insulin+ cells and significantly fewer CD45+ leukocytes in Ad TGF-β1-transfected islet grafts. Also, islet β-cell apoptosis was significantly decreased whereas apoptosis of graft-infiltrating leukocytes was significantly increased in Ad TGF-β1-transfected islet grafts. These observations demonstrate that overexpression of TGF-β1, by gene transfection of NOD mouse islets, protects islet β-cells from apoptosis and autoimmune destruction and delays diabetes recurrence after islet transplantation.
Collapse
Affiliation(s)
| | - Yvonne Marcoux
- Department of Surgery, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Aziz Ghahary
- Department of Surgery, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Alex Rabinovitch
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| |
Collapse
|
56
|
Amisten S, Mohammad Al-Amily I, Soni A, Hawkes R, Atanes P, Persaud SJ, Rorsman P, Salehi A. Anti-diabetic action of all-trans retinoic acid and the orphan G protein coupled receptor GPRC5C in pancreatic β-cells. Endocr J 2017; 64:325-338. [PMID: 28228611 DOI: 10.1507/endocrj.ej16-0338] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Pancreatic islets express high levels of the orphan G-protein coupled receptor C5C (GPRC5C), the function of which remains to be established. Here we have examined the role of GPRC5C in the regulation of insulin secretion and β-cell survival and proliferation using human and mouse pancreatic islets. The expression of GPRC5C was analysed by RNA-sequencing, qPCR, western blotting and confocal microscopy. Insulin secretion and cell viability were determined by RIA and MTS assays, respectively. GPRC5C mRNA expression and protein level were reduced in the islets from type-2 diabetic donors. RNA sequencing in human islets revealed GPRC5C expression correlated with the expression of genes controlling apoptosis, cell survival and proliferation. A reduction in Gprc5c mRNA and protein expression was observed in islets isolated from old mice (>46 weeks of age) compared to that in islets from newborn (<3 weeks) mice. Down-regulation of Gprc5c led to both moderately reduced glucose-stimulated insulin release and also reduced cAMP content in mouse islets. Potentiation of glucose-stimulated insulin secretion concomitant with enhanced islet cAMP level by all-trans retinoic acid (ATRA) was attenuated upon Gprc5c-KD. ATRA also increased [Ca+2]i in Huh7-cells. Gprc5c over expression in Huh7 cells was associated with increased ERK1/2 activity. Gprc5c-KD in clonal MIN6c4 cells reduced cell proliferation and in murine islets increased apoptosis and the sensitivity of primary islet cells to a cocktail of pro-apoptotic cytokines. Our results demonstrate that agents activating GPRC5C represent a novel modality for the treatment and/or prevention of diabetes by restoring and/or maintaining functional β-cell mass.
Collapse
Affiliation(s)
- Stefan Amisten
- The Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Vasu S, McGahon MK, Moffett RC, Curtis TM, Conlon JM, Abdel-Wahab YHA, Flatt PR. Esculentin-2CHa(1-30) and its analogues: stability and mechanisms of insulinotropic action. J Endocrinol 2017; 232:423-435. [PMID: 28115493 DOI: 10.1530/joe-16-0453] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 01/03/2017] [Indexed: 12/17/2022]
Abstract
The insulin-releasing effects, cellular mechanisms of action and anti-hyperglycaemic activity of 10 analogues of esculentin-2CHa lacking the cyclic C-terminal domain (CKISKQC) were evaluated. Analogues of the truncated peptide, esculentin-2CHa(1-30), were designed for plasma enzyme resistance and increased biological activity. Effects of those analogues on insulin release, cell membrane integrity, membrane potential, intracellular Ca2+ and cAMP levels were determined using clonal BRIN-BD11 cells. Their acute effects on glucose tolerance were investigated using NIH Swiss mice. d-Amino acid substitutions at positions 7(Arg), 15(Lys) and 23(Lys) and fatty acid (l-octanoate) attachment to Lys at position 15 of esculentin-2CHa(1-30) conveyed resistance to plasma enzyme degradation whilst preserving insulin-releasing activity. Analogues, [d-Arg7,d-Lys15,d-Lys23]-esculentin-2CHa(1-30) and Lys15-octanoate-esculentin-2CHa(1-30), exhibiting most promising profiles and with confirmed effects on both human insulin-secreting cells and primary mouse islets were selected for further analysis. Using chemical inhibition of adenylate cyclase, protein kinase C or phospholipase C pathways, involvement of PLC/PKC-mediated insulin secretion was confirmed similar to that of CCK-8. Diazoxide, verapamil and Ca2+ omission inhibited insulin secretion induced by the esculentin-2CHa(1-30) analogues suggesting an action on KATP and Ca2+ channels also. Consistent with this, the analogues depolarised the plasma membrane and increased intracellular Ca2+ Evaluation with fluorescent-labelled esculentin-2CHa(1-30) indicated membrane action, with internalisation; however, patch-clamp experiments suggested that depolarisation was not due to the direct inhibition of KATP channels. Acute administration of either analogue to NIH Swiss mice improved glucose tolerance and enhanced insulin release similar to that observed with GLP-1. These data suggest that multi-acting analogues of esculentin-2CHa(1-30) may prove useful for glycaemic control in obesity-diabetes.
Collapse
Affiliation(s)
- Srividya Vasu
- SAAD Centre for Pharmacy & DiabetesSchool of Biomedical Sciences, University of Ulster, Coleraine, UK
| | - Mary K McGahon
- Centre for Experimental MedicineQueens University of Belfast, Belfast, UK
| | - R Charlotte Moffett
- SAAD Centre for Pharmacy & DiabetesSchool of Biomedical Sciences, University of Ulster, Coleraine, UK
| | - Tim M Curtis
- Centre for Experimental MedicineQueens University of Belfast, Belfast, UK
| | - J Michael Conlon
- SAAD Centre for Pharmacy & DiabetesSchool of Biomedical Sciences, University of Ulster, Coleraine, UK
| | - Yasser H A Abdel-Wahab
- SAAD Centre for Pharmacy & DiabetesSchool of Biomedical Sciences, University of Ulster, Coleraine, UK
| | - Peter R Flatt
- SAAD Centre for Pharmacy & DiabetesSchool of Biomedical Sciences, University of Ulster, Coleraine, UK
| |
Collapse
|
58
|
Zongyi Y, Funian Z, Hao L, Ying C, Jialin Z, Baifeng L. A rapid, efficient, and economic device and method for the isolation and purification of mouse islet cells. PLoS One 2017; 12:e0171618. [PMID: 28207765 PMCID: PMC5313167 DOI: 10.1371/journal.pone.0171618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/23/2017] [Indexed: 11/28/2022] Open
Abstract
Rapid, efficient, and economic method for the isolation and purification of islets has been pursued by numerous islet-related researchers. In this study, we compared the advantages and disadvantages of our developed patented method with those of commonly used conventional methods (Ficoll-400, 1077, and handpicking methods). Cell viability was assayed using Trypan blue, cell purity and yield were assayed using diphenylthiocarbazone, and islet function was assayed using acridine orange/ethidium bromide staining and enzyme-linked immunosorbent assay-glucose stimulation testing 4 days after cultivation. The results showed that our islet isolation and purification method required 12 ± 3 min, which was significantly shorter than the time required in Ficoll-400, 1077, and HPU groups (34 ± 3, 41 ± 4, and 30 ± 4 min, respectively; P < 0.05). There was no significant difference in islet viability among the four groups. The islet purity, function, yield, and cost of our method were superior to those of the Ficoll-400 and 1077 methods, but inferior to the handpicking method. However, the handpicking method may cause wrist injury and visual impairment in researchers during large-scale islet isolation (>1000 islets). In summary, the MCT method is a rapid, efficient, and economic method for isolating and purifying murine islet cell clumps. This method overcomes some of the shortcomings of conventional methods, showing a relatively higher quality and yield of islets within a shorter duration at a lower cost. Therefore, the current method provides researchers with an alternative option for islet isolation and should be widely generalized.
Collapse
Affiliation(s)
- Yin Zongyi
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
| | - Zou Funian
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
| | - Li Hao
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China
| | - Cheng Ying
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China.,National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China.,Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
| | - Zhang Jialin
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China.,National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China.,Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
| | - Li Baifeng
- Department of Hepatobiliary Surgery and Organ Transplantation, the First Hospital of China Medical University, Shenyang, China.,National Key Lab. of General Surgery, the First Hospital of China Medical University, Shenyang, China.,Multiple Organ Transplantation Institute of the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
59
|
Müller A, Neukam M, Ivanova A, Sönmez A, Münster C, Kretschmar S, Kalaidzidis Y, Kurth T, Verbavatz JM, Solimena M. A Global Approach for Quantitative Super Resolution and Electron Microscopy on Cryo and Epoxy Sections Using Self-labeling Protein Tags. Sci Rep 2017; 7:23. [PMID: 28154417 PMCID: PMC5428382 DOI: 10.1038/s41598-017-00033-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 12/20/2016] [Indexed: 01/19/2023] Open
Abstract
Correlative light and electron microscopy (CLEM) is a powerful approach to investigate the molecular ultrastructure of labeled cell compartments. However, quantitative CLEM studies are rare, mainly due to small sample sizes and the sensitivity of fluorescent proteins to strong fixatives and contrasting reagents for EM. Here, we show that fusion of a self-labeling protein to insulin allows for the quantification of age-distinct insulin granule pools in pancreatic beta cells by a combination of super resolution and transmission electron microscopy on Tokuyasu cryosections. In contrast to fluorescent proteins like GFP organic dyes covalently bound to self-labeling proteins retain their fluorescence also in epoxy resin following high pressure freezing and freeze substitution, or remarkably even after strong chemical fixation. This enables for the assessment of age-defined granule morphology and degradation. Finally, we demonstrate that this CLEM protocol is highly versatile, being suitable for single and dual fluorescent labeling and detection of different proteins with optimal ultrastructure preservation and contrast.
Collapse
Affiliation(s)
- Andreas Müller
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Martin Neukam
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Anna Ivanova
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Anke Sönmez
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Carla Münster
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany
| | - Susanne Kretschmar
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany.,Biotechnology Center of the TU Dresden (BIOTEC), Dresden, Germany
| | - Yannis Kalaidzidis
- Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Dresden, Germany.,Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow, Russia
| | - Thomas Kurth
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany.,Biotechnology Center of the TU Dresden (BIOTEC), Dresden, Germany
| | - Jean-Marc Verbavatz
- Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Dresden, Germany.,Institut Jacques Monod, Université Paris Diderot, Paris, France
| | - Michele Solimena
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany. .,Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany. .,Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Dresden, Germany.
| |
Collapse
|
60
|
Alwahsh SM, Dwyer BJ, Forbes S, Thiel DHV, Lewis PJS, Ramadori G. Insulin Production and Resistance in Different Models of Diet-Induced Obesity and Metabolic Syndrome. Int J Mol Sci 2017; 18:ijms18020285. [PMID: 28134848 PMCID: PMC5343821 DOI: 10.3390/ijms18020285] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/13/2017] [Accepted: 01/17/2017] [Indexed: 12/18/2022] Open
Abstract
The role of the liver and the endocrine pancreas in development of hyperinsulinemia in different types of obesity remains unclear. Sedentary rats (160 g) were fed a low-fat-diet (LFD, chow 13% kcal fat), high-fat-diet (HFD, 35% fat), or HFD+ 30% ethanol+ 30% fructose (HF-EFr, 22% fat). Overnight-fasted rats were culled after one, four or eight weeks. Pancreatic and hepatic mRNAs were isolated for subsequent RT-PCR analysis. After eight weeks, body weights increased three-fold in the LFD group, 2.8-fold in the HFD group, and 2.4-fold in the HF-EFr (p < 0.01). HF-EFr-fed rats had the greatest liver weights and consumed less food during Weeks 4–8 (p < 0.05). Hepatic-triglyceride content increased progressively in all groups. At Week 8, HOMA-IR values, fasting serum glucose, C-peptide, and triglycerides levels were significantly increased in LFD-fed rats compared to that at earlier time points. The greatest plasma levels of glucose, triglycerides and leptin were observed in the HF-EFr at Week 8. Gene expression of pancreatic-insulin was significantly greater in the HFD and HF-EFr groups versus the LFD. Nevertheless, insulin: C-peptide ratios and HOMA-IR values were substantially higher in HF-EFr. Hepatic gene-expression of insulin-receptor-substrate-1/2 was downregulated in the HF-EFr. The expression of phospho-ERK-1/2 and inflammatory-mediators were greatest in the HF-EFr-fed rats. Chronic intake of both LFD and HFD induced obesity, MetS, and intrahepatic-fat accumulation. The hyperinsulinemia is the strongest in rats with the lowest body weights, but having the highest liver weights. This accompanies the strongest increase of pancreatic insulin production and the maximal decrease of hepatic insulin signaling, which is possibly secondary to hepatic fat deposition, inflammation and other factors.
