1
|
Di Piazza E, Todi L, Di Giuseppe G, Soldovieri L, Ciccarelli G, Brunetti M, Quero G, Alfieri S, Tondolo V, Pontecorvi A, Gasbarrini A, Nista EC, Giaccari A, Pani G, Mezza T. Advancing Diabetes Research: A Novel Islet Isolation Method from Living Donors. Int J Mol Sci 2024; 25:5936. [PMID: 38892122 PMCID: PMC11172646 DOI: 10.3390/ijms25115936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Pancreatic islet isolation is critical for type 2 diabetes research. Although -omics approaches have shed light on islet molecular profiles, inconsistencies persist; on the other hand, functional studies are essential, but they require reliable and standardized isolation methods. Here, we propose a simplified protocol applied to very small-sized samples collected from partially pancreatectomized living donors. Islet isolation was performed by digesting tissue specimens collected during surgery within a collagenase P solution, followed by a Lympholyte density gradient separation; finally, functional assays and staining with dithizone were carried out. Isolated pancreatic islets exhibited functional responses to glucose and arginine stimulation mirroring donors' metabolic profiles, with insulin secretion significantly decreasing in diabetic islets compared to non-diabetic islets; conversely, proinsulin secretion showed an increasing trend from non-diabetic to diabetic islets. This novel islet isolation method from living patients undergoing partial pancreatectomy offers a valuable opportunity for targeted study of islet physiology, with the primary advantage of being time-effective and successfully preserving islet viability and functionality. It enables the generation of islet preparations that closely reflect donors' clinical profiles, simplifying the isolation process and eliminating the need for a Ricordi chamber. Thus, this method holds promises for advancing our understanding of diabetes and for new personalized pharmacological approaches.
Collapse
Affiliation(s)
- Eleonora Di Piazza
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
| | - Laura Todi
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
| | - Gianfranco Di Giuseppe
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Laura Soldovieri
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Gea Ciccarelli
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Michela Brunetti
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Giuseppe Quero
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Digestive Surgery Unit, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
- Digestive Surgery Unit, Ospedale Isola Tiberina—Gemelli Isola, 00186 Roma, Italy
| | - Sergio Alfieri
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Digestive Surgery Unit, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
- Digestive Surgery Unit, Ospedale Isola Tiberina—Gemelli Isola, 00186 Roma, Italy
| | - Vincenzo Tondolo
- Digestive Surgery Unit, Ospedale Isola Tiberina—Gemelli Isola, 00186 Roma, Italy
| | - Alfredo Pontecorvi
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Antonio Gasbarrini
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
| | - Enrico Celestino Nista
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
| | - Andrea Giaccari
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Giovambattista Pani
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Teresa Mezza
- Department of Medicine and Translational Surgery, General Pathology Section, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Roma, Italy
| |
Collapse
|
2
|
Cheung P, Thorngren J, Zhang B, Vasylovska S, Lechi F, Persson J, Ståhl S, Löfblom J, Korsgren O, Eriksson J, Lau J, Eriksson O. Preclinical evaluation of Affibody molecule for PET imaging of human pancreatic islets derived from stem cells. EJNMMI Res 2023; 13:107. [PMID: 38100042 PMCID: PMC10724103 DOI: 10.1186/s13550-023-01057-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Beta-cell replacement methods such as transplantation of isolated donor islets have been proposed as a curative treatment of type 1 diabetes, but widespread application is challenging due to shortages of donor tissue and the need for continuous immunosuppressive treatments. Stem-cell-derived islets have been suggested as an alternative source of beta cells, but face transplantation protocols optimization difficulties, mainly due to a lack of available methods and markers to directly monitor grafts survival, as well as their localization and function. Molecular imaging techniques and particularly positron emission tomography has been suggested as a tool for monitoring the fate of islets after clinical transplantation. The integral membrane protein DGCR2 has been demonstrated to be a potential pancreatic islet biomarker, with specific expression on insulin-positive human embryonic stem-cell-derived pancreatic progenitor cells. The candidate Affibody molecule ZDGCR2:AM106 was radiolabeled with fluorine-18 using a novel click chemistry-based approach. The resulting positron emission tomography tracer [18F]ZDGCR2:AM106 was evaluated for binding to recombinant human DGCR2 and cryosections of stem-cell-derived islets, as well as in vivo using an immune-deficient mouse model transplanted with stem-cell-derived islets. Biodistribution of the [18F]ZDGCR2:AM106 was also assessed in healthy rats and pigs. RESULTS [18F]ZDGCR2:AM106 was successfully synthesized with high radiochemical purity and yield via a pretargeting approach. [18F]ZDGCR2:AM106 retained binding to recombinant human DCGR2 as well as to cryosectioned stem-cell-derived islets, but in vivo binding to native pancreatic tissue in both rat and pig was low. However, in vivo uptake of [18F]ZDGCR2:AM106 in stem-cell-derived islets transplanted in the immunodeficient mice was observed, albeit only within the early imaging frames after injection of the radiotracer. CONCLUSION Targeting of DGCR2 is a promising approach for in vivo detection of stem-cell-derived islets grafts by molecular imaging. The synthesis of [18F]ZDGCR2:AM106 was successfully performed via a pretargeting method to label a site-specific covalently bonded fluorine-18 to the Affibody molecule. However, the rapid washout of [18F]ZDGCR2:AM106 from the stem-cell-derived islets graft indicates that dissociation kinetics can be improved. Further studies using alternative binders of similar classes with improved binding potential are warranted.
Collapse
Affiliation(s)
- Pierre Cheung
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Julia Thorngren
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Bo Zhang
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | | | - Francesco Lechi
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jonas Persson
- Department of Protein Science, Division of Protein Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Stefan Ståhl
- Department of Protein Science, Division of Protein Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, Division of Protein Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jonas Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
3
|
Omar-Hmeadi M, Guček A, Barg S. Local PI(4,5)P 2 signaling inhibits fusion pore expansion during exocytosis. Cell Rep 2023; 42:112036. [PMID: 36701234 DOI: 10.1016/j.celrep.2023.112036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 11/04/2022] [Accepted: 01/11/2023] [Indexed: 01/26/2023] Open
Abstract
Phosphatidylinositol(4,5)bisphosphate (PI(4,5)P2) is an important signaling phospholipid that is required for regulated exocytosis and some forms of endocytosis. The two processes share a topologically similar pore structure that connects the vesicle lumen with the outside. Widening of the fusion pore during exocytosis leads to cargo release, while its closure initiates kiss&run or cavicapture endocytosis. We show here, using live-cell total internal reflection fluorescence (TIRF) microscopy of insulin granule exocytosis, that transient accumulation of PI(4,5)P2 at the release site recruits components of the endocytic fission machinery and stalls the late fusion pore expansion that is required for peptide release. The absence of clathrin differentiates this mechanism from clathrin-mediated endocytosis. Knockdown of phosphatidylinositol-phosphate-5-kinase-1c or optogenetic recruitment of 5-phosphatase reduces PI(4,5)P2 transients and accelerates fusion pore expansion, suggesting that acute PI(4,5)P2 synthesis is involved. Thus, local phospholipid signaling inhibits fusion pore expansion and peptide release through an unconventional endocytic mechanism.
Collapse
Affiliation(s)
- Muhmmad Omar-Hmeadi
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, 751 23 Uppsala, Sweden
| | - Alenka Guček
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, 751 23 Uppsala, Sweden
| | - Sebastian Barg
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, 751 23 Uppsala, Sweden.
| |
Collapse
|
4
|
[ 18F]MK-7246 for Positron Emission Tomography Imaging of the Beta-Cell Surface Marker GPR44. Pharmaceutics 2023; 15:pharmaceutics15020499. [PMID: 36839820 PMCID: PMC9962486 DOI: 10.3390/pharmaceutics15020499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The progressive loss of beta-cell mass is a hallmark of diabetes and has been suggested as a complementary approach to studying the progression of diabetes in contrast to the beta-cell function. Non-invasive nuclear medicinal imaging techniques such as Positron Emission Tomography using radiation emitting tracers have thus been suggested as more viable methodologies to visualize and quantify the beta-cell mass with sufficient sensitivity. The transmembrane G protein-coupled receptor GPR44 has been identified as a biomarker for monitoring beta-cell mass. MK-7246 is a GPR44 antagonist that selectively binds to GPR44 with high affinity and good pharmacokinetic properties. Here, we present the synthesis of MK-7246, radiolabeled with the positron emitter fluorine-18 for preclinical evaluation using cell lines, mice, rats and human pancreatic cells. Here, we have described a synthesis and radiolabeling method for producing [18F]MK-7246 and its precursor compound. Preclinical assessments demonstrated the strong affinity and selectivity of [18F]MK-7246 towards GPR44. Additionally, [18F]MK-7246 exhibited excellent metabolic stability, a fast clearance profile from blood and tissues, qualifying it as a promising radioactive probe for GPR44-directed PET imaging.
