1
|
Cai X, Cao J, Wang L, Zou J, Li R, Sun P, Ding X, Zhang B, Liu Z, Pei X, Yang J, Zhan Y, Liu N, Liu T, Liang R, Gao J, Wang S. Liraglutide Protects Pancreatic Islet From Ischemic Injury by Reducing Oxidative Stress and Activating Akt Signaling During Cold Preservation to Improve Islet Transplantation Outcomes. Transplantation 2024; 108:e156-e169. [PMID: 38578708 DOI: 10.1097/tp.0000000000004949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
BACKGROUND Islet transplantation is a promising therapy for patients with type 1 diabetes. However, ischemic injury to the donor islets during cold preservation leads to reduced islet quality and compromises transplant outcome. Several studies imply that liraglutide, a glucagon-like peptide-1 receptor agonist, has a positive effect on promoting islet survival, but its impact on islet cold-ischemic injury remains unexplored. Therefore, the aim of this study was to investigate whether liraglutide can improve islet transplantation efficacy by inhibiting cold-ischemic injury and to explore the underlying mechanisms. METHODS Liraglutide was applied in a mouse pancreas preservation model and a human islets cold-preservation model, and islet viability, function, oxidative stress levels were evaluated. Furthermore, islet transplantation was performed in a syngeneic mouse model and a human-to-nude mouse islet xenotransplantation model. RESULTS The supplementation of liraglutide in preservation solution improved islet viability, function, and reduced cell apoptosis. Liraglutide inhibited the oxidative stress of cold-preserved pancreas or islets through upregulating the antioxidant enzyme glutathione levels, inhibiting reactive oxygen species accumulation, and maintaining the mitochondrial membrane integrity, which is associated with the activation of Akt signaling. Furthermore, the addition of liraglutide during cold preservation of donor pancreas or donor islets significantly improved the subsequent transplant outcomes in both syngeneic mouse islet transplantation model and human-to-nude mouse islet xenotransplantation model. CONCLUSIONS Liraglutide protects islets from cold ischemia-related oxidative stress during preservation and hence improved islet transplantation outcomes, and this protective effect of liraglutide in islets is associated with the activation of Akt signaling.
Collapse
Affiliation(s)
- Xiangheng Cai
- School of Medicine, Nankai University, Tianjin, China
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jinglin Cao
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Le Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jiaqi Zou
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Rui Li
- Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Peng Sun
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Xuejie Ding
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Boya Zhang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Zewen Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Xirui Pei
- First Clinical Department, The First Hospital of China Medical University, China Medical University, Shenyang, China
| | - Jiuxia Yang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Yixiang Zhan
- School of Medicine, Nankai University, Tianjin, China
| | - Na Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Tengli Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Rui Liang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, China
| | - Shusen Wang
- School of Medicine, Nankai University, Tianjin, China
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| |
Collapse
|
2
|
Holter MM, Phuong DJ, Lee I, Saikia M, Weikert L, Fountain S, Anderson ET, Fu Q, Zhang S, Sloop KW, Cummings BP. 14-3-3-zeta mediates GLP-1 receptor agonist action to alter α cell proglucagon processing. SCIENCE ADVANCES 2022; 8:eabn3773. [PMID: 35867787 PMCID: PMC9307243 DOI: 10.1126/sciadv.abn3773] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
Recent studies demonstrate that α cells contribute to glucose-stimulated insulin secretion (GSIS). Glucagon-like peptide-1 receptor (GLP-1R) agonists potently potentiate GSIS, making these drugs useful for diabetes treatment. However, the role of α and β cell paracrine interactions in the effects of GLP-1R agonists is undefined. We previously found that increased β cell GLP-1R signaling activates α cell GLP-1 expression. Here, we characterized the bidirectional paracrine cross-talk by which α and β cells communicate to mediate the effects of the GLP-1R agonist, liraglutide. We find that the effect of liraglutide to enhance GSIS is blunted by α cell ablation in male mice. Furthermore, the effect of β cell GLP-1R signaling to activate α cell GLP-1 is mediated by a secreted protein factor that is regulated by the signaling protein, 14-3-3-zeta, in mouse and human islets. These data refine our understanding of GLP-1 pharmacology and identify 14-3-3-zeta as a potential target to enhance α cell GLP-1 production.
Collapse
Affiliation(s)
- Marlena M. Holter
- Department of Biomedical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, NY, USA
| | - Daryl J. Phuong
- Department of Biomedical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, NY, USA
| | - Isaac Lee
- Department of Biomedical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, NY, USA
| | - Mridusmita Saikia
- Department of Biomedical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, NY, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, NY, USA
| | - Lisa Weikert
- Department of Biomedical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, NY, USA
| | - Samantha Fountain
- Department of Biomedical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, NY, USA
| | - Elizabeth T. Anderson
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Qin Fu
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Kyle W. Sloop
- Diabetes and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Bethany P. Cummings
- Department of Biomedical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, NY, USA
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
3
|
Lee SM, Kim D, Kwak KM, Khin PP, Lim OK, Kim KW, Kim BJ, Jun HS. Comparison of the Effects of Liraglutide on Islet Graft Survival Between Local and Systemic Delivery. Cell Transplant 2021; 29:963689720971245. [PMID: 33172296 PMCID: PMC7784585 DOI: 10.1177/0963689720971245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Islet transplantation has emerged as a promising treatment for type 1 diabetes mellitus. Liraglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, protects beta cells after islet transplantation by improving glycemic control through several mechanisms. In this study, we compared the effects of local pretreatment and systemic treatment with liraglutide on islet transplantation in a diabetic mouse model. Streptozotocin (STZ)-induced diabetic C57BL/6 mice were transplanted with syngeneic islets under the kidney capsule. Isolated islets were either locally treated with liraglutide before transplantation or mice were treated systemically by intraperitoneal injection after islet transplantation. Local pretreatment of islets with liraglutide was more effective in increasing body weight, decreasing hemoglobin A1c levels, and lowering blood glucose levels in STZ-diabetic mice transplanted with islets. Local pretreatment was also more effective in increasing insulin secretion and islet survival in STZ-diabetic mice. Histological analysis of the transplantation site revealed fewer apoptotic cells following local pretreatment compared with systemic injection of liraglutide. These findings indicate that liraglutide administered once locally before transplantation might have superior effects on islet preservation than systemic administration.
Collapse
Affiliation(s)
- Song Mi Lee
- College of Pharmacy and Gachon Institute Pharmaceutical Science, Gachon University, Yeonsu-gu, Incheon, Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Yeonsu-gu, Incheon, Korea.,Both the authors contributed equally to this article
| | - Donghee Kim
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Yeonsu-gu, Incheon, Korea.,Both the authors contributed equally to this article
| | - Kyung Min Kwak
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Yeonsu-gu, Incheon, Korea
| | - Phyu Phyu Khin
- College of Pharmacy and Gachon Institute Pharmaceutical Science, Gachon University, Yeonsu-gu, Incheon, Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Yeonsu-gu, Incheon, Korea
| | - Oh Kyung Lim
- Department of Rehabilitation Medicine, Gachon University Gil Medical Center, Namdong-gu, Incheon, Korea
| | - Kwang-Won Kim
- Department of Internal Medicine, Gachon University Gil Medical Center, Namdong-gu, Incheon, Korea
| | - Byung-Joon Kim
- Department of Internal Medicine, Gachon University Gil Medical Center, Namdong-gu, Incheon, Korea
| | - Hee-Sook Jun
- College of Pharmacy and Gachon Institute Pharmaceutical Science, Gachon University, Yeonsu-gu, Incheon, Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Yeonsu-gu, Incheon, Korea.,Gil Medical Research Institute, Gil Hospital, Namdong-gu, Incheon, Korea
| |
Collapse
|
4
|
Verhoeff K, Henschke SJ, Marfil-Garza BA, Dadheech N, Shapiro AMJ. Inducible Pluripotent Stem Cells as a Potential Cure for Diabetes. Cells 2021; 10:cells10020278. [PMID: 33573247 PMCID: PMC7911560 DOI: 10.3390/cells10020278] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 02/07/2023] Open
Abstract
Over the last century, diabetes has been treated with subcutaneous insulin, a discovery that enabled patients to forego death from hyperglycemia. Despite novel insulin formulations, patients with diabetes continue to suffer morbidity and mortality with unsustainable costs to the health care system. Continuous glucose monitoring, wearable insulin pumps, and closed-loop artificial pancreas systems represent an advance, but still fail to recreate physiologic euglycemia and are not universally available. Islet cell transplantation has evolved into a successful modality for treating a subset of patients with ‘brittle’ diabetes but is limited by organ donor supply and immunosuppression requirements. A novel approach involves generating autologous or immune-protected islet cells for transplant from inducible pluripotent stem cells to eliminate detrimental immune responses and organ supply limitations. In this review, we briefly discuss novel mechanisms for subcutaneous insulin delivery and define their shortfalls. We describe embryological development and physiology of islets to better understand their role in glycemic control and, finally, discuss cell-based therapies for diabetes and barriers to widespread use. In response to these barriers, we present the promise of stem cell therapy, and review the current gaps requiring solutions to enable widespread use of stem cells as a potential cure for diabetes.