Collapse
Affiliation(s)
- Salamah M Alwahsh
- Clinic for Gastroenterology and Endocrinology, University Medical Center, Georg-August-University Goettingen, Goettingen D-37075, Germany.
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK.
| | - Benjamin J Dwyer
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK.
| | - Shareen Forbes
- Endocrinology Unit, University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| | - David H van Thiel
- Advanced Liver and Gastrointestinal Disease Center, Chicago, IL 60611, USA.
| | | | - Giuliano Ramadori
- Clinic for Gastroenterology and Endocrinology, University Medical Center, Georg-August-University Goettingen, Goettingen D-37075, Germany.
| |
Collapse
|
61
|
Tsukita S, Yamada T, Takahashi K, Munakata Y, Hosaka S, Takahashi H, Gao J, Shirai Y, Kodama S, Asai Y, Sugisawa T, Chiba Y, Kaneko K, Uno K, Sawada S, Imai J, Katagiri H. MicroRNAs 106b and 222 Improve Hyperglycemia in a Mouse Model of Insulin-Deficient Diabetes via Pancreatic β-Cell Proliferation. EBioMedicine 2016; 15:163-172. [PMID: 27974246 PMCID: PMC5233820 DOI: 10.1016/j.ebiom.2016.12.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 11/16/2016] [Accepted: 12/04/2016] [Indexed: 01/01/2023] Open
Abstract
Major symptoms of diabetes mellitus manifest, once pancreatic β-cell numbers have become inadequate. Although natural regeneration of β-cells after injury is very limited, bone marrow (BM) transplantation (BMT) promotes their regeneration through undetermined mechanism(s) involving inter-cellular (BM cell-to-β-cell) crosstalk. We found that two microRNAs (miRNAs) contribute to BMT-induced β-cell regeneration. Screening murine miRNAs in serum exosomes after BMT revealed 42 miRNAs to be increased. Two of these miRNAs (miR-106b-5p and miR-222-3p) were shown to be secreted by BM cells and increased in pancreatic islet cells after BMT. Treatment with the corresponding anti-miRNAs inhibited BMT-induced β-cell regeneration. Furthermore, intravenous administration of the corresponding miRNA mimics promoted post-injury β-cell proliferation through Cip/Kip family down-regulation, thereby ameliorating hyperglycemia in mice with insulin-deficient diabetes. Thus, these identified miRNAs may lead to the development of therapeutic strategies for diabetes. BMT regenerates β-cells in mice with STZ-induced diabetes and increases miR-106b and miR-222 in serum exosomes and islets. Inhibition with anti-miRs against these miRs suppresses BMT-induced β-cell regeneration. Injection of miR-106b and miR-222 mimics promotes β-cell proliferation and improves hyperglycemia in STZ-treated mice.
Regeneration of pancreatic β-cells is a promising therapeutic strategy not only for type 1 diabetes but also for certain forms of type 2 diabetes. However, natural regeneration of β-cells hardly ever occurs. Interestingly, bone marrow transplantation (BMT) has been shown to promote β-cell regeneration through an undetermined mechanism(s). In this study, we found that two microRNAs (miR-106b/-222) contribute to BMT-induced β-cell proliferation. Inhibition of miR-106b/-222 using specific anti-miRNAs significantly suppressed BMT-induced β-cell proliferation. Furthermore, intravenously administered miR-106b/222 promoted β-cell proliferation, thereby ameliorating hyperglycemia in mice with insulin-deficient diabetes. Thus, these identified miRNAs may lead to novel therapeutic strategies for diabetes.
Collapse
Affiliation(s)
- Sohei Tsukita
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Tetsuya Yamada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan; Centre for Metabolic Diseases, Tohoku University Graduate School of Medicine, Japan.
| | - Kei Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Yuichiro Munakata
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Shinichiro Hosaka
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Hironobu Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Junhong Gao
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan; Tohoku University Frontier Research Institute for Interdisciplinary Science, Miyagi, Japan
| | - Yuta Shirai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Shinjiro Kodama
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Yoichiro Asai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Takashi Sugisawa
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Yumiko Chiba
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Kenji Uno
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Shojiro Sawada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Junta Imai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Japan; Centre for Metabolic Diseases, Tohoku University Graduate School of Medicine, Japan; Japan Agency for Medical Research and Development, CREST, Japan
| |
Collapse
|
62
|
Lundquist I, Mohammed Al-Amily I, Meidute Abaraviciene S, Salehi A. Metformin Ameliorates Dysfunctional Traits of Glibenclamide- and Glucose-Induced Insulin Secretion by Suppression of Imposed Overactivity of the Islet Nitric Oxide Synthase-NO System. PLoS One 2016; 11:e0165668. [PMID: 27820841 PMCID: PMC5098820 DOI: 10.1371/journal.pone.0165668] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/14/2016] [Indexed: 01/15/2023] Open
Abstract
Metformin lowers diabetic blood glucose primarily by reducing hepatic gluconeogenesis and increasing peripheral glucose uptake. However, possible effects by metformin on beta-cell function are incompletely understood. We speculated that metformin might positively influence insulin secretion through impacting the beta-cell nitric oxide synthase (NOS)-NO system, a negative modulator of glucose-stimulated insulin release. In short-time incubations with isolated murine islets either glibenclamide or high glucose augmented insulin release associated with increased NO production from both neural and inducible NOS. Metformin addition suppressed the augmented NO generation coinciding with amplified insulin release. Islet culturing with glibenclamide or high glucose revealed pronounced fluorescence of inducible NOS in the beta-cells being abolished by metformin co-culturing. These findings were reflected in medium nitrite-nitrate levels. A glucose challenge following islet culturing with glibenclamide or high glucose revealed markedly impaired insulin response. Metformin co-culturing restored this response. Culturing murine islets and human islets from controls and type 2 diabetics with high glucose or high glucose + glibenclamide induced a pronounced decrease of cell viability being remarkably restored by metformin co-culturing. We show here, that imposed overactivity of the beta-cell NOS-NO system by glibenclamide or high glucose leads to insulin secretory dysfunction and reduced cell viability and also, importantly, that these effects are relieved by metformin inhibiting beta-cell NO overproduction from both neural and inducible NOS thus ameliorating a concealed negative influence by NO induced by sulfonylurea treatment and/or high glucose levels. This double-edged effect of glibenclamide on the beta-cellsuggests sulfonylurea monotherapy in type 2 diabetes being avoided.
Collapse
Affiliation(s)
- Ingmar Lundquist
- Department of Clinical Science, SUS, Division of Islet Cell Physiology University of Lund, Malmö, Sweden
- Department of Experimental Medical Science, University of Lund, Lund, Sweden
| | - Israa Mohammed Al-Amily
- Department of Clinical Science, SUS, Division of Islet Cell Physiology University of Lund, Malmö, Sweden
| | - Sandra Meidute Abaraviciene
- Department of Clinical Science, SUS, Division of Islet Cell Physiology University of Lund, Malmö, Sweden
- Dept. of Physiology, Biochemistry, Microbiology and Laboratory Medicine Vilnius University, and Dep. of Regenerative Medicine, State Research Institute Center for Innovative Medicine, Vilnius, Lithuania
| | - Albert Salehi
- Department of Clinical Science, SUS, Division of Islet Cell Physiology University of Lund, Malmö, Sweden
- Department of Neuroscience and Physiology, Metabolic Research Unit, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
63
|
Owolabi BO, Ojo OO, Srinivasan DK, Conlon JM, Flatt PR, Abdel-Wahab YHA. Glucoregulatory, endocrine and morphological effects of [P5K]hymenochirin-1B in mice with diet-induced glucose intolerance and insulin resistance. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:769-81. [DOI: 10.1007/s00210-016-1243-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/04/2016] [Indexed: 12/25/2022]
|
64
|
Taguchi Y, Allende ML, Mizukami H, Cook EK, Gavrilova O, Tuymetova G, Clarke BA, Chen W, Olivera A, Proia RL. Sphingosine-1-phosphate Phosphatase 2 Regulates Pancreatic Islet β-Cell Endoplasmic Reticulum Stress and Proliferation. J Biol Chem 2016; 291:12029-38. [PMID: 27059959 DOI: 10.1074/jbc.m116.728170] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Indexed: 11/06/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a sphingolipid metabolite that regulates basic cell functions through metabolic and signaling pathways. Intracellular metabolism of S1P is controlled, in part, by two homologous S1P phosphatases (SPPases), 1 and 2, which are encoded by the Sgpp1 and Sgpp2 genes, respectively. SPPase activity is needed for efficient recycling of sphingosine into the sphingolipid synthesis pathway. SPPase 1 is important for skin homeostasis, but little is known about the functional role of SPPase 2. To identify the functions of SPPase 2 in vivo, we studied mice with the Sgpp2 gene deleted. In contrast to Sgpp1(-/-) mice, Sgpp2(-/-) mice had normal skin and were viable into adulthood. Unexpectedly, WT mice expressed Sgpp2 mRNA at high levels in pancreatic islets when compared with other tissues. Sgpp2(-/-) mice had normal pancreatic islet size; however, they exhibited defective adaptive β-cell proliferation that was demonstrated after treatment with either a high-fat diet or the β-cell-specific toxin, streptozotocin. Importantly, β-cells from untreated Sgpp2(-/-) mice showed significantly increased expression of proteins characteristic of the endoplasmic reticulum stress response compared with β-cells from WT mice, indicating a basal islet defect. Our results show that Sgpp2 deletion causes β-cell endoplasmic reticulum stress, which is a known cause of β-cell dysfunction, and reveal a juncture in the sphingolipid recycling pathway that could impact the development of diabetes.
Collapse
Affiliation(s)
| | | | - Hiroki Mizukami
- the Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Emily K Cook
- From the Genetics of Development and Disease Branch
| | | | | | | | | | - Ana Olivera
- the Laboratory of Allergic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892 and
| | | |
Collapse
|
65
|
Ma D, Duan W, Li Y, Wang Z, Li S, Gong N, Chen G, Chen Z, Wan C, Yang J. PD-L1 Deficiency within Islets Reduces Allograft Survival in Mice. PLoS One 2016; 11:e0152087. [PMID: 26990974 PMCID: PMC4798758 DOI: 10.1371/journal.pone.0152087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/08/2016] [Indexed: 12/21/2022] Open
Abstract
Background Islet transplantation may potentially cure type 1 diabetes mellitus (T1DM). However, immune rejection, especially that induced by the alloreactive T-cell response, remains a restraining factor for the long-term survival of grafted islets. Programmed death ligand-1 (PD-L1) is a negative costimulatory molecule. PD-L1 deficiency within the donor heart accelerates allograft rejection. Here, we investigate whether PD-L1 deficiency in donor islets reduces allograft survival time. Methods Glucose Stimulation Assays were performed to evaluate whether PD-L1 deficiency has detrimental effects on islet function. Islets isolated from PDL1-deficient mice or wild- type (WT) mice (C57BL/6j) were implanted beneath the renal capsule of streptozotocin (STZ)-induced diabetic BALB/c mice. Blood glucose levels and graft survival time after transplantation were monitored. Moreover, we analyzed the residual islets, infiltrating immune cells and alloreactive cells from the recipients. Results PD-L1 deficiency within islets does not affect islet function. However, islet PD-L1 deficiency increased allograft rejection and was associated with enhanced inflammatory cell infiltration and recipient T-cell alloreactivity. Conclusions This is the first report to demonstrate that PD-L1 deficiency accelerated islet allograft rejection and regulated recipient alloimmune responses.
