51
|
Moore SJ, Gala-Lopez BL, Pepper AR, Pawlick RL, Shapiro AMJ. Bioengineered stem cells as an alternative for islet cell transplantation. World J Transplant 2015; 5:1-10. [PMID: 25815266 PMCID: PMC4371156 DOI: 10.5500/wjt.v5.i1.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 02/18/2014] [Accepted: 10/29/2014] [Indexed: 02/05/2023] Open
Abstract
Type 1 diabetes is an autoimmune and increasingly prevalent condition caused by immunological destruction of beta cells. Insulin remains the mainstay of therapy. Endeavours in islet transplantation have clearly demonstrated that type 1 diabetes is treatable by cellular replacement. Many challenges remain with this approach. The opportunity to use bioengineered embryonic or adult pluripotential stem cells, or islets derived from porcine xenograft sources could address future demands, but are still associated with considerable challenges. This detailed review outlines current progress in clinical islet transplantation, and places this in perspective for the remarkable scientific advances now occurring in stem cell and regenerative medicine approaches in the treatment of future curative treatment of diabetes.
Collapse
|
52
|
Schaschkow A, Mura C, Bietiger W, Peronet C, Langlois A, Bodin F, Dissaux C, Bruant-Rodier C, Pinget M, Jeandidier N, Juszczak MT, Sigrist S, Maillard E. Impact of an autologous oxygenating matrix culture system on rat islet transplantation outcome. Biomaterials 2015; 52:180-8. [PMID: 25818424 DOI: 10.1016/j.biomaterials.2015.02.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/29/2015] [Accepted: 02/01/2015] [Indexed: 10/23/2022]
Abstract
Disruption of the pancreatic islet environment combined with the decrease in oxygen supply that occurs during isolation leads to poor islet survival. The aim of this study was to validate the benefit of using a plasma-based scaffold supplemented with perfluorodecalin to improve islet transplantation outcome. Rat islets were cultured in three conditions: i) control group, ii) plasma based-matrix (P-matrix), and iii) P-matrix supplemented with emulsified perfluorodecalin. After 24 h culture, matrix/cell contacts (Integrinβ1, p-FAK/FAK, p-Akt/Akt), survival (caspase 3, TUNEL, FDA/PI), function, and HIF-1α translocation were assessed. Afterwards, P-matrices were dissolved and the islets were intraportally transplanted. Graft function was monitored for 31 days with glycaemia and C-peptide follow up. Inflammation was assessed by histology (macrophage and granulocyte staining) and thrombin/anti-thrombin complex measurement. Islet survival correlated with an increase in integrin, FAK, and Akt activation in P-matrices and function was maintained. Perfluorodecalin supplementation decreased translocation of HIF-1α in the nucleus and post-transplantation islet structure was better preserved in P-matrices, but a quicker activation of IBMIR resulted in early loss of graft function. "Oxygenating" P-matrices provided a real benefit to islet survival and resistance in vivo. However, intraportal transplantation is not suitable for this kind of culture due to IBMIR; thus, alternative sites must be explored.
Collapse
Affiliation(s)
- A Schaschkow
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, Strasbourg, France
| | - C Mura
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, Strasbourg, France
| | - W Bietiger
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, Strasbourg, France
| | - C Peronet
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, Strasbourg, France
| | - A Langlois
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, Strasbourg, France
| | - F Bodin
- Service de chirurgie Plastique et maxillo faciale, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - C Dissaux
- Service de chirurgie Plastique et maxillo faciale, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - C Bruant-Rodier
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, Strasbourg, France; Service de chirurgie Plastique et maxillo faciale, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - M Pinget
- Structure d'Endocrinologie, Diabète-Nutrition et Addictologie, Pôle NUDE, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - N Jeandidier
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, Strasbourg, France; Structure d'Endocrinologie, Diabète-Nutrition et Addictologie, Pôle NUDE, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - M T Juszczak
- Department of Vascular Surgery, John Radcliffe Hospital, Oxford, United Kingdom
| | - S Sigrist
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, Strasbourg, France
| | - E Maillard
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Bld René Leriche, Strasbourg, France.
| |
Collapse
|
53
|
Shin JY, Jeong JH, Han J, Bhang SH, Jeong GJ, Haque MR, Al-Hilal TA, Noh M, Byun Y, Kim BS. Transplantation of heterospheroids of islet cells and mesenchymal stem cells for effective angiogenesis and antiapoptosis. Tissue Eng Part A 2015; 21:1024-35. [PMID: 25344077 DOI: 10.1089/ten.tea.2014.0022] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Although islet transplantation has been suggested as an alternative therapy for type 1 diabetes, there are efficiency concerns that are attributed to poor engraftment of transplanted islets. Hypoxic condition and delayed vasculogenesis induce necrosis and apoptosis of the transplanted islets. To overcome these limitations in islet transplantation, heterospheroids (HSs), which consist of rat islet cells (ICs) and human bone marrow-derived mesenchymal stem cells (hMSCs), were transplanted to the kidney and liver. The HSs cultured under the hypoxic condition system exhibited a significant increase in antiapoptotic gene expression in ICs. hMSCs in the HSs secreted angiogenic and antiapoptotic proteins. With the HS system, ICs and hMSCs were successfully located in the same area of the liver after transplantation of HSs through the portal vein, whereas the transplantation of islets and the dissociated hMSCs did not result in localization of transplanted ICs and hMSCs in the same area. HS transplantation resulted in an increase in angiogenesis at the transplantation area and a decrease in the apoptosis of transplanted ICs after transplantation into the kidney subcapsule compared with transplantation of islet cell clusters (ICCs). Insulin production levels of ICs were higher in the HS transplantation group compared with the ICC transplantation group. The HS system may be a more efficient transplantation method than the conventional methods for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Jung-Youn Shin
- 1 School of Chemical and Biological Engineering, Seoul National University , Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Kourtzelis I, Ferreira A, Mitroulis I, Ricklin D, Bornstein SR, Waskow C, Lambris JD, Chavakis T. Complement inhibition in a xenogeneic model of interactions between human whole blood and porcine endothelium. Horm Metab Res 2015; 47:36-42. [PMID: 25350518 PMCID: PMC4383746 DOI: 10.1055/s-0034-1390452] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Xenotransplantation (xeno-Tx) is considered as an alternative solution to overcome the shortage of human donor organs. However, the success of xeno-Tx is hindered by immune reactions against xenogeneic cells (e. g. of porcine origin). More specifically, activation of innate immune mechanisms such as complement and triggering of the coagulation cascade occur shortly after xeno-Tx, and adhesion of human leukocytes to porcine endothelium is another early critical step mediating the immune attack. To investigate the therapeutic potential of complement inhibition in the context of xenogeneic interactions, we have employed a whole-blood model in the present study. Incubation of human blood with porcine endothelial cells (PAECs) led to activation of complement and coagulation as well as to increased leukocyte adhesion. The observed responses can be attributed to the pig-to-human xenogeneicity, since the presence of human endothelium induced a minor cellular and plasmatic inflammatory response. Importantly, complement inhibition using a potent complement C3 inhibitor, compstatin analogue Cp40, abrogated the adhesion of leukocytes and, more specifically, the attachment of neutrophils to porcine endothelium. Moreover, Cp40 inhibited the activation of PAECs and leukocytes, since the levels of the adhesion molecules E-selectin, ICAM-1, ICAM-2, and VCAM-1 on PAECs and the surface expression of integrin CD11b on neutrophils were significantly decreased. Along the same line, inhibition of CD11b resulted in decreased leukocyte adhesion. Taken together, our findings provide a better understanding of the mechanisms regulating the acute innate immune complications in the context of xeno-Tx and could pave the way for complement-targeting therapeutic interventions.
Collapse
Affiliation(s)
- I. Kourtzelis
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany
| | - A. Ferreira
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany
| | - I. Mitroulis
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - D. Ricklin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - S. R. Bornstein
- Department of Internal Medicine, Technische Universität Dresden, Dresden, Germany
| | - C. Waskow
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute of Immunology, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - J. D. Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - T. Chavakis
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
- Department of Internal Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
55
|
Kourtzelis I, Magnusson PU, Kotlabova K, Lambris JD, Chavakis T. Regulation of Instant Blood Mediated Inflammatory Reaction (IBMIR) in Pancreatic Islet Xeno-Transplantation: Points for Therapeutic Interventions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 865:171-88. [DOI: 10.1007/978-3-319-18603-0_11] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
56
|
Thromboinflammation in Therapeutic Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 865:3-17. [DOI: 10.1007/978-3-319-18603-0_1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
57
|
Teramura Y, Asif S, Ekdahl KN, Nilsson B. Cell Surface Engineering for Regulation of Immune Reactions in Cell Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 865:189-209. [PMID: 26306451 DOI: 10.1007/978-3-319-18603-0_12] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Transplantation of the pancreatic islets of Langerhans (islets) is a promising cell therapy for treating insulin-dependent type 1 diabetes mellitus. Islet transplantation is a minimally-invasive technique involving relatively simple surgery. However, after intraportal transplantation, the transplanted islets are attacked by the recipient's immune system, because they activate a number of systems, including coagulation, complement response, inflammation, immune rejection, and recurrence of autoimmune disease. We have developed a surface modification and microencapsulation technique that protects cells and islets with biomaterials and bioactive substances, which may be useful in clinical settings. This approach employs amphiphilic polymers, which can interact with lipid bilayer membranes, without increasing cell volume. Molecules attached to these polymers can protect transplanted cells and islets from attack by the host immune system. We expect that this surface modification technique will improve graft survival in clinical islet transplantation.