Collapse
Affiliation(s)
- Kevin Verhoeff
- Department of Surgery, University of Alberta, Edmonton, AB T6G 2B7, Canada;
- Correspondence: ; Tel.: +1-780-984-1836
| | - Sarah J. Henschke
- Department of Emergency Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada;
| | | | - Nidheesh Dadheech
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada;
| | - Andrew Mark James Shapiro
- FRCS (Eng) FRCSC MSM FCAHS, Clinical Islet Transplant Program, Alberta Diabetes Institute, Department of Surgery, Canadian National Transplant Research Program, Edmonton, AB T6G 2B7, Canada;
| |
Collapse
|
5
|
Juang JH, Chen CY, Kao CW, Huang YW, Chiu TY, Chen CT. Implanted islet mass influences the effects of dipeptidyl peptidase-IV inhibitor LAF237 on transplantation outcomes in diabetic mice. Biomed J 2020; 44:S210-S217. [PMID: 35300943 PMCID: PMC9068567 DOI: 10.1016/j.bj.2020.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/10/2020] [Accepted: 10/06/2020] [Indexed: 01/06/2023] Open
Abstract
Background Previous studies showed inconsistent Results of the effects of dipeptidyl peptidase (DPP)-IV inhibitors on syngeneic mouse islet transplantation. We hypothesized that the implanted islet numbers are critical for the effects of DPP-IV inhibitors on the outcomes of transplantation. Methods One hundred and fifty or three hundred islets were syngeneically transplanted under the renal capsule of each streptozocin-diabetic C57BL/6 mouse and recipients were then treated without or with LAF237 (10 mg/kg/day, po) for 6 weeks. After transplantation, recipients’ blood glucose, body weight and intraperitoneal glucose tolerance test (IPGTT) were followed-up periodically. The graft was removed for the measurement of β-cell mass at 6 weeks. Results In recipients with 150 islets, it was not significantly different between the LAF237- treated group (n = 14) and control group (n = 14) in terms of the blood glucose, body weight, glucose tolerance at 2, 4 and 6 weeks or the graft β-cell mass at 6 weeks. In contrast, in recipients with 300 islets, the LAF237-treated group (n = 24) did have a lower area under the curve of the IPGTT at 4 weeks (p = 0.0237) and 6 weeks (p = 0.0113) as well as more graft β-cell mass at 6 weeks (0.655 ± 0.008 mg vs. 0.435 ± 0.006 mg, p = 0.0463) than controls (n = 24). Conclusions Our findings revealed 6-week treatment of LAF237 improves glucose tolerance and increases graft β-cell mass in diabetic mice transplanted with a sufficient number but not a marginal number of islets. These indicate that the effects of DPP-IV inhibitors are influenced by the implanted islet mass.
Collapse
Affiliation(s)
- Jyuhn-Huarng Juang
- Division of Endocrinology and Metabolism, Center for Tissue Engineering, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Chen-Yi Chen
- Division of Endocrinology and Metabolism, Center for Tissue Engineering, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chen-Wei Kao
- Division of Endocrinology and Metabolism, Center for Tissue Engineering, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yu-Wen Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Tai-Yu Chiu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
6
|
Stock AA, Manzoli V, De Toni T, Abreu MM, Poh YC, Ye L, Roose A, Pagliuca FW, Thanos C, Ricordi C, Tomei AA. Conformal Coating of Stem Cell-Derived Islets for β Cell Replacement in Type 1 Diabetes. Stem Cell Reports 2020; 14:91-104. [PMID: 31839542 PMCID: PMC6962554 DOI: 10.1016/j.stemcr.2019.11.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 11/08/2019] [Accepted: 11/15/2019] [Indexed: 12/29/2022] Open
Abstract
The scarcity of donors and need for immunosuppression limit pancreatic islet transplantation to a few patients with labile type 1 diabetes. Transplantation of encapsulated stem cell-derived islets (SC islets) might extend the applicability of islet transplantation to a larger cohort of patients. Transplantation of conformal-coated islets into a confined well-vascularized site allows long-term diabetes reversal in fully MHC-mismatched diabetic mice without immunosuppression. Here, we demonstrated that human SC islets reaggregated from cryopreserved cells display glucose-stimulated insulin secretion in vitro. Importantly, we showed that conformally coated SC islets displayed comparable in vitro function with unencapsulated SC islets, with conformal coating permitting physiological insulin secretion. Transplantation of SC islets into the gonadal fat pad of diabetic NOD-scid mice revealed that both unencapsulated and conformal-coated SC islets could reverse diabetes and maintain human-level euglycemia for more than 80 days. Overall, these results provide support for further evaluation of safety and efficacy of conformal-coated SC islets in larger species.
Collapse
Affiliation(s)
- Aaron A Stock
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Vita Manzoli
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA
| | - Teresa De Toni
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Maria M Abreu
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA
| | | | - Lillian Ye
- Semma Therapeutics, Inc., Cambridge, MA 02142, USA
| | - Adam Roose
- Semma Therapeutics, Inc., Cambridge, MA 02142, USA
| | | | - Chris Thanos
- Semma Therapeutics, Inc., Cambridge, MA 02142, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
7
|
Knudsen LB. Inventing Liraglutide, a Glucagon-Like Peptide-1 Analogue, for the Treatment of Diabetes and Obesity. ACS Pharmacol Transl Sci 2019; 2:468-484. [PMID: 32259078 DOI: 10.1021/acsptsci.9b00048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Indexed: 01/08/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) has been in focus since the early 1980s as a long looked for incretin hormone, released from the gastrointestinal tract and with an important effect on glucose-dependent insulin secretion, providing efficient glucose lowering, with little risk for hypoglycemia. The enzyme dipeptidyl peptidase-4 (DPP-4) degrades GLP-1 very fast, and the remaining metabolite is cleared rapidly by the kidneys. Liraglutide is a fatty acid acylated analogue of GLP-1 that provides efficacy for 24 h/day. The mechanism of action for liraglutide is reviewed in detail with focus on pancreatic efficacy and safety, thyroid safety, and weight loss mechanism. Evolving science hypothesizes that GLP-1 has important effects on atherosclerosis, relevant for the cardiovascular benefit seen in the treatment of diabetes and obesity. Also, GLP-1 may be relevant in neurodegenerative diseases.
Collapse
Affiliation(s)
- Lotte Bjerre Knudsen
- Global Drug Discovery, Novo Nordisk, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| |
Collapse
|
8
|
Sun S, Zhang H, Shan K, Sun T, Lin M, Jia L, Chen YQ. Effect of Different Cereal Peptides on the Development of Type 1 Diabetes is Associated with Their Anti‐inflammatory Ability: In Vitro and In Vivo Studies. Mol Nutr Food Res 2019; 63:e1800987. [DOI: 10.1002/mnfr.201800987] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/20/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Suling Sun
- School of MedicineJiangnan University Wuxi 214122 P. R. China
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Kai Shan
- School of MedicineJiangnan University Wuxi 214122 P. R. China
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Tianjun Sun
- Department of Biochemistry & Molecular Biology & Center for Blood ResearchUniversity of British Columbia 2350 Health Sciences Mall Vancouver British Columbia V6T 1Z3 Canada
| | - Mengyuan Lin
- Wuxi Maternal and Child Health Hospital P. R. China
| | - Lingling Jia
- School of MedicineJiangnan University Wuxi 214122 P. R. China
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Yong Q. Chen
- School of MedicineJiangnan University Wuxi 214122 P. R. China
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- Departments of Cancer Biology and BiochemistryWake Forest School of Medicine Winston‐Salem NC 27157 USA
| |
Collapse
|
9
|
Gao M, Deng XL, Liu ZH, Song HJ, Zheng J, Cui ZH, Xiao KL, Chen LL, Li HQ. Liraglutide protects β-cell function by reversing histone modification of Pdx-1 proximal promoter in catch-up growth male rats. J Diabetes Complications 2018; 32:985-994. [PMID: 30177467 DOI: 10.1016/j.jdiacomp.2018.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/17/2018] [Accepted: 08/01/2018] [Indexed: 01/08/2023]
Abstract
AIMS Catch-up growth after a period of nutritional deprivation in adulthood is related to the onset of metabolic disorders. This process involves chromatin remodelling of the Pdx-1 gene in pancreas. The objective of this study was to determine the chromatin remodelling mechanism of GLP-1 analogue Liraglutide upon Pdx-1 in catch-up growth rats in vivo and in vitro. METHODS Five-week-old male specific pathogen free (SPF) Wistar rats were randomly divided into normal group, catch-up growth group and Liraglutide group. Hyperglycemic clamp test and glucose-stimulated insulin secretion test were carried out to evaluate β-cell function in vivo and in vitro. The histone H3 modification changes at the Pdx-1 proximal promoter were assessed by chromatin immunoprecipitation. RESULTS The catch-up growth state was characterized by less recruitment of histone H3 lysine4 trimethylation and histone H3 acetylation and more recruitment of histone H3 lysine9 dimethylation at the Pdx-1 proximal promoter. Liraglutide treatment reversed these epigenetic changes and increased Pdx-1 expression, which could be abrogated by GLP-1 receptor antagonist Exendin 9-39. The β-cell function of catch-up growth rats was improved after Liraglutide treatment. CONCLUSIONS The protective effects of Liraglutide on pancreatic islet β-cell function may be related to histone H3 modification at the Pdx-1 proximal promoter during catch-up growth and could be used to treat catch-up growth-related metabolic disorders.