Collapse
Affiliation(s)
- Dongxia Ma
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, Hubei Province, P. R. China
| | - Wu Duan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Yakun Li
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, Hubei Province, P. R. China
| | - Zhimin Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, Hubei Province, P. R. China
| | - Shanglin Li
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, Hubei Province, P. R. China
| | - Nianqiao Gong
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, Hubei Province, P. R. China
| | - Gang Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, Hubei Province, P. R. China
| | - Zhishui Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, Hubei Province, P. R. China
| | - Chidan Wan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
- * E-mail: (JY); (CW)
| | - Jun Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, Hubei Province, P. R. China
- * E-mail: (JY); (CW)
| |
Collapse
|
66
|
Proinsulin and heat shock protein 90 as biomarkers of beta-cell stress in the early period after onset of type 1 diabetes. Transl Res 2016; 168:96-106.e1. [PMID: 26397425 PMCID: PMC4839287 DOI: 10.1016/j.trsl.2015.08.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/12/2015] [Accepted: 08/28/2015] [Indexed: 11/23/2022]
Abstract
Rapid evaluation of therapies designed to preserve β cells in persons with type 1 diabetes (T1D) is hampered by limited availability of sensitive β-cell health biomarkers. In particular, biomarkers elucidating the presence and degree of β-cell stress are needed. We characterized β-cell secretory activity and stress in 29 new-onset T1D subjects (10.6 ± 3.0 years, 55% male) at diagnosis and then 8.2 ± 1.2 weeks later at first clinic follow-up. We did comparisons with 16 matched healthy controls. We evaluated hemoglobin A1c (HbA1c), β-cell function (random C-peptide [C] and proinsulin [PI]), β-cell stress (PI:C ratio), and the β-cell stress marker heat shock protein (HSP)90 and examined these parameters' relationships with clinical and laboratory characteristics at diagnosis. Mean diagnosis HbA1c was 11.3% (100 mmol/mol) and 7.6% (60 mmol/mol) at follow-up. C-peptide was low at diagnosis (P < 0.001 vs controls) and increased at follow-up (P < 0.001) to comparable with controls. PI did not differ from controls at diagnosis but increased at follow-up (P = 0.003) signifying increased release of PI alongside improved insulin secretion. PI:C ratios and HSP90 concentrations were elevated at both time points. Younger subjects had lower C-peptide and greater PI, PI:C, and HSP90. We also examined islets isolated from prediabetic nonobese diabetic mice and found that HSP90 levels were increased ∼4-fold compared with those in islets isolated from matched CD1 controls, further substantiating HSP90 as a marker of β-cell stress in T1D. Our data indicate that β-cell stress can be assessed using PI:C and HSP90. This stress persists after T1D diagnosis. Therapeutic approaches to reduce β-cell stress in new-onset T1D should be considered.
Collapse
|
67
|
Arefanian H, Tredget EB, Mok DCM, Ramji Q, Rafati S, Rodriguez-Barbosa J, Korbutt GS, Rajotte RV, Gill RG, Rayat GR. Porcine Islet-Specific Tolerance Induced by the Combination of Anti-LFA-1 and Anti-CD154 mAbs is Dependent on PD-1. Cell Transplant 2016; 25:327-42. [DOI: 10.3727/096368915x688506] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We previously demonstrated that short-term administration of a combination of anti-LFA-1 and anti-CD154 monoclonal antibodies (mAbs) induces tolerance to neonatal porcine islet (NPI) xenografts that is mediated by regulatory T cells (Tregs) in B6 mice. In this study, we examined whether the coinhibitory molecule PD-1 is required for the induction and maintenance of tolerance to NPI xenografts. We also determined whether tolerance to NPI xenografts could be extended to allogeneic mouse or xenogeneic rat islet grafts since we previously demonstrated that tolerance to NPI xenografts could be extended to second-party NPI xenografts. Finally, we determined whether tolerance to NPI xenografts could be extended to allogeneic mouse or second-party porcine skin grafts. Diabetic B6 mice were transplanted with 2,000 NPIs under the kidney capsule and treated with short-term administration of a combination of anti-LFA-1 and anti-CD154 mAbs. Some of these mice were also treated simultaneously with anti-PD-1 mAb at >150 days posttransplantation. Spleen cells from some of the tolerant B6 mice were used for proliferation assays or were injected into B6 rag-/- mice with established islet grafts from allogeneic or xenogeneic donors. All B6 mice treated with anti-LFA-1 and anti-CD154 mAbs achieved and maintained normoglycemia until the end of the study; however, some mice that were treated with anti-PD-1 mAb became diabetic. All B6 rag-/- mouse recipients of first- and second-party NPIs maintained normoglycemia after reconstitution with spleen cells from tolerant B6 mice, while all B6 rag-/- mouse recipients of allogeneic mouse or xenogeneic rat islets rejected their grafts after cell reconstitution. Tolerant B6 mice rejected their allogeneic mouse or xenogeneic second-party porcine skin grafts while remaining normoglycemic until the end of the study. These results show that porcine islet-specific tolerance is dependent on PD-1, which could not be extended to skin grafts.
Collapse
Affiliation(s)
- Hossein Arefanian
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
- Endocrinology and Metabolism Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Pancreatic Islet Biology and Transplantation Unit, Dasman Diabetes Institute, Kuwait, Dasman, Kuwait
| | - Eric B. Tredget
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Dereck C. M. Mok
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Qahir Ramji
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Shahin Rafati
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Jose Rodriguez-Barbosa
- Institute of Biomedicine (Immunobiology), University of Leon, Campus de Vegazana s/n, Leon, Spain
| | - Gregory S. Korbutt
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Ray V. Rajotte
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Ron G. Gill
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Gina R. Rayat
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
68
|
Ramírez-Domínguez M. Isolation of Mouse Pancreatic Islets of Langerhans. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 938:25-34. [DOI: 10.1007/978-3-319-39824-2_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
69
|
Acute intravenous glucose load impairs early insulin secretion and insulin content in islet β cells in mice. Life Sci 2016; 144:148-55. [DOI: 10.1016/j.lfs.2015.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 11/14/2015] [Accepted: 12/04/2015] [Indexed: 01/04/2023]
|
70
|
Ramírez-Domínguez M. Historical Background of Pancreatic Islet Isolation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 938:1-9. [PMID: 27586418 DOI: 10.1007/978-3-319-39824-2_1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Until the discovery of insulin in the twentieth century, diabetes mellitus was a mortal disease with an unclear origin and physiology. Despite the appearance of the concept in an Egyptian papyrus dated c.1550 BC, and the documentation of its study by ancient Chinese, the term "diabetes" was only coined by the Greek Aretaeus in the second century AD. In Europe, the study of diabetes was largely ignored until the seventeenth century, when the characteristic sweet flavor of diabetic urine was first described. However, the link between diabetes and the pancreas was not discovered until 1889 by Minkowski and von Mering, long after the first description of the pancreatic islets by Paul Langerhans in 1869. One of the most significant milestones in the field was the discovery of insulin by Banting and collaborators in 1922, which led to the therapeutic development of insulin administration as a life-saving intervention for type 1 diabetic patients. On the other hand, the isolation of islets was first reported by Bensley in 1911, a critical technical achievement that paved the way for clinical islet transplantation. Here we discuss the history of islet isolation, since the firsts studies of diabetes by ancient civilizations to the birth and parallel evolution of islet isolation and transplantation.
Collapse
Affiliation(s)
- Miriam Ramírez-Domínguez
- Laboratory of Cell Therapy of Diabetes, Department of Pediatrics, Faculty of Medicine and Odontology, Hospital Cruces, University of the Basque Country (UPV/EHU), Barrio Sarriena, s/n, 48940, Leioa, Biscay, Spain.
| |
Collapse
|
71
|
Ojo OO, Srinivasan DK, Owolabi BO, Vasu S, Conlon JM, Flatt PR, Abdel-Wahab YHA. Esculentin-2CHa-Related Peptides Modulate Islet Cell Function and Improve Glucose Tolerance in Mice with Diet-Induced Obesity and Insulin Resistance. PLoS One 2015; 10:e0141549. [PMID: 26512980 PMCID: PMC4626215 DOI: 10.1371/journal.pone.0141549] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/10/2015] [Indexed: 01/08/2023] Open
Abstract
The frog skin host-defense peptide esculentin-2CHa (GFSSIFRGVA10KFASKGLGK D20LAKLGVDLVA30CKISKQC) displays antimicrobial, antitumor, and immunomodulatory properties. This study investigated the antidiabetic actions of the peptide and selected analogues. Esculentin-2CHa stimulated insulin secretion from rat BRIN-BD11 clonal pancreatic β-cells at concentrations greater than 0.3 nM without cytotoxicity by a mechanism involving membrane depolarization and increase of intracellular Ca2+. Insulinotropic activity was attenuated by activation of KATP channels, inhibition of voltage-dependent Ca2+ channels and chelation of extracellular Ca2+. The [L21K], [L24K], [D20K, D27K] and [C31S,C37S] analogues were more potent but less effective than esculentin-2CHa whereas the [L28K] and [C31K] analogues were both more potent and produced a significantly (P < 0.001) greater maximum response. Acute administration of [L28K]esculentin-2CHa (75 nmol/kg body weight) to high fat fed mice with obesity and insulin resistance enhanced glucose tolerance and insulin secretion. Twice-daily administration of this dose of [L28K]esculentin-2CHa for 28 days had no significant effect on body weight, food intake, indirect calorimetry or body composition. However, mice exhibited decreased non-fasting plasma glucose (P < 0.05), increased non-fasting plasma insulin (P < 0.05) as well as improved glucose tolerance and insulin secretion (P < 0.01) following both oral and intraperitoneal glucose loads. Impaired responses of isolated islets from high fat fed mice to established insulin secretagogues were restored by [L28K]esculentin-2CHa treatment. Peptide treatment was accompanied by significantly lower plasma and pancreatic glucagon levels and normalization of α-cell mass. Circulating triglyceride concentrations were decreased but plasma cholesterol and LDL concentrations were not significantly affected. The data encourage further investigation of the potential of esculentin-2CHa related peptides for treatment of patients with type 2 diabetes.
Collapse
Affiliation(s)
- Opeolu O. Ojo
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, United Kingdom
- School of Sport, Health and Bioscience, University of East London, Stratford, E15 4LZ, United Kingdom
| | - Dinesh K. Srinivasan
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, United Kingdom
| | - Bosede O. Owolabi
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, United Kingdom
| | - Srividya Vasu
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, United Kingdom
| | - J. Michael Conlon
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, United Kingdom
| | - Peter R. Flatt
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, United Kingdom
| | - Yasser H. A. Abdel-Wahab
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, United Kingdom
- * E-mail:
| |
Collapse
|
72
|
Li T, Ma R, Zhu JY, Wang FS, Huang L, Leng XS. PD-1/PD-L1 costimulatory pathway-induced mouse islet transplantation immune tolerance. Transplant Proc 2015; 47:165-70. [PMID: 25645798 DOI: 10.1016/j.transproceed.2014.10.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 10/28/2014] [Indexed: 11/25/2022]
Abstract
BACKGROUND Programmed death-1/PD-1 ligand-1 (PD-1/PD-L1) costimulatory signals may play an important role in T-cell-induced immune response. The aim of this study is to investigate the role of the PD-1/PD-L1 costimulatory pathway on immunotolerance induction in mouse pancreatic islet transplantation. METHODS Full-length mouse PD-L1 cDNA was subcloned into pShuttle-GFP-CMV(-) shuttle plasmid. The product was cut by certain restriction endonuclease and ligated with pAdxsi vector. The adenovirus bone plasmid was transformed into DH5α-competent bacteria. After linearization, the recombined adenovirus DNA was transfected into 293 cells for package and amplification. Streptozotocin was injected intraperitoneally into C57BL/6 (H-2(b)) mouse to induce diabetic model recipient. Recipients were randomly divided into 3 groups. Group A was the control. Group B and group C were injected with Ad-EGFP and Ad-PD-L1 through the tail vein, respectively, 1 day before islet transplantation. The 300 to 400 islets of DBA/2 (H-2(d)) were transplanted into the renal subcapsular space of the diabetic model recipient. We monitored and analyzed the blood glucose levels and the survival time of grafts after transplantation. RESULTS Recombinant adenovirus Ad-PD-L1 had high efficiency expression of PD-L1 in recipient mouse. The blood glucose concentration of mice in the Ad-PD-L1 gene treatment group was obviously lower than that of the control and Ad-EGFP treatment groups and was stable and kept within the normal range at post-transplant 21 days. The survival time of grafts in the Ad-PD-L1 group (27.6 ± 3.5 days) was significantly longer than in the control (7.8 ± 0.33 days) and Ad-EGFP groups (7.6 ± 0.59 days), P < .01. Mixed lymphocyte response showed a specific decrease reaction of recipient lymphocyte vs donor lymphocyte. Flow cytometry detection showed that unsplit cells occupied 90% of recipient mouse lymphocytes, but unsplit cells among normal C57BL/6 mouse lymphocytes without Ad-PD-L1 gene treatment were 51 in the control group. The differences between them were significant (P < .01). CONCLUSION Recombinant adenovirus Ad-PD-L1 has been successfully constructed. In mouse pancreatic islet transplantation, it can suppress the activation of recipient T lymphocyte through the PD-1/PD-L1 costimulatory pathway, and significantly prolong the survival time of grafts.
