1
|
Shang KM, Suzuki T, Kato H, Toyoda T, Tai YC, Komatsu H. Oxygen dynamics and delivery strategies to enhance beta cell replacement therapy. Am J Physiol Cell Physiol 2025; 328:C1667-C1684. [PMID: 40204281 DOI: 10.1152/ajpcell.00984.2024] [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: 12/13/2024] [Revised: 01/06/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
Beta cell replacement therapy via pancreatic islet transplantation offers a promising treatment for type 1 diabetes as an alternative to insulin injections. However, posttransplantation oxygenation remains a critical challenge; isolated islets from donors lose vascularity and rely on slow oxygen diffusion for survival until revascularization occurs in the host tissue. This often results in significant hypoxia-induced acute graft loss. Overcoming the oxygenation barrier is crucial for advancing islet transplantation. This review is structured in three sections: the first examines oxygen dynamics in islet transplantation, focusing on factors affecting oxygen supply, including vascularity. It highlights oxygen dynamics specific to both transplant sites and islet grafts, with particular attention to extrahepatic sites such as subcutaneous tissue. The second section explores current oxygen delivery strategies, categorized into two main approaches: augmenting oxygen supply and enhancing effective oxygen solubility. The final section addresses key challenges, such as the lack of a clearly defined oxygen threshold for islet survival and the limited precision in measuring oxygen levels within small islet constructs. Recent advancements addressing these challenges are introduced. By deepening the understanding of oxygen dynamics and identifying current obstacles, this review aims to guide the development of innovative strategies for future research and clinical applications. These advancements are anticipated to enhance transplantation outcomes and bring us closer to a cure for type 1 diabetes.
Collapse
Affiliation(s)
- Kuang-Ming Shang
- Department of Medical Engineering, California Institute of Technology, Pasadena, California, United States
| | - Tomoharu Suzuki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hiroyuki Kato
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Taro Toyoda
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yu-Chong Tai
- Department of Medical Engineering, California Institute of Technology, Pasadena, California, United States
| | - Hirotake Komatsu
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| |
Collapse
|
2
|
Wong JM, Pepper AR. Status of islet transplantation and innovations to sustainable outcomes: novel sites, cell sources, and drug delivery strategies. FRONTIERS IN TRANSPLANTATION 2024; 3:1485444. [PMID: 39553396 PMCID: PMC11565603 DOI: 10.3389/frtra.2024.1485444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024]
Abstract
Islet transplantation (ITx) is an effective means to restore physiologic glycemic regulation in those living with type 1 diabetes; however, there are a handful of barriers that prevent the broad application of this functionally curative procedure. The restricted cell supply, requisite for life-long toxic immunosuppression, and significant immediate and gradual graft attrition limits the procedure to only those living with brittle diabetes. While intraportal ITx is the primary clinical site, portal vein-specific factors including low oxygen tension and the instant blood-mediated inflammatory reaction are detrimental to initial engraftment and long-term function. These factors among others prevent the procedure from granting recipients long-term insulin independence. Herein, we provide an overview of the status and limitations of ITx, and novel innovations that address the shortcomings presented. Despite the marked progress highlighted in the review from as early as the initial islet tissue transplantation in 1893, ongoing efforts to improve the procedure efficacy and success are also explored. Progress in identifying unlimited cell sources, more favourable transplant sites, and novel drug delivery strategies all work to broaden ITx application and reduce adverse outcomes. Exploring combination of these approaches may uncover synergies that can further advance the field of ITx in providing sustainable functional cures. Finally, the potential of biomaterial strategies to facilitate immune evasion and local immune modulation are featured and may underpin successful application in alternative transplant sites.
Collapse
Affiliation(s)
| | - Andrew R. Pepper
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
3
|
Li S, Fan L, Viktoria U, Oleksandr P, Li Z, Zhang W, Deng B. Effect of resuscitation of cryopreserved porcine adrenal glands at 26 °C on their recovery and functioning under xenotransplantation. Cryobiology 2024; 115:104895. [PMID: 38616031 DOI: 10.1016/j.cryobiol.2024.104895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
The study is devoted to the effect of lowered resuscitation temperature (26 °C) on cryopreserved porcine adrenal glands functional activity in vitro and in vivo under xenotransplantation. The adrenals were collected from newborn pigs, cryopreserved with 5 % DMSO at a rate of 1 °C/min, resuscitated at 26 or 37 °C for 48 h (5 % CO2, DMEM), embedded into small intestinal submucosa, and transplanted to bilaterally adrenalectomized rats. It has been shown that the glands resuscitated at 26 °C have suppressed free-radical processes and can produce cortisol and aldosterone in vitro, and may lead to elevated blood levels of these hormones. Moreover, the adrenal grafts maintain blood glucose levels and promote the formation of glycogen stores. Thus, the resuscitation at 26 °C can improve the quality of grafts and favor the introduction and application of the cryopreserved organs and tissues for transplantation in clinical and experimental practice.
Collapse
Affiliation(s)
- Shasha Li
- College of Basic Medical and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang City, China.
| | - Lingling Fan
- College of Basic Medical and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang City, China.
| | - Ustichenko Viktoria
- Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, Kharkiv, Ukraine.
| | - Pakhomov Oleksandr
- Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, Kharkiv, Ukraine.
| | - Zhongjie Li
- College of Basic Medical and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang City, China.
| | - Wenlu Zhang
- College of Basic Medical and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang City, China.
| | - Bo Deng
- College of Basic Medical and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang City, China.
| |
Collapse
|
4
|
Hasse JM, Meng S, Silpe S, Naziruddin B. Nutrition challenges following total pancreatectomy with islet autotransplantation. Nutr Clin Pract 2024; 39:86-99. [PMID: 38213274 DOI: 10.1002/ncp.11106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 01/13/2024] Open
Abstract
Total pancreatectomy with islet autotransplantation (TPIAT) is a surgical treatment option for patients with chronic pancreatitis who have not responded to other therapies. TP offers pain relief whereas IAT preserves beta cell mass to reduce endocrine insufficiency. During the surgical procedure, the entire pancreas is removed. Islet cells from the pancreas are then isolated, purified, and infused into the liver via the portal vein. Successful TPIAT relieves pain for a majority of patients but is not without obstacles, specifically gastrointestinal, exocrine, and endocrine challenges. The postoperative phase can be complicated by gastrointestinal symptoms causing patients to have difficulty regaining adequate oral intake. Enteral nutrition is frequently provided as a bridge to oral diet. Patients undergoing TPIAT must be monitored for macronutrient and micronutrient deficiencies following the procedure. Exocrine insufficiency must be treated lifelong with pancreatic enzyme replacement therapy. Endocrine function must be monitored and exogenous insulin provided in the postoperative phase; however, a majority of patients undergoing TPIAT require little or no long-term insulin. Although TPIAT can be a successful option for patients with chronic pancreatitis, nutrition-related concerns must be addressed for optimal recovery.
Collapse
Affiliation(s)
- Jeanette M Hasse
- Baylor Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, Texas, USA
| | - Shumei Meng
- Division of Endocrinology, Internal Medicine, Baylor University Medical Center, Dallas, Texas, USA
| | - Stephanie Silpe
- Baylor Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, Texas, USA
| | - Bashoo Naziruddin
- Islet Cell Laboratory, Baylor Research Institute, Baylor Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, Texas, USA
| |
Collapse
|
5
|
Yabe SG, Fukuda S, Nishida J, Takeda F, Okochi H. The functional maturity of grafted human pluripotent stem cell derived-islets (hSC-Islets) evaluated by the glycemic set point during blood glucose normalizing process in diabetic mice. Heliyon 2023; 9:e19972. [PMID: 37809993 PMCID: PMC10559575 DOI: 10.1016/j.heliyon.2023.e19972] [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: 05/27/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 10/10/2023] Open
Abstract
Human pluripotent stem cell (hPSCs) derived-pancreatic islets (hSC-islets) are good candidates for cell replacement therapy for patients with diabetes as substitutes for deceased donor-derived islets, because they are pluripotent and have infinite proliferation potential. Grafted hSC-islets ameliorate hyperglycemia in diabetic mice; however, several weeks are needed to normalize the hyperglycemia. These data suggest hSC-islets require maturation, but their maturation process in vivo is not yet fully understood. In this study, we utilized two kinds of streptozotocin (STZ)-induced diabetes model mice by changing the administration timing in order to examine the time course of maturation of hSC-islets and the effects of hyperglycemia on their maturation. We found no hyperglycemia in immune-compromised mice when hSC-islets had been transplanted under their kidney capsules in advance, and STZ was administered 4 weeks after transplantation. Of note, the blood glucose levels of those mice were stably maintained under 100 mg/dl 10 weeks after transplantation; this is lower than the mouse glycemic set point (120-150 mg/dl), suggesting that hSC-islets control blood glucose levels to the human glycemic set point. We confirmed that gene expression of maturation markers of pancreatic beta cells tended to upregulate during 4 weeks after transplantation. Periodical histological analysis revealed that revascularization was observed as early as 1 week after transplantation, but reinnervation in the grafted hSC-islets was not detected at all, even 15 weeks after transplantation. In conclusion, our hSC-islets need at least 4 weeks to mature, and the human glycemic set point is a good index for evaluating ultimate maturity for hSC-islets in vivo.
Collapse
Affiliation(s)
- Shigeharu G. Yabe
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Satsuki Fukuda
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Junko Nishida
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Fujie Takeda
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Hitoshi Okochi
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo, 162-8655, Japan
| |
Collapse
|
6
|
Wang Z, Jiang Z, Lu R, Kou L, Zhao YZ, Yao Q. Formulation strategies to provide oxygen-release to contrast local hypoxia for transplanted islets. Eur J Pharm Biopharm 2023; 187:130-140. [PMID: 37105362 DOI: 10.1016/j.ejpb.2023.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/08/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Islet transplantation refers to the transfusion of healthy islet cells into the diabetic recipients and reconstruction of their endogenous insulin secretion to achieve insulin independence. It is a minimally invasive surgery that holds renewed prospect as a therapeutic method for type 1 diabetes mellitus. However, poor oxygenation in the early post-transplantation period is considered as one of the major causes of islet loss and dysfunction. Due to the metabolism chacteristics, islets required a high supply of oxygen for cell survival while a hypoxia environment would lead to severe islet loss and graft failure. Emerging strategies have been proposed, including providing external oxygen and speeding up revascularization. From the perspective of formulation science, it is feasible and practical to protect transplanted islets by oxygen-release before revascularization as opposed to local hypoxia. In this study, we review the potential formulation strategies that could provide oxygen-release by either delivering external oxygen or triggering localized oxygen generation for transplanted islets.
Collapse
Affiliation(s)
- Zeqing Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhikai Jiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ruijie Lu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Longfa Kou
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Qing Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| |
Collapse
|
7
|
Jeyagaran A, Lu CE, Zbinden A, Birkenfeld AL, Brucker SY, Layland SL. Type 1 diabetes and engineering enhanced islet transplantation. Adv Drug Deliv Rev 2022; 189:114481. [PMID: 36002043 PMCID: PMC9531713 DOI: 10.1016/j.addr.2022.114481] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 01/24/2023]
Abstract
The development of new therapeutic approaches to treat type 1 diabetes mellitus (T1D) relies on the precise understanding and deciphering of insulin-secreting β-cell biology, as well as the mechanisms responsible for their autoimmune destruction. β-cell or islet transplantation is viewed as a potential long-term therapy for the millions of patients with diabetes. To advance the field of insulin-secreting cell transplantation, two main research areas are currently investigated by the scientific community: (1) the identification of the developmental pathways that drive the differentiation of stem cells into insulin-producing cells, providing an inexhaustible source of cells; and (2) transplantation strategies and engineered transplants to provide protection and enhance the functionality of transplanted cells. In this review, we discuss the biology of pancreatic β-cells, pathology of T1D and current state of β-cell differentiation. We give a comprehensive view and discuss the different possibilities to engineer enhanced insulin-secreting cell/islet transplantation from a translational perspective.
