1
|
Dai L, Wang Q. Targeting ferroptosis: opportunities and challenges of mesenchymal stem cell therapy for type 1 diabetes mellitus. Stem Cell Res Ther 2025; 16:47. [PMID: 39901210 PMCID: PMC11792594 DOI: 10.1186/s13287-025-04188-7] [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] [Received: 10/09/2024] [Accepted: 01/24/2025] [Indexed: 02/05/2025] Open
Abstract
Type 1 diabetes mellitus (T1DM) is characterized by progressive β-cell death, leading to β-cell loss and insufficient insulin secretion. Mesenchymal stem cells (MSCs) transplantation is currently one of the most promising methods for β-cell replacement therapy. However, recent studies have shown that ferroptosis is not only one of the key mechanisms of β-cell death, but also one of the reasons for extensive cell death within a short period of time after MSCs transplantation. Ferroptosis is a new type of regulated cell death (RCD) characterized by iron-dependent accumulation of lipid peroxides. Due to the weak antioxidant capacity of β-cells, they are susceptible to cytotoxic stimuli such as oxidative stress (OS), and are therefore susceptible to ferroptosis. Transplanted MSCs are also extremely susceptible to perturbations in their microenvironment, especially OS, which can weaken their antioxidant capacity and induce MSCs death through ferroptosis. In the pathophysiological process of T1DM, a large amount of reactive oxygen species (ROS) are produced, causing OS. Therefore, targeting ferroptosis may be a key way to protect β-cells and improve the therapeutic effect of MSCs transplantation. This review reviews the research related to ferroptosis of β-cells and MSCs, and summarizes the currently developed strategies that help inhibit cell ferroptosis. This study aims to help understand the ferroptosis mechanism of β-cell death and MSCs death after transplantation, emphasize the importance of targeting ferroptosis for protecting β-cells and improving the survival and function of transplanted MSCs, and provide a new research direction for stem cells transplantation therapy of T1DM in the future.
Collapse
Affiliation(s)
- Le Dai
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, 126 Xiantai Avenue, Changchun City, Jilin Province, China
| | - Qing Wang
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, 126 Xiantai Avenue, Changchun City, Jilin Province, China.
| |
Collapse
|
2
|
Qin T, Smink AM, de Vos P. Enhancing longevity of immunoisolated pancreatic islet grafts by modifying both the intracapsular and extracapsular environment. Acta Biomater 2023:S1742-7061(23)00362-8. [PMID: 37392934 DOI: 10.1016/j.actbio.2023.06.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/02/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic metabolic disease characterized by autoimmune destruction of pancreatic β cells. Transplantation of immunoisolated pancreatic islets might treat T1DM in the absence of chronic immunosuppression. Important advances have been made in the past decade as capsules can be produced that provoke minimal to no foreign body response after implantation. However, graft survival is still limited as islet dysfunction may occur due to chronic damage to islets during islet isolation, immune responses induced by inflammatory cells, and nutritional issues for encapsulated cells. This review summarizes the current challenges for promoting longevity of grafts. Possible strategies for improving islet graft longevity are also discussed, including supplementation of the intracapsular milieu with essential survival factors, promotion of vascularization and oxygenation near capsules, modulation of biomaterials, and co-transplantation of accessory cells. Current insight is that both the intracapsular as well as the extracapsular properties should be improved to achieve long-term survival of islet-tissue. Some of these approaches reproducibly induce normoglycemia for more than a year in rodents. Further development of the technology requires collective research efforts in material science, immunology, and endocrinology. STATEMENT OF SIGNIFICANCE: Islet immunoisolation allows for transplantation of insulin producing cells in absence of immunosuppression and might facilitate the use of xenogeneic cell sources or grafting of cells obtained from replenishable cell sources. However, a major challenge to date is to create a microenvironment that supports long-term graft survival. This review provides a comprehensive overview of the currently identified factors that have been demonstrated to be involved in either stimulating or reducing islet graft survival in immunoisolating devices and discussed current strategies to enhance the longevity of encapsulated islet grafts as treatment for type 1 diabetes. Although significant challenges remain, interdisciplinary collaboration across fields may overcome obstacles and facilitate the translation of encapsulated cell therapy from the laboratory to clinical application.
Collapse
Affiliation(s)
- Tian Qin
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, EA 11, 9713 GZ, Groningen, The Netherlands.
| | - Alexandra M Smink
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, EA 11, 9713 GZ, Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, EA 11, 9713 GZ, Groningen, The Netherlands
| |
Collapse
|
3
|
Brandhorst D, Brandhorst H, Acreman S, Johnson PRV. The ischaemic preconditioning paradox and its implications for islet isolation from heart-beating and non heart-beating donors. Sci Rep 2022; 12:19321. [PMID: 36369239 PMCID: PMC9652462 DOI: 10.1038/s41598-022-23862-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
Abstract
The impact of ischaemia can severely damage procured donor organs for transplantation. The pancreas, and pancreatic islets in particular, is one of the most sensitive tissues towards hypoxia. The present study was aimed to assess the effect of hypoxic preconditioning (HP) performed ex-vivo in islets isolated from heart-beating donor (HBD) and non heart-beating donor (NHBD) rats. After HP purified islets were cultured for 24 h in hypoxia followed by islet characterisation. Post-culture islet yields were significantly lower in sham-treated NHBD than in HBD. This difference was reduced when NHBD islets were preconditioned. Similar results were observed regarding viability, apoptosis and in vitro function. Reactive oxygen species generation after hypoxic culture was significantly enhanced in sham-treated NHBD than in HBD islets. Again, this difference could be diminished through HP. qRT-PCR revealed that HP decreases pro-apoptotic genes but increases HIF-1 and VEGF. However, the extent of reduction and augmentation was always substantially higher in preconditioned NHBD than in HBD islets. Our findings indicate a lower benefit of HBD islets from HP than NHBD islets. The ischaemic preconditioning paradox suggests that HP should be primarily applied to islets from marginal donors. This observation needs evaluation in human islets.
Collapse
Affiliation(s)
- Daniel Brandhorst
- Research Group for Islet Transplantation, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
| | - Heide Brandhorst
- Research Group for Islet Transplantation, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Samuel Acreman
- Research Group for Islet Transplantation, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Paul R V Johnson
- Research Group for Islet Transplantation, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| |
Collapse
|
4
|
Elumalai S, Karunakaran U, Moon JS, Won KC. Ferroptosis Signaling in Pancreatic β-Cells: Novel Insights & Therapeutic Targeting. Int J Mol Sci 2022; 23:13679. [PMID: 36430158 PMCID: PMC9690757 DOI: 10.3390/ijms232213679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022] Open
Abstract
Metabolic stress impairs pancreatic β-cell survival and function in diabetes. Although the pathophysiology of metabolic stress is complex, aberrant tissue damage and β-cell death are brought on by an imbalance in redox equilibrium due to insufficient levels of endogenous antioxidant expression in β-cells. The vulnerability of β-cells to oxidative damage caused by iron accumulation has been linked to contributory β-cell ferroptotic-like malfunction under diabetogenic settings. Here, we take into account recent findings on how iron metabolism contributes to the deregulation of the redox response in diabetic conditions as well as the ferroptotic-like malfunction in the pancreatic β-cells, which may offer insights for deciphering the pathomechanisms and formulating plans for the treatment or prevention of metabolic stress brought on by β-cell failure.