Collapse
Affiliation(s)
- Ming Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China; Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Xiu-Ling Deng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Zhen-Hua Liu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Hui-Jie Song
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China; Department of Endocrinology, Wuhan No.1 Hospital, Wuhan 430022, Hubei, China
| | - Juan Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Zhen-Hai Cui
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Kang-Li Xiao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Lu-Lu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Hui-Qing Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China.
| |
Collapse
|
10
|
Bellin MD, Beilman GJ, Dunn TB, Pruett TL, Sutherland DER, Chinnakotla S, Hodges JS, Lane A, Ptacek P, Berry KL, Hering BJ, Moran A. Sitagliptin Treatment After Total Pancreatectomy With Islet Autotransplantation: A Randomized, Placebo-Controlled Study. Am J Transplant 2017; 17:443-450. [PMID: 27459721 PMCID: PMC5266635 DOI: 10.1111/ajt.13979] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/19/2016] [Accepted: 07/12/2016] [Indexed: 01/25/2023]
Abstract
Insulin independence after total pancreatectomy and islet autotransplant (TPIAT) for chronic pancreatitis is limited by a high rate of postprocedure beta cell apoptosis. Endogenous glucagon-like peptide-1 and glucose-dependent insulinotropic peptide, which are increased by dipeptidyl peptidase 4 inhibitor therapy (sitagliptin) may protect against beta cell apoptosis. To determine the effect of sitagliptin after TPIAT, 83 adult TPIAT recipients were randomized to receive sitagliptin (n = 54) or placebo (n = 29) for 12 months after TPIAT. At 12 and 18 months after TPIAT, participants were assessed for insulin independence; metabolic testing was performed with mixed meal tolerance testing and frequent sample intravenous glucose tolerance testing. Insulin independence did not differ between the sitagliptin and placebo groups at 12 months (42% vs. 45%, p = 0.82) or 18 months (36% vs. 44%, p = 0.48). At 12 months, insulin dose was 9.0 (standard error 1.7) units/day and 7.9 (2.2) units/day in the sitagliptin and placebo groups, respectively (p = 0.67) and at 18 months 10.3 (1.9) and 7.1 (2.6) units/day, respectively (p = 0.32). Hemoglobin A1c levels and insulin secretory measures were similar in the two groups, as were adverse events. In conclusion, sitagliptin could be safely administered but did not improve metabolic outcomes after TPIAT.
Collapse
Affiliation(s)
- M D Bellin
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - G J Beilman
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - T B Dunn
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - T L Pruett
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - D E R Sutherland
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - S Chinnakotla
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - J S Hodges
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - A Lane
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - P Ptacek
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - K L Berry
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - B J Hering
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| | - A Moran
- Departments of Pediatrics, Surgery, Biostatistics, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN
| |
Collapse
|
11
|
Langlois A, Dal S, Vivot K, Mura C, Seyfritz E, Bietiger W, Dollinger C, Peronet C, Maillard E, Pinget M, Jeandidier N, Sigrist S. Improvement of islet graft function using liraglutide is correlated with its anti-inflammatory properties. Br J Pharmacol 2016; 173:3443-3453. [PMID: 27515367 PMCID: PMC5120160 DOI: 10.1111/bph.13575] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/27/2016] [Accepted: 07/20/2016] [Indexed: 01/15/2023] Open
Abstract
Background and Purpose Liraglutide improves the metabolic control of diabetic animals after islet transplantation. However, the mechanisms underlying this effect remain unknown. The objective of this study was to evaluate the anti‐inflammatory and anti‐oxidative properties of liraglutide on rat pancreatic islets in vitro and in vivo. Experimental Approach In vitro, rat islets were incubated with 10 μmol·L−1 liraglutide for 12 and 24 h. Islet viability functionality was assessed. The anti‐inflammatory properties of liraglutide were evaluated by measuring CCL2, IL‐6 and IL‐10 secretion and macrophage chemotaxis. The anti‐oxidative effect of liraglutide was evaluated by measuring intracellular ROS and the total anti‐oxidative capacity. In vivo, 1000 islets were cultured for 24 h with or without liraglutide and then transplanted into the liver of streptozotocin‐induced diabetic Lewis rats with or without injections of liraglutide. Effects of liraglutide on metabolic control were evaluated for 1 month. Key Results Islet viability and function were preserved and enhanced with liraglutide treatment. Liraglutide decreased CCL2 and IL‐6 secretion and macrophage activation after 12 h of culture, while IL‐10 secretion was unchanged. However, intracellular levels of ROS were increased with liraglutide treatment at 12 h. This result was correlated with an increase of anti‐oxidative capacity. In vivo, liraglutide decreased macrophage infiltration and reduced fasting blood glucose in transplanted rats. Conclusions and Implications The beneficial effects of liraglutide on pancreatic islets appear to be linked to its anti‐inflammatory and anti‐oxidative properties. These findings indicated that analogues of glucagon‐like peptide‐1 could be used to improve graft survival.
Collapse
Affiliation(s)
- A Langlois
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - S Dal
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - K Vivot
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - C Mura
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - E Seyfritz
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - W Bietiger
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - C Dollinger
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - C Peronet
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - E Maillard
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - M Pinget
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Service d'Endocrinologie, Diabète, Maladies Métaboliques, Pôle NUDE, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - N Jeandidier
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Service d'Endocrinologie, Diabète, Maladies Métaboliques, Pôle NUDE, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - S Sigrist
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
12
|
Functionality and antidiabetic utility of β- and L-cell containing pseudoislets. Exp Cell Res 2016; 344:201-9. [DOI: 10.1016/j.yexcr.2016.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 12/12/2022]
|
13
|
Langlois A, Mura C, Bietiger W, Seyfritz E, Dollinger C, Peronet C, Maillard E, Pinget M, Jeandidier N, Sigrist S. In Vitro and In Vivo Investigation of the Angiogenic Effects of Liraglutide during Islet Transplantation. PLoS One 2016; 11:e0147068. [PMID: 26974949 PMCID: PMC4790919 DOI: 10.1371/journal.pone.0147068] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/27/2015] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION This study investigated the angiogenic properties of liraglutide in vitro and in vivo and the mechanisms involved, with a focus on Hypoxia Inducible Factor-1α (HIF-1α) and mammalian target of rapamycin (mTOR). MATERIALS AND METHODS Rat pancreatic islets were incubated in vitro with 10 μmol/L of liraglutide (Lira) for 12, 24 and 48 h. Islet viability was studied by fluorescein diacetate/propidium iodide staining and their function was assessed by glucose stimulation. The angiogenic effect of liraglutide was determined in vitro by the measure of vascular endothelial growth factor (VEGF) secretion using enzyme-linked immunosorbent assay and by the evaluation of VEGF and platelet-derived growth factor-α (PDGFα) expression with quantitative polymerase chain reaction technic. Then, in vitro and in vivo, angiogenic property of Lira was evaluated using immunofluorescence staining targeting the cluster of differentiation 31 (CD31). To understand angiogenic mechanisms involved by Lira, HIF-1α and mTOR activation were studied using western blotting. In vivo, islets (1000/kg body-weight) were transplanted into diabetic (streptozotocin) Lewis rats. Metabolic control was assessed for 1 month by measuring body-weight gain and fasting blood glucose. RESULTS Islet viability and function were respectively preserved and enhanced (p<0.05) with Lira, versus control. Lira increased CD31-positive cells, expression of VEGF and PDGFα (p<0.05) after 24 h in culture. Increased VEGF secretion versus control was also observed at 48 h (p<0.05). Moreover, Lira activated mTOR (p<0.05) signalling pathway. In vivo, Lira improved vascular density (p<0.01), body-weight gain (p<0.01) and reduced fasting blood glucose in transplanted rats (p<0.001). CONCLUSION The beneficial effects of liraglutide on islets appeared to be linked to its angiogenic properties. These findings indicated that glucagon-like peptide-1 analogues could be used to improve transplanted islet revascularisation.
Collapse
Affiliation(s)
- Allan Langlois
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Carole Mura
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - William Bietiger
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Elodie Seyfritz
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Camille Dollinger
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Claude Peronet
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Elisa Maillard
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Michel Pinget
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
- Service d’Endocrinologie, Diabète, Maladies Métaboliques, Pôle NUDE, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| | - Nathalie Jeandidier
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
- Service d’Endocrinologie, Diabète, Maladies Métaboliques, Pôle NUDE, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| | - Séverine Sigrist
- UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| |
Collapse
|
14
|
Abdulreda MH, Rodriguez-Diaz R, Caicedo A, Berggren PO. Liraglutide Compromises Pancreatic β Cell Function in a Humanized Mouse Model. Cell Metab 2016; 23:541-6. [PMID: 26876561 PMCID: PMC4785083 DOI: 10.1016/j.cmet.2016.01.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/30/2015] [Accepted: 01/15/2016] [Indexed: 01/29/2023]
Abstract
Incretin mimetics are frequently used in the treatment of type 2 diabetes because they potentiate β cell response to glucose. Clinical evidence showing short-term benefits of such therapeutics (e.g., liraglutide) is abundant; however, there have been several recent reports of unexpected complications in association with incretin mimetic therapy. Importantly, clinical evidence on the potential effects of such agents on the β cell and islet function during long-term, multiyear use remains lacking. We now show that prolonged daily liraglutide treatment of >200 days in humanized mice, transplanted with human pancreatic islets in the anterior chamber of the eye, is associated with compromised release of human insulin and deranged overall glucose homeostasis. These findings raise concern about the chronic potentiation of β cell function through incretin mimetic therapy in diabetes.
