1
|
Cai X, Zhang M, Zou J, Wang L, Zhan Y, Li D, Jiang T, Alim N, Liu Z, Yang J, Liu N, Liu T, Sun P, Ding X, Zhang B, Liu Z, Wang X, Liang R, Cai J, Gao J, Cao J, Wang S. A novel self-assembling peptide nanofiber hydrogel with glucagon-like peptide-1 functionality enhances islet survival to improve islet transplantation outcome in diabetes treatment. J Nanobiotechnology 2024; 22:792. [PMID: 39716238 DOI: 10.1186/s12951-024-03072-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024] Open
Abstract
Islet transplantation is a promising therapy for diabetes, yet the limited survival and functionality of transplanted islet grafts hinder optimal outcomes. Glucagon-like peptide-1 (GLP-1), an endogenous hormone, has shown potential to enhance islet survival and function; however, its systemic administration can result in poor localization and undesirable side effects. To address these challenges, we developed a novel peptide-based nanofiber hydrogel incorporating GLP-1 functionality for localized delivery. By conjugating the FFG tripeptide (a self-assembling motif derived from phenylalanine-phenylalanine-glycine) to the C-terminus of native GLP-1, we engineered GLP-1-FFG, a self-assembling peptide that forms a robust nanofiber structure resistant to enzymatic degradation. When GLP-1-FFG co-assembles with the biotin-DFYIGSRGD peptide (referred to as SupraGel), a self-assembling supramolecular polypeptide hydrogel we previously identified containing motifs derived from extracellular matrix components, the resulting hydrogel (SupraGel + GLP-1-FFG) creates a stable nanofibrous network with excellent rheological properties. In vitro, this nanofiber hydrogel significantly improves islet function and survival. Bulk RNA sequencing results demonstrate that the hydrogel suppresses the expression of hypoxia-related genes, downregulates pro-inflammatory genes, and upregulates genes associated with islet function. Further analysis reveals that these effects are related to the activation of the AKT signaling pathway. In a syngeneic mouse islet transplantation model, the localized application of SupraGel + GLP-1-FFG at the renal subcapsular islet transplant site significantly enhanced the efficacy of marginal-dose islet transplantation, as shown by improved glycemic control, faster and higher rates of diabetes reversal, better glucose tolerance, and greater islet graft survival in diabetic recipient mice. This innovative nanotechnology-based hydrogel offers a promising strategy for enhancing the efficacy of islet grafts in transplantation therapy.
Collapse
Affiliation(s)
- Xiangheng Cai
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
- Organ Transplantation Center, The Institute of Transplantation Science, The Affiliated Hospital of Qingdao University, Qingdao, 266035, China
| | - Mengnan Zhang
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jiaqi Zou
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Le Wang
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Yixiang Zhan
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Dandan Li
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Tingsheng Jiang
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Nijat Alim
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Zhaoce Liu
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Jiuxia Yang
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Na Liu
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Tengli Liu
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Peng Sun
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Xuejie Ding
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Boya Zhang
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Zewen Liu
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Xuelian Wang
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Rui Liang
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China
| | - Jinzhen Cai
- Organ Transplantation Center, The Institute of Transplantation Science, The Affiliated Hospital of Qingdao University, Qingdao, 266035, China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Jinglin Cao
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China.
| | - Shusen Wang
- NHC Key Laboratory for Critical Care Medicine, School of Medicine, Tianjin First Central Hospital, Research Institute of Transplant Medicine, Organ Transplant Center, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
2
|
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
|
3
|
Durak A, Turan B. Liraglutide provides cardioprotection through the recovery of mitochondrial dysfunction and oxidative stress in aging hearts. J Physiol Biochem 2022:10.1007/s13105-022-00939-9. [PMID: 36515811 DOI: 10.1007/s13105-022-00939-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 12/03/2022] [Indexed: 12/15/2022]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists improve cardiovascular dysfunction via the pleiotropic effects behind their receptor action. However, it is unknown whether they have a cardioprotective action in the hearts of the elderly. Therefore, we examined the effects of GLP-1R agonist liraglutide treatment (LG, 4 weeks) on the systemic parameters of aged rats (24-month-old) compared to those of adult rats (6-month-old) such as electrocardiograms (ECGs) and systolic and diastolic blood pressure (SBP and DBP). At the cellular level, the action potential (AP) parameters, ionic currents, and Ca2+ regulation were examined in freshly isolated ventricular cardiomyocytes. The LG treatment of aged rats significantly ameliorated the prolongation of QRS duration and increased both SBP and DBP together with recovery in plasma oxidant and antioxidant statuses. The prolonged AP durations and depolarized membrane potentials of the isolated cardiomyocytes from the aged rats were normalized via recoveries in K+ channel currents with LG treatment. The alterations in Ca2+ regulation including leaky-ryanodine receptors (RyR2) could be also ameliorated via recoveries in Na+/Ca2+ exchanger currents with this treatment. A direct LG treatment of isolated aged rat cardiomyocytes could recover the depolarized mitochondrial membrane potential, the increase in both reactive oxygen and nitrogen species (ROS and RNS), and the cytosolic Na+ level, although the Na+ channel currents were not affected by aging. Interestingly, LG treatment of aged rat cardiomyocytes provided a significant inhibition of activated sodium-glucose co-transporter-2 (SGLT2) and recoveries in the depressed insulin receptor substrate 1 (IRS1) and increased protein kinase G (PKG). The recovery in the ratio of phospho-endothelial nitric oxide (pNOS3) level to NOS3 protein level in LG-treated cardiomyocytes implies the involvement of LG-associated inhibition of oxidative stress-induced injury via IRS1-eNOS-PKG pathway in the aging heart. Overall, our data, for the first time, provide important information on the direct cardioprotective effects of GLP-1R agonism with LG in the hearts of aged rats through an examination of recoveries in mitochondrial dysfunction, and both levels of ROS and RNS in left ventricular cardiomyocytes.