Collapse
Affiliation(s)
- Suma Elumalai
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
| | - Udayakumar Karunakaran
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
| | - Jun-Sung Moon
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
- Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu 42415, Korea
| | - Kyu-Chang Won
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
- Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu 42415, Korea
| |
Collapse
|
5
|
Encapsulation Strategies for Pancreatic Islet Transplantation without Immune Suppression. CURRENT STEM CELL REPORTS 2021. [DOI: 10.1007/s40778-021-00190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
6
|
Feng S, Mao S, Dou J, Li W, Li H, Lin JM. An open-space microfluidic chip with fluid walls for online detection of VEGF via rolling circle amplification. Chem Sci 2019; 10:8571-8576. [PMID: 31803431 PMCID: PMC6839512 DOI: 10.1039/c9sc02974e] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022] Open
Abstract
We report an open-space microfluidic chip with fluid walls, integrating functions of cell culture and online detection of secreted proteins controlled by the interfacial tension value.
Despite traditional poly-dimethyl siloxane (PDMS) microfluidic devices having great potential in various biological studies, they are limited by sophisticated fabrication processes and low utilization. An easily controlled microfluidic platform with high efficiency and low cost is desperately required. In this work, we present an open-space microfluidic chip with fluid walls, integrating cell culture and online semi-quantitative detection of vascular endothelial growth factor (VEGF) via rolling circle amplification (RCA) reaction. In comparison with conventional co-culture detecting platforms, this method features the prominent advantages of saving reagents and time, a simplified chip fabrication process, and avoiding additional assistance for online detection with the help of an interfacial tension valve. On such a multi-functional microfluidic chip, cells (human umbilical vein endothelial cells and malignant glioma cells) could maintain regular growth and cell viability. VEGF could be detected with excellent specificity and good linearity in the range of 10–250 pg mL–1. Meanwhile, VEGF secreted by malignant glioma cells was also detected online and obviously increased when cells were stimulated by deferoxamine (DFO) to mimic a hypoxic microenvironment. The designed biochip with fluid walls provides a new perspective for micro-total analysis and could be promisingly applied in future clinical diagnosis and drug analysis.
Collapse
Affiliation(s)
- Shuo Feng
- Department of Chemistry , Beijing Key Laboratory of Microanalytical Methods and Instrumentation , MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| | - Sifeng Mao
- Department of Chemistry , Beijing Key Laboratory of Microanalytical Methods and Instrumentation , MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| | - Jinxin Dou
- Department of Chemistry , Beijing Key Laboratory of Microanalytical Methods and Instrumentation , MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| | - Weiwei Li
- Department of Chemistry , Beijing Key Laboratory of Microanalytical Methods and Instrumentation , MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| | - Haifang Li
- Department of Chemistry , Beijing Key Laboratory of Microanalytical Methods and Instrumentation , MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| | - Jin-Ming Lin
- Department of Chemistry , Beijing Key Laboratory of Microanalytical Methods and Instrumentation , MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| |
Collapse
|
7
|
Danggui Sini Decoction Protected Islet Endothelial Cell Survival from Hypoxic Damage via PI3K/Akt/eNOS Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:5421023. [PMID: 30108656 PMCID: PMC6077529 DOI: 10.1155/2018/5421023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023]
Abstract
Danggui Sini decoction (DSD) is a traditional Chinese decoction, which is wildly applied and showed to be effective in ameliorating ischemia-related symptoms. However, the mechanisms of DSD action in ischemic damage remain to be fully clarified. Pancreatic islet endothelial cells are pivotal constituent of islet microvasculature, with high vulnerability to hypoxic injuries. Here, using MST1 cell, a pancreatic islet endothelial cell-line, as a model, we investigated the effects of DSD on hypoxia-stimulated endothelial cell lesions and its underlying mechanisms. We found that DSD-Containing Serum (DSD-CS), collected from DSD-treated rats, could efficiently protect MST1 survival and proliferation from Cobalt chloride (CoCl2) induced damage, including cell viability, proliferation, and tube formation. Furthermore, DSD-CS restored the activity of PI3K/Akt/eNOS signaling inhibited by CoCl2 in MST1 cells. The protective effect of DSD-CS could be blocked by the specific PI3K/Akt/eNOS inhibitor LY294002, suggesting that DSD-CS protection of MST1 cell survival from hypoxia was mediated by PI3K/Akt/eNOS pathway. In conclusion, DSD treatment protected MST1 survival from hypoxic injuries via PI3K/Akt/eNOS pathway, indicating its role in protecting microvascular endothelial cells.
Collapse
|
8
|
Bruni A, Bornstein S, Linkermann A, Shapiro AMJ. Regulated Cell Death Seen through the Lens of Islet Transplantation. Cell Transplant 2018; 27:890-901. [PMID: 29845882 PMCID: PMC6050903 DOI: 10.1177/0963689718766323] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Clinical islet transplantation effectively restores euglycemia and corrects glycosylated
hemoglobin in labile type 1 diabetes mellitus (T1DM). Despite marked improvements in islet
transplantation outcomes, acute islet cell death remains a substantial obstacle that
compromises long-term engraftment outcomes. Multiple organ donors are routinely required
to achieve insulin independence. Therapeutic agents that ameliorate cell death and/or
control injury-related inflammatory cascades offer potential to improve islet transplant
success. Apoptotic cell death has been identified as a major contributor to cellular
demise and therapeutic strategies that subvert initiation and consequences of apoptotic
cell death have shown promise in pre-clinical models. Indeed, in numerous pathologies and
diseases apoptosis has been the most extensively described form of regulated cell death.
However, recent identification of novel, alternative regulated cell death pathways in
other disease states and solid organ transplantation suggest that these additional
pathways may also have substantial relevance in islet transplantation. These regulated,
non-apoptotic cell death pathways exhibit distinct biochemical characteristics but have
yet to be fully characterized within islet transplantation. We review herein the various
regulated cell death pathways and highlight their relative potential contributions to
islet viability, engraftment failure and islet dysfunction.
Collapse
Affiliation(s)
- Antonio Bruni
- 1 Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,2 Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Stefan Bornstein
- 3 Division of Nephrology, Medical Clinic 3, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Andreas Linkermann
- 3 Division of Nephrology, Medical Clinic 3, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - A M James Shapiro
- 1 Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,2 Department of Surgery, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
9
|
Lee SH, Park HS, Yang Y, Lee EY, Kim JW, Khang G, Yoon KH. Improvement of islet function and survival by integration of perfluorodecalin into microcapsules in vivo and in vitro. J Tissue Eng Regen Med 2018; 12:e2110-e2122. [PMID: 29330944 DOI: 10.1002/term.2643] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/04/2017] [Accepted: 01/02/2018] [Indexed: 12/24/2022]
Abstract
Hypoxic injury of islets is a major obstacle for encapsulated islet transplantation into the peritoneal cavity. To improve oxygen delivery to encapsulated islets, we integrated 20% of the oxygen carrier material, perfluorodecalin (PFD), in alginate capsules mixed with islets (PFD-alginate). Integration of PFD clearly improved islet viability and decreased reactive oxygen species production compared to islets encapsulated with alginate only (alginate) and naked islets exposed to hypoxia in vitro. In PFD-alginate capsules, HIF-1α expression was minimal, and insulin expression was well maintained. Furthermore, the best islet function represented by glucose-stimulated insulin secretion was observed for the PFD-alginate capsules in hypoxic condition. For the in vivo study, the same number of naked islets and encapsulated islets (alginate and PFD-alginate) was transplanted into streptozotocin-induced diabetic mice. Nonfasting blood glucose levels and the area under the curve for glucose based on intraperitoneal glucose tolerance tests in the PFD-alginate group were lower than in the alginate group. The harvested islets stained positive for insulin in all groups, but the ratio of dead cell area was 4 times higher in the alginate group than in the PFD-alginate group. In conclusion, integration of PFD in alginate microcapsules improved islet function and survival by minimizing the hypoxic damage of islets after intraperitoneal transplantation.
