1
|
Graham ML, Ramachandran S, Singh A, Moore MEG, Flanagan EB, Azimzadeh A, Burlak C, Mueller KR, Martins K, Anazawa T, Balamurugan AN, Bansal-Pakala P, Murtaugh MP, O’Brien TD, Papas KK, Spizzo T, Schuurman HJ, Hancock WW, Hering BJ. Clinically available immunosuppression averts rejection but not systemic inflammation after porcine islet xenotransplant in cynomolgus macaques. Am J Transplant 2022; 22:745-760. [PMID: 34704345 PMCID: PMC9832996 DOI: 10.1111/ajt.16876] [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: 05/26/2021] [Revised: 09/30/2021] [Accepted: 10/19/2021] [Indexed: 01/25/2023]
Abstract
A safe, efficacious, and clinically applicable immunosuppressive regimen is necessary for islet xenotransplantation to become a viable treatment option for diabetes. We performed intraportal transplants of wild-type adult porcine islets in 25 streptozotocin-diabetic cynomolgus monkeys. Islet engraftment was good in 21, partial in 3, and poor in 1 recipient. Median xenograft survival was 25 days with rapamycin and CTLA4Ig immunosuppression. Adding basiliximab induction and maintenance tacrolimus to the base regimen significantly extended median graft survival to 147 days (p < .0001), with three animals maintaining insulin-free xenograft survival for 265, 282, and 288 days. We demonstrate that this regimen suppresses non-Gal anti-pig antibody responses, circulating effector memory T cell expansion, effector function, and infiltration of the graft. However, a chronic systemic inflammatory state manifested in the majority of recipients with long-term graft survival indicated by increased neutrophil to lymphocyte ratio, IL-6, MCP-1, CD40, and CRP expression. This suggests that this immunosuppression regimen fails to regulate innate immunity and resulting inflammation is significantly associated with increased incidence and severity of adverse events making this regimen unacceptable for translation. Additional studies are needed to optimize a maintenance regimen for regulating the innate inflammatory response.
Collapse
Affiliation(s)
- Melanie L. Graham
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | | | - Amar Singh
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Meghan E. G. Moore
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN
| | - E. Brian Flanagan
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Agnes Azimzadeh
- Department of Surgery, University of Maryland, Baltimore, MD
| | - Christopher Burlak
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Kate R. Mueller
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Kyra Martins
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| | - Takayuki Anazawa
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | | | - Pratima Bansal-Pakala
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Michael P. Murtaugh
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| | - Timothy D. O’Brien
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN
| | - Klearchos K. Papas
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | | | - Henk-J. Schuurman
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN,Spring Point Project, Minneapolis, MN
| | - Wayne W. Hancock
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Bernhard. J. Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| |
Collapse
|
2
|
Imura T, Inagaki A, Igarashi Y, Goto M. Optimization of dye solutions for detecting damaged pancreatic tissues during islet isolation procedures. PLoS One 2021; 16:e0255733. [PMID: 34388180 PMCID: PMC8362985 DOI: 10.1371/journal.pone.0255733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/22/2021] [Indexed: 12/29/2022] Open
Abstract
We previously reported that dye was effective to prevent the leakage of enzyme solutions from pancreatic glands during an islet isolation procedure. However, the dye used for islet isolation has not yet been optimized. In this study, we focused on pyoktanin blue (PB), diagnogreen (DG), and indigo carmine (IC) as potential candidates among clinically established dyes. A serial dilution assay was performed to determine minimal effective concentrations of each dye for detecting damaged pancreatic tissues. According to the outcome of serial dilution assays, double minimum effective concentrations of each dye were used for in vitro toxicity assays on islets and used in the isolation procedure to investigate whether they adversely affect islet isolation efficiency. The evaluations included islet yield, ADP/ATP, ATP/DNA, glucose stimulation test, and insulin/DNA assays. Islet viability cultured with PB contained medium was significantly lower than the other dyes. DG and IC appeared to be non-toxic to the islets. In isolation experiments, the islet yield in the DG group was considerably lower than that in the Control group, suggesting that DG might inhibit enzyme activity. The present study demonstrates that IC could be a promising candidate for an effective dye to detect damaged pancreatic tissues without affecting the enzyme activity and islet quality.
Collapse
Affiliation(s)
- Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Yasuhiro Igarashi
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Masafumi Goto
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
3
|
Axelsson AS, Mahdi T, Nenonen HA, Singh T, Hänzelmann S, Wendt A, Bagge A, Reinbothe TM, Millstein J, Yang X, Zhang B, Gusmao EG, Shu L, Szabat M, Tang Y, Wang J, Salö S, Eliasson L, Artner I, Fex M, Johnson JD, Wollheim CB, Derry JMJ, Mecham B, Spégel P, Mulder H, Costa IG, Zhang E, Rosengren AH. Sox5 regulates beta-cell phenotype and is reduced in type 2 diabetes. Nat Commun 2017; 8:15652. [PMID: 28585545 PMCID: PMC5467166 DOI: 10.1038/ncomms15652] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 04/10/2017] [Indexed: 01/09/2023] Open
Abstract
Type 2 diabetes (T2D) is characterized by insulin resistance and impaired insulin secretion, but the mechanisms underlying insulin secretion failure are not completely understood. Here, we show that a set of co-expressed genes, which is enriched for genes with islet-selective open chromatin, is associated with T2D. These genes are perturbed in T2D and have a similar expression pattern to that of dedifferentiated islets. We identify Sox5 as a regulator of the module. Sox5 knockdown induces gene expression changes similar to those observed in T2D and diabetic animals and has profound effects on insulin secretion, including reduced depolarization-evoked Ca2+-influx and β-cell exocytosis. SOX5 overexpression reverses the expression perturbations observed in a mouse model of T2D, increases the expression of key β-cell genes and improves glucose-stimulated insulin secretion in human islets from donors with T2D. We suggest that human islets in T2D display changes reminiscent of dedifferentiation and highlight SOX5 as a regulator of β-cell phenotype and function.
Collapse
Affiliation(s)
- A S Axelsson
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - T Mahdi
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden.,Medical Research Center, Hawler Medical University, 44001 Erbil, Iraq
| | - H A Nenonen
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - T Singh
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - S Hänzelmann
- Institute of Biomedical Engineering, RWTH Aachen University Hospital, Pauwelstr 19, 52074 Aachen, Germany
| | - A Wendt
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - A Bagge
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - T M Reinbothe
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - J Millstein
- Sage Bionetworks, 1100 Fairview Avenue N, Seattle, Washington 98109, USA
| | - X Yang
- Sage Bionetworks, 1100 Fairview Avenue N, Seattle, Washington 98109, USA.,Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Dr East, Los Angeles, California 90095, USA
| | - B Zhang
- Sage Bionetworks, 1100 Fairview Avenue N, Seattle, Washington 98109, USA.,Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, New York 10029, USA
| | - E G Gusmao
- Institute of Biomedical Engineering, RWTH Aachen University Hospital, Pauwelstr 19, 52074 Aachen, Germany
| | - L Shu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Dr East, Los Angeles, California 90095, USA
| | - M Szabat
- Diabetes Research Group, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 5358-2350 Health Sciences Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | - Y Tang
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden.,Key Lab of Hormones and Development, Ministry of Health, Metabolic Diseases Hospital, Tianjin Medical University, Tianjin 300070, China
| | - J Wang
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden.,Department of Emergency, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, China
| | - S Salö
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - L Eliasson
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - I Artner
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - M Fex
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - J D Johnson
- Diabetes Research Group, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, 5358-2350 Health Sciences Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | - C B Wollheim
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden.,Department of Cell Physiology and Metabolism, University Medical Center, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - J M J Derry
- Sage Bionetworks, 1100 Fairview Avenue N, Seattle, Washington 98109, USA
| | - B Mecham
- Trialomics, 6310 12th Avenue NE, Seattle, Washington 98115, USA
| | - P Spégel
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden.,Centre for Analysis and Synthesis, Department of Chemistry, Lund University, SE-221 00 Lund, Sweden
| | - H Mulder
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - I G Costa
- Institute of Biomedical Engineering, RWTH Aachen University Hospital, Pauwelstr 19, 52074 Aachen, Germany
| | - E Zhang
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - A H Rosengren
- Lund University Diabetes Center, CRC 91-11 SUS, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden.,Sage Bionetworks, 1100 Fairview Avenue N, Seattle, Washington 98109, USA.,Department of Neuroscience and Physiology, University of Gothenburg, Box 100, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
4
|
Gibson WT, Hayden MR. Mycophenolate mofetil and animal models. Lupus 2016. [DOI: 10.1177/0961203306071675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mycophenolate mofetil (MMF), is the morpholinoethyl ester of mycophenolic acid (MPA). Though initially developed as an anti-rejection treatment, MMF is beginning to find application in more common immune-mediated diseases. MMF has been shown to be effective against transplant-associated vascular disease, lupus and other inflammatory diseases via multiple mechanisms in several animal models. MMF treatment blocks the proliferation of T cells and B cells, attenuates the production of autoreactive IgG and IgM, diminishes complement deposition, and reduces the production of multiple proinflammatory cytokines including TNF-α, IFN-γ, IL-2, IL-3, IL-4, IL-5, IL-6 and IL-12. It also increases production of the anti-inflammatory mediator IL-10. In addition, MMF reduces the infiltration of immune cells into sites of inflammation by interfering with the expression of cell-surface molecules critical for this process, including MHC class II, CD40, CD80, CD86, I-A, and ICAM-1. Additional mechanisms involving mannosylation and N-linked glycosylation of cell-surface molecules are only beginning to be investigated. This article will focus on the contribution of animal models of disease as investigational tools in the development of MMF as an immunomodulatory drug. The use of mice, rats, rabbits, monkeys, baboons and interspecific xenografts will be discussed.