Collapse
Affiliation(s)
- Abiramy Jeyagaran
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; NMI Natural and Medical Sciences Institute at the University Tübingen, 72770 Reutlingen, Germany
| | - Chuan-En Lu
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Aline Zbinden
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, German Center for Diabetes Research (DZD e.V.), Munich, Germany
| | - Sara Y Brucker
- Department of Women's Health, Eberhard Karls University, 72076 Tübingen, Germany
| | - Shannon L Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; Department of Women's Health, Eberhard Karls University, 72076 Tübingen, Germany.
| |
Collapse
|
8
|
Opara A, Jost A, Dagogo-Jack S, Opara EC. Islet cell encapsulation - Application in diabetes treatment. Exp Biol Med (Maywood) 2021; 246:2570-2578. [PMID: 34666516 PMCID: PMC8669170 DOI: 10.1177/15353702211040503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In this minireview, we briefly outline the hallmarks of diabetes, the distinction between type 1 and type 2 diabetes, the global incidence of diabetes, and its associated comorbidities. The main goal of the review is to highlight the great potential of encapsulated pancreatic islet transplantation to provide a cure for type 1 diabetes. Following a short overview of the different approaches to islet encapsulation, we provide a summary of the merits and demerits of each approach of the encapsulation technology. We then discuss various attempts to clinical translation with each model of encapsulation as well as the factors that have mitigated the full clinical realization of the promise of the encapsulation technology, the progress that has been made and the challenges that remain to be overcome. In particular, we pay significant attention to the emerging strategies to overcome these challenges. We believe that these strategies to enhance the performance of the encapsulated islet constructs discussed herein provide good platforms for additional work to achieve successful clinical translation of the encapsulated islet technology.
Collapse
Affiliation(s)
- Amoge Opara
- Diabetes Section, Biologics Delivery Technologies, Reno, NV 89502, USA
| | - Alec Jost
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Sam Dagogo-Jack
- Division of Endocrinology, Diabetes & Metabolism, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Emmanuel C Opara
- Diabetes Section, Biologics Delivery Technologies, Reno, NV 89502, USA
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences (SBES), Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
9
|
Tissue Engineering Strategies for Improving Beta Cell Transplantation Outcome. CURRENT TRANSPLANTATION REPORTS 2021. [DOI: 10.1007/s40472-021-00333-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Abstract
Purpose of Review
Beta cell replacement therapy as a form of islet transplantation is a promising alternative therapy with the possibility to make selected patients with type 1 diabetes (T1D) insulin independent. However, this technique faces challenges such as extensive activation of the host immune system post-transplantation, lifelong need for immunosuppression, and the scarcity of islet donor pancreas. Advancement in tissue engineering strategies can improve these challenges and allow for a more widespread application of this therapy. This review will discuss the recent development and clinical translation of tissue engineering strategies in beta cell replacement therapy.
Recent Findings
Tissue engineering offers innovative solutions for producing unlimited glucose responsive cells and fabrication of appropriate devices/scaffolds for transplantation applications. Generation of pancreatic organoids with supporting cells in biocompatible biomaterials is a powerful technique to improve the function of insulin-producing cell clusters. Fabrication of physical barriers such as encapsulation strategies can protect the cells from the host immune system and allow for graft retrieval, although this strategy still faces major challenges to fully restore physiological glucose regulation.
Summary
The three main components of tissue engineering strategies including the generation of stem cell-derived insulin-producing cells and organoids and the possibilities for therapeutic delivery of cell-seeded devices to extra-hepatic sites need to come together in order to provide safe and functional insulin-producing devices for clinical beta cell replacement therapy.
Collapse
|
10
|
Liu Y, Yang M, Cui Y, Yao Y, Liao M, Yuan H, Gong G, Deng S, Zhao G. A novel prevascularized tissue-engineered chamber as a site for allogeneic and xenogeneic islet transplantation to establish a bioartificial pancreas. PLoS One 2020; 15:e0234670. [PMID: 33270650 PMCID: PMC7714105 DOI: 10.1371/journal.pone.0234670] [Citation(s) in RCA: 2] [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: 05/29/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
Although sites for clinical or experimental islet transplantation are well established, pancreatic islet survival and function in these locations remain unsatisfactory. A possible factor that might account for this outcome is local hypoxia caused by the limited blood supply. Here, we modified a prevascularized tissue-engineered chamber (TEC) that facilitated the viability and function of the seeded islets in vivo by providing a microvascular network prior to transplantation. TECs were created, filled with Growth Factor-Matrigel™ (Matrigel™) and then implanted into the groins of mice with streptozotocin-induced diabetes. The degree of microvascularization in each TECs was analyzed by histology, real-time PCR, and Western blotting. Three hundred syngeneic islets were seeded into each chamber on days 0, 14, and 28 post-chamber implantation, and 300, 200, or 100 syngeneic islets were seeded into additional chambers on day 28 post-implantation, respectively. Furthermore, allogeneic or xenogeneic islet transplantation is a potential solution for organ shortage. The feasibility of TECs as transplantation sites for islet allografts or xenografts and treatment with anti-CD45RB and/or anti-CD40L (MR-1) was therefore explored. A highly developed microvascularized network was established in each TEC on day 28 post-implantation. Normalization of blood glucose levels in diabetic mice was negatively correlated with the duration of prevascularization and the number of seeded syngeneic islets. Combined treatment with anti-CD45RB and MR-1 resulted in long-term survival of the grafts following allotransplantation (5/5, 100%) and xenotransplantation (16/20, 80%). Flow cytometry demonstrated that the frequency of CD4+Foxp3-Treg and CD4+IL-4+-Th2 cells increased significantly after tolerogenic xenograft transplantation, while the number of CD4+IFN-γ-Th1 cells decreased. These findings demonstrate that highly developed microvascularized constructs can facilitate the survival of transplanted islets in a TECs, implying its potential application as artificial pancreas in the future.
Collapse
Affiliation(s)
- Yanzhuo Liu
- Department of Gastrointestinal, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Maozhu Yang
- Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, Sichuan Province, China
| | - Yuanyuan Cui
- Department of Gastrointestinal, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Yuanyuan Yao
- Department of Gastrointestinal, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Minxue Liao
- Department of Gastrointestinal, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Hao Yuan
- Department of Gastrointestinal, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Guojin Gong
- Department of Gastrointestinal Surgery, Xi Chang People’s Hospital, Xi Chang, Sichuan Province, China
| | - Shaoping Deng
- Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, Sichuan Province, China
| | - Gaoping Zhao
- Department of Gastrointestinal, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- Organ Transplantation Translational Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, Sichuan Province, China
- * E-mail:
| |
Collapse
|
11
|
Figueiredo H, Figueroa ALC, Garcia A, Fernandez-Ruiz R, Broca C, Wojtusciszyn A, Malpique R, Gasa R, Gomis R. Targeting pancreatic islet PTP1B improves islet graft revascularization and transplant outcomes. Sci Transl Med 2020; 11:11/497/eaar6294. [PMID: 31217339 DOI: 10.1126/scitranslmed.aar6294] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/16/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
Deficient vascularization is a major driver of early islet graft loss and one of the primary reasons for the failure of islet transplantation as a viable treatment for type 1 diabetes. This study identifies the protein tyrosine phosphatase 1B (PTP1B) as a potential modulator of islet graft revascularization. We demonstrate that grafts of pancreatic islets lacking PTP1B exhibit increased revascularization, which is accompanied by improved graft survival and function, and recovery of normoglycemia and glucose tolerance in diabetic mice transplanted with PTP1B-deficient islets. Mechanistically, we show that the absence of PTP1B leads to activation of hypoxia-inducible factor 1α-independent peroxisome proliferator-activated receptor γ coactivator 1α/estrogen-related receptor α signaling and enhanced expression and production of vascular endothelial growth factor A (VEGF-A) by β cells. These observations were reproduced in human islets. Together, these findings reveal that PTP1B regulates islet VEGF-A production and suggest that this phosphatase could be targeted to improve islet transplantation outcomes.
Collapse
Affiliation(s)
- Hugo Figueiredo
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,University of Barcelona, 08036 Barcelona, Spain.,Escuela de Medicina y Ciencias de la Salud, Dept. Medicina Cardiovascular y Metabolómica, Tecnológico de Monterrey, 66278 San Pedro Garza García, Nuevo León, Mexico
| | - Ana Lucia C Figueroa
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,University of Barcelona, 08036 Barcelona, Spain
| | - Ainhoa Garcia
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Rebeca Fernandez-Ruiz
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Christophe Broca
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, 34295 Montpellier, France
| | - Anne Wojtusciszyn
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, 34295 Montpellier, France.,Department of Endocrinology, Diabetes and Nutrition, University Hospital of Montpellier, Lapeyronie Hospital, 34295 Montpellier, France.,Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Rita Malpique
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Rosa Gasa
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Ramon Gomis
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain. .,University of Barcelona, 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.,Universitat Oberta de Catalunya (UOC), 08018 Barcelona, Spain.,Department of Endocrinology and Nutrition, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
12
|
Komatsu H, Gonzalez N, Salgado M, Cook CA, Li J, Rawson J, Omori K, Tai Y, Kandeel F, Mullen Y. A subcutaneous pancreatic islet transplantation platform using a clinically applicable, biodegradable Vicryl mesh scaffold ‐ an experimental study. Transpl Int 2020; 33:806-818. [DOI: 10.1111/tri.13607] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 12/27/2019] [Accepted: 03/17/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Hirotake Komatsu
- Department of Translational Research & Cellular Therapeutics Beckman Research Institute of City of Hope Duarte CA USA
| | - Nelson Gonzalez
- Department of Translational Research & Cellular Therapeutics Beckman Research Institute of City of Hope Duarte CA USA
| | - Mayra Salgado
- Department of Translational Research & Cellular Therapeutics Beckman Research Institute of City of Hope Duarte CA USA
| | - Colin A. Cook
- Department of Electrical Engineering California Institute of Technology Pasadena CA USA
| | - Junfeng Li
- Department of Translational Research & Cellular Therapeutics Beckman Research Institute of City of Hope Duarte CA USA
| | - Jeffrey Rawson
- Department of Translational Research & Cellular Therapeutics Beckman Research Institute of City of Hope Duarte CA USA
| | - Keiko Omori
- Department of Translational Research & Cellular Therapeutics Beckman Research Institute of City of Hope Duarte CA USA
| | - Yu‐Chong Tai
- Department of Electrical Engineering California Institute of Technology Pasadena CA USA
| | - Fouad Kandeel
- Department of Translational Research & Cellular Therapeutics Beckman Research Institute of City of Hope Duarte CA USA
| | - Yoko Mullen
- Department of Translational Research & Cellular Therapeutics Beckman Research Institute of City of Hope Duarte CA USA
| |
Collapse
|
13
|
Addison P, Fatakhova K, Rodriguez Rilo HL. Considerations for an Alternative Site of Islet Cell Transplantation. J Diabetes Sci Technol 2020; 14:338-344. [PMID: 31394934 PMCID: PMC7196852 DOI: 10.1177/1932296819868495] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Islet cell transplantation has been limited most by poor graft survival. Optimizing the site of transplantation could improve clinical outcomes by minimizing required donor cells, increasing graft integration, and simplifying the transplantation and monitoring process. In this article, we review the history and significant human and animal data for clinically relevant sites, including the liver, spleen, and kidney subcapsule, and identify promising new sites for further research. While the liver was the first studied site and has been used the most in clinical practice, the majority of transplanted islets become necrotic. We review the potential causes for graft death, including the instant blood-mediated inflammatory reaction, exposure to immunosuppressive agents, and low oxygen tension. Significant research exists on alternative sites for islet cell transplantation, suggesting a promising future for patients undergoing pancreatectomy.