Collapse
Affiliation(s)
- Sang-Ho Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Heon-Seok Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yeoree Yang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji-Won Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Gilson Khang
- Department of Polymer Nano Science and Technology, Department of BIN Fusion Technology and BK-21 Polymer BIN Fusion Research Team, Chonbuk National University, Jeonju, South Korea
| | - Kun-Ho Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
10
|
Harb G, Poh YC, Pagliuca F. Stem Cell-Derived Insulin-Producing β Cells to Treat Diabetes. CURRENT TRANSPLANTATION REPORTS 2017. [DOI: 10.1007/s40472-017-0161-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
11
|
Abstract
Transplantation of pancreatic islets encapsulated within immuno-protective microcapsules is a strategy that has the potential to overcome graft rejection without the need for toxic immunosuppressive medication. However, despite promising preclinical studies, clinical trials using encapsulated islets have lacked long-term efficacy, and although generally considered clinically safe, have not been encouraging overall. One of the major factors limiting the long-term function of encapsulated islets is the host's immunological reaction to the transplanted graft which is often manifested as pericapsular fibrotic overgrowth (PFO). PFO forms a barrier on the capsule surface that prevents the ingress of oxygen and nutrients leading to islet cell starvation, hypoxia and death. The mechanism of PFO formation is still not elucidated fully and studies using a pig model have tried to understand the host immune response to empty alginate microcapsules. In this review, the varied strategies to overcome or reduce PFO are discussed, including alginate purification, altering microcapsule geometry, modifying alginate chemical composition, co-encapsulation with immunomodulatory cells, administration of pharmacological agents, and alternative transplantation sites. Nanoencapsulation technologies, such as conformal and layer-by-layer coating technologies, as well as nanofiber, thin-film nanoporous devices, and silicone based NanoGland devices are also addressed. Finally, this review outlines recent progress in imaging technologies to track encapsulated cells, as well as promising perspectives concerning the production of insulin-producing cells from stem cells for encapsulation.
Collapse
Affiliation(s)
- Vijayaganapathy Vaithilingam
- Materials Science and Engineering, Commonwealth Scientific and Industrial Research Organization (CSIRO), North Ryde, New South Wales, Australia
| | - Sumeet Bal
- Materials Science and Engineering, Commonwealth Scientific and Industrial Research Organization (CSIRO), North Ryde, New South Wales, Australia
| | - Bernard E Tuch
- School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
12
|
Qu Z, Jiang C, Wu J, Ding Y. Lenalidomide induces apoptosis and inhibits angiogenesis via caspase-3 and VEGF in hepatocellular carcinoma cells. Mol Med Rep 2016; 14:4781-4786. [DOI: 10.3892/mmr.2016.5797] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 09/23/2016] [Indexed: 11/06/2022] Open
|
13
|
Esfahani M, Karimi F, Afshar S, Niknazar S, Sohrabi S, Najafi R. Prolyl hydroxylase inhibitors act as agents to enhance the efficiency of cell therapy. Expert Opin Biol Ther 2015; 15:1739-55. [PMID: 26325448 DOI: 10.1517/14712598.2015.1084281] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION In stem cell-based therapy as a subtype of regenerative medicine, stem cells can be used to replace or repair injured tissue and cells in order to treat disease. Stem cells have the ability to integrate into injured areas and produce new cells via processes of proliferation and differentiation. Several studies have demonstrated that hypoxia increases self-renewal, proliferation and post-homing differentiation of stem cells through the regulation of hypoxia-inducible factor-1 (HIF-1)-mediated gene expression. Thus, pharmacological interventions including prolyl hydroxylase (PHD) inhibitors are considered as promising solutions for stem cell-based therapy. PHD inhibitors stabilize the HIF-1 and activate its pathway through preventing proteasomal degradation of HIF-1. AREAS COVERED This review focuses on the role of hypoxia, HIF-1 and especially PHD inhibitors on cell therapy. PHD structure and function are discussed as well as their inhibitors. In addition, we have investigated several preclinical studies in which PHD inhibitors improved the efficiency of cell-based therapies. EXPERT OPINION The data reviewed here suggest that PHD inhibitors are effective operators in improving stem cell therapy. However, because of some limitations, these compounds should be properly examined before clinical application.
Collapse
Affiliation(s)
- Maryam Esfahani
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Fatemeh Karimi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Saeid Afshar
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Somayeh Niknazar
- b 2 Shahid Beheshti University of Medical Science, Hearing Disorders Research Center , Tehran, the Islamic Republic of Iran
| | - Sareh Sohrabi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Rezvan Najafi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| |
Collapse
|
14
|
Pancreatic Islet Survival and Engraftment Is Promoted by Culture on Functionalized Spider Silk Matrices. PLoS One 2015; 10:e0130169. [PMID: 26090859 PMCID: PMC4474965 DOI: 10.1371/journal.pone.0130169] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 05/18/2015] [Indexed: 01/08/2023] Open
Abstract
Transplantation of pancreatic islets is one approach for treatment of diabetes, however, hampered by the low availability of viable islets. Islet isolation leads to disruption of the environment surrounding the endocrine cells, which contributes to eventual cell death. The reestablishment of this environment is vital, why we herein investigated the possibility of using recombinant spider silk to support islets in vitro after isolation. The spider silk protein 4RepCT was formulated into three different formats; 2D-film, fiber mesh and 3D-foam, in order to provide a matrix that can give the islets physical support in vitro. Moreover, cell-binding motifs from laminin were incorporated into the silk protein in order to create matrices that mimic the natural cell environment. Pancreatic mouse islets were thoroughly analyzed for adherence, necrosis and function after in vitro maintenance on the silk matrices. To investigate their suitability for transplantation, we utilized an eye model which allows in vivo imaging of engraftment. Interestingly, islets that had been maintained on silk foam during in vitro culture showed improved revascularization. This coincided with the observation of preserved islet architecture with endothelial cells present after in vitro culture on silk foam. Selected matrices were further evaluated for long-term preservation of human islets. Matrices with the cell-binding motif RGD improved human islet maintenance (from 36% to 79%) with preserved islets architecture and function for over 3 months in vitro. The islets established cell-matrix contacts and formed vessel-like structures along the silk. Moreover, RGD matrices promoted formation of new, insulin-positive islet-like clusters that were connected to the original islets via endothelial cells. On silk matrices with islets from younger donors (<35 year), the amount of newly formed islet-like clusters found after 1 month in culture were almost double compared to the initial number of islets added.