Collapse
Affiliation(s)
- WT Gibson
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - MR Hayden
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
5
|
Synergistic Effect of Neutral Protease and Clostripain on Rat Pancreatic Islet Isolation. Transplantation 2016; 99:1349-55. [PMID: 25803499 DOI: 10.1097/tp.0000000000000662] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Islet isolation currently requires collagenase, neutral protease and other components. Thermolysin (TL) from Bacillus thermoproteolyticus is the gold standard neutral protease. However, we speculated that neutral protease derived from Clostridium histolyticum (Ch; ChNP) would be biologically superior for islet isolation. Tryptic-like activity has also been reported to be important. Therefore, we focused on clostripain (CP), since it is one of the main proteases in Clostridium histolyticum which possesses tryptic-like activity. We then examined the synergistic effects of highly purified ChNP and CP on rat islet isolation. METHODS The same amount of collagenase was used in all four groups (TL, ChNP, TL+CP and ChNP+CP; n = 12/group). The efficiency was evaluated by the islet yield and function. An immunohistochemical analysis, in vitro digestion assay for each enzyme component and evaluation of the activation of endogenous exocrine proteases during islet isolation were also performed. RESULTS The islet yield of the TL group was significantly higher than that of the ChNP group (P < 0.01). The islet yield was dose dependently increased in the ChNP+CP group, but was decreased in the TL + CP group. The islet yield in the ChNP + CP group was significantly higher than that in the TL group, but their islet function was similar. Different specificities for laminin, especially laminin-511, were observed in the TL, ChNP, and CP groups. CONCLUSIONS Clostripain had a strong synergistic effect with ChNP, but not with TL. Therefore, ChNP and CP, in combination with collagenase derived from the same bacteria, may effectively increase the isolation efficiency without affecting the quality of islets.
Collapse
|
6
|
Bowers DT, Botchwey EA, Brayman KL. Advances in Local Drug Release and Scaffolding Design to Enhance Cell Therapy for Diabetes. TISSUE ENGINEERING. PART B, REVIEWS 2015; 21:491-503. [PMID: 26192271 DOI: 10.1089/ten.teb.2015.0275] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Islet transplant is a curative treatment for insulin-dependent diabetes. However, challenges, including poor tissue survival and a lack of efficient engraftment, must be overcome. An encapsulating or scaffolding material can act as a vehicle for agents carefully chosen for the islet transplant application. From open porous scaffolds to spherical capsules and conformal coatings, greater immune protection is often accompanied by greater distances to microvasculature. Generating a local oxygen supply from the implant material or encouraging vessel growth through the release of local factors can create an oxygenated engraftment site. Intricately related to the vascularization response, inflammatory interaction with the cell supporting implant is a long-standing hurdle to material-based islet transplant. Modulation of the immune responses to the islets as well as the material itself must be considered. To match the post-transplant complexity, the release rate can be tuned to orchestrate temporal responses. Material degradation properties can be utilized in passive approaches or external stimuli and biological cues in active approaches. A combination of multiple carefully chosen factors delivered in an agent-specialized manner is considered by this review to improve the long-term function of islets transplanted in scaffolding and encapsulating materials.
Collapse
Affiliation(s)
- Daniel T Bowers
- 1 Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia
- 2 Department of Surgery, University of Virginia , Charlottesville, Virginia
| | - Edward A Botchwey
- 3 Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University , Atlanta, Georgia
| | - Kenneth L Brayman
- 1 Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia
- 2 Department of Surgery, University of Virginia , Charlottesville, Virginia
| |
Collapse
|
7
|
Fujio A, Murayama K, Yamagata Y, Watanabe K, Imura T, Inagaki A, Ohbayashi N, Shima H, Sekiguchi S, Fujimori K, Igarashi K, Ohuchi N, Satomi S, Goto M. Collagenase H is crucial for isolation of rat pancreatic islets. Cell Transplant 2013; 23:1187-98. [PMID: 23768818 DOI: 10.3727/096368913x668654] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The role(s) of collagenase G (ColG) and collagenase H (ColH) during pancreatic islet isolation remains controversial, possibly due to the enzyme blends used in the previous studies. We herein examined the role of ColG and ColH using highly pure enzyme blends of recombinant collagenase of each subtype. Rat pancreases were digested using thermolysin, together with ColG, ColH, or ColG/ColH (n = 9, respectively). No tryptic-like activity was detected in any components of the enzyme blends. The efficiency of the collagenase subtypes was evaluated by islet yield and function. Immunohistochemical analysis, in vitro collagen digestion assay, and mass spectrometry were also performed to examine the target matrix components of the crucial collagenase subtype. The islet yield was highest in the ColG/ColH group (4,101 ± 460 islet equivalents). A substantial number of functional islets (2,811 ± 581 islet equivalents) was obtained in the ColH group, whereas no islets were retrieved in the ColG group. Mass spectrometry demonstrated that ColH reacts with collagen I and III. In the immunohistochemical analysis, both collagen I and III were located in exocrine tissues, although collagen III expression was more pronounced. The collagen digestion assay showed that collagen III was more effectively digested by ColH than by ColG. The present study reveals that ColH is crucial, while ColG plays only a supporting role, in rat islet isolation. In addition, collagen III appears to be one of the key targets of ColH.
Collapse
Affiliation(s)
- Atsushi Fujio
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Ductal injection does not increase the islet yield or function after cold storage in a vascular perfusion model. PLoS One 2012; 7:e42319. [PMID: 22900012 PMCID: PMC3416830 DOI: 10.1371/journal.pone.0042319] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 07/03/2012] [Indexed: 11/19/2022] Open
Abstract
Several studies have reported that pancreatic ductal preservation greatly improved the islet yield and function after cold storage. However, these studies were devoid of appropriate controls, such as vascular perfusion, which is routinely performed to preserve organs in the clinical setting. In this study, we created a vascular perfusion model using inbred rats, and investigated the effect of ductal injection on the islet yield and function after cold storage. Rat pancreases after 10 h cold ischemia were classified as follows: without ductal/vascular perfusion; with ductal injection; with vascular perfusion; and with ductal/vascular perfusion. The islet yield, function, viability, release of inflammatory mediators, and pathological changes in the exocrine tissues were assessed in the Hanks' Balanced Salt Solution (HBSS) model. The islet yield was also assesed by introducing University of Wisconsin Solution (UWS) and Histidine-Tryptophan-Ketoglutarate solution (HTK), which are the standard clinical preservation solutions. In the HBSS model, ductal injection and vascular perfusion significantly improved the islet yield compared with the control group. However, ductal injection showed no additional effects on the islet yield, function, viability and suppressing the release of inflammatory mediators when vascular perfusion was performed. Although ductal injection significantly decreased the apoptosis of exocrine cells, no beneficial effect on vacuolation was observed. In contrast, vascular perfusion significantly suppressed vacuolation in the exocrine tissues. Likewise, in the UWS and HTK model, ductal injection and vascular perfusion improved the islet yield compared with the control group. Nevertheless, the combination group showed no additional effects. These data suggest that ductal injection has no additional effect on islet yield and function after cold storage in a vascular perfusion model. We propose that ductal injection can be an effective and simple alternative for vascular perfusion prior to pancreas harvest, but is not necessary in most cases, since vascular perfusion is routinely performed.
Collapse
|
9
|
Rosengren AH, Braun M, Mahdi T, Andersson SA, Travers ME, Shigeto M, Zhang E, Almgren P, Ladenvall C, Axelsson AS, Edlund A, Pedersen MG, Jonsson A, Ramracheya R, Tang Y, Walker JN, Barrett A, Johnson PR, Lyssenko V, McCarthy MI, Groop L, Salehi A, Gloyn AL, Renström E, Rorsman P, Eliasson L. Reduced insulin exocytosis in human pancreatic β-cells with gene variants linked to type 2 diabetes. Diabetes 2012; 61:1726-33. [PMID: 22492527 PMCID: PMC3379663 DOI: 10.2337/db11-1516] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The majority of genetic risk variants for type 2 diabetes (T2D) affect insulin secretion, but the mechanisms through which they influence pancreatic islet function remain largely unknown. We functionally characterized human islets to determine secretory, biophysical, and ultrastructural features in relation to genetic risk profiles in diabetic and nondiabetic donors. Islets from donors with T2D exhibited impaired insulin secretion, which was more pronounced in lean than obese diabetic donors. We assessed the impact of 14 disease susceptibility variants on measures of glucose sensing, exocytosis, and structure. Variants near TCF7L2 and ADRA2A were associated with reduced glucose-induced insulin secretion, whereas susceptibility variants near ADRA2A, KCNJ11, KCNQ1, and TCF7L2 were associated with reduced depolarization-evoked insulin exocytosis. KCNQ1, ADRA2A, KCNJ11, HHEX/IDE, and SLC2A2 variants affected granule docking. We combined our results to create a novel genetic risk score for β-cell dysfunction that includes aberrant granule docking, decreased Ca(2+) sensitivity of exocytosis, and reduced insulin release. Individuals with a high risk score displayed an impaired response to intravenous glucose and deteriorating insulin secretion over time. Our results underscore the importance of defects in β-cell exocytosis in T2D and demonstrate the potential of cellular phenotypic characterization in the elucidation of complex genetic disorders.
Collapse
Affiliation(s)
- Anders H. Rosengren
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
- Corresponding author: Anders H. Rosengren, , or Lena Eliasson,
| | - Matthias Braun
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Taman Mahdi
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Sofia A. Andersson
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Mary E. Travers
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
| | - Makoto Shigeto
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
| | - Enming Zhang
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Peter Almgren
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Claes Ladenvall
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Annika S. Axelsson
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Anna Edlund
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Morten Gram Pedersen
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Anna Jonsson
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Reshma Ramracheya
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
| | - Yunzhao Tang
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
- Key Laboratory of Hormones and Development, Ministry of Health, China, Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| | - Jonathan N. Walker
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Oxford National Institute for Health Research Biomedical Research Centre, Churchill Hospital, Oxford, U.K
| | - Amy Barrett
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
| | - Paul R.V. Johnson
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Oxford National Institute for Health Research Biomedical Research Centre, Churchill Hospital, Oxford, U.K
- Nuffield Department of Surgery, University of Oxford, Oxford, U.K
| | - Valeriya Lyssenko
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Mark I. McCarthy
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Oxford National Institute for Health Research Biomedical Research Centre, Churchill Hospital, Oxford, U.K
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
| | - Leif Groop
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Albert Salehi
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Anna L. Gloyn
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Oxford National Institute for Health Research Biomedical Research Centre, Churchill Hospital, Oxford, U.K
| | - Erik Renström
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, U.K
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Oxford National Institute for Health Research Biomedical Research Centre, Churchill Hospital, Oxford, U.K
| | - Lena Eliasson
- Lund University Diabetes Centre, Department of Clinical Sciences Malmo, Lund University, Malmo, Sweden
- Corresponding author: Anders H. Rosengren, , or Lena Eliasson,
| |
Collapse
|
10
|
Abstract
Transplantation therapy for humans is limited by insufficient availability of donor organs and outcomes are complicated by the toxicity of immunosuppressive drugs. Xenotransplantation is a strategy to overcome supply problems. Implantation of tissue obtained early during embryogenesis is a way to reduce immunogenicity of transplants. Insulin-producing cells originating from embryonic pig pancreas obtained very early following initiation of organogenesis [embryonic day 28 (E28)] engraft long-term in non-immune suppressed diabetic rats or rhesus macaques. Recently, we demonstrated engraftment of morphologically similar cells originating from adult porcine islets of Langerhans (islets) in rats previously transplanted with E28 pig pancreatic primordia. Our findings are consistent with induction of tolerance to a cell component of porcine islets induced by previous transplantation of embryonic pig pancreas, a phenomenon we designate organogenetic tolerance. Induction of organogenetic tolerance to porcine islets in humans with diabetes mellitus would enable the use of pigs as islet donors with no host immune suppression requirement. Adaptation of methodology for transplanting embryonic organs other than pancreas so as to induce organogenetic tolerance would revolutionize transplantation therapy.