Collapse
|
5
|
Omar-Hmeadi M, Liu L, Echeverry S, Barg S. Quantification of Secretory Granule Exocytosis by TIRF Imaging and Capacitance Measurements. Methods Mol Biol 2023; 2565:179-186. [PMID: 36205894 DOI: 10.1007/978-1-0716-2671-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Hormones and neurotransmitters are released from (neuro)endocrine cells by regulated exocytosis of secretory granules. During exocytosis, the granule membrane fuses with the plasma membrane, which allows release of the stored content into the bloodstream or the surrounding tissue. Here, we give a detailed description of two complementary methods to observe and quantify exocytosis in single cells: high-resolution TIRF microscopy and patch-clamp capacitance recordings. Precise stimulation of exocytosis is achieved by local pressure application or voltage-clamp depolarizations. While the chapter is focused on insulin-secreting cells as an accessible and disease-relevant model system, the methodology is applicable to a wide variety of secretory cells including chromaffin and PC12 cells.
Collapse
Affiliation(s)
| | - Liangwen Liu
- Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Sebastian Barg
- Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
6
|
Teratani T, Kasahara N, Fujimoto Y, Sakuma Y, Miki A, Goto M, Sata N, Kitayama J. Mesenchymal Stem Cells Secretions Enhanced ATP Generation on Isolated Islets during Transplantation. Islets 2022; 14:69-81. [PMID: 35034568 PMCID: PMC8765074 DOI: 10.1080/19382014.2021.2022423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The success of islet transplantation in both basic research and clinical settings has proven that cell therapy has the potential to cure diabetes. Islets intended for transplantation are inevitably subjected to damage from a number of sources, including ischemic injury during removal and delivery of the donor pancreas, enzymatic digestion during islet isolation, and reperfusion injury after transplantation in the recipient. Here, we found that protein factors secreted by porcine adipose-tissue mesenchymal stem cells (AT-MSCs) were capable of activating preserved porcine islets. A conditioned medium was prepared from the supernatant obtained by culturing porcine AT-MSCs for 2 days in serum-free medium. Islets were preserved at 4°C in University of Wisconsin solution during transportation and then incubated at 37°C in RPMI-1620 medium with fractions of various molecular weights prepared from the conditioned medium. After treatment with certain fractions of the AT-MSC secretions, the intracellular ATP levels of the activated islets had increased to over 160% of their initial values after 4 days of incubation. Our novel system may be able to restore the condition of isolated islets after transportation or preservation and may help to improve the long-term outcome of islet transplantation.Abbreviations: AT-MSC, adipose-tissue mesenchymal stem cell; Cas-3, caspase-3; DAPI, 4,6-diamidino-2-phenylindole; DTZ, dithizone; ES cell, embryonic stem cell; FITC, fluorescein isothiocyanate; IEQ, islet equivalent; INS, insulin; iPS cell, induced pluripotent stem cell; Luc-Tg rat, luciferase-transgenic rat; PCNA, proliferating cell nuclear antigen; PDX1, pancreatic and duodenal homeobox protein-1; UW, University of Wisconsin; ZO1, zona occludens 1.
Collapse
Affiliation(s)
- Takumi Teratani
- Division of Translational Research, Jichi Medical University, Tochigi, Japan
- Department of Surgery, Jichi Medical University, Tochigi, Japan
- CONTACT Takumi Teratani Division of Clinical Investigation, Jichi Medical University, 3311-1, Yakushiji, Shimotsukeshi, Tochigi329-0498, Japan
| | - Naoya Kasahara
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | | | - Yasunaru Sakuma
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Atsushi Miki
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Masafumi Goto
- New Industry Creation Hatchery Center, Tohoku University, Miyagi, Japan
| | - Naohiro Sata
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Joji Kitayama
- Division of Translational Research, Jichi Medical University, Tochigi, Japan
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
7
|
Granlund L, Hedin A, Korsgren O, Skog O, Lundberg M. Altered microvasculature in pancreatic islets from subjects with type 1 diabetes. PLoS One 2022; 17:e0276942. [PMID: 36315525 PMCID: PMC9621430 DOI: 10.1371/journal.pone.0276942] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022] Open
Abstract
AIMS The transcriptome of different dissociated pancreatic islet cells has been described in enzymatically isolated islets in both health and disease. However, the isolation, culturing, and dissociation procedures likely affect the transcriptome profiles, distorting the biological conclusions. The aim of the current study was to characterize the cells of the islets of Langerhans from subjects with and without type 1 diabetes in a way that reflects the in vivo situation to the highest possible extent. METHODS Islets were excised using laser capture microdissection directly from frozen pancreatic tissue sections obtained from organ donors with (n = 7) and without (n = 8) type 1 diabetes. Transcriptome analysis of excised samples was performed using AmpliSeq. Consecutive pancreatic sections were used to estimate the proportion of beta-, alpha-, and delta cells using immunofluorescence and to examine the presence of CD31 positive endothelial regions using immunohistochemistry. RESULTS The proportion of beta cells in islets from subjects with type 1 diabetes was reduced to 0% according to both the histological and transcriptome data, and several alterations in the transcriptome were derived from the loss of beta cells. In total, 473 differentially expressed genes were found in the islets from subjects with type 1 diabetes. Functional enrichment analysis showed that several of the most upregulated gene sets were related to vasculature and angiogenesis, and histologically, vascular density was increased in subjects with type 1 diabetes. Downregulated in type 1 diabetes islets was the gene set epithelial mesenchymal transition. CONCLUSION A number of transcriptional alterations are present in islets from subjects with type 1 diabetes. In particular, several gene sets related to vasculature and angiogenesis are upregulated and there is an increased vascular density, suggesting an altered microvasculature in islets from subjects with type 1 diabetes. By studying pancreatic islets extracted directly from snap-frozen pancreatic tissue, this study reflects the in vivo situation to a high degree and gives important insights into islet pathophysiology in type 1 diabetes.
Collapse
Affiliation(s)
- Louise Granlund
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Anders Hedin
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Marcus Lundberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
8
|
Development of a novel method for measuring tissue oxygen pressure to improve the hypoxic condition in subcutaneous islet transplantation. Sci Rep 2022; 12:14731. [PMID: 36042259 PMCID: PMC9427780 DOI: 10.1038/s41598-022-19189-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/25/2022] [Indexed: 11/08/2022] Open
Abstract
Subcutaneous tissue is a promising site for islet transplantation, but poor engraftment, due to hypoxia and low vascularity, hinders its prevalence. However, oxygen partial pressure (pO2) of the subcutaneous space (SC) and other sites were reported to be equivalent in several previous reports. This contradiction may be based on accidental puncture to the indwelling micro-vessels in target tissues. We therefore developed a novel optical sensor system, instead of a conventional Clark-type needle probe, for measuring tissue pO2 and found that pO2 of the SC was extremely low in comparison to other sites. To verify the utility of this method, we transplanted syngeneic rat islets subcutaneously into diabetic recipients under several oxygenation conditions using an oxygen delivery device, then performed pO2 measurement, glucose tolerance, and immunohistochemistry. The optical sensor system was validated by correlating the pO2 values with the transplanted islet function. Interestingly, this novel technique revealed that islet viability estimated by ATP/DNA assay reduced to less than 75% by hypoxic condition at the SC, indicating that islet engraftment may substantially improve if the pO2 levels reach those of the renal subcapsular space. Further refinements for a hypoxic condition using the present technique may contribute to improving the efficiency of subcutaneous islet transplantation.
Collapse
|
9
|
De Paep DL, Van Hulle F, Ling Z, Vanhoeij M, Hilbrands R, Distelmans W, Gillard P, Keymeulen B, Pipeleers D, Jacobs-Tulleneers-Thevissen D. Utility of Islet Cell Preparations From Donor Pancreases After Euthanasia. Cell Transplant 2022; 31:9636897221096160. [PMID: 35583214 PMCID: PMC9125111 DOI: 10.1177/09636897221096160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Patients fulfilling criteria for euthanasia can choose to donate their organs after circulatory death [donors after euthanasia (DCD V)]. This study assesses the outcome of islet cell isolation from DCD V pancreases. A procedure for DCD V procurement provided 13 pancreases preserved in Institut Georges Lopez-1 preservation solution and following acirculatory warm ischemia time under 10 minutes. Islet cell isolation outcomes are compared with those from reference donors after brain death (DBD, n = 234) and a cohort of donors after controlled circulatory death (DCD III, n = 29) procured under the same conditions. Islet cell isolation from DCD V organs resulted in better in vitro outcome than for selected DCD III or reference DBD organs. A 50% higher average beta cell number before and after culture and a higher average beta cell purity (35% vs 24% and 25%) was observed, which led to more frequent selection for our clinical protocol (77% of isolates vs 50%). The functional capacity of a DCD V islet cell preparation was illustrated by its in vivo effect following intraportal transplantation in a type 1 diabetes patient: injection of 2 million beta cells/kg body weight (1,900 IEQ/kg body weight) at 39% insulin purity resulted in an implant with functional beta cell mass that represented 30% of that in non-diabetic controls. In conclusion, this study describes procurement and preservation conditions for donor organs after euthanasia, which allow preparation of cultured islet cells, that more frequently meet criteria for clinical use than those from DBD or DCD III organs.
