1
|
Rohban R, Martins CP, Esni F. Advanced therapy to cure diabetes: mission impossible is now possible? Front Cell Dev Biol 2024; 12:1484859. [PMID: 39629270 PMCID: PMC11611888 DOI: 10.3389/fcell.2024.1484859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Cell and Gene therapy are referred to as advanced therapies that represent overlapping fields of regenerative medicine. They have similar therapeutic goals such as to modify cellular identity, improve cell function, or fight a disease. These two therapeutic avenues, however, possess major differences. While cell therapy involves introduction of new cells, gene therapy entails introduction or modification of genes. Furthermore, the aim of cell therapy is often to replace, or repair damaged tissue, whereas gene therapy is used typically as a preventive approach. Diabetes mellitus severely affects the quality of life of afflicted individuals and has various side effects including cardiovascular, ophthalmic disorders, and neuropathy while putting enormous economic pressure on both the healthcare system and the patient. In recent years, great effort has been made to develop cutting-edge therapeutic interventions for diabetes treatment, among which cell and gene therapies stand out. This review aims to highlight various cell- and gene-based therapeutic approaches leading to the generation of new insulin-producing cells as a topmost "panacea" for treating diabetes, while deliberately avoiding a detailed molecular description of these approaches. By doing so, we aim to target readers who are new to the field and wish to get a broad helicopter overview of the historical and current trends of cell- and gene-based approaches in β-cell regeneration.
Collapse
Affiliation(s)
- Rokhsareh Rohban
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Christina P. Martins
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Farzad Esni
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
- UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- McGowan Institute for regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
2
|
Maestas MM, Bui MH, Millman JR. Recent progress in modeling and treating diabetes using stem cell-derived islets. Stem Cells Transl Med 2024; 13:949-958. [PMID: 39159002 PMCID: PMC11465181 DOI: 10.1093/stcltm/szae059] [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: 05/02/2024] [Accepted: 07/12/2024] [Indexed: 08/21/2024] Open
Abstract
Stem cell-derived islets (SC-islets) offer the potential to be an unlimited source of cells for disease modeling and the treatment of diabetes. SC-islets can be genetically modified, treated with chemical compounds, or differentiated from patient derived stem cells to model diabetes. These models provide insights into disease pathogenesis and vulnerabilities that may be targeted to provide treatment. SC-islets themselves are also being investigated as a cell therapy for diabetes. However, the transplantation process is imperfect; side effects from immunosuppressant use have reduced SC-islet therapeutic potential. Alternative methods to this include encapsulation, use of immunomodulating molecules, and genetic modification of SC-islets. This review covers recent advances using SC-islets to understand different diabetes pathologies and as a cell therapy.
Collapse
Affiliation(s)
- Marlie M Maestas
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, United States
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Maggie H Bui
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Jeffrey R Millman
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, United States
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, United States
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States
| |
Collapse
|
3
|
Hogrebe NJ, Ishahak M, Millman JR. Developments in stem cell-derived islet replacement therapy for treating type 1 diabetes. Cell Stem Cell 2023; 30:530-548. [PMID: 37146579 PMCID: PMC10167558 DOI: 10.1016/j.stem.2023.04.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/20/2023] [Accepted: 04/05/2023] [Indexed: 05/07/2023]
Abstract
The generation of islet-like endocrine clusters from human pluripotent stem cells (hPSCs) has the potential to provide an unlimited source of insulin-producing β cells for the treatment of diabetes. In order for this cell therapy to become widely adopted, highly functional and well-characterized stem cell-derived islets (SC-islets) need to be manufactured at scale. Furthermore, successful SC-islet replacement strategies should prevent significant cell loss immediately following transplantation and avoid long-term immune rejection. This review highlights the most recent advances in the generation and characterization of highly functional SC-islets as well as strategies to ensure graft viability and safety after transplantation.
Collapse
Affiliation(s)
- Nathaniel J Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA.
| | - Matthew Ishahak
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA; Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, USA.
| |
Collapse
|
4
|
Ahmed HH, Aglan HA, Beherei HH, Mabrouk M, Mahmoud NS. The promising role of hypoxia-resistant insulin-producing cells in ameliorating diabetes mellitus in vivo. Future Sci OA 2022; 8:FSO811. [PMID: 36248064 PMCID: PMC9540411 DOI: 10.2144/fsoa-2022-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/25/2022] [Indexed: 11/23/2022] Open
Abstract
AIM This study aimed to evaluate the efficacy of hypoxia-persistent insulin-producing cells (IPCs) against diabetes in vivo. MATERIALS & METHODS Mesenchymal stem cells (MSCs) differentiation into IPCs in the presence of Se/Ti (III) or CeO2 nanomaterials. IPCs were subjected to hypoxia and hypoxia genes were analyzed. PKH-26-labeled IPCs were infused in diabetic rats to evaluate their anti-diabetic potential. RESULTS MSCs were differentiated into functional IPCs. IPCs exhibited overexpression of anti-apoptotic genes and down-expression of hypoxia and apoptotic genes. IPCs implantation elicited glucose depletion and elevated insulin, HK and G6PD levels. They provoked VEGF and PDX-1 upregulation and HIF-1α and Caspase-3 down-regulation. IPCs transplantation ameliorated the destabilization of pancreatic tissue architecture. CONCLUSION The chosen nanomaterials were impressive in generating hypoxia-resistant IPCs that could be an inspirational strategy for curing diabetes.