Collapse
Affiliation(s)
- Marc R Hammerman
- Renal Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
11
|
Goto M, Imura T, Inagaki A, Ogawa N, Yamaya H, Fujimori K, Kurokawa Y, Satomi S. The impact of ischemic stress on the quality of isolated pancreatic islets. Transplant Proc 2011; 42:2040-2. [PMID: 20692402 DOI: 10.1016/j.transproceed.2010.05.101] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Although the ischemic stress of donated organs has been shown to have strong negative effects on islet recovery, the impact on islet quality remains uncertain. In the present study, therefore, we examined the influence of ischemic stress on the expression of inflammatory mediators among isolated islets. MATERIALS AND METHODS Islets were isolated from adult porcine pancreata subjected to 16-hour cold ischemia time (CIT) in addition to 40-minute warm ischemia time (WIT). We evaluated the islet yield, islet loss during the first 24 hours in culture, adenosine diphosphate (ADP)/adenosine triphosphate (ATP) ratio, ATP/DNA ratio, glucose-stimulated respiratory activity, in vivo bioassay, and the expression of inflammatory mediators (tissue factor [TF], [MCP-1], macrophage migration inhibitory factor) on the isolated islets. We also analyzed ATP/DNA ratios of the exocrine tissues during isolation procedures. RESULTS The islet yield, survival rate during culture, and glucose-stimulated respiratory activity were significantly lower in cases of 16-hour CIT plus 40-minute WIT compared with the control group (P < .0001, .0006, and .002, respectively). In contrast, ADP/ATP ratio as well as TF and MCP-1 expressions on the isolated islets were higher among the ischemic group (P = .005, .16, and .005, respectively). During isolation procedures, the ATP/DNA of the exocrine tissues was extremely lower in the ischemic compared to the control group (P < .0001). Notably, however, both ATP/DNA and ADP/ATP ratio of isolated islets were well preserved even in the ischemic group (P = .45 and .40). DISCUSSION These data suggest that ischemic stress during the preservation period negatively affects the energy status of exocrine tissues. Destruction of the exocrine tissues, in combination with warm ischemic stress during the isolation procedures, subsequently decreases isolated islet activity, inducing the expression of inflammatory mediators.
Collapse
Affiliation(s)
- M Goto
- Tohoku University International Advanced Research and Education Organization, Tohoku University, Sendai, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Halley K, Dyson EL, Kaur G, Mital P, Uong PM, Dass B, Crowell SN, Dufour JM. Delivery of a therapeutic protein by immune-privileged Sertoli cells. Cell Transplant 2010; 19:1645-57. [PMID: 20719072 DOI: 10.3727/096368910x516628] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Immune-privileged Sertoli cells survive long term after allogeneic or xenogeneic transplantation without the use of immunosuppressive drugs, suggesting they could be used as a vehicle to deliver therapeutic proteins. As a model to test this, we engineered Sertoli cells to transiently produce basal levels of insulin and then examined their ability to lower blood glucose levels after transplantation into diabetic SCID mice. Mouse and porcine Sertoli cells transduced with a recombinant adenoviral vector containing furin-modified human proinsulin cDNA expressed insulin mRNA and secreted insulin protein. Transplantation of 5-20 million insulin-expressing porcine Sertoli cells into diabetic SCID mice significantly decreased blood glucose levels in a dose-dependent manner, with 20 million Sertoli cells decreasing blood glucose levels to 9.8 ± 2.7 mM. Similar results were obtained when 20 million insulin-positive, BALB/c mouse Sertoli cells were transplanted; blood glucose levels dropped to 6.3 ± 2.4 mM and remained significantly lower for 5 days. To our knowledge, this is the first study to demonstrate Sertoli cells can be engineered to produce and secrete a clinically relevant factor that has a therapeutic effect, thus supporting the concept of using immune-privileged Sertoli cells as a potential vehicle for gene therapy.
Collapse
Affiliation(s)
- Katelyn Halley
- Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Rogers SA, Mohanakumar T, Liapis H, Hammerman MR. Engraftment of cells from porcine islets of Langerhans and normalization of glucose tolerance following transplantation of pig pancreatic primordia in nonimmune-suppressed diabetic rats. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:854-64. [PMID: 20581052 DOI: 10.2353/ajpath.2010.091193] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Transplantation therapy for human diabetes is limited by the toxicity of immunosuppressive drugs. However, even if toxicity can be minimalized, there will still be a shortage of human donor organs. Xenotransplantation of porcine islets may be a strategy to overcome these supply problems. Xenotransplantation in mesentery of pig pancreatic primordia obtained very early during organogenesis [embryonic day 28 (E28)] can obviate the need for immune suppression in rats or rhesus macaques. Here, in rats transplanted previously with E28 pig pancreatic primordia in the mesentery, we show normalization of glucose tolerance in nonimmune-suppressed streptozotocin-diabetic LEW rats and insulin and porcine proinsulin mRNA-expressing cell engraftment in the kidney following implantation of porcine islets beneath the renal capsule. Donor cell engraftment was confirmed using fluorescent in situ hybridization for the porcine X chromosome and electron microscopy. In contrast, cells from islets did not engraft in the kidney without prior transplantation of E28 pig pancreatic primordia in the mesentery. This is the first report of prolonged engraftment and sustained normalization of glucose tolerance following transplantation of porcine islets in nonimmune-suppressed, immune-competent rodents. The data are consistent with tolerance induction to a cell component of porcine islets induced by previous transplantation of E28 pig pancreatic primordia.
Collapse
Affiliation(s)
- Sharon A Rogers
- Renal Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
14
|
Saito Y, Goto M, Maya K, Ogawa N, Fujimori K, Kurokawa Y, Satomi S. Brain Death in Combination with Warm Ischemic Stress during Isolation Procedures Induces the Expression of Crucial Inflammatory Mediators in the Isolated Islets. Cell Transplant 2010; 19:775-82. [DOI: 10.3727/096368910x508889] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tissue factor (TF) and monocyte chemoattractant protein-1 (MCP-1) expressed on the islets have been identified as the main trigger of the instant blood-mediated inflammatory reaction (IBMIR) in islet transplantation. Because the key steps that directly induce TF and MCP-1 remain to be determined, we focused on the influence of brain death (BD) on TF and MCP-1 expression in the pancreatic tissues and isolated islets using a rodent model. TF and MCP-1 mRNA levels in the pancreatic tissues were similar between the BD and the control group. However, TF and MCP-1 mRNA in the fresh islets of the BD group were significantly higher than that of the control group ( p < 0.01). BD may thus be suggested to be of great importance as an initiator of TF and MCP-1 induction in the isolated islets. Furthermore, the upregulation of crucial inflammatory mediators induced by BD could be exacerbated by warm ischemic damage during digestion procedures. In the present study, the islet yield and purity were affected by BD. However, almost no influences were observed with respect to islet viability, indicating that the expression of inflammatory mediators rather than islet viability is more susceptible to BD. According to the change in time course of TF and MCP-1 expression in the isolated islets, the selected time point for islet infusion in current clinical islet transplantation was thus shown to be at its worst level, at least with respect to the damage caused by BD and ischemic stress. In conclusion, BD in combination with warm ischemic stress during isolation procedures induces a high expression of TF and MCP-1 in the isolated islets. In order to reduce the expression of crucial inflammatory mediators in the islet grafts, the management of the pancreas from brain-dead donors with early anti-inflammatory treatments is thus warranted.
Collapse
Affiliation(s)
- Yukihiko Saito
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
| | - Masafumi Goto
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
- Tohoku University International Advanced Research and Education Organization, Tohoku University, Sendai, Japan
| | - Kozue Maya
- Tohoku University International Advanced Research and Education Organization, Tohoku University, Sendai, Japan
| | - Norihiko Ogawa
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
| | - Keisei Fujimori
- Medical Safety Management Office, Tohoku University, Sendai, Japan
| | - Yoshimochi Kurokawa
- Tohoku University Innovation of New Biomedical Engineering Center, Tohoku University, Sendai, Japan
| | - Susumu Satomi
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
| |
Collapse
|
15
|
Rijkelijkhuizen JKRA, Töns A, Terpstra OT, Bouwman E. Transplantation of Long-term Cultured Porcine Islets in the Rat: Prolonged Graft Survival and Recipient Growth on Reduced Immunosuppression. Cell Transplant 2010; 19:387-98. [DOI: 10.3727/096368909x484257] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
To evaluate whether further improvement in porcine islet xenotransplantation is feasible, a number of questions were addressed. Earlier we showed significant improvement in the nude mouse of the porcine islets by selection through long-term culture. Now these islets were tested in the stringent pig-to-rat model. Islets were isolated from adult pigs, cultured for 1.5–3 weeks and transplanted to rats. Possible rejection mechanisms were assessed by interference of the cellular response with cyclosporine A (CsA), blocking macrophages with gadolinium chloride (GdCl), and suppressing the humoral response with cyclophosphamide. Modifications in graft size and condition were analyzed. Untreated control recipients showed primary nonfunction (PNF). CsA treatment could fully overcome PNF and resulted in graft survival from 10 to over 134 days. Rejection was the main cause of function loss. Although rejection could not be prevented by intensifying the induction therapy, increased maintenance immunosuppression effectively blocked rejection, albeit at the expense of toxicity. Blocking the humoral response was ineffective; all grafts showed PNF. In contrast, depletion of macrophages fully prevented PNF. Combination of GdCl and CsA gave no additional effect, and grafts were rejected between 57 and 162 days. Generally, graft survivals were similar to those reported in the literature; however, long-term cultured islets required much less maintenance immunosuppression. Cessation of graft function was not always due to rejection; in some cases “islet exhaustion” was found, possibly caused by discrepancy between the graft size and the rapidly growing recipient. Neither the presence of damaged islet tissue in the graft nor the size of the graft exerted any influence on graft survival. On rejection, no real infiltration of the graft was seen; destruction gradually processed from the outside. The good functional capability of the cultured islets was illustrated by disappearance of the clinical symptoms and increase in body weight, which almost doubled in the long-term survivors.
