1
|
Opara A, Canning P, Alwan A, Opara EC. Challenges and Perspectives for Future Considerations in the Bioengineering of a Bioartificial Pancreas. Ann Biomed Eng 2024; 52:1795-1803. [PMID: 36913086 DOI: 10.1007/s10439-023-03180-7] [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: 10/21/2022] [Accepted: 02/25/2023] [Indexed: 03/14/2023]
Abstract
There is an unrelenting interest in the development of a reliable bioartificial pancreas construct since the first description of this technology of encapsulated islets by Lim and Sun in 1980 because it promised to be a curative treatment for Type 1 Diabetes Mellitus (T1DM). Despite the promise of the concept of encapsulated islets, there are still some challenges that impede the full realization of the clinical potential of the technology. In this review, we will first present the justification for continued research and development of this technology. Next, we will review key barriers that impede progress in this field and discuss strategies that can be used to design a reliable construct capable of effective long-term performance after transplantation in diabetic patients. Finally, we will share our perspectives on areas of additional work for future research and development of the technology.
Collapse
Affiliation(s)
- Amoge Opara
- Diabetes Section, Biologics Delivery Technologies, Reno, NV, 89502, USA
| | - Priyadarshini Canning
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Abdelrahman Alwan
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, 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
|
2
|
Geng Z, Zhang Q, Li T, Huang T, Wang H, Zhou Q, Deng S, Zhao Y, Li Y, Cheng C, Gonelle-Gispert C, Buhler LH, Wang Y. Advantages of the retroperitoneal retrocolic space as the transplant site for encapsulated xenogeneic islets. Xenotransplantation 2023; 30:e12787. [PMID: 36454040 DOI: 10.1111/xen.12787] [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: 07/18/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVE Islet allotransplantation has demonstrated improved clinical outcomes using the hepatic portal vein as the standard infusion method. However, the current implantation site is not ideal due to the short-term thrombotic and long-term immune destruction. Meanwhile, the shortage of human organ donors further limits its application. To find a new strategy, we tested a new polymer combination for islet encapsulation and transplantation. Meanwhile, we explored a new site for xenogeneic islet transplantation in mice. METHOD We synthesized a hydrogel combining alginate plus poly-ethylene-imine (Alg/PEI) for the encapsulation of rat, neonatal porcine, and human islets. Transplantation was performed into the retroperitoneal retro-colic space of diabetic mice. Control mice received free islets under the kidney capsule or encapsulated islets into the peritoneum. The biochemical indexes were measured, and the transplanted islets were harvested for immunohistochemical staining of insulin and glucagon. RESULTS Mice receiving encapsulated rat, porcine and human islets transplanted into the retroperitoneal space maintained normoglycemia for a median of 275, 145.5, and 146 days, respectively. In contrast, encapsulated xenogeneic islets transplanted into the peritoneum, maintained function for a median of 61, 95.5, and 82 days, respectively. Meanwhile, xenogeneic islets transplanted free into the kidney capsule lost their function within 3 days after transplantation. Immunohistochemical staining of encapsulated rat, porcine and human islets, retrieved from the retroperitoneal space, allowed to distinguish morphological normal insulin expressing β- and glucagon expressing α-cells at 70, 60, and 100 days post-transplant, respectively. CONCLUSION Transplantation of Alg/PEI encapsulated xenogeneic islets into the retroperitoneal space provides a valuable new implantation strategy for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Zhen Geng
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qi Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ting Li
- Department of Rheumatology, Wenjiang District People's Hospital, Chengdu, China
| | - Ting Huang
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Institute of Organ Transplantation, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiao Zhou
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shaoping Deng
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Institute of Organ Transplantation, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanshuang Zhao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanjiao Li
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Leo H Buhler
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, China
| |
Collapse
|
3
|
Honarpisheh M, Lei Y, Zhang Y, Pehl M, Kemter E, Kraetzl M, Lange A, Wolf E, Wolf-van Buerck L, Seissler J. Formation of Re-Aggregated Neonatal Porcine Islet Clusters Improves In Vitro Function and Transplantation Outcome. Transpl Int 2022; 35:10697. [PMID: 36685665 PMCID: PMC9846776 DOI: 10.3389/ti.2022.10697] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
Neonatal porcine islet-like cell clusters (NPICCs) are a promising source for islet cell transplantation. Excellent islet quality is important to achieve a cure for type 1 diabetes. We investigated formation of cell clusters from dispersed NPICCs on microwell cell culture plates, evaluated the composition of re-aggregated porcine islets (REPIs) and compared in vivo function by transplantation into diabetic NOD-SCID IL2rγ-/- (NSG) mice with native NPICCs. Dissociation of NPICCs into single cells and re-aggregation resulted in the formation of uniform REPI clusters. A higher prevalence of normoglycemia was observed in diabetic NSG mice after transplantation with a limited number (n = 1500) of REPIs (85.7%) versus NPICCs (n = 1500) (33.3%) (p < 0.05). Transplanted REPIs and NPICCs displayed a similar architecture of endocrine and endothelial cells. Intraperitoneal glucose tolerance tests revealed an improved beta cell function after transplantation of 1500 REPIs (AUC glucose 0-120 min 6260 ± 305.3) as compared to transplantation of 3000 native NPICCs (AUC glucose 0-120 min 8073 ± 536.2) (p < 0.01). Re-aggregation of single cells from dissociated NPICCs generates cell clusters with excellent functionality and improved in vivo function as compared to native NPICCs.
Collapse
Affiliation(s)
- M. Honarpisheh
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Y. Lei
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - Y. Zhang
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - M. Pehl
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - E. Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - M. Kraetzl
- Chair for Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - A. Lange
- Chair for Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - E. Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - L. Wolf-van Buerck
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| | - J. Seissler
- Medizinische Klinik und Poliklinik IV, Diabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
4
|
Pignatelli C, Campo F, Neroni A, Piemonti L, Citro A. Bioengineering the Vascularized Endocrine Pancreas: A Fine-Tuned Interplay Between Vascularization, Extracellular-Matrix-Based Scaffold Architecture, and Insulin-Producing Cells. Transpl Int 2022; 35:10555. [PMID: 36090775 PMCID: PMC9452644 DOI: 10.3389/ti.2022.10555] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022]
Abstract
Intrahepatic islet transplantation is a promising β-cell replacement strategy for the treatment of type 1 diabetes. Instant blood-mediated inflammatory reactions, acute inflammatory storm, and graft revascularization delay limit islet engraftment in the peri-transplant phase, hampering the success rate of the procedure. Growing evidence has demonstrated that islet engraftment efficiency may take advantage of several bioengineering approaches aimed to recreate both vascular and endocrine compartments either ex vivo or in vivo. To this end, endocrine pancreas bioengineering is an emerging field in β-cell replacement, which might provide endocrine cells with all the building blocks (vascularization, ECM composition, or micro/macro-architecture) useful for their successful engraftment and function in vivo. Studies on reshaping either the endocrine cellular composition or the islet microenvironment have been largely performed, focusing on a single building block element, without, however, grasping that their synergistic effect is indispensable for correct endocrine function. Herein, the review focuses on the minimum building blocks that an ideal vascularized endocrine scaffold should have to resemble the endocrine niche architecture, composition, and function to foster functional connections between the vascular and endocrine compartments. Additionally, this review highlights the possibility of designing bioengineered scaffolds integrating alternative endocrine sources to overcome donor organ shortages and the possibility of combining novel immune-preserving strategies for long-term graft function.
Collapse
Affiliation(s)
- Cataldo Pignatelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Campo
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Alessia Neroni
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
5
|
Canning P, Alwan A, Khalil F, Zhang Y, Opara EC. Perspectives and Challenges on the Potential Use of Exosomes in Bioartificial Pancreas Engineering. Ann Biomed Eng 2022; 50:1177-1186. [PMID: 35804253 DOI: 10.1007/s10439-022-03004-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/30/2022] [Indexed: 11/01/2022]
Abstract
Exosomes are enclosed within a single outer membrane and exemplify a specific subtype of secreted vesicles. Exosomes transfer signalling molecules, including microRNAs (miRNAs), messenger RNA (mRNA), fatty acids, proteins, and growth factors, making them a promising therapeutic tool. In routine bioartificial pancreas fabrication, cells are immobilized in polymeric hydrogels lacking attachment capability for cells and other biological cues. In this opinion article, we will discuss the potential role that exosomes and their specific biofactors may play to improve and sustain the function of this bioartificial construct. We will particularly discuss the challenges associated with their isolation and characterization. Since stem cells are an attractive source of exosomes, we will present the advantages of using exosomes in place of stem cells in medical devices including the bioartificial pancreas. We will provide literature evidence of active biofactors in exosomes to support their incorporation in the matrix of encapsulated islets. This will include their potential beneficial effect on hypoxic injury to encapsulated islets. In summary, we propose that the biofactors contained in secreted exosomes have significant potential to enhance the performance of islets encapsulated in polymeric material hydrogels with perm-selective properties to provide immunoisolation for islet transplants as an insulin delivery platform in diabetes.
Collapse
Affiliation(s)
- Priyadarshini Canning
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Abdelrahman Alwan
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Fatma Khalil
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Emmanuel C Opara
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA. .,Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27-57, USA.
| |
Collapse
|
6
|
Viability and Functionality of Neonatal Porcine Islet-like Cell Clusters Bioprinted in Alginate-Based Bioinks. Biomedicines 2022; 10:biomedicines10061420. [PMID: 35740440 PMCID: PMC9220255 DOI: 10.3390/biomedicines10061420] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
The transplantation of pancreatic islets can prevent severe long-term complications in diabetes mellitus type 1 patients. With respect to a shortage of donor organs, the transplantation of xenogeneic islets is highly attractive. To avoid rejection, islets can be encapsulated in immuno-protective hydrogel-macrocapsules, whereby 3D bioprinted structures with macropores allow for a high surface-to-volume ratio and reduced diffusion distances. In the present study, we applied 3D bioprinting to encapsulate the potentially clinically applicable neonatal porcine islet-like cell clusters (NICC) in alginate-methylcellulose. The material was additionally supplemented with bovine serum albumin or the human blood plasma derivatives platelet lysate and fresh frozen plasma. NICC were analysed for viability, proliferation, the presence of hormones, and the release of insulin in reaction to glucose stimulation. Bioprinted NICC are homogeneously distributed, remain morphologically intact, and show a comparable viability and proliferation to control NICC. The number of insulin-positive cells is comparable between the groups and over time. The amount of insulin release increases over time and is released in response to glucose stimulation over 4 weeks. In summary, we show the successful bioprinting of NICC and could demonstrate functionality over the long-term in vitro. Supplementation resulted in a trend for higher viability, but no additional benefit on functionality was observed.