Collapse
Affiliation(s)
- Hanaa H Ahmed
- Hormones Department, Medical Research & Clinical Studies Institute, National Research Centre, Giza, 12622, Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, 12622, Egypt
| | - Hadeer A Aglan
- Hormones Department, Medical Research & Clinical Studies Institute, National Research Centre, Giza, 12622, Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, 12622, Egypt
| | - Hanan H Beherei
- Refractories, Ceramics & Building Materials Department, Advanced Materials Technology & Mineral Resources Research Institute, National Research Centre, Giza, 12622, Egypt
| | - Mostafa Mabrouk
- Refractories, Ceramics & Building Materials Department, Advanced Materials Technology & Mineral Resources Research Institute, National Research Centre, Giza, 12622, Egypt
| | - Nadia S Mahmoud
- Hormones Department, Medical Research & Clinical Studies Institute, National Research Centre, Giza, 12622, Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, 12622, Egypt
| |
Collapse
|
5
|
Mooranian A, Ionescu CM, Walker D, Jones M, Wagle SR, Kovacevic B, Chester J, Foster T, Johnston E, Kuthubutheen J, Brown D, Atlas MD, Mikov M, Al-Salami H. Single-Cellular Biological Effects of Cholesterol-Catabolic Bile Acid-Based Nano/Micro Capsules as Anti-Inflammatory Cell Protective Systems. Biomolecules 2022; 12:biom12010073. [PMID: 35053221 PMCID: PMC8773943 DOI: 10.3390/biom12010073] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 01/27/2023] Open
Abstract
Recent studies in our laboratories have shown promising effects of bile acids in ➀ drug encapsulation for oral targeted delivery (via capsule stabilization) particularly when encapsulated with Eudragit NM30D® and ➁ viable-cell encapsulation and delivery (via supporting cell viability and biological activities, postencapsulation). Accordingly, this study aimed to investigate applications of bile acid-Eudragit NM30D® capsules in viable-cell encapsulation ready for delivery. Mouse-cloned pancreatic β-cell line was cultured and cells encapsulated using bile acid-Eudragit NM30D® capsules, and capsules' images, viability, inflammation, and bioenergetics of encapsulated cells assessed. The capsules' thermal and chemical stability assays were also assessed to ascertain an association between capsules' stability and cellular biological activities. Bile acid-Eudragit NM30D® capsules showed improved cell viability (e.g., F1 < F2 & F8; p < 0.05), insulin, inflammatory profile, and bioenergetics as well as thermal and chemical stability, compared with control. These effects were formulation-dependent and suggest, overall, that changes in ratios of bile acids to Eudragit NM30D® can change the microenvironment of the capsules and subsequent cellular biological activities.
Collapse
Affiliation(s)
- Armin Mooranian
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Corina Mihaela Ionescu
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Daniel Walker
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Melissa Jones
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Susbin Raj Wagle
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Bozica Kovacevic
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Jacqueline Chester
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Thomas Foster
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Edan Johnston
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | | | - Daniel Brown
- Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia;
| | - Marcus D. Atlas
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Momir Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, 21101 Novi Sad, Serbia;
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
- Correspondence: ; Tel.: +61-8-9266-9816; Fax: +61-8-9266-2769
| |
Collapse
|
6
|
Xu Y, Chen J, Zhou H, Wang J, Song J, Xie J, Guo Q, Wang C, Huang Q. Effects and mechanism of stem cells from human exfoliated deciduous teeth combined with hyperbaric oxygen therapy in type 2 diabetic rats. Clinics (Sao Paulo) 2020; 75:e1656. [PMID: 32520222 PMCID: PMC7247751 DOI: 10.6061/clinics/2020/e1656] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/10/2020] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVES Mesenchymal stem cells (MSCs) are potentially ideal for type 2 diabetes treatment, owing to their multidirectional differentiation ability and immunomodulatory properties. Here we investigated whether the stem cells from human exfoliated deciduous teeth (SHED) in combination with hyperbaric oxygen (HBO) could treat type 2 diabetic rats, and explored the underlying mechanism. METHODS SD rats were used to generate a type 2 diabetes model, which received stem cell therapy, HBO therapy, or both together. Before and after treatment, body weight, blood glucose, and serum insulin, blood lipid, pro-inflammatory cytokines (tumor necrosis factor-alpha and interleukin-6), and urinary proteins were measured and compared. After 6 weeks, rats were sacrificed and their organs were subjected to hematoxylin and eosin staining and immunofluorescence staining for insulin and glucagon; apoptosis and proliferation were analyzed in islet cells. Structural changes in islets were observed under an electron microscope. Expression levels of Pdx1, Ngn3, and Pax4 mRNAs in the pancreas were assessed by real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS In comparison with diabetic mice, those treated with the combination or SHE therapy showed decreased blood glucose, insulin resistance, serum lipids, and pro-inflammatory cytokines and increased body weight and serum insulin. The morphology and structure of pancreatic islets improved, as evident from an increase in insulin-positive cells and a decrease in glucagon-positive cells. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining of islet cells revealed the decreased apoptosis index, while Ki67 and proliferating cell nuclear antigen staining showed increased proliferation index. Pancreatic expression of Pdx1, Ngn3, and Pax4 was upregulated. CONCLUSION SHED combined with HBO therapy was effective for treating type 2 diabetic rats. The underlying mechanism may involve SHED-mediated increase in the proliferation and trans-differentiation of islet β-cells and decrease in pro-inflammatory cytokines and apoptosis of islets.