Collapse
Affiliation(s)
| | - Annemiek Töns
- Department of Surgery, Leiden University Medical Centre, Leiden, The Netherlands
| | - Onno T. Terpstra
- Department of Surgery, Leiden University Medical Centre, Leiden, The Netherlands
| | - Eelco Bouwman
- Department of Surgery, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
16
|
Pepper AR, Gall C, Mazzuca DM, Melling CWJ, White DJG. Diabetic rats and mice are resistant to porcine and human insulin: flawed experimental models for testing islet xenografts. Xenotransplantation 2010; 16:502-10. [PMID: 20042050 DOI: 10.1111/j.1399-3089.2009.00548.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Islet transplantation is potentially a promising therapy for the restoration of carbohydrate control to diabetic patients. However, the global application of islet transplantation requires a ubiquitous source of beta cells. The xenotransplantation of porcine islets would provide such a source. Success in porcine islet xenografting has been achieved in diabetic primates. However, there are few reports of reversal of diabetes with porcine islet xenografts in rodent models of diabetes, relative to the number of successful rodent experiments performed as allografts. Here we report for the first time the inability of porcine (and human) insulin to control blood glucose levels in diabetic rodents determined by a series of dose escalating studies. METHODS Insulin was administered intravenously to streptozotocin induced diabetic Lewis rats, Balb/c and athymic Balb/c mice (n = 5 per group) at the following doses: Group I "physiological dose" (pd) of 0.16 U/kg for a total dose of 40 mU to a 250 g rat. Group II received 0.64 U/kg (4xpd), group III 1.6 U/kg (10xpd) and group IV 6.4 U/kg (40xpd). Blood glucose levels were monitored in each animal at seven time points: 0 (pre-injection), 10 min, 20 min, 30 min, 45 min, 1 h, 1.5 h, 2 h and 3 h post-injection. Serum insulin levels were also determined. RESULTS Diabetic Lewis rats achieved a maximum reduction in blood glucose from 22.1 +/- 1.8mmol/l to 8.0 +/- 3.1 mmol/l (a 63.7% reduction), 90 minutes post-injection of 6.4 U/kg dose of porcine insulin (40xpd). Human insulin was less effective at reducing blood glucose levels in rats than porcine insulin (P < 0.001). Porcine insulin reduced blood glucose levels in Balb/c mice from a mean of 18.2 +/- 2.1 mmol/l to a hypoglycemic minimum of 1.26 +/- 0.18 mmol/l a reduction of 93.0%, 60 min post-injection of the maximum dose of 6.4 U/kg. Balb/c mice were significantly more responsive to porcine insulin than Lewis rats at doses of 0.64 U/kg (P < 0.001), 1.6 U/kg (P < 0.05) and 6.4 U/kg (P < 0.001). Athymic Balb/c nude mice reached a maximum reduction in blood glucose from 21.6 +/- 1.8 mmol/l to 3.6 +/- 0.9 mmol/l (a 83.4% reduction) 120 min post-injection at a dose of 6.4 U/kg. Overall, athymic Balb/c nude mice were more resistant to porcine insulin than immunocompetent Balb/c mice at doses of 0.64 U/kg (P < 0.001), 1.6 U/kg (P < 0.001) and 6.4 U/kg (P < 0.05). Insulin diluent alone marginally increased blood glucose levels in all animals tested. CONCLUSIONS Our results suggest that restoration of normoglycemia in diabetic rodents is not ideal for testing porcine islets xenografts since the reversals of diabetes in these species requires 20 to 40 times the dose of porcine insulin used in humans.
Collapse
Affiliation(s)
- Andrew R Pepper
- Department of Pathology, University of Western Ontario, London, Canada
| | | | | | | | | |
Collapse
|
17
|
Jin SM, Kim KS, Lee SY, Gong CH, Park SK, Yu JE, Yeom SC, Yoon TW, Ha J, Park CG, Kim SJ. Enhanced prediction of porcine islet yield and posttransplant outcome using a combination of quantitative histomorphometric parameters and flow cytometry. Cell Transplant 2009; 19:299-311. [PMID: 19951461 DOI: 10.3727/096368909x481638] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Prediction of islet yield and posttransplant outcome is essential for clinical porcine islet xenotransplantation. Although several histomorphometric parameters of biopsied porcine pancreases are predictive of islet yield, their role in the prediction of in vivo islet potency is unknown. We investigated which histomorphometrical parameter best predicts islet yield and function, and determined whether it enhanced the predictive value of in vitro islet function tests for the prediction of posttransplant outcome. We analyzed the histomorphometry of pancreases from which 60 adult pig islet isolations were obtained. Islet function was assessed using the beta-cell viability index based on flow cytometry analysis, oxygen consumption rate, ADP/ATP ratio, and/or concurrent transplantation into NOD/SCID mice. Receiver operating characteristic (ROC) analysis revealed that only islet equivalent (IEQ)/cm(2) and the number of islets >200 microm in diameter significantly predicted an islet yield of >2000 IEQ/g (p < 0.001 for both) and in vivo islet potency (p = 0.024 and p = 0.019, respectively). Although not predictive of islet yield, a high proportion of large islets (>100 microm in diameter) best predicted diabetes reversal (p = 0.001). Multiple regression analysis revealed that the beta-cell viability index (p = 0.003) and the proportion of islets >100 microm in diameter (p = 0.048) independently predicted mean posttransplant blood glucose level (BGL). When BGL was estimated using both these parameters [area under the ROC curve (AUC), 0.868; 95% confidence interval (CI), 0.730-1.006], it predicted posttransplant outcome more accurately than the beta-cell viability index alone (AUC, 0.742; 95% CI, 0.544-0.939). In conclusion, we identified the best histomorphometric predictors of islet yield and posttransplant outcome. This further enhanced the predictive value of the flow cytometry analysis. These parameters should be useful for predicting islet yield and in vivo potency before clinical adult porcine islet xenotransplantation.
Collapse
Affiliation(s)
- Sang-Man Jin
- Xenotransplantation Research Center, Seoul National University Hospital, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
A novel predictive method for assessing the quality of isolated pancreatic islets using scanning electrochemical microscopy. Transplant Proc 2009; 41:311-3. [PMID: 19249542 DOI: 10.1016/j.transproceed.2008.10.075] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 09/11/2008] [Accepted: 10/20/2008] [Indexed: 11/22/2022]
Abstract
INTRODUCTION The current methods for evaluating islet potency are not useful in clinical transplantation. Therefore, we need reliable, rapid methods enabling accurate prediction of islet quality. MATERIALS AND METHODS We evaluated respiratory activity using scanning electrochemical microscopy (SECM), glucose-stimulated respiratory activity, glucose-stimulated insulin release, ADP/ATP assays, insulin/DNA levels, and Trypan blue exclusion tests as predictive methods for the ability of isolated rat islets to cure syngeneic diabetic rats. RESULTS Although glucose-stimulated respiratory activity, basal respiratory activity, ADP/ATP ratio, and glucose-stimulated insulin release were significantly correlated with the outcome of transplantation into diabetic rats, there was no correlation between outcomes, insulin/DNA ratios, and Trypan blue exclusion tests. The glucose-stimulated respiratory activity in islet preparations that could cure diabetic rats was significantly greater than those unable to cure diabetes. Rat islets with >1.5-fold glucose-stimulated respiratory activity consistently cured diabetic rats, whereas those with a value <1.5 hardly cured any rats. CONCLUSION Measurement of the glucose-stimulated respiratory activity using SECM technique is a novel method that may be useful as a rapid, potent predictor of the outcome of clinical islet transplantation.
Collapse
|
19
|
Saito Y, Goto M, Maya K, Ogawa N, Fujimori K, Kurokawa Y, Satomi S. The Influence of Brain Death on Tissue Factor Expression in the Pancreatic Tissues and Isolated Islets in Rats. Transplant Proc 2009; 41:307-10. [DOI: 10.1016/j.transproceed.2008.10.080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 09/11/2008] [Accepted: 10/15/2008] [Indexed: 10/21/2022]
|
20
|
Kumagai-Braesch M, Ekberg H, Wang F, Osterholm C, Ehrnfelt C, Sharma A, Lindeborg E, Holgersson J, Corbascio M. Anti-LFA-1 Improves Pig Islet Xenograft Function in Diabetic Mice When Long-Term Acceptance Is Induced by CTLA4Ig/Anti-CD40L. Transplantation 2007; 83:1259-67. [PMID: 17496544 DOI: 10.1097/01.tp.0000261722.02697.75] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND It has been previously demonstrated that addition of anti-LFA-1 to a combination of CTLA4Ig and anti-CD40L induces the permanent acceptance of dopaminergic fetal pig xenografts when transplanted into the brain of wild-type mice. The purpose of this study was to test whether this costimulation blockade also can induce acceptance of adult pig islets transplanted to C57BL/6 mice with streptozotocin-induced diabetes. METHODS Recipients were treated with CTLA4Ig/anti-CD40L+/-anti-LFA-1 or isotype control antibodies during the first week after transplantation. Half of the costimulation blockade-treated recipients had their grafts removed after 8 weeks. The other half was observed up to 5 months. RESULTS Recipients treated with CTLA4Ig/anti-CD40L/anti-LFA-1 had significantly lower blood glucose and gained more weight than CTLA4Ig/anti-CD40L-treated recipients. CTLA4Ig/anti-CD40L-treated recipients exhibited unstable blood glucose. IPGTT of these recipients revealed a slow recovery to normal blood glucose levels at week 4. In comparison, CTLA4Ig/anti-CD40L/anti-LFA-1 treated recipients exhibited a significantly superior glucose clearance. CTLA4Ig/anti-CD40L+/-anti-LFA-1 treated recipients did not produce anti-pig IgG, whereas control antibody-treated mice did. CD4+ T cells from costimulation blockade-treated recipients proliferated less than CD4+ T cells from control antibody-treated mice when co-cultured with syngeneic antigen presenting cells loaded with pig islet antigens. CONCLUSIONS CTLA4Ig/anti-CD40L/anti-LFA-1-treated recipients had superior islet function compared with CTLA4Ig/anti-CD40L-treated recipients. However, both costimulation blockade regimens led to islet graft acceptance up to 5 months after a 1-week treatment.