Collapse
Affiliation(s)
- Diedert L De Paep
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium.,Beta Cell Bank, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Surgery, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Freya Van Hulle
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Zhidong Ling
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium.,Beta Cell Bank, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marian Vanhoeij
- Department of Surgery, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robert Hilbrands
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium.,Diabetes Clinic, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wim Distelmans
- Supportive and Palliative Care, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pieter Gillard
- Diabetes Clinic, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Bart Keymeulen
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium.,Diabetes Clinic, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Daniel Pipeleers
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Daniel Jacobs-Tulleneers-Thevissen
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium.,Beta Cell Bank, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Surgery, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
10
|
Bergström M, Yao M, Müller M, Korsgren O, von Zur-Mühlen B, Lundgren T. Autologous regulatory T cells in clinical intraportal allogenic pancreatic islet transplantation. Transpl Int 2021; 34:2816-2823. [PMID: 34787936 DOI: 10.1111/tri.14163] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/20/2021] [Accepted: 11/07/2021] [Indexed: 01/19/2023]
Abstract
Allogeneic islet transplantation in type 1 diabetes requires lifelong immunosuppression to prevent graft rejection. This medication can cause adverse effects and increases the susceptibility for infections and malignancies. Adoptive therapies with regulatory T cells (Tregs) have shown promise in reducing the need for immunosuppression in human transplantation settings but have previously not been evaluated in islet transplantation. In this study, five patients with type 1 diabetes undergoing intraportal allogeneic islet transplantation were co-infused with polyclonal autologous Tregs under a standard immunosuppressive regimen. Patients underwent leaukapheresis from which Tregs were purified by magnetic-activated cell sorting (MACS) and cryopreserved until transplantation. Dose ranges of 0.14-1.27 × 106 T cells per kilo bodyweight were transplanted. No negative effects were seen related to the Treg infusion, regardless of cell dose. Only minor complications related to the immunosuppressive drugs were reported. This first-in-man study of autologous Treg infusion in allogenic pancreatic islet transplantation shows that the treatment is safe and feasible. Based on these results, future efficacy studies will be developed under the label of advanced therapeutic medical products (ATMP), using modified or expanded Tregs with the aim of minimizing the need for chronic immunosuppressive medication in islet transplantation.
Collapse
Affiliation(s)
- Marcus Bergström
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Uppsala, Sweden
| | - Ming Yao
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden.,Department of Transplantation Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Malin Müller
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Uppsala, Sweden
| | - Bengt von Zur-Mühlen
- Department of Surgical Sciences, Transplantation Surgery, Uppsala University, Uppsala, Sweden
| | - Torbjörn Lundgren
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden.,Department of Transplantation Surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
11
|
Yao M, Domogatskaya A, Ågren N, Watanabe M, Tokodai K, Brines M, Cerami A, Ericzon BG, Kumagai-Braesch M, Lundgren T. Cibinetide Protects Isolated Human Islets in a Stressful Environment and Improves Engraftment in the Perspective of Intra Portal Islet Transplantation. Cell Transplant 2021; 30:9636897211039739. [PMID: 34498509 PMCID: PMC8436319 DOI: 10.1177/09636897211039739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
During intra-portal pancreatic islet transplantation (PITx), innate immune reactions such as the instant blood mediated inflammatory reaction (IBMIR) cause an immediate loss of islets. The non-hematopoietic erythropoietin analogue cibinetide has previously shown islet-protective effects in mouse PITx. Herein, we aimed to confirm cibinetide's efficacy on human islets, and to characterize its effect on IBMIR. We cultured human islets with pro-inflammatory cytokines for 18 hours with or without cibinetide. ATP content and caspase 3/7 activity were measured. Dynamic glucose perfusion assay was used to evaluate islet function. To evaluate cibinetides effect on IBMIR, human islets were incubated in heparinized polyvinyl chloride tubing system with ABO compatible blood and rotated for 60 minutes to mimic the portal vein system. Moreover, human islets were transplanted into athymic mice livers via the portal vein with or without perioperative cibinetide treatment. The mice were sacrificed six days following transplantation and the livers were analyzed for human insulin and serum for human C-peptide levels. Histological examination of recipient livers to evaluate islet graft infiltration by CD11b+ cells was performed. Our results show that cibinetide maintained human islet ATP levels and reduced the caspase 3/7 activity during culture with pro-inflammatory cytokines and improved their insulin secreting capacity. In the PVC loop system, administration of cibinetide reduced the IBMIR-induced platelet consumption. In human islet to athymic mice PITx, cibinetide treatment showed an increased amount of human insulin in the livers and higher serum human C-peptide, while histological examination of the livers showed reduced infiltration of pro-inflammatory CD11b+ cells around islets grafts compared to the controls. In summary, Cibinetide protected isolated human islets in a pro-inflammatory milieu and reduced IBMIR related platelet consumption. It improved engraftment of human islets in athymic mice. The study confirms that cibinetide is a promising agent to be used in clinical PITx.
Collapse
Affiliation(s)
- Ming Yao
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Anna Domogatskaya
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Nils Ågren
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Masaaki Watanabe
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Kazuaki Tokodai
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | | | | | - Bo-Göran Ericzon
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Makiko Kumagai-Braesch
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Torbjörn Lundgren
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, and Department of Transplantation Surgery, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| |
Collapse
|
12
|
Imura T, Inagaki A, Igarashi Y, Goto M. Optimization of dye solutions for detecting damaged pancreatic tissues during islet isolation procedures. PLoS One 2021; 16:e0255733. [PMID: 34388180 PMCID: PMC8362985 DOI: 10.1371/journal.pone.0255733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/22/2021] [Indexed: 12/29/2022] Open
Abstract
We previously reported that dye was effective to prevent the leakage of enzyme solutions from pancreatic glands during an islet isolation procedure. However, the dye used for islet isolation has not yet been optimized. In this study, we focused on pyoktanin blue (PB), diagnogreen (DG), and indigo carmine (IC) as potential candidates among clinically established dyes. A serial dilution assay was performed to determine minimal effective concentrations of each dye for detecting damaged pancreatic tissues. According to the outcome of serial dilution assays, double minimum effective concentrations of each dye were used for in vitro toxicity assays on islets and used in the isolation procedure to investigate whether they adversely affect islet isolation efficiency. The evaluations included islet yield, ADP/ATP, ATP/DNA, glucose stimulation test, and insulin/DNA assays. Islet viability cultured with PB contained medium was significantly lower than the other dyes. DG and IC appeared to be non-toxic to the islets. In isolation experiments, the islet yield in the DG group was considerably lower than that in the Control group, suggesting that DG might inhibit enzyme activity. The present study demonstrates that IC could be a promising candidate for an effective dye to detect damaged pancreatic tissues without affecting the enzyme activity and islet quality.
Collapse
Affiliation(s)
- Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Yasuhiro Igarashi
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Masafumi Goto
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
13
|
Doppenberg JB, Leemkuil M, Engelse MA, Krikke C, de Koning EJP, Leuvenink HGD. Hypothermic oxygenated machine perfusion of the human pancreas for clinical islet isolation: a prospective feasibility study. Transpl Int 2021; 34:1397-1407. [PMID: 34036616 PMCID: PMC8456912 DOI: 10.1111/tri.13927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 11/28/2022]
Abstract
Due to an increasing scarcity of pancreases with optimal donor characteristics, islet isolation centers utilize pancreases from extended criteria donors, such as from donation after circulatory death (DCD) donors, which are particularly susceptible to prolonged cold ischemia time (CIT). We hypothesized that hypothermic machine perfusion (HMP) can safely increase CIT. Five human DCD pancreases were subjected to 6 h of oxygenated HMP. Perfusion parameters, apoptosis, and edema were measured prior to islet isolation. Five human DBD pancreases were evaluated after static cold storage (SCS). Islet viability, and in vitro and in vivo functionality in diabetic mice were analyzed. Islets were isolated from HMP pancreases after 13.4 h [12.9–14.5] CIT and after 9.2 h [6.5–12.5] CIT from SCS pancreases. Histological analysis of the pancreatic tissue showed that HMP did not induce edema nor apoptosis. Islets maintained >90% viable during culture, and an appropriate in vitro and in vivo function in mice was demonstrated after HMP. The current study design does not permit to demonstrate that oxygenated HMP allows for cold ischemia extension; however, the successful isolation of functional islets from discarded human DCD pancreases after performing 6 h of oxygenated HMP indicates that oxygenated HMP may be a useful technology for better preservation of pancreases.