Collapse
Affiliation(s)
- T Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - R Ma
- Department of Surgical Intensive Care Unit, Peking University People's Hospital, Beijing, China
| | - J Y Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China.
| | - F S Wang
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - L Huang
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - X S Leng
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| |
Collapse
|
73
|
Owolabi BO, Ojo OO, Srinivasan DK, Conlon JM, Flatt PR, Abdel-Wahab YHA. In vitro and in vivo insulinotropic properties of the multifunctional frog skin peptide hymenochirin-1B: a structure-activity study. Amino Acids 2015; 48:535-47. [PMID: 26439377 DOI: 10.1007/s00726-015-2107-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 09/24/2015] [Indexed: 11/29/2022]
Abstract
Hymenochirin-1b (Hym-1B; IKLSPETKDNLKKVLKGAIKGAIAVAKMV.NH2) is a cationic, α-helical amphibian host-defense peptide with antimicrobial, anticancer, and immunomodulatory properties. This study investigates the abilities of the peptide and nine analogues containing substitutions of Pro(5), Glu(6), and Asp(9) by either L-lysine or D-lysine to stimulate insulin release in vitro using BRIN-BD11 clonal β cells or isolated mouse islets and in vivo using mice fed a high-fat diet to produce obesity and insulin resistance. Hym-1B produced a significant and concentration-dependent increase in the rate of insulin release from BRIN-BD11 cells without cytotoxicity at concentrations up to 1 µM with a threshold concentration of 1 nM. The threshold concentrations for the analogues were: [P5K], [E6K], [D9K], [P5K, E6K] and [E6K, D9k] 0.003 nM, [E6K, D9K] and [D9k] 0.01 nM, [P5K, D9K] 0.1 nM and [E6k] 0.3 nM. All peptides displayed cytotoxicity at concentrations ≥1 µM except the [P5K] and [D9k] analogues which were non-toxic at 3 µM. The potency and maximum rate of insulin release from mouse islets produced by the [P5K] peptide were significantly greater than produced by Hym-1B. Neither Hym-1B nor the [P5K] analogue at 1 µM concentration had an effect on membrane depolarization or intracellular Ca(2+). The [P5K] analogue (1 µM) produced a significant increase in cAMP concentration in BRIN-BD11 cells and stimulated GLP-1 secretion from GLUTag cells. Down-regulation of the protein kinase A pathway by overnight incubation with forskolin completely abolished the insulin-releasing effects of [P5K]hym-1B. Intraperitoneal administration of the [P5K] and [D9k] analogues (75 nmol/kg body weight) to high-fat-fed mice with insulin resistance significantly enhanced glucose tolerance with a concomitant increase in insulin secretion. We conclude that [P5K]hym-1B and [D9k]hym-1B show potential for development into anti-diabetic agents.
Collapse
Affiliation(s)
- Bosede O Owolabi
- SAAD Centre for Pharmacy and Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, Northern Ireland, UK
| | - Opeolu O Ojo
- SAAD Centre for Pharmacy and Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, Northern Ireland, UK
- School of Health, Sport and Bioscience, University of East London, Stratford, E15 4LZ, UK
| | - Dinesh K Srinivasan
- SAAD Centre for Pharmacy and Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, Northern Ireland, UK
| | - J Michael Conlon
- SAAD Centre for Pharmacy and Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, Northern Ireland, UK
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, Northern Ireland, UK
| | - Yasser H A Abdel-Wahab
- SAAD Centre for Pharmacy and Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, Northern Ireland, UK.
| |
Collapse
|
74
|
Yamashita S, Ohashi K, Utoh R, Okano T, Yamamoto M. Human Laminin Isotype Coating for Creating Islet Cell Sheets. CELL MEDICINE 2015; 8:39-46. [PMID: 26858907 DOI: 10.3727/215517915x689029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Our experimental approach toward the development of new islet-based treatment for diabetes mellitus has been the creation of a monolayered islet cell construct (islet cell sheet), followed by its transplantation into a subcutaneous pocket. Previous studies describe rat laminin-5 (chain composition: α3, β3, γ2) as a suitable extracellular matrix (ECM) for surfaces comprised of a coated temperature-responsive polymer, poly(N-isopropylacrylamide) (PIPAAm). To progress toward the clinical application of this approach, the present study attempted to identify an optimal human ECM as a coating material on PIPAAm surfaces, which allowed islet cells to attach on the surfaces and subsequently to be harvested as a monolithic cell sheet. Dispersed rat islet cells were seeded onto PIPAAm dishes coated with various human laminin isotypes: human laminin (HL)-211, HL-332, HL-411, HL-511, and HL-placenta. Plating efficiency at day 1, the confluency at day 3, and glucose-stimulated insulin secretion test at day 3 were performed. The highest value of plating efficiency was found in the HL-332-PIPAAm group (83.1 ± 0.7%). The HL-332-PIPAAm group also showed the highest cellular confluency (98.6 ± 0.5%). Islet cells cultured on the HL-332-PIPAAm surfaces showed a positive response in the glucose-stimulated insulin secretion test. By reducing culture temperature from 37°C to 20°C in the HL-332-PIPAAm group, cells were able to be harvested as a monolithic islet sheet. The present study showed that HL-332 was an optimal human-derived ECM on a PIPAAm coating for preparing islet cell sheets.
Collapse
Affiliation(s)
- Shingo Yamashita
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan; †Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Kazuo Ohashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan; †Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan; ‡iPS Cell-based Projects on Cell Transplantation and Cell Dynamics, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Rie Utoh
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University , Shinjuku-ku, Tokyo , Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University , Shinjuku-ku, Tokyo , Japan
| | - Masakazu Yamamoto
- † Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University , Shinjuku-ku, Tokyo , Japan
| |
Collapse
|
75
|
Nerve Growth Factor Improves Survival and Function of Transplanted Islets Via TrkA-mediated β Cell Proliferation and Revascularization. Transplantation 2015; 99:1132-43. [PMID: 25806408 DOI: 10.1097/tp.0000000000000655] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Nerve growth factor (NGF), which plays important roles in promoting growth and differentiation of nerve cells, has recently been reported as a regulator in pancreatic β cells in terms of insulin releasing function. In this study, we examined whether NGF stimulation would promote islet graft survival and function in islet transplantation. METHODS We found that supplementation of cultured islets with NGF improved the viability of islet cells and induced the production of insulin, vascular endothelial growth factor, and cellular proliferative markers. Because a specific inhibitor of TrkA, K252a, blocked all these effects, we propose that the TrkA receptor is the mediator of NGF stimulation. RESULTS After transplantation to the kidney subcapsule and liver of syngenic diabetic mice, a higher rate of normoglycemic achievement, increased serum insulin, and improved glucose tolerance were observed in the mice transplanted with NGF-pretreated islet grafts. Histological analysis revealed higher expression of insulin and vascular endothelial growth factor, an increase in proliferative β cells, and revascularization in NGF-pretreated islet grafts without activation of any inflammatory cells. CONCLUSIONS The NGF treatment can therefore serve as a new and promising therapeutic tool for improving islet graft viability and function in islet transplantation.
Collapse
|
76
|
Hata T, Sakata N, Yoshimatsu G, Tsuchiya H, Fukase M, Ishida M, Aoki T, Katayose Y, Egawa S, Unno M. Cholestatic Liver Injury After Biliary Reconstruction Impairs Transplanted Islet Viability and Function. Am J Transplant 2015; 15:2085-95. [PMID: 25908212 DOI: 10.1111/ajt.13266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 02/03/2015] [Accepted: 02/11/2015] [Indexed: 01/25/2023]
Abstract
Islet autotransplantation following total pancreatectomy differs from allograft transplantation with respect to the requirement of biliary reconstruction. Although it is known that careful consideration should be given to postoperative cholestatic liver injury after biliary reconstruction, its direct effects on transplanted islets have not been completely elucidated. In this study, we developed a murine model of postoperative cholestatic liver injury after biliary reconstruction with islet autotransplantation that involved syngeneic intraportal islet transplantation into chemically induced diabetic mice and common bile duct ligation. We assessed the viability and function of the transplanted islets. The impaired viability of transplanted islets and increased blood glucose levels indicated restoration of the diabetic state after common bile duct ligation in this murine model. Furthermore, impaired islet viability and function occurred earlier in the transplanted islets than in the surrounding liver tissues, which was consistent with the faster and higher expression of oxidative stress markers in the transplanted islets. Transplanted islets may be more vulnerable to oxidative stress caused by cholestatic liver injury than the surrounding liver tissue. Therefore, patients should be intensively managed after total pancreatectomy with islet autotransplantation to preserve viability and function of the transplanted islets.
Collapse
Affiliation(s)
- T Hata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - N Sakata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - G Yoshimatsu
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - H Tsuchiya
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - M Fukase
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - M Ishida
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - T Aoki
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Y Katayose
- Division of Integrated Surgery and Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - S Egawa
- Division of International Cooperation for Disaster Medicine, International Research Institute of Disaster Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - M Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan.,Division of Integrated Surgery and Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
77
|
Hamano K, Nakagawa Y, Ohtsu Y, Li L, Medina J, Tanaka Y, Masuda K, Komatsu M, Kojima I. Lactisole inhibits the glucose-sensing receptor T1R3 expressed in mouse pancreatic β-cells. J Endocrinol 2015; 226:57-66. [PMID: 25994004 DOI: 10.1530/joe-15-0102] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2015] [Indexed: 11/08/2022]
Abstract
Glucose activates the glucose-sensing receptor T1R3 and facilitates its own metabolism in pancreatic β-cells. An inhibitor of this receptor would be helpful in elucidating the physiological function of the glucose-sensing receptor. The present study was conducted to examine whether or not lactisole can be used as an inhibitor of the glucose-sensing receptor. In MIN6 cells, in a dose-dependent manner, lactisole inhibited insulin secretion induced by sweeteners, acesulfame-K, sucralose and glycyrrhizin. The IC50 was ∼4 mmol/l. Lactisole attenuated the elevation of cytoplasmic Ca2+ concentration ([Ca2+]c) evoked by sucralose and acesulfame-K but did not affect the elevation of intracellular cAMP concentration ([cAMP]c) induced by these sweeteners. Lactisole also inhibited the action of glucose in MIN6 cells. Thus, lactisole significantly reduced elevations of intracellular [NADH] and intracellular [ATP] induced by glucose, and also inhibited glucose-induced insulin secretion. To further examine the effect of lactisole on T1R3, we prepared HEK293 cells stably expressing mouse T1R3. In these cells, sucralose elevated both [Ca2+]c and [cAMP]c. Lactisole attenuated the sucralose-induced increase in [Ca2+]c but did not affect the elevation of [cAMP]c. Finally, lactisole inhibited insulin secretion induced by a high concentration of glucose in mouse islets. These results indicate that the mouse glucose-sensing receptor was inhibited by lactisole. Lactisole may be useful in assessing the role of the glucose-sensing receptor in mouse pancreatic β-cells.