Collapse
Affiliation(s)
- Makiko Kumagai-Braesch
- Department of Transplantation Surgery, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Dai W, Kloner RA. Myocardial regeneration by human amniotic fluid stem cells: Challenges to be overcome. J Mol Cell Cardiol 2007; 42:730-2. [PMID: 17362984 DOI: 10.1016/j.yjmcc.2007.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Accepted: 01/29/2007] [Indexed: 10/23/2022]
|
22
|
Kim JH, Kim HI, Lee KW, Yu JE, Kim SH, Park HS, Park CG, Ihm SH, Ha J, Kim SJ, Lee HK, Ahn C, Park KS. Influence of strain and age differences on the yields of porcine islet isolation: extremely high islet yields from SPF CMS miniature pigs. Xenotransplantation 2007; 14:60-6. [PMID: 17214705 DOI: 10.1111/j.1399-3089.2006.00364.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Porcine pancreas is a potential source of material for islet xenotransplantation. However, the difficulty in isolating islets, because of their fragility and the variability of isolation outcome in donor age and breed, represents a major obstacle to porcine islet xenotransplantation. In this study, we compared the islet isolation yield of specific pathogen-free (SPF) Chicago Medical School (CMS) miniature pigs with that of another miniature pig breed and market pigs from a local slaughterhouse. METHODS Nine adult CMS miniature (ACM) pigs (>12 months), six young CMS miniature (YCM) pigs (6-7 months), four adult Prestige World Genetics (PWG) miniature (APM) pigs (>12 months), and 13 adult market (AM) pigs from a local slaughterhouse were used for islet isolation. RESULTS The islet yield per gram of pancreas from ACM pigs (9589 +/- 2823 IEQ/g) was significantly higher than that from APM pigs (1752 +/- 874 IEQ/g, P < 0.05), AM pigs (1931 +/- 947 IEQ/g, P < 0.05), or YCM pigs (3460 +/- 1985 IEQ/g, P < 0.05). Isolated islets from ACM pigs were significantly larger than those from AM pigs or YCM pigs. The in vitro and in vivo function of isolated islets showed no difference among experimental groups. The pancreases of ACM pigs contained higher mean islet volume density percentages and larger size of islets than those of AM or APM pigs. CONCLUSIONS We isolated extremely high yields of well-functioning islets from ACM pigs bred under SPF conditions. SPF CMS miniature pigs should be one of the best porcine islet donors for clinical porcine islet xenotransplantation.
Collapse
Affiliation(s)
- Jae Hyeon Kim
- Department of Internal Medicine, Seoul National University Boramae Hospital, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Chiavegato A, Bollini S, Pozzobon M, Callegari A, Gasparotto L, Taiani J, Piccoli M, Lenzini E, Gerosa G, Vendramin I, Cozzi E, Angelini A, Iop L, Zanon GF, Atala A, De Coppi P, Sartore S. Human amniotic fluid-derived stem cells are rejected after transplantation in the myocardium of normal, ischemic, immuno-suppressed or immuno-deficient rat. J Mol Cell Cardiol 2006; 42:746-59. [PMID: 17300799 DOI: 10.1016/j.yjmcc.2006.12.008] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Revised: 11/23/2006] [Accepted: 12/18/2006] [Indexed: 01/14/2023]
Abstract
Human amniotic fluid-derived stem (AFS) cells, similarly to embryonic stem cells, could possess privileged immunological characteristics suitable for a successful transplantation even in a discordant xenograft system. We investigated whether AFS cells could be fruitfully used in a rat model of myocardial infarction. c-kit immunomagnetic-sorted AFS cells were characterized by flow cytometric analysis and cytospins as well as reverse-transcription polymerase chain reaction, Western blotting and immunocytochemistry for cardiovascular differentiation markers. In vitro, AFS cell phenotypic conversion was assayed by cardiovascular-specific induction media or co-cultured with rat neonatal cardiomyocytes. AFS cells showed mRNAs and/or protein for endothelial (angiopoietin, CD146) and smooth muscle (smoothelin) cells, and cardiomyocyte (Nkx2.5, MLC-2v, GATA-4, beta-MyHC) markers. Acquisition of a cardiomyocyte-like phenotype in rare AFS cells could be seen only in co-cultures with rat neonatal cells. In vivo, AFS cells xenotransplantated in a rat model of myocardial infarction, with or without cyclosporine treatment, or in intact heart from immuno-competent or immuno-deficient animals were acutely rejected due to the different recruitment of recipient CD4(+), CD8(+) T and B lymphocytes, NK cells and macrophages. This reaction is most likely to be linked to expression of B7 co-stimulatory molecules CD80 and CD86 as well as macrophage marker CD68 on AFS cells. Xenotransplanted AFS cells gave also rise in some animals to cell masses in the subendocardium and myocardium suggestive of a process of chondro-osteogenic differentiation. Despite AFS cells in vitro can differentiate to some extent to cells of cardiovascular lineages, their in vivo use in xenotransplantation for cell therapy of myocardial infarction is hampered by their peculiar immunogenic properties and phenotypic instability.
Collapse
Affiliation(s)
- Angela Chiavegato
- Deparment of Biomedical Sciences, University of Padua, Viale G. Colombo, 3 35121 Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Goto M, Holgersson J, Kumagai-Braesch M, Korsgren O. The ADP/ATP ratio: A novel predictive assay for quality assessment of isolated pancreatic islets. Am J Transplant 2006; 6:2483-7. [PMID: 16869808 DOI: 10.1111/j.1600-6143.2006.01474.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The current standard assays for islet product release criteria are unable to predict the outcome after clinical islet transplantation. Therefore, establishment of reliable and rapid assays enabling pre-transplantation prediction of islet potency is warranted. In the present study, we have evaluated the adenosine diphosphate (ADP)/adenosine triphosphate (ATP) test, the glucose-stimulated insulin release, the loss of islets during the first 24 h in culture, and the insulin/deoxyribonucleic acid as predictive assays for the ability of isolated porcine islets to cure athymic mice with streptozotocin-induced diabetes. From the results presented, it is concluded that the measurement of the ADP/ATP ratio was the only test that correlated with transplantation outcome. In summary, we propose that the ADP/ATP assay is worthwhile as applied to human islet transplantation and seek to validate it as a rapid and potent predictor of transplant outcome.
Collapse
Affiliation(s)
- M Goto
- Tohoku University, Biomedical Engineering Research Organization, Sendai, Japan.
| | | | | | | |
Collapse
|
25
|
Lindeborg E, Kumagai-Braesch M, Möller E. Phenotypic and functional characterization of human T cell clones indirectly activated against adult pig islet cells. Xenotransplantation 2006; 13:41-52. [PMID: 16497211 DOI: 10.1111/j.1399-3089.2005.00257.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Xenotransplanted patients produce xenospecific IgG1 antibodies directed against epitopes other than Galalpha1,3Gal. IgG1 antibody production is believed to be dependent upon T cell help. Therefore, as a natural continuation of our work aimed at characterizing the xenoimmune antibody response against pig islet cells, we have also examined the T cell response. T cell reactivity against islet cells is believed to result from indirect antigen presentation, and our in vitro study was designed to mimic the response in vivo. The main purpose of this study was to characterize the phenotype, the immunological specificity and the functional capacity of indirectly activated T cell clones, reactive against pig islet cell antigens. MATERIALS AND METHODS Human T cell clones, activated against pig islet cells in the presence of autologous antigen-presenting cells, were produced from limiting dilutions of bulk cultures. Clonality was investigated by T cell receptor Vbeta (TcRVbeta) expression analysis. Clonal specificity was studied in proliferation assays using different pig cells as stimulators. ELISpot experiments were performed to detect cytokine production patterns. The cytotoxic capacity of the clones was assessed using standard cell-mediated lysis tests and different porcine and human target cells. Several long-term bulk cultures of human lymphocytes, indirectly activated against pig islet cells, maintained for up to 60 days, were used as a control for possible bias in the selection of the clones. RESULTS Nineteen CD4+ TcRValphabeta+ T cell clones were recovered. No activation of natural killer T cells or gammadelta-T cells was recorded. There was no bias in the TcRVbeta-usage. The immunological specificity differed between clones; some were specifically reactive against pig islet cell antigens, while others were reactive with antigens present on a variety of pig cells. All clones produced a broad spectrum of cytokines, e.g. interferon (IFN)-gamma, tumor necrosis factor (TNF)-alpha, interleukin (IL)5, IL10 and IL13, with no evidence of bias for a particular phenotype. None of the T cell clones were cytotoxic against pig islet cells, but two clones were cytotoxic against pig phytohemagglutinin (PHA)-blasts. CONCLUSION The analysis of several, indirectly activated, human CD4+ T cell clones shows that the response against pig islet cells is heterogeneous both with regard to immunological specificity and functional characteristics. This heterogeneity was further confirmed by analysis of the long-term bulk cultures of human lymphocytes, indirectly activated against pig islet cells.
Collapse
Affiliation(s)
- Ellinor Lindeborg
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Huddinge, Sweden.
| | | | | |
Collapse
|
26
|
He Z, Wang F, Kumagai-Braesch M, Permert J, Holgersson J. Long-term gene expression and metabolic control exerted by lentivirus-transduced pancreatic islets. Xenotransplantation 2006; 13:195-203. [PMID: 16756562 DOI: 10.1111/j.1399-3089.2006.00274.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Genetic modification of non-human islets before transplantation may provide means by which they can escape immunity and, thus, be used in a human host. To accomplish this, efficient gene transfer methods are needed. Lentiviral vectors are transgene vehicles capable of stably transducing a variety of primary, post-mitotic cells including islets. METHODS We investigated whether lentiviral transduction impaired rat pancreatic islet function long term. Following transduction, the gross morphology, viability and in vitro functionality of islets were evaluated by microscopy, adenylate nucleotide and insulin secretion assays, respectively. Further, in vivo functionality of transduced islets was assessed by transplanting the islets under the kidney capsule of diabetic nude mice. RESULTS All transduced islets contained green fluorescent protein (GFP)-positive cells. In single cell suspensions prepared from transduced islets, 33+/-8% (n = 3) of dispersed islet cells were GFP-positive. The ADP/ATP ratio was 0.07+/-0.01 for transduced islets and 0.06+/-0.01 for controls (normal range <0.11). No morphological changes were observed in transduced islets. Further, basal insulin secretion was comparable between the two islet groups. When transduced and non-transduced islets were challenged with insulin secretagogues, they showed similar increases in insulin release. Transduced and non-transduced islets were equally effective in normalizing blood glucose when transplanted into diabetic nude mice. Euglycemia was maintained for 8 weeks until the graft-bearing kidney was removed. Intense green fluorescence was seen in removed islet grafts. Histology revealed preserved islet morphology, with abundant insulin-producing cells, few apoptotic cells and infiltrating leukocytes in both transduced and non-transduced grafts. CONCLUSIONS Lentivirus transduction does not affect islet morphology or function. Lentiviral vectors will allow genetic modifications to be performed in islets before transplantation--modifications that can improve engraftment and/or prevent xenograft rejection.