Collapse
Affiliation(s)
- Jason B Doppenberg
- Transplantation Center, Leiden University Medical Center, Leiden, the Netherlands
| | - Marjolein Leemkuil
- Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - Marten A Engelse
- Transplantation Center, Leiden University Medical Center, Leiden, the Netherlands
| | - Christina Krikke
- Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - Eelco J P de Koning
- Transplantation Center, Leiden University Medical Center, Leiden, the Netherlands
| | - Henri G D Leuvenink
- Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
14
|
Nagaya M, Hasegawa K, Uchikura A, Nakano K, Watanabe M, Umeyama K, Matsunari H, Osafune K, Kobayashi E, Nakauchi H, Nagashima H. Feasibility of large experimental animal models in testing novel therapeutic strategies for diabetes. World J Diabetes 2021; 12:306-330. [PMID: 33889282 PMCID: PMC8040081 DOI: 10.4239/wjd.v12.i4.306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/30/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes is among the top 10 causes of death in adults and caused approximately four million deaths worldwide in 2017. The incidence and prevalence of diabetes is predicted to increase. To alleviate this potentially severe situation, safer and more effective therapeutics are urgently required. Mice have long been the mainstay as preclinical models for basic research on diabetes, although they are not ideally suited for translating basic knowledge into clinical applications. To validate and optimize novel therapeutics for safe application in humans, an appropriate large animal model is needed. Large animals, especially pigs, are well suited for biomedical research and share many similarities with humans, including body size, anatomical features, physiology, and pathophysiology. Moreover, pigs already play an important role in translational studies, including clinical trials for xenotransplantation. Progress in genetic engineering over the past few decades has facilitated the development of transgenic animals, including porcine models of diabetes. This article discusses features that attest to the attractiveness of genetically modified porcine models of diabetes for testing novel treatment strategies using recent technical advances.
Collapse
Affiliation(s)
- Masaki Nagaya
- Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
- Department of Immunology, St. Marianna University School of Medicine, Kawasaki 261-8511, Kanagawa, Japan
| | - Koki Hasegawa
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
| | - Ayuko Uchikura
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
| | - Kazuaki Nakano
- Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
- Research and Development, PorMedTec Co. Ltd, Kawasaki 214-0034, Kanagawa, Japan
| | - Masahito Watanabe
- Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
- Research and Development, PorMedTec Co. Ltd, Kawasaki 214-0034, Kanagawa, Japan
| | - Kazuhiro Umeyama
- Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
- Research and Development, PorMedTec Co. Ltd, Kawasaki 214-0034, Kanagawa, Japan
| | - Hitomi Matsunari
- Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Kyoto, Japan
| | - Eiji Kobayashi
- Department of Organ Fabrication, Keio University School of Medicine, Shinjuku 160-8582, Tokyo, Japan
| | - Hiromitsu Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine, Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, United States
- Division of Stem Cell Therapy, Institute of Medical Science, The University of Tokyo, Minato 108-8639, Tokyo, Japan
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki 214-8571, Kanagawa, Japan
| |
Collapse
|
15
|
Saitoh Y, Inagaki A, Fathi I, Imura T, Nishimaki H, Ogasawara H, Matsumura M, Miyagi S, Yasunami Y, Unno M, Kamei T, Goto M. Improvement of hepatocyte engraftment by co-transplantation with pancreatic islets in hepatocyte transplantation. J Tissue Eng Regen Med 2021; 15:361-374. [PMID: 33484496 PMCID: PMC8048420 DOI: 10.1002/term.3170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 12/05/2020] [Accepted: 01/06/2021] [Indexed: 12/15/2022]
Abstract
Because of the fragility of isolated hepatocytes, extremely poor engraftment of transplanted hepatocytes remains a severe issue in hepatocyte transplantation. Therefore, improving hepatocyte engraftment is necessary to establish hepatocyte transplantation as a standard therapy. Since the pancreatic islets are known to have favorable autocrine effects, we hypothesized that the transplanted islets might influence not only the islets but also the nearby hepatocytes, subsequently promoting engraftment. We evaluated the effects of islet co-transplantation using an analbuminemic rat model (in vivo model). Furthermore, we established a mimicking in vitro model to investigate the underlying mechanisms. In an in vivo model, the hepatocyte engraftment was significantly improved only when the islets were co-transplanted to the nearby hepatocytes (p < 0.001). Moreover, the transplanted hepatocytes appeared to penetrate the renal parenchyma together with the co-transplanted islets. In an in vitro model, the viability of cultured hepatocytes was also improved by coculture with pancreatic islets. Of particular interest, the coculture supernatant alone could also exert beneficial effects comparable to islet coculture. Although insulin, VEGF, and GLP-1 were selected as candidate crucial factors using the Bio-Plex system, beneficial effects were partially counteracted by anti-insulin receptor antibodies. In conclusion, this study demonstrated that islet co-transplantation improves hepatocyte engraftment, most likely due to continuously secreted crucial factors, such as insulin, in combination with providing favorable circumstances for hepatocyte engraftment. Further refinements of this approach, especially regarding substitutes for islets, could be a promising strategy for improving the outcomes of hepatocyte transplantation.
Collapse
Affiliation(s)
- Yoshikatsu Saitoh
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Ibrahim Fathi
- Division of Transplantation and Regenerative MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Takehiro Imura
- Division of Transplantation and Regenerative MedicineTohoku University Graduate School of MedicineSendaiJapan
| | - Hiroyasu Nishimaki
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Hiroyuki Ogasawara
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Muneyuki Matsumura
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Shigehito Miyagi
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | | | - Michiaki Unno
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Takashi Kamei
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Masafumi Goto
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
- Division of Transplantation and Regenerative MedicineTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
16
|
Wassmer CH, Perrier Q, Combescure C, Pernin N, Parnaud G, Cottet-Dumoulin D, Brioudes E, Bellofatto K, Lebreton F, Berishvili E, Lablanche S, Kessler L, Wojtusciszyn A, Buron F, Borot S, Bosco D, Berney T, Lavallard V. Impact of ischemia time on islet isolation success and posttransplantation outcomes: A retrospective study of 452 pancreas isolations. Am J Transplant 2021; 21:1493-1502. [PMID: 32986297 DOI: 10.1111/ajt.16320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/03/2020] [Accepted: 09/04/2020] [Indexed: 01/25/2023]
Abstract
Many variables impact islet isolation, including pancreas ischemia time. The ischemia time upper limit that should be respected to avoid a negative impact on the isolation outcome is not well defined. We have performed a retrospective analysis of all islet isolations in our center between 2008 and 2018. Total ischemia time, cold ischemia time, and organ removal time were analyzed. Isolation success was defined as an islet yield ≥200 000 IEQ. Of the 452 pancreases included, 288 (64%) were successfully isolated. Probability of isolation success showed a significant decrease after 8 hours of total ischemia time, 7 hours of cold ischemia time, and 80 minutes of organ removal time. Although we observed an impact of ischemia time on islet yield, a probability of isolation success of 50% was still present even when total ischemia time exceeds 12 hours. Posttransplantation clinical outcomes were assessed in 32 recipients and no significant difference was found regardless of ischemia time. These data indicate that although shorter ischemia times are associated with better islet isolation outcomes, total ischemia time >12 hours can provide excellent results in appropriately selected donors.