Collapse
Affiliation(s)
- Kunihisa Hamano
- Institute for Molecular and Cellular RegulationGunma University, Maebashi 371-8512, JapanDepartment of General MedicineNational Defense Medical College, Tokorozawa, JapanSuntory Institute for Bioorganic ResearchOsaka, JapanDepartment of Internal MedicineShinshu University School of Medicine, Matsumoto, Japan Institute for Molecular and Cellular RegulationGunma University, Maebashi 371-8512, JapanDepartment of General MedicineNational Defense Medical College, Tokorozawa, JapanSuntory Institute for Bioorganic ResearchOsaka, JapanDepartment of Internal MedicineShinshu University School of Medicine, Matsumoto, Japan
| | - Yuko Nakagawa
- Institute for Molecular and Cellular RegulationGunma University, Maebashi 371-8512, JapanDepartment of General MedicineNational Defense Medical College, Tokorozawa, JapanSuntory Institute for Bioorganic ResearchOsaka, JapanDepartment of Internal MedicineShinshu University School of Medicine, Matsumoto, Japan
| | - Yoshiaki Ohtsu
- Institute for Molecular and Cellular RegulationGunma University, Maebashi 371-8512, JapanDepartment of General MedicineNational Defense Medical College, Tokorozawa, JapanSuntory Institute for Bioorganic ResearchOsaka, JapanDepartment of Internal MedicineShinshu University School of Medicine, Matsumoto, Japan
| | - Longfei Li
- Institute for Molecular and Cellular RegulationGunma University, Maebashi 371-8512, JapanDepartment of General MedicineNational Defense Medical College, Tokorozawa, JapanSuntory Institute for Bioorganic ResearchOsaka, JapanDepartment of Internal MedicineShinshu University School of Medicine, Matsumoto, Japan
| | - Johan Medina
- Institute for Molecular and Cellular RegulationGunma University, Maebashi 371-8512, JapanDepartment of General MedicineNational Defense Medical College, Tokorozawa, JapanSuntory Institute for Bioorganic ResearchOsaka, JapanDepartment of Internal MedicineShinshu University School of Medicine, Matsumoto, Japan
| | - Yuji Tanaka
- Institute for Molecular and Cellular RegulationGunma University, Maebashi 371-8512, JapanDepartment of General MedicineNational Defense Medical College, Tokorozawa, JapanSuntory Institute for Bioorganic ResearchOsaka, JapanDepartment of Internal MedicineShinshu University School of Medicine, Matsumoto, Japan
| | - Katsuyoshi Masuda
- Institute for Molecular and Cellular RegulationGunma University, Maebashi 371-8512, JapanDepartment of General MedicineNational Defense Medical College, Tokorozawa, JapanSuntory Institute for Bioorganic ResearchOsaka, JapanDepartment of Internal MedicineShinshu University School of Medicine, Matsumoto, Japan
| | - Mitsuhisa Komatsu
- Institute for Molecular and Cellular RegulationGunma University, Maebashi 371-8512, JapanDepartment of General MedicineNational Defense Medical College, Tokorozawa, JapanSuntory Institute for Bioorganic ResearchOsaka, JapanDepartment of Internal MedicineShinshu University School of Medicine, Matsumoto, Japan
| | - Itaru Kojima
- Institute for Molecular and Cellular RegulationGunma University, Maebashi 371-8512, JapanDepartment of General MedicineNational Defense Medical College, Tokorozawa, JapanSuntory Institute for Bioorganic ResearchOsaka, JapanDepartment of Internal MedicineShinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
78
|
Over-nutrient environment during both prenatal and postnatal development increases severity of islet injury, hyperglycemia, and metabolic disorders in the offspring. J Physiol Biochem 2015; 71:391-403. [PMID: 26048534 DOI: 10.1007/s13105-015-0419-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/28/2015] [Indexed: 12/18/2022]
Abstract
Prenatal and postnatal over-nutrition has emerged as a new health issue contributing to metabolic disorders in early development of the offspring. Accumulating evidence has suggested that adverse prenatal and postnatal environments gave rise to the predisposition to metabolic syndromes including hyperglycemia, obesity, and diabetes. However, little research has concentrated on the effects of exposures to both adverse conditions before and after birth of the offspring. In this study, we aimed to investigate whether prenatal and postnatal over-nutrition is able to cause metabolic disorders to female mice feed on high-fat/fructose diet (HFFD) as well as their offspring. Female mice were fed on either HFFD or a normal chow diet (NC), while their offspring were divided into four experimental groups as NC/NC, HFFD/NC, NC/HFFD, and HFFD/HFFD (prenatal/postnatal diet order), respectively. Both NC/HFFD and HFFD/HFFD offspring exhibited obvious body weight and fat content gain, hyperglycemia, and severe insulin resistance. Interestingly, when compared to NC/HFFD offspring, the HFFD/HFFD offspring exhibited more severe alterations in their metabolism and dysfunctions on pancreatic β-cells, suggesting a potential impact of prenatal HFFD on the programming of pancreatic β-cell deficiency in the fetus. Meanwhile, the results from HFFD/NC mice indicated that a balance diet after birth partially compensated the adverse prenatal HFFD impact. In conclusion, this study demonstrated that prenatal and postnatal over-nutrition increases severity of islet injury, hyperglycemia, and metabolic disorders in the offspring.
Collapse
|
79
|
Sakata N, Sax N, Yoshimatsu G, Tsuchiya H, Kato S, Aoki T, Ishida M, Katayose Y, Egawa S, Kodama T, Unno M. Enhanced ultrasonography using a nano/microbubble contrast agent for islet transplantation. Am J Transplant 2015; 15:1531-42. [PMID: 25846610 DOI: 10.1111/ajt.13152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 11/30/2014] [Accepted: 12/09/2014] [Indexed: 01/25/2023]
Abstract
Recent basic and clinical studies have assessed the use of highly sensitive imaging modalities for visualizing transplanted islets. We investigated the utility of enhanced ultrasonography, combined with fluorescent acoustic liposome nano/microbubbles (FALs), for evaluating angiogenesis and the endocrine function of transplanted islets. BALB/c mice were classified into three groups: Diabetic mice that underwent syngeneic islet transplantation into the subrenal capsule and achieved normoglycemia (Tx group); those that failed to achieve normoglycemia (Tx-DM group); and those not receiving any treatment (DM group). Mice were examined by FAL-enhanced high frequency ultrasonography. The echogenicity of the islets increased rapidly within the first minute after injection of FALs and remained at a higher level in the Tx group, while small increases were observed in the other two groups. In histological assessments, fluorescently stained erythrocytes could be seen in and around the transplanted islets, indicating that the transplanted islets were enhanced by infusion of FALs via vessel networks between the engrafted islets and tissue. Furthermore, the echogenicity correlated significantly with endocrine parameters, including blood glucose (BG), serum insulin, and the BG change in the glucose tolerance test. In conclusion, the echogenicity of the islets under FAS-enhanced ultrasonosonography correlated with the endocrine status of transplanted islets.
Collapse
Affiliation(s)
- N Sakata
- Department of Surgery, Tohoku University, Sendai, Japan
| | - N Sax
- Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - G Yoshimatsu
- Department of Surgery, Tohoku University, Sendai, Japan
| | - H Tsuchiya
- Department of Surgery, Tohoku University, Sendai, Japan
| | - S Kato
- Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - T Aoki
- Department of Surgery, Tohoku University, Sendai, Japan
| | - M Ishida
- Department of Surgery, Tohoku University, Sendai, Japan
| | - Y Katayose
- Department of Surgery, Tohoku University, Sendai, Japan.,Division of Integrated Surgery and Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - S Egawa
- Division of International Cooperation for Disaster Medicine, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - T Kodama
- Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - M Unno
- Department of Surgery, Tohoku University, Sendai, Japan
| |
Collapse
|
80
|
Yamada T, Cavelti-Weder C, Caballero F, Lysy PA, Guo L, Sharma A, Li W, Zhou Q, Bonner-Weir S, Weir GC. Reprogramming Mouse Cells With a Pancreatic Duct Phenotype to Insulin-Producing β-Like Cells. Endocrinology 2015; 156:2029-38. [PMID: 25836667 PMCID: PMC4430605 DOI: 10.1210/en.2014-1987] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Reprogramming technology has opened the possibility of converting one cell type into another by forced expression of transgenes. Transduction of adenoviral vectors encoding 3 pancreatic transcription factors, Pdx1, Ngn3, and MafA, into mouse pancreas results in direct reprogramming of exocrine cells to insulin-producing β-like cells. We hypothesized that cultured adult pancreatic duct cells could be reprogrammed to become insulin-producing β-cells by adenoviral-mediated expression of this same combination of factors. Exocrine were isolated from adult mouse insulin 1 promoter (MIP)-green fluorescent protein (GFP) transgenic mice to allow new insulin-expressing cells to be detected by GFP fluorescence. Cultured cells were transduced by an adenoviral vector carrying a polycistronic construct Ngn3/Pdx1/MafA/mCherry (Ad-M3C) or mCherry sequence alone as a control vector. In addition, the effects of glucagon-like peptide-1 (GLP-1) receptor agonist, exendin-4 (Ex-4) on the reprogramming process were examined. GFP(+) cells appeared 2 days after Ad-M3C transduction; the reprogramming efficiency was 8.6 ± 2.6% by day 4 after transduction. Ad-M3C also resulted in increased expression of β-cell markers insulin 1 and 2, with enhancement by Ex-4. Expression of other β-cell markers, neuroD and GLP-1 receptor, were also significantly up-regulated. The amount of insulin release into the media and insulin content of the cells were significantly higher in the Ad-M3C-transduced cells; this too was enhanced by Ex-4. The transduced cells did not secrete insulin in response to increased glucose, indicating incomplete differentiation to β-cells. Thus, cultured murine adult pancreatic cells with a duct phenotype can be directly reprogrammed to insulin-producing β-like cells by adenoviral delivery of 3 pancreatic transcription factors.
Collapse
Affiliation(s)
- Takatsugu Yamada
- Section on Islet Cell and Regenerative Biology (T.Y., C.C.-W., F.C., P.A.L., L.G., A.S., S.B.-W., G.C.W.), Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215; and Department of Stem Cell and Regenerative Biology (W.L., Q.Z.), Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138
| | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Diethyldithiocarbamate-mediated zinc ion chelation reveals role of Cav2.3 channels in glucagon secretion. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:953-64. [DOI: 10.1016/j.bbamcr.2015.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/28/2014] [Accepted: 01/03/2015] [Indexed: 12/13/2022]
|
82
|
Zhou J, Xu G, Yan J, Li K, Bai Z, Cheng W, Huang K. Rehmannia glutinosa (Gaertn.) DC. polysaccharide ameliorates hyperglycemia, hyperlipemia and vascular inflammation in streptozotocin-induced diabetic mice. JOURNAL OF ETHNOPHARMACOLOGY 2015; 164:229-38. [PMID: 25698243 DOI: 10.1016/j.jep.2015.02.026] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 01/19/2015] [Accepted: 02/09/2015] [Indexed: 05/22/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rehmannia glutinosa (Gaertn.) DC. (RG) has been widely used as traditional Chinese herbal medicine for treatment of diabetes and its complications. The polysaccharide fraction of RG has been proposed to possess hypoglycemic effect by intraperitoneal administration, however, the mechanisms responsible for the hypoglycemic effect of RG polysaccharide (RGP) remain poorly understood. Here we studied the anti-hyperglycemic and anti-hyperlipidemic effect of oral administration of a purified RGP and its underlying mechanisms in streptozotocin (STZ)-induced diabetic mice. MATERIALS AND METHODS The preliminary structure of RGP was determined by GC and FT-IR. Mice were injected with STZ to induce type 1 diabetes. RGP at doses of 20, 40 and 80 mg/kg/day was orally administered to mice for 4 weeks, and metformin was used as positive control. After 4 weeks, the blood biochemical parameters, the pancreatic insulin contents, in vitro insulin secretion, the hepatic glycogen contents and mRNA expression of phosphoenolpyruvate carboxyl kinase (PEPCK) were assayed. RESULTS RGP was composed of rhamnose, arabinose, mannose, glucose and galactose in the molar ratio of 1.00:1.26:0.73:16.45:30.40 with the average molecular weight of 63.5 kDa. RGP administration significantly decreased the blood levels of glucose, total cholesterol, triglycerides, low density lipoprotein-cholesterol, and increased the blood levels of high density lipoprotein-cholesterol and insulin in diabetic mice, concurrent with increases in body weights and pancreatic insulin contents. The in vitro study revealed that RGP significantly enhanced both basal and glucose-stimulated insulin secretions, as well as islet insulin contents in the pancreatic islets of diabetic mice. Moreover, RGP reversed the increased mRNA expression of PEPCK and the reduced glycogen contents in the liver of diabetic mice. Furthermore, RGP exhibited potent anti-inflammatory and anti-oxidative activities, as evidenced by the decreased blood levels of TNF-α, IL-6, monocyte chemoattractant protein-1, MDA, and also the elevated blood levels of SOD and GPx activities in diabetic mice. CONCLUSIONS Taken together, RGP can effectively ameliorate hyperglycemia, hyperlipemia, vascular inflammation and oxidative stress in STZ-induced diabetic mice, and thus may be a potential therapeutic option for type 1 diabetes.