Collapse
|
15
|
Park HS, Kim JW, Lee SH, Yang HK, Ham DS, Sun CL, Hong TH, Khang G, Park CG, Yoon KH. Antifibrotic effect of rapamycin containing polyethylene glycol-coated alginate microcapsule in islet xenotransplantation. J Tissue Eng Regen Med 2015; 11:1274-1284. [DOI: 10.1002/term.2029] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 02/05/2015] [Accepted: 03/20/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Heon-Seok Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St Mary's Hospital, College of Medicine; Catholic University of Korea; Seoul Republic of Korea
| | - Ji-Won Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St Mary's Hospital, College of Medicine; Catholic University of Korea; Seoul Republic of Korea
- Convergent Research Consortium for Immunologic Disease, Seoul St Mary's Hospital; Catholic University of Korea; Seoul Republic of Korea
| | - Seung-Hwan Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St Mary's Hospital, College of Medicine; Catholic University of Korea; Seoul Republic of Korea
| | - Hae Kyung Yang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St Mary's Hospital, College of Medicine; Catholic University of Korea; Seoul Republic of Korea
| | - Dong-Sik Ham
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St Mary's Hospital, College of Medicine; Catholic University of Korea; Seoul Republic of Korea
| | - Cheng-Lin Sun
- Department of Endocrinology and Metabolism; First Hospital of Jilin University; Changchun Jilin People's Republic of China
| | - Tae Ho Hong
- Department of Surgery, College of Medicine; Catholic University of Korea; Seoul Republic of Korea
| | - Gilson Khang
- Department of Polymer Nano Science and Technology, Department of BIN Fusion Technology and BK-21 Polymer BIN Fusion Research Team; Chonbuk National University, Dukjin; Jeonju Republic of Korea
| | - Chung-Gyu Park
- Department of Microbiology and Immunology, Translational Xenotransplantation Research Centre, Cancer Research Institute, Biomedical Research Institute, College of Medicine; Seoul National University; Republic of Korea
| | - Kun-Ho Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St Mary's Hospital, College of Medicine; Catholic University of Korea; Seoul Republic of Korea
- Convergent Research Consortium for Immunologic Disease, Seoul St Mary's Hospital; Catholic University of Korea; Seoul Republic of Korea
| |
Collapse
|
16
|
Jung IR, Choi SE, Jung JG, Lee SA, Han SJ, Kim HJ, Kim DJ, Lee KW, Kang Y. Involvement of iron depletion in palmitate-induced lipotoxicity of beta cells. Mol Cell Endocrinol 2015; 407:74-84. [PMID: 25779532 DOI: 10.1016/j.mce.2015.03.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/28/2015] [Accepted: 03/10/2015] [Indexed: 12/28/2022]
Abstract
High levels of plasma free fatty acid are thought to contribute to the loss of pancreatic beta-cells in type 2 diabetes. In particular, saturated fatty acid such as palmitate or stearate can induce apoptosis in cultured beta cells (lipotoxicity). Endoplasmic reticulum stress is a critical mediator of free fatty acid-induced lipotoxicity. Recently, disorders in mitochondrial respiratory metabolism have been linked to lipotoxicity. Since iron is a critical component of respiratory metabolism, this study is initiated to determine whether abnormal iron metabolism is involved in palmitate-induced beta cell death. Immunoblotting analysis showed that treatment of INS-1 beta cells with palmitate reduced the level of transferrin receptor 1 (TfR1), but increased the level of heavy chain ferritin (FTH). In addition, palmitate reduced intracellular labile iron pool. Whereas iron depletion through treatment with iron-chelators deferoxamine or deferasirox augmented palmitate-induced cell death, iron supplementation with ferric chloride, ferrous sulfate, or holo-transferrin significantly protected cells against palmitate-induced death. Furthermore, overexpression of TfR1 reduced palmitate-induced cell death, whereas knockdown of TfR1 augmented cell death. In particular, treatment with deferoxamine increased the level of endoplasmic reticulum (ER) stress markers phospho-PERK, phospho-eIF2α, CHOP and phospho-c-Jun N-terminal kinase. Treatment with chemical chaperone significantly protected cells against deferoxamine-induced apoptosis. Iron supplementation also protected cells against palmitate-induced primary islet death. These data suggest that iron depletion plays an important role in palmitate-induced beta cell death through inducing ER stress. Therefore, attempts to block iron depletion might be able to prevent beta cell loss in type 2 diabetes.
Collapse
Affiliation(s)
- Ik-Rak Jung
- Department of Physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea; Department of Biomedical Science, The Graduate School, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Sung-E Choi
- Department of Physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Jong-Gab Jung
- Department of Biomedical Science, The Graduate School, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea; Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Sang-A Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Kwan-Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea; Department of Biomedical Science, The Graduate School, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea.
| |
Collapse
|
17
|
Hansen JB, Moen IW, Mandrup-Poulsen T. Iron: the hard player in diabetes pathophysiology. Acta Physiol (Oxf) 2014; 210:717-32. [PMID: 24521359 DOI: 10.1111/apha.12256] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/09/2014] [Accepted: 02/03/2014] [Indexed: 12/14/2022]
Abstract
The interest in the role of ferrous iron in diabetes pathophysiology has been revived by recent evidence of iron as an important determinant of pancreatic islet inflammation and as a biomarker of diabetes risk and mortality. The iron metabolism in the β-cell is complex. Excess free iron is toxic, but at the same time, iron is required for normal β-cell function and thereby glucose homeostasis. In the pathogenesis of diabetes, iron generates reactive oxygen species (ROS) by participating in the Fenton chemistry, which can induce oxidative damage and apoptosis. The aim of this review is to present and discuss recent evidence, suggesting that iron is a key pathogenic factor in both type 1 and type 2 diabetes with a focus on inflammatory pathways. Pro-inflammatory cytokine-induced β-cell death is not fully understood, but may include iron-induced ROS formation resulting in dedifferentiation by activation of transcription factors, activation of the mitochondrial apoptotic machinery or of other cell death mechanisms. The pro-inflammatory cytokine IL-1β facilitates divalent metal transporter 1 (DMT1)-induced β-cell iron uptake and consequently ROS formation and apoptosis, and we propose that this mechanism provides the relay between inflammation and oxidative β-cell damage. Iron chelation may be a potential therapeutic approach to reduce disease severity and mortality among diabetes patients. However, the therapeutic effect and safety of iron reduction need to be tested in clinical trials before dietary interventions or the use of iron chelation therapy titrated to avoid anaemia.
Collapse
Affiliation(s)
- J. B. Hansen
- Section for Endocrinological Research; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Physiology; University of Toronto; Toronto ON Canada
| | - I. W. Moen
- Section for Endocrinological Research; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
| | - T. Mandrup-Poulsen
- Section for Endocrinological Research; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Molecular Medicine and Surgery; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
18
|
Nourmohammadzadeh M, Lo JF, Bochenek M, Mendoza-Elias JE, Wang Q, Li Z, Zeng L, Qi M, Eddington DT, Oberholzer J, Wang Y. Microfluidic array with integrated oxygenation control for real-time live-cell imaging: effect of hypoxia on physiology of microencapsulated pancreatic islets. Anal Chem 2013; 85:11240-9. [PMID: 24083835 DOI: 10.1021/ac401297v] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In this article, we present a novel microfluidic islet array based on a hydrodynamic trapping principle. The lab-on-a-chip studies with live-cell multiparametric imaging allow understanding of physiological and pathophysiological changes of microencapsulated islets under hypoxic conditions. Using this microfluidic array and imaging analysis techniques, we demonstrate that hypoxia impairs the function of microencapsulated islets at the single islet level, showing a heterogeneous pattern reflected in intracellular calcium signaling, mitochondrial energetic, and redox activity. Our approach demonstrates an improvement over conventional hypoxia chambers that is able to rapidly equilibrate to true hypoxia levels through the integration of dynamic oxygenation. This work demonstrates the feasibility of array-based cellular analysis and opens up new modality to conduct informative analysis and cell-based screening for microencapsulated pancreatic islets.
Collapse
Affiliation(s)
- Mohammad Nourmohammadzadeh
- Department of Surgery/Transplant, University of Illinois at Chicago , 840 South Wood Street, Room 502, Chicago, Illinois 60612
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Cantley J, Walters SN, Jung MH, Weinberg A, Cowley MJ, Whitworth PT, Kaplan W, Hawthorne WJ, O'connell PJ, Weir G, Grey ST. A Preexistent Hypoxic Gene Signature Predicts Impaired Islet Graft Function and Glucose Homeostasis. Cell Transplant 2013; 22:2147-59. [DOI: 10.3727/096368912x658728] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We examined whether hypoxic exposure prior to the event of transplantation would have a positive or negative effect upon later islet graft function. Mouse islets exposed to hypoxic culture were transplanted into syngeneic recipients. Islet graft function, β-cell physiology, as well as molecular changes were examined. Expression of hypoxia-response genes in human islets pre- and posttransplant was examined by microarray. Hypoxia-preexposed murine islet grafts provided poor glycemic control in their syngeneic recipients, marked by persistent hyperglycemia and pronounced glucose intolerance with failed first- and second-phase glucose-stimulated insulin secretion in vivo. Mechanistically, hypoxic preexposure stabilized HIF-1α with a concomitant increase in hypoxic-response genes including LDHA, and a molecular gene set, which would favor glycolysis and lactate production and impair glucose sensing. Indeed, static incubation studies showed that hypoxia-exposed islets exhibited dysregulated glucose responsiveness with elevated basal insulin secretion. Isolated human islets, prior to transplantation, express a characteristic hypoxia-response gene expression signature, including high levels of LDHA, which is maintained posttransplant. Hypoxic preexposure of an islet graft drives a HIF-dependent switch to glycolysis with subsequent poor glycemic control and loss of glucose-stimulated insulin secretion (GSIS). Early intervention to reverse or prevent these hypoxia-induced metabolic gene changes may improve clinical islet transplantation.