Collapse
Affiliation(s)
- Aysegul Durak
- Department of Biophysics, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Belma Turan
- Department of Biophysics, Lokman Hekim University Faculty of Medicine, Ankara, Turkey.
| |
Collapse
|
4
|
Shadboorestan A, Eftekhari S, Mottaghi- Dastjerdi N, Shahparvari R, Tarighi P, Jahandar H, Faghihi H, Montazeri H. Metformin exerts synergistic anti-proliferative effects with liraglutide in human umbilical vein endothelial cells (HUVECs). IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:506-513. [PMID: 35656067 PMCID: PMC9150808 DOI: 10.22038/ijbms.2022.64117.14117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 11/06/2022]
Abstract
Objectives Metformin (Met) and liraglutide (Lira) have been approved to treat type 2 diabetes mellitus and have cardioprotective effects. Materials and Methods Human umbilical vein endothelial cells (HUVECs) were incubated with Met, Lira, or their combination in this study. Results Results showed that the synergistic inhibitory effect of the two drugs on HUVECs proliferation was significant (75%) after 48 hr drug exposure. In addition, either Lira or Met alone had a marked tendency to inhibit the migration of HUVECs (42% and 39%). Almost a complete inhibition (97%) was demonstrated in combinational use after 48 hr treatment. After combining these two drugs, the apoptosis rate raised to 68%, which was a significant approval of synergistic apoptosis induction of Met and Lira. The combinational group indicated a substantial increase in VEGF, PDGF, and MMP-9 at 24 hr compared with the control. Conclusion This study showed that combination therapy with Lira and Met could effectively reduce cell proliferation, induce apoptosis, and inhibit cell migration in the HUVECs. This study provides evidence to support using Met in combination with Lira as a treatment option for patients with type-2 diabetes and cancer.
Collapse
Affiliation(s)
- Amir Shadboorestan
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- These authors contributed equally to this work
| | - Samane Eftekhari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- These authors contributed equally to this work
| | - Negar Mottaghi- Dastjerdi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Rezvan Shahparvari
- Pharmaceutical Sciences Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran Iran
| | - Parastoo Tarighi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hoda Jahandar
- Pharmaceutical Sciences Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran Iran
| | - Homa Faghihi
- Department of Pharmaceutics, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Montazeri
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
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: 6] [Impact Index Per Article: 1.5] [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
|
6
|
Wang L, Tang Y, He H, Wei W. Liraglutide restores late cardioprotective effects of remote preconditioning in diabetic rats via activation of hydrogen sulfide and nuclear factor erythroid 2-related factor 2 signaling pathway. Acta Cir Bras 2021; 36:e360207. [PMID: 33656046 PMCID: PMC7909944 DOI: 10.1590/acb360207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/23/2021] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The present study explored the influence of liraglutide on remote preconditioning-mediated cardioprotection in diabetes mellitus along with the role of nuclear factor erythroid 2-related factor 2 (Nrf2), hypoxia inducible factor (HIF-1α) and hydrogen sulfide (H2S). METHODS Streptozotocin was given to rats to induce diabetes mellitus and rats were kept for eight weeks. Four cycles of ischemia and reperfusion were given to hind limb to induce remote preconditioning. After 24 h, hearts were isolated and subjected to 30 min of ischemia and 120 min of reperfusion on Langendorff system. Liraglutide was administered along with remote preconditioning. Cardiac injury was assessed by measuring the release of creatine kinase (CK-MB), cardiac troponin (cTnT) and development of left ventricular developed pressure. After ischemia-reperfusion, hearts were homogenized to measure the nuclear cytoplasmic ratio of Nrf2, H2S and HIF-1α levels. RESULTS In diabetic rats, there was more pronounced injury and the cardioprotective effects of remote preconditioning were not observed. Administration of liraglutide restored the cardioprotective effects of remote preconditioning in a dose-dependent manner. Moreover, liraglutide increased the Nrf2, H2S and HIF-1α levels in remote preconditioning-subjected diabetic rats. CONCLUSIONS Liraglutide restores the lost cardioprotective effects of remote preconditioning in diabetes by increasing the expression of Nrf2, H2S and HIF-1α.