Collapse
Affiliation(s)
- Jun Zhou
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, PR China.
| | - Gang Xu
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, PR China
| | - Junyan Yan
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, PR China
| | - Kaicheng Li
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, PR China
| | - Zhaoshuai Bai
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, PR China
| | - Weinan Cheng
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, PR China
| | - Kaixun Huang
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, PR China.
| |
Collapse
|
83
|
Kooptiwut S, Hanchang W, Semprasert N, Junking M, Limjindaporn T, Yenchitsomanus PT. Testosterone reduces AGTR1 expression to prevent β-cell and islet apoptosis from glucotoxicity. J Endocrinol 2015; 224:215-24. [PMID: 25512346 DOI: 10.1530/joe-14-0397] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hypogonadism in men is associated with an increased incidence of type 2 diabetes. Supplementation with testosterone has been shown to protect pancreatic β-cell against apoptosis due to toxic substances including streptozotocin and high glucose. One of the pathological mechanisms of glucose-induced pancreatic β-cell apoptosis is the induction of the local rennin-angiotensin-aldosterone system (RAAS). The role of testosterone in regulation of the pancreatic RAAS is still unknown. This study aims to investigate the protective action of testosterone against glucotoxicity-induced pancreatic β-cell apoptosis via alteration of the pancreatic RAAS pathway. Rat insulinoma cell line (INS-1) cells or isolated male mouse islets were cultured in basal and high-glucose media in the presence or absence of testosterone, losartan, and angiotensin II (Ang II), then cell apoptosis, cleaved caspase 3 expression, oxidative stress, and expression of angiotensin II type 1 receptor (AGTR1) and p47(phox) mRNA and protein were measured. Testosterone and losartan showed similar effects in reducing pancreatic β-cell apoptosis. Testosterone significantly reduced expression of AGTR1 protein in INS-1 cells cultured in high-glucose medium or high-glucose medium with Ang II. Testosterone decreased the expression of AGTR1 and p47(phox) mRNA and protein in comparison with levels in cells cultured in high-glucose medium alone. Furthermore, testosterone attenuated superoxide production when co-cultured with high-glucose medium. In contrast, when cultured in basal glucose, supplementation of testosterone did not have any effect on cell apoptosis, oxidative stress, and expression of AGT1R and p47(phox). In addition, high-glucose medium did not increase cleaved caspase 3 in AGTR1 knockdown experiments. Thus, our results indicated that testosterone prevents pancreatic β-cell apoptosis due to glucotoxicity through reduction of the expression of ATGR1 and its signaling pathway.
Collapse
Affiliation(s)
- Suwattanee Kooptiwut
- Department of PhysiologyDepartment of AnatomyDivision of Molecular MedicineDepartment of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Wanthanee Hanchang
- Department of PhysiologyDepartment of AnatomyDivision of Molecular MedicineDepartment of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Namoiy Semprasert
- Department of PhysiologyDepartment of AnatomyDivision of Molecular MedicineDepartment of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Mutita Junking
- Department of PhysiologyDepartment of AnatomyDivision of Molecular MedicineDepartment of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Thawornchai Limjindaporn
- Department of PhysiologyDepartment of AnatomyDivision of Molecular MedicineDepartment of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Pa-thai Yenchitsomanus
- Department of PhysiologyDepartment of AnatomyDivision of Molecular MedicineDepartment of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
84
|
Yoshimatsu G, Sakata N, Tsuchiya H, Minowa T, Takemura T, Morita H, Hata T, Fukase M, Aoki T, Ishida M, Motoi F, Naitoh T, Katayose Y, Egawa S, Unno M. The co-transplantation of bone marrow derived mesenchymal stem cells reduced inflammation in intramuscular islet transplantation. PLoS One 2015; 10:e0117561. [PMID: 25679812 PMCID: PMC4332659 DOI: 10.1371/journal.pone.0117561] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 12/27/2014] [Indexed: 12/12/2022] Open
Abstract
Aims/Hypothesis Although the muscle is one of the preferable transplant sites in islet transplantation, its transplant efficacy is poor. Here we attempted to determine whether an intramuscular co-transplantation of mesenchymal stem cells (MSCs) could improve the outcome. Methods We co-cultured murine islets with MSCs and then analyzed the morphological changes, viability, insulin-releasing function (represented by the stimulation index), and gene expression of the islets. We also transplanted 500 islets intramuscularly with or without 5 × 105 MSCs to diabetic mice and measured their blood glucose level, the glucose changes in an intraperitoneal glucose tolerance test, and the plasma IL-6 level. Inflammation, apoptosis, and neovascularization in the transplantation site were evaluated histologically. Results The destruction of islets tended to be prevented by co-culture with MSCs. The stimulation index was significantly higher in islets co-cultured with MSCs (1.78 ± 0.59 vs. 7.08 ± 2.53; p = 0.0025). In terms of gene expression, Sult1c2, Gstm1, and Rab37 were significantly upregulated in islets co-cultured with MSCs. Although MSCs were effective in the in vitro assays, they were only partially effective in facilitating intramuscular islet transplantation. Co-transplanted MSCs prevented an early inflammatory reaction from the islets (plasma IL-6; p = 0.0002, neutrophil infiltration; p = 0.016 inflammatory area; p = 0.021), but could not promote neovascularization in the muscle, resulting in the failure of many intramuscular transplanted islets to engraft. Conclusions In conclusion, co-culturing and co-transplanting MSCs is potentially useful in islet transplantation, especially in terms of anti-inflammation, but further augmentation for an anti-apoptosis effect and neovascularization is necessary.
Collapse
Affiliation(s)
- Gumpei Yoshimatsu
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoaki Sakata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Haruyuki Tsuchiya
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Minowa
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Japan
| | - Taro Takemura
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Japan
| | - Hiromi Morita
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Japan
| | - Tatsuo Hata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masahiko Fukase
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeshi Aoki
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masaharu Ishida
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fuyuhiko Motoi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeshi Naitoh
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu Katayose
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan; Division of Integrated Surgery and Oncology, Tohoku University Hospital, Sendai, Japan
| | - Shinichi Egawa
- Division of International Cooperation for Disaster Medicine, Tohoku University, Sendai, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
85
|
Ojo OO, Srinivasan DK, Owolabi BO, Flatt PR, Abdel-Wahab YH. Beneficial effects of tigerinin-1R on glucose homeostasis and beta cell function in mice with diet-induced obesity-diabetes. Biochimie 2015; 109:18-26. [DOI: 10.1016/j.biochi.2014.11.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/28/2014] [Indexed: 11/29/2022]
|
86
|
Li S, Vaziri ND, Masuda Y, Hajighasemi-Ossareh M, Robles L, Le A, Vo K, Chan JY, Foster CE, Stamos MJ, Ichii H. Pharmacological activation of Nrf2 pathway improves pancreatic islet isolation and transplantation. Cell Transplant 2015; 24:2273-83. [PMID: 25581574 DOI: 10.3727/096368915x686210] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Oxidative stress is a major cause of islet damage and loss during the islet isolation process. The Nrf2 pathway plays a critical role in protecting the cells against oxidative stress. The aim of this study was to investigate the effect of an Nrf2 activator (dh404) on islet isolation and transplantation in a rodent model. Islet isolation was conducted using Nrf2-deficient and wild-type mice and vehicle-treated and Nrf2 activator (dh404)-treated rats. Islet yield, viability, and Nrf2 pathway activity were determined. An in vivo islet potency test was done. Islet yield and viability in Nrf2-deficient mice was significantly lower compared to wild-type (p < 0.05) mice. Furthermore, administration of dh404 to normal Sprague-Dawley rats enhanced nuclear translocation of Nrf2 and elevated HO-1 expression in the pancreas. Islet yield and viability in dh404-treated rats was significantly higher compared to the vehicle-treated group (p < 0.05). The diabetes cure rate in nude mice with chemically induced diabetes was significantly greater in those transplanted with islets from the dh404-treated group (6/9) than vehicle-treated rats (2/9, p < 0.05). The Nrf2 pathway plays a significant role in protecting islets against stress caused by the isolation process. Pharmacological activation of the Nrf2 pathway significantly increased HO-1 expression, improved islet yield, viability, and function after transplantation.
Collapse
Affiliation(s)
- Shiri Li
- Department of Surgery, University of California, Irvine, CA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Ojo O, Srinivasan D, Owolabi B, Conlon J, Flatt P, Abdel-Wahab Y. Magainin-AM2 improves glucose homeostasis and beta cell function in high-fat fed mice. Biochim Biophys Acta Gen Subj 2015; 1850:80-7. [DOI: 10.1016/j.bbagen.2014.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/22/2014] [Accepted: 10/13/2014] [Indexed: 01/01/2023]
|
88
|
Rady B, Chen Y, Vaca P, Wang Q, Wang Y, Salmon P, Oberholzer J. Overexpression ofE2F3promotes proliferation of functional human β cells without induction of apoptosis. Cell Cycle 2014; 12:2691-702. [DOI: 10.4161/cc.25834] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
89
|
Johnson JS, Kono T, Tong X, Yamamoto WR, Zarain-Herzberg A, Merrins MJ, Satin LS, Gilon P, Evans-Molina C. Pancreatic and duodenal homeobox protein 1 (Pdx-1) maintains endoplasmic reticulum calcium levels through transcriptional regulation of sarco-endoplasmic reticulum calcium ATPase 2b (SERCA2b) in the islet β cell. J Biol Chem 2014; 289:32798-810. [PMID: 25271154 DOI: 10.1074/jbc.m114.575191] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Although the pancreatic duodenal homeobox 1 (Pdx-1) transcription factor is known to play an indispensable role in β cell development and secretory function, recent data also implicate Pdx-1 in the maintenance of endoplasmic reticulum (ER) health. The sarco-endoplasmic reticulum Ca(2+) ATPase 2b (SERCA2b) pump maintains a steep Ca(2+) gradient between the cytosol and ER lumen. In models of diabetes, our data demonstrated loss of β cell Pdx-1 that occurs in parallel with altered SERCA2b expression, whereas in silico analysis of the SERCA2b promoter revealed multiple putative Pdx-1 binding sites. We hypothesized that Pdx-1 loss under inflammatory and diabetic conditions leads to decreased SERCA2b levels and activity with concomitant alterations in ER health. To test this, siRNA-mediated knockdown of Pdx-1 was performed in INS-1 cells. The results revealed reduced SERCA2b expression and decreased ER Ca(2+), which was measured using fluorescence lifetime imaging microscopy. Cotransfection of human Pdx-1 with a reporter fused to the human SERCA2 promoter increased luciferase activity 3- to 4-fold relative to an empty vector control, and direct binding of Pdx-1 to the proximal SERCA2 promoter was confirmed by chromatin immunoprecipitation. To determine whether restoration of SERCA2b could rescue ER stress induced by Pdx-1 loss, Pdx1(+/-) mice were fed a high-fat diet. Isolated islets demonstrated an increased spliced-to-total Xbp1 ratio, whereas SERCA2b overexpression reduced the Xbp1 ratio to that of wild-type controls. Together, these results identify SERCA2b as a novel transcriptional target of Pdx-1 and define a role for altered ER Ca(2+) regulation in Pdx-1-deficient states.