Collapse
Affiliation(s)
- Charles-Henri Wassmer
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Quentin Perrier
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Christophe Combescure
- Division of Clinical Epidemiology, Faculty of Medicine, University of Geneva, and Geneva University Hospitals, Geneva, Switzerland
| | - Nadine Pernin
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Géraldine Parnaud
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - David Cottet-Dumoulin
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Estelle Brioudes
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Kevin Bellofatto
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Fanny Lebreton
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland.,Institute of Medical Research, Ilia State University, Tbilisi, Georgia
| | - Sandrine Lablanche
- Endocrinology Department, Grenoble Alpes University Hospital, Grenoble, France
| | - Laurence Kessler
- Department of Diabetology, University Hospital, Strasbourg, France.,Federation of Translational Medicine of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Anne Wojtusciszyn
- Department of Endocrinology, Diabetes, and Nutrition, Montpellier University Hospital, Montpellier, France.,Laboratory of Cell Therapy of Diabetes, Institute of Functional Genomics, Mixed Research Unit, French National Center for Scientific Research 5203, Inserm U1191, University of Montpellier, Montpellier, France
| | - Fanny Buron
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Sophie Borot
- Endocrinology Department, Besancon University Hospital, Besancon, France
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Vanessa Lavallard
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| |
Collapse
|
17
|
Uitbeijerse BS, Nijhoff MF, Sont JK, de Koning EJP. Fasting parameters for estimation of stimulated β cell function in islet transplant recipients with or without basal insulin treatment. Am J Transplant 2021; 21:297-306. [PMID: 32524720 PMCID: PMC7818182 DOI: 10.1111/ajt.16135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/11/2020] [Accepted: 05/27/2020] [Indexed: 01/25/2023]
Abstract
In order to assess β cell secretory capacity after islet transplantation, standardized mixed meal stimulation tests are often used. But these tests are cumbersome and the effect of exogenous insulin on the test results is unclear. The aim of our study was to determine to what extent fasting glycemic indices can estimate stimulated β cell function in islet transplant recipients with and without basal insulin. In total 100 mixed meal stimulation tests, including 31 with concurrent basal insulin treatment, were performed in 36 islet transplant recipients. In a multivariate model, fasting C-peptide and fasting glucose together estimated peak C-peptide with R2 = .87 and area under the curve (AUC) C-peptide with a R2 = .93. There was a larger increase of glucose during tests in which exogenous insulin was used (+7.9 vs +5.3 mmol/L, P < .001) and exogenous insulin use was associated with a slightly lower estimated peak C-peptide (relative change: -15%, P = .02). In islet transplant recipients the combination of fasting C-peptide and glucose can be used to accurately estimate stimulated β cell function after a mixed meal stimulation test, whether exogenous basal insulin is present or not. These data indicate that graft function can be reliably determined during exogenous insulin treatment and that regular islet graft stimulation tests can be minimized.
Collapse
Affiliation(s)
- Bas S. Uitbeijerse
- Department of Internal MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Michiel F. Nijhoff
- Department of Internal MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Jacob K. Sont
- Department of Biomedical Data SciencesSection Medical Decision MakingLeiden University Medical CenterLeidenthe Netherlands
| | - Eelco J. P. de Koning
- Department of Internal MedicineLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
18
|
Jonsson A, Hedin A, Müller M, Skog O, Korsgren O. Transcriptional profiles of human islet and exocrine endothelial cells in subjects with or without impaired glucose metabolism. Sci Rep 2020; 10:22315. [PMID: 33339897 PMCID: PMC7749106 DOI: 10.1038/s41598-020-79313-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/04/2020] [Indexed: 11/21/2022] Open
Abstract
In experimental studies, pancreatic islet microvasculature is essential for islet endocrine function and mass, and islet vascular morphology is altered in diabetic subjects. Even so, almost no information is available concerning human islet microvascular endothelial cell (MVEC) physiology and gene expression. In this study, islets and exocrine pancreatic tissue were acquired from organ donors with normoglycemia or impaired glucose metabolism (IGM) immediately after islet isolation. Following single-cell dissociation, primary islet- and exocrine MVECs were obtained through fluorescence-activated cell sorting (FACS) and transcriptional profiles were generated using AmpliSeq. Multiple gene sets involved in general vascular development and extracellular matrix remodeling were enriched in islet MVEC. In exocrine MVEC samples, multiple enriched gene sets that relate to biosynthesis and biomolecule catabolism were found. No statistically significant enrichment was found in gene sets related to autophagy or endoplasmic reticulum (ER) stress. Although ample differences were found between islet- and exocrine tissue endothelial cells, no differences could be observed between normoglycemic donors and donors with IGM at gene or gene set level. Our data is consistent with active angiogenesis and vascular remodeling in human islets and support the notion of ongoing endocrine pancreas tissue repair and regeneration even in the adult human.
Collapse
Affiliation(s)
- Alexander Jonsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Anders Hedin
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Malin Müller
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
19
|
Heterogeneity of Human Pancreatic Islet Isolation Around Europe: Results of a Survey Study. Transplantation 2020; 104:190-196. [PMID: 31365472 DOI: 10.1097/tp.0000000000002777] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Europe is currently the most active region in the field of pancreatic islet transplantation, and many of the leading groups are actually achieving similar good outcomes. Further collaborative advances in the field require the standardization of islet cell product isolation processes, and this work aimed to identify differences in the human pancreatic islet isolation processes within European countries. METHODS A web-based questionnaire about critical steps, including donor selection, pancreas processing, pancreas perfusion and digestion, islet counting and culture, islet quality evaluation, microbiological evaluation, and release criteria of the product, was completed by isolation facilities participating at the Ninth International European Pancreas and Islet Transplant Association (EPITA) Workshop on Islet-Beta Cell Replacement in Milan. RESULTS Eleven islet isolation facilities completed the questionnaire. The facilities reported 445 and 53 islet isolations per year over the last 3 years from deceased organ donors and pancreatectomized patients, respectively. This activity resulted in 120 and 40 infusions per year in allograft and autograft recipients, respectively. Differences among facilities emerged in donor selection (age, cold ischemia time, intensive care unit length, amylase concentration), pancreas procurement, isolation procedures (brand and concentration of collagenase, additive, maximum acceptable digestion time), quality evaluation, and release criteria for transplantation (glucose-stimulated insulin secretion tests, islet numbers, and purity). Moreover, even when a high concordance about the relevance of one parameter was evident, thresholds for the acceptance were different among facilities. CONCLUSIONS The result highlighted the presence of a heterogeneity in the islet cell product process and product release criteria.
Collapse
|
20
|
Omar-Hmeadi M, Lund PE, Gandasi NR, Tengholm A, Barg S. Paracrine control of α-cell glucagon exocytosis is compromised in human type-2 diabetes. Nat Commun 2020; 11:1896. [PMID: 32312960 PMCID: PMC7171169 DOI: 10.1038/s41467-020-15717-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/23/2020] [Indexed: 01/05/2023] Open
Abstract
Glucagon is released from pancreatic α-cells to activate pathways that raise blood glucose. Its secretion is regulated by α-cell-intrinsic glucose sensing and paracrine control through insulin and somatostatin. To understand the inadequately high glucagon levels that contribute to hyperglycemia in type-2 diabetes (T2D), we analyzed granule behavior, exocytosis and membrane excitability in α-cells of 68 non-diabetic and 21 T2D human donors. We report that exocytosis is moderately reduced in α-cells of T2D donors, without changes in voltage-dependent ion currents or granule trafficking. Dispersed α-cells have a non-physiological V-shaped dose response to glucose, with maximal exocytosis at hyperglycemia. Within intact islets, hyperglycemia instead inhibits α-cell exocytosis, but not in T2D or when paracrine inhibition by insulin or somatostatin is blocked. Surface expression of somatostatin-receptor-2 is reduced in T2D, suggesting a mechanism for the observed somatostatin resistance. Thus, elevated glucagon in human T2D may reflect α-cell insensitivity to paracrine inhibition at hyperglycemia. Glucagon is elevated Type-2 diabetes, which contributes to poor glucose control in patients with the disease. Here the authors report that secretion of the hormone is controlled by paracrine inhibition, and that resistance of α-cells to somatostatin can explain hyperglucagonemia in type-2 diabetes.
Collapse
Affiliation(s)
- Muhmmad Omar-Hmeadi
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Per-Eric Lund
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Nikhil R Gandasi
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Anders Tengholm
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Sebastian Barg
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden.
| |
Collapse
|
21
|
Nilsson J, Fardoos R, Hansen L, Lövkvist H, Pietras K, Holmberg D, Schmidt-Christensen A. Recruited fibroblasts reconstitute the peri-islet membrane: a longitudinal imaging study of human islet grafting and revascularisation. Diabetologia 2020; 63:137-148. [PMID: 31701200 PMCID: PMC6890581 DOI: 10.1007/s00125-019-05018-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 08/22/2019] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS Rapid and adequate islet revascularisation and restoration of the islet-extracellular matrix (ECM) interaction are significant factors influencing islet survival and function of the transplanted islets in individuals with type 1 diabetes. Because the ECM encapsulating the islets is degraded during islet isolation, understanding the process of revascularisation and engraftment after transplantation is essential and needs further investigation. METHODS Here we apply a longitudinal and high-resolution imaging approach to investigate the dynamics of the pancreatic islet engraftment process up to 11 months after transplantation. Human and mouse islet grafts were inserted into the anterior chamber of the mouse eye, using a NOD.ROSA-tomato.Rag2-/- or B6.ROSA-tomato host allowing the investigation of the expansion of host vs donor cells and the contribution of host cells to aspects such as promoting the encapsulation and vascularisation of the graft. RESULTS A fibroblast-like stromal cell population of host origin rapidly migrates to ensheath the transplanted islet and aid in the formation of a basement membrane-like structure. Moreover, we show that the vessel network, while reconstituted by host endothelial cells, still retains the overall architecture of the donor islets. CONCLUSIONS/INTERPRETATION In this transplantation situation the fibroblast-like stromal cells appear to take over as main producers of ECM or act as a scaffold for other ECM-producing cells to reconstitute a peri-islet-like basement membrane. This may have implications for our understanding of long-term graft rejection and for the design of novel strategies to interfere with this process.