Collapse
Affiliation(s)
- Yifeng Xu
- Department of Endocrinology, Changhai Hospital, the First Affiliated Hospital of the Naval Medical University, Shanghai 200433, China
- Department of Endocrinology, Air Force Hospital of Northern Theater Command of PLA, Shenyang 110042, China
| | - Jin Chen
- Department of Endocrinology, Changhai Hospital, the First Affiliated Hospital of the Naval Medical University, Shanghai 200433, China
| | - Hui Zhou
- Department of Out-patient, Changning retired cadre retreat of Shanghai garrison command, Shanghai 200050, China
| | - Jing Wang
- Department of Endocrinology, Changhai Hospital, the First Affiliated Hospital of the Naval Medical University, Shanghai 200433, China
- Department of Internal Medcine, Hotan Country People’s Hospital of Xinjiang, Hotan Country 848000, China
| | - Jingyun Song
- Department of Endocrinology, Changhai Hospital, the First Affiliated Hospital of the Naval Medical University, Shanghai 200433, China
| | - Junhao Xie
- Department of Endocrinology, Changhai Hospital, the First Affiliated Hospital of the Naval Medical University, Shanghai 200433, China
| | - Qingjun Guo
- Department of Endocrinology, Changhai Hospital, the First Affiliated Hospital of the Naval Medical University, Shanghai 200433, China
| | - Chaoqun Wang
- Department of Endocrinology, Changhai Hospital, the First Affiliated Hospital of the Naval Medical University, Shanghai 200433, China
| | - Qin Huang
- Department of Endocrinology, Changhai Hospital, the First Affiliated Hospital of the Naval Medical University, Shanghai 200433, China
- *Corresponding author. E-mail:
| |
Collapse
|
7
|
Ernst AU, Bowers DT, Wang LH, Shariati K, Plesser MD, Brown NK, Mehrabyan T, Ma M. Nanotechnology in cell replacement therapies for type 1 diabetes. Adv Drug Deliv Rev 2019; 139:116-138. [PMID: 30716349 PMCID: PMC6677642 DOI: 10.1016/j.addr.2019.01.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/17/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022]
Abstract
Islet transplantation is a promising long-term, compliance-free, complication-preventing treatment for type 1 diabetes. However, islet transplantation is currently limited to a narrow set of patients due to the shortage of donor islets and side effects from immunosuppression. Encapsulating cells in an immunoisolating membrane can allow for their transplantation without the need for immunosuppression. Alternatively, "open" systems may improve islet health and function by allowing vascular ingrowth at clinically attractive sites. Many processes that enable graft success in both approaches occur at the nanoscale level-in this review we thus consider nanotechnology in cell replacement therapies for type 1 diabetes. A variety of biomaterial-based strategies at the nanometer range have emerged to promote immune-isolation or modulation, proangiogenic, or insulinotropic effects. Additionally, coating islets with nano-thin polymer films has burgeoned as an islet protection modality. Materials approaches that utilize nanoscale features manipulate biology at the molecular scale, offering unique solutions to the enduring challenges of islet transplantation.
Collapse
Affiliation(s)
- Alexander U Ernst
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Daniel T Bowers
- 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
| | - Kaavian Shariati
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Mitchell D Plesser
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Natalie K Brown
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Tigran Mehrabyan
- 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
|
8
|
Kim JS, Jung Y, Kim SH, Shin JS, Kim SH, Park CG. Vascularization of PLGA-based bio-artificial beds by hypoxia-preconditioned mesenchymal stem cells for subcutaneous xenogeneic islet transplantation. Xenotransplantation 2018; 26:e12441. [PMID: 30054954 DOI: 10.1111/xen.12441] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/06/2018] [Accepted: 06/11/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Subcutaneous tissue is an attractive extra-hepatic heterotopic site for islet transplantation; however, poor oxygen tension and blood supply during early engraftment of implanted islets have limited the use of this site in clinical applications. METHODS This study investigated the vascularization potential of hypoxia-preconditioned mesenchymal stem cells (3% O2 ; hypo-MSCs) in PLGA-based bio-artificial beds for subsequent subcutaneous islet transplantation. Sheet-typed polymeric PLGA scaffolds coated with hypo-MSCs or normo-MSCs (MSCs cultured under normoxia conditions, 21% O2 ) were implanted subcutaneously in mice. RESULTS Compared to normo-MSCs, hypo-MSCs significantly enhanced vasculogenesis, both on the interior and exterior surfaces of the implanted PLGA devices, which peaked 4 weeks after implantation. Further, infusion of porcine islets inside the prevascularized PLGA bed restored normal glycemic control in 6 of 6 STZ-induced diabetic mice. The mass of the marginal islet was approximately 2000 IEQs, which is comparable to that required for the renal subcapsular space, a highly vascularized site. CONCLUSIONS Therefore, PLGA-based bio-artificial devices prevascularized with hypo-MSCs could be a useful modality for successful subcutaneous islet transplantation, which is of high clinical relevance.