Collapse
Affiliation(s)
| | | | - Xin Tong
- Cellular and Integrative Physiology and
| | | | - Angel Zarain-Herzberg
- the Departamento de Bioquimica, Facultad de Medicina, National Autonomous University of México, México City, 04510 México
| | - Matthew J Merrins
- the Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and Department of Biomolecular Chemistry, University of Wisconsin Madison School of Medicine and Public Health, Madison, Wisconsin 53705
| | - Leslie S Satin
- the Department of Pharmacology and Brehm Center for Diabetes Research, University of Michigan Medical School, Ann Arbor, Michigan 48105
| | - Patrick Gilon
- the Pôle d'Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, 1348 Belgium, and
| | - Carmella Evans-Molina
- From the Departments of Biochemistry and Molecular Biology, Medicine, and Cellular and Integrative Physiology and the Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, the Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana 46202
| |
Collapse
|
90
|
Robles L, Vaziri ND, Li S, Masuda Y, Takasu C, Takasu M, Vo K, Farzaneh SH, Stamos MJ, Ichii H. Dimethyl fumarate protects pancreatic islet cells and non-endocrine tissue in L-arginine-induced chronic pancreatitis. PLoS One 2014; 9:e107111. [PMID: 25198679 PMCID: PMC4157838 DOI: 10.1371/journal.pone.0107111] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 08/06/2014] [Indexed: 12/12/2022] Open
Abstract
Background Chronic pancreatitis (CP) is a progressive disorder resulting in the destruction and fibrosis of the pancreatic parenchyma which ultimately leads to impairment of the endocrine and exocrine functions. Dimethyl Fumarate (DMF) was recently approved by FDA for treatment of patients with multiple sclerosis. DMF's unique anti-oxidant and anti-inflammatory properties make it an interesting drug to test on other inflammatory conditions. This study was undertaken to determine the effects of DMF on islet cells and non-endocrine tissue in a rodent model of L-Arginine-induced CP. Methods Male Wistar rats fed daily DMF (25 mg/kg) or vehicle by oral gavage were given 5 IP injections of L-Arginine (250 mg/100 g×2, 1 hr apart). Rats were assessed with weights and intra-peritoneal glucose tolerance tests (IPGTT, 2 g/kg). Islets were isolated and assessed for islet mass and viability with flow cytometry. Non-endocrine tissue was assessed for histology, myeloperoxidase (MPO), and lipid peroxidation level (MDA). In vitro assessments included determination of heme oxygenase (HO-1) protein expression by Western blot. Results Weight gain was significantly reduced in untreated CP group at 6 weeks. IPGTT revealed significant impairment in untreated CP group and its restoration with DMF therapy (P <0.05). Untreated CP rats had pancreatic atrophy, severe acinar architectural damage, edema, and fatty infiltration as well as elevated MDA and MPO levels, which were significantly improved by DMF treatment. After islet isolation, the volume of non-endocrine tissue was significantly smaller in untreated CP group. Although islet counts were similar in the two groups, islet viability was significantly reduced in untreated CP group and improved with DMF treatment. In vitro incubation of human pancreatic tissue with DMF significantly increased HO-1 expression. Conclusion Administration of DMF attenuated L-Arginine-induced CP and islet function in rats. DMF treatment could be a possible strategy to improve clinical outcome in patients with CP.
Collapse
Affiliation(s)
- Lourdes Robles
- Departments of Surgery and Medicine, University of California Irvine, Irvine, CA, United States of America
| | - Nosratola D. Vaziri
- Departments of Surgery and Medicine, University of California Irvine, Irvine, CA, United States of America
| | - Shiri Li
- Departments of Surgery and Medicine, University of California Irvine, Irvine, CA, United States of America
| | - Yuichi Masuda
- Departments of Surgery and Medicine, University of California Irvine, Irvine, CA, United States of America
| | - Chie Takasu
- Departments of Surgery and Medicine, University of California Irvine, Irvine, CA, United States of America
| | - Mizuki Takasu
- Departments of Surgery and Medicine, University of California Irvine, Irvine, CA, United States of America
| | - Kelly Vo
- Departments of Surgery and Medicine, University of California Irvine, Irvine, CA, United States of America
| | - Seyed H. Farzaneh
- Departments of Surgery and Medicine, University of California Irvine, Irvine, CA, United States of America
| | - Michael J. Stamos
- Departments of Surgery and Medicine, University of California Irvine, Irvine, CA, United States of America
| | - Hirohito Ichii
- Departments of Surgery and Medicine, University of California Irvine, Irvine, CA, United States of America
- * E-mail:
| |
Collapse
|
91
|
Andreassen KV, Feigh M, Hjuler ST, Gydesen S, Henriksen JE, Beck-Nielsen H, Christiansen C, Karsdal MA, Henriksen K. A novel oral dual amylin and calcitonin receptor agonist (KBP-042) exerts antiobesity and antidiabetic effects in rats. Am J Physiol Endocrinol Metab 2014; 307:E24-33. [PMID: 24801386 DOI: 10.1152/ajpendo.00121.2014] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study investigated a novel oral dual amylin and calcitonin receptor agonist (DACRA), KBP-042, in head-to-head comparison with salmon calcitonin (sCT) with regard to in vitro receptor pharmacology, ex vivo pancreatic islet studies, and in vivo proof of concept studies in diet-induced obese (DIO) and Zucker diabetic fatty (ZDF) rats. In vitro, KBP-042 demonstrated superior binding affinity and activation of amylin and calcitonin receptors, and ex vivo, KBP-042 exerted inhibitory action on stimulated insulin and glucagon release from isolated islets. In vivo, KBP-042 induced a superior and pronounced reduction in food intake in conjunction with a sustained pair-fed corrected weight loss in DIO rats. Concomitantly, KBP-042 improved glucose homeostasis and reduced hyperinsulinemia and hyperleptinemia in conjunction with enhanced insulin sensitivity. In ZDF rats, KBP-042 induced a superior attenuation of diabetic hyperglycemia and alleviated impaired glucose and insulin tolerance. Concomitantly, KBP-042 preserved insulinotropic and induced glucagonostatic action, ultimately preserving pancreatic insulin and glucagon content. In conclusion, oral KBP-042 is a novel DACRA, which exerts antiobesity and antidiabetic efficacy by dual modulation of insulin sensitivity and directly decelerating stress on the pancreatic α- and β-cells. These results could provide the basis for oral KBP-042 as a novel therapeutic agent in type 2 diabetes.
Collapse
Affiliation(s)
| | - Michael Feigh
- Nordic Bioscience, Herlev, Denmark; and Diabetes Research Centre, Department of Endocrinology, University of Southern Denmark, Odense, Denmark
| | | | | | - Jan Erik Henriksen
- Diabetes Research Centre, Department of Endocrinology, University of Southern Denmark, Odense, Denmark
| | - Henning Beck-Nielsen
- Diabetes Research Centre, Department of Endocrinology, University of Southern Denmark, Odense, Denmark
| | | | | | | |
Collapse
|
92
|
PTBP1 is required for glucose-stimulated cap-independent translation of insulin granule proteins and Coxsackieviruses in beta cells. Mol Metab 2014; 3:518-30. [PMID: 25061557 PMCID: PMC4099505 DOI: 10.1016/j.molmet.2014.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 12/15/2022] Open
Abstract
Glucose and GLP-1 stimulate not only insulin secretion, but also the post-transcriptional induction of insulin granule biogenesis. This process involves the nucleocytoplasmic translocation of the RNA binding protein PTBP1. Binding of PTBP1 to the 3'-UTRs of mRNAs for insulin and other cargoes of beta cell granules increases their stability. Here we show that glucose enhances also the binding of PTBP1 to the 5'-UTRs of these transcripts, which display IRES activity, and their translation exclusively in a cap-independent fashion. Accordingly, glucose-induced biosynthesis of granule cargoes was unaffected by pharmacological, genetic or Coxsackievirus-mediated inhibition of cap-dependent translation. Infection with Coxsackieviruses, which also depend on PTBP1 for their own cap-independent translation, reduced instead granule stores and insulin release. These findings provide insight into the mechanism for glucose-induction of insulin granule production and on how Coxsackieviruses, which have been implicated in the pathogenesis of type 1 diabetes, can foster beta cell failure.
Collapse
Key Words
- Beta cells
- CV, Coxsackievirus
- Diabetes
- ER, endoplasmic reticulum
- EV, Enterovirus
- F, Faulkner
- FL, firefly luciferase
- IRES, internal ribosomal entry site
- ITAF, IRES-trans-acting factor
- Insulin
- MCA, MIN6 cell adapted
- PABP, poly(A)-binding protein
- PC, prohormone convertase
- PTBP1, polypyrimidine tract-binding protein 1
- Polypyrimidine tract-binding protein
- S6K1, p70S6 Kinase 1
- Secretory granules
- T1D, type 1 diabetes
- Translation
- UTR, untranslated region
- Virus
- eIF4E-V5, eIF4E tagged at its C-terminus with a V5-epitope
- mTORC1, mammalian Target Of Rapamycin Complex 1
Collapse
|
93
|
Zuellig RA, Cavallari G, Gerber P, Tschopp O, Spinas GA, Moritz W, Lehmann R. Improved physiological properties of gravity-enforced reassembled rat and human pancreatic pseudo-islets. J Tissue Eng Regen Med 2014; 11:109-120. [PMID: 24737702 DOI: 10.1002/term.1891] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 12/20/2013] [Accepted: 02/26/2014] [Indexed: 01/01/2023]
Abstract
Previously we demonstrated the superiority of small islets vs large islets in terms of function and survival after transplantation, and we generated reaggregated rat islets (pseudo-islets) of standardized small dimensions by the hanging-drop culture method (HDCM). The aim of this study was to generate human pseudo-islets by HDCM and to evaluate and compare the physiological properties of rat and human pseudo-islets. Isolated rat and human islets were dissociated into single cells and incubated for 6-14 days by HDCM. Newly formed pseudo-islets were analysed for dimensions, morphology, glucose-stimulated insulin secretion (GSIS) and total insulin content. The morphology of reaggregated human islets was similar to that of native islets, while rat pseudo-islets had a reduced content of α and δ cells. GSIS of small rat and human pseudo-islets (250 cells) was increased up to 4.0-fold (p < 0.01) and 2.5-fold (p < 0.001), respectively, when compared to their native counterparts. Human pseudo-islets showed a more pronounced first-phase insulin secretion as compared to intact islets. GSIS was inversely correlated to islet size, and small islets (250 cells) contained up to six-fold more insulin/cell than large islets (1500 cells). Tissue loss with this new technology could be reduced to 49.2 ± 1.5% in rat islets, as compared to the starting amount. With HDCM, pseudo-islets of standardized size with similar cellular composition and improved biological function can be generated, which compensates for tissue loss during production. Transplantation of small pseudo-islets may represent an attractive strategy to improve graft survival and function, due to better oxygen and nutrient supply during the phase of revascularization. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- R A Zuellig
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - G Cavallari
- Nephrology, Dialysis and Transplantation Unit (Stefoni), S.Orsola-Malpighi Hospital, University of Bologna, Italy
| | - P Gerber
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - O Tschopp
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - G A Spinas
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - W Moritz
- InSphero AG, Schlieren, Switzerland
| | - R Lehmann
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Switzerland
| |
Collapse
|
94
|
Jun Y, Kang AR, Lee JS, Park SJ, Lee DY, Moon SH, Lee SH. Microchip-based engineering of super-pancreatic islets supported by adipose-derived stem cells. Biomaterials 2014; 35:4815-26. [PMID: 24636217 DOI: 10.1016/j.biomaterials.2014.02.045] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/23/2014] [Indexed: 12/13/2022]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic disorder characterized by targeted autoimmune-mediated destruction of the β cells of Langerhans within pancreatic islets. Currently, islet transplantation is the only curative therapy; however, donor shortages and cellular damage during the isolation process critically limit the use of this approach. Here, we describe a method for creating viable and functionally potent islets for successful transplantation by co-culturing single primary islet cells with adipose-derived stem cells (ADSCs) in concave microwells. We observed that the ADSCs segregated from the islet cells, eventually yielding purified islet spheroids in the three-dimensional environment. Thereafter, the ADSC-exposed islet spheroids showed significantly different ultrastructural morphologies, higher viability, and enhanced insulin secretion compared to mono-cultured islet spheroids. This suggests that ADSCs may have a significant potential to protect islet cells from damage during culture, and may be employed to improve islet cell survival and function prior to transplantation. In vivo experiments involving xenotransplantation of microfiber-encapsulated spheroids into a mouse model of diabetes revealed that co-culture-transplanted mice maintained their blood glucose levels longer than mono-culture-transplanted mice, and required less islet mass to reverse diabetes. This method for culturing islet spheroids could potentially help overcome the cell shortages that have limited clinical applications and could possibly be developed into a bioartificial pancreas.