Collapse
Affiliation(s)
- Midhat H Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, FL 33136, USA.
| | - Rayner Rodriguez-Diaz
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, Miami, FL 33136, USA
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, Miami, FL 33136, USA
| | - Per-Olof Berggren
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, FL 33136, USA; The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm SE-17176, Sweden.
| |
Collapse
|
15
|
Gala-Lopez BL, Pepper AR, Pawlick RL, O'Gorman D, Kin T, Bruni A, Abualhassan N, Bral M, Bautista A, Manning Fox JE, Young LG, MacDonald PE, Shapiro AMJ. Antiaging Glycopeptide Protects Human Islets Against Tacrolimus-Related Injury and Facilitates Engraftment in Mice. Diabetes 2016; 65:451-62. [PMID: 26581595 DOI: 10.2337/db15-0764] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 11/10/2015] [Indexed: 02/05/2023]
Abstract
Clinical islet transplantation has become an established treatment modality for selected patients with type 1 diabetes. However, a large proportion of transplanted islets is lost through multiple factors, including immunosuppressant-related toxicity, often requiring more than one donor to achieve insulin independence. On the basis of the cytoprotective capabilities of antifreeze proteins (AFPs), we hypothesized that supplementation of islets with synthetic AFP analog antiaging glycopeptide (AAGP) would enhance posttransplant engraftment and function and protect against tacrolimus (Tac) toxicity. In vitro and in vivo islet Tac exposure elicited significant but reversible reduction in insulin secretion in both mouse and human islets. Supplementation with AAGP resulted in improvement of islet survival (Tac(+) vs. Tac+AAGP, 31.5% vs. 67.6%, P < 0.01) coupled with better insulin secretion (area under the curve: Tac(+) vs. Tac+AAGP, 7.3 vs. 129.2 mmol/L/60 min, P < 0.001). The addition of AAGP reduced oxidative stress, enhanced insulin exocytosis, improved apoptosis, and improved engraftment in mice by decreasing expression of interleukin (IL)-1β, IL-6, keratinocyte chemokine, and tumor necrosis factor-α. Finally, transplant efficacy was superior in the Tac+AAGP group and was similar to islets not exposed to Tac, despite receiving continuous treatment for a limited time. Thus, supplementation with AAGP during culture improves islet potency and attenuates long-term Tac-induced graft dysfunction.
Collapse
Affiliation(s)
- Boris L Gala-Lopez
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada Canadian National Transplant Research Program, University of Alberta, Edmonton, Alberta, Canada
| | - Andrew R Pepper
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada Canadian National Transplant Research Program, University of Alberta, Edmonton, Alberta, Canada
| | - Rena L Pawlick
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Doug O'Gorman
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Tatsuya Kin
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Antonio Bruni
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada Canadian National Transplant Research Program, University of Alberta, Edmonton, Alberta, Canada
| | - Nasser Abualhassan
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada Canadian National Transplant Research Program, University of Alberta, Edmonton, Alberta, Canada
| | - Mariusz Bral
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Austin Bautista
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Jocelyn E Manning Fox
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Lachlan G Young
- ProtoKinetix Inc., Vancouver, Vancouver, British Columbia, Canada
| | - Patrick E MacDonald
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - A M James Shapiro
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada Canadian National Transplant Research Program, University of Alberta, Edmonton, Alberta, Canada Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
16
|
Renner S, Blutke A, Streckel E, Wanke R, Wolf E. Incretin actions and consequences of incretin-based therapies: lessons from complementary animal models. J Pathol 2015; 238:345-58. [DOI: 10.1002/path.4655] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/02/2015] [Accepted: 10/05/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Simone Renner
- Gene Centre, Centre for Innovative Medical Models (CiMM) and German Centre for Diabetes Research (DZD); Ludwig-Maximilians-Universität München; Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology, Centre for Clinical Veterinary Medicine; Ludwig-Maximilians-Universität München; Germany
| | - Elisabeth Streckel
- Gene Centre, Centre for Innovative Medical Models (CiMM) and German Centre for Diabetes Research (DZD); Ludwig-Maximilians-Universität München; Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Centre for Clinical Veterinary Medicine; Ludwig-Maximilians-Universität München; Germany
| | - Eckhard Wolf
- Gene Centre, Centre for Innovative Medical Models (CiMM) and German Centre for Diabetes Research (DZD); Ludwig-Maximilians-Universität München; Germany
| |
Collapse
|
17
|
Carlessi R, Lemos NE, Dias AL, Oliveira FS, Brondani LA, Canani LH, Bauer AC, Leitão CB, Crispim D. Exendin-4 protects rat islets against loss of viability and function induced by brain death. Mol Cell Endocrinol 2015; 412:239-50. [PMID: 25976662 DOI: 10.1016/j.mce.2015.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 02/01/2023]
Abstract
Islet quality loss after isolation from brain-dead donors still hinders the implementation of human islet transplantation for treatment of type 1 diabetes. In this scenario, systemic inflammation elicited by donor brain death (BD) is among the main factors influencing islet viability and functional impairment. Exendin-4 is largely recognized to promote anti-inflammatory and cytoprotective effects on β-cells. Therefore, we hypothesized that administration of exendin-4 to brain-dead donors might improve islet survival and insulin secretory capabilities. Here, using a rat model of BD, we demonstrate that exendin-4 administration to the brain-dead donors increases both islet viability and glucose-stimulated insulin secretion. In this model, exendin-4 treatment produced a significant decrease in interleukin-1β expression in the pancreas. Furthermore, exendin-4 treatment increased the expression of superoxide dismutase-2 and prevented BD-induced elevation in uncoupling protein-2 expression. Such observations were accompanied by a reduction in gene expression of two genes often associated with endoplasmic reticulum (ER) stress response in freshly isolated islets from treated animals, C/EBP homologous protein and immunoglobulin heavy-chain binding protein. As ER stress response has been shown to be triggered by and to participate in cytokine-induced β-cell death, we suggest that exendin-4 might exert its beneficial effects through alleviation of pancreatic inflammation and oxidative stress, which in turn could prevent islet ER stress and β-cell death. Our findings might unveil a novel strategy to preserve islet quality from brain-dead donors. After testing in the human pancreatic islet transplantation setting, this approach might sum to the ongoing effort to achieve consistent and successful single-donor islet transplantation.
Collapse
Affiliation(s)
- Rodrigo Carlessi
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; School of Biomedical Sciences, CHIRI - Biosciences, Curtin University, Perth, Western Australia 6845, Australia
| | - Natália E Lemos
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ana L Dias
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Fernanda S Oliveira
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Letícia A Brondani
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luis H Canani
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andrea C Bauer
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Cristiane B Leitão
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daisy Crispim
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
18
|
Nomoto H, Miyoshi H, Furumoto T, Oba K, Tsutsui H, Miyoshi A, Kondo T, Tsuchida K, Atsumi T, Manda N, Kurihara Y, Aoki S. A Comparison of the Effects of the GLP-1 Analogue Liraglutide and Insulin Glargine on Endothelial Function and Metabolic Parameters: A Randomized, Controlled Trial Sapporo Athero-Incretin Study 2 (SAIS2). PLoS One 2015; 10:e0135854. [PMID: 26284918 PMCID: PMC4540491 DOI: 10.1371/journal.pone.0135854] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/26/2015] [Indexed: 02/06/2023] Open
Abstract
Objectives GLP-1 improves hyperglycemia, and it has been reported to have favorable effects on atherosclerosis. However, it has not been fully elucidated whether GLP-1 is able to improve endothelial function in patients with type 2 diabetes. Therefore, we investigated the efficacy of the GLP-1 analogue, liraglutide on endothelial function and glycemic metabolism compared with insulin glargine therapy. Materials and Methods In this multicenter, prospective randomized parallel-group comparison study, 31 diabetic outpatients (aged 60.3 ± 10.3 years with HbA1c levels of 8.6 ± 0.8%) with current metformin and/or sulfonylurea treatment were enrolled and randomly assigned to receive liraglutide or glargine therapy once daily for 14 weeks. Flow mediated dilation (FMD), a comprehensive panel of hemodynamic parameters (Task Force Monitor), and serum metabolic markers were assessed before and after the treatment period. Results A greater reduction (worsening) in %FMD was observed in the glargine group, although this change was not statistically different from the liraglutide group (liraglutide; 5.7 to 5.4%, glargine 6.7 to 5.7%). The augmentation index, C-peptide index, derivatives of reactive oxygen metabolites and BMI were significantly improved in the liraglutide group. Central systolic blood pressure and NT-proBNP also tended to be improved in the liraglutide-treated group, while improvements in HbA1c levels were similar between groups. Cardiac index, blood pressure and most other metabolic parameters were not different. Conclusions Regardless of glycemic improvement, early liraglutide therapy did not affect endothelial function but may provide favorable effects on beta-cell function and cardioprotection in type 2 diabetics without advanced atherosclerosis. Trial Registration UMIN Clinical Trials Registry System as trial ID UMIN000005331.
Collapse
Affiliation(s)
- Hiroshi Nomoto
- Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hideaki Miyoshi
- Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- * E-mail:
| | - Tomoo Furumoto
- Department of Cardiovascular Medicine, NTT East Japan Sapporo Hospital, Sapporo, Japan
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Koji Oba
- Department of Biostatistics, School of Public Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Interfaculty Initiative in Information Studies, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Arina Miyoshi
- Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Takuma Kondo
- Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | - Tatsuya Atsumi
- Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | | | | | | |
Collapse
|
19
|
Rudinsky AJ, Adin CA, Borin-Crivellenti S, Rajala-Schultz P, Hall MJ, Gilor C. Pharmacology of the glucagon-like peptide-1 analog exenatide extended-release in healthy cats. Domest Anim Endocrinol 2015; 51:78-85. [PMID: 25594949 DOI: 10.1016/j.domaniend.2014.12.003] [Citation(s) in RCA: 15] [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] [Received: 08/26/2014] [Revised: 12/08/2014] [Accepted: 12/11/2014] [Indexed: 11/20/2022]
Abstract
Exenatide extended-release (ER) is a microencapsulated formulation of the glucagon-like peptide 1-receptor agonist exenatide. It has a protracted pharmacokinetic profile that allows a once-weekly injection with comparable efficacy to insulin with an improved safety profile in type II diabetic people. Here, we studied the pharmacology of exenatide ER in 6 healthy cats. A single subcutaneous injection of exenatide ER (0.13 mg/kg) was administered on day 0. Exenatide concentrations were measured for 12 wk. A hyperglycemic clamp (target = 225 mg/dL) was performed on days -7 (clamp I) and 21 (clamp II) with measurements of insulin and glucagon concentrations. Glucose tolerance was defined as the amount of glucose required to maintain hyperglycemia during the clamp. Continuous glucose monitoring was performed on weeks 0, 2, and 6 after injection. Plasma concentrations of exenatide peaked at 1 h and 4 wk after injection. Comparing clamp I with clamp II, fasting blood glucose decreased (mean ± standard deviation = -11 ± 8 mg/dL, P = 0.02), glucose tolerance improved (median [range] +33% [4%-138%], P = 0.04), insulin concentrations increased (+36.5% [-9.9% to 274.1%], P = 0.02), and glucagon concentrations decreased (-4.7% [0%-12.1%], P = 0.005). Compared with preinjection values on continuous glucose monitoring, glucose concentrations decreased and the frequency of readings <50 mg/dL increased at 2 and 6 wk after injection of exenatide ER. This did not correspond to clinical hypoglycemia. No other side effects were observed throughout the study. Exenatide ER was safe and effective in improving glucose tolerance 3 wk after a single injection. Further evaluation is needed to determine its safety, efficacy, and duration of action in diabetic cats.