Collapse
Affiliation(s)
- Jung-Sik Kim
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, Korea
| | - Su Hee Kim
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, Korea
| | - Jun-Seop Shin
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Hyun Kim
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, Korea
| | - Chung-Gyu Park
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Biomedical Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Zhang D, Liu Y, Tang Y, Wang X, Li Z, Li R, Ti Z, Gao W, Bai J, Lv Y. Increased mitochondrial fission is critical for hypoxia-induced pancreatic beta cell death. PLoS One 2018; 13:e0197266. [PMID: 29768513 PMCID: PMC5955518 DOI: 10.1371/journal.pone.0197266] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 04/30/2018] [Indexed: 01/27/2023] Open
Abstract
Hypoxia-mediated pancreatic beta cell death is one of the main causes of pancreatic beta celldeath, which leads to the loss of functional pancreatic beta cell mass and type 1 diabetes andtype 2 diabetes.However, the molecular mechanisms that control life and death of pancreatic beta cells remain poorly understood. Here we showed that mitochondrial fission was strongly induced in pancreatic beta cellsmainly due to an elevation of DRP1S616 phosphorylation through HIF-1αactivation and subsequent DRP1 mitochondrial translocation. Hypoxia-induced pancreatic beta cell death can be reversed by the inhibition of mitochondrial fission viaDRP1 knockdown. We further demonstrated that hypoxia-induced mitochondrial fission untightened the cristae formation, which subsequently triggers mitochondrial cytochrome c release and consequent caspase activation. Moreover, treatment with mitochondrial division inhibitor-1 (Mdivi-1), a specific inhibitor of DRP1-mediated mitochondrial fission, significantly suppressedbeta cell death in vitro, indicating a promising therapeutic strategy for treatment of diabetes.Taken together, our results reveal a crucial role for the DRP1-mediated mitochondrial fission in hypoxia-induced beta cell death, which provides a strong evidence for thisprocess as drug target indiabetestreatment.
Collapse
Affiliation(s)
- Da Zhang
- Department of Hepatobiliary Surgery, Institute of Advanced Surgical Technology and Engineering, Shaanxi Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of General Surgery, Xi'an No.3 Hospital, Xi’an, China
| | - Yanfang Liu
- Department of Ophthalmology, Xi'an Children’s Hospital,Xi’an, China
| | - Yao Tang
- Department of General Surgery, Xi'an No.3 Hospital, Xi’an, China
| | - Xiaofeng Wang
- Department of General Surgery, Xi'an No.3 Hospital, Xi’an, China
| | - Zhichao Li
- Department of General Surgery, Xi'an No.3 Hospital, Xi’an, China
| | - Rui Li
- Department of General Surgery, Xi'an No.3 Hospital, Xi’an, China
| | - Zhenyu Ti
- Department of General Surgery, Xi'an No.3 Hospital, Xi’an, China
| | - Weidong Gao
- Department of General Surgery, Xi'an No.3 Hospital, Xi’an, China
| | - Jigang Bai
- Department of Hepatobiliary Surgery, Institute of Advanced Surgical Technology and Engineering, Shaanxi Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yi Lv
- Department of Hepatobiliary Surgery, Institute of Advanced Surgical Technology and Engineering, Shaanxi Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- * E-mail:
| |
Collapse
|
10
|
Abstract
Review of emerging advances and persisting challenges in the engineering and translation of islet encapsulation technologies.
Collapse
Affiliation(s)
| | - Long-Hai Wang
- Department of Biological and Environmental Engineering
- Cornell University
- Ithaca
- USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering
- Cornell University
- Ithaca
- USA
| |
Collapse
|
11
|
Kumar S, Marriott CE, Alhasawi NF, Bone AJ, Macfarlane WM. The role of tumour suppressor PDCD4 in beta cell death in hypoxia. PLoS One 2017; 12:e0181235. [PMID: 28750063 PMCID: PMC5531437 DOI: 10.1371/journal.pone.0181235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 06/28/2017] [Indexed: 12/31/2022] Open
Abstract
Objective Hypoxia is known to induce pancreatic beta cell dysfunction and apoptosis. Changes in Programmed Cell Death Gene 4 (PDCD4) expression have previously been linked with beta cell neogenesis and function. Our aim was to investigate the effects of hypoxia on cell viability, PDCD4 expression and subcellular localisation. Methods MIN6 beta cells and ARIP ductal cells were exposed to 1% (hypoxia) or 21% O2 (normoxia) for 12 or 24 hours. MTT assay, HPI staining, scanning electron microscopy, western blotting and immunocytochemistry analyses were performed to determine the effect of hypoxia on cell viability, morphology and PDCD4 expression. Results 24 hour exposure to hypoxia resulted in ~70% loss of beta cell viability (P<0.001) compared to normoxia. Both HPI staining and SEM analysis demonstrated beta cell apoptosis and necrosis after 12 hours exposure to hypoxia. ARIP cells also displayed hypoxia-induced apoptosis and altered surface morphology after 24 hours, but no significant growth difference (p>0.05) was observed between hypoxic and normoxic conditions. Significantly higher expression of PDCD4 was observed in both beta cells (P<0.001) and ductal (P<0.01) cells under hypoxic conditions compared to controls. PDCD4 expression was localised to the cytoplasm of both beta cells and ductal cells, with no observed effects of hypoxia, normoxia or serum free conditions on intracellular shuttling of PDCD4. Conclusion These findings indicate that hypoxia-induced expression of PDCD4 is associated with increased beta cell death and suggests that PDCD4 may be an important factor in regulating beta cell survival during hypoxic stress.