Collapse
Affiliation(s)
- Yuji Teramura
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan,
| | | | | | | |
Collapse
|
58
|
Alleviation of instant blood-mediated inflammatory reaction in autologous conditions through treatment of human islets with NF-κB inhibitors. Transplantation 2014; 98:578-84. [PMID: 24798306 DOI: 10.1097/tp.0000000000000107] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND The instant blood-mediated inflammatory response (IBMIR) has been shown as a major factor that causes damage to transplanted islets. Withaferin A (WA), an inhibitor of nuclear factor (NF) κB, was shown to suppress the inflammatory response in islets and improve syngeneic islet graft survival in mice. We investigated how treating islets with NF-κB inhibitors affected IBMIR using an in vitro human autologous blood islet model. METHODS Human islets were pretreated with or without NF-κB inhibitors WA or CAY10512 before mixing autologous blood in a miniaturized in vitro tube model. Plasma samples were collected at multiple time points and used for the measurement of C-peptide, proinsulin, thrombin-antithrombin (TAT) complex, and a panel of proinflammatory cytokines. Infiltration of neutrophils into islets was analyzed using immunohistochemistry. RESULTS Rapid release of C-peptide and proinsulin was observed 3 hr after mixing islets and blood in the control group, but not in the NF-κB inhibitor-treated groups, whereas TAT levels were elevated in all three groups with a peak at 6 hr. Significant elevation of proinflammatory cytokines was observed in the control group after 3 hr, but not in the treatment groups. Significant inhibition of neutrophil infiltration was also observed in the WA group compared with the control (P<0.001) and CAY10512 (P<0.001) groups. CONCLUSIONS A miniaturized in vitro tube model can be useful in investigating IBMIR. The presence of NF-κB inhibitor could alleviate IBMIR, thus improving the survival of transplanted islets. Protection of islets in the peritransplant phase may improve long-term graft outcomes.
Collapse
|
59
|
Shin JS, Kim JS, Kim JM, Jang JY, Kim YH, Kim HJ, Park CG. Minimizing immunosuppression in islet xenotransplantation. Immunotherapy 2014; 6:419-30. [PMID: 24815782 DOI: 10.2217/imt.14.14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pancreatic islet transplantation is a promising treatment option for Type 1 diabetes, but organ supply shortage limits its wide adoption. Pig islets are the most promising alternative source and many important measures such as donor animal selection, pig islet production release criteria, preclinical data and zoonosis surveillance prior to human clinical trials have been put forward as a consensus through the efforts of the International Xenotransplantation Association. To bring pig islet transplantation to clinical reality, the development of clinically applicable immunosuppression regimens and methods to minimize immunosuppression to reduce side effects should be established. This review encompasses immune rejection mechanisms in islet xenotransplantation, immunosuppression regimens that have enabled long-term graft survival in pig-to-nonhuman primate experiments and strategies for minimizing immunosuppression in islet xenotransplantation. By thoroughly examining the drugs that are currently available and in development and their individual targets within the immune response, the best strategy for enabling clinical trials of pig islets for Type 1 diabetes will be proposed.
Collapse
Affiliation(s)
- Jun-Seop Shin
- Translational Xenotransplantation Research Center, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 110-799, Korea
| | | | | | | | | | | | | |
Collapse
|
60
|
Suszynski TM, Avgoustiniatos ES, Stein SA, Falde EJ, Hammer BE, Papas KK. Assessment of tissue-engineered islet graft viability by fluorine magnetic resonance spectroscopy. Transplant Proc 2014; 43:3221-5. [PMID: 22099762 DOI: 10.1016/j.transproceed.2011.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
INTRODUCTION Despite significant progress in the last decade, islet transplantation remains an experimental therapy for a limited number of patients with type 1 diabetes. Tissue-engineered approaches may provide promising alternatives to the current clinical protocol and would benefit greatly from concurrent development of graft quality assessment techniques. This study was designed to evaluate whether viability of tissue-engineered islet grafts can be assessed using fluorine magnetic resonance spectroscopy ((19)F-MRS), by the noninvasive measurement of oxygen partial pressure (pO(2)) and the subsequent calculation of islet oxygen consumption rate (OCR). METHODS Scaffolds composed of porcine plasma were seeded with human islets and perfluorodecalin. Each graft was covered with the same volume of culture media in a Petri dish. Four scaffolds were seeded with various numbers (0-8000) of islet equivalents (IE) aliquoted from the same preparation. After randomizing run order, grafts were examined by (19)F-MRS at 37°C using a 5T spectrometer and a single-loop surface coil placed underneath. A standard inversion recovery sequence was used to obtain characteristic (19)F spin-lattice relaxation times (T1), which were converted to steady-state average pO(2) estimates using a previously determined linear calibration (R(2) = 1.000). Each condition was assessed using replicate (19)F-MRS measurements (n = 6-8). RESULTS Grafts exhibited IE dose-dependent increases in T1 and decreases in pO(2) estimates. From the difference between scaffold pO(2) estimates and ambient pO(2), the islet preparation OCR was calculated to be 95 ± 12 (mean ± standard error of the mean) nmol/(min·mg DNA) using theoretical modeling. This value compared well with OCR values measured using established methods for human islet preparations. CONCLUSIONS (19)F-MRS can be used for noninvasive pre- and possibly posttransplant assessment of tissue-engineered islet graft viability by estimating the amount of viable, oxygen-consuming tissue in a scaffold.
Collapse
Affiliation(s)
- T M Suszynski
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | |
Collapse
|
61
|
Nilsson B, Teramura Y, Ekdahl KN. The role and regulation of complement activation as part of the thromboinflammation elicited in cell therapies. Mol Immunol 2014; 61:185-90. [PMID: 24998801 DOI: 10.1016/j.molimm.2014.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 06/09/2014] [Indexed: 02/07/2023]
Abstract
Cell therapies in which the cells come into direct contact with blood and other body fluids are emerging treatment procedures for patients with various diseases, such as diabetes mellitus, liver insufficiency, and graft-versus-host disease. However, despite recent progress, these procedures are associated with tissue loss caused by thromboinflammatory reactions. These deleterious reactions involve the activation of the complement and coagulation cascades and platelet and leukocyte activation, ultimately resulting in clot formation and damage to the implanted cells. In this concept review, we discuss the basic mechanisms underlying the thrombininflammatory process, with special reference to the engagement of complement and emerging strategies for the therapeutic regulation of these reactions that include the use of selective systemic inhibitors and various procedures to coat the surfaces of the cells. The coating procedures may also be applied to other treatment modalities in which similar mechanisms are involved, including whole organ transplantation, treatment with biomaterials in contact with blood, and extracorporeal procedures.