Collapse
|
7
|
Mou L, Shi G, Cooper DK, Lu Y, Chen J, Zhu S, Deng J, Huang Y, Ni Y, Zhan Y, Cai Z, Pu Z. Current Topics of Relevance to the Xenotransplantation of Free Pig Islets. Front Immunol 2022; 13:854883. [PMID: 35432379 PMCID: PMC9010617 DOI: 10.3389/fimmu.2022.854883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Pig islet xenotransplantation is a potential treatment for patients with type 1 diabetes. Current efforts are focused on identifying the optimal pig islet source and overcoming the immunological barrier. The optimal age of the pig donors remains controversial since both adult and neonatal pig islets have advantages. Isolation of adult islets using GMP grade collagenase has significantly improved the quantity and quality of adult islets, but neonatal islets can be isolated at a much lower cost. Certain culture media and coculture with mesenchymal stromal cells facilitate neonatal islet maturation and function. Genetic modification in pigs affords a promising strategy to prevent rejection. Deletion of expression of the three known carbohydrate xenoantigens (Gal, Neu5Gc, Sda) will certainly be beneficial in pig organ transplantation in humans, but this is not yet proven in islet transplantation, though the challenge of the '4th xenoantigen' may prove problematic in nonhuman primate models. Blockade of the CD40/CD154 costimulation pathway leads to long-term islet graft survival (of up to 965 days). Anti-CD40mAbs have already been applied in phase II clinical trials of islet allotransplantation. Fc region-modified anti-CD154mAbs successfully prevent the thrombotic complications reported previously. In this review, we discuss (I) the optimal age of the islet-source pig, (ii) progress in genetic modification of pigs, (iii) the immunosuppressive regimen for pig islet xenotransplantation, and (iv) the reduction in the instant blood-mediated inflammatory reaction.
Collapse
Affiliation(s)
- Lisha Mou
- Department of Hepatopancreatobiliary Surgery, Shenzhen Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Guanghan Shi
- Department of Hepatopancreatobiliary Surgery, Shenzhen Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- Faculty of Arts and Science, University of Toronto, Toronto, ON, Canada
| | - David K.C. Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, United States
| | - Ying Lu
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Jiao Chen
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Shufang Zhu
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Jing Deng
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Yuanyuan Huang
- Department of Life Science, Bellevue College, Bellevue, WA, United States
| | - Yong Ni
- Department of Hepatopancreatobiliary Surgery, Shenzhen Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Yongqiang Zhan
- Department of Hepatopancreatobiliary Surgery, Shenzhen Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Zhiming Cai
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Shenzhen Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Zuhui Pu
- Imaging Department, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
8
|
Mouré A, Bekir S, Bacou E, Pruvost Q, Haurogné K, Allard M, De Beaurepaire L, Bosch S, Riochet D, Gauthier O, Blancho G, Soulillou JP, Poncelet D, Mignot G, Courcoux P, Jegou D, Bach JM, Mosser M. Optimization of an O 2-balanced bioartificial pancreas for type 1 diabetes using statistical design of experiment. Sci Rep 2022; 12:4681. [PMID: 35304495 PMCID: PMC8933496 DOI: 10.1038/s41598-022-07887-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 02/03/2022] [Indexed: 01/17/2023] Open
Abstract
A bioartificial pancreas (BAP) encapsulating high pancreatic islets concentration is a promising alternative for type 1 diabetes therapy. However, the main limitation of this approach is O2 supply, especially until graft neovascularization. Here, we described a methodology to design an optimal O2-balanced BAP using statistical design of experiment (DoE). A full factorial DoE was first performed to screen two O2-technologies on their ability to preserve pseudo-islet viability and function under hypoxia and normoxia. Then, response surface methodology was used to define the optimal O2-carrier and islet seeding concentrations to maximize the number of viable pseudo-islets in the BAP containing an O2-generator under hypoxia. Monitoring of viability, function and maturation of neonatal pig islets for 15 days in vitro demonstrated the efficiency of the optimal O2-balanced BAP. The findings should allow the design of a more realistic BAP for humans with high islets concentration by maintaining the O2 balance in the device.
Collapse
Affiliation(s)
- Anne Mouré
- Oniris, INRAE, IECM, USC 1383, 44300, Nantes, France
| | - Sawsen Bekir
- Oniris, INRAE, IECM, USC 1383, 44300, Nantes, France
| | - Elodie Bacou
- Oniris, INRAE, IECM, USC 1383, 44300, Nantes, France
| | | | | | - Marie Allard
- Oniris, INRAE, IECM, USC 1383, 44300, Nantes, France
| | | | - Steffi Bosch
- Oniris, INRAE, IECM, USC 1383, 44300, Nantes, France
| | - David Riochet
- SSR Pédiatriques ESEAN-APF France Handicap, Nantes University Hospital, Nantes, France
| | - Olivier Gauthier
- Oniris, Nantes Université, INSERM, RMeS, UMR 1229, F-44000, Nantes, France
| | - Gilles Blancho
- CRTI, UMR 1064, INSERM, Nantes Université, 44000, Nantes, France
- ITUN, CHU Nantes, 44000, Nantes, France
| | - Jean-Paul Soulillou
- CRTI, UMR 1064, INSERM, Nantes Université, 44000, Nantes, France
- ITUN, CHU Nantes, 44000, Nantes, France
| | - Denis Poncelet
- GEPEA, UMR CNRS 6144 FR, Nantes Université, 44000, Nantes, France
| | | | | | | | | | | |
Collapse
|
9
|
Quizon MJ, García AJ. Engineering β Cell Replacement Therapies for Type 1 Diabetes: Biomaterial Advances and Considerations for Macroscale Constructs. ANNUAL REVIEW OF PATHOLOGY 2022; 17:485-513. [PMID: 34813353 DOI: 10.1146/annurev-pathol-042320-094846] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
While significant progress has been made in treatments for type 1 diabetes (T1D) based on exogenous insulin, transplantation of insulin-producing cells (islets or stem cell-derived β cells) remains a promising curative strategy. The current paradigm for T1D cell therapy is clinical islet transplantation (CIT)-the infusion of islets into the liver-although this therapeutic modality comes with its own limitations that deteriorate islet health. Biomaterials can be leveraged to actively address the limitations of CIT, including undesired host inflammatory and immune responses, lack of vascularization, hypoxia, and the absence of native islet extracellular matrix cues. Moreover, in efforts toward a clinically translatable T1D cell therapy, much research now focuses on developing biomaterial platforms at the macroscale, at which implanted platforms can be easily retrieved and monitored. In this review, we discuss how biomaterials have recently been harnessed for macroscale T1D β cell replacement therapies.
Collapse
Affiliation(s)
- Michelle J Quizon
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA; ,
| | - Andrés J García
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA; ,
| |
Collapse
|
10
|
Arefanian H, Ramji Q, Gupta N, Spigelman AF, Grynoch D, MacDonald PE, Mueller TF, Gazda LS, Rajotte RV, Rayat GR. Yield, cell composition, and function of islets isolated from different ages of neonatal pigs. Front Endocrinol (Lausanne) 2022; 13:1032906. [PMID: 36619563 PMCID: PMC9811407 DOI: 10.3389/fendo.2022.1032906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/01/2022] [Indexed: 12/24/2022] Open
Abstract
The yield, cell composition, and function of islets isolated from various ages of neonatal pigs were characterized using in vitro and in vivo experimental models. Islets from 7- and 10-day-old pigs showed significantly better function both in vitro and in vivo compared to islets from 3- and 5-day-old pigs however, the islet yield from 10-day-old pigs were significantly less than those obtained from the other pigs. Since islets from 3-day-old pigs were used in our previous studies and islets from 7-day-old pigs reversed diabetes more efficiently than islets from other groups, we further evaluated the function of these islets post-transplantation. B6 rag-/- mouse recipients of various numbers of islets from 7-day-old pigs achieved normoglycemia faster and showed significantly improved response to glucose challenge compared to the recipients of the same numbers of islets from 3-day-old pigs. These results are in line with the findings that islets from 7-day-old pigs showed reduced voltage-dependent K+ (Kv) channel activity and their ability to recover from post-hypoxia/reoxygenation stress. Despite more resident immune cells and immunogenic characteristics detected in islets from 7-day-old pigs compared to islets from 3-day-old pigs, the combination of anti-LFA-1 and anti-CD154 monoclonal antibodies are equally effective at preventing the rejection of islets from both age groups of pigs. Collectively, these results suggest that islets from various ages of neonatal pigs vary in yield, cellular composition, and function. Such parameters may be considered when defining the optimal pancreas donor for islet xenotransplantation studies.
Collapse
Affiliation(s)
- Hossein Arefanian
- Alberta Diabetes Institute, Ray Rajotte Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Qahir Ramji
- Alberta Diabetes Institute, Ray Rajotte Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Nancy Gupta
- Alberta Diabetes Institute, Ray Rajotte Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Aliya F. Spigelman
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Donald Grynoch
- Alberta Precision Labs, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Patrick E. MacDonald
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Thomas F. Mueller
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | | | - Ray V. Rajotte
- Alberta Diabetes Institute, Ray Rajotte Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Gina R. Rayat, ; Ray V. Rajotte,
| | - Gina R. Rayat
- Alberta Diabetes Institute, Ray Rajotte Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Gina R. Rayat, ; Ray V. Rajotte,
| |
Collapse
|
11
|
Purich K, Cai H, Yang B, Xu Z, Tessier AG, Black A, Hung RW, Boivin E, Xu B, Wu P, Zhang B, Xin D, Fallone BG, Rajotte RV, Wu Y, Rayat GR. MRI monitoring of transplanted neonatal porcine islets labeled with polyvinylpyrrolidone-coated superparamagnetic iron oxide nanoparticles in a mouse model. Xenotransplantation 2021; 29:e12720. [PMID: 34850455 DOI: 10.1111/xen.12720] [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: 05/04/2021] [Revised: 09/25/2021] [Accepted: 10/22/2021] [Indexed: 11/27/2022]
Abstract
Islet transplantation is a potential treatment option for certain patients with type 1 diabetes; however, it still faces barriers to widespread use, including the lack of tools to monitor islet grafts post-transplantation. This study investigates whether labeling neonatal porcine islets (NPI) with polyvinylpyrrolidone-coated superparamagnetic iron oxide nanoparticles (PVP-SPIO) affects their function, and whether this nanoparticle can be utilized to monitor NPI xenografts with magnetic resonance imaging (MRI) in a mouse model. In vitro, PVP-SPIO-labeled NPI in an agarose gel was visualized clearly by MRI. PVP-SPIO-labeled islets were then transplanted under the kidney capsules of immunodeficient nondiabetic and diabetic mice. All diabetic mice that received transplantation of PVP-SPIO-labeled islets reached normoglycemia. Grafts appeared as hypo-intense areas on MRI and were distinguishable from the surrounding tissues. Following injection of spleen cells from immunocompetent mice, normoglycemic recipient mice became diabetic and islet grafts showed an increase in volume, accompanied by a mixed signal on MRI. Overall, this study demonstrates that PVP-SPIO did not affect the function of NPI that PVP-SPIO-labeled islets were easily seen on MRI, and changes in MRI signals following rejection suggest a potential use of PVP-SPIO-labeled islets to monitor graft viability.