Collapse
Affiliation(s)
- James Cantley
- Diabetes and Obesity Research Program, Garvan Institute, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Stacey N. Walters
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
- Immunology Program, Garvan Institute, Darlinghurst, New South Wales, Australia
| | - Min-Ho Jung
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Anita Weinberg
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
- Immunology Program, Garvan Institute, Darlinghurst, New South Wales, Australia
| | - Mark J. Cowley
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
- Cancer Program, Garvan Institute, Darlinghurst, New South Wales, Australia
| | - P. Tess Whitworth
- Diabetes and Obesity Research Program, Garvan Institute, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Warren Kaplan
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
- Peter Wills Bioinformatics Centre, Garvan Institute, Darlinghurst, New South Wales, Australia
| | - Wayne J. Hawthorne
- The Centre for Transplant and Renal Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Philip J. O'connell
- The Centre for Transplant and Renal Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Gordon Weir
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Shane T. Grey
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
- Immunology Program, Garvan Institute, Darlinghurst, New South Wales, Australia
| |
Collapse
|
20
|
Vaithilingam V, Kollarikova G, Qi M, Larsson R, Lacik I, Formo K, Marchese E, Oberholzer J, Guillemin GJ, Tuch BE. Beneficial effects of coating alginate microcapsules with macromolecular heparin conjugates-in vitro and in vivo study. Tissue Eng Part A 2013; 20:324-34. [PMID: 23971677 DOI: 10.1089/ten.tea.2013.0254] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Pericapsular fibrotic overgrowth (PFO) is associated with poor survival of encapsulated pancreatic islets. Modification of the microcapsule membrane aimed at preventing PFO should improve graft survival. This study investigated the effect of macromolecular Corline Heparin Conjugate (CHC) binding on intrinsic properties of alginate microcapsules and assessed the anti-fibrotic potential of this strategy both in vitro and in vivo. CHC was bound to alginate microcapsules using a layer-by-layer approach incorporating avidin. CHC binding to alginate microcapsule was visualized by confocal microscopy. Effects of CHC binding on microcapsule size, strength, and permeability were assessed, and the anti-clotting activity of bound CHC was determined by coagulation assay. Effect of CHC binding on the viability of encapsulated human islets was assessed in vitro, and their ability to function was assessed both in vitro and in vivo in diabetic immunodeficient mice. The potential of bound CHC to reduce PFO was assessed in vivo in different rat transplantation models. Confocal microscopy demonstrated a uniform coating of CHC onto the surface of microcapsules. CHC binding affected neither size nor permeability but significantly increased the tensile strength of alginate microcapsules by ~1.3-fold. The bound CHC molecules were stable and retained their anti-clotting activity for 3 weeks in culture. CHC binding affected neither viability nor function of the encapsulated human islets in vitro. In vivo CHC binding did not compromise islet function, and diabetes was reversed in all recipients with mice exhibiting lower blood glucose levels similar to controls in oral glucose tolerance tests. CHC binding was beneficial and significantly reduced PFO in both syngeneic and allogeneic rat transplantation models by ~65% and ~43%, respectively. In conclusion, our results show a new method to successfully coat CHC on alginate microcapsules and demonstrate its beneficial effect in increasing capsule strength and reduce PFO. This strategy has the potential to improve graft survival of encapsulated human islets.
Collapse
Affiliation(s)
- Vijayaganapathy Vaithilingam
- 1 Department of Pharmacology, School of Medical Sciences, Faculty of Medicine, University of New South Wales , Randwick, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Park JB, Jeong JH, Lee M, Lee DY, Byun Y. Xenotransplantation of exendin-4 gene transduced pancreatic islets using multi-component (alginate, poly-L-lysine, and polyethylene glycol) microcapsules for the treatment of type 1 diabetes mellitus. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2013; 24:2045-2057. [PMID: 23905775 DOI: 10.1080/09205063.2013.823071] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This study proposed that microencapsulation of exendin-4 gene transduced islets using alginate, poly-L-lysine, and polyethylene glycol could lead to increased viability and functionality of islets in a rat to mouse xenograft model. The stability of the microcapsules was determined using an osmotic pressure test and a rotational stress test. Exendin-4 gene was transduced into pancreatic islets using lenti-viral vectors and the transduced islets were encapsulated using multi-component microcapsules mentioned above. Both viability and functionality of microencapsulated islets were evaluated in both in vitro and in vivo xenograft model. The viabilities of the unmodified islets (control) and the exendin-4 transduced islets (test) on 14th day were 18.6 ± 11.1 and 49.2 ± 13.4%, respectively (p < 0.05). The stimulation index of the control and the test groups was 2.3 ± 1.7 and 3.0 ± 1.6, respectively. The mean survival times (MST) of the control and the test groups were 20.2 ± 8.0 and 35.2 ± 10.0 days, respectively (p < 0.05). Significant differences in MST suggested that transduction of exendin-4 gene had a great potential to increase the function of encapsulated islets. In conclusion, exendin-4 gene transduced islets encapsulated by poly(ethylene glycol) conjugated alginate/PLL microcapsules significantly improved both viability and functionality of encapsulated islets.
Collapse
Affiliation(s)
- Jun-Beom Park
- a Graduate School of Convergence Science and Technology, Department of Molecular Medicine and Biopharmaceutical Sciences , Seoul National University , Seoul , 151742 , South Korea
| | | | | | | | | |
Collapse
|
22
|
Ma Z, Moruzzi N, Catrina SB, Hals I, Oberholzer J, Grill V, Björklund A. Preconditioning with associated blocking of Ca2+ inflow alleviates hypoxia-induced damage to pancreatic β-cells. PLoS One 2013; 8:e67498. [PMID: 23935835 PMCID: PMC3723782 DOI: 10.1371/journal.pone.0067498] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 05/20/2013] [Indexed: 01/06/2023] Open
Abstract
Objective Beta cells of pancreatic islets are susceptible to functional deficits and damage by hypoxia. Here we aimed to characterize such effects and to test for and pharmacological means to alleviate a negative impact of hypoxia. Methods and Design Rat and human pancreatic islets were subjected to 5.5 h of hypoxia after which functional and viability parameters were measured subsequent to the hypoxic period and/or following a 22 h re-oxygenation period. Preconditioning with diazoxide or other agents was usually done during a 22 h period prior to hypoxia. Results Insulin contents decreased by 23% after 5.5 h of hypoxia and by 61% after a re-oxygenation period. Preconditioning with diazoxide time-dependently alleviated these hypoxia effects in rat and human islets. Hypoxia reduced proinsulin biosynthesis (3H-leucine incorporation into proinsulin) by 35%. Preconditioning counteracted this decrease by 91%. Preconditioning reduced hypoxia-induced necrosis by 40%, attenuated lowering of proteins of mitochondrial complexes I–IV and enhanced stimulation of HIF-1-alpha and phosphorylated AMPK proteins. Preconditioning by diazoxide was abolished by co-exposure to tolbutamide or elevated potassium (i.e. conditions which increase Ca2+ inflow). Preconditioning with nifedipine, a calcium channel blocker, partly reproduced effects of diazoxide. Both diazoxide and nifedipine moderately reduced basal glucose oxidation whereas glucose-induced oxygen consumption (tested with diazoxide) was unaffected. Preconditioning with diaxoxide enhanced insulin contents in transplants of rat islets to non-diabetic rats and lowered hyperglycemia vs. non-preconditioned islets in streptozotocin-diabetic rats. Preconditioning of human islet transplants lowered hyperglycemia in streptozotocin-diabetic nude mice. Conclusions 1) Prior blocking of Ca2+ inflow associates with lesser hypoxia-induced damage, 2) preconditioning affects basal mitochondrial metabolism and accelerates activation of hypoxia-reactive and potentially protective factors, 3) results indicate that preconditioning by K+-ATP-channel openers has therapeutic potential for islet transplantations.