Collapse
Affiliation(s)
- Bo Nilsson
- Dept. of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory, Uppsala University, SE-751 85 Uppsala, Sweden.
| | - Yuji Teramura
- Dept. of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory, Uppsala University, SE-751 85 Uppsala, Sweden; Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo 113-8656, Japan
| | - Kristina N Ekdahl
- Dept. of Immunology, Genetics and Pathology (IGP), Rudbeck Laboratory, Uppsala University, SE-751 85 Uppsala, Sweden; Linnæus Center of Biomaterials Chemistry, Linnæus University, SE-391 82 Kalmar, Sweden
| |
Collapse
|
62
|
Abdelli S, Papas KK, Mueller KR, Murtaugh MP, Hering BJ, Bonny C. Regulation of the JNK3 signaling pathway during islet isolation: JNK3 and c-fos as new markers of islet quality for transplantation. PLoS One 2014; 9:e99796. [PMID: 24983249 PMCID: PMC4077704 DOI: 10.1371/journal.pone.0099796] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/19/2014] [Indexed: 12/22/2022] Open
Abstract
Stress conditions generated throughout pancreatic islet processing initiate the activation of pro-inflammatory pathways and beta-cell destruction. Our goal is to identify relevant and preferably beta-specific markers to assess the activation of beta-cell stress and apoptotic mechanisms, and therefore the general quality of the islet preparation prior to transplantation. Protein expression and activation were analyzed by Western blotting and kinase assays. ATP measurements were performed by a luminescence-based assay. Oxygen consumption rate (OCR) was measured based on standard protocols using fiber optic sensors. Total RNA was used for gene expression analyzes. Our results indicate that pancreas digestion initiates a potent stress response in the islets by activating two stress kinases, c-Jun N-terminal Kinase (JNK) and p38. JNK1 protein levels remained unchanged between different islet preparations and following culture. In contrast, levels of JNK3 increased after islet culture, but varied markedly, with a subset of preparations bearing low JNK3 expression. The observed changes in JNK3 protein content strongly correlated with OCR measurements as determined by the Spearman's rank correlation coefficient rho in the matching islet samples, while inversely correlating with c-fos mRNA expression . In conclusion, pancreas digestion recruits JNK and p38 kinases that are known to participate to beta-cell apoptosis. Concomitantly, the islet isolation alters JNK3 and c-fos expression, both strongly correlating with OCR. Thus, a comparative analysis of JNK3 and c-fos expression before and after culture may provide for novel markers to assess islet quality prior to transplantation. JNK3 has the advantage over all other proposed markers to be islet-specific, and thus to provide for a marker independent of non-beta cell contamination.
Collapse
Affiliation(s)
- Saida Abdelli
- Departement of Medical Genetics, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Klearchos K. Papas
- Department of Surgery, University of Arizona, Institute for Cellular Transplantation, Tucson, Arizona, United States of America
| | - Kate R. Mueller
- Department of Surgery, University of Arizona, Institute for Cellular Transplantation, Tucson, Arizona, United States of America
| | - Mike P. Murtaugh
- Department of Veterinary and Biomedical Sciences, St. Paul, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Bernhard J. Hering
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Christophe Bonny
- Departement of Medical Genetics, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
63
|
Bruni A, Gala-Lopez B, Pepper AR, Abualhassan NS, Shapiro AMJ. Islet cell transplantation for the treatment of type 1 diabetes: recent advances and future challenges. Diabetes Metab Syndr Obes 2014; 7:211-23. [PMID: 25018643 PMCID: PMC4075233 DOI: 10.2147/dmso.s50789] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Islet transplantation is a well-established therapeutic treatment for a subset of patients with complicated type I diabetes mellitus. Prior to the Edmonton Protocol, only 9% of the 267 islet transplant recipients since 1999 were insulin independent for >1 year. In 2000, the Edmonton group reported the achievement of insulin independence in seven consecutive patients, which in a collaborative team effort propagated expansion of clinical islet transplantation centers worldwide in an effort to ameliorate the consequences of this disease. To date, clinical islet transplantation has established improved success with insulin independence rates up to 5 years post-transplant with minimal complications. In spite of marked clinical success, donor availability and selection, engraftment, and side effects of immunosuppression remain as existing obstacles to be addressed to further improve this therapy. Clinical trials to improve engraftment, the availability of insulin-producing cell sources, as well as alternative transplant sites are currently under investigation to expand treatment. With ongoing experimental and clinical studies, islet transplantation continues to be an exciting and attractive therapy to treat type I diabetes mellitus with the prospect of shifting from a treatment for some to a cure for all.
Collapse
Affiliation(s)
- Anthony Bruni
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Boris Gala-Lopez
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Andrew R Pepper
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Nasser S Abualhassan
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - AM James Shapiro
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
64
|
The Role of the Alternative Complement Pathway in Early Graft Loss After Intraportal Porcine Islet Xenotransplantation. Transplantation 2014; 97:999-1008. [DOI: 10.1097/tp.0000000000000069] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
65
|
SoRelle JA, Kanak MA, Itoh T, Horton JM, Naziruddin B, Kane RR. Comparison of surface modification chemistries in mouse, porcine, and human islets. J Biomed Mater Res A 2014; 103:869-77. [DOI: 10.1002/jbm.a.35229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 05/01/2014] [Accepted: 05/13/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Jeffrey A. SoRelle
- Institute of Biomedical Studies; Baylor University; Waco Texas 76798-7224
| | - Mazhar A. Kanak
- Institute of Biomedical Studies; Baylor University; Waco Texas 76798-7224
| | - Takeshi Itoh
- Baylor Research Institute; Baylor University Medical Center; Dallas Texas 75204
| | - Joshua M. Horton
- Institute of Biomedical Studies; Baylor University; Waco Texas 76798-7224
| | - Bashoo Naziruddin
- Institute of Biomedical Studies; Baylor University; Waco Texas 76798-7224
- Annette C. and Harold C. Simmons Transplant Institute; Baylor University Medical Center; Dallas Texas 75246
| | - Robert R. Kane
- Institute of Biomedical Studies; Baylor University; Waco Texas 76798-7224
- Department of Chemistry and Biochemistry; Baylor University; Waco Texas 76798-9348
| |
Collapse
|
66
|
Kanak MA, Takita M, Kunnathodi F, Lawrence MC, Levy MF, Naziruddin B. Inflammatory response in islet transplantation. Int J Endocrinol 2014; 2014:451035. [PMID: 24883060 PMCID: PMC4021753 DOI: 10.1155/2014/451035] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/04/2014] [Accepted: 02/12/2014] [Indexed: 12/23/2022] Open
Abstract
Islet cell transplantation is a promising beta cell replacement therapy for patients with brittle type 1 diabetes as well as refractory chronic pancreatitis. Despite the vast advancements made in this field, challenges still remain in achieving high frequency and long-term successful transplant outcomes. Here we review recent advances in understanding the role of inflammation in islet transplantation and development of strategies to prevent damage to islets from inflammation. The inflammatory response associated with islets has been recognized as the primary cause of early damage to islets and graft loss after transplantation. Details on cell signaling pathways in islets triggered by cytokines and harmful inflammatory events during pancreas procurement, pancreas preservation, islet isolation, and islet infusion are presented. Robust control of pre- and peritransplant islet inflammation could improve posttransplant islet survival and in turn enhance the benefits of islet cell transplantation for patients who are insulin dependent. We discuss several potent anti-inflammatory strategies that show promise for improving islet engraftment. Further understanding of molecular mechanisms involved in the inflammatory response will provide the basis for developing potent therapeutic strategies for enhancing the quality and success of islet transplantation.
Collapse
Affiliation(s)
- Mazhar A. Kanak
- Institute for Biomedical Studies, Baylor University, Waco, TX 76712, USA
| | - Morihito Takita
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX 75204, USA
| | - Faisal Kunnathodi
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX 75204, USA
| | | | - Marlon F. Levy
- Baylor Annette C. and Harold C. Simmons Transplant Institute, 3410 Worth Street, Dallas, TX 75246, USA
| | - Bashoo Naziruddin
- Baylor Annette C. and Harold C. Simmons Transplant Institute, 3410 Worth Street, Dallas, TX 75246, USA
| |
Collapse
|
67
|
Vadori M, Cozzi E. Immunological challenges and therapies in xenotransplantation. Cold Spring Harb Perspect Med 2014; 4:a015578. [PMID: 24616201 DOI: 10.1101/cshperspect.a015578] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Xenotransplantation, or the transplantation of cells, tissues, or organs between different species, was proposed a long time ago as a possible solution to the worldwide shortage of human organs and tissues for transplantation. In this setting, the pig is currently seen as the most likely candidate species. In the last decade, progress in this field has been remarkable and includes a better insight into the immunological mechanisms underlying the rejection process. Several immunological hurdles nonetheless remain, such as the strong antibody-mediated and innate or adaptive cellular immune responses linked to coagulation derangements, precluding indefinite xenograft survival. This article reviews our current understanding of the immunological mechanisms involved in xenograft rejection and the potential strategies that may enable xenotransplantation to become a clinical reality in the not-too-distant future.
Collapse
Affiliation(s)
- Marta Vadori
- CORIT (Consortium for Research in Organ Transplantation), Legnaro, 35020 Padua, Italy
| | | |
Collapse
|
68
|
Zhu HT, Wang WL, Yu L, Wang B. Pig-islet xenotransplantation: recent progress and current perspectives. Front Surg 2014; 1:7. [PMID: 25593932 PMCID: PMC4287008 DOI: 10.3389/fsurg.2014.00007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/07/2014] [Indexed: 01/23/2023] Open
Abstract
Islet xenotransplantation is one prospective treatment to bridge the gap between available human cells and needs of patients with diabetes. Pig represents an ideal candidate for obtaining such available cells. However, potential clinical application of pig islet still faces obstacles including inadequate yield of high-quality functional islets and xenorejection of the transplants. Adequate amounts of available islets can be obtained by selection of a suitable pathogen-free source herd and the development of isolation and purification method. Several studies demonstrated the feasibility of successful preclinical pig-islet xenotransplantation and provided insights and possible mechanisms of xenogeneic immune recognition and rejection. Particularly promising is the achievement of long-term insulin independence in diabetic models by means of distinct islet products and novel immunotherapeutic strategies. Nonetheless, further efforts are needed to obtain much more safety and efficacy data to translate these findings into clinic.