Collapse
Affiliation(s)
- Kieran Purich
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Haolei Cai
- Department of Surgery, 2nd Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Bin Yang
- Department of Surgery, 2nd Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Zhihao Xu
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anthony G Tessier
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Physics, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Adnan Black
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ryan W Hung
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
| | - Eric Boivin
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Baoyou Xu
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ping Wu
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Bo Zhang
- Department of Surgery, 2nd Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Dong Xin
- Department of Surgery, 2nd Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Biagio Gino Fallone
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Physics, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Ray V Rajotte
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Yulian Wu
- Department of Surgery, 2nd Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Gina R Rayat
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
12
|
Montanari E, Szabó L, Balaphas A, Meyer J, Perriraz-Mayer N, Pimenta J, Giraud MN, Egger B, Gerber-Lemaire S, Bühler L, Gonelle-Gispert C. Multipotent mesenchymal stromal cells derived from porcine exocrine pancreas improve insulin secretion from juvenile porcine islet cell clusters. Xenotransplantation 2021; 28:e12666. [PMID: 33538027 DOI: 10.1111/xen.12666] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/30/2020] [Accepted: 11/26/2020] [Indexed: 01/03/2023]
Abstract
Neonatal and juvenile porcine islet cell clusters (ICC) present an unlimited source for islet xenotransplantation to treat type 1 diabetes patients. We isolated ICC from pancreata of 14 days old juvenile piglets and characterized their maturation by immunofluorescence and insulin secretion assays. Multipotent mesenchymal stromal cells derived from exocrine tissue of same pancreata (pMSC) were characterized for their differentiation potential and ability to sustain ICC insulin secretion in vitro and in vivo. Isolation of ICC resulted in 142 ± 50 × 103 IEQ per pancreas. Immunofluorescence staining revealed increasing presence of insulin-positive beta cells between day 9 and 21 in culture and insulin content per 500IEC of ICC increased progressively over time from 1178.4 ± 450 µg/L to 4479.7 ± 1954.2 µg/L from day 7 to 14, P < .001. Highest glucose-induced insulin secretion by ICC was obtained at day 7 of culture and reached a fold increase of 2.9 ± 0.4 compared to basal. Expansion of adherent cells from the pig exocrine tissue resulted in a homogenous CD90+ , CD34- , and CD45- fibroblast-like cell population and differentiation into adipocytes and chondrocytes demonstrated their multipotency. Insulin release from ICC was increased in the presence of pMSC and dependent on cell-cell contact (glucose-induced fold increase: ICC alone: 1.6 ± 0.2; ICC + pMSC + contact: 3.2 ± 0.5, P = .0057; ICC + pMSC no-contact: 1.9 ± 0.3; theophylline stimulation: alone: 5.4 ± 0.7; pMSC + contact: 8.4 ± 0.9, P = .013; pMSC no-contact: 5.2 ± 0.7). After transplantation of encapsulated ICC using Ca2+ -alginate (alg) microcapsules into streptozotocin-induced diabetic and immunocompetent mice, transient normalization of glycemia was obtained up to day 7 post-transplant, whereas ICC co-encapsulated with pMSC did not improve glycemia and showed increased pericapsular fibrosis. We conclude that pMSC derived from juvenile porcine exocrine pancreas improves insulin secretion of ICC by direct cell-cell contact. For transplantation purposes, the use of pMSC to support beta-cell function will depend on the development of new anti-fibrotic polymers and/or on genetically modified pigs with lower immunogenicity.
Collapse
Affiliation(s)
- Elisa Montanari
- Surgical Research Unit, CMU-1, University Hospitals of Geneva, Geneva, Switzerland
| | - Luca Szabó
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, EPFL SB ISIC SCI-SB-SG, Lausanne, Switzerland
| | - Alexandre Balaphas
- Surgical Research Unit, CMU-1, University Hospitals of Geneva, Geneva, Switzerland
| | - Jeremy Meyer
- Surgical Research Unit, CMU-1, University Hospitals of Geneva, Geneva, Switzerland
| | - Nadja Perriraz-Mayer
- Surgical Research Unit, CMU-1, University Hospitals of Geneva, Geneva, Switzerland
| | - Joel Pimenta
- Surgical Research Unit, CMU-1, University Hospitals of Geneva, Geneva, Switzerland
| | - Marie-Noelle Giraud
- Cardiology, Dpt EMC, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Bernhard Egger
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, EPFL SB ISIC SCI-SB-SG, Lausanne, Switzerland
| | - Leo Bühler
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Carmen Gonelle-Gispert
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
13
|
Zbinden A, Urbanczyk M, Layland SL, Becker L, Marzi J, Bosch M, Loskill P, Duffy GP, Schenke-Layland K. Collagen and Endothelial Cell Coculture Improves β-Cell Functionality and Rescues Pancreatic Extracellular Matrix. Tissue Eng Part A 2020; 27:977-991. [PMID: 33023407 DOI: 10.1089/ten.tea.2020.0250] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The use of biomaterials and biomaterial functionalization is a promising approach to support pancreatic islet viability posttransplantation in an effort to reduce insulin dependence for patients afflicted with diabetes mellitus type 1. Extracellular matrix (ECM) proteins are known to impact numerous reparative functions in the body. Assessing how endogenously expressed pancreatic ECM proteins are affected by posttransplant-like hypoxic conditions may provide significant insights toward the development of tissue-engineered therapeutic strategies to positively influence β-cell survival, proliferation, and functionality. Here, we investigated the expression of three relevant groups of pancreatic ECM proteins in human native tissue, including basement membrane (BM) proteins (collagen type 4 [COL4], laminins [LAM]), proteoglycans (decorin [DCN], nidogen-1 [NID1]), and fibril-forming proteins (fibronectin [FN], collagen type 1 [COL1]). In an in vitro hypoxia model, we identified that ECM proteins were differently affected by hypoxic conditions, contributing to an overall loss of β-cell functionality. The use of a COL1 hydrogel as carrier material demonstrated a protective effect on β-cells mitigating the effect of hypoxia on proteoglycans as well as fibril-forming protein expression, supporting β-cell functionality in hypoxia. We further showed that providing endothelial cells (ECs) into the COL1 hydrogel improves β-cell response as well as the expression of relevant BM proteins. Our data show that β-cells benefit from a microenvironment composed of structure-providing COL1 with the incorporation of ECs to withstand the harsh conditions of hypoxia. Such hydrogels support β-cell survival and can serve as an initial source of ECM proteins to allow cell engraftment while preserving cell functionality posttransplantation. Impact statement Expression analysis identifies hypoxia-induced pathological changes in extracellular matrix (ECM) homeostasis as potential targets to support β-cell transplants by encapsulation in biomaterials for the treatment of diabetes mellitus. A collagen-1 hydrogel is shown to attenuate the effect of hypoxia on β-cells and their ECM expression. The functionalization of the hydrogel with endothelial cells increases the β-cell response to glucose and rescues essential basement membrane proteins.
Collapse
Affiliation(s)
- Aline Zbinden
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Max Urbanczyk
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Shannon L Layland
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Lucas Becker
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University Tübingen, Tübingen, Germany
| | - Julia Marzi
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University Tübingen, Tübingen, Germany.,NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Mariella Bosch
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Peter Loskill
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany.,Fraunhofer IGB, Stuttgart, Germany
| | - Garry P Duffy
- Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Katja Schenke-Layland
- Department of Bioengineering, Eberhard Karls University Tübingen, Tübingen, Germany.,Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University Tübingen, Tübingen, Germany.,NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany.,Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland.,Cardiovascular Research Laboratories, Department of Medicine/Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
14
|
Salama BF, Seeberger KL, Korbutt GS. Fibrin supports subcutaneous neonatal porcine islet transplantation without the need for pre‐vascularization. Xenotransplantation 2019; 27:e12575. [DOI: 10.1111/xen.12575] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/14/2019] [Accepted: 11/20/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Bassem F. Salama
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Department of Surgery University of Alberta Edmonton Alberta Canada
| | - Karen L. Seeberger
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Department of Surgery University of Alberta Edmonton Alberta Canada
| | - Gregory S. Korbutt
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Department of Surgery University of Alberta Edmonton Alberta Canada
| |
Collapse
|
15
|
Navigating Two Roads to Glucose Normalization in Diabetes: Automated Insulin Delivery Devices and Cell Therapy. Cell Metab 2019; 29:545-563. [PMID: 30840911 DOI: 10.1016/j.cmet.2019.02.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 12/23/2022]
Abstract
Incredible strides have been made since the discovery of insulin almost 100 years ago. Insulin formulations have improved dramatically, glucose levels can be measured continuously, and recently first-generation biomechanical "artificial pancreas" systems have been approved by regulators around the globe. However, still only a small fraction of patients with diabetes achieve glycemic goals. Replacement of insulin-producing cells via transplantation shows significant promise, but is limited in application due to supply constraints (cadaver-based) and the need for chronic immunosuppression. Over the past decade, significant progress has been made to address these barriers to widespread implementation of a cell therapy. Can glucose levels in people with diabetes be normalized with artificial pancreas systems or via cell replacement approaches? Here we review the road ahead, including the challenges and opportunities of both approaches.
Collapse
|
16
|
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: 2.0] [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
|
17
|
Oxygenation strategies for encapsulated islet and beta cell transplants. Adv Drug Deliv Rev 2019; 139:139-156. [PMID: 31077781 DOI: 10.1016/j.addr.2019.05.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 04/19/2019] [Accepted: 05/04/2019] [Indexed: 02/06/2023]
Abstract
Human allogeneic islet transplantation (ITx) is emerging as a promising treatment option for qualified patients with type 1 diabetes. However, widespread clinical application of allogeneic ITx is hindered by two critical barriers: the need for systemic immunosuppression and the limited supply of human islet tissue. Biocompatible, retrievable immunoisolation devices containing glucose-responsive insulin-secreting tissue may address both critical barriers by enabling the more effective and efficient use of allogeneic islets without immunosuppression in the near-term, and ultimately the use of a cell source with a virtually unlimited supply, such as human stem cell-derived β-cells or xenogeneic (porcine) islets with minimal or no immunosuppression. However, even though encapsulation methods have been developed and immunoprotection has been successfully tested in small and large animal models and to a limited extent in proof-of-concept clinical studies, the effective use of encapsulation approaches to convincingly and consistently treat diabetes in humans has yet to be demonstrated. There is increasing consensus that inadequate oxygen supply is a major factor limiting their clinical translation and routine implementation. Poor oxygenation negatively affects cell viability and β-cell function, and the problem is exacerbated with the high-density seeding required for reasonably-sized clinical encapsulation devices. Approaches for enhanced oxygen delivery to encapsulated tissues in implantable devices are therefore being actively developed and tested. This review summarizes fundamental aspects of islet microarchitecture and β-cell physiology as well as encapsulation approaches highlighting the need for adequate oxygenation; it also evaluates existing and emerging approaches for enhanced oxygen delivery to encapsulation devices, particularly with the advent of β-cell sources from stem cells that may enable the large-scale application of this approach.
Collapse
|
18
|
Abstract
β cell replacement with either pancreas or islet transplantation has progressed immensely over the last decades with current 1- and 5-year insulin independence rates of approximately 85% and 50%, respectively. Recent advances are largely attributed to improvements in immunosuppressive regimen, donor selection, and surgical technique. However, both strategies are compromised by a scarce donor source. Xenotransplantation offers a potential solution by providing a theoretically unlimited supply of islets, but clinical application has been limited by concerns for a potent immune response against xenogeneic tissue. β cell clusters derived from embryonic or induced pluripotent stem cells represent another promising unlimited source of insulin producing cells, but clinical application is pending further advances in the function of the β cell like clusters. Exciting developments and rapid progress in all areas of β cell replacement prompted a lively debate by members of the young investigator committee of the International Pancreas and Islet Transplant Association at the 15th International Pancreas and Islet Transplant Association Congress in Melbourne and at the 26th international congress of The Transplant Society in Hong Kong. This international group of young investigators debated which modality of β cell replacement would predominate the landscape in 10 years, and their arguments are summarized here.