Collapse
Affiliation(s)
- Julia Nilsson
- Department of Experimental Medical Science, Lund University, 221 84, Lund, Sweden
- Lund University Diabetes Centre, Malmö, Sweden
| | - Rabiah Fardoos
- Department of Experimental Medical Science, Lund University, 221 84, Lund, Sweden
| | - Lisbeth Hansen
- Department of Experimental Medical Science, Lund University, 221 84, Lund, Sweden
| | - Håkan Lövkvist
- Department of Clinical Sciences, Lund, Neurology, Lund University, Lund, Sweden
- Clinical Studies Sweden - Forum South, Unit for Medical Statistics and Epidemiology, Skåne University Hospital, Lund, Sweden
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, BioCARE, Lund University, Lund, Sweden
| | - Dan Holmberg
- Department of Experimental Medical Science, Lund University, 221 84, Lund, Sweden
- Lund University Diabetes Centre, Malmö, Sweden
| | - Anja Schmidt-Christensen
- Department of Experimental Medical Science, Lund University, 221 84, Lund, Sweden.
- Lund University Diabetes Centre, Malmö, Sweden.
| |
Collapse
|
22
|
Nguyen PM, Gandasi NR, Xie B, Sugahara S, Xu Y, Idevall-Hagren O. The PI(4)P phosphatase Sac2 controls insulin granule docking and release. J Cell Biol 2019; 218:3714-3729. [PMID: 31533953 PMCID: PMC6829663 DOI: 10.1083/jcb.201903121] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/20/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Insulin granule biogenesis involves transport to, and stable docking at, the plasma membrane before priming and fusion. Defects in this pathway result in impaired insulin secretion and are a hallmark of type 2 diabetes. We now show that the phosphatidylinositol 4-phosphate phosphatase Sac2 localizes to insulin granules in a substrate-dependent manner and that loss of Sac2 results in impaired insulin secretion. Sac2 operates upstream of granule docking, since loss of Sac2 prevented granule tethering to the plasma membrane and resulted in both reduced granule density and number of exocytic events. Sac2 levels correlated positively with the number of docked granules and exocytic events in clonal β cells and with insulin secretion in human pancreatic islets, and Sac2 expression was reduced in islets from type 2 diabetic subjects. Taken together, we identified a phosphoinositide switch on the surface on insulin granules that is required for stable granule docking at the plasma membrane and impaired in human type 2 diabetes.
Collapse
Affiliation(s)
- Phuoc My Nguyen
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Nikhil R Gandasi
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Beichen Xie
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Sari Sugahara
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Yingke Xu
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
| | | |
Collapse
|
23
|
Open Randomized Multicenter Study to Evaluate Safety and Efficacy of Low Molecular Weight Sulfated Dextran in Islet Transplantation. Transplantation 2019; 103:630-637. [PMID: 30211831 DOI: 10.1097/tp.0000000000002425] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND When transplanted human pancreatic islets are exposed to blood during intraportal infusion, an innate immune response is triggered. This instant blood-mediated inflammatory reaction (IBMIR) activates the coagulation and complement cascades and leads to the destruction of 25% of all transplanted islets within minutes, contributing to the need, in most patients, for islets from more than 1 donor. Low molecular dextran sulfate (LMW-DS) has been shown in experimental settings to inhibit IBMIR. METHODS The Clinical Islet Transplantation consortium 01 study was a phase II, multicenter, open label, active control, randomized study. Twenty-four subjects were randomized to peritransplant intraportal and systemic treatment with either LMW-DS or heparin, targeting an activated partial thromboplastin time of 150 ± 10 seconds and 50 ± 5 seconds, respectively. C-peptide response was measured with a mixed meal tolerance test at 75 and 365 days after transplant. RESULTS Low molecular dextran sulfate was safe and well tolerated with similar observed adverse events (mostly attributed to immunosuppression) as in the heparin arm. There was no difference in the primary endpoint (stimulated C-peptide 75 ± 5 days after the first transplant) between the 2 arms (1.33 ± 1.10 versus 1.56 ± 1.36 ng/mL, P = 0.66). Insulin requirement, metabolic parameters, Clarke and HYPO score, quality of life, and safety were similar between the 2 treatments groups. CONCLUSIONS Even with low dosing, LMW-DS showed similar efficacy in preventing IBMIR to promote islet engraftment when compared to "state-of-the art" treatment with heparin. Furthermore, no substantial differences in the efficacy and safety endpoints were detected, providing important information for future studies with more optimal dosing of LMW-DS for the prevention of IBMIR in islet transplantation.
Collapse
|
24
|
Helker CSM, Mullapudi ST, Mueller LM, Preussner J, Tunaru S, Skog O, Kwon HB, Kreuder F, Lancman JJ, Bonnavion R, Dong PDS, Looso M, Offermanns S, Korsgren O, Spagnoli FM, Stainier DYR. A whole organism small molecule screen identifies novel regulators of pancreatic endocrine development. Development 2019; 146:dev.172569. [PMID: 31142539 DOI: 10.1242/dev.172569] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 05/08/2019] [Indexed: 12/16/2022]
Abstract
An early step in pancreas development is marked by the expression of the transcription factor Pdx1 within the pancreatic endoderm, where it is required for the specification of all endocrine cell types. Subsequently, Pdx1 expression becomes restricted to the β-cell lineage, where it plays a central role in β-cell function. This pivotal role of Pdx1 at various stages of pancreas development makes it an attractive target to enhance pancreatic β-cell differentiation and increase β-cell function. In this study, we used a newly generated zebrafish reporter to screen over 8000 small molecules for modulators of pdx1 expression. We found four hit compounds and validated their efficacy at different stages of pancreas development. Notably, valproic acid treatment increased pancreatic endoderm formation, while inhibition of TGFβ signaling led to α-cell to β-cell transdifferentiation. HC toxin, another HDAC inhibitor, enhances β-cell function in primary mouse and human islets. Thus, using a whole organism screening strategy, this study identified new pdx1 expression modulators that can be used to influence different steps in pancreas and β-cell development.