Collapse
Affiliation(s)
- Hai-Tao Zhu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi’an Jiaotong University, Xi’an, China
| | - Wan-Li Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi’an Jiaotong University, Xi’an, China
| | - Liang Yu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi’an Jiaotong University, Xi’an, China
| | - Bo Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
69
|
Meier RPH, Seebach JD, Morel P, Mahou R, Borot S, Giovannoni L, Parnaud G, Montanari E, Bosco D, Wandrey C, Berney T, Bühler LH, Muller YD. Survival of free and encapsulated human and rat islet xenografts transplanted into the mouse bone marrow. PLoS One 2014; 9:e91268. [PMID: 24625569 PMCID: PMC3953382 DOI: 10.1371/journal.pone.0091268] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 02/10/2014] [Indexed: 01/19/2023] Open
Abstract
Bone marrow was recently proposed as an alternative and potentially immune-privileged site for pancreatic islet transplantation. The aim of the present study was to assess the survival and rejection mechanisms of free and encapsulated xenogeneic islets transplanted into the medullary cavity of the femur, or under the kidney capsule of streptozotocin-induced diabetic C57BL/6 mice. The median survival of free rat islets transplanted into the bone marrow or under the kidney capsule was 9 and 14 days, respectively, whereas that of free human islets was shorter, 7 days (bone marrow) and 10 days (kidney capsule). Infiltrating CD8+ T cells and redistributed CD4+ T cells, and macrophages were detected around the transplanted islets in bone sections. Recipient mouse splenocytes proliferated in response to donor rat stimulator cells. One month after transplantation under both kidney capsule or into bone marrow, encapsulated rat islets had induced a similar degree of fibrotic reaction and still contained insulin positive cells. In conclusion, we successfully established a small animal model for xenogeneic islet transplantation into the bone marrow. The rejection of xenogeneic islets was associated with local and systemic T cell responses and macrophage recruitment. Although there was no evidence for immune-privilege, the bone marrow may represent a feasible site for encapsulated xenogeneic islet transplantation.
Collapse
Affiliation(s)
- Raphael P. H. Meier
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Jörg D. Seebach
- Division of Clinical Immunology and Allergology, Department of Internal Medicine, University Hospital and Medical Faculty, Geneva, Switzerland
| | - Philippe Morel
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Redouan Mahou
- Institut d’Ingénierie Biologique et Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Sophie Borot
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Laurianne Giovannoni
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Geraldine Parnaud
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Elisa Montanari
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Christine Wandrey
- Institut d’Ingénierie Biologique et Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Leo H. Bühler
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Yannick D. Muller
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- Division of Clinical Immunology and Allergology, Department of Internal Medicine, University Hospital and Medical Faculty, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
70
|
Dietary zinc supplementation to the donor improves insulin secretion after islet transplantation in chemically induced diabetic rats. Pancreas 2014; 43:236-9. [PMID: 24518501 DOI: 10.1097/mpa.0000000000000093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Zinc (Zn) is related to insulin synthesis, storage, and secretion. This study demonstrates the effects of Zn supplementation in donor rats on the outcomes of islet transplantation. METHODS Donor rats received 3 different regimens of dietary Zn supplementation for 2 weeks before undergoing pancreas donation: a standard diet containing Zn at 50 ppm (control), 1 ppm (low-Zn group) or 1000 ppm (high-Zn group), respectively. Diabetic recipient rats underwent islet transplantation, and the blood glucose levels and insulin secretion were monitored for 7 days after transplantation. RESULTS The serum and pancreatic Zn levels at the time of donation were significantly lower in the low-Zn group (48.8 ± 25.5 µg/dL and 11.3 ± 1.9 µg/g) and higher in the high-Zn group (147.3 ± 17.6 µg/dL and 18.7 ± 2.2 µg/g) when compared with those observed in the controls (118.7 ± 7.9 µg/dL and 14.6 ± 2.0 µg/g) (P < 0.05). The blood glucose levels became re-elevated 2 days after transplantation in rats receiving islet grafts from the controls and the low-Zn groups. In contrast, in the rats that received islets from the high-Zn groups, these were maintained within a reference range (P < 0.01). CONCLUSIONS These data indicate that a Zn-rich diet for donor rats improves the function of islet grafts in chemically induced diabetic rats.
Collapse
|
71
|
Naziruddin B, Iwahashi S, Kanak MA, Takita M, Itoh T, Levy MF. Evidence for instant blood-mediated inflammatory reaction in clinical autologous islet transplantation. Am J Transplant 2014; 14:428-37. [PMID: 24447621 DOI: 10.1111/ajt.12558] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 10/21/2013] [Accepted: 10/22/2013] [Indexed: 01/25/2023]
Abstract
A nonspecific inflammatory and thrombotic reaction termed instant blood-mediated inflammatory reaction (IBMIR) has been reported when allogenic or xenogenic islets come into contact with blood. This reaction is known to cause significant loss of transplanted islets. We hypothesized that IBMIR occurs in patients undergoing total pancreatectomy followed by autologous islet transplantation (TP-AIT) and tested this hypothesis in 24 patients and in an in vitro model. Blood samples drawn during the peritransplant period showed a significant and rapid increase of thrombin-anti-thrombin III complex (TAT) and C-peptide during islet infusion, which persisted for up to 3 h, along with a decreased platelet count. A concomitant increase in levels of inflammatory proteins IL-6, IL-8 and interferon-inducible protein-10 was observed. An in vitro model composed of pure islets plus autologous blood also demonstrated significantly increased levels of TAT (p<0.05), C-peptide (p<0.05), tumor necrosis factor-alpha (p<0.05) and MCP-1 (p<0.05), as well as strong tissue factor expression in islets. Islet viability decreased significantly but was rescued by the presence of low-molecular-weight dextran sulfate. In conclusion, AIT-induced elevation of TAT and destruction of islets suggests that IBMIR might occur during AIT. Modulating this process may help improve islet engraftment and the insulin independence rate in TP-AIT patients.
Collapse
Affiliation(s)
- B Naziruddin
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center at Dallas, Dallas, TX
| | | | | | | | | | | |
Collapse
|
72
|
Chhabra P, Brayman KL. Overcoming barriers in clinical islet transplantation: current limitations and future prospects. Curr Probl Surg 2014; 51:49-86. [PMID: 24411187 DOI: 10.1067/j.cpsurg.2013.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
73
|
Fine D, Grattoni A, Goodall R, Bansal SS, Chiappini C, Hosali S, van de Ven AL, Srinivasan S, Liu X, Godin B, Brousseau L, Yazdi IK, Fernandez-Moure J, Tasciotti E, Wu HJ, Hu Y, Klemm S, Ferrari M. Silicon micro- and nanofabrication for medicine. Adv Healthc Mater 2013; 2:632-66. [PMID: 23584841 PMCID: PMC3777663 DOI: 10.1002/adhm.201200214] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 08/31/2012] [Indexed: 12/13/2022]
Abstract
This manuscript constitutes a review of several innovative biomedical technologies fabricated using the precision and accuracy of silicon micro- and nanofabrication. The technologies to be reviewed are subcutaneous nanochannel drug delivery implants for the continuous tunable zero-order release of therapeutics, multi-stage logic embedded vectors for the targeted systemic distribution of both therapeutic and imaging contrast agents, silicon and porous silicon nanowires for investigating cellular interactions and processes as well as for molecular and drug delivery applications, porous silicon (pSi) as inclusions into biocomposites for tissue engineering, especially as it applies to bone repair and regrowth, and porous silica chips for proteomic profiling. In the case of the biocomposites, the specifically designed pSi inclusions not only add to the structural robustness, but can also promote tissue and bone regrowth, fight infection, and reduce pain by releasing stimulating factors and other therapeutic agents stored within their porous network. The common material thread throughout all of these constructs, silicon and its associated dielectrics (silicon dioxide, silicon nitride, etc.), can be precisely and accurately machined using the same scalable micro- and nanofabrication protocols that are ubiquitous within the semiconductor industry. These techniques lend themselves to the high throughput production of exquisitely defined and monodispersed nanoscale features that should eliminate architectural randomness as a source of experimental variation thereby potentially leading to more rapid clinical translation.