Collapse
Affiliation(s)
- Yesl Jun
- Biotechnology-Medical Science, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-701, Republic of Korea
| | - Ah Ran Kang
- Biotechnology-Medical Science, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-701, Republic of Korea
| | - Jae Seo Lee
- Biotechnology-Medical Science, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-701, Republic of Korea
| | - Soon-Jung Park
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 143-701, Republic of Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791, Republic of Korea
| | - Sung-Hwan Moon
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 143-701, Republic of Korea
| | - Sang-Hoon Lee
- Biotechnology-Medical Science, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-701, Republic of Korea; Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 136-703, Republic of Korea.
| |
Collapse
|
95
|
Tsai MJ, Yang-Yen HF, Chiang MK, Wang MJ, Wu SS, Chen SH. TCTP is essential for β-cell proliferation and mass expansion during development and β-cell adaptation in response to insulin resistance. Endocrinology 2014; 155:392-404. [PMID: 24248465 DOI: 10.1210/en.2013-1663] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The perinatal period is critical for β-cell mass establishment, which is characterized by a transient burst in proliferation to increase β-cell mass in response to the need for glucose homeostasis throughout life. In adulthood, the ability of β-cells to grow, proliferate, and expand their mass is also characteristic of pathological states of insulin resistance. Translationally controlled tumor-associated protein (TCTP), an evolutionarily highly conserved protein that is implicated in cell growth and proliferation, has been identified as a novel glucose-regulated survival-supporting protein in pancreatic β-cells. In this study, the enhanced β-cell proliferation detected both during the perinatal developmental period and in insulin-resistant states in high-fat diet-fed mice was found to parallel the expression of TCTP in pancreatic β-cells. Specific knockout of TCTP in β-cells led to increased expression of total and nuclear Forkhead box protein O1 and tumor suppressor protein 53, and decreased expression of p70S6 kinase phosphorylation and cyclin D2 and cyclin-dependent kinase 2. This resulted in decreased β-cell proliferation and growth, reduced β-cell mass, and insulin secretion. Together, these effects led to hyperglycemia. These observations suggest that TCTP is essential for β-cell mass expansion during development and β-cell adaptation in response to insulin resistance.
Collapse
Affiliation(s)
- Ming-Jen Tsai
- PhD Program in Pharmacology and Toxicology (M.J.-T., S.-H.C.) and Department of Pharmacology (S.-S.W., S.-H.C.), School of Medicine, Tzu Chi University, Hualien 970, Taiwan; Departments of Emergency Medicine (M.J.-T.) and Medical Research (M.-J.W.), Neuro-Medical Scientific Center, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan; Institutes of Molecular Biology (H.-F.Y.-Y.), Academia Sinica, Taipei 115, Taiwan; and Department of Life Science and Institute of Molecular Biology (M.-K.C.), National Chung-Cheng University, Chia-Yi 621, Taiwan
| | | | | | | | | | | |
Collapse
|
96
|
Topographical arrangement of α- and β-cells within neo-islet tissues engineered by islet cell sheet transplantation in mice. Transplant Proc 2014; 45:1881-4. [PMID: 23769062 DOI: 10.1016/j.transproceed.2013.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 01/21/2013] [Indexed: 11/24/2022]
Abstract
BACKGROUND We established a procedure to engineer therapeutic neo-islets in subcutaneous spaces in mice by transplanting contiguous layers of islet cell sheets. In this study, we investigated the cellular arrangements of α and β within these engineered neo-islets in vivo as a function of time after sheet transplantation. METHODS AND RESULTS Temperature-responsive culture dishes optimized for dispersed islet cell culture were prepared by covalently immobilizing a temperature-responsive polymer poly(N-isopropylacrylamide) (PIPAAm) on plastic dishes followed by laminin-5 coating. Dispersed islet cells obtained from Lewis rats were plated onto the PIPAAm dishes. After reaching confluence at day 2, islet cells were harvested as uniformly spread islet cell sheets by lowering the culture temperature from 37°C to 20°C for 20 minutes. Islet sheet transplantation was performed to creat neo-islet tissues in the subcutaneous spaces of SCID mice with streptozotocin-induced diabetes. This neo-islet engineering approach successfully lowered mouse blood glucose levels, achieving euglycemia at day 5 and thereafter. Histologic analyses of samples obtained at day 4 revealed that neo-islet tissues in the subcutaneous spaces showed heterogeneous cellular alignment of α and β cells. In contrast, analyses of samples at days 14 and 60 revealed α and β cells predominantly located at the peripheral and central parts of the engineered tissues, respectively. CONCLUSIONS Reassembly of α and β cells occurred in neo-islet tissues engineered by sheet transplantation. The unique cellular arrangements in neo-islet tissues, which were similar to those in naïve pancreatic islets, may contribute to their longevity and long-term function.
Collapse
|
97
|
Wang X, Lei XG, Wang J. Malondialdehyde regulates glucose-stimulated insulin secretion in murine islets via TCF7L2-dependent Wnt signaling pathway. Mol Cell Endocrinol 2014; 382:8-16. [PMID: 24035868 DOI: 10.1016/j.mce.2013.09.003] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/30/2013] [Accepted: 09/03/2013] [Indexed: 11/29/2022]
Abstract
Evidence showed strong relations between malondialdehyde (MDA) levels and different pathological stages of diabetes. Here, an explicit system with freshly isolated islets and precisely controlled MDA gradient was employed to investigate the physiological effect of MDA on GSIS. Promoter analysis, pathway mapping, Q-PCR profiling, and siRNA verification were performed to clarify the intracellular signaling pathways. MDA at a moderately high level (5 and 10μM) promoted GSIS and accompanied with ATP/ADP ratio, cytosolic Ca(2+) level, and key regulators (GK, GLUT2, PDX1, and UCP2) changes in islets. Both upstream (PI3K and p-AKT) and downstream (TCF7L2 and TCF7) factors of Wnt pathway showed greatest changes. Knockdown of TCF7L2 blocked the MDA-induced GSIS elevation. These results indicated that MDA acted as a signaling messenger and regulated islet GSIS mainly through Wnt pathway. Therefore, the elevated MDA level and up-regulated Wnt signaling pathway could be an etiological factor in the development of hyperinsulinemia and type 2 diabetes.
Collapse
Affiliation(s)
- Xinhui Wang
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, Guangdong 510006, China; Animal Science Department, Cornell University, Ithaca, NY 14853, USA
| | - Xin Gen Lei
- Animal Science Department, Cornell University, Ithaca, NY 14853, USA.
| | - Jufang Wang
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
98
|
Wang X, Yun JW, Lei XG. Glutathione peroxidase mimic ebselen improves glucose-stimulated insulin secretion in murine islets. Antioxid Redox Signal 2014; 20:191-203. [PMID: 23795780 PMCID: PMC3887434 DOI: 10.1089/ars.2013.5361] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AIMS Glutathione peroxidase (GPX) mimic ebselen and superoxide dismutase (SOD) mimic copper diisopropylsalicylate (CuDIPs) were used to rescue impaired glucose-stimulated insulin secretion (GSIS) in islets of GPX1 and(or) SOD1-knockout mice. RESULTS Ebselen improved GSIS in islets of all four tested genotypes. The rescue in the GPX1 knockout resulted from a coordinated transcriptional regulation of four key GSIS regulators and was mediated by the peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α)-mediated signaling pathways. In contrast, CuDIPs improved GSIS only in the SOD1 knockout and suppressed gene expression of the PGC-1α pathway. INNOVATION Islets from the GPX1 and(or) SOD1 knockout mice provided metabolically controlled intracellular hydrogen peroxide (H2O2) and superoxide conditions for the present study to avoid confounding effects. Bioinformatics analyses of gene promoters and expression profiles guided the search for upstream signaling pathways to link the ebselen-initiated H2O2 scavenging to downstream key events of GSIS. The RNA interference was applied to prove PGC-1α as the main mediator for that link. CONCLUSION Our study revealed a novel metabolic use and clinical potential of ebselen in rescuing GSIS in the GPX1-deficient islets and mice, along with distinct differences between the GPX and SOD mimics in this regard. These findings highlight the necessities and opportunities of discretional applications of various antioxidant enzyme mimics in treating insulin secretion disorders. REBOUND TRACK: This work was rejected during standard peer review and rescued by Rebound Peer Review (Antioxid Redox Signal 16: 293-296, 2012) with the following serving as open reviewers: Regina Brigelius-Flohe, Vadim Gladyshev, Dexing Hou, and Holger Steinbrenner.
Collapse
Affiliation(s)
- Xinhui Wang
- 1 Department of Animal Science, Cornell University , Ithaca, New York
| | | | | |
Collapse
|
99
|
Medina A, Nakagawa Y, Ma J, Li L, Hamano K, Akimoto T, Ninomiya Y, Kojima I. Expression of the glucose-sensing receptor T1R3 in pancreatic islet: changes in the expression levels in various nutritional and metabolic states. Endocr J 2014; 61:797-805. [PMID: 24898279 DOI: 10.1507/endocrj.ej14-0221] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We reported recently that the taste type 1 receptor 3 (T1R3), a subunit of the sweet taste receptor, functions as a cell-surface glucose-sensing receptor in pancreatic β-cells. In the present study, we investigated the expression of T1R3 in pancreatic islets. mRNA for T1R2 and T1R3 was detected in mouse pancreatic islets. Quantitatively, the mRNA expression level of T1R2 was less than 1% of that of T1R3. Immunohistochemically, T1R3 was abundantly expressed in mouse islets whereas T1R2 was barely detected. Most immunoreactive T1R3 was colocalized with insulin and almost all β-cells were positive for T1R3. In addition, T1R3 was expressed in some portion of α-cells. Immunoreactivity of T1R3 in β-cells was markedly reduced in fed mice compared to those in fasting mice. In contrast, mRNA for T1R3 was not different in islets of fasting and fed mice. Glucose-induced insulin-secretion was higher in islets obtained from fasting mice compared to those from fed mice. The expression of T1R3 was markedly reduced in islets of ob/ob mice compared to those of control mice. Similarly, the expression of T1R3 was reduced in islet of db/db mice. In addition, the expression of T1R3 was markedly reduced in β-cells of fatty diabetic rats and GK rats, models of obese and non-obese type 2 diabetes, respectively. These results indicate that T1R3 is expressed mainly in β-cells and the expression levels are different depending upon the nutritional and metabolic conditions.
Collapse
Affiliation(s)
- Anya Medina
- Institute for Molecular & Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Tatarkiewicz K, Hargrove DM, Jodka CM, Gedulin BR, Smith PA, Hoyt JA, Lwin A, Collins L, Mamedova L, Levy OE, D'Souza L, Janssen S, Srivastava V, Ghosh SS, Parkes DG. A novel long-acting glucose-dependent insulinotropic peptide analogue: enhanced efficacy in normal and diabetic rodents. Diabetes Obes Metab 2014; 16:75-85. [PMID: 23859463 PMCID: PMC4237114 DOI: 10.1111/dom.12181] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 05/21/2013] [Accepted: 07/11/2013] [Indexed: 12/18/2022]
Abstract
AIM Glucose-dependent insulinotropic peptide (GIP) is an incretin hormone that is released from intestinal K cells in response to nutrient ingestion. We aimed to investigate the therapeutic potential of the novel N- and C-terminally modified GIP analogue AC163794. METHODS AC163794 was synthesized by solid-phase peptide synthesis. Design involved the substitution of the C-terminus tail region of the dipeptidyl peptidase IV (DPP-IV)-resistant GIP analogue [d-Ala(2) ]GIP(1-42) with the unique nine amino acid tail region of exenatide. The functional activity and binding of AC163794 to the GIP receptor were evaluated in RIN-m5F β-cells. In vitro metabolic stability was tested in human plasma and kidney membrane preparations. Acute insulinotropic effects were investigated in isolated mouse islets and during an intravenous glucose tolerance test in normal and diabetic Zucker fatty diabetic (ZDF) rats. The biological actions of AC163794 were comprehensively assessed in normal, ob/ob and high-fat-fed streptozotocin (STZ)-induced diabetic mice. Acute glucoregulatory effects of AC163794 were tested in diet-induced obese mice treated subchronically with AC3174, the exendatide analogue [Leu(14) ] exenatide. Human GIP or [d-Ala(2) ]GIP(1-42) were used for comparison. RESULTS AC163794 exhibited nanomolar functional GIP receptor potency in vitro similar to GIP and [d-Ala(2) ]GIP(1-42). AC163794 was metabolically more stable in vitro and displayed longer duration of insulinotropic action in vivo versus GIP and [d-Ala(2) ]GIP(1-42). In diabetic mice, AC163794 improved HbA1c through enhanced insulinotropic action, partial restoration of pancreatic insulin content and improved insulin sensitivity with no adverse effects on fat storage and metabolism. AC163794 provided additional baseline glucose-lowering when injected to mice treated with AC3174. CONCLUSIONS These studies support the potential use of a novel GIP analogue AC163794 for the treatment of type 2 diabetes.
Collapse
|