Collapse
Affiliation(s)
- A J Rudinsky
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - C A Adin
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - S Borin-Crivellenti
- FAPESP (#2013/00027-6) and FCAV, Universidade Estadual Paulista (UNESP), Jaboticabal, Sao Paulo, Brazil
| | - P Rajala-Schultz
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - M J Hall
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - C Gilor
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
20
|
Gleizes C, Constantinescu A, Abbas M, Bouhadja H, Zobairi F, Kessler L, Toti F. Liraglutide protects Rin-m5f β cells by reducing procoagulant tissue factor activity and apoptosis prompted by microparticles under conditions mimicking Instant Blood-Mediated Inflammatory Reaction. Transpl Int 2014; 27:733-40. [DOI: 10.1111/tri.12286] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/02/2013] [Accepted: 02/14/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Céline Gleizes
- EA 7293; Vascular and Tissular Stress in Transplantation; Federation of Translational Medicine of Strasbourg; Faculty of Medicine; University of Strasbourg; Strasbourg France
| | - Andrei Constantinescu
- EA 7293; Vascular and Tissular Stress in Transplantation; Federation of Translational Medicine of Strasbourg; Faculty of Medicine; University of Strasbourg; Strasbourg France
- Department of Parasitology and Parasitic Diseases and Animal Biology; Faculty of Veterinary Medicine; University of Agronomical Sciences and Veterinary Medicine; Bucharest Romania
| | - Malak Abbas
- EA 7293; Vascular and Tissular Stress in Transplantation; Federation of Translational Medicine of Strasbourg; Faculty of Medicine; University of Strasbourg; Strasbourg France
- Faculty of Sciences; Lebanon University; Hadath Lebanon
| | - Houda Bouhadja
- Faculty of Pharmacy; University of Strasbourg; UMR7213 CNRS; Laboratory of Biophotonics and Pharmacology; Illkirch France
| | - Fatiha Zobairi
- EA 7293; Vascular and Tissular Stress in Transplantation; Federation of Translational Medicine of Strasbourg; Faculty of Medicine; University of Strasbourg; Strasbourg France
| | - Laurence Kessler
- EA 7293; Vascular and Tissular Stress in Transplantation; Federation of Translational Medicine of Strasbourg; Faculty of Medicine; University of Strasbourg; Strasbourg France
- Department of Diabetology; University Hospital; Strasbourg Cedex France
| | - Florence Toti
- Faculty of Pharmacy; University of Strasbourg; UMR7213 CNRS; Laboratory of Biophotonics and Pharmacology; Illkirch France
| |
Collapse
|
21
|
Chhabra P, Brayman KL. Overcoming barriers in clinical islet transplantation: current limitations and future prospects. Curr Probl Surg 2014; 51:49-86. [PMID: 24411187 DOI: 10.1067/j.cpsurg.2013.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
22
|
Marcovecchio ML, Chiarelli F. An update on the pharmacotherapy options for pediatric diabetes. Expert Opin Biol Ther 2014; 14:355-64. [PMID: 24387753 DOI: 10.1517/14712598.2014.874413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Diabetes mellitus is a frequent endocrine disease during childhood and adolescence. Achieving a good glycemic control is of paramount importance to avoid short- and long-term complications and to allow a normal growth and quality of life. AREAS COVERED This review offers an update on current available treatment strategies for type 1 and type 2 diabetes approved for use in children and adolescents. EXPERT OPINION Although many progresses have been made in the field of diabetes management in children and adolescents, there are still several problems to deal with. With regard to type 1 diabetes, insulin remains the main and essential therapeutic strategy. However, the main issue is to develop a system that allows more physiological insulin coverage and reduces the risk of hypoglycemia and weight gain. Adjunct therapies would be invaluable for patients struggling to achieve an acceptable glycemic control. Treatment of type 2 diabetes is based on lifestyle interventions and metformin is the first-line drug for children older than 10 years. As for type 1 diabetes, there is a strong need for developing new drugs to be used alone or in combination.
Collapse
Affiliation(s)
- M Loredana Marcovecchio
- University of Chieti, Department of Paediatrics , Via dei Vestini 5, 66100 Chieti , Italy +0039 0871 358015 ; +0039 0871 574538 ;
| | | |
Collapse
|
23
|
Effect of Liraglutide on endoplasmic reticulum stress in diabetes. Biochem Biophys Res Commun 2013; 441:133-8. [DOI: 10.1016/j.bbrc.2013.10.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 11/24/2022]
|
24
|
Wang Y, Qi M, McGarrigle JJ, Rady B, Davis M, Vaca P, Oberholzer J. Use of glucagon-like peptide-1 agonists to improve islet graft performance. Curr Diab Rep 2013; 13:723-32. [PMID: 23925432 PMCID: PMC3888204 DOI: 10.1007/s11892-013-0402-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human islet transplantation is an effective and promising therapy for type I diabetes. However, long-term insulin independence is both difficult to achieve and inconsistent. De novo or early administration of incretin-based drugs is being explored for improving islet engraftment. In addition to its glucose-dependent insulinotropic effects, incretins also lower postprandial glucose excursion by inhibiting glucagon secretion, delaying gastric emptying, and can protect beta-cell function. Incretin therapy has so far proven clinically safe and tolerable with little hypoglycemic risk. The present review aims to highlight the new frontiers in research involving incretins from both in vitro and in vivo animal studies in the field of islet transplant. It also provides an overview of the current clinical status of incretin usage in islet transplantation in the management of type I diabetes.
Collapse
Affiliation(s)
- Yong Wang
- . 312-996-0851(W), 312-996-7913(Fax). Department of Surgery/Transplant, University of Illinois at Chicago, Chicago, IL 60612
| | - Meirigeng Qi
- . 312-996-0530(W), 312-996-7913(Fax). Department of Surgery/Transplant, University of Illinois at Chicago, Chicago, IL 60612
| | - James J. McGarrigle
- . 312-996-8316(W), 312-996-7913(Fax). Department of Surgery/Transplant, University of Illinois at Chicago, Chicago, IL 60612
| | - Brian Rady
- 312-996-8316(W), 312-996-7913(Fax). Department of Surgery/Transplant, University of Illinois at Chicago, Chicago, IL 60612
| | - Maureen Davis
- . 312-996-8316(W), 312-996-7913(Fax). Department of Surgery/Transplant, University of Illinois at Chicago, Chicago, IL 60612
| | - Pilar Vaca
- . 312-996-8316(W), 312-996-7913(Fax). Department of Surgery/Transplant, University of Illinois at Chicago, Chicago, IL 60612
| | - Jose Oberholzer
- . 312-996-6771(W), 312-996-7961(Fax). Department of Surgery/Transplant, University of Illinois at Chicago, Chicago, IL 60612
| |
Collapse
|
25
|
Abstract
Early innate inflammatory reaction strongly affects islet engraftment and survival after intrahepatic transplantation. This early immune response is triggered by ischemia-reperfusion injury and instant blood mediated inflammatory reaction (IBMIR) occurring hours and days after islet infusion. Evidence in both mouse model and in human counterpart suggest the involvement of coagulation, complement system, and proinflammatory chemokines/cytokines. Identification and targeting of pathway(s), playing a role as "master regulator(s)" in post-transplant detrimental inflammatory events, is now mandatory to improve islet transplantation success. This review will focus on inflammatory pathway(s) differentially modulated by islet isolation and mainly associated with the early post-transplant events. Moreover, we will take into account anti-inflammatory strategies that have been tested at 2 levels: on the graft, ex vivo, during islet culture (i.e., donor) and/or on the graft site, in vivo, early after islet infusion (i.e., recipient).
Collapse
Affiliation(s)
- Antonio Citro
- Beta Cell Biology Unit, Diabetes Research Institute, San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy,
| | | | | |
Collapse
|
26
|
Abstract
Clinical islet transplantation has progressed considerably over the past 12 years, and >750 patients with type 1 diabetes have received islet transplants internationally over this time. Many countries are beginning to accept the transition from research to accepted and funded clinical care, especially for patients with brittle control that cannot be stabilized by more conventional means. Major challenges remain, including the need for more than one donor, and the requirement for potent, chronic immunosuppression. Combining immunological tolerance both to allo- and autoantigens, and a limitless expandable source of stem cell- or xenograft-derived insulin-secreting cells represent remaining hurdles in moving this effective treatment to a potential cure for all those with type 1 or 2 diabetes.