Collapse
Affiliation(s)
- Poppy Addison
- Donald and Barbara Zucker School of
Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Pancreas Disease Center, Northwell
Health System, Manhasset, NY, USA
| | - Karina Fatakhova
- Donald and Barbara Zucker School of
Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Pancreas Disease Center, Northwell
Health System, Manhasset, NY, USA
| | - Horacio L. Rodriguez Rilo
- Donald and Barbara Zucker School of
Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Pancreas Disease Center, Northwell
Health System, Manhasset, NY, USA
- Horacio L. Rodriguez Rilo, MD, Pancreas
Disease Center, 350 Lakeville Road, New Hyde Park, NY 11042, USA.
| |
Collapse
|
14
|
Delaune V, Toso C, Kahler-Quesada A, Slits F, Gex Q, Kaya G, Lavallard V, Orci LA, Peloso A, Lacotte S. Antibody-induced NKG2D blockade in a rat model of intraportal islet transplantation leads to a deleterious reaction. Transpl Int 2020; 33:675-688. [PMID: 32003082 DOI: 10.1111/tri.13589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/04/2019] [Accepted: 01/28/2020] [Indexed: 11/30/2022]
Abstract
Intraportal islet transplantation is plagued by an acute destruction of transplanted islets. Amongst the first responders, NK cells and macrophages harbour an activating receptor, NKG2D, recognizing ligands expressed by stressed cells. We aimed to determine whether islet NKG2D ligand expression increases with culture time, and to analyse the impact of antibody-induced NKG2D blockade in islet transplantation. NKG2D-ligand expression was analysed in rat and human islets. Syngeneic marginal mass intraportal islet transplantations were performed in rats: control group, recipients transplanted with NKG2D-recombinant-treated islets (recombinant group), and recipients treated with a mouse anti-rat anti-NKG2D antibody and transplanted with recombinant-treated islets (antibody-recombinant group). Islets demonstrated increased gene expression of NKG2D ligands with culture time. Blockade of NKG2D on NK cells decreased in vitro cytotoxicity against islets. Recipients from the control and recombinant groups showed similar metabolic results; conversely, treatment with the antibody resulted in lower diabetes reversal. The antibody depleted circulating and liver NK cells in recipients, who displayed increased macrophage infiltration of recipient origin around the transplanted islets. In vitro blockade of NKG2D ligands had no impact on early graft function. Systemic treatment of recipients with an anti-NKG2D antibody was deleterious to the islet graft, possibly through an antibody-dependent cell-mediated cytotoxicity reaction.
Collapse
Affiliation(s)
- Vaihere Delaune
- Divisions of Abdominal and Transplantation Surgery, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland.,Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christian Toso
- Divisions of Abdominal and Transplantation Surgery, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland.,Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Arianna Kahler-Quesada
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Florence Slits
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Quentin Gex
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gürkan Kaya
- Division of Dermatology and Venereology, Department of Internal Medicine Specialties, Geneva University Hospitals, Geneva, Switzerland
| | - Vanessa Lavallard
- Cell Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Lorenzo Annibale Orci
- Divisions of Abdominal and Transplantation Surgery, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland.,Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Andrea Peloso
- Divisions of Abdominal and Transplantation Surgery, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland.,Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Stéphanie Lacotte
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
15
|
Hadavi E, Leijten J, Engelse M, de Koning E, Jonkheijm P, Karperien M, van Apeldoorn A. Microwell Scaffolds Using Collagen-IV and Laminin-111 Lead to Improved Insulin Secretion of Human Islets. Tissue Eng Part C Methods 2020; 25:71-81. [PMID: 30632461 DOI: 10.1089/ten.tec.2018.0336] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IMPACT STATEMENT This research deals with finding a proper bioengineering strategy to improve the outcome of islets transplantation for treatment of type 1 diabetes. It is focused on the mimicking of islet extracellular matrix niche in microwell islet delivery devices to improve their endocrine function.
Collapse
Affiliation(s)
- Elahe Hadavi
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Jeroen Leijten
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Marten Engelse
- 2 Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eelco de Koning
- 2 Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,3 Hubrecht Institute, Utrecht, The Netherlands
| | - Pascal Jonkheijm
- 4 Bioinspired Molecular Engineering Laboratory and Molecular Nanofabrication Group, MESA+ Institute for Nanotechnology, University of Twente, Enschede, The Netherlands
| | - Marcel Karperien
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Aart van Apeldoorn
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Enschede, The Netherlands.,5 Department of Complex Tissue Regeneration, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
16
|
Bowers DT, Song W, Wang LH, Ma M. Engineering the vasculature for islet transplantation. Acta Biomater 2019; 95:131-151. [PMID: 31128322 PMCID: PMC6824722 DOI: 10.1016/j.actbio.2019.05.051] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/13/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022]
Abstract
The microvasculature in the pancreatic islet is highly specialized for glucose sensing and insulin secretion. Although pancreatic islet transplantation is a potentially life-changing treatment for patients with insulin-dependent diabetes, a lack of blood perfusion reduces viability and function of newly transplanted tissues. Functional vasculature around an implant is not only necessary for the supply of oxygen and nutrients but also required for rapid insulin release kinetics and removal of metabolic waste. Inadequate vascularization is particularly a challenge in islet encapsulation. Selectively permeable membranes increase the barrier to diffusion and often elicit a foreign body reaction including a fibrotic capsule that is not well vascularized. Therefore, approaches that aid in the rapid formation of a mature and robust vasculature in close proximity to the transplanted cells are crucial for successful islet transplantation or other cellular therapies. In this paper, we review various strategies to engineer vasculature for islet transplantation. We consider properties of materials (both synthetic and naturally derived), prevascularization, local release of proangiogenic factors, and co-transplantation of vascular cells that have all been harnessed to increase vasculature. We then discuss the various other challenges in engineering mature, long-term functional and clinically viable vasculature as well as some emerging technologies developed to address them. The benefits of physiological glucose control for patients and the healthcare system demand vigorous pursuit of solutions to cell transplant challenges. STATEMENT OF SIGNIFICANCE: Insulin-dependent diabetes affects more than 1.25 million people in the United States alone. Pancreatic islets secrete insulin and other endocrine hormones that control glucose to normal levels. During preparation for transplantation, the specialized islet blood vessel supply is lost. Furthermore, in the case of cell encapsulation, cells are protected within a device, further limiting delivery of nutrients and absorption of hormones. To overcome these issues, this review considers methods to rapidly vascularize sites and implants through material properties, pre-vascularization, delivery of growth factors, or co-transplantation of vessel supporting cells. Other challenges and emerging technologies are also discussed. Proper vascular growth is a significant component of successful islet transplantation, a treatment that can provide life-changing benefits to patients.
Collapse
Affiliation(s)
- Daniel T Bowers
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Wei Song
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Long-Hai Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
17
|
Mouré A, Bacou E, Bosch S, Jegou D, Salama A, Riochet D, Gauthier O, Blancho G, Soulillou J, Poncelet D, Olmos E, Bach J, Mosser M. Extracellular hemoglobin combined with an O
2
‐generating material overcomes O
2
limitation in the bioartificial pancreas. Biotechnol Bioeng 2019; 116:1176-1189. [DOI: 10.1002/bit.26913] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/05/2018] [Accepted: 12/26/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Anne Mouré
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| | - Elodie Bacou
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| | - Steffi Bosch
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| | - Dominique Jegou
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| | - Apolline Salama
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
- Centre de Recherche en Transplantation et Immunologie UMR 1064INSERM, Université de NantesNantes France
| | - David Riochet
- Service de Pédiatrie des Maladies ChroniquesCHU de NantesNantes France
| | | | - Gilles Blancho
- Centre de Recherche en Transplantation et Immunologie UMR 1064INSERM, Université de NantesNantes France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU NantesNantes France
| | - Jean‐Paul Soulillou
- Centre de Recherche en Transplantation et Immunologie UMR 1064INSERM, Université de NantesNantes France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU NantesNantes France
| | - Denis Poncelet
- Department of Process Engineering for Environment and Food Laboratory (GEPEA)UMR CNRS 6144, OnirisNantes France
| | - Eric Olmos
- Laboratoire Réactions et Génie des Procédés (LRGP)Université de Lorraine, CNRSNancy France
| | - Jean‐Marie Bach
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| | - Mathilde Mosser
- Immuno-Endocrinology Unit (IECM), Oniris, INRA, Université Bretagne LoireNantes France
| |
Collapse
|
18
|
Komatsu H, Cook CA, Gonzalez N, Medrano L, Salgado M, Sui F, Li J, Kandeel F, Mullen Y, Tai YC. Oxygen transporter for the hypoxic transplantation site. Biofabrication 2018; 11:015011. [PMID: 30524058 PMCID: PMC9851375 DOI: 10.1088/1758-5090/aaf2f0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Cell transplantation is a promising treatment for complementing lost function by replacing new cells with a desired function, e.g. pancreatic islet transplantation for diabetics. To prevent cell obliteration, oxygen supply is critical after transplantation, especially until the graft is sufficiently re-vascularized. To supply oxygen during this period, we developed a chemical-/electrical-free implantable oxygen transporter that delivers oxygen to the hypoxic graft site from ambient air by diffusion potential. This device is simply structured using a biocompatible silicone-based body that holds islets, connected to a tube that opens outside the body. In computational simulations, the oxygen transporter increased the oxygen level to >120 mmHg within grafts; in contrast, a control device that did not transport oxygen showed <6.5 mmHg. In vitro experiments demonstrated similar results. To test the effectiveness of the oxygen transporter in vivo, we transplanted pancreatic islets, which are susceptible to hypoxia, subcutaneously into diabetic rats. Islets transplanted using the oxygen transporter showed improved graft viability and cellular function over the control device. These results indicate that our oxygen transporter, which is safe and easily fabricated, effectively supplies oxygen locally. Such a device would be suitable for multiple clinical applications, including cell transplantations that require changing a hypoxic microenvironment into an oxygen-rich site.
Collapse
Affiliation(s)
- Hirotake Komatsu
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA.,Corresponding author: Hirotake Komatsu,
| | - Colin A. Cook
- Department of Electrical Engineering, California Institute of Technology, 1200 E. California Blvd., MC 136-93, Pasadena, CA 91125, USA
| | - Nelson Gonzalez
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Leonard Medrano
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Mayra Salgado
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Feng Sui
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Junfeng Li
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Fouad Kandeel
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Yoko Mullen
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | - Yu-Chong Tai
- Department of Electrical Engineering, California Institute of Technology, 1200 E. California Blvd., MC 136-93, Pasadena, CA 91125, USA
| |
Collapse
|
19
|
McQuilling JP, Sittadjody S, Pendergraft S, Farney AC, Opara EC. Applications of particulate oxygen-generating substances (POGS) in the bioartificial pancreas. Biomater Sci 2018; 5:2437-2447. [PMID: 29051963 DOI: 10.1039/c7bm00790f] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Type-1 Diabetes (T1D) is a devastating autoimmune disorder which results in the destruction of beta cells within the pancreas. A promising treatment strategy for T1D is the replacement of the lost beta cell mass through implantation of immune-isolated microencapsulated islets referred to as the bioartificial pancreas. The goal of this approach is to restore blood glucose regulation and prevent the long-term comorbidities of T1D without the need for immunosuppressants. A major requirement in the quest to achieve this goal is to address the oxygen needs of islet cells. Islets are highly metabolically active and require a significant amount of oxygen for normal function. During the process of isolation, microencapsulation, and processing prior to transplantation, the islets' oxygen supply is disrupted, and a large amount of islet cells are therefore lost due to extended hypoxia, thus creating a major barrier to clinical success with this treatment. In this work, we have investigated the oxygen generating compounds, sodium percarbonate (SPO) and calcium peroxide (CPO) as potential supplemental oxygen sources for islets during isolation and encapsulation before and immediately after transplantation. First, SPO particles were used as an oxygen source for islets during isolation. Secondly, silicone films containing SPO were used to provide supplemental oxygen to islets for up to 4 days in culture. Finally, CPO was used as an oxygen source for encapsulated cells by co-encapsulating CPO particles with islets in permselective alginate microspheres. These studies provide an important proof of concept for the utilization of these oxygen generating materials to prevent beta cell death caused by hypoxia.