Collapse
Affiliation(s)
- Daniel Fine
- Department of Nanomedicine, The Methodist Hospital Research Institute, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Nishimura R, Nishioka S, Fujisawa I, Shiku H, Shimada M, Sekiguchi S, Fujimori K, Ushiyama A, Matsue T, Ohuchi N, Satomi S, Goto M. Tacrolimus inhibits the revascularization of isolated pancreatic islets. PLoS One 2013; 8:e56799. [PMID: 23613708 PMCID: PMC3629082 DOI: 10.1371/journal.pone.0056799] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 01/16/2013] [Indexed: 01/10/2023] Open
Abstract
AIMS Immunosuppressive drugs could be crucial factors for a poor outcome after islet allotransplantation. Unlike rapamycin, the effects of tacrolimus, the current standard immunosuppressant used in islet transplantation, on graft revascularization remain unclear. We examined the effects of tacrolimus on islet revascularization using a highly sensitive imaging system, and analyzed the gene expression in transplanted islets by introducing laser microdissection techniques. METHODS Islets isolated from C57BL/6-Tg (CAG-EGFP) mice were transplanted into the nonmetallic dorsal skinfold chamber on the recipients. Balb/c athymic mice were used as recipients and were divided into two groups: including a control group (n = 9) and tacrolimus-treated group (n = 7). The changes in the newly-formed vessels surrounding the islet grafts were imaged and semi-quantified using multi-photon laser-scanning microscopy and a Volocity system. Gene expression in transplanted islets was analyzed by the BioMark dynamic system. RESULTS The revascularization process was completed within 14 days after pancreatic islet transplantation at subcutaneous sites. The newly-formed vascular volume surrounding the transplanted islets in the tacrolimus-treated group was significantly less than that in the control group (p<0.05). Although the expression of Vegfa (p<0.05) and Ccnd1 (p<0.05) was significantly upregulated in the tacrolimus-treated group compared with that of the control group, no differences were observed between the groups in terms of other types of gene expression. CONCLUSIONS The present study demonstrates that tacrolimus inhibits the revascularization of isolated pancreatic islets without affecting the characteristics of the transplanted grafts. Further refinements of this immunosuppressive regimen, especially regarding the revascularization of islet grafts, could improve the outcome of islet allotransplantation.
Collapse
Affiliation(s)
- Ryuichi Nishimura
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
| | - Sho Nishioka
- Graduate School of Environmental Studies, Tohoku University, Sendai, Japan
| | - Ikuma Fujisawa
- Graduate School of Environmental Studies, Tohoku University, Sendai, Japan
| | - Hitoshi Shiku
- Graduate School of Environmental Studies, Tohoku University, Sendai, Japan
| | - Miki Shimada
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| | - Satoshi Sekiguchi
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
| | - Keisei Fujimori
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
| | - Akira Ushiyama
- Department of Environmental Health, National Institute of Public Health, Wako, Japan
| | - Tomokazu Matsue
- WorldPremier InternationalResearch Center Initiative Advanced Institute for Materials Research, Tohoku University, Sendai, Japan
| | - Noriaki Ohuchi
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
| | - Susumu Satomi
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
| | - Masafumi Goto
- Division of Advanced Surgical Science and Technology, Tohoku University, Sendai, Japan
- New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
- * E-mail:
| |
Collapse
|
75
|
Inhibition of instant blood-mediated inflammatory responses by co-immobilization of sCR1 and heparin on islets. Biomaterials 2013; 34:5019-24. [PMID: 23578554 DOI: 10.1016/j.biomaterials.2013.03.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 03/14/2013] [Indexed: 02/03/2023]
Abstract
Intraportal transplantation of islets of Langerhans is followed by marked islet loss, mainly caused by instant blood-mediated inflammatory responses (IBMIR). We previously developed a method of co-immobilizing sCR1 and heparin on islets. Here we examined whether this process could reduce islet loss following intraportal islet transplantation in a syngeneic mouse model. sCR1-heparin islets or unmodified islet controls were transplanted into the livers of streptozotocin-induced diabetic mice. Transplantation of 100 and 125 sCR1-heparin islets normalized blood glucose levels in 8 of 9 (88.9%) and 9 of 9 diabetic mice (100%), respectively, whereas transplantation of 100 and 125 non-treated islets induced normoglycemia in 0 of 9 and 2 of 9 diabetic mice, respectively. Fibrin staining and plasma insulin measurements indicated that, compared to non-treated islets, sCR1-heparin islet transplantation was associated with fewer blood clots around islets, and significantly less insulin leakage from damaged islets at 1 h post-transplantation. Long-term follow-up of the sCR1-heparin islet group showed islet cells in the livers and insulin expression. In conclusion, co-immobilization of sCR1 and heparin on islets could effectively reduce islet damage by IBMIR, and might be useful to enable transplantation with only one donor and one recipient.
Collapse
|
76
|
Im BH, Jeong JH, Haque MR, Lee DY, Ahn CH, Kim JE, Byun Y. The effects of 8-arm-PEG-catechol/heparin shielding system and immunosuppressive drug, FK506 on the survival of intraportally allotransplanted islets. Biomaterials 2013; 34:2098-106. [DOI: 10.1016/j.biomaterials.2012.11.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 11/20/2012] [Indexed: 01/26/2023]
|
77
|
Jung HS, Kim MJ, Hong SH, Lee YJ, Kang S, Lee H, Chung SS, Park JS, Park KS. The potential of endothelial colony-forming cells to improve early graft loss after intraportal islet transplantation. Cell Transplant 2013; 23:273-83. [PMID: 23294520 DOI: 10.3727/096368912x661364] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Early graft loss in islet transplantation means that a large amount of donor islets is required. Endothelial cells and endothelial colony-forming cells (ECFCs) have been reported to improve instant blood-mediated inflammatory reaction (IBMIR) in vitro. In this study, we examined if ECFC-coated porcine islets would prevent early graft loss in vivo. Human ECFCs were prepared from cord blood and cocultured with islets to make composite grafts. Diabetic nude mice underwent intraportal transplantation. Blood glucose levels were monitored, and morphological examination of the grafts along with analysis of the components of IBMIR and inflammatory reaction were performed with the liver tissues. The ECFC-coated islets significantly decreased blood glucose levels immediately after transplantation compared to the uncoated islets. Composite ECFC islet grafts were observed in the liver sections, associated with a more insulin(+) area compared to that of the uncoated group within 48 h after transplantation. Deposition of CD41a, C5b-9, and CD11b(+) cells was also decreased in the ECFC-coated group. Expression of porcine HMGB1 and mouse TNF-α was increased in the transplantated groups compared to the sham operation group, with a trend of a decreasing trend across the uncoated group, the ECFC-coated group, and the sham group. We demonstrated that the composite ECFC porcine islets transplanted into the portal vein of nude mice improved early graft loss and IBMIR in vivo.
Collapse
Affiliation(s)
- Hye Seung Jung
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
van der Windt DJ, Bottino R, Kumar G, Wijkstrom M, Hara H, Ezzelarab M, Ekser B, Phelps C, Murase N, Casu A, Ayares D, Lakkis FG, Trucco M, Cooper DK. Clinical islet xenotransplantation: how close are we? Diabetes 2012; 61:3046-55. [PMID: 23172951 PMCID: PMC3501885 DOI: 10.2337/db12-0033] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 06/06/2012] [Indexed: 01/27/2023]
Affiliation(s)
- Dirk J. van der Windt
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Rita Bottino
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Division of Immunogenetics, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Goutham Kumar
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Martin Wijkstrom
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Hidetaka Hara
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Mohamed Ezzelarab
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Burcin Ekser
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Surgery, Transplantation and Advanced Technologies, Vascular Surgery and Organ Transplant Unit, University Hospital of Catania, Catania, Italy
| | | | - Noriko Murase
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Anna Casu
- Diabetes Unit, Department of Medicine, Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (ISMETT), Palermo, Italy
| | | | - Fadi G. Lakkis
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Massimo Trucco
- Division of Immunogenetics, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - David K.C. Cooper
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
79
|
van der Windt DJ, Marigliano M, He J, Votyakova TV, Echeverri GJ, Ekser B, Ayares D, Lakkis FG, Cooper DKC, Trucco M, Bottino R. Early islet damage after direct exposure of pig islets to blood: has humoral immunity been underestimated? Cell Transplant 2012; 21:1791-802. [PMID: 22776064 DOI: 10.3727/096368912x653011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Currently, islet transplantation as a cell therapeutic option for type 1 diabetes occurs via islet injection into the portal vein. Direct contact between islets and blood is a pathophysiological "provocation" that results in the instant blood-mediated inflammatory reaction (IBMIR) and is associated with early islet loss. However, the nature of the various insults on the islets in the blood stream remains mostly unknown. To gain insight into the mechanisms, we utilized a simplified in vitro model in which islets were exposed to blood in different clinically relevant but increasingly challenging, autologous, allogeneic, and xenogeneic combinations. Irrespective of the blood type and species compatibility, islets triggered blood clotting. Islet damage was worse as islet, and blood compatibility diminished, with substantial islet injury after exposure of porcine islets to human blood. Islet damage involved membrane leakage, antibody deposition, complement activation, positive staining for the membrane attack complex, and mitochondrial dysfunction. Islet damage occurred even after exposure to plasma only, and specific complement inactivation and neutralization of IgM substantially prevented islet damage, indicating the importance of humoral immunity. Efficacious measures are needed to reduce this injury, especially in view of a potential clinical use of porcine islets to treat diabetes.