Collapse
Affiliation(s)
- Michael McCall
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, Alberta T6G 2B7, Canada
| | | |
Collapse
|
27
|
Shapiro AMJ. Islet transplantation in type 1 diabetes: ongoing challenges, refined procedures, and long-term outcome. Rev Diabet Stud 2012; 9:385-406. [PMID: 23804275 DOI: 10.1900/rds.2012.9.385] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Remarkable progress has been made in islet transplantation over a span of 40 years. Once just an experimental curiosity in mice, this therapy has moved forward, and can now provide robust therapy for highly selected patients with type 1 diabetes (T1D), refractory to stabilization by other means. This progress could not have occurred without extensive dynamic international collaboration. Currently, 1,085 patients have undergone islet transplantation at 40 international sites since the Edmonton Protocol was reported in 2000 (752 allografts, 333 autografts), according to the Collaborative Islet Transplant Registry. The long-term results of islet transplantation in selected centers now match registry data of pancreas-alone transplantation, with 6 sites reporting five-year insulin independence rates ≥50%. Islet transplantation has been criticized for the use of multiple donor pancreas organs, but progress has also occurred in single-donor success, with 10 sites reporting increased single-donor engraftment. The next wave of innovative clinical trial interventions will address instant blood-mediated inflammatory reaction (IBMIR), apoptosis, and inflammation, and will translate into further marked improvements in single-donor success. Effective control of auto- and alloimmunity is the key to long-term islet function, and high-resolution cellular and antibody-based assays will add considerable precision to this process. Advances in immunosuppression, with new antibody-based targeting of costimulatory blockade and other T-B cellular signaling, will have further profound impact on the safety record of immunotherapy. Clinical trials will move forward shortly to test out new human stem cell derived islets, and in parallel trials will move forward, testing pig islets for compatibility in patients. Induction of immunological tolerance to self-islet antigens and to allografts is a difficult challenge, but potentially within our grasp.
Collapse
Affiliation(s)
- A M James Shapiro
- Clinical Islet Transplant Program, University of Alberta, 2000 College Plaza, 8215 112th Street, Edmonton AB Canada T6G 2C8.
| |
Collapse
|
28
|
Mori Y, Ohtsuka T, Tsutsumi K, Yasui T, Ueda J, Takahata S, Nakamura M, Tanaka M. Different incretin responses after pancreatoduodenectomy and distal pancreatectomy. Pancreas 2012; 41:455-60. [PMID: 22422137 DOI: 10.1097/mpa.0b013e3182319d7c] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are known as incretins to stimulate insulin secretion. The aims of this study were to investigate the postoperative β-cell function and hormonal responses of GLP-1 and GIP after pancreatoduodenectomy (PD) and distal pancreatectomy (DP). METHODS Oral glucose tolerance tests were performed in 34 patients (20 PD and 14 DP) before and 1 month after operation. The changes in the serum glucose and insulin concentrations, homeostasis model assessment of insulin resistance, and pancreatic β-cell function (BCF) were analyzed. GLP-1 and GIP were also measured. RESULTS There was no patient with postoperative deterioration of glucose tolerance after PD, whereas impairment of glucose metabolism was observed after DP. Homeostasis model assessment of insulin resistance decreased after PD, whereas those after DP showed no change. The postoperative BCF were lower than preoperative values in both groups. GLP-1 increased after DP but not after PD, whereas GIP decreased after PD but not after DP. CONCLUSIONS The changes in glucose metabolism and incretin responses were different between PD and DP. The increased level of GLP-1 after DP might reflect the relatively insufficient BCF; and thus, perioperative administration of GLP-1 might improve the diabetic condition after DP.
Collapse
Affiliation(s)
- Yasuhisa Mori
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Affiliation(s)
- A J Garber
- Division of Endocrinology, Diabetes and Metabolism Baylor College of Medicine Houston, TX 77030, USA.
| |
Collapse
|
30
|
Advances and challenges in islet transplantation: islet procurement rates and lessons learned from suboptimal islet transplantation. J Transplant 2011; 2011:979527. [PMID: 22235361 PMCID: PMC3253477 DOI: 10.1155/2011/979527] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Accepted: 10/04/2011] [Indexed: 01/22/2023] Open
Abstract
The initial step in successful islet transplantation is procurement of healthy donor islets. Given the limited number of donor pancreata selected for islet isolation and that islets from multiple donors are typically required to obtain insulin independence, it is critical to improve pancreas procurement rates and yield of islets for transplantation. Islets are delicate microorgans that are susceptible to apoptosis, hypoxia, and ischemia during isolation, culture, and the peritransplant period. Once the islets are engrafted, both prompt revascularization and protection from beta-cell death and graft rejection are key to secure long-term survival and function. To facilitate the engraftment of more robust islets suitable for combating the challenging isolation period and proinflammatory transplantation milieu, numerous approaches have been employed to prevent beta-cell dysfunction and death including immune modulation, prevention of apoptosis and hypoxia, as well as stimulation of growth factors, angiogenesis, and reinnervation. In addition to briefly discussing islet isolation procedures, procurement rates, and islet transplantation, the relevant literature pertaining to successful suboptimal islet transplantation is reviewed to provide insight into potential approaches to balance the limited supply of available donor islets.
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW The current review addresses a critical need in clinical islet transplantation, namely the routine transition from the requirement of two to four donors down to one donor per recipient. The ability to achieve single-donor islet transplantation will provide many more islet grafts for treatment of an ever-expanding patient base with type 1 diabetes (T1DM) with poor glycemic control. Avoiding exposure of recipients to multiple different donor human leukocyte associated (HLA) antigens is critical if risk of donor sensitization is to be avoided. This point is important as further islet or pancreas transplants in the remote future or the potential future need for a solid organ kidney transplant may become prohibitive if the recipient is sensitized. RECENT FINDINGS This review addresses systematically all areas that contribute to the success or failure of single-donor islet engraftment, beginning with donor-related factors, optimizing islet isolation and culture conditions, and describes a series of strategies in the treatment of the recipient to prevent inflammation, apoptosis, islet thrombosis, and improve metabolic functional outcome, all of which will lead to improved single-donor engraftment success. SUMMARY If single-donor islet transplantation can be achieved routinely, therapy will become more widely available, more accepted by the transplant community (currently pancreas transplantation requires only a single donor), and this situation will have a major impact overall as an effective treatment option in T1DM.
Collapse
Affiliation(s)
- A M James Shapiro
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
32
|
Maciver AH, McCall MD, Edgar RL, Thiesen AL, Bigam DL, Churchill TA, Shapiro AJ. Sirolimus drug-eluting, hydrogel-impregnated polypropylene mesh reduces intra-abdominal adhesion formation in a mouse model. Surgery 2011; 150:907-15. [DOI: 10.1016/j.surg.2011.06.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 06/15/2011] [Indexed: 11/26/2022]
|
33
|
Merani S, McCall M, Pawlick RL, Edgar RL, Davis J, Toso C, Emamaullee JA, Kin T, Shapiro AMJ. AEB071 (sotrastaurin) does not exhibit toxic effects on human islets in vitro, nor after transplantation into immunodeficient mice. Islets 2011; 3:338-43. [PMID: 21934354 DOI: 10.4161/isl.3.6.17766] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AEB071 (AEB, sotrastaurin), a specific inhibitor of protein kinase C, reduces T-lymphocyte activation and cytokine release. AEB delays islet allograft rejection in rats and prevents rejection when combined with cyclosporine. Since many immunosuppressive agents have toxic effects on the function of transplanted islets, we investigated whether this was also the case with AEB. Human islets were transplanted into Rag-knockout mice randomly assigned to vehicle control, AEB or sirolimus treatment groups. Non-fasting blood glucose levels, body weight and glucose tolerance was measured in recipients. In a separate experiment, human islets were cultured in the presence of AEB and assayed for glucose dependent insulin secretion and level of β-cell apoptosis. Eighty-six percent of the AEB-treated recipients achieved normoglycemia following transplant (compared with none in sirolimus-treated group, p < 0.05). AEB-treated recipients exhibited similar glucose homeostasis as vehicle-treated controls, which was better than in sirolimus-treated recipients. Human islets cultured with AEB showed similar rates of β-cell apoptosis (p = 0.98 by one-way ANOVA) and glucose stimulated insulin secretion (p = 0.15) as those cultured with vehicle. These results suggest that AEB is not associated with toxic effects on islet engraftment or function. AEB appears to be an appropriate immunosuppressive candidate for clinical trials in islet transplantation.
Collapse
Affiliation(s)
- Shaheed Merani
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Clinical islet transplantation has transitioned from curiosity to realistic therapy over the past decade. An estimated 750 patients have undergone intraportal islet-alone transplantation over this period, and a smaller subset received combined islet-kidney transplants. The primary benefit of successful islet transplantation has been to eliminate severe, recurrent hypoglycemia, a problem that has been hard to eliminate by other means in 15% of those with type 1 diabetes. The secondary benefit of independence from insulin has attracted patients, but has had limited sustainability previously, especially with a single-donor graft, but recent results from four independent centers suggest marked improvement in long-term outcome, with 5-year results now approximating solitary pancreas transplantation. Emerging data confirm that islet transplantation can stabilize and reverse several secondary diabetic complications similar to whole pancreas transplantation, but larger, head-to-head trials are needed to compare islet transplantation with best medical therapies. Current goals are to extend durability, and to make islet transplantation more widely available for patients in need. Governmental and health insurance providers in several countries now reimburse islet transplantation as part of clinical care. As the safety of the procedure and of adjunctive immunosuppressive therapies improve, and benefit accrues over potential risk, islet transplantation will be offered earlier in the course of the disease, including newly diagnosed children. The role of islet transplantation in type 2 diabetes has yet to be defined. We review the current status of islet transplantation, and discuss current and future immunosuppressive protocols that will pave the way to more broad application of cellular replacement in diabetes.