Collapse
Affiliation(s)
- Zhong He
- Division of Clinical Immunology, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | | | | | | | | |
Collapse
|
27
|
Rijkelijkhuizen JKRA, van der Burg MPM, Töns A, Terpstra OT, Bouwman E. Pretransplant culture selects for high-quality porcine islets. Pancreas 2006; 32:403-7. [PMID: 16670623 DOI: 10.1097/01.mpa.0000220866.87658.b2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The pig is generally considered a suitable alternative donor for clinical islet transplantation. However, adult pig islets are difficult to isolate and culture, often behave variably in in vitro assays, and do not consistently cure diabetic nude mice. In this study, we compared the in vivo function of freshly isolated and cultured adult porcine islets by transplantation in diabetic nude mice. METHODS Freshly isolated and cultured islets were transplanted in different doses to diabetic nude mice (N = 48). RESULTS Average islet yield was 1924 islet-equivalents per gram of pancreas, purity 96%, and the viability that was measured by acridine orange and propidium iodide was greater than 80% in all freshly isolated islet preparations. Grafts of freshly isolated islets failed to reduce hyperglycemia in 17 of 18 recipients. Although after 1 day of culture islet recovery was only 21%, grafts of these islets cured 12 of 17 mice. After 7 to 14 days of culture, the recovery had decreased to 11%; however, these islets reversed hyperglycemia in all mice (13/13) and showed shorter time-to-normoglycemia and more tightly regulated blood glucose. CONCLUSIONS Although freshly isolated adult porcine islets survive culture and transplantation poorly, islets selected by prolonged culture are of high potential.
Collapse
|
28
|
Figliuzzi M, Plati T, Cornolti R, Adobati F, Fagiani A, Rossi L, Remuzzi G, Remuzzi A. Biocompatibility and function of microencapsulated pancreatic islets. Acta Biomater 2006; 2:221-7. [PMID: 16701881 DOI: 10.1016/j.actbio.2005.12.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2005] [Revised: 10/24/2005] [Accepted: 12/01/2005] [Indexed: 11/21/2022]
Abstract
Encapsulation of pancreatic islets in alginate is used to protect against xenogenic rejection in different animal models. In this study, several factors, including differences in alginate composition, the presence or absence of xenogenic islet tissue and a transient immunosuppression, were investigated in a model of bovine islet transplantation in rats. A pure alginate with predominantly guluronic acid (Manugel) and an ultrapure low viscosity guluronic acid alginate (UP-LVG) were used. When microcapsules of Manugel or UP-LVG containing 16,000 bovine islet equivalents were transplanted in diabetic rats, we observed normoglycemia for 8.3+/-0.7 (range 6-12 days) and 7.5+/-0.2 days (range 7-8 days) on average, respectively. To ameliorate immunoprotection of alginate microcapsules we repeated the same experiments using transient immunosuppressive therapy. Low doses of cyclosporin A (CyA) administered for 18 days after implantation increased the time in normoglycemia, which averaged 27+/-3 days (range 8-55 days) in Manugel capsules while in UP-LVG capsules it averaged 18+/-8 days (range 3-39 days). The surface of recovered capsules showed less capsules free of overgrowth in Manugel with respect to UP-LVG alginate. These data were comparable with those observed in empty microcapsules similarly implanted, indicating that the capsular overgrowth was not promoted by the presence of xenogenic islet tissue. In recovered Manugel capsules the percentage of capsules without fibrotic overgrowth was higher than that observed without CyA. The same observation was made in empty capsules. These observations indicate that a combination of a highly purified alginate and short-term immunosuppression prolong islet function in a model of xenotransplantation.
Collapse
Affiliation(s)
- Marina Figliuzzi
- Department of Biomedical Engineering, Mario Negri Institute for Pharmacological Research, Via Gavazzeni, 11, 24125 Bergamo, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
N/A, 沈 滨, 刘 晓. N/A. Shijie Huaren Xiaohua Zazhi 2005; 13:1885-1888. [DOI: 10.11569/wcjd.v13.i15.1885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
30
|
Fotiadis C, Xekouki P, Papalois AE, Antonakis PT, Sfiniadakis I, Flogeras D, Karampela E, Zografos G. Effects of mycophenolate mofetil vs cyclosporine administration on graft survival and function after islet allotransplantation in diabetic rats. World J Gastroenterol 2005; 11:2733-8. [PMID: 15884112 PMCID: PMC4305906 DOI: 10.3748/wjg.v11.i18.2733] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To develop an experimental model of islet allotran-splantation in diabetic rats and to determine the positive or adverse effects of MMF as a single agent.
METHODS: Thirty-six male Wistar rats and 18 male Lewis rats were used as recipients and donors respectively. Diabetes was induced by the use of streptozotocin (60 mg/kg) intraperitoneally. Unpurified islets were isolated using the collagenase digestion technique and transplanted into the splenic parenchyma. The recipients were randomly assigned to one of the following three groups: group A (control group) had no immunosuppression; group B received cyclosporine (CsA) (5 mg/kg); group C received mycophenolate mofetil (MMF) (20 mg/kg). The animals were killed on the 12th d. Blood and grafted tissues were obtained for laboratory and histological assessment.
RESULTS: Median allograft survival was significantly higher in the two therapy groups than that in the controls (10 and 12 d for CsA and MMF respectively vs 0 d for the control group, P<0.01). No difference in allograft survival between the CsA and MMF groups was found. However, MMF had less renal and hepatic toxicity and allowed weight gain.
CONCLUSION: Monotherapy with MMF for immunosu-ppression was safe in an experimental model of islet allotransplantation and was equally effective with cyclosporine, with less toxicity.
Collapse
Affiliation(s)
- Constantin Fotiadis
- 3rd Department of Surgery, Athens University Medical School, 12 Diligiani str, 14561 Athens, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Animal models have been used extensively in diabetes research. Early studies used pancreatectomised dogs to confirm the central role of the pancreas in glucose homeostasis, culminating in the discovery and purification of insulin. Today, animal experimentation is contentious and subject to legal and ethical restrictions that vary throughout the world. Most experiments are carried out on rodents, although some studies are still performed on larger animals. Several toxins, including streptozotocin and alloxan, induce hyperglycaemia in rats and mice. Selective inbreeding has produced several strains of animal that are considered reasonable models of Type 1 diabetes, Type 2 diabetes and related phenotypes such as obesity and insulin resistance. Apart from their use in studying the pathogenesis of the disease and its complications, all new treatments for diabetes, including islet cell transplantation and preventative strategies, are initially investigated in animals. In recent years, molecular biological techniques have produced a large number of new animal models for the study of diabetes, including knock-in, generalized knock-out and tissue-specific knockout mice.
Collapse
Affiliation(s)
- D A Rees
- Department of Medicine, University of Wales College of Medicine, Cardiff, UK
| | | |
Collapse
|
32
|
Ehrnfelt C, Kumagai-Braesch M, Uzunel M, Holgersson J. Adult porcine islets produce MCP-1 and recruit human monocytes in vitro. Xenotransplantation 2004; 11:184-94. [PMID: 14962280 DOI: 10.1046/j.1399-3089.2003.00104.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Type 1 diabetes can be cured by transplantation of isolated pancreatic islets. Because of the shortage of human donor tissue, adult porcine islets (APIs) constitute a possible alternative tissue source. Upon intraportal injection, islets are subjected to an instant blood-mediated inflammatory reaction (IBMIR) leading to blood clotting, leukocyte islet-infiltration, islet damage and insulin release. Xenogeneic islets surviving IBMIR are rejected in a cellular process involving CD4(+) T lymphocytes and macrophages. We have investigated whether APIs themselves produce and secrete chemokines and/or inflammatory cytokines that may contribute to IBMIR and/or cell-mediated rejection. APIs, cultured for 1, 4, 8 and 11 days post-isolation, expressed mRNA for monocyte chemoattractant protein-1 (MCP-1), IL-1beta and TNF-alpha. API culture supernatants induced migration of human monocytes, which was significantly blocked by an anti-human MCP-1 antibody (Ab). Immunohistochemistry revealed MCP-1 in the cytoplasm of alpha- and beta-cells in isolated islets and in islets in situ. However, APIs or their supernatants were not able to activate human aortic endothelial cells (HAECs) in vitro, and neither IL-1beta nor TNF-alpha were detected by enzyme-linked immunosorbent assay (ELISA) in API culture supernatants. Both recombinant porcine IL-1beta and TNF-alpha were able to activate human endothelial cells (ECs) inducing CD62E and CD106 expression as analyzed by flow cytometry. In conclusion, MCP-1 secreted by APIs may contribute to both IBMIR and rejection by attracting monocytes into the islet; monocytes which upon transformation into macrophages will potentiate antigen presentation and execute islet rejection.
Collapse
Affiliation(s)
- Cecilia Ehrnfelt
- Division of Clinical Immunology, Karolinska Institutet, Huddinge University Hospital AB, Stockholm, Sweden.
| | | | | | | |
Collapse
|
33
|
Lindeborg E, Kumagai-Braesch M, Tibell A, Christensson B, Möller E. Biological activity of pig islet-cell reactive IgG antibodies in xenotransplanted diabetic patients. Xenotransplantation 2004; 11:457-70. [PMID: 15303983 DOI: 10.1111/j.1399-3089.2004.00168.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND The IgG antibody response in type I diabetic patients, transplanted with fetal pig islet-like cell-clusters, was investigated using purified immunoglobulin G (IgG) fractions from sera collected 7 to 9 yr after transplantation. From our earlier studies, we knew that the immunological specificities of xenoreactive IgG1 and IgG2 antibodies are different, and that IgG1 antibodies, in contrast to the IgG2 population, are mainly directed against non-Galalpha1,3Gal epitopes. In this study our goal was to establish whether xenoreactive IgG1 and IgG2 antibodies react with pig islet cells and, if so, to identify the target cell type, the biological function as well as the specificity of such antibodies for islet cell antigens. Sera from xenotransplanted patients were compared with those of patients with diabetes, selected for high titres of islet-cell specific autoantibodies. METHODS IgG antibody fractions from patient sera were purified on a protein G column. Surface expression of target antigens was studied using flow cytometry as well as immunofluorescence microscopy. The biological function of islet-cell reactive sera was tested using antibody dependent cellular cytotoxicity with both xenogeneic adult pig islet cells and allogeneic human islet cells as targets. Antibody specificity was assessed using 2D Western blots with both fetal and adult pig islet as well as human islet cell antigenic preparations. RESULTS Some of the diabetic patients, who have been transplanted with xenogeneic fetal pig islet cells, continue to produce xenospecific IgG1 and IgG2 antibodies for 7 to 9 yr post-transplantation. A separate analysis of IgG1 and IgG2 antibodies showed that IgG1 antibodies react with pig islet beta cells, whereas IgG2 antibodies mainly react with non-endocrine pig cells. Such antibodies are xenospecific, as they were found to mediate antibody-dependent cellular cytotoxicity of adult pig, but not human islet target cells. The reverse was true for antibodies from non-transplanted diabetic patients with high titres of autoantibodies against beta cells. Fluorescence analysis as well as 2D gel Western blots revealed that the reactivity was variable between patient samples, indicating that target antigens for non-Galalpha1,3Gal-specific antibodies are heterogeneous. CONCLUSION Thus, xenotransplantation of diabetic patients induces islet-beta cell reactive xenospecific IgG1 antibodies, which are biologically active and can mediate antibody-dependent cellular cytotoxicity of pig islet cells.