Collapse
Affiliation(s)
| | - Yinyan Tang
- The Forth People’s Hospital of Yongzhou, China
| | - Huimin He
- The Forth People’s Hospital of Yongzhou, China
| | | |
Collapse
|
7
|
Saikia M, Holter MM, Donahue LR, Lee IS, Zheng QC, Wise JL, Todero JE, Phuong DJ, Garibay D, Coch R, Sloop KW, Garcia-Ocana A, Danko CG, Cummings BP. GLP-1 receptor signaling increases PCSK1 and β cell features in human α cells. JCI Insight 2021; 6:141851. [PMID: 33554958 PMCID: PMC7934853 DOI: 10.1172/jci.insight.141851] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is an incretin hormone that potentiates glucose-stimulated insulin secretion. GLP-1 is classically produced by gut L cells; however, under certain circumstances α cells can express the prohormone convertase required for proglucagon processing to GLP-1, prohormone convertase 1/3 (PC1/3), and can produce GLP-1. However, the mechanisms through which this occurs are poorly defined. Understanding the mechanisms by which α cell PC1/3 expression can be activated may reveal new targets for diabetes treatment. Here, we demonstrate that the GLP-1 receptor (GLP-1R) agonist, liraglutide, increased α cell GLP-1 expression in a β cell GLP-1R-dependent manner. We demonstrate that this effect of liraglutide was translationally relevant in human islets through application of a new scRNA-seq technology, DART-Seq. We found that the effect of liraglutide to increase α cell PC1/3 mRNA expression occurred in a subcluster of α cells and was associated with increased expression of other β cell-like genes, which we confirmed by IHC. Finally, we found that the effect of liraglutide to increase bihormonal insulin+ glucagon+ cells was mediated by the β cell GLP-1R in mice. Together, our data validate a high-sensitivity method for scRNA-seq in human islets and identify a potentially novel GLP-1-mediated pathway regulating human α cell function.
Collapse
Affiliation(s)
- Mridusmita Saikia
- Department of Biomedical Sciences and
- Baker Institute for Animal Health, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | | | | | | | | | | | | | | | | | - Reilly Coch
- Cayuga Medical Center, Ithaca, New York, USA
| | - Kyle W Sloop
- Diabetes and Complications, Lilly Research Laboratories, Lilly, Indianapolis, Indiana, USA
| | | | - Charles G Danko
- Department of Biomedical Sciences and
- Baker Institute for Animal Health, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | | |
Collapse
|
8
|
Keshtkar S, Kaviani M, Jabbarpour Z, Sabet Sarvestani F, Ghahremani MH, Esfandiari E, Hossein Aghdaei M, Nikeghbalian S, Shamsaeefar A, Geramizadeh B, Azarpira N. Hypoxia-Preconditioned Wharton's Jelly-Derived Mesenchymal Stem Cells Mitigate Stress-Induced Apoptosis and Ameliorate Human Islet Survival and Function in Direct Contact Coculture System. Stem Cells Int 2020; 2020:8857457. [PMID: 33381188 PMCID: PMC7759420 DOI: 10.1155/2020/8857457] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/08/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
Protection of isolated pancreatic islets against hypoxic and oxidative damage-induced apoptosis is essential during a pretransplantation culture period. A beneficial approach to maintain viable and functional islets is the coculture period with mesenchymal stem cells (MSCs). Hypoxia preconditioning of MSCs (Hpc-MSCs) for a short time stimulates the expression and secretion of antiapoptotic, antioxidant, and prosurvival factors. The aim of the present study was to evaluate the survival and function of human islets cocultured with Hpc-MSCs. Wharton's jelly-derived MSCs were subjected to hypoxia (5% O2: Hpc) or normoxia (20% O2: Nc) for 24 hours and then cocultured with isolated human islets in direct and indirect systems. Assays of viability and apoptosis, along with the production of reactive oxygen species (ROS), hypoxia-inducible factor 1-alpha (HIF-1α), apoptotic pathway markers, and vascular endothelial growth factor (VEGF) in the islets, were performed. Insulin and C-peptide secretions as islet function were also evaluated. Hpc-MSCs and Nc-MSCs significantly reduced the ROS production and HIF-1α protein aggregation, as well as downregulation of proapoptotic proteins and upregulation of antiapoptotic marker along with increment of VEGF secretion in the cocultured islet. However, the Hpc-MSCs groups were better than Nc-MSCs cocultured islets. Hpc-MSCs in both direct and indirect coculture systems improved the islet survival, while promotion of function was only significant in the direct cocultured cells. Hpc potentiated the cytoprotective and insulinotropic effects of MSCs on human islets through reducing stressful markers, inhibiting apoptosis pathway, enhancing prosurvival factors, and promoting insulin secretion, especially in direct coculture system, suggesting the effective strategy to ameliorate the islet quality for better transplantation outcomes.