Collapse
Affiliation(s)
- A M James Shapiro
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB T6G 2C8, Canada.
| |
Collapse
|
35
|
The efficacy and safety of liraglutide. Int J Clin Pharm 2011; 33:740-9. [DOI: 10.1007/s11096-011-9552-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Accepted: 08/07/2011] [Indexed: 11/25/2022]
|
36
|
Grover GJ, Koetzner L, Wicks J, Gahler RJ, Lyon MR, Reimer RA, Wood S. Effects of the Soluble Fiber Complex PolyGlycopleX on Glucose Homeostasis and Body Weight in Young Zucker Diabetic Rats. Front Pharmacol 2011; 2:47. [PMID: 21922008 PMCID: PMC3168379 DOI: 10.3389/fphar.2011.00047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Accepted: 07/26/2011] [Indexed: 11/13/2022] Open
Abstract
Dietary fiber can reduce insulin resistance, body weight, and hyperlipidemia depending on fiber type, water solubility, and viscosity. PolyGlycopleX(®) (PGX(®)) is a natural, novel water soluble, non-starch polysaccharide complex that with water forms a highly viscous gel compared to other naturally occurring dietary fiber. We determined the effect of dietary PGX(®) vs. cellulose and inulin on the early development of insulin resistance, body weight, hyperlipidemia, and glycemia-induced tissue damage in young Zucker diabetic rats (ZDFs) in fasted and non-fasted states. ZDFs (5 weeks old) were fed a diet containing 5% (wgt/wgt) cellulose, inulin, or PGX(®) for 8 weeks. Body weight, lipids, insulin, and glucose levels were determined throughout the study and homeostasis model assessment (HOMA) was used to measure insulin sensitivity throughout the study in fasted animals. At study termination, insulin sensitivity (oral glucose tolerance test, OGTT) and kidney, liver, and pancreatic histopathology were determined. Body weight and food intake were significantly reduced by PGX(®) vs. inulin and cellulose. Serum insulin in fasted and non-fasted states was significantly reduced by PGX(®) as was non-fasted blood glucose. Insulin resistance, measured as a HOMA score, was significantly reduced by PGX(®) in weeks 5 through 8 as well as terminal OGTT scores in fed and fasted states. Serum total cholesterol was also significantly reduced by PGX(®). PGX(®) significantly reduced histological kidney and hepatic damage in addition to reduced hepatic steatosis and cholestasis. A greater mass of pancreatic β-cells was found in the PGX(®) group. PGX(®) therefore may be a useful dietary additive in the control of the development of the early development of the metabolic syndrome.
Collapse
Affiliation(s)
- Gary James Grover
- Department of Pharmacology, Eurofins-Product Safety LaboratoriesDayton, NJ, USA
- Department of Physiology and Biophysics, Robert Wood Johnson Medical SchoolPiscataway, NJ, USA
| | - Lee Koetzner
- Department of Pharmacology, Eurofins-Product Safety LaboratoriesDayton, NJ, USA
| | | | | | - Michael R. Lyon
- Canadian Centre for Functional MedicineCoquitlam, BC, Canada
- Food, Nutrition and Health Program, University of British ColumbiaVancouver, BC, Canada
| | - Raylene A. Reimer
- Faculty of Kinesiology, Department of Biochemistry and Molecular Biology, University of CalgaryCalgary, AB, Canada
| | - Simon Wood
- Canadian Centre for Functional MedicineCoquitlam, BC, Canada
| |
Collapse
|
37
|
Suen CS, Burn P. The potential of incretin-based therapies in type 1 diabetes. Drug Discov Today 2011; 17:89-95. [PMID: 21920456 DOI: 10.1016/j.drudis.2011.08.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 08/25/2011] [Accepted: 08/30/2011] [Indexed: 02/07/2023]
Abstract
Finding a cure for type 1 diabetes (T1D) has been elusive. Incretin-based therapies, since their approval, have demonstrated their clinical utilities in type 2 diabetes (T2D). Yet, their potential clinical benefits in T1D remain to be appraised. GLP-1, in addition to its insulinotropic action in alleviating hyperglycemia, possesses beneficial effects in protecting progressive impairment of pancreatic β-cell function, preservation of β-cell mass and suppression of glucagon secretion, gastric emptying and appetite. Preclinical data using incretin-based therapies in diabetic NOD mice demonstrated additional effects including immuno-modulation, anti-inflammation and β-cell regeneration. Thus, data accumulated hold the promise that incretin-based therapies may be effective in delaying the new-onset, halting the further progression, or reversing T1D in subjects with newly diagnosed or long-standing, established disease.
Collapse
Affiliation(s)
- Chen S Suen
- The Sanford Project, Sanford Research, Sanford Health and Department of Pediatrics, Sanford School of Medicine of The University of South Dakota, 2301 East 60th Street North, Sioux Falls, SD 57104, USA
| | | |
Collapse
|
38
|
Abstract
Management guidelines recommend metformin as the first-line therapy for most patients with type 2 diabetes uncontrolled by diet and exercise. Efficacy with metformin therapy is usually of limited duration, which necessitates the early introduction of one or two additional oral agents or the initiation of injections, glucagon-like peptide-1 (GLP-1) agonists or insulin. Although safe and effective, metformin monotherapy has been associated with gastrointestinal side effects (≈20% of treated patients in randomized studies) and is contraindicated in patients with renal insufficiency or severe liver disease. Patients treated with a sulphonylurea are at increased risk for hypoglycaemia and moderate weight gain, whereas those receiving a thiazolidinedione are subject to an increased risk of weight gain, oedema, heart failure or fracture. Weight gain and hypoglycaemia are associated with insulin use. Thus, there is an unmet need for a safe and efficacious add-on agent after initial-therapy failure. Evidence suggests that incretin-based agents, such as GLP-1 receptor agonists and dipeptidyl peptidase-4 inhibitors, can successfully achieve glycaemic targets and potentially provide cardiovascular and β-cell-function benefits. This review will examine current approaches for treating type 2 diabetes and discuss the place of incretin therapies, mainly GLP-1 agonists, in the type 2 diabetes treatment spectrum.
Collapse
Affiliation(s)
- B Charbonnel
- Department of Endocrinology and Diabetes, University Hospital, Nantes, France.
| | | |
Collapse
|
39
|
Van Belle TL, Coppieters KT, Von Herrath MG. Type 1 Diabetes: Etiology, Immunology, and Therapeutic Strategies. Physiol Rev 2011; 91:79-118. [DOI: 10.1152/physrev.00003.2010] [Citation(s) in RCA: 673] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease in which destruction or damaging of the beta-cells in the islets of Langerhans results in insulin deficiency and hyperglycemia. We only know for sure that autoimmunity is the predominant effector mechanism of T1D, but may not be its primary cause. T1D precipitates in genetically susceptible individuals, very likely as a result of an environmental trigger. Current genetic data point towards the following genes as susceptibility genes: HLA, insulin, PTPN22, IL2Ra, and CTLA4. Epidemiological and other studies suggest a triggering role for enteroviruses, while other microorganisms might provide protection. Efficacious prevention of T1D will require detection of the earliest events in the process. So far, autoantibodies are most widely used as serum biomarker, but T-cell readouts and metabolome studies might strengthen and bring forward diagnosis. Current preventive clinical trials mostly focus on environmental triggers. Therapeutic trials test the efficacy of antigen-specific and antigen-nonspecific immune interventions, but also include restoration of the affected beta-cell mass by islet transplantation, neogenesis and regeneration, and combinations thereof. In this comprehensive review, we explain the genetic, environmental, and immunological data underlying the prevention and intervention strategies to constrain T1D.
Collapse
Affiliation(s)
- Tom L. Van Belle
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Ken T. Coppieters
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Matthias G. Von Herrath
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| |
Collapse
|
40
|
Abstract
Type 2 diabetes occurs due to a relative deficit in β-cell mass or function. Glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), cholecystokinin (CCK), and gastrin are gastrointestinal hormones that are secreted in response to nutrient intake, regulating digestion, insulin secretion, satiety, and β-cell mass. In this review, we focus upon β-cell mass regulation. β-cell mass expands through β-cell proliferation and islet neogenesis; β-cell mass is lost via apoptosis. GLP-1 and GIP are well-studied gastrointestinal hormones and influence β-cell proliferation, apoptosis, and islet neogenesis. CCK regulates β-cell apoptosis and mitogenesis, and gastrin stimulates islet neogenesis. GLP-1 and GIP bind to G protein-coupled receptors and regulate β-cell mass via multiple signaling pathways. The protein kinase A pathway is central to this process because it directly regulates proliferative and anti-apoptotic genes and transactivates several signaling cascades, including Akt and mitogen-activated protein kinases. However, the signaling pathways downstream of G protein-coupled CCK receptors that influence β-cell mass remain unidentified. Gastrointestinal hormones integrate nutrient signals from the gut to the β-cell, regulating insulin secretion and β-cell mass adaptation.