Collapse
Affiliation(s)
- Dirk J van der Windt
- Division of Immunogenetics, Department of Pediatrics, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Jackson AM, Kanak MA, Grishman EK, Chaussabel D, Levy MF, Naziruddin B. Gene expression changes in human islets exposed to type 1 diabetic serum. Islets 2012; 4:312-9. [PMID: 22885994 PMCID: PMC3496656 DOI: 10.4161/isl.21510] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A major obstacle to the success of islet cell transplantation as a standard treatment for labile type 1 diabetes mellitus is the immediate loss of up to 70% of the transplanted islet mass. Activation of the complement cascade and coagulation factors has been implicated in initiating the destruction of the islet graft. In this study, we analyzed the gene expression changes in islet cells following exposure to type 1 diabetes mellitus serum (T1DM). Isolated human pancreatic islet cells were cultured for 2 d to stabilize islet cell gene expression. Cultured islets were divided into three groups for treatment as follows: group 1 was treated with autologous donor serum, while groups two and three were treated with sera from ABO-matched allogeneic donors or autoantibody positive type 1 diabetic patient, respectively. Complement was detected using anti-C3 FITC and CH50 assay. Islet gene expression was analyzed using Illumina micro-array technology. Results were confirmed using real-time PCR. Immunofluorescent imaging demonstrated complement deposition only in the T1DM condition. Gene array and class prediction analysis generated a list of 50 genes that were able to predict the effect of T1DM serum on islets. Quantitative PCR corroborated microarray results. Both techniques demonstrated upregulation of MMP9 (243%), IL-1β (255%), IL-11 (220%), IL-12A (132%), RAD (343%) and a concomitant downregulation of IL-1RN (64%) in islets treated with T1DM serum. Islets treated with T1DM serum overexpressed genes associated with angiogenesis while decreasing transcription of genes that protect islets from inflammatory cytokines and reactive oxygen species.
Collapse
Affiliation(s)
| | - Mazhar A. Kanak
- Institute of Biomedical Studies; Baylor University; Waco, TX USA
| | | | | | - Marlon F. Levy
- Baylor Simmons Transplant Institute; Baylor University Medical Center; Dallas, TX USA
| | - Bashoo Naziruddin
- Institute of Biomedical Studies; Baylor University; Waco, TX USA
- Baylor Simmons Transplant Institute; Baylor University Medical Center; Dallas, TX USA
- Correspondence to: Bashoo Naziruddin,
| |
Collapse
|
81
|
Control of instant blood-mediated inflammatory reaction to improve islets of Langerhans engraftment. Curr Opin Organ Transplant 2012; 16:620-6. [PMID: 21971510 DOI: 10.1097/mot.0b013e32834c2393] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Transplantation of islets of Langerhans is an emerging treatment procedure for patients with severe type 1 diabetes, but despite recent progress the procedure is associated with massive tissue loss caused by an inflammatory reaction termed instant blood-mediated inflammatory reaction (IBMIR). This reaction involves activation of the complement and coagulation cascades, ultimately resulting in clot formation and infiltration of leukocytes into the islets, which leads to disruption of islet integrity and islet destruction. RECENT FINDINGS In this review we discuss basic mechanisms underlying the IBMIR and emerging strategies for therapeutic regulation of the IBMIR. These include the use of selective inhibitors of the coagulation and complement systems, different procedures to coat the surface of the islets as well as the development of composite islet-endothelial cell grafts. SUMMARY The IBMIR is a major cause of tissue loss in clinical islet transplantation, and most likely in other cell therapies in which cells are exposed to blood. Thus, it is an obvious target for therapeutic intervention. Due to its complexity, it is necessary to use different strategies to control the IBMIR.
Collapse
|
82
|
Ekser B, Ezzelarab M, Hara H, van der Windt DJ, Wijkstrom M, Bottino R, Trucco M, Cooper DKC. Clinical xenotransplantation: the next medical revolution? Lancet 2012; 379:672-83. [PMID: 22019026 DOI: 10.1016/s0140-6736(11)61091-x] [Citation(s) in RCA: 250] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The shortage of organs and cells from deceased individuals continues to restrict allotransplantation. Pigs could provide an alternative source of tissue and cells but the immunological challenges and other barriers associated with xenotransplantation need to be overcome. Transplantation of organs from genetically modified pigs into non-human primates is now not substantially limited by hyperacute, acute antibody-mediated, or cellular rejection, but other issues have become more prominent, such as development of thrombotic microangiopathy in the graft or systemic consumptive coagulopathy in the recipient. To address these problems, pigs that express one or more human thromboregulatory or anti-inflammatory genes are being developed. The results of preclinical transplantation of pig cells--eg, islets, neuronal cells, hepatocytes, or corneas--are much more encouraging than they are for organ transplantation, with survival times greater than 1 year in all cases. Risk of transfer of an infectious microorganism to the recipient is small.
Collapse
Affiliation(s)
- Burcin Ekser
- Thomas E Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
83
|
Sakata N, Sumi S, Yoshimatsu G, Goto M, Egawa S, Unno M. Encapsulated islets transplantation: Past, present and future. World J Gastrointest Pathophysiol 2012; 3:19-26. [PMID: 22368783 PMCID: PMC3284522 DOI: 10.4291/wjgp.v3.i1.19] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 12/04/2011] [Accepted: 02/08/2012] [Indexed: 02/06/2023] Open
Abstract
Islet transplantation could become an ideal treatment for severe diabetes to prevent hypoglycemia shock and irreversible diabetic complications, once some of the major and unresolved obstacles are overcome, including limited donor supplies and side effects caused by permanent immunosuppressant use. Approximately 30 years ago, some groups succeeded in improving the blood glucose of diabetic animals by transplanting encapsulated islets with semi-permeable membranes consisting of polymer. A semi-permeable membrane protects both the inner islets from mechanical stress and the recipient's immune system (both cellular and humoral immunities), while allowing bidirectional diffusion of nutrients, oxygen, glucose, hormones and wastes, i.e., immune-isolation. This device, which enables immune-isolation, is called encapsulated islets or bio-artificial pancreas. Encapsulation with a semi-permeable membrane can provide some advantages: (1) this device protects transplanted cells from the recipient's immunity even if the xenogeneic islets (from large animals such as pig) or insulin-producing cells are derived from cells that have the potential for differentiation (some kinds of stem cells). In other words, the encapsulation technique can resolve the problem of limited donor supplies; and (2) encapsulation can reduce or prevent chronic administration of immunosuppressants and, therefore, important side effects otherwise induced by immunosuppressants. And now, many novel encapsulated islet systems have been developed and are being prepared for testing in a clinical setting.
Collapse
|
84
|
Abstract
Xenotransplantation, the transplantation of cells, tissues, or organs between different species, has the potential to overcome the current shortage of human organs and tissues for transplantation. In the last decade, the progress made in the field is remarkable, suggesting that clinical xenotransplantation procedures, particularly those involving cells, may become a reality in the not-too-distant future. However, several hurdles remain, mainly immunological barriers, physiological discrepancies, and safety issues, making xenotransplantion a complex and multidisciplinary discipline.
Collapse
|
85
|
Thompson P, Badell IR, Lowe M, Cano J, Song M, Leopardi F, Avila J, Ruhil R, Strobert E, Korbutt G, Rayat G, Rajotte R, Iwakoshi N, Larsen CP, Kirk AD. Islet xenotransplantation using gal-deficient neonatal donors improves engraftment and function. Am J Transplant 2011; 11:2593-602. [PMID: 21883917 PMCID: PMC3226931 DOI: 10.1111/j.1600-6143.2011.03720.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Significant deficiencies in understanding of xenospecific immunity have impeded the success of preclinical trials in xenoislet transplantation. Although galactose-α1,3-galactose, the gal epitope, has emerged as the principal target of rejection in pig-to-primate models of solid organ transplant, the importance of gal-specific immunity in islet xenotransplant models has yet to be clearly demonstrated. Here, we directly compare the immunogenicity, survival and function of neonatal porcine islets (NPIs) from gal-expressing wild-type (WT) or gal-deficient galactosyl transferase knockout (GTKO) donors. Paired diabetic rhesus macaques were transplanted with either WT (n = 5) or GTKO (n = 5) NPIs. Recipient blood glucose, transaminase and serum xenoantibody levels were used to monitor response to transplant. Four of five GTKO versus one of five WT recipients achieved insulin-independent normoglycemia; transplantation of WT islets resulted in significantly greater transaminitis. The WT NPIs were more susceptible to antibody and complement binding and destruction in vitro. Our results confirm that gal is an important variable in xenoislet transplantation. The GTKO NPI recipients have improved rates of normoglycemia, likely due to decreased susceptibility of xenografts to innate immunity mediated by complement and preformed xenoantibody. Therefore, the use of GTKO donors is an important step toward improved consistency and interpretability of results in future xenoislet studies.