Collapse
Affiliation(s)
- Somayeh Keshtkar
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Jabbarpour
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Hossein Ghahremani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elaheh Esfandiari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Saman Nikeghbalian
- Shiraz Organ Transplant Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Shamsaeefar
- Shiraz Organ Transplant Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bita Geramizadeh
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Keshtkar S, Kaviani M, Sarvestani FS, Ghahremani MH, Aghdaei MH, Al-Abdullah IH, Azarpira N. Exosomes derived from human mesenchymal stem cells preserve mouse islet survival and insulin secretion function. EXCLI JOURNAL 2020; 19:1064-1080. [PMID: 33013264 PMCID: PMC7527509 DOI: 10.17179/excli2020-2451] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022]
Abstract
Islet cell death and loss of function after isolation and before transplantation is considered a key barrier to successful islet transplantation outcomes. Mesenchymal stem cells (MSCs) have been used to protect isolated islets owing to their paracrine potential partially through the secretion of vascular endothelial growth factor (VEGF). The paracrine functions of MSCs are also mediated, at least in part, by the release of extracellular vesicles including exosomes. In the present study, we examined (i) the effect of exosomes from human MSCs on the survival and function of isolated mouse islets and (ii) whether exosomes contain VEGF and the potential impact of exosomal VEGF on the survival of mouse islets. Isolated mouse islets were cultured for three days with MSC-derived exosomes (MSC-Exo), MSCs, or MSC-conditioned media without exosomes (MSC-CM-without-Exo). We investigated the effects of the exosomes, MSCs, and conditioned media on islet viability, apoptosis and function. Besides the expression of apoptotic and pro-survival genes, the production of human and mouse VEGF proteins was evaluated. The MSCs and MSC-Exo, but not the MSC-CM-without-Exo, significantly decreased the percentage of apoptotic cells and increased islet viability following the downregulation of pro-apoptotic genes and the upregulation of pro-survival factors, as well as the promotion of insulin secretion. Human VEGF was observed in the isolated exosomes, and the gene expression and protein production of mouse VEGF significantly increased in islets cultured with MSC-Exo. MSC-derived exosomes are as efficient as parent MSCs for mitigating cell death and improving islet survival and function. This cytoprotective effect was probably mediated by VEGF transfer, suggesting a pivotal strategy for ameliorating islet transplantation outcomes.
Collapse
Affiliation(s)
- Somayeh Keshtkar
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Hossein Ghahremani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ismail H Al-Abdullah
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA/USA
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
10
|
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: 0.2] [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
|
11
|
Menger MM, Nalbach L, Wrublewsky S, Glanemann M, Gu Y, Laschke MW, Menger MD, Ampofo E. Darbepoetin-α increases the blood volume flow in transplanted pancreatic islets in mice. Acta Diabetol 2020; 57:1009-1018. [PMID: 32221724 PMCID: PMC8318962 DOI: 10.1007/s00592-020-01512-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/04/2020] [Indexed: 11/30/2022]
Abstract
AIMS The minimal-invasive transplantation of pancreatic islets is a promising approach to treat diabetes mellitus type 1. However, islet transplantation is still hampered by the insufficient process of graft revascularization, leading to a poor clinical outcome. Accordingly, the identification of novel compounds, which accelerate and improve the revascularization of transplanted islets, is of great clinical interest. Previous studies have shown that darbepoetin (DPO)-α, a long lasting analogue of erythropoietin, is capable of promoting angiogenesis. Hence, we investigated in this study whether DPO improves the revascularization of transplanted islets. METHODS Islets were isolated from green fluorescent protein-positive FVB/N donor mice and transplanted into dorsal skinfold chambers of FVB/N wild-type animals, which were treated with DPO low dose (2.5 µg/kg), DPO high dose (10 µg/kg) or vehicle (control). The revascularization was assessed by repetitive intravital fluorescence microscopy over an observation period of 14 days. Subsequently, the cellular composition of the grafts was analyzed by immunohistochemistry. RESULTS The present study shows that neither low- nor high-dose DPO treatment accelerates the revascularization of free pancreatic islet grafts. However, high-dose DPO treatment increased the blood volume flow of the transplanted islet. CONCLUSIONS These findings demonstrated that DPO treatment does not affect the revascularization of transplanted islets. However, the glycoprotein increases the blood volume flow of the grafts, which results in an improved microvascular function and may facilitate successful transplantation.
Collapse
Affiliation(s)
- Maximilian M Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Lisa Nalbach
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Selina Wrublewsky
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Matthias Glanemann
- Department for General, Visceral, Vascular and Pediatric Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Yuan Gu
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany.
| |
Collapse
|
12
|
Abdelrazik Soliman NG, Abdel-Hamid AA, El-Hawwary AA, Ellakkany A. Impact of liraglutide on microcirculation in experimental diabetic cardiomyopathy. Acta Histochem 2020; 122:151533. [PMID: 32197755 DOI: 10.1016/j.acthis.2020.151533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 03/01/2020] [Accepted: 03/02/2020] [Indexed: 12/23/2022]
Abstract
Liraglutide is a new therapy used in diabetes and its effect on diabetic complications particularly cardiovascular ones is still under investigated. In our research, we tried to study the effect of liraglutide on experimental diabetic cardiomyopathy (DCM) induced by streptozotocin. We found that liraglutide nearly preserved normal myocardiac structure and significantly protected against myocardiac inflammation and fibrosis that was found in DCM group, p < 0.05. It also increased the density of coronary arteriolar vasculature markedly indicated by significant increase in α SMA (p < 0.05) compared to both DCM and non-diabetic (ND) groups. Moreover, liraglutide decreased TNFα and increased VEGF proteins expression (P < 0.05) compared to DCM group. Conclusion, liraglutide may have a very important role in protecting against experimentally induced diabetic cardiomyopathy by preventing the degenerative changes in the cardiomyocytes and the associated fibrosis, inflammation and decreased vasculature at structural and molecular levels.