Collapse
Affiliation(s)
- Sandeep Kumar
- Diabetes Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Claire E. Marriott
- Diabetes Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Nouf F. Alhasawi
- Diabetes Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Adrian J. Bone
- Diabetes Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Wendy M. Macfarlane
- Diabetes Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
- * E-mail:
| |
Collapse
|
12
|
Rodriguez-Brotons A, Bietiger W, Peronet C, Langlois A, Magisson J, Mura C, Sookhareea C, Polard V, Jeandidier N, Zal F, Pinget M, Sigrist S, Maillard E. Comparison of Perfluorodecalin and HEMOXCell as Oxygen Carriers for Islet Oxygenation in an In Vitro Model of Encapsulation. Tissue Eng Part A 2016; 22:1327-1336. [PMID: 27796164 DOI: 10.1089/ten.tea.2016.0064] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Transplantation of encapsulated islets in a bioartificial pancreas is a promising alternative to free islet cell therapy to avoid immunosuppressive regimens. However, hypoxia, which can induce a rapid loss of islets, is a major limiting factor. The efficiency of oxygen delivery in an in vitro model of bioartificial pancreas involving hypoxia and confined conditions has never been investigated. Oxygen carriers such as perfluorocarbons and hemoglobin might improve oxygenation. To verify this hypothesis, this study aimed to identify the best candidate of perfluorodecalin (PFD) or HEMOXCell® to reduce cellular hypoxia in a bioartificial pancreas in an in vitro model of encapsulation ex vivo. The survival, hypoxia, and inflammation markers and function of rat islets seeded at 600 islet equivalents (IEQ)/cm2 and under 2% pO2 were assessed in the presence of 50 μg/mL of HEMOXCell or 10% PFD with or without adenosine. Both PFD and HEMOXCell increased the cell viability and decreased markers of hypoxia (hypoxia-inducible factor mRNA and protein). In these culture conditions, adenosine had deleterious effects, including an increase in cyclooxygenase-2 and interleukin-6, in correlation with unregulated proinsulin release. Despite the effectiveness of PFD in decreasing hypoxia, no restoration of function was observed and only HEMOXCell had the capacity to restore insulin secretion to a normal level. Thus, it appeared that the decrease in cell hypoxia as well as the intrinsic superoxide dismutase activity of HEMOXCell were both mandatory to maintain islet function under hypoxia and confinement. In the context of islet encapsulation in a bioartificial pancreas, HEMOXCell is the candidate of choice for application in vivo.
Collapse
Affiliation(s)
| | - William Bietiger
- 1 Université de Strasbourg, Centre Européen d'Etude du Diabète, Strasbourg, France
| | - Claude Peronet
- 1 Université de Strasbourg, Centre Européen d'Etude du Diabète, Strasbourg, France
| | - Allan Langlois
- 1 Université de Strasbourg, Centre Européen d'Etude du Diabète, Strasbourg, France
| | | | - Carole Mura
- 1 Université de Strasbourg, Centre Européen d'Etude du Diabète, Strasbourg, France
| | - Cynthia Sookhareea
- 1 Université de Strasbourg, Centre Européen d'Etude du Diabète, Strasbourg, France
| | - Valerie Polard
- 4 HEMARINA Aéropôle Centre , Biotechnopôle, Morlaix, France
| | - Nathalie Jeandidier
- 1 Université de Strasbourg, Centre Européen d'Etude du Diabète, Strasbourg, France .,2 Structure d'Endocrinologie, Diabète-Nutrition et Addictologie, Pôle NUDE, Hôpitaux Universitaires de Strasbourg (HUS) , Strasbourg, France
| | - Franck Zal
- 4 HEMARINA Aéropôle Centre , Biotechnopôle, Morlaix, France
| | - Michel Pinget
- 1 Université de Strasbourg, Centre Européen d'Etude du Diabète, Strasbourg, France .,2 Structure d'Endocrinologie, Diabète-Nutrition et Addictologie, Pôle NUDE, Hôpitaux Universitaires de Strasbourg (HUS) , Strasbourg, France
| | - Séverine Sigrist
- 1 Université de Strasbourg, Centre Européen d'Etude du Diabète, Strasbourg, France
| | - Elisa Maillard
- 1 Université de Strasbourg, Centre Européen d'Etude du Diabète, Strasbourg, France
| |
Collapse
|
13
|
Vivot K, Benahmed MA, Seyfritz E, Bietiger W, Elbayed K, Ruhland E, Langlois A, Maillard E, Pinget M, Jeandidier N, Gies JP, Namer IJ, Sigrist S, Reix N. A Metabolomic Approach ( 1H HRMAS NMR Spectroscopy) Supported by Histology to Study Early Post-transplantation Responses in Islet-transplanted Livers. Int J Biol Sci 2016; 12:1168-1180. [PMID: 27766032 PMCID: PMC5069439 DOI: 10.7150/ijbs.15189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/28/2016] [Indexed: 11/05/2022] Open
Abstract
Intrahepatic transplantation of islets requires a lot of islets because more than 50% of the graft is lost during the 24 hours following transplantation. We analyzed, in a rat model, early post-transplantation inflammation using systemic inflammatory markers, or directly in islet-transplanted livers by immunohistochemistry. 1H HRMAS NMR was employed to investigate metabolic responses associated with the transplantation. Inflammatory markers (Interleukin-6, α2-macroglobulin) are not suitable to follow islet reactions as they are not islet specific. To study islet specific inflammatory events, immunohistochemistry was performed on sections of islet transplanted livers for thrombin (indicator of the instant blood-mediated inflammatory reaction (IBMIR)) and granulocytes and macrophages. We observed a specific correlation between IBMIR and granulocyte and macrophage infiltration after 12 h. In parallel, we identified a metabolic response associated with transplantation: after 12 h, glucose, alanine, aspartate, glutamate and glutathione were significantly increased. An increase of glucose is a marker of tissue degradation, and could be explained by immune cell infiltration. Alanine, aspartate and glutamate are inter-connected in a common metabolic pathway known to be activated during hypoxia. An increase of glutathione revealed the presence of antioxidant protection. In this study, IBMIR visualization combined with 1H HRMAS NMR facilitated the characterization of cellular and molecular pathways recruited following islet transplantation.