Collapse
Affiliation(s)
- P Thompson
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - IR Badell
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - M Lowe
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - J Cano
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - M Song
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - F Leopardi
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - J Avila
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - R Ruhil
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - E Strobert
- Yerkes National Primate Research Center, Atlanta, GA, USA 30322
| | - G Korbutt
- Surgical-Medical Research Institute, University of Alberta, Edmonton, AB, T6G 2N8, Canada
| | - G Rayat
- Surgical-Medical Research Institute, University of Alberta, Edmonton, AB, T6G 2N8, Canada
| | - R Rajotte
- Surgical-Medical Research Institute, University of Alberta, Edmonton, AB, T6G 2N8, Canada
| | - N Iwakoshi
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - CP Larsen
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| | - AD Kirk
- Emory Transplant Center, Emory University, Atlanta, GA, USA 30322
| |
Collapse
|
86
|
Chhabra P, Brayman KL. Current status of immunomodulatory and cellular therapies in preclinical and clinical islet transplantation. J Transplant 2011; 2011:637692. [PMID: 22046502 PMCID: PMC3199196 DOI: 10.1155/2011/637692] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 07/11/2011] [Indexed: 02/08/2023] Open
Abstract
Clinical islet transplantation is a β-cell replacement strategy that represents a possible definitive intervention for patients with type 1 diabetes, offering substantial benefits in terms of lowering daily insulin requirements and reducing incidences of debilitating hypoglycemic episodes and unawareness. Despite impressive advances in this field, a limiting supply of islets, inadequate means for preventing islet rejection, and the deleterious diabetogenic and nephrotoxic side effects associated with chronic immunosuppressive therapy preclude its wide-spread applicability. Islet transplantation however allows a window of opportunity for attempting various therapeutic manipulations of islets prior to transplantation aimed at achieving superior transplant outcomes. In this paper, we will focus on the current status of various immunosuppressive and cellular therapies that promote graft function and survival in preclinical and clinical islet transplantation with special emphasis on the tolerance-inducing capacity of regulatory T cells as well as the β-cells regenerative capacity of stem cells.
Collapse
Affiliation(s)
- Preeti Chhabra
- Department of Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Kenneth L. Brayman
- Department of Surgery, University of Virginia, Charlottesville, VA 22908, USA
- Division of Transplantation, Department of Surgery, University of Virginia, Charlottesville, VA 22908, USA
- The Center for Cellular Transplantation and Therapeutics, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
87
|
Abstract
The therapy of type 1 diabetes is an open challenging problem. The restoration of normoglycemia and insulin independence in immunosuppressed type 1 diabetic recipients of islet allotransplantation has shown the potential of a cell-based diabetes therapy. Even if successful, this approach poses a problem of scarce tissue supply. Xenotransplantation can be the answer to this limited donor availability and, among possible candidate tissues for xenotransplantation, porcine islets are the closest to a future clinical application. Xenotransplantation, with pigs as donors, offers the possibility of using healthy, living, and genetically modified islets from pathogen-free animals available in unlimited number of islets. Several studies in the pig-to-nonhuman primate model demonstrated the feasibility of successful preclinical islet xenotransplantation and have provided insights into the critical events and possible mechanisms of immune recognition and rejection of xenogeneic islet grafts. Particularly promising results in the achievement of prolonged insulin independence were obtained with newly developed, genetically modified pigs islets able to produce immunoregulatory products, using different implantation sites, and new immunotherapeutic strategies. Nonetheless, further efforts are needed to generate additional safety and efficacy data in nonhuman primate models to safely translate these findings into the clinic.
Collapse
Affiliation(s)
- Marco Marigliano
- Division of Immunogenetics, Department of Pediatrics, Rangos Research Center, Children’s Hospital of Pittsburgh, 6th floor, Room 6126, 4401 Penn Avenue, Pittsburgh, PA 15224 USA
- Regional Center for Diabetes in Children and Adolescents, Salesi’s Hospital, Via Corridoni 11, 60123 Ancona, Italy
| | - Suzanne Bertera
- Division of Immunogenetics, Department of Pediatrics, Rangos Research Center, Children’s Hospital of Pittsburgh, 6th floor, Room 6126, 4401 Penn Avenue, Pittsburgh, PA 15224 USA
| | - Maria Grupillo
- Division of Immunogenetics, Department of Pediatrics, Rangos Research Center, Children’s Hospital of Pittsburgh, 6th floor, Room 6126, 4401 Penn Avenue, Pittsburgh, PA 15224 USA
- RiMeD Foundation, Palermo, Italy
| | - Massimo Trucco
- Division of Immunogenetics, Department of Pediatrics, Rangos Research Center, Children’s Hospital of Pittsburgh, 6th floor, Room 6126, 4401 Penn Avenue, Pittsburgh, PA 15224 USA
| | - Rita Bottino
- Division of Immunogenetics, Department of Pediatrics, Rangos Research Center, Children’s Hospital of Pittsburgh, 6th floor, Room 6126, 4401 Penn Avenue, Pittsburgh, PA 15224 USA
| |
Collapse
|
88
|
Kahraman S, Dirice E, Hapil FZ, Ertosun MG, Ozturk S, Griffith TS, Sanlioglu S, Sanlioglu AD. Tracing of islet graft survival by way of in vivo fluorescence imaging. Diabetes Metab Res Rev 2011; 27:575-83. [PMID: 21584921 DOI: 10.1002/dmrr.1216] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND To increase the success rate in xenogeneic islet transplantation, proper assessment of graft mass is required following transplantation. For this reason, we aimed to develop a suitable fluorescence imaging system to monitor islet xenograft survival in diabetic mice. METHODS Adenovirus vector encoding enhanced green fluorescent protein-transduced rat pancreatic islets were transplanted under the renal capsule of streptozotocin-induced diabetic mice and the fluorescence signal was quantified over time using a cooled charge-coupled device. Non-fasting blood glucose levels were recorded during the same period. Insulin release from transduced and control islets was detected via enzyme-linked immunosorbent assay. RESULTS Adenovirus vector encoding enhanced green fluorescent protein infection did not alter the function or survival of pancreatic islets post transduction. A direct correlation was found between the number of islets (250-750) transplanted under the kidney capsule and the blood glucose recovery. CONCLUSIONS Fluorescence imaging appears to be a useful tool for quantitative assessment of islet cell viability post transplantation and could permit earlier detection of graft rejection.
Collapse
Affiliation(s)
- Sevim Kahraman
- Department of Medical Biology and Genetics, Human Gene and Cell Therapy Center of Akdeniz University Hospitals and Clinics, Antalya 07058, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Subcapsular fetal pig pancreas fragment transplantation provides normal blood glucose control in a preclinical model of diabetes. Transplantation 2011; 91:515-21. [PMID: 21183867 DOI: 10.1097/tp.0b013e3182079474] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Identifying a limitless source of β-cells that survive transplantation into a neovascularised site and provide normal blood glucose control remains an important goal in the development of pancreatic islet xenotransplantation. It was our hypothesis that fetal porcine pancreas fragments could achieve these objectives, and this was tested in a large preclinical animal model. RESEARCH DESIGN AND METHODS Inbred "Westran Pig" fetal porcine pancreas fragments were transplanted beneath the splenic capsule into syngeneic Westran Pig recipients without immunosuppression, and 3 months later, a total native pancreatectomy was performed to demonstrate function. RESULTS Histologic analysis showed appropriate development of islet-like structures up to and beyond 120 days after transplantation. After native pancreatectomy, recipients survived more than 100 days without exogenous insulin and with normal glucose homeostasis as assessed by normal glucose tolerance tests, K values, and normal glucagon secretion. CONCLUSIONS This study confirms that fetal pig islet tissue has the potential to mature and function normally in a neovascularised site, hence, avoiding the innate immune destruction that occurs when islet tissue is exposed directly to the circulation.