Collapse
|
19
|
Smith KE, Purvis WG, Davis MA, Min CG, Cooksey AM, Weber CS, Jandova J, Price ND, Molano DS, Stanton JB, Kelly AC, Steyn LV, Lynch RM, Limesand SW, Alexander M, Lakey JRT, Seeberger K, Korbutt GS, Mueller KR, Hering BJ, McCarthy FM, Papas KK. In vitro characterization of neonatal, juvenile, and adult porcine islet oxygen demand, β-cell function, and transcriptomes. Xenotransplantation 2018; 25:e12432. [PMID: 30052287 DOI: 10.1111/xen.12432] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/20/2018] [Accepted: 05/24/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND There is currently a shortage of human donor pancreata which limits the broad application of islet transplantation as a treatment for type 1 diabetes. Porcine islets have demonstrated potential as an alternative source, but a study evaluating islets from different donor ages under unified protocols has yet to be conducted. METHODS Neonatal porcine islets (NPI; 1-3 days), juvenile porcine islets (JPI; 18-21 days), and adult porcine islets (API; 2+ years) were compared in vitro, including assessments of oxygen consumption rate, membrane integrity determined by FDA/PI staining, β-cell proliferation, dynamic glucose-stimulated insulin secretion, and RNA sequencing. RESULTS Oxygen consumption rate normalized to DNA was not significantly different between ages. Membrane integrity was age dependent, and API had the highest percentage of intact cells. API also had the highest glucose-stimulated insulin secretion response during a dynamic insulin secretion assay and had 50-fold higher total insulin content compared to NPI and JPI. NPI and JPI had similar glucose responsiveness, β-cell percentage, and β-cell proliferation rate. Transcriptome analysis was consistent with physiological assessments. API transcriptomes were enriched for cellular metabolic and insulin secretory pathways, while NPI exhibited higher expression of genes associated with proliferation. CONCLUSIONS The oxygen demand, membrane integrity, β-cell function and proliferation, and transcriptomes of islets from API, JPI, and NPI provide a comprehensive physiological comparison for future studies. These assessments will inform the optimal application of each age of porcine islet to expand the availability of islet transplantation.
Collapse
Affiliation(s)
- Kate E Smith
- Department of Physiological Sciences, University of Arizona, Tucson, AZ, USA.,Department of Surgery, University of Arizona, Tucson, AZ, USA
| | | | - Melissa A Davis
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Catherine G Min
- Department of Physiological Sciences, University of Arizona, Tucson, AZ, USA.,Department of Surgery, University of Arizona, Tucson, AZ, USA
| | - Amanda M Cooksey
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Craig S Weber
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Jana Jandova
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | | | - Diana S Molano
- Department of Surgery, University of Arizona, Tucson, AZ, USA
| | | | - Amy C Kelly
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Leah V Steyn
- Department of Surgery, University of Arizona, Tucson, AZ, USA
| | - Ronald M Lynch
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Sean W Limesand
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Michael Alexander
- Department of Surgery, University of California-Irvine, Orange, CA, USA
| | | | - Karen Seeberger
- Department of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, AL, Canada
| | - Gregory S Korbutt
- Department of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, AL, Canada
| | - Kate R Mueller
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Bernhard J Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Fiona M McCarthy
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | | |
Collapse
|
20
|
Smith KE, Johnson RC, Papas KK. Update on cellular encapsulation. Xenotransplantation 2018; 25:e12399. [DOI: 10.1111/xen.12399] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 03/27/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Kate E. Smith
- Department of Physiological Sciences; University of Arizona; Tucson AZ USA
- Department of Surgery; University of Arizona; Tucson AZ USA
| | | | | |
Collapse
|
21
|
Li C, Yang B, Xu Z, Boivin E, Black M, Huang W, Xu B, Wu P, Zhang B, Li X, Chen K, Wu Y, Rayat GR. Protective effect of cyanidin-3-O-glucoside on neonatal porcine islets. J Endocrinol 2017; 235:237-249. [PMID: 28931557 DOI: 10.1530/joe-17-0141] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 09/20/2017] [Indexed: 12/13/2022]
Abstract
Oxidative stress is a major cause of islet injury and dysfunction during isolation and transplantation procedures. Cyanidin-3-O-glucoside (C3G), which is present in various fruits and vegetables especially in Chinese bayberry, shows a potent antioxidant property. In this study, we determined whether C3G could protect neonatal porcine islets (NPI) from reactive oxygen species (H2O2)-induced injury in vitro and promote the function of NPI in diabetic mice. We found that C3G had no deleterious effect on NPI and that C3G protected NPI from damage induced by H2O2 Significantly higher hemeoxygenase-1 (HO1) gene expression was detected in C3G-treated NPI compared to untreated islets before and after transplantation (P < 0.05). Western blot analysis showed a significant increase in the levels of phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2) and phosphatidylinositol 3-kinase (PI3K/Akt) proteins in C3G-treated NPI compared to untreated islets. C3G induced the nuclear translocation of nuclear erythroid 2-related factor 2 (NRF2) and the significant elevation of HO1 protein. Recipients of C3G-treated NPI with or without C3G-supplemented drinking water achieved normoglycemia earlier compared to recipients of untreated islets. Mice that received C3G-treated islets with or without C3G-supplemented water displayed significantly lower blood glucose levels at 5-10 weeks post-transplantation compared to mice that received untreated islets. Mice that received C3G-treated NPI and C3G-supplemented drinking water had significantly (P < 0.05) lower blood glucose levels at 7 and 8 weeks post-transplantation compared to mice that received C3G-treated islets. These findings suggest that C3G has a beneficial effect on NPI through the activation of ERK1/2- and PI3K/AKT-induced NRF2-mediated HO1 signaling pathway.
Collapse
Affiliation(s)
- Chao Li
- Department of SurgeryThe Second Affiliated Hospital of Zhejiang University, Hanghzou, Zhejiang, China
| | - Bin Yang
- Department of SurgeryThe Second Affiliated Hospital of Zhejiang University, Hanghzou, Zhejiang, China
| | - Zhihao Xu
- Department of SurgeryRay Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Eric Boivin
- Department of SurgeryRay Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mazzen Black
- Department of SurgeryRay Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Wenlong Huang
- Department of SurgeryRay Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Baoyou Xu
- Department of SurgeryRay Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ping Wu
- Department of SurgeryRay Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Bo Zhang
- Department of SurgeryThe Second Affiliated Hospital of Zhejiang University, Hanghzou, Zhejiang, China
| | - Xian Li
- Department of HorticultureCollege of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kunsong Chen
- Department of HorticultureCollege of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yulian Wu
- Department of SurgeryThe Second Affiliated Hospital of Zhejiang University, Hanghzou, Zhejiang, China
| | - Gina R Rayat
- Department of SurgeryRay Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
22
|
Ma X, Yang C, Zhang J, Wang J, Li W, Xu C, Rong P, Ye B, Wu M, Jiang J, Yi S, Wang W. Culturing with modified EGM2 medium enhances porcine neonatal islet-like cell clusters resistance to apoptosis in islet xenotransplantation. Xenotransplantation 2017; 25. [PMID: 29131417 DOI: 10.1111/xen.12358] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/12/2017] [Accepted: 09/12/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Neonatal pig islet-like cell clusters (NICC) are an attractive source of insulin-producing tissue for potential transplantation treatment of type 1 diabetic patients. However, a considerable loss of NICC after their transplantation due to apoptosis resulted from islet isolation and instant blood-mediated inflammatory reaction remains to be overcome. METHODS EGM2 medium depleted with hydrocortisone and supplemented with 50 mmol/L isobutylmethylxanthine, 10 mmol/L nicotinamide, and 10 mmol/L glucose was used to culture NICC at day 1, the day after isolation and changed every other day. NICC cultured with EGM2 or control Ham's F-10 medium were collected at day 7 of culture for the following assays. The viability of NICC was evaluated by AO/EB staining and FACS. Static assay and oxygen consumption rate analysis were performed to assess the function of NICC. Insulin and glucagon gene expression were measured by real-time PCR. Tubing loops model and TUNEL assay were performed to confirm the apoptosis-resistant ability of NICC cultured with modified EGM2 medium. Serum starvation and hypoxia treatment were used to test the tolerant capability of NICC in the microenvironment of hypoxia/nutrient deficiency in vitro. The molecules involved in apoptosis pathways in NICC were analyzed by Western blotting. RESULTS Compared with Ham's F-10 medium, culturing NICC with EGM2 medium led to increased number and viability of NICC with higher stimulation index, upregulated gene expression of both insulin and glucagon, and enhanced mitochondria function. Furthermore, fewer modified EGM2 medium cultured NICC were found under apoptosis when evaluated in an in vitro tubing loop model of IBMIR. Moreover, EGM2 medium cultured NICC demonstrated much less apoptotic cells under either serum starvation or hypoxia condition than their Ham's F-10 medium cultured counterparts. The enhanced capability of EGM2 cultured NICC to resist apoptosis was associated with their elevated protein levels of anti-apoptotic Bcl-2 family member Mcl-1. CONCLUSION Culturing NICC with EGM2 provides a simple and effective approach not only to increase NICC yield, viability, and maturation but also to enhance their resistance to apoptosis to preserve the initial graft mass for successful islet xenotransplantation.
Collapse
Affiliation(s)
- Xiaoqian Ma
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Cejun Yang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Juan Zhang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Jia Wang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Wei Li
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Chang Xu
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Pengfei Rong
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Bin Ye
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| | - Minghua Wu
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianhui Jiang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Shounan Yi
- Center for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Wei Wang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Engineering and Technology Research Center for Xenotransplantation of Human Province, Changsha, Hunan, China
| |
Collapse
|
23
|
|
24
|
Abraham S, Kuppan P, Raj S, Salama B, Korbutt GS, Montemagno CD. Developing Hybrid Polymer Scaffolds Using Peptide Modified Biopolymers for Cell Implantation. ACS Biomater Sci Eng 2017; 3:2215-2222. [DOI: 10.1021/acsbiomaterials.7b00383] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sinoj Abraham
- IngenuityLab, National Institute for Nanotechnology, 11421 Saskatchewan Drive NW, Edmonton, Alberta T6G 2M9, Canada
| | | | - Shammy Raj
- IngenuityLab, National Institute for Nanotechnology, 11421 Saskatchewan Drive NW, Edmonton, Alberta T6G 2M9, Canada
| | | | | | - Carlo D. Montemagno
- IngenuityLab, National Institute for Nanotechnology, 11421 Saskatchewan Drive NW, Edmonton, Alberta T6G 2M9, Canada
| |
Collapse
|
25
|
LEA29Y expression in transgenic neonatal porcine islet-like cluster promotes long-lasting xenograft survival in humanized mice without immunosuppressive therapy. Sci Rep 2017; 7:3572. [PMID: 28620237 PMCID: PMC5472587 DOI: 10.1038/s41598-017-03913-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/05/2017] [Indexed: 02/02/2023] Open
Abstract
Genetically engineered pigs are a promising source for islet cell transplantation in type 1 diabetes, but the strong human anti-pig immune response prevents its successful clinical application. Here we studied the efficacy of neonatal porcine islet-like cell clusters (NPICCs) overexpressing LEA29Y, a high-affinity variant of the T cell co-stimulation inhibitor CTLA-4Ig, to engraft and restore normoglycemia after transplantation into streptozotocin-diabetic NOD-SCID IL2rγ−/− (NSG) mice stably reconstituted with a human immune system. Transplantation of INSLEA29Y expressing NPICCs resulted in development of normal glucose tolerance (70.4%) and long-term maintenance of normoglycemia without administration of immunosuppressive drugs. All animals transplanted with wild-type NPICCs remained diabetic. Immunohistological examinations revealed a strong peri- and intragraft infiltration of wild-type NPICCs with human CD45+ immune cells consisting of predominantly CD4+ and CD8+ lymphocytes and some CD68+ macrophages and FoxP3+ regulatory T cells. Significantly less infiltrating lymphocytes and only few macrophages were observed in animals transplanted with INSLEA29Y transgenic NPICCs. This is the first study providing evidence that beta cell-specific LEA29Y expression is effective for NPICC engraftment in the presence of a humanized immune system and it has a long-lasting protective effect on inhibition of human anti-pig xenoimmunity. Our findings may have important implications for the development of a low-toxic protocol for porcine islet transplantation in patients with type 1 diabetes.