Collapse
Affiliation(s)
- John P McQuilling
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | | | | | | | | |
Collapse
|
20
|
Komatsu H, Rawson J, Barriga A, Gonzalez N, Mendez D, Li J, Omori K, Kandeel F, Mullen Y. Posttransplant oxygen inhalation improves the outcome of subcutaneous islet transplantation: A promising clinical alternative to the conventional intrahepatic site. Am J Transplant 2018; 18:832-842. [PMID: 28898528 DOI: 10.1111/ajt.14497] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/29/2017] [Accepted: 09/06/2017] [Indexed: 01/25/2023]
Abstract
Subcutaneous tissue is a promising site for islet transplantation, due to its large area and accessibility, which allows minimally invasive procedures for transplantation, graft monitoring, and removal of malignancies as needed. However, relative to the conventional intrahepatic transplantation site, the subcutaneous site requires a large number of islets to achieve engraftment success and diabetes reversal, due to hypoxia and low vascularity. We report that the efficiency of subcutaneous islet transplantation in a Lewis rat model is significantly improved by treating recipients with inhaled 50% oxygen, in conjunction with prevascularization of the graft bed by agarose-basic fibroblast growth factor. Administration of 50% oxygen increased oxygen tension in the subcutaneous site to 140 mm Hg, compared to 45 mm Hg under ambient air. In vitro, islets cultured under 140 mm Hg oxygen showed reduced central necrosis and increased insulin release, compared to those maintained in 45 mm Hg oxygen. Six hundred syngeneic islets subcutaneously transplanted into the prevascularized graft bed reversed diabetes when combined with postoperative 50% oxygen inhalation for 3 days, a number comparable to that required for intrahepatic transplantation; in the absence of oxygen treatment, diabetes was not reversed. Thus, we show oxygen inhalation to be a simple and promising approach to successfully establishing subcutaneous islet transplantation.
Collapse
Affiliation(s)
- H Komatsu
- Division of Developmental and Translational Diabetes and Endocrinology Research, Department of Diabetes and Metabolic Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - J Rawson
- Division of Developmental and Translational Diabetes and Endocrinology Research, Department of Diabetes and Metabolic Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - A Barriga
- Division of Developmental and Translational Diabetes and Endocrinology Research, Department of Diabetes and Metabolic Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - N Gonzalez
- Division of Developmental and Translational Diabetes and Endocrinology Research, Department of Diabetes and Metabolic Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - D Mendez
- Division of Developmental and Translational Diabetes and Endocrinology Research, Department of Diabetes and Metabolic Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - J Li
- Division of Developmental and Translational Diabetes and Endocrinology Research, Department of Diabetes and Metabolic Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - K Omori
- Division of Developmental and Translational Diabetes and Endocrinology Research, Department of Diabetes and Metabolic Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - F Kandeel
- Division of Developmental and Translational Diabetes and Endocrinology Research, Department of Diabetes and Metabolic Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Y Mullen
- Division of Developmental and Translational Diabetes and Endocrinology Research, Department of Diabetes and Metabolic Research, Beckman Research Institute of City of Hope, Duarte, CA, USA
| |
Collapse
|
21
|
Song L, Sun Z, Kim DS, Gou W, Strange C, Dong H, Cui W, Gilkeson G, Morgan KA, Adams DB, Wang H. Adipose stem cells from chronic pancreatitis patients improve mouse and human islet survival and function. Stem Cell Res Ther 2017; 8:192. [PMID: 28854965 PMCID: PMC5577777 DOI: 10.1186/s13287-017-0627-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/09/2017] [Accepted: 07/03/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Chronic pancreatitis has surgical options including total pancreatectomy to control pain. To avoid surgical diabetes, the explanted pancreas can have islets harvested and transplanted. Immediately following total pancreatectomy with islet autotransplantation (TP-IAT), many islet cells die due to isolation and transplantation stresses. The percentage of patients remaining insulin free after TP-IAT is therefore low. We determined whether cotransplantation of adipose-derived mesenchymal stem cells (ASCs) from chronic pancreatitis patients (CP-ASCs) would protect islets after transplantation. METHODS In a marginal mass islet transplantation model, islets from C57BL/6 mice were cotransplanted with CP-ASCs into syngeneic streptozotocin-treated diabetic mice. Treatment response was defined by the percentage of recipients reaching normoglycemia, and by the area under the curve for glucose and c-peptide in a glucose tolerance test. Macrophage infiltration, β-cell apoptosis, and islet graft vasculature were measured in transplanted islet grafts by immunohistochemistry. mRNA expression profiling of 84 apoptosis-related genes in islet grafts transplanted alone or with CP-ASCs was measured by the RT2 Profiler™ Apoptosis PCR Array. The impact of insulin-like growth factor-1 (IGF-1) on islet apoptosis was determined in islets stimulated with cytokines (IL-1β and IFN-γ) in the presence and absence of CP-ASC conditioned medium. RESULTS CP-ASC-treated mice were more often normoglycemic compared to mice receiving islets alone. ASC cotransplantation reduced macrophage infiltration, β-cell death, suppressed expression of TNF-α and Bcl-2 modifying factor (BMF), and upregulated expressions of IGF-1 and TNF Receptor Superfamily Member 11b (TNFRSF11B) in islet grafts. Islets cultured in conditioned medium from CP-ASCs showed reduced cell death. This protective effect was diminished when IGF-1 was blocked in the conditioned medium by the anti-IGF-1 antibody. CONCLUSION Cotransplantation of islets with ASCs from the adipose of chronic pancreatitis patients improved islet survival and islet function after transplantation. The effects are in part mediated by paracrine secretion of IGF-1, suppression of inflammation, and promotion of angiogenesis. ASCs from chronic pancreatitis patients have the potential to be used as a synergistic therapy to enhance the efficacy of islet transplantation following pancreatectomy.
Collapse
Affiliation(s)
- Lili Song
- Department of Surgery, Medical University of South Carolina, BSB 641, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Zhen Sun
- Department of Surgery, Medical University of South Carolina, BSB 641, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Do-Sung Kim
- Department of Surgery, Medical University of South Carolina, BSB 641, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Wenyu Gou
- Department of Surgery, Medical University of South Carolina, BSB 641, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Charlie Strange
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Huansheng Dong
- Department of Surgery, Medical University of South Carolina, BSB 641, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Wanxing Cui
- Medstar Georgetown University Hospital, Washington, DC, USA
| | - Gary Gilkeson
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Katherine A Morgan
- Department of Surgery, Medical University of South Carolina, BSB 641, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - David B Adams
- Department of Surgery, Medical University of South Carolina, BSB 641, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, BSB 641, 173 Ashley Avenue, Charleston, SC, 29425, USA. .,Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
22
|
Juang JH, Hsu BRS, Kuo CH, Ueng SWN. Beneficial Effects of Hyperbaric Oxygen Therapy on Islet Transplantation. Cell Transplant 2017; 11:95-101. [PMID: 28853948 DOI: 10.3727/096020198389825] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We envisage that hyperbaric oxygen (HBO) would ameliorate islet anoxia, preventing early graft failure. Thus, treatment of HBO to diabetic recipients should improve the outcome of islet transplantation. We tested this hypothesis by syngeneically transplanting insufficient number of islets (150 islets) into streptozotocin-diabetic C57BL/6 mice, each followed by HBO (2.4 ATA, 100% O2) therapy for 1.5 h from day 0 to 28, once daily (group A) or twice daily (group B), or from day 5 to 28, once daily (group C) or twice daily (group D), 6 days/week. Recipients without HBO treatment served as controls. At day 28 after transplantation, groups B, C, and D gained weight and had lower blood glucose compared with their baseline values. In addition, groups B and D had higher insulin content of the graft than the controls. To determine the optimal timing of HBO therapy, groups B and D were compared with recipients treated with HBO twice daily, 6 days/week, from day -14 to 0 (group E) and from day -14 to 28 (group F). At day 28 after transplantation, groups B, D, E, and F had significantly lower blood glucose than their individual baseline values and higher insulin content of the graft than controls. But only group F had more β-cell mass of the graft than controls. These findings lend credence to the expectation that peritransplant application of adequate frequency of HBO to diabetic recipients would enhance the performance and growth of the islet graft, resulting in an improvement of the outcome of the transplantation.
Collapse
Affiliation(s)
- Jyuhn-Huarng Juang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Brend Ray-Sea Hsu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chien-Hung Kuo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Steve Wen-Neng Ueng
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
23
|
Wang W, Gu Y, Miyamoto M, Hori H, Nagata N, Balamurugan AN, Touma M, Sakurai T, Inoue K. Effect of Basic Fibroblast Growth Factor on Insulin Secretion from Microencapsulated Pancreatic Islets: An In Vitro Study. Cell Transplant 2017. [DOI: 10.3727/000000001783986521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Microencapsulation of pancreatic islets represents a potentially effective method to prevent graft rejection in allotransplantation or xenotransplantation without the need of immunosuppression. Adequate insulin secretion and glucose responsiveness of microencapsulated pancreatic islets has been regarded as a prerequisite for successful transplantation. The microencapsulated pancreatic islets were respectively cultured in bFGF+ RPMI-1640 medium (bFGF+) or bFGF- RPMI-1640 medium (bFGF-) for 21 days. The functional activities of microencapsulated pancreatic islets were assessed by measuring basal insulin secretion and stimulated insulin release at different time points. The results revealed that microencapsulated pancreatic islets in the presence of bFGF demonstrated an increase in basal insulin secretion. Furthermore, microencapsulated pancreatic islets in the presence of bFGF demonstrated a marked stimulated insulin release and relative stability of stimulation indices (SI). The results in the perifusion study showed that microencapsulated pancreatic islets in the presence of bFGF maintained good glucose responsiveness over the course of culture period as well. These results indicate that bFGF has a beneficial effect on insulin secretion from microencapsulated pancreatic islets during in vitro culture. New strategies for preserving and improving function of microencapsulated pancreatic islets prior to transplantation may be developed by application of growth factors or other factors.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Yuanjun Gu
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Masaaki Miyamoto
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Hiroshi Hori
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Natsuki Nagata
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - A. N. Balamurugan
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Maki Touma
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Tomonori Sakurai
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Kazutomo Inoue
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
24
|
Carlsson PO, Mattsson G. Oxygen Tension and Blood Flow in Relation to Revascularization in Transplanted Adult and Fetal Rat Pancreatic Islets. Cell Transplant 2017. [DOI: 10.3727/000000002783985251] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We have previously recorded a decreased oxygen tension and blood flow in syngeneically transplanted rat pancreatic islets. The present study related measurements of oxygen tension and blood flow to the vascular density in such grafts implanted beneath the renal capsule. We also evaluated whether transplanted fetal islets are better revascularized than adult islets, and if the degree of revascularization is directly related to the islet vascular endothelial growth factor (VEGF) production. Tissue pO2 was measured using Clark microelectrodes, whereas islet graft blood flow was measured with laser-Doppler flowmetry. The vascular density of endogenous and transplanted islets was quantified in histological specimens stained with the lectin Bandeiraea simplicifolia (BS-1). Tissue pO2 in the transplanted adult and fetal islet grafts was similar and markedly lower than in the endogenous islets. The blood perfusion of both the adult and fetal islet grafts was 60–65% of that in the renal cortex. Administration of d-glucose did not affect tissue pO2 in either the endogenous or transplanted islets, nor graft blood perfusion. The number of capillaries found in the transplanted adult and fetal islets was similar and markedly lower than in endogenous islets. However, in the connective tissue stroma, which constituted ~20% of all islet grafts, the vascular density was higher than in the corresponding endocrine parts of these grafts. Incubated adult islets released higher amounts of VEGF than fetal islets. In conclusion, the previously described low oxygen tension of syngeneically transplanted adult rat islets is related to a low vascular density. Similar low oxygen tension and vascular density are seen in grafted fetal islets. The amount of VEGF production does not correlate to the degree of revascularization of the grafts.