Collapse
Affiliation(s)
- Jeremy A Lavine
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | |
Collapse
|
41
|
Cummings BP, Stanhope KL, Graham JL, Baskin DG, Griffen SC, Nilsson C, Sams A, Knudsen LB, Raun K, Havel PJ. Chronic administration of the glucagon-like peptide-1 analog, liraglutide, delays the onset of diabetes and lowers triglycerides in UCD-T2DM rats. Diabetes 2010; 59:2653-61. [PMID: 20622169 PMCID: PMC3279561 DOI: 10.2337/db09-1564] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The efficacy of liraglutide, a human glucagon-like peptide-1 (GLP-1) analog, to prevent or delay diabetes in UCD-T2DM rats, a model of polygenic obese type 2 diabetes, was investigated. RESEARCH DESIGN AND METHODS At 2 months of age, male rats were divided into three groups: control, food-restricted, and liraglutide. Animals received liraglutide (0.2 mg/kg s.c.) or vehicle injections twice daily. Restricted rats were food restricted to equalize body weights to liraglutide-treated rats. Half of the animals were followed until diabetes onset, whereas the other half of the animals were killed at 6.5 months of age for tissue collection. RESULTS Before diabetes onset energy intake, body weight, adiposity, and liver triglyceride content were higher in control animals compared with restricted and liraglutide-treated rats. Energy-restricted animals had lower food intake than liraglutide-treated animals to maintain the same body weights, suggesting that liraglutide increases energy expenditure. Liraglutide treatment delayed diabetes onset by 4.1 ± 0.8 months compared with control (P < 0.0001) and by 1.3 ± 0.8 months compared with restricted animals (P < 0.05). Up to 6 months of age, energy restriction and liraglutide treatment lowered fasting plasma glucose and A1C concentrations compared with control animals. In contrast, liraglutide-treated animals exhibited lower fasting plasma insulin, glucagon, and triglycerides compared with both control and restricted animals. Furthermore, energy-restricted and liraglutide-treated animals exhibited more normal islet morphology. CONCLUSIONS Liraglutide treatment delays the development of diabetes in UCD-T2DM rats by reducing energy intake and body weight, and by improving insulin sensitivity, improving lipid profiles, and maintaining islet morphology.
Collapse
Affiliation(s)
- Bethany P. Cummings
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California
- Department of Nutrition, University of California, Davis, Davis, California
| | - Kimber L. Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California
- Department of Nutrition, University of California, Davis, Davis, California
| | - James L. Graham
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California
- Department of Nutrition, University of California, Davis, Davis, California
| | - Denis G. Baskin
- Research and Development Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, Washington, and the Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, Washington
| | - Steven C. Griffen
- Department of Internal Medicine, University of California, Davis, Sacramento, California
| | - Cecilia Nilsson
- Biology and Pharmacology Management, Novo Nordisk, Maaloev, Denmark
| | - Anette Sams
- Diabetes Inflammation Department, Novo Nordisk, Gentofte, Denmark
| | - Lotte B. Knudsen
- Biology and Pharmacology Management, Novo Nordisk, Maaloev, Denmark
| | - Kirsten Raun
- Biology and Pharmacology Management, Novo Nordisk, Maaloev, Denmark
| | - Peter J. Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California
- Department of Nutrition, University of California, Davis, Davis, California
- Corresponding author: Peter J. Havel,
| |
Collapse
|
42
|
Affiliation(s)
- Thierry Berney
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|
43
|
Toso C, McCall M, Emamaullee J, Merani S, Davis J, Edgar R, Pawlick R, Kin T, Knudsen LB, Shapiro AMJ. Liraglutide, a long-acting human glucagon-like peptide 1 analogue, improves human islet survival in culture. Transpl Int 2010; 23:259-65. [PMID: 19821955 DOI: 10.1111/j.1432-2277.2009.00984.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The culture of human islets is associated with approximately 10-20% islet loss, occasionally preventing transplantation. Preconditioning of the islets to improve postculture yields would be of immediate benefit, with the potential to increase both the number of transplanted patients and their metabolic reserve. In this study, the effect of liraglutide, a long-acting human glucagon-like peptide 1 analogue, on cultured human islets was examined. Culture with liraglutide (1 micromol/l) was associated with a preservation of islet mass (significantly more islets at 24 and 48 h, compared to control; P < or = 0.05 at 24 and 48 h) and with the presence of larger islets (P < or = 0.05 at 48 h). These observations were supported by reduced apoptosis rates after 24 h of treatment. We also demonstrated that human islet engraftment is improved in C57Bl/6-RAG(-/-) mice treated with liraglutide 200 microg/kg sc twice daily (P < or = 0.05), suggesting that liraglutide should be continued after transplantation. Overall, these data demonstrate the beneficial effect of liraglutide on cultured human islets, preserving islet mass. They support the design of clinical studies looking at the effect of liraglutide in clinical islet transplantation.
Collapse
Affiliation(s)
- Christian Toso
- Clinical Islet Transplant Program, Department of Surgery, University of Alberta, Edmonton, AB, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Montanya E, Sesti G. A review of efficacy and safety data regarding the use of liraglutide, a once-daily human glucagon-like peptide 1 analogue, in the treatment of type 2 diabetes mellitus. Clin Ther 2009; 31:2472-88. [PMID: 20109994 DOI: 10.1016/j.clinthera.2009.11.034] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2009] [Indexed: 11/19/2022]
Affiliation(s)
- Eduard Montanya
- Endocrine Unit, University Hospital of Bellvitge, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain.
| | | |
Collapse
|
45
|
Abstract
Glucose-dependent insulinotropic polypeptide (GIP or gastric inhibitory polypeptide) is a 42-amino-acid hormone, secreted from the enteroendocrine K cells, which has insulin-releasing and extrapancreatic glucoregulatory actions. However, the unfavourable pharmacokinetic profile and the weak biological effects of native GIP limit its effectiveness for the treatment of type 2 diabetes. To overcome this, longer-acting GIP agonists exhibiting enzymatic stability and enhanced bioactivity have been generated and successfully tested in animal models of diabetes. Thus, GIP receptor agonists offer one of the newest classes of potential antidiabetic drug. GIP is also known to play a role in lipid metabolism and fat deposition. Accordingly, both genetic and chemical ablation of GIP signalling in mice with obesity-diabetes can protect against, or even reverse many of the obesity-associated metabolic disturbances. Strong parallels exist with the beneficial metabolic effects of Roux-en-Y gastric bypass in obese, insulin-resistant humans that surgically ablates GIP-secreting K cells. The purpose of this article is to highlight the therapeutic potential of GIP-based therapeutics in the treatment of type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Nigel Irwin
- School of Biomedical Sciences, University of Ulster, Cromore Road, Coleraine, Co. Londonderry, BT52 1SA, Northern Ireland, UK.
| | | |
Collapse
|
46
|
Emamaullee JA, Merani S, Toso C, Kin T, Al-Saif F, Truong W, Pawlick R, Davis J, Edgar R, Lock J, Bonner-Weir S, Knudsen LB, Shapiro AMJ. Porcine marginal mass islet autografts resist metabolic failure over time and are enhanced by early treatment with liraglutide. Endocrinology 2009; 150:2145-52. [PMID: 19131571 DOI: 10.1210/en.2008-1116] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although insulin independence is maintained in most islet recipients at 1 yr after transplant, extended follow-up has revealed that many patients will eventually require insulin therapy. Previous studies have shown that islet autografts are prone to chronic failure in large animals and humans, suggesting that nonimmunological events contribute to islet graft functional decay. Early intervention with therapies that promote graft stability should provide a measurable benefit over time. In this study, the efficacy of the long-acting glucagon-like peptide-1 analog liraglutide was explored in a porcine marginal mass islet autograft transplant model. Incubation with liraglutide enhanced porcine islet survival and function after prolonged culture. Most vehicle-treated (83%) and liraglutide-treated (80%) animals became insulin independent after islet autotransplantation. Although liraglutide therapy did not improve insulin independence rates or blood glucose levels after transplant, a significant increase in insulin secretion and acute-phase insulin response was observed in treated animals. Surprisingly, no evidence for deterioration of graft function was observed in any of the transplanted animals over more than 18 months of follow-up despite significant weight gain; in fact, an enhanced response to glucose developed over time even in control animals. Histological analysis showed that intraportally transplanted islets remained highly insulin positive, retained alpha-cells, and did not form amyloid deposits. This study demonstrates that marginal mass porcine islet autografts have stable long-term function, even in the presence of an increasing metabolic demand. These results are discrepant with previous large animal studies and suggest that porcine islets may be resistant to metabolic failure.
Collapse
Affiliation(s)
- Juliet A Emamaullee
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada T6G 2E1.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine produced by many tissues including pancreatic beta-cells. METHODS This study investigates the impact of MIF on islet transplantation using MIF knock-out (MIFko) mice. RESULTS Early islet function, assessed with a syngeneic marginal islet mass transplant model, was enhanced when using MIFko islets (P<0.05 compared with wild-type [WT] controls). This result was supported by increased in vitro resistance of MIFko islets to apoptosis (terminal deoxynucleotide tranferase-mediated dUTP nick-end labeling assay), and by improved glucose metabolism (lower blood glucose levels, reduced glucose areas under curve and higher insulin release during intraperitoneal glucose challenges, and in vitro in the absence of MIF, P<0.01). The beneficial impact of MIFko islets was insufficient to delay allogeneic islet rejection. However, the rejection of WT islet allografts was marginally delayed in MIFko recipients by 6 days when compared with WT recipient (P<0.05). This effect is supported by the lower activity of MIF-deficient macrophages, assessed in vitro and in vivo by cotransplantation of islet/macrophages. Leukocyte infiltration of the graft and donor-specific lymphocyte activity (mixed lymphocyte reaction, interferon gamma ELISPOT) were similar in both groups. CONCLUSION These data indicate that targeting MIF has the potential to improve early function after syngeneic islet transplantation, but has only a marginal impact on allogeneic rejection.
Collapse
|