Collapse
|
26
|
Hayward JA, Ellis CE, Seeberger K, Lee T, Salama B, Mulet-Sierra A, Kuppan P, Adesida A, Korbutt GS. Cotransplantation of Mesenchymal Stem Cells With Neonatal Porcine Islets Improve Graft Function in Diabetic Mice. Diabetes 2017; 66:1312-1321. [PMID: 28246290 DOI: 10.2337/db16-1068] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 02/19/2017] [Indexed: 11/13/2022]
Abstract
Mesenchymal stem cells (MSCs) possess immunoregulatory, anti-inflammatory, and proangiogenic properties and, therefore, have the potential to improve islet engraftment and survival. We assessed the effect human bone marrow-derived MSCs have on neonatal porcine islets (NPIs) in vitro and determined islet engraftment and metabolic outcomes when cotransplanted in a mouse model. NPIs cocultured with MSCs had greater cellular insulin content and increased glucose-stimulated insulin secretion. NPIs were cotransplanted with or without MSCs in diabetic B6.129S7-Rag1tm1Mom/J mice. Blood glucose and weight were monitored until reversal of diabetes; mice were then given an oral glucose tolerance test. Islet grafts were assessed for the degree of vascularization and total cellular insulin content. Cotransplantation of NPIs and MSCs resulted in significantly earlier normoglycemia and vascularization, improved glucose tolerance, and increased insulin content. One experiment conducted with MSCs from a donor with an autoimmune disorder had no positive effects on transplant outcomes. Cotransplantation of human MSCs with NPIs demonstrated a beneficial metabolic effect likely as a result of earlier islet vascularization and improved islet engraftment. In addition, donor pathology of MSCs can influence the functional capacity of MSCs.
Collapse
Affiliation(s)
- Julie A Hayward
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Cara E Ellis
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Karen Seeberger
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Timothy Lee
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Bassem Salama
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | | | - Purushothaman Kuppan
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Adetola Adesida
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Gregory S Korbutt
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Diabetes is medical and social burden affecting millions around the world. Despite intensive therapy, insulin fails to maintain adequate glucose homeostasis and often results in episodes of hypoglycemic unawareness. Islet transplantation is a propitious replacement therapy, and incremental improvements in islet isolation and immunosuppressive drugs have made this procedure a feasible option. Shortage of donors, graft loss, and toxic immunosuppressive agents are few of many hurdles against making human allogenic islet transplantation a routine procedure. RECENT FINDINGS Xenografts-especially pig islets-offer a logical alternative source for islets. Current preclinical studies have revealed problems such as optimal islet source, zoonosis, and immune rejection. These issues are slowing clinical application. Genetically modified pigs, encapsulation devices, and new immune-suppressive regimens can confer graft protection. In addition, extrahepatic transplant sites are showing promising results. Notwithstanding few approved clinical human trials, and available data from non-human primates, recent reports indicate that porcine islets are closer to be the promising solution to cure diabetes.
Collapse
Affiliation(s)
- Bassem F Salama
- Department of Surgery, University of Alberta, 5.002 Li Ka Shing Bldg, 8602 112 Street, Edmonton, AB, T6G 2E1, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Gregory S Korbutt
- Department of Surgery, University of Alberta, 5.002 Li Ka Shing Bldg, 8602 112 Street, Edmonton, AB, T6G 2E1, Canada.
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
28
|
Muthyala S, Safley S, Gordan K, Barber G, Weber C, Sambanis A. The effect of hypoxia on free and encapsulated adult porcine islets-an in vitro study. Xenotransplantation 2016; 24. [PMID: 28247506 DOI: 10.1111/xen.12275] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 08/18/2016] [Accepted: 09/07/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Adult porcine islets (APIs) constitute a promising alternative to human islets in treating type 1 diabetes. The intrahepatic site has been used in preclinical primate studies of API xenografts; however, an estimated two-thirds of donor islets are destroyed after intraportal infusion due to a number of factors, including the instant blood-mediated inflammatory reaction (IBMIR), immunosuppressant toxicity, and poor reestablishment of extracellular matrix connections. Intraperitoneal (ip) transplantation of non-vascularized encapsulated islets offers several advantages over intrahepatic transplantation of free islets, including avoidance of IBMIR, immunoprotection, accommodation of a larger graft volume, and reduced risk of hemorrhage. However, there exists evidence that the peritoneal site is hypoxic, which likely impedes islet function. METHODS We tested the effect of hypoxia (2%-5% oxygen or pO2 : 15.2-38.0 mm Hg) on free and encapsulated APIs over a period of 6 days in culture. Free and encapsulated APIs under normoxia served as controls. Islet viability was evaluated with a viability/cytotoxicity assay using calcein AM and ethidium bromide on days 1, 3, and 6 of culture. Alamar blue assay was used to measure the metabolic activity on days 1 and 6. Insulin in spent medium was assayed by ELISA on days 1 and 6. RESULTS Viability staining indicated that free islet clusters lost their integrity and underwent severe necrosis under hypoxia; encapsulated islets remained intact, even when they began to undergo necrosis. Under hypoxia, the metabolic activity and insulin secretion (normalized to metabolic activity) of both free and encapsulated islets decreased relative to islets cultured under normoxic conditions. CONCLUSIONS Hypoxia (2%-5% oxygen or pO2 : 15.2-38.0 mm Hg) affects the viability, metabolic activity, and insulin secretion of both free and encapsulated APIs over a six-day culture period. Encapsulation augments islet integrity under hypoxia, but it does not prevent loss of viability, metabolic activity, or insulin secretion.
Collapse
Affiliation(s)
- Sudhakar Muthyala
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Susan Safley
- Department of Surgery, Emory University, Atlanta, GA, USA
| | - Kereen Gordan
- Department of Surgery, Emory University, Atlanta, GA, USA
| | - Graham Barber
- Department of Surgery, Emory University, Atlanta, GA, USA
| | - Collin Weber
- Department of Surgery, Emory University, Atlanta, GA, USA
| | - Athanassios Sambanis
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,W.M. Keck Foundation, Los Angeles, CA, USA
| |
Collapse
|
29
|
Cooper DKC, Ezzelarab MB, Hara H, Iwase H, Lee W, Wijkstrom M, Bottino R. The pathobiology of pig-to-primate xenotransplantation: a historical review. Xenotransplantation 2016; 23:83-105. [PMID: 26813438 DOI: 10.1111/xen.12219] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/22/2015] [Indexed: 12/16/2022]
Abstract
The immunologic barriers to successful xenotransplantation are related to the presence of natural anti-pig antibodies in humans and non-human primates that bind to antigens expressed on the transplanted pig organ (the most important of which is galactose-α1,3-galactose [Gal]), and activate the complement cascade, which results in rapid destruction of the graft, a process known as hyperacute rejection. High levels of elicited anti-pig IgG may develop if the adaptive immune response is not prevented by adequate immunosuppressive therapy, resulting in activation and injury of the vascular endothelium. The transplantation of organs and cells from pigs that do not express the important Gal antigen (α1,3-galactosyltransferase gene-knockout [GTKO] pigs) and express one or more human complement-regulatory proteins (hCRP, e.g., CD46, CD55), when combined with an effective costimulation blockade-based immunosuppressive regimen, prevents early antibody-mediated and cellular rejection. However, low levels of anti-non-Gal antibody and innate immune cells and/or platelets may initiate the development of a thrombotic microangiopathy in the graft that may be associated with a consumptive coagulopathy in the recipient. This pathogenic process is accentuated by the dysregulation of the coagulation-anticoagulation systems between pigs and primates. The expression in GTKO/hCRP pigs of a human coagulation-regulatory protein, for example, thrombomodulin, is increasingly being associated with prolonged pig graft survival in non-human primates. Initial clinical trials of islet and corneal xenotransplantation are already underway, and trials of pig kidney or heart transplantation are anticipated within the next few years.
Collapse
Affiliation(s)
- David K C Cooper
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mohamed B Ezzelarab
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hidetaka Hara
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hayato Iwase
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Whayoung Lee
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Martin Wijkstrom
- The Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rita Bottino
- Institute for Cellular Therapeutics, Allegheny-Singer Research Institute, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Song S, Roy S. Progress and challenges in macroencapsulation approaches for type 1 diabetes (T1D) treatment: Cells, biomaterials, and devices. Biotechnol Bioeng 2016; 113:1381-402. [PMID: 26615050 DOI: 10.1002/bit.25895] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/30/2015] [Accepted: 11/24/2015] [Indexed: 12/15/2022]
Abstract
Macroencapsulation technology has been an attractive topic in the field of treatment for Type 1 diabetes due to mechanical stability, versatility, and retrievability of the macro-capsule design. Macro-capsules can be categorized into extravascular and intravascular devices, in which solute transport relies either on diffusion or convection, respectively. Failure of macroencapsulation strategies can be due to limited regenerative capacity of the encased insulin-producing cells, sub-optimal performance of encapsulation biomaterials, insufficient immunoisolation, excessive blood thrombosis for vascular perfusion devices, and inadequate modes of mass transfer to support cell viability and function. However, significant technical advancements have been achieved in macroencapsulation technology, namely reducing diffusion distance for oxygen and nutrients, using pro-angiogenic factors to increase vascularization for islet engraftment, and optimizing membrane permeability and selectivity to prevent immune attacks from host's body. This review presents an overview of existing macroencapsulation devices and discusses the advances based on tissue-engineering approaches that will stimulate future research and development of macroencapsulation technology. Biotechnol. Bioeng. 2016;113: 1381-1402. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shang Song
- Department of Bioengineering and Therapeutic Sciences, University of California-San Francisco, San Francisco, California 94158
| | - Shuvo Roy
- Department of Bioengineering and Therapeutic Sciences, University of California-San Francisco, San Francisco, California 94158.
| |
Collapse
|
31
|
Quiskamp N, Bruin JE, Kieffer TJ. Differentiation of human pluripotent stem cells into β-cells: Potential and challenges. Best Pract Res Clin Endocrinol Metab 2015; 29:833-47. [PMID: 26696513 DOI: 10.1016/j.beem.2015.10.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) hold great potential as the basis for cell-based therapies of degenerative diseases, including diabetes. Current insulin-based therapies for diabetes do not prevent hyperglycaemia or the associated long-term organ damage. While transplantation of pancreatic islets can achieve insulin independence and improved glycemic control, it is limited by donor tissue scarcity, challenges of purifying islets from the pancreas, and the need for immunosuppression to prevent rejection of transplants. Large-scale production of β-cells from stem cells is a promising alternative. Recent years have seen considerable progress in the optimization of in vitro differentiation protocols to direct hESCs/iPSCs into mature insulin-secreting β-cells and clinical trials are now under way to test the safety and efficiency of hESC-derived pancreatic progenitor cells in patients with type 1 diabetes. Here, we discuss key milestones leading up to these trials in addition to recent developments and challenges for hESC/iPSC-based diabetes therapies and disease modeling.