Collapse
Affiliation(s)
- Ellinor Lindeborg
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.
| | | | | | | | | |
Collapse
|
34
|
Goto M, Maeda A, Elfman L, Suling KM, Wood JC, Patience C, Groth CG, Wennberg L. No transmission of porcine endogenous retrovirus after transplantation of adult porcine islets into diabetic nude mice and immunosuppressed rats. Xenotransplantation 2004; 11:340-6. [PMID: 15196128 DOI: 10.1111/j.1399-3089.2004.00144.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The aim of this study was to investigate whether transmission of porcine endogenous retrovirus (PERV) occurs in a model of diabetes reversal by the xenotransplantation of adult porcine islets (APIs) into immunoincompetent diabetic rodents. METHODS Black-6 nu/nu mice and Lewis rats were immunosuppressed with cyclosporin A (CsA) and FTY 720, and rendered diabetic with streptozotocin. Purified APIs were transplanted into the renal subcapsular space; 5,000 islet equivalents (IEQs) were used in the nude mice (n = 4) and 40,000 IEQs in the rats (n = 4). The nude mice were sacrificed at 75 days after transplantation. In order to confirm chronic xenograft function, the graft-bearing kidney was removed prior to sacrifice. The rats were followed until xenograft rejection, at which time they were sacrificed. Immediately after sacrifice, tissue samples (liver, spleen, and small intestine) were taken for analysis. Quantitative polymerase chain reaction (PCR) was used to assess evidence of PERV transmission, and porcine cell chimerism. RESULTS All animals became normoglycemic within 48 h of transplantation. The nude mice remained normoglycemic during the 75-day study period, with removal of the graft-bearing kidney resulting in prompt hyperglycemia. The rats remained normoglycemic until xenograft rejection, which occurred at 66 +/- 28 days. Despite the evidence of porcine cell microchimerism in recipients, real-time PCR detected no evidence of PERV transmission in any of the tissue specimens tested. CONCLUSIONS There was no evidence of PERV transmission following transplantation of pig islets into diabetic nude mice and immunosuppressed rats.
Collapse
Affiliation(s)
- Masafumi Goto
- Department of Transplantation Surgery, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Tang TH, Li CL, Li X, Jiang FQ, Zhang YK, Ren HQ, Su SS, Jiang GS. Immune tolerance in pancreatic islet xenotransplantation. World J Gastroenterol 2004; 10:1457-61. [PMID: 15133853 PMCID: PMC4656284 DOI: 10.3748/wjg.v10.i10.1457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To observe the effect of tail vein injection with donor hepatocytes and/or splenocytes on the islet xenotransplantation rejection.
METHODS: New-born male pigs and BALB/C mice were selected as donors and recipients respectively. Islet xenotransplantation was performed in recipients just after the third time of tail vein injection with donor hepatocytes and/or splenocytes. Macrophage phagocytosis, NK(natural killing cell) killing activity, T lymphocyte transforming function of spleen cells, antibody forming function of B lymphocytes, and T lymphocyte subsets were taken to monitor transplantation rejection. The effects of this kind of transplantation were indicated as variation of blood glucose and survival days of recipients.
RESULTS: The results showed that streptozotocin (STZ) could induce diabetes mellitus models of mice. The pre-injection of donor hepatocytes, splenocytes or their mixture by tail vein injection was effective in preventing donor islet transplantation from rejection, which was demonstrated by the above-mentioned immunological marks. Each group of transplantation could decrease blood glucose in recipients and increase survival days. Pre-injection of mixture of donor hepatocytes and splenocytes was more effective in preventing rejection as compared with that of donor hepatocyte or splenocyte pre-injection respectively.
CONCLUSION: Pre-injection of donor hepatocytes, splenocytes or their mixture before donor islet transplantation is a good way in preventing rejection.
Collapse
Affiliation(s)
- Tian-Hua Tang
- Department of Hemato-oncology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jingshi Road 89, Jinan 250062, Shandong Province, China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Goto M, Johansson H, Maeda A, Elgue G, Korsgren O, Nilsson B. Low molecular weight dextran sulfate prevents the instant blood-mediated inflammatory reaction induced by adult porcine islets. Transplantation 2004; 77:741-7. [PMID: 15021838 DOI: 10.1097/01.tp.0000114872.26990.4f] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND One of the main obstacles to clinical islet xenotransplantation is the injurious instant blood-mediated inflammatory reaction (IBMIR) that causes rapid binding of platelets to the islet surface, activation of the coagulation and complement systems, and leukocyte infiltration of the islets when the islets are exposed to blood. METHODS This study assesses the effect of low molecular weight dextran sulfate (LMW-DS) on IBMIR induced by porcine islets in an in vitro tubing loop assay using human blood and in an in vivo model using diabetic athymic mice. RESULTS In vitro experiments demonstrated that platelet consumption, coagulation, and complement activation were already reduced in the presence of LMW-DS at 0.01 mg/mL, and that at 0.1 mg/mL, LMW-DS prevented IBMIR. Immunohistochemical investigation showed that the leukocyte infiltration was abrogated at the highest dose. In vivo experiments showed that the transplanted pig islets survived for a significantly longer period in recipients treated with LMW-DS, and morphologic examination of transplanted islets showed a reduction in IBMIR analogous to that demonstrated by in vitro studies. CONCLUSIONS Given that LMW-DS has been used in clinical studies without serious adverse reactions, it has potential as a drug candidate that can control the strong innate immune response induced by pig islets when transplanted through the portal vein.
Collapse
Affiliation(s)
- Masafumi Goto
- Department of Radiology, Oncology and Clinical Immunology, Division of Clinical Immunology, the Rudbeck Laboratory, Uppsala, Sweden.
| | | | | | | | | | | |
Collapse
|
37
|
Rogers SA, Chen F, Talcott M, Hammerman MR. Islet cell engraftment and control of diabetes in rats after transplantation of pig pancreatic anlagen. Am J Physiol Endocrinol Metab 2004; 286:E502-9. [PMID: 14678953 DOI: 10.1152/ajpendo.00445.2003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The insufficient supply of tissue, loss posttransplantation, and limited potential for expansion of beta-cells restrict the use of islet allotransplantation for diabetes. A way to overcome the supply and expansion problems is to xenotransplant embryonic tissue. We have shown that whole rat pancreatic anlagen isotransplanted into the omentum of rats, or xenotransplanted into costimulatory blocked mice, undergo growth and differentiate into islets surrounded by stoma without exocrine tissue. Isotransplants normalize glucose tolerance in diabetic hosts. Here, we show that embryonic day 29 porcine pancreas transplanted into the omentum of adult diabetic rats undergoes endocrine tissue differentiation over 20 wk and normalizes body weights and glucose tolerance. Unlike rat-to-rodent transplants, individual alpha- and beta-cells engraft without a stromal component, and no immunosuppression is required for pig-to-rat transplants. Herein is described a novel means to effect the xenotransplantation of individual islet cells across a highly disparate barrier.
Collapse
Affiliation(s)
- Sharon A Rogers
- Renal Division, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
38
|
Abstract
The number of donor human pancreas organs that can be transplanted directly or used for islet of Langerhans isolation is limited. We and others have shown that it is possible to 'grow' new pancreatic tissue in situ by transplanting embryonic organ-specific pancreatic precursor cells. This technology takes advantage of the fact that selective development of islets takes place post transplantation of embryonic pancreas and that the developing organ can attract its blood supply from an appropriate vascular bed post transplantation, enabling the transplantation of pancreas in 'cellular' form. Whole pancreatic anlagen implanted into a host peritoneum develop into a novel organ consisting of functional islets of Langerhans surrounded by stroma or individual alpha and beta cells within omental fat. Transplantation of developing pancreas to achieve organogenesis of its endocrine components could lead to a novel treatment for diabetes mellitus.
Collapse
Affiliation(s)
- Marc R Hammerman
- Endocrine Division, Renal Division, Box 8126, Departments of Medicine, and Cell Biology and Physiology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA.
| |
Collapse
|
39
|
Zhang L, Qi Z, Wu D, Shan S, Ekberg H. Additive Effects of Leflunomide and Tacrolimus in Prevention of Islet Xenograft Rejection. Scand J Immunol 2004; 59:255-60. [PMID: 15030575 DOI: 10.1111/j.0300-9475.2004.01401.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Leflunomide is a low molecular weight immunosuppressive drug which inhibits the enzymes dehydroorotate dehydrogenase and protein tyrosine kinase, both of which are important components in the immune response. As the mechanisms of action of leflunomide and tacrolimus are different, we postulated an additive or synergistic effect of the two drugs and investigated the effects of leflunomide alone, or in combination with a suboptimal dose of tacrolimus, on xenogeneic islet transplantation in a rat-to-mouse model. A total of 1200-1500 rat islets were transplanted under the left kidney capsule of streptozotocin-induced diabetic BALB/c mice. The median survival time (MST) of the untreated group was 6 days. Leflunomide at 5, 10 and 20 mg/kg/d administrated for 10 days significantly prolonged MST to 10, 16 and 20 days. A dose of tacrolimus (2 mg/kg/d) was associated with a graft survival of 9 (range 6-12) days; most grafts rejected during ongoing therapy. When tacrolimus (2 mg/kg/d) was combined with leflunomide (10 mg/kg/d), the survival time of the islet xenografts was increased further to 22 days, significantly longer than with leflunomide or tacrolimus alone. In summary, our findings demonstrate that leflunomide prolonged xenogeneic islet graft survival, and that its immunosuppressive effect was improved when combined with tacrolimus.