Collapse
Affiliation(s)
- Zuheng Ma
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Noah Moruzzi
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Hals
- Institute of Cancer Research and Molecular Medicine, The Medical Faculty, Norwegian University of Science and Technology, Trondheim, Norway
| | - José Oberholzer
- Department of Transplant/Surgery, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Valdemar Grill
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Institute of Cancer Research and Molecular Medicine, The Medical Faculty, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St. Olav University Hospital, 7006 Trondheim, Norway
| | - Anneli Björklund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
23
|
Robles L, Storrs R, Lamb M, Alexander M, Lakey JRT. Current status of islet encapsulation. Cell Transplant 2013; 23:1321-48. [PMID: 23880554 DOI: 10.3727/096368913x670949] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell encapsulation is a method of encasing cells in a semipermeable matrix that provides a permeable gradient for the passage of oxygen and nutrients, but effectively blocks immune-regulating cells from reaching the graft, preventing rejection. This concept has been described as early as the 1930s, but it has exhibited substantial achievements over the last decade. Several advances in encapsulation engineering, chemical purification, applications, and cell viability promise to make this a revolutionary technology. Several obstacles still need to be overcome before this process becomes a reality, including developing a reliable source of islets or insulin-producing cells, determining the ideal biomaterial to promote graft function, reducing the host response to the encapsulation device, and ultimately a streamlined, scaled-up process for industry to be able to efficiently and safely produce encapsulated cells for clinical use. This article provides a comprehensive review of cell encapsulation of islets for the treatment of type 1 diabetes, including a historical perspective, current research findings, and future studies.
Collapse
Affiliation(s)
- Lourdes Robles
- Department of Surgery, University of California Irvine, Irvine, CA, USA
| | | | | | | | | |
Collapse
|
24
|
Choi JS, Koh IU, Lee HJ, Kim WH, Song J. Effects of excess dietary iron and fat on glucose and lipid metabolism. J Nutr Biochem 2013; 24:1634-44. [PMID: 23643521 DOI: 10.1016/j.jnutbio.2013.02.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 01/09/2013] [Accepted: 02/06/2013] [Indexed: 12/11/2022]
Abstract
PURPOSE Diets rich in fat and energy are associated with metabolic syndrome (MS). Increased body iron stores have been recognized as a feature of MS. High-fat diets (HFs), excess iron loading and MS are closely associated, but the mechanism linking them has not been clearly defined. We investigated the interaction between dietary fat and dietary Fe in the context of glucose and lipid metabolism in the body. METHODS C57BL6/J mice were divided into four groups and fed the modified AIN-93G low-fat diet (LF) and HF with adequate or excess Fe for 7 weeks. The Fe contents were increased by adding carbonyl iron (2% of diet weight) (LF+Fe and HF+Fe). RESULTS High iron levels increased blood glucose levels but decreased high-density lipoprotein cholesterol levels. The HF group showed increases in plasma levels of glucose and insulin and insulin resistance. HF+Fe mice showed greater changes. Representative indices of iron status, such hepatic and plasma Fe levels, were not altered further by the HF. However, both the HF and excess iron loading changed the hepatic expression of hepcidin and ferroportin. The LF+Fe, HF and HF+Fe groups showed greater hepatic fat accumulation compared with the LF group. These changes were paralleled by alterations in the levels of enzymes related to hepatic gluconeogenesis and lipid synthesis, which could be due to increases in mitochondrial dysfunction and oxidative stress. CONCLUSIONS High-fat diets and iron overload are associated with insulin resistance, modified hepatic lipid and iron metabolism and increased mitochondrial dysfunction and oxidative stress.
Collapse
Affiliation(s)
- Joo Sun Choi
- Division of Metabolic Diseases, Center for Biomedical Sciences, Korea National Institute of Health, Cheongwon-gun, Chungbuk-do 363-951, South Korea
| | | | | | | | | |
Collapse
|
25
|
Najafi R, Sharifi AM. Deferoxamine preconditioning potentiates mesenchymal stem cell homing in vitro and in streptozotocin-diabetic rats. Expert Opin Biol Ther 2013; 13:959-72. [PMID: 23536977 DOI: 10.1517/14712598.2013.782390] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Today, cell therapy is considered a promising alternative in treatment of several diseases such as type 1 diabetes. Loss of transplanted stem cell and more importantly scarcity in the number of cells reaching to target tissue is a major obstacle in cell therapy. There is evidences showing that deferoxamine (DFO), an iron chelator, increases the mobilization and homing of progenitor cells through increasing the stability of hypoxia-inducible factor 1α (HIF-1α) protein. In this study, the effect of DFO on some factors involved in homing of bone marrow-derived mesenchymal stem cell was investigated, and the other objectives of this research were to determine whether DFO is able to increase migration and subsequent homing of mesenchymal stem cell (MSCs) both in vitro and in vivo in streptozotocin-diabetic rats. RESEARCH DESIGN AND METHODS MSCs were treated by DFO in minimal essential medium α (αMEM) for 24 h. The expression and localization of HIF-1α were evaluated by western blotting and immunocytochemistry. The expression of C-X-C chemokine receptor type 4 (CXCR-4) and chemokine receptor 2 (CCR2) were assessed by western blotting and RT-PCR. The activity of matrix metalloproteinases (MMP) -2 and -9 were measured by gelatin zymography. Finally, in vitro migration of MSCs toward different concentrations of stromal cell-derived factor and monocyte chemotactic protein-1 were also evaluated. To demonstrate the homing of MSCs in vivo, DFO-treated chloromethyl-benzamidodialkylcarbocyanine-labeled MSCs were injected into the tail vein of rats, and the number of stained MSCs reaching to the pancreas were determined after 24 h. RESULTS In DFO-treated MSCs, expression of HIF-1α (p < 0.001), CXCR4 (p < 0.001), CCR2 (p < 0.001), and the activity of MMP-2 (p < 0.01) and MMP-9 (p < 0.05) were significantly increased compared to control groups. Elevation of HIF-1α, upregulation of CXCR4/CCR2 and higher activity of MMP-2/MMP-9 in DFO-treated MSCs were reversed by 2-methoxyestradiol (2-ME; 5 μmol), a HIF-1α inhibitor. The in vitro migrations as well as in vivo homing of DFO-treated MSCs were also significantly higher than control groups (p < 0.05). CONCLUSIONS Preconditioning of MSCs by DFO prior to transplantation could increase homing of MSCs through affecting some chemokine receptors as well as proteases involved and eventually improving the efficacy of cell therapy.