Collapse
Affiliation(s)
- Nina Quiskamp
- Laboratory of Molecular and Cellular Medicine, Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| | - Jennifer E Bruin
- Laboratory of Molecular and Cellular Medicine, Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine, Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
32
|
Wolf-van Buerck L, Schuster M, Baehr A, Mayr T, Guethoff S, Abicht J, Reichart B, Nam-Apostolopoulos YC, Klymiuk N, Wolf E, Seissler J. Engraftment and reversal of diabetes after intramuscular transplantation of neonatal porcine islet-like clusters. Xenotransplantation 2015; 22:443-50. [PMID: 26490671 DOI: 10.1111/xen.12201] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 09/16/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Intraportal infusion is currently the method of choice for clinical islet cell transplantation but suffers from poor efficacy. As the liver may not represent an optimal transplantation site for Langerhans islets, we examined the potential of neonatal porcine islet-like clusters (NPICCs) to engraft in skeletal muscle as an alternative transplantation site. METHODS Neonatal porcine islet-like clusters were isolated from 2- to 5-day-old piglets and either transplanted under the kidney capsule (s.k.) or injected into the lower hindlimb muscle (i.m.) of streptozotocin-diabetic NOD-SCID IL2rγ(-/-) (NSG) mice. Survival, vascularization, maturation, and functional activity were analyzed by intraperitoneal glucose tolerance testing and immunohistochemical analyses. RESULTS Intramuscular transplantation of NPICCs resulted in development of normoglycemia and restored glucose homeostasis. Time to reversal of diabetes and glucose tolerance (AUC glucose and AUC insulin) did not significantly differ as compared to s.k. transplantation. Intramuscular grafts exhibited rapid neovascularization and graft composition with cytokeratin-positive ductal cells and beta cells at post-transplant weeks 2 and 8 and after establishment of normoglycemia was comparable in both groups. CONCLUSIONS Intramuscular injection represents a minimally invasive but efficient alternative for transplantation of NPICCs and, thus, offers an attractive alternative site for xenotransplantation approaches. These findings may have important implications for improving the outcome and the monitoring of pig islet xenotransplantation.
Collapse
Affiliation(s)
- Lelia Wolf-van Buerck
- Medizinische Klinik und Poliklinik IV-Campus Innenstadt, Diabetes Zentrum, Klinikum der Ludwig-Maximilians-Universität, München, Germany.,Transregio Collaborative Research Center 127, Ludwig-Maximilians-Universität, München, Germany
| | - Marion Schuster
- Medizinische Klinik und Poliklinik IV-Campus Innenstadt, Diabetes Zentrum, Klinikum der Ludwig-Maximilians-Universität, München, Germany.,Transregio Collaborative Research Center 127, Ludwig-Maximilians-Universität, München, Germany
| | - Andrea Baehr
- Transregio Collaborative Research Center 127, Ludwig-Maximilians-Universität, München, Germany.,Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität, München, Germany
| | - Tanja Mayr
- Transregio Collaborative Research Center 127, Ludwig-Maximilians-Universität, München, Germany.,Department of Cardiac Surgery, Ludwig-Maximilians-Universität, München, Germany
| | - Sonja Guethoff
- Transregio Collaborative Research Center 127, Ludwig-Maximilians-Universität, München, Germany.,Department of Cardiac Surgery, Ludwig-Maximilians-Universität, München, Germany
| | - Jan Abicht
- Transregio Collaborative Research Center 127, Ludwig-Maximilians-Universität, München, Germany.,Department of Anesthesiology, Ludwig-Maximilians-Universität, München, Germany
| | - Bruno Reichart
- Transregio Collaborative Research Center 127, Ludwig-Maximilians-Universität, München, Germany
| | | | - Nikolai Klymiuk
- Transregio Collaborative Research Center 127, Ludwig-Maximilians-Universität, München, Germany.,Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität, München, Germany
| | - Eckhard Wolf
- Transregio Collaborative Research Center 127, Ludwig-Maximilians-Universität, München, Germany.,Chair for Molecular Animal Breeding and Biotechnology and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität, München, Germany
| | - Jochen Seissler
- Medizinische Klinik und Poliklinik IV-Campus Innenstadt, Diabetes Zentrum, Klinikum der Ludwig-Maximilians-Universität, München, Germany.,Transregio Collaborative Research Center 127, Ludwig-Maximilians-Universität, München, Germany
| |
Collapse
|
33
|
Ellis C, Lyon JG, Korbutt GS. Optimization and Scale-up Isolation and Culture of Neonatal Porcine Islets: Potential for Clinical Application. Cell Transplant 2015; 25:539-47. [PMID: 26377964 DOI: 10.3727/096368915x689451] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
One challenge that must be overcome to allow transplantation of neonatal porcine islets (NPIs) to become a clinical reality is defining a reproducible and scalable protocol for the efficient preparation of therapeutic quantities of clinical grade NPIs. In our standard protocol, we routinely isolate NPIs from a maximum of four pancreases, requiring tissue culture in 16 Petri dishes (four per pancreas) in Ham's F10 and bovine serum albumin (BSA). We have now developed a scalable and technically simpler protocol that allows us to isolate NPIs from a minimum of 12 pancreases at a time by employing automated tissue chopping, collagenase digestion in a single vessel, and tissue culture/media changes in 75% fewer Petri dishes. For culture, BSA is replaced with human serum albumin and supplemented with Z-VAD-FMK general caspase inhibitor and a protease inhibitor cocktail. The caspase inhibitor was added to the media for only the first 90 min of culture. NPIs isolated using the scalable protocol had significantly more cellular insulin recovered (56.9 ± 1.4 µg) when compared to the standard protocol (15.0 ± 0.5 µg; p < 0.05). Compared to our standard protocol, recovery of β-cells (6.0 × 10(6) ± 0.2 vs. 10.0 × 10(6) ± 0.4; p < 0.05) and islet equivalents (35,135 ± 186 vs. 41,810 ± 226; p < 0.05) was significantly higher using the scalable protocol. During a static glucose stimulation assay, the SI of islets isolated by the standard protocol were significantly lower than the scale-up protocol (4.3 ± 0.2 vs. 5.5 ± 0.1; p < 0.05). Mice transplanted with NPIs using the scalable protocol had significantly lower blood glucose levels than the mice that receiving NPIs from the standard protocol (p < 0.01) and responded significantly better to a glucose tolerance test. Based on the above findings, this improved simpler scalable protocol is a significantly more efficient means for preparing therapeutic quantities of clinical grade NPIs.
Collapse
Affiliation(s)
- Cara Ellis
- Department of Surgery, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
34
|
Nagaraju S, Bertera S, Tanaka T, Hara H, Rayat GR, Wijkstrom M, Ayares D, Trucco M, Cooper DKC, Bottino R. In vitro exposure of pig neonatal isletlike cell clusters to human blood. Xenotransplantation 2015; 22:317-24. [PMID: 26179209 DOI: 10.1111/xen.12178] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/07/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Pig islet grafts have been successful in treating diabetes in animal models. One remaining question is whether neonatal pig isletlike cell clusters (NICC) are resistant to the early loss of islets from the instant blood-mediated inflammatory reaction (IBMIR). METHODS Neonatal isletlike cell clusters were harvested from three groups of piglets-(i) wild-type (genetically unmodified), (ii) α1,3-galactosyltransferase gene-knockout (GTKO)/CD46, and (iii) GTKO/CD46/CD39. NICC samples were mixed with human blood in vitro, and the following measurements were made-antibody binding; complement activation; speed of islet-induced coagulation; C-peptide; glutamic acid decarboxylase (GAD65) release; viability. RESULTS Time to coagulation and viability were both reduced in all groups compared to freshly drawn non-anticoagulated human blood and autologous combinations, respectively. Antibody binding to the NICC occurred in all groups. CONCLUSIONS Neonatal isletlike cell clusters were subject to humoral injury with no difference associated to their genetic characteristics.
Collapse
Affiliation(s)
- Santosh Nagaraju
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Suzanne Bertera
- Division of Immunogenetics, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Takayuki Tanaka
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Hidetaka Hara
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Gina R Rayat
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Martin Wijkstrom
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Massimo Trucco
- Division of Immunogenetics, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - David K C Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rita Bottino
- Division of Immunogenetics, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
35
|
Nagaraju S, Bottino R, Wijkstrom M, Trucco M, Cooper DKC. Islet xenotransplantation: what is the optimal age of the islet-source pig? Xenotransplantation 2014; 22:7-19. [DOI: 10.1111/xen.12130] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 06/26/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Santosh Nagaraju
- Thomas E. Starzl Transplantation Institute; University of Pittsburgh Medical Center; Pittsburgh PA USA
| | - Rita Bottino
- Division of Immunogenetics; Department of Pediatrics; Children's Hospital of Pittsburgh; University of Pittsburgh Medical Center; Pittsburgh PA USA
| | - Martin Wijkstrom
- Thomas E. Starzl Transplantation Institute; University of Pittsburgh Medical Center; Pittsburgh PA USA
| | - Massimo Trucco
- Division of Immunogenetics; Department of Pediatrics; Children's Hospital of Pittsburgh; University of Pittsburgh Medical Center; Pittsburgh PA USA
| | - David K. C. Cooper
- Thomas E. Starzl Transplantation Institute; University of Pittsburgh Medical Center; Pittsburgh PA USA
| |
Collapse
|
36
|
Loganathan G, Graham M, Spizzo T, Tiwari M, Lockridge A, Soltani S, Wilhelm J, Balamurugan A, Hering B. Pretreatment of Donor Pigs With a Diet Rich in Soybean Oil Increases the Yield of Isolated Islets. Transplant Proc 2014; 46:1945-9. [DOI: 10.1016/j.transproceed.2014.05.078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
37
|
Krishnan R, Arora RP, Alexander M, White SM, Lamb MW, Foster CE, Choi B, Lakey JRT. Noninvasive evaluation of the vascular response to transplantation of alginate encapsulated islets using the dorsal skin-fold model. Biomaterials 2013; 35:891-8. [PMID: 24176195 DOI: 10.1016/j.biomaterials.2013.10.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/02/2013] [Indexed: 01/25/2023]
Abstract
Alginate encapsulation reduces the risk of transplant rejection by evading immune-mediated cell injury and rejection; however, poor vascular perfusion results in graft failure. Since existing imaging models are incapable of quantifying the vascular response to biomaterial implants after transplantation, in this study, we demonstrate the use of in vivo laser speckle imaging (LSI) and wide-field functional imaging (WiFI) to monitor the microvascular environment surrounding biomaterial implants. The vascular response to two islet-containing biomaterial encapsulation devices, alginate microcapsules and a high-guluronate alginate sheet, was studied and compared after implantation into the mouse dorsal window chamber (N = 4 per implant group). Images obtained over a 14-day period using LSI and WiFI were analyzed using algorithms to quantify blood flow, hemoglobin oxygen saturation and vascular density. Using our method, we were able to monitor the changes in the peri-implant microvasculature noninvasively without the use of fluorescent dyes. Significant changes in blood flow, hemoglobin oxygen saturation and vascular density were noted as early as the first week post-transplant. The dorsal window chamber model enables comparison of host responses to transplanted biomaterials. Future experiments will study the effect of changes in alginate composition on the vascular and immune responses.