Collapse
Affiliation(s)
- Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Göran Mattsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
25
|
Suijkerbuijk SJE, van Rheenen J. From good to bad: Intravital imaging of the hijack of physiological processes by cancer cells. Dev Biol 2017; 428:328-337. [PMID: 28473106 DOI: 10.1016/j.ydbio.2017.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 12/23/2022]
Abstract
Homeostasis of tissues is tightly regulated at the cellular, tissue and organismal level. Interestingly, tumor cells have found ways to hijack many of these physiological processes at all the different levels. Here we review how intravital microscopy techniques have provided new insights into our understanding of tissue homeostasis and cancer progression. In addition, we highlight the different strategies that tumor cells have adopted to use these physiological processes for their own benefit. We describe how visualization of these dynamic processes in living mice has broadened to our view on cancer initiation and progression.
Collapse
Affiliation(s)
- Saskia J E Suijkerbuijk
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands
| | - Jacco van Rheenen
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
26
|
Delaune V, Berney T, Lacotte S, Toso C. Intraportal islet transplantation: the impact of the liver microenvironment. Transpl Int 2017; 30:227-238. [DOI: 10.1111/tri.12919] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 11/09/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Vaihere Delaune
- Hepatology and Transplantation Laboratory; Department of Surgery; Faculty of Medicine; University of Geneva; Geneva Switzerland
- Divisions of Abdominal and Transplantation Surgery; Department of Surgery; Geneva University Hospitals; Geneva Switzerland
| | - Thierry Berney
- Divisions of Abdominal and Transplantation Surgery; Department of Surgery; Geneva University Hospitals; Geneva Switzerland
- Cell Transplantation Laboratory; Department of Surgery; Faculty of Medicine; University of Geneva; Geneva Switzerland
| | - Stéphanie Lacotte
- Hepatology and Transplantation Laboratory; Department of Surgery; Faculty of Medicine; University of Geneva; Geneva Switzerland
| | - Christian Toso
- Hepatology and Transplantation Laboratory; Department of Surgery; Faculty of Medicine; University of Geneva; Geneva Switzerland
- Divisions of Abdominal and Transplantation Surgery; Department of Surgery; Geneva University Hospitals; Geneva Switzerland
| |
Collapse
|
27
|
Berclaz C, Szlag D, Nguyen D, Extermann J, Bouwens A, Marchand PJ, Nilsson J, Schmidt-Christensen A, Holmberg D, Grapin-Botton A, Lasser T. Label-free fast 3D coherent imaging reveals pancreatic islet micro-vascularization and dynamic blood flow. BIOMEDICAL OPTICS EXPRESS 2016; 7:4569-4580. [PMID: 27895996 PMCID: PMC5119596 DOI: 10.1364/boe.7.004569] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/16/2016] [Accepted: 10/03/2016] [Indexed: 05/14/2023]
Abstract
In diabetes, pancreatic β-cells play a key role. These cells are clustered within structures called islets of Langerhans inside the pancreas and produce insulin, which is directly secreted into the blood stream. The dense vascularization of islets of Langerhans is critical for maintaining a proper regulation of blood glucose homeostasis and is known to be affected from the early stage of diabetes. The deep localization of these islets inside the pancreas in the abdominal cavity renders their in vivo visualization a challenging task. A fast label-free imaging method with high spatial resolution is required to study the vascular network of islets of Langerhans. Based on these requirements, we developed a label-free and three-dimensional imaging method for observing islets of Langerhans using extended-focus Fourier domain Optical Coherence Microscopy (xfOCM). In addition to structural imaging, this system provides three-dimensional vascular network imaging and dynamic blood flow information within islets of Langerhans. We propose our method to deepen the understanding of the interconnection between diabetes and the evolution of the islet vascular network.
Collapse
Affiliation(s)
- Corinne Berclaz
- Laboratoire d’Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne (EPFL),1015 Lausanne,
Switzerland
| | - Daniel Szlag
- Laboratoire d’Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne (EPFL),1015 Lausanne,
Switzerland
| | - David Nguyen
- Laboratoire d’Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne (EPFL),1015 Lausanne,
Switzerland
| | - Jérôme Extermann
- Laboratoire d’Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne (EPFL),1015 Lausanne,
Switzerland
- Hepia, University of Applied Science of Western Switzerland, 1202 Genève,
Switzerland
| | - Arno Bouwens
- Laboratoire d’Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne (EPFL),1015 Lausanne,
Switzerland
| | - Paul J. Marchand
- Laboratoire d’Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne (EPFL),1015 Lausanne,
Switzerland
| | | | | | - Dan Holmberg
- EMV Immunology, Lund University, 22100 Lund,
Sweden
| | | | - Theo Lasser
- Laboratoire d’Optique Biomédicale, Ecole Polytechnique Fédérale de Lausanne (EPFL),1015 Lausanne,
Switzerland
| |
Collapse
|
28
|
Shahbazov R, Yoshimatsu G, Haque WZ, Khan OS, Saracino G, Lawrence MC, Kim PT, Onaca N, Naziruddin B, Levy MF. Clinical effectiveness of a pylorus-preserving procedure on total pancreatectomy with islet autotransplantation. Am J Surg 2016; 213:1065-1071. [PMID: 27760705 DOI: 10.1016/j.amjsurg.2016.09.051] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/07/2016] [Accepted: 09/12/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND The impact of pylorus preserving procedures (PP) on total pancreatectomy with islet autotransplantation (TPIAT) has not been examined. This study aimed to investigate the clinical impact of the PP on TPIAT. METHODS The Baylor Simmons Transplant Institute database was queried to identify seventy-three patients who underwent TPIAT from 2006 to 2014. All patients were investigated in postoperative complications, long-term nutritional status, and graft function. RESULTS Patients with PP did not face worse outcomes in terms of delayed gastric emptying and length of hospital stay. Also, nutritional status and metabolic outcome, such as body weight, serum albumin level, serum vitamin level, HbA1c level, graft survival rate and insulin independent rate, were similar between both groups. CONCLUSIONS Clinical results including the graft function indicated that patients undergoing TPIAT with PP did not amplify surgical complications such as delayed gastric emptying and showed no significant advantage of nutrition and metabolic outcome.
Collapse
Affiliation(s)
- Rauf Shahbazov
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | | | - Waqas Z Haque
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX, USA
| | - Omar S Khan
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX, USA
| | - Giovanna Saracino
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX, USA
| | | | - Peter T Kim
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Nicholas Onaca
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Bashoo Naziruddin
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX, USA.
| | - Marlon F Levy
- Transplant Division, Department of Surgery, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| |
Collapse
|
29
|
Christoffersson G, von Herrath MG. A Deeper Look into Type 1 Diabetes - Imaging Immune Responses during Onset of Disease. Front Immunol 2016; 7:313. [PMID: 27574523 PMCID: PMC4983548 DOI: 10.3389/fimmu.2016.00313] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 08/02/2016] [Indexed: 12/28/2022] Open
Abstract
Cytotoxic T lymphocytes execute the killing of insulin-producing beta cells during onset of type 1 diabetes mellitus (T1D). The research community has come far in dissecting the major events in the development of this disease, but still the trigger and high-resolved information of the immunological events leading up to beta cell loss are missing. During the past decades, intravital imaging of immune responses has led to significant scientific breakthroughs in diverse models of disease, including T1D. Dynamic imaging of immune cells at the pancreatic islets during T1D onset has been made possible through the development of both advanced microscopes, and animal models that allow long-term immobilization of the pancreas. The use of these modalities has revealed a milling microenvironment at the pancreatic islets during disease onset with a plethora of active players. Clues to answering the remaining questions in this disease may lie in intravital imaging, including how key immune cells traffic to and from the pancreas, and how cells interact at this target tissue. This review highlights and discusses recent studies, models, and techniques focused to understand the immune responses during T1D onset through intravital imaging.
Collapse
Affiliation(s)
- Gustaf Christoffersson
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology , La Jolla, CA , USA
| | - Matthias G von Herrath
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA; Novo Nordisk Diabetes Research and Development Center, Seattle, WA, USA
| |
Collapse
|
30
|
Lablanche S, Cottet-Rousselle C, Argaud L, Laporte C, Lamarche F, Richard MJ, Berney T, Benhamou PY, Fontaine E. Respective effects of oxygen and energy substrate deprivation on beta cell viability. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:629-39. [PMID: 25868875 DOI: 10.1016/j.bbabio.2015.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 03/30/2015] [Accepted: 04/05/2015] [Indexed: 12/25/2022]
Abstract
Deficit in oxygen and energetic substrates delivery is a key factor in islet loss during islet transplantation. Permeability transition pore (PTP) is a mitochondrial channel involved in cell death. We have studied the respective effects of oxygen and energy substrate deprivation on beta cell viability as well as the involvement of oxidative stress and PTP opening. Energy substrate deprivation for 1h followed by incubation in normal conditions led to a cyclosporin A (CsA)-sensitive-PTP-opening in INS-1 cells and human islets. Such a procedure dramatically decreased INS-1 cells viability except when transient removal of energy substrates was performed in anoxia, in the presence of antioxidant N-acetylcysteine (NAC) or when CsA or metformin inhibited PTP opening. Superoxide production increased during removal of energy substrates and increased again when normal energy substrates were restored. NAC, anoxia or metformin prevented the two phases of oxidative stress while CsA prevented the second one only. Hypoxia or anoxia alone did not induce oxidative stress, PTP opening or cell death. In conclusion, energy substrate deprivation leads to an oxidative stress followed by PTP opening, triggering beta cell death. Pharmacological prevention of PTP opening during islet transplantation may be a suitable option to improve islet survival and graft success.