Collapse
Affiliation(s)
- R Najafi
- Tehran University of Medical Sciences, School of Medicine, Razi Drug Research Center, Department of Pharmacology, Tehran, Iran
| | | |
Collapse
|
26
|
Hals IK, Rokstad AM, Strand BL, Oberholzer J, Grill V. Alginate microencapsulation of human islets does not increase susceptibility to acute hypoxia. J Diabetes Res 2013; 2013:374925. [PMID: 24364039 PMCID: PMC3864170 DOI: 10.1155/2013/374925] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/08/2013] [Indexed: 01/06/2023] Open
Abstract
Islet transplantation in diabetes is hampered by the need of life-long immunosuppression. Encapsulation provides partial immunoprotection but could possibly limit oxygen supply, a factor that may enhance hypoxia-induced beta cell death in the early posttransplantation period. Here we tested susceptibility of alginate microencapsulated human islets to experimental hypoxia (0.1-0.3% O2 for 8 h, followed by reoxygenation) on viability and functional parameters. Hypoxia reduced viability as measured by MTT by 33.8 ± 3.5% in encapsulated and 42.9 ± 5.2% in nonencapsulated islets (P < 0.2). Nonencapsulated islets released 37.7% (median) more HMGB1 compared to encapsulated islets after hypoxic culture conditions (P < 0.001). Glucose-induced insulin release was marginally affected by hypoxia. Basal oxygen consumption was equally reduced in encapsulated and nonencapsulated islets, by 22.0 ± 6.1% versus 24.8 ± 5.7%. Among 27 tested cytokines/chemokines, hypoxia increased the secretion of IL-6 and IL-8/CXCL8 in both groups of islets, whereas an increase of MCP-1/CCL2 was seen only with nonencapsulated islets. Conclusion. Alginate microencapsulation of human islets does not increase susceptibility to acute hypoxia. This is a positive finding in relation to potential use of encapsulation for islet transplantation.
Collapse
Affiliation(s)
- I. K. Hals
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Postbox 8905, 7491 Trondheim, Norway
- *I. K. Hals:
| | - A. M. Rokstad
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Postbox 8905, 7491 Trondheim, Norway
| | - B. L. Strand
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Postbox 8905, 7491 Trondheim, Norway
- Department of Biotechnology, Faculty of Natural Sciences and Technology, Norwegian University of Science and Technology, 7034 Trondheim, Norway
| | - J. Oberholzer
- Department of Surgery, University of Illinois, IL at Chicago, Chicago, IL 60612, USA
| | - V. Grill
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Postbox 8905, 7491 Trondheim, Norway
- Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, Postbox 3250, 7006 Trondheim, Norway
| |
Collapse
|
27
|
Hansen JB, Tonnesen MF, Madsen AN, Hagedorn PH, Friberg J, Grunnet LG, Heller RS, Nielsen AØ, Størling J, Baeyens L, Anker-Kitai L, Qvortrup K, Bouwens L, Efrat S, Aalund M, Andrews NC, Billestrup N, Karlsen AE, Holst B, Pociot F, Mandrup-Poulsen T. Divalent metal transporter 1 regulates iron-mediated ROS and pancreatic β cell fate in response to cytokines. Cell Metab 2012; 16:449-61. [PMID: 23000401 DOI: 10.1016/j.cmet.2012.09.001] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 06/27/2012] [Accepted: 08/27/2012] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS) contribute to target-cell damage in inflammatory and iron-overload diseases. Little is known about iron transport regulation during inflammatory attack. Through a combination of in vitro and in vivo studies, we show that the proinflammatory cytokine IL-1β induces divalent metal transporter 1 (DMT1) expression correlating with increased β cell iron content and ROS production. Iron chelation and siRNA and genetic knockdown of DMT1 expression reduce cytokine-induced ROS formation and cell death. Glucose-stimulated insulin secretion in the absence of cytokines in Dmt1 knockout islets is defective, highlighting a physiological role of iron and ROS in the regulation of insulin secretion. Dmt1 knockout mice are protected against multiple low-dose streptozotocin and high-fat diet-induced glucose intolerance, models of type 1 and type 2 diabetes, respectively. Thus, β cells become prone to ROS-mediated inflammatory damage via aberrant cellular iron metabolism, a finding with potential general cellular implications.
Collapse
Affiliation(s)
- Jakob Bondo Hansen
- Center for Medical Research Methodology, Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Human apo-lactoferrin as a physiological mimetic of hypoxia stabilizes hypoxia-inducible factor-1 alpha. Biometals 2012; 25:1247-59. [DOI: 10.1007/s10534-012-9586-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 09/06/2012] [Indexed: 01/02/2023]
|
29
|
Jia X, Sharma A, Kumagai-Braesch M, Wernerson AM, Sörenby AK, Yamamoto S, Wang F, Tibell AB. Exendin-4 increases the expression of hypoxia-inducible factor-1α in rat islets and preserves the endocrine cell volume of both free and macroencapsulated islet grafts. Cell Transplant 2012; 21:1269-83. [PMID: 22405036 DOI: 10.3727/096368911x627408] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
In this study, we evaluated the effects of exendin-4 on free and encapsulated islet grafts in a rodent model. We also investigated the role of a transcription factor, hypoxia-inducible factor-1 (HIF-1), in mediating the beneficial effects of exendin-4. Diabetic athymic mice were transplanted with free rat islets under the kidney capsule or with macroencapsulated rat islets SC with or without exendin-4, islet preculture (exendin-4 0.1 nM for 20 h), and/or recipient treatment (IP 100 ng/day, day 0-7). The mice were followed for 4 weeks and the graft function and β-cell volume were evaluated. The effects of exendin-4 on islet HIF-1α mRNA and protein expression and on ATP content in a rat insulinoma cell line (INS-1E) were also examined. Preculture with exendin-4 followed by recipient treatment improved the outcome of both free (73% graft function vs. 26% in controls, p = 0.03) and macroencapsulated islet grafts (100% vs. 25% in controls, p = 0.02). In macroencapsulated grafts, the exendin-4-treated group had significantly larger endocrine volume, less graft necrosis, and more blood vessels around the capsule. In rat islets cultured with exendin-4, HIF-1α mRNA and protein expression were significantly enhanced. ATP content was increased in exendin-4-treated INS-1E cells under hypoxic conditions. The improved functional outcome after transplantation of a marginal islet mass with a brief initial treatment with exendin-4 is related to a larger surviving endocrine cell volume. Exendin-4 may improve islet graft resistance to hypoxia during the peritransplant period by increasing the expression of HIF-1α.
Collapse
Affiliation(s)
- Xiaohui Jia
- CLINTEC, Division of Transplantation Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Vaithilingam V, Kollarikova G, Qi M, Lacik I, Oberholzer J, Guillemin GJ, Tuch BE. Effect of prolonged gelling time on the intrinsic properties of barium alginate microcapsules and its biocompatibility. J Microencapsul 2011; 28:499-507. [PMID: 21827357 DOI: 10.3109/02652048.2011.586067] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Pericapsular fibrotic overgrowth (PFO) may be attributed to an immune response against microcapsules themselves or to antigen shedding through microcapsule pores from encapsulated islet tissue. Modification of microcapsules aimed at reducing pore size should prevent PFO and improve graft survival. This study investigated the effect of increased gelling time (20 vs. 2 min) in barium chloride on intrinsic properties of alginate microcapsules and tested their biocompatibility in vivo. Prolonged gelling time affected neither permeability nor size of the microcapsules. However, prolonged gelling time for 20 min produced brittle microcapsules compared to 2 min during compression test. Encapsulation of human islets in both types of microcapsules affected neither islet viability nor function. The presence of PFO when transplanted into a large animal model such as baboon and its absence in small animal models such as rodents suggest that the host immune response towards alginate microcapsules is species rather than alginate specific.