Collapse
Affiliation(s)
- Rahul Krishnan
- Department of Surgery, University of California Irvine, Orange, CA 92868, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Current therapies for the treatment of type 1 diabetes include daily administration of exogenous insulin and, less frequently, whole-pancreas or islet transplantation. Insulin injections often result in inaccurate insulin doses, exposing the patient to hypo- and/or hyperglycemic episodes that lead to long-term complications. Islet transplantation is also limited by lack of high-quality islet donors, early graft failure, and chronic post-transplant immunosuppressive treatment. These barriers could be circumvented by designing a safe and efficient strategy to restore insulin production within the patient's body. Porcine islets have been considered as a possible alternative source of transplantable insulin-producing cells to replace human cadaveric islets. More recently, embryonic or induced pluripotent stem cells have also been examined for their ability to differentiate in vitro into pancreatic endocrine cells. Alternatively, it may be feasible to generate new β-cells by ectopic expression of key transcription factors in endogenous non-β-cells. Finally, engineering surrogate β-cells by in vivo delivery of the insulin gene to specific tissues is also being studied as a possible therapy for type 1 diabetes. In the present review, we discuss these different approaches to restore insulin production.
Collapse
Affiliation(s)
- Eva Tudurí
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
39
|
van der Windt DJ, Bottino R, Kumar G, Wijkstrom M, Hara H, Ezzelarab M, Ekser B, Phelps C, Murase N, Casu A, Ayares D, Lakkis FG, Trucco M, Cooper DK. Clinical islet xenotransplantation: how close are we? Diabetes 2012; 61:3046-55. [PMID: 23172951 PMCID: PMC3501885 DOI: 10.2337/db12-0033] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 06/06/2012] [Indexed: 01/27/2023]
Affiliation(s)
- Dirk J. van der Windt
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Rita Bottino
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Division of Immunogenetics, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Goutham Kumar
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Martin Wijkstrom
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Hidetaka Hara
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Mohamed Ezzelarab
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Burcin Ekser
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Surgery, Transplantation and Advanced Technologies, Vascular Surgery and Organ Transplant Unit, University Hospital of Catania, Catania, Italy
| | | | - Noriko Murase
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Anna Casu
- Diabetes Unit, Department of Medicine, Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (ISMETT), Palermo, Italy
| | | | - Fadi G. Lakkis
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Massimo Trucco
- Division of Immunogenetics, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - David K.C. Cooper
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
40
|
Yeom HJ, Koo OJ, Yang J, Cho B, Hwang JI, Park SJ, Hurh S, Kim H, Lee EM, Ro H, Kang JT, Kim SJ, Won JK, O'Connell PJ, Kim H, Surh CD, Lee BC, Ahn C. Generation and characterization of human heme oxygenase-1 transgenic pigs. PLoS One 2012; 7:e46646. [PMID: 23071605 PMCID: PMC3465346 DOI: 10.1371/journal.pone.0046646] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Accepted: 09/03/2012] [Indexed: 12/12/2022] Open
Abstract
Xenotransplantation using transgenic pigs as an organ source is a promising strategy to overcome shortage of human organ for transplantation. Various genetic modifications have been tried to ameliorate xenograft rejection. In the present study we assessed effect of transgenic expression of human heme oxygenase-1 (hHO-1), an inducible protein capable of cytoprotection by scavenging reactive oxygen species and preventing apoptosis caused by cellular stress during inflammatory processes, in neonatal porcine islet-like cluster cells (NPCCs). Transduction of NPCCs with adenovirus containing hHO-1 gene significantly reduced apoptosis compared with the GFP-expressing adenovirus control after treatment with either hydrogen peroxide or hTNF-α and cycloheximide. These protective effects were diminished by co-treatment of hHO-1 antagonist, Zinc protoporphyrin IX. We also generated transgenic pigs expressing hHO-1 and analyzed expression and function of the transgene. Human HO-1 was expressed in most tissues, including the heart, kidney, lung, pancreas, spleen and skin, however, expression levels and patterns of the hHO-1 gene are not consistent in each organ. We isolate fibroblast from transgenic pigs to analyze protective effect of the hHO-1. As expected, fibroblasts derived from the hHO-1 transgenic pigs were significantly resistant to both hydrogen peroxide damage and hTNF-α and cycloheximide-mediated apoptosis when compared with wild-type fibroblasts. Furthermore, induction of RANTES in response to hTNF-α or LPS was significantly decreased in fibroblasts obtained from the hHO-1 transgenic pigs. These findings suggest that transgenic expression of hHO-1 can protect xenografts when exposed to oxidative stresses, especially from ischemia/reperfusion injury, and/or acute rejection mediated by cytokines. Accordingly, hHO-1 could be an important candidate molecule in a multi-transgenic pig strategy for xenotransplantation.
Collapse
Affiliation(s)
- Hye-Jung Yeom
- Transplantation Research Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Ok Jae Koo
- Transplantation Research Institute, College of Medicine, Seoul National University, Seoul, Korea
- Designed Animal Resource Center and Biotransplant Research Institute, Seoul National University Green-Bio Research Complex, Gangwon-do, Korea
| | - Jaeseok Yang
- Transplantation Research Institute, College of Medicine, Seoul National University, Seoul, Korea
- Transplantation Center, Seoul National University Hospital, Seoul, Korea
| | - Bumrae Cho
- Transplantation Research Institute, College of Medicine, Seoul National University, Seoul, Korea
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Jong-Ik Hwang
- Graduate School of Medicine, Laboratory of G Protein Coupled Receptors, Korea University, Seoul, Korea
| | - Sol Ji Park
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Sunghoon Hurh
- Transplantation Research Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Hwajung Kim
- Transplantation Research Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Eun Mi Lee
- Transplantation Research Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Han Ro
- Transplantation Research Institute, College of Medicine, Seoul National University, Seoul, Korea
- Transplantation Center, Seoul National University Hospital, Seoul, Korea
| | - Jung Taek Kang
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Su Jin Kim
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Jae-Kyung Won
- Molecular Pathology Center, Seoul National University Cancer Hospital, Seoul, Korea
| | - Philip J. O'Connell
- The Center for Transplant Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, New South Wales, Australia
| | - Hyunil Kim
- Optifarm Solution Inc., Seonggeo-eup, Cheonan, Korea
| | - Charles D. Surh
- The Scripps Research Institute, La Jolla, California, United States of America
| | - Byeong-Chun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
- Designed Animal Resource Center and Biotransplant Research Institute, Seoul National University Green-Bio Research Complex, Gangwon-do, Korea
- * E-mail: (AC); (B-CL)
| | - Curie Ahn
- Transplantation Research Institute, College of Medicine, Seoul National University, Seoul, Korea
- Designed Animal Resource Center and Biotransplant Research Institute, Seoul National University Green-Bio Research Complex, Gangwon-do, Korea
- Transplantation Center, Seoul National University Hospital, Seoul, Korea
- Division of Nephrology, Seoul National University College of Medicine, Seoul, Korea
- * E-mail: (AC); (B-CL)
| |
Collapse
|
41
|
Montane J, Klimek-Abercrombie A, Potter KJ, Westwell-Roper C, Bruce Verchere C. Metabolic stress, IAPP and islet amyloid. Diabetes Obes Metab 2012; 14 Suppl 3:68-77. [PMID: 22928566 DOI: 10.1111/j.1463-1326.2012.01657.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Amyloid forms within pancreatic islets in type 2 diabetes from aggregates of the β-cell peptide islet amyloid polypeptide (IAPP). These aggregates are toxic to β-cells, inducing β-cell death and dysfunction, as well as inciting islet inflammation. The β-cell is subject to a number of other stressors, including insulin resistance and hyperglycaemia, that may contribute to amyloid formation by increasing IAPP production by the β-cell. β-Cell dysfunction, evident as impaired glucose-stimulated insulin secretion and defective prohormone processing and exacerbated by metabolic stress, is also a likely prerequisite for islet amyloid formation to occur in type 2 diabetes. Islet transplants in patients with type 1 diabetes face similar stressors, and are subject to rapid amyloid formation and impaired proinsulin processing associated with progressive loss of β-cell function and mass. Declining β-cell mass is predicted to increase metabolic demand on remaining β-cells, promoting a feed-forward cycle of β-cell decline.
Collapse
Affiliation(s)
- J Montane
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | |
Collapse
|
42
|
Adenovirus-mediated heme oxygenase-1 gene transfer to neonatal porcine islet-like cluster cells: the effects on gene expression and protection from cell stress. BIOCHIP JOURNAL 2012. [DOI: 10.1007/s13206-012-6108-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
43
|
Induction of protective genes leads to islet survival and function. J Transplant 2011; 2011:141898. [PMID: 22220267 PMCID: PMC3246756 DOI: 10.1155/2011/141898] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 09/01/2011] [Indexed: 12/16/2022] Open
Abstract
Islet transplantation is the most valid approach to the treatment of type 1 diabetes. However, the function of transplanted islets is often compromised since a large number of β cells undergo apoptosis induced by stress and the immune rejection response elicited by the recipient after transplantation. Conventional treatment for islet transplantation is to administer immunosuppressive drugs to the recipient to suppress the immune rejection response mounted against transplanted islets. Induction of protective genes in the recipient (e.g., heme oxygenase-1 (HO-1), A20/tumor necrosis factor alpha inducible protein3 (tnfaip3), biliverdin reductase (BVR), Bcl2, and others) or administration of one or more of the products of HO-1 to the donor, the islets themselves, and/or the recipient offers an alternative or synergistic approach to improve islet graft survival and function. In this perspective, we summarize studies describing the protective effects of these genes on islet survival and function in rodent allogeneic and xenogeneic transplantation models and the prevention of onset of diabetes, with emphasis on HO-1, A20, and BVR. Such approaches are also appealing to islet autotransplantation in patients with chronic pancreatitis after total pancreatectomy, a procedure that currently only leads to 1/3 of transplanted patients being diabetes-free.
Collapse
|
44
|
Thompson P, Badell IR, Lowe M, Cano J, Song M, Leopardi F, Avila J, Ruhil R, Strobert E, Korbutt G, Rayat G, Rajotte R, Iwakoshi N, Larsen CP, Kirk AD. Islet xenotransplantation using gal-deficient neonatal donors improves engraftment and function. Am J Transplant 2011; 11:2593-602. [PMID: 21883917 PMCID: PMC3226931 DOI: 10.1111/j.1600-6143.2011.03720.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Significant deficiencies in understanding of xenospecific immunity have impeded the success of preclinical trials in xenoislet transplantation. Although galactose-α1,3-galactose, the gal epitope, has emerged as the principal target of rejection in pig-to-primate models of solid organ transplant, the importance of gal-specific immunity in islet xenotransplant models has yet to be clearly demonstrated. Here, we directly compare the immunogenicity, survival and function of neonatal porcine islets (NPIs) from gal-expressing wild-type (WT) or gal-deficient galactosyl transferase knockout (GTKO) donors. Paired diabetic rhesus macaques were transplanted with either WT (n = 5) or GTKO (n = 5) NPIs. Recipient blood glucose, transaminase and serum xenoantibody levels were used to monitor response to transplant. Four of five GTKO versus one of five WT recipients achieved insulin-independent normoglycemia; transplantation of WT islets resulted in significantly greater transaminitis. The WT NPIs were more susceptible to antibody and complement binding and destruction in vitro. Our results confirm that gal is an important variable in xenoislet transplantation. The GTKO NPI recipients have improved rates of normoglycemia, likely due to decreased susceptibility of xenografts to innate immunity mediated by complement and preformed xenoantibody. Therefore, the use of GTKO donors is an important step toward improved consistency and interpretability of results in future xenoislet studies.