Collapse
|
13
|
Abstract
Our understanding of the role of the vascular endothelium has evolved over the past 2 decades, with the recognition that it is a dynamically regulated organ and that it plays a nodal role in a variety of physiological and pathological processes. Endothelial cells (ECs) are not only a barrier between the circulation and peripheral tissues, but also actively regulate vascular tone, blood flow, and platelet function. Dysregulation of ECs contributes to pathological conditions such as vascular inflammation, atherosclerosis, hypertension, cardiomyopathy, retinopathy, neuropathy, and cancer. The close anatomic relationship between vascular endothelium and highly vascularized metabolic organs/tissues suggests that the crosstalk between ECs and these organs is vital for both vascular and metabolic homeostasis. Numerous reports support that hyperlipidemia, hyperglycemia, and other metabolic stresses result in endothelial dysfunction and vascular complications. However, how ECs may regulate metabolic homeostasis remains poorly understood. Emerging data suggest that the vascular endothelium plays an unexpected role in the regulation of metabolic homeostasis and that endothelial dysregulation directly contributes to the development of metabolic disorders. Here, we review recent studies about the pivotal role of ECs in glucose and lipid homeostasis. In particular, we introduce the concept that the endothelium adjusts its barrier function to control the transendothelial transport of fatty acids, lipoproteins, LPLs (lipoprotein lipases), glucose, and insulin. In addition, we summarize reports that ECs communicate with metabolic cells through EC-secreted factors and we discuss how endothelial dysregulation contributes directly to the development of obesity, insulin resistance, dyslipidemia, diabetes mellitus, cognitive defects, and fatty liver disease.
Collapse
Affiliation(s)
- Xinchun Pi
- From the Section of Athero & Lipo, Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P., L.X.)
| | - Liang Xie
- From the Section of Athero & Lipo, Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.P., L.X.)
| | - Cam Patterson
- University of Arkansas for Medical Sciences, Little Rock (C.P.)
| |
Collapse
|
14
|
Bowers DT, Song W, Wang LH, Ma M. Engineering the vasculature for islet transplantation. Acta Biomater 2019; 95:131-151. [PMID: 31128322 PMCID: PMC6824722 DOI: 10.1016/j.actbio.2019.05.051] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/13/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022]
Abstract
The microvasculature in the pancreatic islet is highly specialized for glucose sensing and insulin secretion. Although pancreatic islet transplantation is a potentially life-changing treatment for patients with insulin-dependent diabetes, a lack of blood perfusion reduces viability and function of newly transplanted tissues. Functional vasculature around an implant is not only necessary for the supply of oxygen and nutrients but also required for rapid insulin release kinetics and removal of metabolic waste. Inadequate vascularization is particularly a challenge in islet encapsulation. Selectively permeable membranes increase the barrier to diffusion and often elicit a foreign body reaction including a fibrotic capsule that is not well vascularized. Therefore, approaches that aid in the rapid formation of a mature and robust vasculature in close proximity to the transplanted cells are crucial for successful islet transplantation or other cellular therapies. In this paper, we review various strategies to engineer vasculature for islet transplantation. We consider properties of materials (both synthetic and naturally derived), prevascularization, local release of proangiogenic factors, and co-transplantation of vascular cells that have all been harnessed to increase vasculature. We then discuss the various other challenges in engineering mature, long-term functional and clinically viable vasculature as well as some emerging technologies developed to address them. The benefits of physiological glucose control for patients and the healthcare system demand vigorous pursuit of solutions to cell transplant challenges. STATEMENT OF SIGNIFICANCE: Insulin-dependent diabetes affects more than 1.25 million people in the United States alone. Pancreatic islets secrete insulin and other endocrine hormones that control glucose to normal levels. During preparation for transplantation, the specialized islet blood vessel supply is lost. Furthermore, in the case of cell encapsulation, cells are protected within a device, further limiting delivery of nutrients and absorption of hormones. To overcome these issues, this review considers methods to rapidly vascularize sites and implants through material properties, pre-vascularization, delivery of growth factors, or co-transplantation of vessel supporting cells. Other challenges and emerging technologies are also discussed. Proper vascular growth is a significant component of successful islet transplantation, a treatment that can provide life-changing benefits to patients.
Collapse
Affiliation(s)
- Daniel T Bowers
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Wei Song
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Long-Hai Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
15
|
Keshtkar S, Kaviani M, Jabbarpour Z, Geramizadeh B, Motevaseli E, Nikeghbalian S, Shamsaeefar A, Motazedian N, Al-Abdullah IH, Ghahremani MH, Azarpira N. Protective effect of nobiletin on isolated human islets survival and function against hypoxia and oxidative stress-induced apoptosis. Sci Rep 2019; 9:11701. [PMID: 31406275 PMCID: PMC6690971 DOI: 10.1038/s41598-019-48262-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/31/2019] [Indexed: 12/29/2022] Open
Abstract
Islets transplantation, as a treatment of type 1 diabetes, faces challenges, including the loss of islets in the process of isolation and pre-transplantation due to cellular stresses-induced apoptosis. Accordingly, the optimization of culture plays a decisive role in the transplantation success. In this study, we evaluated the effect of nobiletin on the cultured human islets. Isolated human islets were treated by different concentrations of nobiletin and cultured for 24 and 72 hours. Then, the islets viability, apoptosis, insulin and C-peptide secretion, and apoptosis markers were evaluated. Also, the production of reactive oxygen species (ROS), hypoxia inducible factor 1 alpha (HIF-1α), and its target genes in the islets were examined. Our findings showed that the islets were encountered with hypoxia and oxidative stress after isolation and during culture. These insults induced apoptosis and reduced viability during culture period. Moreover, the secretion of insulin and C-peptide decreased. Nobiletin treatments significantly improved the islets survival through reduction of HIF-1α and ROS production and suppression of apoptosis, along with increased islets function. Islet protective effect of nobiletin might be related to its anti-oxidant, anti-apoptotic and insulinotropic properties. Hence, in order to achieve viable and functional islets for clinical transplantation, the application of nobiletin during pre-transplantation period is useful.