Collapse
Affiliation(s)
- Kevin Vivot
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Malika A. Benahmed
- ICube UMR 7357, Université de Strasbourg, CNRS, IMIS, 4 rue Kirschleger, 67085 Strasbourg, France
- Service de Biophysique et Médecine Nucléaire, Hôpitaux Universitaires de Strasbourg, 1 avenue Molière, 67100 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Faculté de médecine, Strasbourg, France
| | - Elodie Seyfritz
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - William Bietiger
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Karim Elbayed
- ICube UMR 7357, Université de Strasbourg, CNRS, IMIS, 4 rue Kirschleger, 67085 Strasbourg, France
| | - Elisa Ruhland
- ICube UMR 7357, Université de Strasbourg, CNRS, IMIS, 4 rue Kirschleger, 67085 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Faculté de médecine, Strasbourg, France
| | - Allan Langlois
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Elisa Maillard
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Michel Pinget
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
- Service d'Endocrinologie - Diabète et Maladies métaboliques, Hôpitaux Universitaires de Strasbourg, 1 place de l'Hôpital, 67091 Strasbourg, France
| | - Nathalie Jeandidier
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
- Service d'Endocrinologie - Diabète et Maladies métaboliques, Hôpitaux Universitaires de Strasbourg, 1 place de l'Hôpital, 67091 Strasbourg, France
| | - Jean-Pierre Gies
- UMR 7034 CNRS, Faculté de Pharmacie, Université de Strasbourg, 67 401 Illkirch, France
| | - Izzie-Jacques Namer
- ICube UMR 7357, Université de Strasbourg, CNRS, IMIS, 4 rue Kirschleger, 67085 Strasbourg, France
- Service de Biophysique et Médecine Nucléaire, Hôpitaux Universitaires de Strasbourg, 1 avenue Molière, 67100 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Faculté de médecine, Strasbourg, France
| | - Séverine Sigrist
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200 Strasbourg, France
| | - Nathalie Reix
- ICube UMR 7357, Université de Strasbourg, CNRS, IMIS, 4 rue Kirschleger, 67085 Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Faculté de médecine, Strasbourg, France
- Laboratoire de biochimie et biologie moléculaire, Hôpitaux Universitaires de Strasbourg, 1 place de l'Hôpital, 67091 Strasbourg, France
| |
Collapse
|
14
|
Bartlett ST, Markmann JF, Johnson P, Korsgren O, Hering BJ, Scharp D, Kay TWH, Bromberg J, Odorico JS, Weir GC, Bridges N, Kandaswamy R, Stock P, Friend P, Gotoh M, Cooper DKC, Park CG, O'Connell P, Stabler C, Matsumoto S, Ludwig B, Choudhary P, Kovatchev B, Rickels MR, Sykes M, Wood K, Kraemer K, Hwa A, Stanley E, Ricordi C, Zimmerman M, Greenstein J, Montanya E, Otonkoski T. Report from IPITA-TTS Opinion Leaders Meeting on the Future of β-Cell Replacement. Transplantation 2016; 100 Suppl 2:S1-44. [PMID: 26840096 PMCID: PMC4741413 DOI: 10.1097/tp.0000000000001055] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/07/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Stephen T. Bartlett
- Department of Surgery, University of Maryland School of Medicine, Baltimore MD
| | - James F. Markmann
- Division of Transplantation, Massachusetts General Hospital, Boston MA
| | - Paul Johnson
- Nuffield Department of Surgical Sciences and Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bernhard J. Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - David Scharp
- Prodo Laboratories, LLC, Irvine, CA
- The Scharp-Lacy Research Institute, Irvine, CA
| | - Thomas W. H. Kay
- Department of Medicine, St. Vincent’s Hospital, St. Vincent's Institute of Medical Research and The University of Melbourne Victoria, Australia
| | - Jonathan Bromberg
- Division of Transplantation, Massachusetts General Hospital, Boston MA
| | - Jon S. Odorico
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI
| | - Gordon C. Weir
- Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Nancy Bridges
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Raja Kandaswamy
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Peter Stock
- Division of Transplantation, University of San Francisco Medical Center, San Francisco, CA
| | - Peter Friend
- Nuffield Department of Surgical Sciences and Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Mitsukazu Gotoh
- Department of Surgery, Fukushima Medical University, Fukushima, Japan
| | - David K. C. Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
| | - Chung-Gyu Park
- Xenotransplantation Research Center, Department of Microbiology and Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Phillip O'Connell
- The Center for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, NSW, Australia
| | - Cherie Stabler
- Diabetes Research Institute, School of Medicine, University of Miami, Coral Gables, FL
| | - Shinichi Matsumoto
- National Center for Global Health and Medicine, Tokyo, Japan
- Otsuka Pharmaceutical Factory inc, Naruto Japan
| | - Barbara Ludwig
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden and DZD-German Centre for Diabetes Research, Dresden, Germany
| | - Pratik Choudhary
- Diabetes Research Group, King's College London, Weston Education Centre, London, United Kingdom
| | - Boris Kovatchev
- University of Virginia, Center for Diabetes Technology, Charlottesville, VA
| | - Michael R. Rickels
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Coulmbia University Medical Center, New York, NY
| | - Kathryn Wood
- Nuffield Department of Surgical Sciences and Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Kristy Kraemer
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Albert Hwa
- Juvenile Diabetes Research Foundation, New York, NY
| | - Edward Stanley
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Monash University, Melbourne, VIC, Australia
| | - Camillo Ricordi
- Diabetes Research Institute, School of Medicine, University of Miami, Coral Gables, FL
| | - Mark Zimmerman
- BetaLogics, a business unit in Janssen Research and Development LLC, Raritan, NJ
| | - Julia Greenstein
- Discovery Research, Juvenile Diabetes Research Foundation New York, NY
| | - Eduard Montanya
- Bellvitge Biomedical Research Institute (IDIBELL), Hospital Universitari Bellvitge, CIBER of Diabetes and Metabolic Diseases (CIBERDEM), University of Barcelona, Barcelona, Spain
| | - Timo Otonkoski