Collapse
Affiliation(s)
- P Thompson
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - IR Badell
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - M Lowe
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - J Cano
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - M Song
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - F Leopardi
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - J Avila
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - R Ruhil
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - E Strobert
- Yerkes National Primate Research Center, Atlanta, GA, USA 30322
| | - G Korbutt
- Surgical-Medical Research Institute, University of Alberta, Edmonton, AB, T6G 2N8, Canada
| | - G Rayat
- Surgical-Medical Research Institute, University of Alberta, Edmonton, AB, T6G 2N8, Canada
| | - R Rajotte
- Surgical-Medical Research Institute, University of Alberta, Edmonton, AB, T6G 2N8, Canada
| | - N Iwakoshi
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - CP Larsen
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - AD Kirk
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| |
Collapse
|
45
|
Subcapsular fetal pig pancreas fragment transplantation provides normal blood glucose control in a preclinical model of diabetes. Transplantation 2011; 91:515-21. [PMID: 21183867 DOI: 10.1097/tp.0b013e3182079474] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Identifying a limitless source of β-cells that survive transplantation into a neovascularised site and provide normal blood glucose control remains an important goal in the development of pancreatic islet xenotransplantation. It was our hypothesis that fetal porcine pancreas fragments could achieve these objectives, and this was tested in a large preclinical animal model. RESEARCH DESIGN AND METHODS Inbred "Westran Pig" fetal porcine pancreas fragments were transplanted beneath the splenic capsule into syngeneic Westran Pig recipients without immunosuppression, and 3 months later, a total native pancreatectomy was performed to demonstrate function. RESULTS Histologic analysis showed appropriate development of islet-like structures up to and beyond 120 days after transplantation. After native pancreatectomy, recipients survived more than 100 days without exogenous insulin and with normal glucose homeostasis as assessed by normal glucose tolerance tests, K values, and normal glucagon secretion. CONCLUSIONS This study confirms that fetal pig islet tissue has the potential to mature and function normally in a neovascularised site, hence, avoiding the innate immune destruction that occurs when islet tissue is exposed directly to the circulation.
Collapse
|
46
|
Wang P, Yigit MV, Medarova Z, Wei L, Dai G, Schuetz C, Moore A. Combined small interfering RNA therapy and in vivo magnetic resonance imaging in islet transplantation. Diabetes 2011; 60:565-71. [PMID: 21270267 PMCID: PMC3028356 DOI: 10.2337/db10-1400] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Recent advances in human islet transplantation are hampered by significant graft loss shortly after transplantation and inability to follow islet fate directly. Both issues were addressed by utilizing a dual-purpose therapy/imaging small interfering RNA (siRNA)-nanoparticle probe targeting apoptotic-related gene caspase-3. We expect that treatment with the probe would result in significantly better survival of transplanted islets, which could be monitored by in vivo magnetic resonance imaging (MRI). RESEARCH DESIGN AND METHODS We synthesized a probe consisting of therapeutic (siRNA to human caspase-3) and imaging (magnetic iron oxide nanoparticles, MN) moieties. In vitro testing of the probe included serum starvation of the islets followed by treatment with the probe. Caspase-3 gene silencing and protein expression were determined by RT-PCR and Western blot, respectively. In vivo studies included serial MRI of NOD-SCID mice transplanted with MN-small interfering (si)Caspase-3-labeled human islets under the left kidney capsule and MN-treated islets under the right kidney capsule. RESULTS Treatment with MN-siCaspase-3 probe resulted in decrease of mRNA and protein expression in serum-starved islets compared with controls. In vivo MRI showed that there were significant differences in the relative volume change between MN-siCaspase-3-treated grafts and MN-labeled grafts. Histology revealed decreased caspase-3 expression and cell apoptosis in MN-siCaspase-3-treated grafts compared with the control side. CONCLUSIONS Our data show the feasibility of combining siRNA therapy and in vivo monitoring of transplanted islets in mice. We observed a protective effect of MN-siCaspase-3 in treated islets both in vitro and in vivo. This study could potentially aid in increasing the success of clinical islet transplantation.
Collapse
Affiliation(s)
- Ping Wang
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mehmet V. Yigit
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Zdravka Medarova
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lingling Wei
- The Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Guangping Dai
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christian Schuetz
- The Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anna Moore
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Corresponding author: Anna Moore,
| |
Collapse
|
47
|
Wu H, Panakanti R, Li F, Mahato RI. XIAP gene expression protects β-cells and human islets from apoptotic cell death. Mol Pharm 2010; 7:1655-66. [PMID: 20677802 DOI: 10.1021/mp100070j] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Islet transplantation has the potential to treat type I diabetes, however, its clinical application is limited due to the massive apoptotic cell death and other post-transplantation challenges to islet grafts. Therefore, the objective of this study was to determine whether ex vivo transduction of rat insulin producing INS-1E cells and human islets with adenoviral vector encoding human X-linked inhibitor of apoptosis (Adv-hXIAP) can protect them from inflammatory cytokines and improve their viability and function. There was dose dependent XIAP gene expression. XIAP expression led to decrease in the activities of caspase 3/7, 8 and 9, resulting in reduced apoptotic cell death induced by a cocktail of inflammatory cytokines such as IL-1β, TNFα, and IFNγ. Prolonged normoglycemic control could be achieved by transplantation of Adv-XIAP transduced human islets under the kidney capsule of streptozotocin induced diabetic NOD-SCID mice. Immunohistological staining of the islets bearing kidney sections at day 42 after transplantation was positive for insulin. Moreover, the protective effect of XIAP was reversed by coadministration of XIAP inhibitor embelin. These results indicate that ex vivo transduction of islets with Adv-XIAP will decrease cytokine induced apoptosis and improve the outcome of islet transplantation.
Collapse
Affiliation(s)
- Hao Wu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38103-3308, USA
| | | | | | | |
Collapse
|
48
|
Islet amyloid deposition limits the viability of human islet grafts but not porcine islet grafts. Proc Natl Acad Sci U S A 2010; 107:4305-10. [PMID: 20160085 DOI: 10.1073/pnas.0909024107] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Islet transplantation is a promising treatment for diabetes but long-term success is limited by progressive graft loss. Aggregates of the beta cell peptide islet amyloid polypeptide (IAPP) promote beta cell apoptosis and rapid amyloid formation occurs in transplanted islets. Porcine islets are an attractive alternative islet source as they demonstrate long-term graft survival. We compared the capacity of transplanted human and porcine islets to form amyloid as an explanation for differences in graft survival. Human islets were transplanted into streptozotocin-diabetic immune-deficient mice. Amyloid deposition was detectable at 4 weeks posttransplantation and was associated with islet graft failure. More extensive amyloid deposition was observed after 8 weeks. By contrast, no amyloid was detected in transplanted neonatal or adult porcine islets that had maintained normoglycemia for up to 195 days. To determine whether differences in IAPP sequence between humans and pigs could explain differences in amyloid formation and transplant viability, we sequenced porcine IAPP. Porcine IAPP differs from the human sequence at 10 positions and includes substitutions predicted to reduce its amyloidogenicity. Synthetic porcine IAPP was considerably less amyloidogenic than human IAPP as determined by transmission electron microscopy, circular dichroism, and thioflavin T binding. Viability assays indicated that porcine IAPP is significantly less toxic to INS-1 beta cells than human IAPP. Our findings demonstrate that species differences in IAPP sequence can explain the lack of amyloid formation and improved survival of transplanted porcine islets. These data highlight the potential of porcine islet transplantation as a therapeutic approach for human diabetes.
Collapse
|
49
|
Abstract
Sensitivity of pancreatic islets to hypoxia is one of the most important of the obstacles responsible for their failure to survive within the recipients. The aim of this study was to compare the in vitro hypoxia tolerance of neonatal and adult rat islet cells and to study the glucose metabolism in these cells after exposure to hypoxia. Islet cells from both age categories were cultured in different hypoxic levels for 24 h and insulin secretion and some metabolites of glucose metabolism were analysed. Glucose-stimulated insulin secretion decreased dramatically in both cell preparations in response to the decrease in oxygen level. The reduction of insulin secretion was more detectable in adult cells and started at 5% O(2), while a significant reduction was obtained at 1% O(2) in neonatal cells. Moreover, basal insulin release of neonatal cells showed an adaptation to hypoxia after a 4-day culture in hypoxia. Intracellular pyruvate was higher in neonatal cells than in adult ones, while no difference in lactate level was observed between them. Similar results to that of pyruvate were observed for adenosine triphosphate (ATP) and the second messenger cyclic adenosine monophosphate (cAMP). The study reveals that neonatal rat islet cells are more hypoxia-tolerant than the adult ones. The most obvious metabolic observation was that both pyruvate and lactate were actively produced in neonatal cells, while adult cells depended mainly on lactate production as an end-product of glycolysis, indicating a more enhanced metabolic flexibility of neonatal cells to utilize the available oxygen and, at the same time, maintain metabolism anaerobically.
Collapse
Affiliation(s)
- Ayman Hyder
- Department of Physiology and Biochemistry of Nutrition, Max Rubner Institute, and Clinical Research Center, Innovation and Technology centre, Kiel, Germany
| | | | | |
Collapse
|
50
|
Yuan Y, Cong C, Zhang J, Wei L, Li S, Chen Y, Tan W, Cheng J, Li Y, Zhao X, Lu Y. Self-assembling peptide nanofiber as potential substrates in islet transplantation. Transplant Proc 2008; 40:2571-4. [PMID: 18929804 DOI: 10.1016/j.transproceed.2008.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hypoxia and reoxygenation (H/R)-induced damage often happens soon after islets are transplantation. The process of islet isolation and purification causes the rapid onset of hypoxia. We sought to develop a functional scaffold to sustain the structure and function of islets as well as to recover some of the surface molecules damaged during isolation, seeking to improve islet transplantation outcomes. Self-assembling peptide nanofiber (SAPNF), a new type of substrate has been shown to be an excellent biological material for neuronal cell culture and tissue engineering in animals. In this study, we investigated the protective effect of SAPNF on damage to rat islets. Freshly prepared rat islets from male Sprague-Dawley rats were seeded in plates coated with (SAPNF-treated group) or without (control group) SAPNF. The islets were then divided into two groups culture under normoxia for 7 days versus exposure to hypoxia (< 1% O2) for 6 hours followed by reoxygenation for 24 hours. The results showed that SAPNF exhibited improving effects on viability and function of cultured islets, protecting the one from H/R-induced damage. In both groups, the stimulation index of SAPNF-treated groups were about two times the controls. SAPNF treatment decreased apoptotic rates of islet cells. These results suggested the usefulness of SAPNF to maintain the viability and function of rat pancreatic islets. SAPNF may be a potential scaffold for clinical islet transplantation.
Collapse
Affiliation(s)
- Y Yuan
- Key Lab of Transplant Engineering and Immunology, Ministry of Health, P R China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|