Collapse
Affiliation(s)
- Somayeh Keshtkar
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Jabbarpour
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bita Geramizadeh
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saman Nikeghbalian
- Shiraz Organ Transplant Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Shamsaeefar
- Shiraz Organ Transplant Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasrin Motazedian
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ismail H Al-Abdullah
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, USA
| | - Mohammad Hossein Ghahremani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pharmacology-Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Shiraz Institute of Stem Cell and Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
16
|
Pop R, Kong SH, Langlois A, Marchegiani F, Shlomovitz E, Legnèr A, Bietiger W, Pinget M, Beaujeux R, Mutter D, Marescaux J, Diana M. Gastrointestinal Hormones Manipulation to Counteract Metabolic Syndrome Using Duodenal Targeted Embolization. Surg Innov 2019; 26:280-292. [PMID: 30920898 DOI: 10.1177/1553350619838098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Targeted embolization of gastrointestinal (GI) arteries can modify hormonal production. We aimed to evaluate the impact of the embolization of the gastroduodenal artery (GDA) on the activity of foregut mucosa. METHODS The GDA's duodenal branch was embolized in 12 Yucatan pigs using 100-300 µm (group A; n = 4) or 300-500 µm (group B; n = 4) microspheres, followed by coiling of the branch. In 4 animals (sham), only saline was injected. The levels of GI hormones (ghrelin, glucose-dependent insulinotropic peptide [GIP], glucagon-like peptide-1 [GLP-1], insulin, peptide YY [PYY], leptin) and the gene expression of sodium-glucose-linked transporter-1 (SGLT-1) and glucose transporter-2 (GLUT-2) were assessed before (T0), 1 hour (T1), 1 month (T2), 3 months (T3), and 6 months (T4) after embolization. RESULTS In group A, a segmental duodenal stenosis occurred in all cases, which required balloon dilatation. There was a significant drop in the baseline glycemia in group A at T1 and T4 versus sham. Ghrelin was reduced in group A versus baseline and versus group B at T2 and T3 and versus sham at T1 and T3. Insulin was significantly lower in group A versus B at T1 and at T4 but not versus sham. SGLT-1 expression increased in B and sham at T4, while it remained stable in group A. GLUT-2 expression increased in sham at T4 but not in A or B. CONCLUSIONS GDA embolization induced a decrease in ghrelin production and influenced expression of glucose carriers in the foregut mucosa.
Collapse
Affiliation(s)
- Raoul Pop
- 1 IHU-Strasbourg, Institute of Image-Guided Surgery, Strasbourg, France
- 2 University Hospital of Strasbourg, Strasbourg, France
| | - Seong-Ho Kong
- 1 IHU-Strasbourg, Institute of Image-Guided Surgery, Strasbourg, France
- 3 Seoul National University Hospital, Seoul, South Korea
| | - Allan Langlois
- 4 European Center for the Study of Diabetes (CEED), Strasbourg, France
| | | | - Eran Shlomovitz
- 1 IHU-Strasbourg, Institute of Image-Guided Surgery, Strasbourg, France
| | - András Legnèr
- 1 IHU-Strasbourg, Institute of Image-Guided Surgery, Strasbourg, France
| | - William Bietiger
- 4 European Center for the Study of Diabetes (CEED), Strasbourg, France
| | - Michel Pinget
- 4 European Center for the Study of Diabetes (CEED), Strasbourg, France
| | - Rémy Beaujeux
- 1 IHU-Strasbourg, Institute of Image-Guided Surgery, Strasbourg, France
- 2 University Hospital of Strasbourg, Strasbourg, France
| | - Didier Mutter
- 1 IHU-Strasbourg, Institute of Image-Guided Surgery, Strasbourg, France
- 2 University Hospital of Strasbourg, Strasbourg, France
- 5 IRCAD Research Institute Against Cancer of the Digestive System, Strasbourg, France
| | - Jacques Marescaux
- 1 IHU-Strasbourg, Institute of Image-Guided Surgery, Strasbourg, France
- 5 IRCAD Research Institute Against Cancer of the Digestive System, Strasbourg, France
| | - Michele Diana
- 1 IHU-Strasbourg, Institute of Image-Guided Surgery, Strasbourg, France
- 2 University Hospital of Strasbourg, Strasbourg, France
- 5 IRCAD Research Institute Against Cancer of the Digestive System, Strasbourg, France
| |
Collapse
|
17
|
Sood A, Swislocki A. Nonglycemic Effects of GLP-1 Agonists: From a Starling to Lizards to People. Metab Syndr Relat Disord 2019; 17:303-313. [PMID: 31145029 DOI: 10.1089/met.2018.0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
With the approval of exenatide in 2005, physicians had a new class of hypoglycemic agents available for the treatment of type 2 diabetes-the glucagon-like peptide-1 receptor agonists (or GLP-1 receptor agonists). As of this writing, there are seven drugs in this class available in the United States. In addition to demonstrating either cardiovascular risk neutrality or overt benefit, as now mandated by the United States Food and Drug Administration (FDA), many of these drugs have other, unexpected actions. It is our goal to outline these actions, some beneficial, some not. We have reviewed English-language articles in this area, not for an exhaustive study, but rather a broad search to define current understanding and perhaps generate further investigation.