Collapse
|
90
|
Attenuation of cross-talk between the complement and coagulation cascades by C5a blockade improves early outcomes after intraportal islet transplantation. Transplantation 2011; 90:1358-65. [PMID: 21197712 DOI: 10.1097/tp.0b013e3181ffb9f5] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Complement 5a factor (C5a) elicits a broad range of proinflammatory effects, including chemotaxis of inflammatory cells and cytokine release. C5a is also linked to the coagulant activity in autoimmune diseases. Therefore, C5a most likely plays a crucial role in the instant blood-mediated inflammatory reaction. METHODS Intraportal transplantation of 2.5 islet equivalents/g of syngeneic rat islet grafts was performed in two groups of streptozotocin-induced diabetic rats: controls and C5a inhibitory peptide (C5aIP)-treated group. RESULTS The thrombin-antithrombin complex was significantly suppressed in the C5aIP group (P=0.003), and both the curative rate and the glucose tolerance were significantly improved in the C5aIP group (P<0.05 and P<0.005, respectively). Expression of tissue factor on granulocytes in recipient livers was up-regulated 1 h after islet infusion (P<0.0001), which was significantly suppressed by C5aIP (P<0.005). However, C5aIP was unable to regulate tissue factor expression on isolated islets. Furthermore, no differences were detected between the groups, regarding infiltration of CD11b-positive cells and deposition of C5b-9 on the islet grafts. CONCLUSIONS These data suggest that C5aIP attenuates cross-talk between the complement and coagulation cascades through suppressing up-regulation of tissue factor expression on leukocytes in recipient livers but not on islet grafts, a process leading to improvement in islet engraftment. Therefore, C5aIP in combination with conventional anticoagulants could be a strong candidate strategy to control the instant blood-mediated inflammatory reaction induced in clinical islet transplantation.
Collapse
|
91
|
Current world literature. Curr Opin Organ Transplant 2010; 15:254-61. [PMID: 20351662 DOI: 10.1097/mot.0b013e328337a8db] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
92
|
Can cells and biomaterials in therapeutic medicine be shielded from innate immune recognition? Trends Immunol 2010; 31:32-8. [PMID: 19836998 DOI: 10.1016/j.it.2009.09.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 09/11/2009] [Accepted: 09/11/2009] [Indexed: 10/20/2022]
Abstract
Biomaterials (e.g. polymers, metals, or ceramics), cell and cell cluster (e.g. pancreatic islets) transplantation are beginning to offer novel treatment modalities for some otherwise intractable diseases. The innate immune system is involved in incompatibility reactions that occur when biomaterials or cells are introduced into the blood circulation. In particular, the complement, coagulation and contact systems are involved in the recognition of biomaterials and cells, eliciting activation of platelets and leukocytes. Such treatments are associated with anaphylactoid and thrombotic reactions, inflammation, and rejection of biomaterials and cells, leading to treatment failures and adverse reactions. We discuss here the new technologies that are being developed to shield the biomaterial and cell surfaces from recognition by the innate immune system.
Collapse
|
93
|
Ekser B, Cooper DKC. Overcoming the barriers to xenotransplantation: prospects for the future. Expert Rev Clin Immunol 2010; 6:219-30. [PMID: 20402385 PMCID: PMC2857338 DOI: 10.1586/eci.09.81] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cross-species transplantation (xenotransplantation) has immense potential to solve the critical need for organs, tissues and cells for clinical transplantation. The increasing availability of genetically engineered pigs is enabling progress to be made in pig-to-nonhuman primate experimental models. Potent pharmacologic immunosuppressive regimens have largely prevented T-cell rejection and a T-cell-dependent elicited antibody response. However, coagulation dysfunction between the pig and primate is proving to be a major problem, and this can result in life-threatening consumptive coagulopathy. This complication is unlikely to be overcome until pigs expressing a human 'antithrombotic' or 'anticoagulant' gene, such as thrombomodulin, tissue factor pathway inhibitor or CD39, become available. Progress in islet xenotransplantation has been more encouraging, and diabetes has been controlled in nonhuman primates for periods in excess of 6 months, although this has usually been achieved using immunosuppressive protocols that might not be clinically applicable. Further advances are required to overcome the remaining barriers.
Collapse
Affiliation(s)
- Burcin Ekser
- Thomas E Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA, and Department of Surgery and Organ Transplantation, University of Padua, Padua, Italy
| | - David KC Cooper
- Thomas E Starzl Transplantation Institute, University of Pittsburgh Medical Center, Starzl Biomedical Science Tower, W1543, 200 Lothrop Street, Pittsburgh, PA 15261, USA, Tel.: +1 412 383 6961, Fax: +1 412 624 1172,
| |
Collapse
|
94
|
Miyagawa S, Yamamoto A, Matsunami K, Wang D, Takama Y, Ueno T, Okabe M, Nagashima H, Fukuzawa M. Complement regulation in the GalT KO era. Xenotransplantation 2010; 17:11-25. [DOI: 10.1111/j.1399-3089.2010.00569.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
95
|
van der Windt DJ, Bottino R, Casu A, Campanile N, Smetanka C, He J, Murase N, Hara H, Ball S, Loveland BE, Ayares D, Lakkis FG, Cooper DKC, Trucco M. Long-term controlled normoglycemia in diabetic non-human primates after transplantation with hCD46 transgenic porcine islets. Am J Transplant 2009; 9:2716-26. [PMID: 19845582 DOI: 10.1111/j.1600-6143.2009.02850.x] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Xenotransplantation of porcine islets into diabetic non-human primates is characterized by (i) an initial massive graft loss possibly due to the instant blood-mediated inflammatory reaction and (ii) the requirement of intensive, clinically unfriendly immunosuppressive therapy. We investigated whether the transgenic expression of a human complement-regulatory protein (hCD46) on porcine islets would improve the outcome of islet xenotransplantation in streptozotocin-induced diabetic Cynomolgus monkeys. Immunosuppression consisted of thymoglobulin, anti-CD154 mAb for costimulation blockade, and mycophenolate mofetil. Following the transplantation of islets from wild-type pigs (n = 2) or from 1,3-galactosyltransferase gene-knockout pigs (n = 2), islets survived for a maximum of only 46 days, as evidenced by return to hyperglycemia and the need for exogenous insulin therapy. The transplantation of islets from hCD46 pigs resulted in graft survival and insulin-independent normoglycemia in four of five monkeys for the 3 months follow-up of the experiment. One normalized recipient, selected at random, was followed for >12 months. Inhibition of complement activation by the expression of hCD46 on the pig islets did not substantially reduce the initial loss of islet mass, rather was effective in limiting antibody-mediated rejection. This resulted in a reduced need for immunosuppression to preserve a sufficient islet mass to maintain normoglycemia long-term.
Collapse
Affiliation(s)
- D J van der Windt
- Division of Immunogenetics, Department of Pediatrics, Children's Hospital of Pittsburgh, of UPMC Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Sakata N, Obenaus A, Chan N, Mace J, Chinnock R, Hathout E. Factors affecting islet graft embolization in the liver of diabetic mice. Islets 2009; 1:26-33. [PMID: 20428330 PMCID: PMC2860388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND: Embolic occlusion of the portal vein due to islet transplantation is one of the major reasons for reduced survival of transplanted islets. In this study, we examined the location of islets as well as the correlation between islet and portal vein size after intraportal islet transplantation and evaluated liver and islet pathology. RESULTS: The liver was divided into peripheral and central sites. Islet and liver apoptosis/necrosis were significantly higher at peripheral sites. In regions without liver apoptosis or necrosis, portal vein diameter was significantly larger and embolic ratios were significantly lower. METHODS: BALB/c mice were intraportally transplanted with 800 islets and the liver was examined at postoperative day (POD) 0 (n = 7), POD 2 (n = 4) and POD 28 (n = 3). Liver specimens were stained for hematoxylin and eosin (necrosis), insulin and TUNEL (apoptosis). We evaluated distance from liver surface to islets, islet and portal vein diameter, embolic ratio (islet diameter/portal vein diameter), apoptosis/necrosis of islets and apoptosis/necrosis of the liver tissue surrounding the islet. CONCLUSION: Transplanted islets and liver tissue exhibited more injury at peripheral sites, in part, due to smaller diameters of portal venules that result in more frequent emboli following islet transplantation.
Collapse
Affiliation(s)
- Naoaki Sakata
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
- Division of Hepato-Biliary Pancreatic Surgery; Department of Surgery; Tohoku University Graduate School of Medicine; Sendai, Japan
| | - Andre Obenaus
- Department of Radiation Medicine, Loma Linda University School of Medicine; Loma Linda, CA USA
- Department of Radiology; Loma Linda University School of Medicine; Loma Linda, CA USA
| | - Nathaniel Chan
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
| | - John Mace
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
| | - Richard Chinnock
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
| | - Eba Hathout
- Islet Transplant Laboratory; Department of Pediatrics; Loma Linda University School of Medicine; Loma Linda, CA USA
- Correspondence to: Eba Hathout;
| |
Collapse
|
97
|
|