Collapse
Affiliation(s)
- Christian S M Helker
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, 61231 Bad Nauheim, Germany .,Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, 35043 Marburg, Germany
| | - Sri-Teja Mullapudi
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, 61231 Bad Nauheim, Germany
| | - Laura M Mueller
- Centre for Stem Cells and Regenerative Medicine, King's College London, London WC2R 2LS, UK
| | - Jens Preussner
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics Core Unit, 61231 Bad Nauheim, Germany
| | - Sorin Tunaru
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, 61231 Bad Nauheim, Germany.,Biochemistry Institute of the Romanian Academy, Department of Enzymology, Bucharest 060031, Romania
| | - Oskar Skog
- Uppsala University, Department of Immunology, Genetics and Pathology, 751 85 Uppsala, Sweden
| | - Hyouk-Bum Kwon
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, 61231 Bad Nauheim, Germany
| | - Florian Kreuder
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, 61231 Bad Nauheim, Germany
| | - Joseph J Lancman
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Remy Bonnavion
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, 61231 Bad Nauheim, Germany
| | - P Duc Si Dong
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Mario Looso
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics Core Unit, 61231 Bad Nauheim, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, 61231 Bad Nauheim, Germany
| | - Ole Korsgren
- Uppsala University, Department of Immunology, Genetics and Pathology, 751 85 Uppsala, Sweden
| | - Francesca M Spagnoli
- Centre for Stem Cells and Regenerative Medicine, King's College London, London WC2R 2LS, UK
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, 61231 Bad Nauheim, Germany
| |
Collapse
|
25
|
Guček A, Gandasi NR, Omar-Hmeadi M, Bakke M, Døskeland SO, Tengholm A, Barg S. Fusion pore regulation by cAMP/Epac2 controls cargo release during insulin exocytosis. eLife 2019; 8:41711. [PMID: 31099751 PMCID: PMC6557626 DOI: 10.7554/elife.41711] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/28/2019] [Indexed: 12/20/2022] Open
Abstract
Regulated exocytosis establishes a narrow fusion pore as initial aqueous connection to the extracellular space, through which small transmitter molecules such as ATP can exit. Co-release of polypeptides and hormones like insulin requires further expansion of the pore. There is evidence that pore expansion is regulated and can fail in diabetes and neurodegenerative disease. Here, we report that the cAMP-sensor Epac2 (Rap-GEF4) controls fusion pore behavior by acutely recruiting two pore-restricting proteins, amisyn and dynamin-1, to the exocytosis site in insulin-secreting beta-cells. cAMP elevation restricts and slows fusion pore expansion and peptide release, but not when Epac2 is inactivated pharmacologically or in Epac2-/- (Rapgef4-/-) mice. Consistently, overexpression of Epac2 impedes pore expansion. Widely used antidiabetic drugs (GLP-1 receptor agonists and sulfonylureas) activate this pathway and thereby paradoxically restrict hormone release. We conclude that Epac2/cAMP controls fusion pore expansion and thus the balance of hormone and transmitter release during insulin granule exocytosis. Insulin is the hormone that signals to the body to take up sugar from the blood. Specialized cells in the pancreas – known as β-cells – release insulin after a meal. Before that, insulin molecules are stored in tiny granules inside the β-cells; these granules must fuse with the cells’ surface membranes to release their contents. The first step in this process creates a narrow pore that allows small molecules, but not the larger insulin molecules, to seep out. The pore then widens to release the insulin. Since the small molecules are known to act locally in the pancreas, it is possible that this “molecular sieve” is biologically important. Yet it is not clear how the pore widens. One of the problems for people with type 2 diabetes is that they release less insulin into the bloodstream. Two kinds of drugs used to treat these patients work by stimulating β-cells to release their insulin. One way to achieve this is by raising the levels of a small molecule called cAMP, which is well known to help prepare insulin granules for release. The cAMP molecule also seems to slow the widening of the pore, and Gucek et al. have now investigated how this happens at a molecular level. By observing individual granules of human β-cells using a special microscope, Gucek et al. could watch how different drugs affect pore widening and content release. They also saw that cAMP activated a protein called Epac2, which then recruited two other proteins – amisyn and dynamin – to the small pores. These two proteins together then closed the pore, rather than expanding it to let insulin out. Type 2 diabetes patients sometimes have high levels of amisyn in their β-cells, which could explain why they do not release enough insulin. The microscopy experiments also revealed that two common anti-diabetic drugs activate Epac2 and prevent the pores from widening, thereby counteracting their positive effect on insulin release. The combined effect is likely a shift in the balance between insulin and the locally acting small molecules. These findings suggest that two common anti-diabetic drugs activate a common mechanism that may lead to unexpected outcomes, possibly even reducing how much insulin the β-cells can release. Future studies in mice and humans will have to investigate these effects in whole organisms.
Collapse
Affiliation(s)
- Alenka Guček
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Nikhil R Gandasi
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Marit Bakke
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Sebastian Barg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
26
|
Miyazaki Y, Murayama K, Fathi I, Imura T, Yamagata Y, Watanabe K, Maeda H, Inagaki A, Igarashi Y, Miyagi S, Shima H, Igarashi K, Kamei T, Unno M, Goto M. Strategy towards tailored donor tissue-specific pancreatic islet isolation. PLoS One 2019; 14:e0216136. [PMID: 31075114 PMCID: PMC6510438 DOI: 10.1371/journal.pone.0216136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 04/15/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Optimizing the collagenase G (ColG):collagenase H (ColH) ratio is a key strategy for achieving tailored donor-tissue specific islet isolation. Collagen V (Col V) and collagen III (Col III) are crucial target matrices of ColG and ColH, respectively. We herein investigated the relevance between the expression of target matrices in pancreatic tissues and influence of ColG:ColH ratio on islet isolation outcome. METHODS Islet isolation was performed in Lewis and SD rats using different ColG:ColH ratios (5:1, 1:1 and 1:5; n = 7/group). The composition of Col III and Col V was examined using immunohistochemical staining, real-time polymerase chain reaction (PCR), Western blotting and mass spectrometry. Chain types in collagen I (Col I) were also assessed using mass spectrometry. RESULTS No beneficial effects were observed by increasing the ColG amount, irrespective of the rat strain. In contrast, the islet yield in Lewis rats was considerably increased by high amounts of ColH but decreased in SD rats, suggesting that Lewis pancreas contains more Col III than SD pancreas. Neither immunohistochemical nor real-time PCR showed correlation with isolation outcome. However, Western blotting revealed that Lewis contained considerably higher amount of Col III than SD (p = 0.10). Likewise, Col-I(α1)/Col-III(α1) and Col-I(α2)/Col-III(α1) were significantly lower in Lewis than in SD rats (p = 0.007, respectively). Furthermore, the isolation outcome was considerably correlated with the composition of homotrimeric Col I. CONCLUSIONS The Col III expression and the composition of homotrimeric Col I in pancreatic tissues determined using mass analyses appeared useful for optimizing the ColG:ColH ratio in islet isolation.
Collapse
Affiliation(s)
- Yuki Miyazaki
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kazutaka Murayama
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Ibrahim Fathi
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Youhei Yamagata
- Department of Applied Biological Chemistry, Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Kimiko Watanabe
- New Industry Creation Hatchery Center, Tohoku University, Sendai, Miyagi, Japan
| | - Hiroshi Maeda
- Department of Applied Biological Chemistry, Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yasuhiro Igarashi
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shigehito Miyagi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroki Shima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- * E-mail:
| |
Collapse
|
27
|
Matsumura M, Imura T, Inagaki A, Ogasawara H, Fukuoka K, Fathi I, Miyagi S, Ohashi K, Unno M, Kamei T, Satomi S, Goto M. A Simple and Useful Predictive Assay for Evaluating the Quality of Isolated Hepatocytes for Hepatocyte Transplantation. Sci Rep 2019; 9:6166. [PMID: 30992529 PMCID: PMC6467914 DOI: 10.1038/s41598-019-42720-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/08/2019] [Indexed: 12/13/2022] Open
Abstract
No optimal assay for assessing isolated hepatocytes before hepatocyte transplantation (HTx) has been established, therefore reliable and rapid assays are warranted. Isolated rat hepatocytes were dipped in a water bath (necrosis model), and were also cultured with Okadaic acid (apoptosis model) or vehicle, followed by cellular assessment including trypan blue exclusion (TBE) viability, ADP /ATP ratio, plating efficiency (PE), DNA quantity and ammonia elimination. Hepatocytes were transplanted into the liver of analbuminemic rats, subsequently engraftment was assessed by serum albumin and the histology of transplanted grafts. In the necrosis model, the ADP/ATP ratio was strongly and negatively correlated with the TBE (R2 = 0.559, P < 0.001). In the apoptosis model, the ADP/ATP ratio assay, PE, DNA quantification and an ammonia elimination test clearly distinguished the groups (P < 0.001, respectively). The ADP/ATP ratio, PE and DNA quantity were well-correlated and the ammonia elimination was slightly correlated with the transplant outcome. TBE could not distinguish the groups and was not correlated with the outcome. The ADP/ATP ratio assay predicted the transplant outcome. PE and DNA quantification may improve the accuracy of the retrospective (evaluations require several days) quality assessment of hepatocytes. The ADP/ATP ratio assay, alone or with a short-term metabolic assay could improve the efficiency of HTx.
Collapse
Affiliation(s)
- Muneyuki Matsumura
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, 980-0872, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, 980-0872, Sendai, Japan
| | - Hiroyuki Ogasawara
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Kengo Fukuoka
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Ibrahim Fathi
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, 980-0872, Sendai, Japan
| | - Shigehito Miyagi
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Kazuo Ohashi
- Graduate School of Pharmaceutical Sciences, Osaka University, 565-0871, Osaka, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Susumu Satomi
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, 980-0872, Sendai, Japan. .,Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, 980-0872, Sendai, Japan.
| |
Collapse
|
28
|
Brandhorst H, Johnson PR, Mönch J, Kurfürst M, Korsgren O, Brandhorst D. Comparison of Clostripain and Neutral Protease as Supplementary Enzymes for Human Islet Isolation. Cell Transplant 2018; 28:176-184. [PMID: 30419762 PMCID: PMC6362525 DOI: 10.1177/0963689718811614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Although human islet transplantation has been established as valid and safe treatment for patients with type 1 diabetes, the utilization rates of human pancreases for clinical islet transplantation are still limited and substantially determined by the quality and composition of collagenase blends. While function and integrity of collagenase has been extensively investigated, information is still lacking about the most suitable supplementary neutral proteases. The present study compared islet isolation outcome after pancreas digestion by means of collagenase used alone or supplemented with either neutral protease (NP), clostripain (CP), or both proteases. Decent amounts of islet equivalents (IEQ) were isolated using collagenase alone (3090 ± 550 IEQ/g), or in combination with NP (2340 ± 450 IEQ/g) or CP (2740 ± 280 IEQ/g). Nevertheless, the proportion of undigested tissue was higher after using collagenase alone (21.1 ± 1.1%, P < 0.05) compared with addition of NP (13.3 ± 2.2%) or CP plus NP (13.7 ± 2.6%). Likewise, the percentage of embedded islets was highest using collagenase only (13 ± 2%) and lowest adding NP plus CP (4 ± 1%, P < 0.01). The latter combination resulted in lowest post-culture overall survival (42.7 ± 3.9%), while highest survival was observed after supplementation with CP (74.5 ± 4.8%, P < 0.01). An insulin response toward glucose challenge was present in all experimental groups, but the stimulation index was significantly decreased using collagenase plus NP (2.0 ± 0.12) compared with supplementation with CP (3.16 ± 0.4, P < 0.001). This study demonstrates for the first time that it is possible to isolate significant numbers of human islets combining collagenase only with CP. The supplementation with CP is an effective means to substantially reduce NP activity, which significantly decreases survival and viability after culture. This will facilitate the manufacturing of enzyme blends with less harmful characteristics.