Collapse
Affiliation(s)
- Ajay Sood
- 1Medical Service, VA Northern California Health Care System, Martinez, California.,2Division of Endocrinology and Metabolism, Department of Internal Medicine, UC Davis School of Medicine, Sacramento, California
| | - Arthur Swislocki
- 1Medical Service, VA Northern California Health Care System, Martinez, California.,2Division of Endocrinology and Metabolism, Department of Internal Medicine, UC Davis School of Medicine, Sacramento, California
| |
Collapse
|
18
|
Chen Y, Zhang X, He J, Xie Y, Yang Y. Delayed Administration of the Glucagon-Like Peptide 1 Analog Liraglutide Promoting Angiogenesis after Focal Cerebral Ischemia in Mice. J Stroke Cerebrovasc Dis 2018; 27:1318-1325. [PMID: 29395648 DOI: 10.1016/j.jstrokecerebrovasdis.2017.12.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/30/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Glucagon-like peptide 1 (GLP-1) analogs administered before or after cerebral ischemia have been shown to provide neuroprotection. Here, we explored whether delayed administration of a GLP-1 analog, liraglutide, could improve long-term functional recovery and promote angiogenesis after stroke. MATERIALS AND METHODS In the present study, mice were established as a focal cerebral cortical ischemia model and were intraperitoneally administered liraglutide or normal saline (NS) daily for 14 consecutive days, starting 1 day after cerebral ischemia. The neurological deficits were evaluated using rotarod test. The microvessel density (MVD) and endothelial cell (EC) proliferation were assessed by immunohistochemical staining. The expression of vascular endothelial growth factor (VEGF) was assessed by Western blot analysis. RESULTS Liraglutide significantly reduced infarct volume and improved the rotarod test scores, compared with mice treated with NS. Liraglutide also greatly increased the MVD and EC proliferation and simultaneously upregulated the expression of VEGF in the cerebral ischemic area. CONCLUSIONS These results demonstrated that liraglutide promoted angiogenesis and long-term recovery of cerebral ischemia through increasing the expression of VEGF.
Collapse
Affiliation(s)
- Yanxia Chen
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiangjian Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei, China.
| | - Junna He
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yanzhao Xie
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yang Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
19
|
Lei Y, Hu L, Yang G, Piao L, Jin M, Cheng X. Dipeptidyl Peptidase-IV Inhibition for the Treatment of Cardiovascular Disease - Recent Insights Focusing on Angiogenesis and Neovascularization. Circ J 2017; 81:770-776. [PMID: 28344207 DOI: 10.1253/circj.cj-16-1326] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dipeptidyl peptidase IV (DPP-IV) is a complex enzyme that acts as a membrane-anchored cell surface exopeptidase and transmits intracellular signals through a small intracellular tail. DPP-IV exists in human blood in a soluble form, and truncates a large number of peptide hormones, chemokines, cytokines, and growth factors in vitro and in vivo. DPP-IV has gained considerable interest as a therapeutic target, and a variety of DPP-IV inhibitors that prolong the insulinotropic effects of glucagon-like peptide-1 (GLP-1) are widely used in clinical settings as antidiabetic drugs. Indeed, DPP-IV is upregulated in proinflammatory states, including obesity and cardiovascular disease with and without diabetes mellitus. Consistent with this maladaptive role, DPP-IV inhibitors seem to exert a protective role in cardiovascular disease. In addition to their GLP-1-dependent vascular protective actions, DPP-IV inhibitors exhibit GLP-1-independent beneficial effects on angiogenesis/neovascularization via several signaling pathways (e.g., stromal cell-derived factor-1α/C-X-C chemokine receptor type-4, vascular endothelial growth factor-A/endothelial nitric oxide synthase, etc.). This review focuses on recent findings in this field, highlighting the role of DPP-IV in therapeutic angiogenesis/neovascularization in ischemic heart disease and peripheral artery disease.