Collapse
Affiliation(s)
- Sandrine Lablanche
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France; Department of Endocrinology, Grenoble University Hospital, Grenoble F-38043, France.
| | - Cécile Cottet-Rousselle
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France
| | | | - Camille Laporte
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France
| | - Frédéric Lamarche
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France
| | - Marie-Jeanne Richard
- Cellular Therapy Unit, EFS Rhône-Alpes, Grenoble University Hospital, Grenoble, France
| | - Thierry Berney
- Cell Isolation and Transplant Center, University of Geneva, Level R, 1 rue Michel Servet, Geneva 4, CH-1211, Switzerland
| | - Pierre-Yves Benhamou
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France; Department of Endocrinology, Grenoble University Hospital, Grenoble F-38043, France
| | - Eric Fontaine
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France; Department of Endocrinology, Grenoble University Hospital, Grenoble F-38043, France
| |
Collapse
|
31
|
Abstract
BACKGROUND The islet size distribution in a preparation may contribute to islet transplant outcomes. At the same islet equivalent (IE) dose, larger islets may exhibit poorer therapeutic value and this may be because of oxygen diffusion limitations that worsen in proportion to islet size. METHODS To test this hypothesis, we studied the impact of islet size index (ISI) and other islet product characteristics on outcomes after islet autotransplant (IAT) in recipients receiving a marginal islet dose (2000-4999 IEs per kg body weight) from January 1, 2009 to June 11, 2012, at the University of Minnesota (n=58). ISI was defined as the number of IE divided by the number of islet particles (IPs) in a preparation; an ISI less than 1 indicates a mean islet diameter that is less than 150 μm. The primary post-IAT outcome was 6-month insulin use status. RESULTS Logistic regression analysis indicate that IPs/kg (P=0.001), IEs/kg (P=0.019), total IPs transplanted (P=0.040), and ISI (P=0.074) were most strongly correlated with the primary outcome. The ISI (mean±standard error) was lower for recipients achieving insulin independence at 6 months (0.71±0.05) versus those partially (0.83±0.05) or completely (1.00±0.07) insulin dependent. The combination of islet dose (expressed as units IPs/kg) and ISI exhibited a sensitivity of 75% and specificity of 74% in predicting insulin independence in this population of patients. CONCLUSION Islet autotransplant recipients of a marginal islet doses were more likely to achieve insulin independence when transplanted with a greater number of smaller islets.
Collapse
|
32
|
Zuellig RA, Cavallari G, Gerber P, Tschopp O, Spinas GA, Moritz W, Lehmann R. Improved physiological properties of gravity-enforced reassembled rat and human pancreatic pseudo-islets. J Tissue Eng Regen Med 2014; 11:109-120. [PMID: 24737702 DOI: 10.1002/term.1891] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 12/20/2013] [Accepted: 02/26/2014] [Indexed: 01/01/2023]
Abstract
Previously we demonstrated the superiority of small islets vs large islets in terms of function and survival after transplantation, and we generated reaggregated rat islets (pseudo-islets) of standardized small dimensions by the hanging-drop culture method (HDCM). The aim of this study was to generate human pseudo-islets by HDCM and to evaluate and compare the physiological properties of rat and human pseudo-islets. Isolated rat and human islets were dissociated into single cells and incubated for 6-14 days by HDCM. Newly formed pseudo-islets were analysed for dimensions, morphology, glucose-stimulated insulin secretion (GSIS) and total insulin content. The morphology of reaggregated human islets was similar to that of native islets, while rat pseudo-islets had a reduced content of α and δ cells. GSIS of small rat and human pseudo-islets (250 cells) was increased up to 4.0-fold (p < 0.01) and 2.5-fold (p < 0.001), respectively, when compared to their native counterparts. Human pseudo-islets showed a more pronounced first-phase insulin secretion as compared to intact islets. GSIS was inversely correlated to islet size, and small islets (250 cells) contained up to six-fold more insulin/cell than large islets (1500 cells). Tissue loss with this new technology could be reduced to 49.2 ± 1.5% in rat islets, as compared to the starting amount. With HDCM, pseudo-islets of standardized size with similar cellular composition and improved biological function can be generated, which compensates for tissue loss during production. Transplantation of small pseudo-islets may represent an attractive strategy to improve graft survival and function, due to better oxygen and nutrient supply during the phase of revascularization. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- R A Zuellig
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - G Cavallari
- Nephrology, Dialysis and Transplantation Unit (Stefoni), S.Orsola-Malpighi Hospital, University of Bologna, Italy
| | - P Gerber
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - O Tschopp
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - G A Spinas
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - W Moritz
- InSphero AG, Schlieren, Switzerland
| | - R Lehmann
- Division of Endocrinology, Diabetes and Clinical Nutrition, University Hospital Zurich, Switzerland
| |
Collapse
|
33
|
Abstract
BACKGROUND Several studies have revealed that posttransplant insulin treatment is beneficial to rest the islet grafts. However, insulin infusion per se is not enough to completely suppress the heavy workload arising caused by postprandial hyperglycemia. Therefore, the present study examined whether short-term fasting combined with insulin treatment could effectively prevent graft exhaustion after intraportal islet transplantation. METHODS A marginal dose of syngeneic rat islet grafts were transplanted intraportally into the control, insulin-treated, and insulin+rest groups of streptozotocin-induced diabetic rats. The control group fed freely without insulin treatment, and the other groups were continuously treated with an optimal amount of insulin to maintain normoglycemia. In addition, the insulin+rest group fasted and received total parenteral nutrition during the 2 weeks after transplantation. RESULTS The curative rate was significantly higher in both the insulin and insulin+rest groups than the control group (P<0.0001). The glucose tolerance, residual graft mass, and graft function were significantly ameliorated in the insulin+rest group, but not in the insulin group, compared to the control group (P<0.01, P=0.03, P=0.001). CONCLUSIONS These data suggest that short-term fasting combined with insulin treatment, especially during the avascular period of the grafts, could therefore be a promising regimen for improving pancreatic islet engraftment in the liver.
Collapse
|
34
|
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
|
35
|
Samikannu B, Chen C, Lingwal N, Padmasekar M, Engel FB, Linn T. Dipeptidyl peptidase IV inhibition activates CREB and improves islet vascularization through VEGF-A/VEGFR-2 signaling pathway. PLoS One 2013; 8:e82639. [PMID: 24349326 PMCID: PMC3859629 DOI: 10.1371/journal.pone.0082639] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 11/04/2013] [Indexed: 01/09/2023] Open
Abstract
Substitution of pancreatic islets is a potential therapy to treat diabetes and it depends on reconstitution of islet’s capillary network. In this study, we addressed the question whether stabilization of Glucagon-Like-Peptide-1 (GLP-1) by inhibiting Dipeptidyl Peptidase-IV (DPP-IV) increases β-cell mass by modulating vascularization. Mouse or porcine donor islets were implanted under kidney capsule of diabetic mice treated with DPP-IV inhibitor sitagliptin. Grafts were analyzed for insulin production, β-cell proliferation and vascularization. In addition, the effect of sitagliptin on sprouting and Vascular Endothelial Growth Factor (VEGF)-A expression was examined ex vivo. The cAMP response element-binding (CREB) and VEGF-A/ Vascular Endothelial Growth Factor Receptor (VEGFR)-2 signaling pathway leading to islet vascularization was explored. Sitagliptin increased mean insulin content of islet grafts and area of insulin-positive tissue as well as β-cell proliferation. Interestingly, sitagliptin treatment also markedly increased endothelial cell proliferation, microvessel density and blood flow. Finally, GLP-1 (7-36) stimulated sprouting and VEGF expression, which was significantly enhanced by sitagliptin- mediated inhibition of DPP-IV. Our in vivo data demonstrate that sitagliptin treatment phosphorylated CREB and induced islet vascularization through VEGF-A/VEGFR-2 signaling pathway. This study paves a new pathway for improvement of islet transplantation in treating diabetes mellitus.
Collapse
Affiliation(s)
- Balaji Samikannu
- Third Medical Clinic and Policlinic, Justus-Liebig-University, Giessen, Germany
- * E-mail:
| | - Chunguang Chen
- CRTD / DFG- Center for Regenerative Therapies Dresden, Paul Langerhans Institut Dresden, Dresden, Germany
| | - Neelam Lingwal
- Third Medical Clinic and Policlinic, Justus-Liebig-University, Giessen, Germany
| | - Manju Padmasekar
- Third Medical Clinic and Policlinic, Justus-Liebig-University, Giessen, Germany
| | - Felix B. Engel
- University Hospital Erlangen, Experimental Renal and Cardiovascular Research, Nephropathology Division, Department of Pathology, Erlangen, Germany
| | - Thomas Linn
- Third Medical Clinic and Policlinic, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
36
|
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
|
37
|
Wittig C, Laschke MW, Scheuer C, Menger MD. Incorporation of bone marrow cells in pancreatic pseudoislets improves posttransplant vascularization and endocrine function. PLoS One 2013; 8:e69975. [PMID: 23875013 PMCID: PMC3715469 DOI: 10.1371/journal.pone.0069975] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 06/14/2013] [Indexed: 01/01/2023] Open
Abstract
Failure of revascularization is known to be the major reason for the poor outcome of pancreatic islet transplantation. In this study, we analyzed whether pseudoislets composed of islet cells and bone marrow cells can improve vascularization and function of islet transplants. Pancreatic islets isolated from Syrian golden hamsters were dispersed into single cells for the generation of pseudoislets containing 4×103 cells. To create bone marrow cell-enriched pseudoislets 2×103 islet cells were co-cultured with 2×103 bone marrow cells. Pseudoislets and bone marrow cell-enriched pseudoislets were transplanted syngeneically into skinfold chambers to study graft vascularization by intravital fluorescence microscopy. Native islet transplants served as controls. Bone marrow cell-enriched pseudoislets showed a significantly improved vascularization compared to native islets and pseudoislets. Moreover, bone marrow cell-enriched pseudoislets but not pseudoislets normalized blood glucose levels after transplantation of 1000 islet equivalents under the kidney capsule of streptozotocin-induced diabetic animals, although the bone marrow cell-enriched pseudoislets contained only 50% of islet cells compared to pseudoislets and native islets. Fluorescence microscopy of bone marrow cell-enriched pseudoislets composed of bone marrow cells from GFP-expressing mice showed a distinct fraction of cells expressing both GFP and insulin, indicating a differentiation of bone marrow-derived cells to an insulin-producing cell-type. Thus, enrichment of pseudoislets by bone marrow cells enhances vascularization after transplantation and increases the amount of insulin-producing tissue. Accordingly, bone marrow cell-enriched pseudoislets may represent a novel approach to increase the success rate of islet transplantation.
Collapse
Affiliation(s)
- Christine Wittig
- Institute for Clinical & Experimental Surgery, University of Saarland, Homburg/Saar, Germany.
| | | | | | | |
Collapse
|
38
|
Tian L, Kim HS, Kim H, Jin X, Jung HS, Park KS, Cho KW, Park S, Moon WK. Changes in metabolic markers in insulin-producing β-cells during hypoxia-induced cell death as studied by NMR metabolomics. J Proteome Res 2013; 12:3738-45. [PMID: 23795807 DOI: 10.1021/pr400315e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This study was designed to investigate changes in the metabolites in the intracellular fluid of the pancreatic β-cell line INS-1 to identify potential early and late biomarkers for predicting hypoxia-induced cell death. INS-1 cells were incubated under normoxic conditions (95% air, 5% CO₂) or hypoxic conditions (1% O₂, 5% CO₂, 95% N₂) for 2, 4, 6, 12, or 24 h. The biological changes indicating the process of cell death were analyzed using the MTT assay, flow cytometry, Western blotting, and immunostaining. Changes in the metabolic profiles from cell lysates were identified using ¹H nuclear magnetic resonance (¹H NMR) spectroscopy, and the spectra were analyzed by the multivariate model Orthogonal Projections to Latent Structure-Discriminant Analysis. Cell viability decreased approximately 40% after 12-24 h of hypoxia, coincident with a high level of cleaved caspase-3. A high level of HIF-1α was detected in the 12-24 h hypoxic conditions. The metabolite profiles were altered according to the degree of exposure to hypoxia. A spectral analysis showed significant differences in creatine-containing compounds at the early stage (2-6 h) and taurine-containing compounds at the late stage (12-24 h), with the detection of HIF-1α and cleaved caspase-3 in cells exposed to hypoxia compared to normoxia. Glycerophosphocholine decreased during the early stage hypoxia. The change in taurine- and creatine-containing compounds and choline species could be involved in the β-cell death process as inhibitors or activators of cell death. Our results imply that assessment by ¹H NMR spectroscopy would be a useful tool to predict the cell death process and to identify molecules regulating hypoxia-induced cell death mechanisms.