Collapse
Affiliation(s)
- L Zhang
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | | | | | | | | |
Collapse
|
40
|
Goto M, Groth CG, Nilsson B, Korsgren O. Intraportal pig islet xenotransplantation into athymic mice as an in vivo model for the study of the instant blood-mediated inflammatory reaction. Xenotransplantation 2004; 11:195-202. [PMID: 14962281 DOI: 10.1046/j.1399-3089.2003.00107.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
One of the main obstacles to successful intraportal islet transplantation is the instant blood-mediated inflammatory reaction (IBMIR) elicited by the isolated islets when exposed to fresh human blood. In the present study, we investigated whether intraportal transplantation of pig islets into diabetic athymic mice could be used as a small animal model to study xenogeneic IBMIR in vivo. Adult porcine islets (APIs) or rat islets were implanted into the portal vein or under the renal subcapsular space of diabetic athymic mice. Graft survival and morphology were evaluated by measuring blood glucose levels and by performing immunohistochemical staining, respectively. Transplantation of rat islets, irrespective of implantation site, cured all diabetic athymic mice. APIs transplanted subcapsularly also cured all diabetic athymic mice, while none of the animals transplanted with an equivalent amount of APIs via the portal vein remained normoglycemic for more than 10 days after transplantation. Immunohistochemical staining on day 7 showed that most of intraportally transplanted APIs were entrapped in clots and infiltrated with CD11b+ leukocytes. Intraportal transplantation of APIs into athymic mice induced IBMIR, thus providing a small animal model for studying xenogeneic IBMIR.
Collapse
Affiliation(s)
- Masafumi Goto
- Department of Radiology, Oncology, and Clinical Immunology, Uppsala University Hospital, Uppsala, Sweden.
| | | | | | | |
Collapse
|
41
|
MacKenzie DA, Hullett DA, Sollinger HW. Xenogeneic transplantation of porcine islets: an overview. Transplantation 2003; 76:887-91. [PMID: 14508349 DOI: 10.1097/01.tp.0000087114.18315.17] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The extreme demand for human organs or tissues for transplantation has driven the search for viable alternatives. Pigs are considered a possible source of tissue for a number of reasons including shared physiology, plentiful supply, short gestation, and, more recently, the generation of transgenic animals. Porcine islets show promise as a source of islets for the treatment of type 1 diabetes mellitus. Porcine islets regulate glucose levels in the same physiologic range as humans, and porcine insulin has been used for years as an exogenous source of insulin for glucose control. In this review, we discuss the advantages and disadvantages of the use of adult or neonatal porcine islets, the immunologic challenges facing transplantation of xenogeneic islets, and the concerns regarding transmission of infectious agents between species. Porcine islets isolated from both adult and neonatal pigs are capable of restoring euglycemia in experimental animal models of diabetes. Adult islets are more difficult to isolate, whereas neonatal islets have great proliferation potential but require several weeks to function posttransplantation. Xenogeneic islets are susceptible to complement-mediated lysis after the binding of preformed natural antibodies and cellular immunity involving both macrophages and CD4+ T cells. In addition, the potential for transmission of porcine endogenous retroviruses, porcine cytomegalovirus, and porcine lymphotropic herpesvirus type 1 are all concerns that must be addressed. Despite the challenges facing xenotransplantation, the extreme need for donor organs and tissues continues to drive progress toward overcoming the unique issues associated with transplantation between species.
Collapse
Affiliation(s)
- Debra A MacKenzie
- University of Wisconsin, Department of Surgery, Clinical Sciences Center, Madison, Wisconsin 53792, USA
| | | | | |
Collapse
|
42
|
Johansson U, Olsson A, Gabrielsson S, Nilsson B, Korsgren O. Inflammatory mediators expressed in human islets of Langerhans: implications for islet transplantation. Biochem Biophys Res Commun 2003; 308:474-9. [PMID: 12914774 DOI: 10.1016/s0006-291x(03)01392-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Expression of immune modulating mediators in human Islets of Langerhans could have important implications for development of autoimmunity in type 1 diabetes and influence the outcome of clinical islet transplantation. Islets obtained from five donors were analyzed at various times after isolation using cDNA array technology. The Atlas Human Cytokine/Receptor and Hematology/Immunology nylon membranes representing 268 genes and 406, respectively, were used and the relative expression of each gene analyzed. Of the 51 gene products identified, high mRNA expression of MCP-1, MIF, VEGF, and thymosin beta-10 was detected in all islet samples. IL-8, IL-1-beta, IL-5R, and INF-gamma antagonist were expressed in islets cultured for 2 days. IL-2R was expressed in islets cultured for more than 6 days. In conclusion, several inflammatory mediators were expressed in isolated islets, particularly at an early stage after isolation, indicating that a few days of culture could be beneficial for the outcome of islet transplantation.
Collapse
Affiliation(s)
- Ulrika Johansson
- Department of Clinical Immunology, The Rudbeck Laboratory, Uppsala University C11, Sweden.
| | | | | | | | | |
Collapse
|
43
|
Maeda A, Goto M, Zhang J, Bennet W, Groth CG, Korsgren O, Wennberg L. Immunosuppression with FTY720 and cyclosporine A inhibits rejection of adult porcine islet xenografts in rats. Transplantation 2003; 75:1409-14. [PMID: 12717240 DOI: 10.1097/01.tp.0000061770.39569.7f] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Our aim was to evaluate the effect of FTY720 in discordant islet xenotransplantation. METHODS Fetal porcine islet-like cell clusters (ICCs) were transplanted into normoglycemic rats that were either left untreated or treated with FTY720 only, with FTY720 plus cyclosporine A (CsA) or with CsA only. Twelve or 24 days after transplantation, graft morphology was evaluated immunohistochemically. Furthermore, adult porcine islets (APIs) were transplanted into diabetic rats immunosuppressed with FTY720 plus CsA. Blood glucose and porcine C-peptide levels were monitored. RESULTS In untreated rats, the ICC xenografts were completely rejected after 12 days. Treatment with CsA had only a marginal effect on the rejection. In animals given FTY720, only the number of infiltrating cells was somewhat reduced. However, at 12 days, no intact ICCs remained. Immunosuppression with FTY720 plus CsA had a marked inhibitory effect on islet xenograft rejection and plentiful morphologically intact ICCs remained. Twelve days after transplantation, only occasional macrophages and T cells could be detected. At 24 days after transplantation, the findings were similar. Furthermore, diabetic rats transplanted with APIs and immunosuppressed with FTY720 plus CsA remained normoglycemic for 53.0+/-15.8 days. In fact, one animal remained normoglycemic for more than 100 days. Serum levels of porcine C-peptide remained at levels similar to those for human C-peptide in healthy individuals. CONCLUSIONS Immunosuppression with FTY720 plus CsA inhibited almost all morphological signs of pig-to-rat islet xenograft rejection for up to 24 days after transplantation. Diabetic rats transplanted with APIs and immunosuppressed with FTY720 plus CsA remained normoglycemic for 53.0+/-15.8 days.
Collapse
Affiliation(s)
- A Maeda
- Department of Transplantation Surgery, Karolinska Institute, Huddinge University Hospital, Huddinge, Sweden
| | | | | | | | | | | | | |
Collapse
|
44
|
Thomas FT, Hutchings A, Contreras J, Wu J, Jiang XL, Eckhoff D, Thomas JM. Islet transplantation in the twenty-first century. Immunol Res 2003; 26:289-96. [PMID: 12403366 DOI: 10.1385/ir:26:1-3:289] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Isolated islet transplantation is poised for clinical application to treat insulin-dependent diabetes. Unlike exogenous insulin therapy, islet transplantation has promise for preventing and/or reversing the dismal secondary complications of diabetes. Islet transplants are arguably the most unique type of allografts, and we discuss their properties, limitations, and potential in this overview. The induction of immunologic tolerance to allow islet grafts to endure and prevail, without the hardship of chronic immunosuppressive therapy, is a major goal in this field. In this context, we discuss our successful results in preclinical models of primate allogeneic and xenogeneic islet graft tolerance.
Collapse
Affiliation(s)
- Frank T Thomas
- Department of Surgery, University of Alabama at Birmingham, 35294, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Affiliation(s)
- Philip O'Connell
- National Pancreas Transplant Unit, University of Sydney at Westmead Hospital, Westmead, Australia
| |
Collapse
|
46
|
Hardinger KL, Wang CD, Schnitzler MA, Miller BW, Jendrisak MD, Shenoy S, Lowell JA, Brennan DC. Prospective, pilot, open-label, short-term study of conversion to leflunomide reverses chronic renal allograft dysfunction. Am J Transplant 2002; 2:867-71. [PMID: 12392293 DOI: 10.1034/j.1600-6143.2002.20909.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Leflunomide (LEF) is a synthetic isoxazole derivative with anti-inflammatory and antiviral properties, which has been reported to prevent acute rejection and delay progression of chronic allograft nephropathy (CAN) in animal models. We performed a pilot, crossover trial in 22 renal transplant recipients who were converted from azathioprine (AZA) or mycophenolate mofetil (MMF) to LEF in an effort to slow progression of renal dysfunction [deteriorating renal function (n = 5), cyclosporine (CyA) nephrotoxicity (n = 4) or biopsy-proven CAN (n = 13)]. Baseline maintenance immunosuppression consisted of CyA, AZA or MMF and prednisone. Six-month postconversion patient and graft survival was 100% and 91%, respectively. Mean serum creatinine 6months preconversion was 2.2 +/- 0.6mg/dL, at initiation was 3.0 +/- 1.1 mg/dL, and 6 months postconversion was 2.8 +/- 1.3 mg/dL. The rate of change in serum creatinine was 35 +/- 39%/6 months preconversion and -5 +/- 21%/6 months postconversion to LEF (p = 0.003). Two patients discontinued LEF for diarrhea and myalgia. No readmissions, increase in liver function tests, infections or acute rejection episodes occurred. Mean CyA levels did not change, 146 +/- 72 ng/ mL pre-LEF vs. 132 +/- 51 ng/mL post-LEF, p = NS. Conversion to LEF reversed progression of chronic renal allograft dysfunction with minimal toxicity.
Collapse
Affiliation(s)
- Karen L Hardinger
- Department of Pharmacy, Barnes-Jewish Hospital, Washington University, St Louis, MO, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Levy GA, Ghanekar A, Mendicino M, Phillips MJ, Grant DR. The present status of xenotransplantation. Transplant Proc 2001; 33:3050-2. [PMID: 11750315 DOI: 10.1016/s0041-1345(01)02304-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- G A Levy
- Department of Medicine, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|