Collapse
|
31
|
Tuch BE, Hughes TC, Evans MDM. Encapsulated pancreatic progenitors derived from human embryonic stem cells as a therapy for insulin-dependent diabetes. Diabetes Metab Res Rev 2011; 27:928-32. [PMID: 22069287 DOI: 10.1002/dmrr.1274] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Cellular-based therapies for insulin-dependent diabetes are potential means of achieving and maintaining normal blood glucose levels (BGL) without the need for insulin administration. Islets isolated from donor pancreases have been the most common tissue used to date, but supply is a limiting factor. The use of human embryonic stem cells (hESC) as a therapy became a possibility with the report that these cells could be differentiated to pancreatic progenitors (PP) over 12 days in vitro. Conversion of PP to glucose-responsive insulin-secreting cells can be achieved by transplanting the progenitors in vivo where cell maturation occurs. To date this step has not been shown under in vitro conditions. METHODS Prior to transplanting, cells are encapsulated in alginate to prevent the immune cells of recipient attacking the graft. The alginate capsules have pores with a molecular weight cut-off of 250 kDa. These are too small to allow entry of immune cells, but large enough for passage of nutrients and insulin. RESULTS Encapsulated insulin-producing cells survive and function when transplanted, and have been shown to normalize BGL when allografted into diabetic mice. As few as 750 encapsulated human islets are sufficient to normalize BGL of diabetic non-obese diabetic severe combined immunodeficient (NOD/SCID) recipient mice for at least 2 months. The safety of transplanting encapsulated human islets as demonstrated by the lack of major adverse events and infection was recently shown in a first-in-human clinical trial. Finally, fetal porcine islet-like cell clusters, which are akin to PP derived from ESC, mature and normalize BGL of diabetic recipient mice with the same efficiency as non-encapsulated clusters placed under the kidney capsule. CONCLUSION Transplanting encapsulated PP, derived from hESCs, into diabetic recipients is the strategy that is now being explored in the Australia Diabetes Therapy Project.
Collapse
Affiliation(s)
- Bernard E Tuch
- Australia Diabetes Therapy Project, Biomaterials, CSIRO Materials Science and Engineering Research Laboratories, Sydney and Melbourne, Australia.
| | | | | |
Collapse
|
32
|
Jahansouz C, Jahansouz C, Kumer SC, Brayman KL. Evolution of β-Cell Replacement Therapy in Diabetes Mellitus: Islet Cell Transplantation. J Transplant 2011; 2011:247959. [PMID: 22013505 PMCID: PMC3195999 DOI: 10.1155/2011/247959] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Accepted: 08/08/2011] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus remains one of the leading causes of morbidity and mortality worldwide. According to the Centers for Disease Control and Prevention, approximately 23.6 million people in the United States are affected. Of these individuals, 5 to 10% have been diagnosed with Type 1 diabetes mellitus (T1DM), an autoimmune disease. Although it often appears in childhood, T1DM may manifest at any age, leading to significant morbidity and decreased quality of life. Since the 1960s, the surgical treatment for diabetes mellitus has evolved to become a viable alternative to insulin administration, beginning with pancreatic transplantation. While islet cell transplantation has emerged as another potential alternative, its role in the treatment of T1DM remains to be solidified as research continues to establish it as a truly viable alternative for achieving insulin independence. In this paper, the historical evolution, procurement, current status, benefits, risks, and ongoing research of islet cell transplantation are explored.
Collapse
Affiliation(s)
- Cyrus Jahansouz
- School of Medicine, University of Virginia, Charlottesville, VA 22102, USA
| | | | | | | |
Collapse
|
33
|
Langlois A, Bietiger W, Seyfritz E, Maillard E, Vivot K, Peronet C, Meyer N, Kessler L, Jeandidier N, Pinget M, Sigrist S. Improvement of Rat Islet Viability during Transplantation: Validation of Pharmacological Approach to Induce VEGF Overexpression. Cell Transplant 2011; 20:1333-42. [DOI: 10.3727/096368910x557182] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Delayed and insufficient revascularization during islet transplantation deprives islets of oxygen and nutrients, resulting in graft failure. Vascular endothelial growth factor (VEGF) could play a critical role in islet revascularization. We aimed to develop pharmacological strategies for VEGF overexpression in pancreatic islets using the iron chelator deferoxamine (DFO), thus avoiding obstacles or safety risks associated with gene therapy. Rat pancreatic islets were infected in vivo using an adenovirus (ADE) encoding human VEGF gene (4.108 pfu/pancreas) or were incubated in the presence of DFO (10 μmol/L). In vitro viability, functionality, and the secretion of VEGF were evaluated in islets 1 and 3 days after treatment. Infected islets or islets incubated with DFO were transplanted into the liver of syngenic diabetic rats and the graft efficiency was estimated in vivo by measuring body weight, glycemia, C-peptide secretion, and animal survival over a period of 2 months. DFO induced transient VEGF overexpression over 3 days, whereas infection with ADE resulted in prolonged VEGF overexpression lasting 14 days; however, this was toxic and decreased islet viability and functionality. The in vivo study showed a decrease in rat deaths after the transplantation of islets treated with DFO or ADE compared with the sham and control group. ADE treatment improved body weight and C-peptide levels. Gene therapy and DFO improved metabolic control in diabetic rats after transplantation, but this effect was limited in the presence of DFO. The pharmacological approach is an interesting strategy for improving graft efficiency during transplantation, but this approach needs to be improved with drugs that are more specific.
Collapse
Affiliation(s)
- A. Langlois
- Centre européen d'étude du Diabète, Strasbourg, France
| | - W. Bietiger
- Centre européen d'étude du Diabète, Strasbourg, France
| | - E. Seyfritz
- Centre européen d'étude du Diabète, Strasbourg, France
| | - E. Maillard
- Centre européen d'étude du Diabète, Strasbourg, France
| | - K. Vivot
- Centre européen d'étude du Diabète, Strasbourg, France
| | - C. Peronet
- Centre européen d'étude du Diabète, Strasbourg, France
| | - N. Meyer
- Faculté de Médecine de Strasbourg, Laboratoire de Biostatistique, Strasbourg, France
| | - L. Kessler
- Service d'endocrinologie, de diabète et des maladies métaboliques, Médicale B Hopital civil, Strasbourg cedex, France
- Université de Strasbourg (UdS), Strasbourg cedex, France
| | - N. Jeandidier
- Service d'endocrinologie, de diabète et des maladies métaboliques, Médicale B Hopital civil, Strasbourg cedex, France
- Université de Strasbourg (UdS), Strasbourg cedex, France
| | - M. Pinget
- Centre européen d'étude du Diabète, Strasbourg, France
- Service d'endocrinologie, de diabète et des maladies métaboliques, Médicale B Hopital civil, Strasbourg cedex, France
- Université de Strasbourg (UdS), Strasbourg cedex, France
| | - S. Sigrist
- Centre européen d'étude du Diabète, Strasbourg, France
| |
Collapse
|
34
|
Vaithilingam V, Tuch BE. Islet transplantation and encapsulation: an update on recent developments. Rev Diabet Stud 2011; 8:51-67. [PMID: 21720673 DOI: 10.1900/rds.2011.8.51] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Human islet transplantation can provide good glycemic control in diabetic recipients without exogenous insulin. However, a major factor limiting its application is the recipient's need to adhere to life-long immunosuppression, something that has serious side effects. Microencapsulating human islets is a strategy that should prevent rejection of the grafted tissue without the need for anti-rejection drugs. Despite promising studies in various animal models, the encapsulated human islets so far have not made an impact in the clinical setting. Many non-immunological and immunological factors such as biocompatibility, reduced immunoprotection, hypoxia, pericapsular fibrotic overgrowth, effects of the encapsulation process and post-transplant inflammation hamper the successful application of this promising technology. In this review, strategies are discussed to overcome the above-mentioned factors and to enhance the survival and function of encapsulated insulin-producing cells, whether in islets or surrogate β-cells. Studies at our center show that barium alginate microcapsules are biocompatible in rodents, but not in humans, raising concerns over the use of rodents to predict outcomes. Studies at our center also show that the encapsulation process had little or no effect on the cellular transcriptome of human islets and on their ability to function either in vitro or in vivo. New approaches incorporating further modifications to the microcapsule surface to prevent fibrotic overgrowth are vital, if encapsulated human islets or β-cell surrogates are to become a viable therapy option for type 1 diabetes in humans.
Collapse
|
35
|
Lau J, Zang G, Carlsson PO. Pancreatic islet transplantation to the liver: how can vascularization problems be resolved? ACTA ACUST UNITED AC 2011. [DOI: 10.2217/dmt.11.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|