Collapse
Affiliation(s)
- Yanna Lei
- Department of ICU, Yanbian University Hospital
| | - Lina Hu
- Department of Public Health, Guilin Medical College
| | - Guang Yang
- Department of Cardiology, Yanbian University Hospital
| | - Limei Piao
- Department of Cardiology, Yanbian University Hospital
| | - Minggen Jin
- Department of ICU, Yanbian University Hospital
| | - Xianwu Cheng
- Department of Cardiology, Yanbian University Hospital.,Institute of Innovation for Future Society, Nagoya University.,Department of Cardiovascular Internal Medicine, Kyung Hee University Hospital, Kyung Hee University
| |
Collapse
|
20
|
Ke J, Wei R, Yu F, Zhang J, Hong T. Liraglutide restores angiogenesis in palmitate-impaired human endothelial cells through PI3K/Akt-Foxo1-GTPCH1 pathway. Peptides 2016; 86:95-101. [PMID: 27777063 DOI: 10.1016/j.peptides.2016.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/21/2016] [Accepted: 10/21/2016] [Indexed: 12/20/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) and its analogues have a beneficial role in cardiovascular system. Here, we aimed to investigate whether liraglutide, a GLP-1 analogue, modulated angiogenesis impaired by palmitic acid (PA) in cultured human umbilical vein endothelial cells (HUVECs). Cells were incubated with liraglutide (3-100 nmol/L) in the presence of PA (0.5mmol/L), and endothelial tube formation was observed and quantified. The protein levels of signaling molecules were analyzed and the specific inhibitors were used to identify the signaling pathways through which liraglutide affected angiogenesis. Results showed that liraglutide ameliorated endothelial tube formation impaired by PA in HUVECs in a dose-dependent manner. Meanwhile, liraglutide increased the phosphorylation of Akt and forkhead box O1 (Foxo1), and upregulated the levels of guanosine 5'-triphosphate cyclohydrolase 1 (GTPCH1) and endothelial nitric oxide synthase (eNOS) in PA-impaired HUVECs. Notably, addition of the PI3K inhibitor LY294002, Foxo1 nuclear export inhibitor trifluoperazine dihydrochloride (TFP), GTPCH1 inhibitor 2,4-diamino-6-hydroxypyrimidine (DAHP) or NOS inhibitor N-nitro-l-arginine-methyl ester (L-NAME) eliminated the angiogenic effect of liraglutide. Moreover, either LY294002 or TFP abolished the liraglutide-induced upregulation of GTPCH1 and eNOS protein levels. In conclusion, liraglutide restores angiogenesis in PA-impaired HUVECs. The effect is mediated via upregulation of GTPCH1 and eNOS levels in a PI3K/Akt-Foxo1-dependent mechanism.
Collapse
Affiliation(s)
- Jing Ke
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Fei Yu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Jingjing Zhang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
21
|
Pujadas G, Drucker DJ. Vascular Biology of Glucagon Receptor Superfamily Peptides: Mechanistic and Clinical Relevance. Endocr Rev 2016; 37:554-583. [PMID: 27732058 DOI: 10.1210/er.2016-1078] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regulatory peptides produced in islet and gut endocrine cells, including glucagon, glucagon-like peptide-1 (GLP-1), GLP-2, and glucose-dependent insulinotropic polypeptide, exert actions with considerable metabolic importance and translational relevance. Although the clinical development of GLP-1 receptor agonists and dipeptidyl peptidase-4 inhibitors has fostered research into how these hormones act on the normal and diseased heart, less is known about the actions of these peptides on blood vessels. Here we review the effects of these peptide hormones on normal blood vessels and highlight their vascular actions in the setting of experimental and clinical vascular injury. The cellular localization and signal transduction properties of the receptors for glucagon, GLP-1, GLP-2, and glucose-dependent insulinotropic polypeptide are discussed, with emphasis on endothelial cells and vascular smooth muscle cells. The actions of these peptides on the control of blood flow, blood pressure, angiogenesis, atherosclerosis, and vascular inflammation are reviewed with a focus on elucidating direct and indirect mechanisms of action. How these peptides traverse the blood-brain barrier is highlighted, with relevance to the use of GLP-1 receptor agonists to treat obesity and neurodegenerative disorders. Wherever possible, we compare actions identified in cell lines and primary cell culture with data from preclinical studies and, when available, results of human investigation, including studies in subjects with diabetes, obesity, and cardiovascular disease. Throughout the review, we discuss pitfalls, limitations, and challenges of the existing literature and highlight areas of controversy and uncertainty. The increasing use of peptide-based therapies for the treatment of diabetes and obesity underscores the importance of understanding the vascular biology of peptide hormone action.
Collapse
Affiliation(s)
- Gemma Pujadas
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| |
Collapse
|
22
|
Cui JP, Zhang MJ, Liu BL. Research progress of mesenchymal stem cells combined with islet transplantation in treatment of type I diabetes mellitus. Shijie Huaren Xiaohua Zazhi 2016; 24:2213-2218. [DOI: 10.11569/wcjd.v24.i14.2213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The most significant feature of type I diabetes is β-cell loss, which results in a series of complications. While β-cell loss occurs, β-cells are ultimately damaged by macrophages and T cells in the presence of inflammatory mediators. Because of this characteristic, five kinds of antibodies are commonly used in clinical practice to diagnose and evaluate β-cell loss, including islet cell antibody, insulin antibody, GAD65, IA-2 and IA-2b. In addition to the HLA gene related factors, environmental factors, such as infection, diet and physiological and psychological factors, are suspected to be causes of this disease. At present, there are many treatments for type I diabetes, and the clinical goal is to control blood glucose, prevent further damage of βcells and control patients' own immune response. In 1992, the discovery of insulin, which converts the fatal diabetes into a chronic disease, to some extent, delayed the progression of microvascular complications; however, it is not able to delay the progression of the disease. β-cell transplantation is currently the only minimally invasive means for reasonable control of blood glucose control disease related complications. Although whole pancreas transplantation can achieve a promising effect to some extent, it is accompanied by high incidence and mortality, as well as lifelong mandatory immune suppression. Bone marrow mesenchymal stem cells transplantation, lipopolysaccharideon (LPS) bone marrow mesenchymal stem cell pretreatment and islet cell exendin-4 liquid preservation reduce warm ischemia time damage and provide new avenues for islet cell transplantation.
Collapse
|