- Children's Hospital and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| |
Collapse
|
15
|
Lazard D, Vardi P, Bloch K. Anti-diabetic and neuroprotective effects of pancreatic islet transplantation into the central nervous system. Diabetes Metab Res Rev 2016; 32:11-20. [PMID: 25708430 DOI: 10.1002/dmrr.2644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/19/2015] [Indexed: 12/19/2022]
Abstract
During the last decades, the central nervous system (CNS) was intensively tested as a site for islet transplantation in different animal models of diabetes. Immunoprivilege properties of intracranial and intrathecal sites were found to delay and reduce rejection of transplanted allo-islets and xeno-islets, especially in the form of dispersed single cells. Insulin released from islets grafted in CNS was shown to cross the blood-brain barrier and to act as a regulator of peripheral glucose metabolism. In diabetic animals, sufficient nutrition and oxygen supply to islets grafted in the CNS provide adequate insulin response to increase glucose level resulting in rapid normoglycemia. In addition to insulin, pancreatic islets produce and secrete several other hormones, as well as neurotrophic and angiogenic factors with potential neuroprotective properties. Recent experimental studies and clinical trials provide a strong support for delivery of islet-derived macromolecules to CNS as a promising strategy to treat various brain disorders. This review article focuses mainly on analysis of current status of intracranial and intrathecal islet transplantations for treatment of experimental diabetes and discusses the possible neuroprotective properties of grafted islets into CNS as a novel therapeutic approach to brain disorders with cognitive dysfunctions characterized by impaired brain insulin signalling. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier
- Brain
- Central Nervous System
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/surgery
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/surgery
- Diabetic Neuropathies/prevention & control
- Disease Models, Animal
- Humans
- Hyperglycemia/prevention & control
- Hypoglycemia/prevention & control
- Insulin/metabolism
- Insulin Resistance
- Insulin Secretion
- Islets of Langerhans Transplantation/adverse effects
- Spinal Cord
- Subarachnoid Space
- Transplantation, Heterologous/adverse effects
- Transplantation, Heterotopic/adverse effects
Collapse
Affiliation(s)
- Daniel Lazard
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Pnina Vardi
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Konstantin Bloch
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| |
Collapse
|
16
|
Atchison N, Swindlehurst G, Papas KK, Tsapatsis M, Kokkoli E. Maintenance of ischemic β cell viability through delivery of lipids and ATP by targeted liposomes. Biomater Sci 2014; 2:548-559. [PMID: 24653833 PMCID: PMC3955996 DOI: 10.1039/c3bm60094g] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Islet transplantation is a promising treatment for type 1 diabetes, but despite the successes, existing challenges prevent widespread application. Ischemia, occurring during pancreas preservation and isolation, as well as after islet transplantation, decreases islet viability and function. We hypothesized that the liposomal delivery of adenosine triphosphate (ATP) could prevent the loss of cell viability during an ischemic insult. In this work we use a model β cell line, INS-1 to probe the liposome/cell interactions and examined the ability of liposomes functionalized with the fibronectin-mimetic peptide PR_b to facilitate the delivery of ATP to ischemic β cells. We demonstrate that PR_b increases the binding and internalization of liposomes to the β cells. Unexpectedly, when comparing the ability of PR_b liposomes with and without ATP to protect INS-1 cells from ischemia we found that both formulations increased cell survival. By probing the functional activity of ischemic cells treated with PR_b functionalized liposomes with and without ATP we find that both lipids and ATP play a role in maintaining cell metabolic activity after an ischemic insult and preventing cell necrosis. This approach may be beneficial for preventing ischemia related damage to islet cells, especially in the organ preservation stage.
Collapse
Affiliation(s)
- Nicole Atchison
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Garrett Swindlehurst
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA. Fax: 612- 626-7246; Tel: 612-626-1185
| | | | - Michael Tsapatsis
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA. Fax: 612- 626-7246; Tel: 612-626-1185
| | - Efrosini Kokkoli
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA. Fax: 612- 626-7246; Tel: 612-626-1185
| |
Collapse
|
17
|
Colton CK. Oxygen supply to encapsulated therapeutic cells. Adv Drug Deliv Rev 2014; 67-68:93-110. [PMID: 24582600 DOI: 10.1016/j.addr.2014.02.007] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 01/06/2014] [Accepted: 02/19/2014] [Indexed: 02/07/2023]
Abstract
Therapeutic cells encapsulated in immunobarrier devices have promise for treatment of a variety of human diseases without immunosuppression. The absence of sufficient oxygen supply to maintain viability and function of encapsulated tissue has been the most critical impediment to progress. Within the framework of oxygen supply limitations, we review the major issues related to development of these devices, primarily in the context of encapsulated islets of Langerhans for treating diabetes, including device designs and materials, supply of tissue, protection from immune rejection, and maintenance of cell viability and function. We describe various defensive measures investigated to enhance survival of transplanted tissue, and we review the diverse approaches to enhancement of oxygen transport to encapsulated tissue, including manipulation of diffusion distances and oxygen permeability of materials, induction of neovascularization with angiogenic factors and vascularizing membranes, and methods for increasing the oxygen concentration adjacent to encapsulated tissue so as to exceed that in the microvasculature. Recent developments, particularly in this latter area, suggest that the field is ready for clinical trials of encapsulated therapeutic cells to treat diabetes.