Collapse
Affiliation(s)
- Lianji Tian
- Department of Biomedical Science, College of Medicine, Medical Research Center, Seoul National University, 101 Daehangno, Jongnogu, Seoul 110-744, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Islet transplantation has been shown to be a viable treatment option for patients afflicted with type 1 diabetes. However, the lack of availablity of human pancreases and the need to use risky immunosuppressive drugs to prevent transplant rejection remain two major obstacles to the routine use of islet transplantation in diabetic patients. Successful development of a bioartificial pancreas using the approach of microencapsulation with perm-selective coating of islets in hydrogels for graft immunoisolation holds tremendous promise for diabetic patients because it has great potential to overcome these two barriers. In this review article, we will discuss the need for a bioartificial pancreas, provide a detailed description of the microencapsulation process, and review the status of the technology in clinical development. We will also critically review the various factors that will need to be taken into consideration in order to achieve the ultimate goal of routine clinical application.
Collapse
Affiliation(s)
- Rajesh A Pareta
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Alan C Farney
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Emmanuel C Opara
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
40
|
Sterkers A, Hubert T, Gmyr V, Torres F, Baud G, Delalleau N, Vantyghem MC, Kerr-Conte J, Caiazzo R, Pattou F. Islet survival and function following intramuscular autotransplantation in the minipig. Am J Transplant 2013; 13:891-898. [PMID: 23496914 DOI: 10.1111/ajt.12136] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 10/22/2012] [Accepted: 11/05/2012] [Indexed: 01/25/2023]
Abstract
The liver may not be an optimal site for islet transplantation due to obstacles by an instant blood-mediated inflammatory response (IBMIR), and low revascularization of transplanted islets. Therefore, intramuscular islet transplantation (IMIT) offers an attractive alternative, based on its simplicity, enabling easier access for noninvasive graft imaging and cell explantation. In this study, we explored the outcome of autologous IMIT in the minipig (n = 30). Using the intramuscular injection technique, we demonstrated by direct histological evidence the rapid revascularization of islets autotransplanted into the gracilius muscle. Islet survival assessment was performed using immunohistochemistry staining for insulin and glucagon up to a period of 6 months. Furthermore, we showed the crucial role of minimizing mechanical trauma to the myofibers and limiting exocrine contamination. Intramuscular islet graft function after transplantation was confirmed by documenting the acute insulin response to intravenous glucose in 5/11 pancreatectomized animals. Graft function after IMIT remained however significantly lower than the function measured in 12 out of 18 minipigs who received a similar islet volume in the liver through intraportal infusion. Collectively, these results demonstrated in a clinically relevant preclinical model, suggest IMIT as a promising alternative to intraportal infusion for the transplantation of β cells in certain medical situations.
Collapse
Affiliation(s)
- A Sterkers
- UMR 859 Biotherapies for diabetes, INSERM, 59000 Lille, France.,UDSL, University of Lille Nord de, France, 59000 Lille, France.,General and Endocrine surgery, CHRU, Lille, 59000 Lille, France
| | - T Hubert
- UMR 859 Biotherapies for diabetes, INSERM, 59000 Lille, France.,UDSL, University of Lille Nord de, France, 59000 Lille, France
| | - V Gmyr
- UMR 859 Biotherapies for diabetes, INSERM, 59000 Lille, France.,UDSL, University of Lille Nord de, France, 59000 Lille, France
| | - F Torres
- UMR 859 Biotherapies for diabetes, INSERM, 59000 Lille, France.,UDSL, University of Lille Nord de, France, 59000 Lille, France.,General and Endocrine surgery, CHRU, Lille, 59000 Lille, France
| | - G Baud
- UMR 859 Biotherapies for diabetes, INSERM, 59000 Lille, France.,UDSL, University of Lille Nord de, France, 59000 Lille, France.,General and Endocrine surgery, CHRU, Lille, 59000 Lille, France
| | - N Delalleau
- UMR 859 Biotherapies for diabetes, INSERM, 59000 Lille, France.,UDSL, University of Lille Nord de, France, 59000 Lille, France
| | - M C Vantyghem
- UMR 859 Biotherapies for diabetes, INSERM, 59000 Lille, France.,UDSL, University of Lille Nord de, France, 59000 Lille, France.,Endocrinology and metabolism, CHRU, Lille, 59000 Lille, France
| | - J Kerr-Conte
- UMR 859 Biotherapies for diabetes, INSERM, 59000 Lille, France.,UDSL, University of Lille Nord de, France, 59000 Lille, France
| | - R Caiazzo
- UMR 859 Biotherapies for diabetes, INSERM, 59000 Lille, France.,UDSL, University of Lille Nord de, France, 59000 Lille, France.,General and Endocrine surgery, CHRU, Lille, 59000 Lille, France
| | - F Pattou
- UMR 859 Biotherapies for diabetes, INSERM, 59000 Lille, France.,UDSL, University of Lille Nord de, France, 59000 Lille, France.,General and Endocrine surgery, CHRU, Lille, 59000 Lille, France.,European Genomic Institute for Diabetes (EGID), FR 3508, 59000 Lille, France
| |
Collapse
|
41
|
Combined strategy of endothelial cells coating, Sertoli cells coculture and infusion improves vascularization and rejection protection of islet graft. PLoS One 2013; 8:e56696. [PMID: 23437215 PMCID: PMC3577699 DOI: 10.1371/journal.pone.0056696] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 01/14/2013] [Indexed: 11/19/2022] Open
Abstract
Improving islet graft revascularization and inhibiting rejection become crucial tasks for prolonging islet graft survival. Endothelial cells (ECs) are the basis of islet vascularization and Sertoli cells (SCs) have the talent to provide nutritional support and exert immunosuppressive effects. We construct a combined strategy of ECs coating in the presence of nutritious and immune factors supplied by SCs in a co-culture system to investigate the effect of vascularization and rejection inhibition for islet graft. In vivo, the combined strategy improved the survival and vascularization as well as inhibited lymphocytes and inflammatory cytokines. In vitro, we found the combinatorial strategy improved the function of islets and the effect of ECs-coating on islets. Combined strategy treated islets revealed higher levels of anti-apoptotic signal molecules (Bcl-2 and HSP-32), survival and function related molecules (PDX-1, Ki-67, ERK1/2 and Akt) and demonstrated increased vascular endothelial growth factor receptor 2 (KDR) and angiogenesis signal molecules (FAk and PLC-γ). SCs effectively inhibited the activation of lymphocyte stimulated by islets and ECs. Predominantly immunosuppressive cytokines could be detected in culture supernatants of the SCs coculture group. These results suggest that ECs-coating and Sertoli cells co-culture or infusion synergistically enhance islet survival and function after transplantation.
Collapse
|
42
|
Kuehl AR, Abshagen K, Eipel C, Laschke MW, Menger MD, Laue M, Vollmar B. External inosculation as a feature of revascularization occurs after free transplantation of murine liver grafts. Am J Transplant 2013; 13:286-98. [PMID: 23205733 DOI: 10.1111/j.1600-6143.2012.04336.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 10/02/2012] [Accepted: 10/09/2012] [Indexed: 01/25/2023]
Abstract
The induction of angiogenesis is essential for successful engraftment of freely transplanted cells or cellular composites. How to augment angiogenesis to ensure an appropriate viability of the grafts is still under investigation. This study evaluated the proangiogenic capability of different syngeneic free liver transplants and elucidated the origin of the newly formed vascular network via use of an eGFP(+) /eGFP(-) (enhanced green fluorescent protein) cross-over design. Using intravital fluorescence microscopy, we found that neonatal and resected murine liver transplants implanted into dorsal skinfold chambers display a significantly enhanced vascularization compared to regular adult transplants. Immunohistochemically, less tissue hypoxia, apoptosis and macrophage infiltration was observed in the neonatal and resected transplants, which is in line with improved vascularization of those grafts. Additionally, electron microscopy revealed morphological hallmarks of liver cells. eGFP(+) liver transplants implanted on eGFP(-) recipients displayed vascular sprouting from the grafts themselves and connection to the recipients` microvasculature, which also undergoes transient proangiogenic response. This process is described as external inosculation, with microvessels exhibiting a chimeric nature of the endothelial lining. These data collectively show that proliferative stimulation is taking effect on angiogenic properties of free transplants and might provide a novel tool for modulating the revascularization of free grafts.
Collapse
Affiliation(s)
- A-R Kuehl
- Institute for Experimental Surgery, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Dufrane D, Gianello P. Macro- or microencapsulation of pig islets to cure type 1 diabetes. World J Gastroenterol 2012; 18:6885-93. [PMID: 23322985 PMCID: PMC3531671 DOI: 10.3748/wjg.v18.i47.6885] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 06/14/2012] [Accepted: 06/28/2012] [Indexed: 02/06/2023] Open
Abstract
Although allogeneic islet transplantation can successfully cure type 1 diabetes, it has limited applicability. For example, organs are in short supply; several human pancreas donors are often needed to treat one diabetic recipient; the intrahepatic site may not be the most appropriate site for islet implantation; and immunosuppressive regimens, which are associated with side effects, are often required to prolong survival of the islet graft. An alternative source of insulin-producing cells would therefore be of major interest. Pigs represent a possible alternative source of beta cells. Grafting of pig islets may appear difficult because of the immunologic species barrier, but pig islets have been shown to function in primates for at least 6 mo with clinically incompatible immunosuppression. Therefore, a bioartificial pancreas made of encapsulated pig islets may resolve issues associated with islet allotransplantation. Although several groups have shown that encapsulated pig islets are functional in small-animal models, less is known about the use of bioartificial pancreases in large-animal models. In this review, we summarize current knowledge of encapsulated pig islets, to determine obstacles to implantation in humans and possible solutions to overcome these obstacles.
Collapse
|
44
|
Williams SJ, Huang HH, Kover K, Moore W, Berkland C, Singh M, Smirnova IV, MacGregor R, Stehno-Bittel L. Reduction of diffusion barriers in isolated rat islets improves survival, but not insulin secretion or transplantation outcome. Organogenesis 2012; 6:115-24. [PMID: 20885858 DOI: 10.4161/org.6.2.10373] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Accepted: 10/20/2009] [Indexed: 01/05/2023] Open
Abstract
For people with type 1 diabetes and severe hypoglycemic unawareness, islet transplants offer hope for improving the quality of life. However, islet cell death occurs quickly during or after transplantation, requiring large quantities of islets per transplant. The purpose of this study was to determine whether poor function demonstrated in large islets was a result of diffusion barriers and if removing those barriers could improve function and transplantation outcomes. Islets were isolated from male DA rats and measured for cell viability, islet survival, glucose diffusion and insulin secretion. Modeling of diffusion barriers was completed using dynamic partial differential equations for a sphere. Core cell death occurred in 100% of the large islets (diameter >150 µm), resulting in poor survival within 7 days after isolation. In contrast, small islets (diameter <100 µm) exhibited good survival rates in culture (91%). Glucose diffusion into islets was tracked with 2-NBDG; 4.2 µm/min in small islets and 2.8 µm/min in large islets. 2-NBDG never permeated to the core cells of islets larger than 150 µm diameter. Reducing the diffusion barrier in large islets improved their immediate and long-term viability in culture. However, reduction of the diffusion barrier in large islets failed to improve their inferior in vitro insulin secretion compared to small islets, and did not return glucose control to diabetic animals following transplantation. Thus, diffusion barriers lead to low viability and poor survival for large islets, but are not solely responsible for the inferior insulin secretion or poor transplantation outcomes of large versus small islets.