Collapse
|
18
|
Chhabra P, Brayman KL. Overcoming barriers in clinical islet transplantation: current limitations and future prospects. Curr Probl Surg 2014; 51:49-86. [PMID: 24411187 DOI: 10.1067/j.cpsurg.2013.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
19
|
Insulin-Like growth factor-II (IGF-II) prevents proinflammatory cytokine-induced apoptosis and significantly improves islet survival after transplantation. Transplantation 2013; 95:671-8. [PMID: 23364485 DOI: 10.1097/tp.0b013e31827fa453] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The early loss of functional islet mass (50-70%) due to apoptosis after clinical transplantation contributes to islet allograft failure. Insulin-like growth factor (IGF)-II is an antiapoptotic protein that is highly expressed in β-cells during development but rapidly decreases in postnatal life. METHODS We used an adenoviral (Ad) vector to overexpress IGF-II in isolated rat islets and investigated its antiapoptotic action against exogenous cytokines interleukin-1β- and interferon-γ-induced islet cell death in vitro. Using an immunocompromised marginal mass islet transplant model, the ability of Ad-IGF-II-transduced rat islets to restore euglycemia in nonobese diabetic/severe combined immunodeficient diabetic recipients was assessed. RESULTS Ad-IGF-II transduction did not affect islet viability or function. Ad-IGF-II cytokine-treated islets exhibited decreased cell death (40% ± 2.8%) versus Ad-GFP and untransduced control islets (63.2% ± 2.5% and 53.6% ± 2.3%, respectively). Ad-IGF-II overexpression during cytokine treatment resulted in a marked reduction in terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling-positive apoptotic cells (8.3% ± 1.4%) versus Ad-GFP control (41% ± 4.2%) and untransduced control islets (46.5% ± 6.2%). Western blot analysis confirmed that IGF-II inhibits apoptosis via activation of the phosphatidylinositol 3-kinase/Akt signaling pathway. Transplantation of IGF-II overexpressing islets under the kidney capsule of diabetic mice restored euglycemia in 77.8% of recipients compared with 18.2% and 47.5% of Ad-GFP and untransduced control islet recipients, respectively (P<0.05, log-rank [Mantel-Cox] test). CONCLUSIONS Antiapoptotic IGF-II decreases apoptosis in vitro and significantly improved islet transplant outcomes in vivo. Antiapoptotic gene transfer is a potentially powerful tool to improve islet survival after transplantation.
Collapse
|
20
|
Grundfest-Broniatowski S. What would surgeons like from materials scientists? WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2013; 5:299-319. [PMID: 23533092 DOI: 10.1002/wnan.1220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Surgery involves the repair, resection, replacement, or improvement of body parts and functions and in numerous ways, surgery should be considered human engineering. There are many areas in which surgical materials could be improved, but surgeons are generally unaware of materials available for use, while materials scientists do not know what surgeons require. This article will review some of the areas where surgeons and materials scientists have interacted in the past and will discuss some of the most pressing problems which remain to be solved. These include better implant materials for hernia repair, breast reconstruction, the treatment of diabetes, vascular stenting and reconstruction, and electrical pacing devices. The combination of tissue engineering and nanomaterials has great potential for application to nearly every aspect of surgery. Tissue engineering will allow cells or artificial organs to be grown for specific uses while nanotechnology will help to ensure maximal biocompatibility. Biosensors will be combined with improved electrodes and pacing devices to control impaired neurological functions.
Collapse
|
21
|
Kim JS, Lim JH, Nam HY, Lim HJ, Shin JS, Shin JY, Ryu JH, Kim K, Kwon IC, Jin SM, Kim HR, Kim SJ, Park CG. In situ application of hydrogel-type fibrin-islet composite optimized for rapid glycemic control by subcutaneous xenogeneic porcine islet transplantation. J Control Release 2012; 162:382-90. [PMID: 22820449 DOI: 10.1016/j.jconrel.2012.07.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 07/12/2012] [Indexed: 11/15/2022]
Abstract
Maximum engraftment of transplanted islets is essential for the clinical application of a subcutaneous site. Significant barriers to the current approaches are associated with their low effectiveness, complexity and unproven biosafety. Here, we evaluated and optimized a fibrin-islet composite for effective glycemic control in a subcutaneous site whose environment is highly hypoxic due to low vascularization potential. In the setting of xenogeneic porcine islet transplantation into the subcutaneous space of a diabetic mouse, the in vivo islet functions were greatly affected by the concentrations of fibrinogen and thrombin. The optimized hydrogel-type fibrin remarkably reduced the marginal islet mass to approximately one tenth that of islets without fibrin. This marginal islet mass was comparable to that in the setting of the subcapsular space of the kidney, which is a highly vascularized organ. Highly vascularized structures were generated inside and on the outer surface of the grafts. A hydrogel-type fibrin-islet composite established early diabetic control within an average of 3.4days after the transplantation. In the mechanistic studies, fibrin promoted local angiogenesis, enhanced islet viability and prevented fragmentation of islets into single cells. In conclusion, in situ application of hydrogel-type fibrin-islet composite may be a promising modality in the clinical success of subcutaneous islet transplantation.
Collapse
Affiliation(s)
- Jung-Sik Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul 110-799, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|