Collapse
Affiliation(s)
- S Janette Williams
- Department of Physical Therapy and Rehabilitation Science, University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kaufman-Francis K, Koffler J, Weinberg N, Dor Y, Levenberg S. Engineered vascular beds provide key signals to pancreatic hormone-producing cells. PLoS One 2012; 7:e40741. [PMID: 22808248 PMCID: PMC3395696 DOI: 10.1371/journal.pone.0040741] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 06/12/2012] [Indexed: 12/13/2022] Open
Abstract
The mechanisms underlying early islet graft failure are not entirely clear, but are thought to involve ischemic injury due to delayed vascularization. We hypothesize that blood vessels play an active role in cell-cell communications supporting islet survival and engraftment. To test this hypothesis and to uncouple endothelial cell (EC)-generated signaling stimuli from their nutritional and gas exchange functions, we developed three dimensional (3D) endothelial vessel networks in engineered pancreatic tissues prepared from islets, fibroblasts and ECs. The tri-culture setup, seeded on highly porous biocompatible polymeric scaffolds closely mimics the natural anatomical context of pancreatic vasculature. Enhanced islet survival correlating with formation of functional tube-like endothelial vessels was demonstrated. Addition of foreskin fibroblasts to islet-endothelial cultures promoted tube-like structure formation, which further supported islet survival as well as insulin secretion. Gene expression profiles of EC growth factors, extracellular matrix (ECM), morphogenes and differentiation markers were significantly different in 2D versus 3D culture systems and were further modified upon addition of fibroblasts. Implantation of prevascularized islets into diabetic mice promoted survival, integration and function of the engrafted engineered tissue, supporting the suggested role of ECs in islet survival. These findings present potential strategies for preparation of transplantable islets with increased survival prospects.
Collapse
Affiliation(s)
- Keren Kaufman-Francis
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
- Biotechnology Interdisciplinary Unit, Technion - Israel Institute of Technology, Haifa, Israel
| | - Jacob Koffler
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
- Biotechnology Interdisciplinary Unit, Technion - Israel Institute of Technology, Haifa, Israel
| | - Noa Weinberg
- Department of Cellular Biochemistry and Human Genetics, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yuval Dor
- Department of Cellular Biochemistry and Human Genetics, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
- * E-mail:
| |
Collapse
|
46
|
Cavallari G, Olivi E, Bianchi F, Neri F, Foroni L, Valente S, La Manna G, Nardo B, Stefoni S, Ventura C. Mesenchymal stem cells and islet cotransplantation in diabetic rats: improved islet graft revascularization and function by human adipose tissue-derived stem cells preconditioned with natural molecules. Cell Transplant 2012; 21:2771-2781. [PMID: 22472472 DOI: 10.3727/096368912x637046] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hypoxia plays an important role in limiting the engraftment, survival, and function of intrahepatically transplanted islets. Mesenchymal stem cells (MSCs) were recently used in animal models of islet transplantation not only to reduce allograft rejection but also to promote revascularization. Among different possible origins, adipose tissue represents a novel and good source of MSCs. Moreover, the capability of adipose tissue-derived stem cells (ASCs) to improve islet graft revascularization was recently reported after hybrid transplantation in mice. Within this context, we have previously shown that hyaluronan esters of butyric and retinoic acids can significantly enhance the rescuing potential of human MSCs (hMSCs). Here we evaluated whether ex vivo preconditioning of human ASCs (hASCs) with a mixture of hyaluronic (HA), butyric (BU), and retinoic (RA) acids may result in optimization of graft revascularization after islet/stem cell intrahepatic cotransplantation in syngeneic diabetic rats. We demonstrated that hASCs exposed to the mixture of molecules are able to increase the secretion of vascular endothelial growth factor (VEGF) as well as the transcription of angiogenic genes, including VEGF, KDR (kinase insert domain receptor), and hepatocyte growth factor (HGF). Rats transplanted with islets cocultured with preconditioned hASCs exhibited a better glycemic control than rats transplanted with an equal volume of islets and control hASCs. Cotransplantation with preconditioned hASCs was also associated with enhanced islet revascularization in vivo, as highlighted by graft morphological analysis. The observed increase in islet graft revascularization and function suggests that our method of stem cell preconditioning may represent a novel strategy to remarkably improve the efficacy of islets-hMSCs cotransplantation.
Collapse
Affiliation(s)
- Giuseppe Cavallari
- Department of General Surgery and Transplantation, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Islet graft survival and function: concomitant culture and transplantation with vascular endothelial cells in diabetic rats. Transplantation 2012; 92:1208-14. [PMID: 22067310 DOI: 10.1097/tp.0b013e3182356ca7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Human islet transplantation is a great potential therapy for type I diabetes. To investigate islet graft survival and function, we recently showed the improved effects after co-culture and co-transplantation with vascular endothelial cells (ECs) in diabetic rats. METHODS ECs were isolated, and the viability of isolated islets was assessed in two groups (standard culture group and co-culture group with ECs). Then streptozotocin-induced diabetic rats were divided into four groups before islet transplantation as follows: group A with infusion of islet grafts; group B with combined vascular ECs and islet grafts; groups C and D as controls with single ECs infusion and phosphate-buffered saline injection, respectively. Blood glucose and insulin concentrations were measured daily. Expression of vascular endothelial growth factor was investigated by immunohistochemical staining. The mean microvascular density was also calculated. RESULTS More than 90% of acridine orange-propidium iodide staining positive islets demonstrated normal morphology while co-cultured with ECs for 7 days. Compared with standard control, insulin release assays showed a significantly higher simulation index in co-culture group except for the first day (P<0.05). After transplantation, there was a significant difference in concentrations of blood glucose and insulin among these groups after 3 days (P<0.05). The mean microvascular density in co-culture group was significantly higher than that in single islet group (P=0.04). CONCLUSION Co-culture with ECs in vitro could improve the survival and function of isolated rat islet, and co-transplantation of islets with ECs could effectively prolong the islet graft survival in diabetic rats.
Collapse
|
48
|
Saito Y, Chan NK, Sakata N, Hathout E. Nerve growth factor is associated with islet graft failure following intraportal transplantation. Islets 2012; 4:24-31. [PMID: 22192949 PMCID: PMC3365801 DOI: 10.4161/isl.18467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Nerve growth factor (NGF) has recently been recognized as an angiogenic factor with an important regulatory role in pancreatic β-cell function. We previously showed that treatment of pancreatic islets with NGF improved their quality and viability. Revascularization and survival of islets transplanted under the kidney capsule were improved by NGF. However, the usefulness of NGF in intraportal islet transplantation was not previously tested. To resolve this problem, we transplanted syngeneic islets (360 islet equivalents per recipient) cultured with or without NGF into the portal vein of streptozotocin-induced diabetic BALB/c mice. Analysis revealed that 44.4% (4/9) of control and 12.5% (1/8) of NGF-treated mice attained normoglycemia (≤ 200 mg/dL) (p = 0.195). NGF-treated islets led to worse graft function (area under the curve of intraperitoneal glucose tolerance tests (IPGTT) on post-operative day (POD) 30, control; 35,800 ± 3,960 min*mg/dl, NGF-treated; 47,900 ± 3,220 min*mg/dl: *p = 0.0348). NGF treatment of islets was also associated with increased graft failure [the percentage of TdT-mediated dUTP-biotin nick-end labeling (TUNEL)-positive and necrotic transplanted islets on POD 5, control; 23.8% (5/21), NGF-treated; 52.9% (9/17): p = 0.0650] following intraportal islet transplantation. Nonviable (TUNEL-positive and necrotic) islets in both groups expressed vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α (HIF-1α). On the other hand, viable (TUNEL-negative and not necrotic) islets in both groups did not express VEGF and HIF-1α. In the present study, pre-transplant NGF treatment was associated with impaired survival and angiogenesis of intraportal islet grafts. The effect of NGF on islet transplantation may significantly vary according to the transplant site.
Collapse
Affiliation(s)
- Yukihiko Saito
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
- Division of Advanced Surgical Science and Technology; Department of Surgery; Tohoku University; Sendai, Japan
| | - Nathaniel K. Chan
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
| | - Naoaki Sakata
- Division of Hepato-Biliary Pancreatic Surgery; Department of Surgery; Tohoku University; Sendai, Japan
| | - Eba Hathout
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
- * Correspondence to: Eba Hathout;
| |
Collapse
|
49
|
Lee BR, Hwang JW, Choi YY, Wong SF, Hwang YH, Lee DY, Lee SH. In situ formation and collagen-alginate composite encapsulation of pancreatic islet spheroids. Biomaterials 2011; 33:837-45. [PMID: 22054535 DOI: 10.1016/j.biomaterials.2011.10.014] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 10/10/2011] [Indexed: 11/19/2022]
Abstract
In this study, we suggest in situ islet spheroid formation and encapsulation on a single platform without replating as a method for producing mono-disperse spheroids and minimizing damage to spheroids during encapsulation. Using this approach, the size of spheroid can be controlled by modulating the size of the concave well. Here, we used 300 μm concave wells to reduce spheroid size and thereby eliminating the central necrosis caused by large volume. As the encapsulation material, we used alginate and collagen-alginate composite (CAC), and evaluated their suitability through diverse in vitro tests, including measurements of viability, oxygen consumption rate (OCR), hypoxic damage to encapsulated spheroids, and insulin secretion. For in situ encapsulation, alginate or CAC was spread over a concave microwell array containing spheroids, and CaCl(2) solution was diffused through a nano-porous dialysis membrane to achieve uniform polymerization, forming convex structures. By this process, the formation of uniform-size islet spheroids and their encapsulation without an intervening replating step was successfully performed. As a control, intact islets were evaluated concurrently. The in vitro test demonstrated excellent performance of CAC-encapsulated spheroids, and on the basis of these results, we transplanted the islet spheroids-encapsulated with CAC into the intraperitoneal cavity of mice with induced diabetes for 4 weeks, and evaluated subsequent glucose control. Intact islets were also transplanted as control to investigate the effect of encapsulation. Transplanted CAC-encapsulated islet spheroids maintained glucose levels below 200 mg/dL for 4 weeks, at which they were still active. At the end of the implantation experiment, we carried out intraperitoneal glucose tolerance test (IPGTT) in mice to investigate whether the implanted islets remained responsive to glucose. The glucose level in mice with CAC-encapsulated islet spheroids dropped below 200 mg/dL 60 min after glucose injection and was stably maintained. In conclusion, the proposed encapsulation method enhances the viability and function of islet spheroids, and protects these spheroids from immune attack.
Collapse
Affiliation(s)
- Bo Ram Lee
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 136-703, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
50
|
Improving islet engraftment by gene therapy. J Transplant 2011; 2011:594851. [PMID: 22132301 PMCID: PMC3202131 DOI: 10.1155/2011/594851] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/22/2011] [Indexed: 11/18/2022] Open
Abstract
Islet cell transplantation is currently the only feasible long-term treatment option for patients with type 1 diabetes. However, the majority of transplanted islets experience damage and apoptosis during the isolation process, a blood-mediated inflammatory microenvironment in the portal vein upon islet infusion, hypoxia induced by the low oxygenated milieu, and poor-revascularization-mediated lack of nutrients, and impaired hormone modulation in the local transplanted site. Strategies using genetic modification methods through overexpression or silencing of those proteins involved in promoting new formation of blood vessels or inhibition of apoptosis may overcome these hurdles and improve islet engraftment outcomes.
Collapse
|