Collapse
Affiliation(s)
- Heide Brandhorst
- 1 Nuffield Department of Surgical Sciences, University of Oxford, UK.,2 Oxford Centre for Diabetes, Endocrinology and Metabolism, UK.,3 Department of Immunology, Genetics and Pathology, University of Uppsala, Sweden
| | - Paul R Johnson
- 1 Nuffield Department of Surgical Sciences, University of Oxford, UK.,2 Oxford Centre for Diabetes, Endocrinology and Metabolism, UK.,4 Oxford NIHR Biomedical Research Centre, UK
| | | | | | - Olle Korsgren
- 3 Department of Immunology, Genetics and Pathology, University of Uppsala, Sweden
| | - Daniel Brandhorst
- 1 Nuffield Department of Surgical Sciences, University of Oxford, UK.,2 Oxford Centre for Diabetes, Endocrinology and Metabolism, UK.,3 Department of Immunology, Genetics and Pathology, University of Uppsala, Sweden
| |
Collapse
|
29
|
Jonsson A, Yngve E, Karlsson M, Ingvast S, Skog O, Korsgren O. Protein Kinase R Is Constitutively Expressed in the Human Pancreas. J Histochem Cytochem 2018; 67:99-105. [PMID: 30265185 DOI: 10.1369/0022155418802838] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Viral infection of the insulin-producing cells in the pancreas has been proposed in the etiology of type 1 diabetes. Protein kinase R (PKR) is a cytoplasmic protein activated through phosphorylation in response to cellular stress and particularly viral infection. As PKR expression in pancreatic beta-cells has been interpreted as a viral footprint, this cross-sectional study aimed at characterizing the PKR expression in non-diabetic human pancreases. PKR expression was evaluated in pancreas tissue from 16 non-diabetic organ donors, using immunohistochemistry, qPCR, and western blot. Immunohistochemistry and western blot showed readily detectable PKR expression in the pancreatic parenchyma. The qPCR detected PKR mRNA in both endocrine and exocrine samples, with a slightly higher expression in the islets. In conclusion, PKR is constitutively expressed in both endocrine and exocrine parts of the pancreas and its expression should not be interpreted as a viral footprint in pancreatic beta cells.
Collapse
Affiliation(s)
- Alexander Jonsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Erik Yngve
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Marie Karlsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sofie Ingvast
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
30
|
Carlsson P, Espes D, Sedigh A, Rotem A, Zimerman B, Grinberg H, Goldman T, Barkai U, Avni Y, Westermark GT, Carlbom L, Ahlström H, Eriksson O, Olerud J, Korsgren O. Transplantation of macroencapsulated human islets within the bioartificial pancreas βAir to patients with type 1 diabetes mellitus. Am J Transplant 2018; 18:1735-1744. [PMID: 29288549 PMCID: PMC6055594 DOI: 10.1111/ajt.14642] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/15/2017] [Accepted: 12/21/2017] [Indexed: 01/25/2023]
Abstract
Macroencapsulation devices provide the dual possibility of immunoprotecting transplanted cells while also being retrievable, the latter bearing importance for safety in future trials with stem cell-derived cells. However, macroencapsulation entails a problem with oxygen supply to the encapsulated cells. The βAir device solves this with an incorporated refillable oxygen tank. This phase 1 study evaluated the safety and efficacy of implanting the βAir device containing allogeneic human pancreatic islets into patients with type 1 diabetes. Four patients were transplanted with 1-2 βAir devices, each containing 155 000-180 000 islet equivalents (ie, 1800-4600 islet equivalents per kg body weight), and monitored for 3-6 months, followed by the recovery of devices. Implantation of the βAir device was safe and successfully prevented immunization and rejection of the transplanted tissue. However, although beta cells survived in the device, only minute levels of circulating C-peptide were observed with no impact on metabolic control. Fibrotic tissue with immune cells was formed in capsule surroundings. Recovered devices displayed a blunted glucose-stimulated insulin response, and amyloid formation in the endocrine tissue. We conclude that the βAir device is safe and can support survival of allogeneic islets for several months, although the function of the transplanted cells was limited (Clinicaltrials.gov: NCT02064309).
Collapse
Affiliation(s)
- Per‐Ola Carlsson
- Department of Medical Cell BiologyUppsala UniversityUppsalaSweden,Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Daniel Espes
- Department of Medical Cell BiologyUppsala UniversityUppsalaSweden,Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Amir Sedigh
- Department of Surgical SciencesUppsala UniversityUppsalaSweden
| | - Avi Rotem
- BetaO2 Technologies LtdRosh HaAyinIsrael
| | | | | | | | | | - Yuval Avni
- BetaO2 Technologies LtdRosh HaAyinIsrael
| | | | - Lina Carlbom
- Department of Surgical SciencesUppsala UniversityUppsalaSweden
| | - Håkan Ahlström
- Department of Surgical SciencesUppsala UniversityUppsalaSweden,Antaros Medical ABMölndalSweden
| | - Olof Eriksson
- Department of Medicinal ChemistryUppsala UniversityUppsalaSweden
| | - Johan Olerud
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| | - Olle Korsgren
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| |
Collapse
|
31
|
Spiers RM, Cross SE, Brown HL, Bateman PA, Vaughan RH, Hughes SJ, Johnson PRV. Development of a Simple In Vitro Assay to Assess Digestion of the Extracellular Matrix of the Human Pancreas by Collagenase Enzyme Blends. Cell Transplant 2018; 27:1039-1046. [PMID: 29954221 PMCID: PMC6158553 DOI: 10.1177/0963689718779778] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Despite huge advances in the field of islet transplantation over the last two decades, current islet isolation methods remain suboptimal, with transplantable yields obtained in less than half of all pancreases processed worldwide. Successful islet isolation is dependent on the ability of collagenase-based enzyme blends to digest extracellular matrix components at the islet–exocrine interface. The limited availability of donor pancreases hinders the use of full-scale islet isolations to characterize pancreas digestion by different enzyme components or blends, or allow the influence of inter-pancreatic variability between donors to be explored. We have developed a method that allows multiple enzyme components to be tested on any one pancreas. Biopsies of 0.5 cm3 were taken from seven standard (age ≥45) and eight young (age ≤35) pancreases. Serial cryosections were treated with Serva collagenase, neutral protease (NP), or the two enzymes together at clinically relevant concentrations. Following digestion, insulin and either collagen IV or laminin-α5 were detected by immunofluorescent labeling. Protein loss at the islet–exocrine interface was semi-quantified morphometrically, with reference to a control section. Differential digestion of the two proteins based on the enzyme components used was seen, with protein digestion significantly influenced by donor age. Treatment with collagenase and NP alone was significantly more effective at digesting collagen IV in the standard donor group, as was the NP mediated digestion of laminin-α5. Collagenase alone was not capable of significantly digesting laminin-α5 in either donor group. Combining the two enzymes ameliorated the age-related differences in the digestion of both proteins. No significant differences in protein loss were detected by the method when analyzed by two independent operators, demonstrating the reproducibility of the assay. The development of this simple yet reproducible assay has implications for both enzyme batch testing and identifying inter-donor digestion variability, while utilizing small amounts of both enzyme and human tissue.
Collapse
Affiliation(s)
- Rebecca M Spiers
- 1 Islet Transplant Research Group, Nuffield Department of Surgical Sciences, University of Oxford, Level 6, John Radcliffe Hospital, Oxford, UK.,2 Oxford Center for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, UK
| | - Sarah E Cross
- 1 Islet Transplant Research Group, Nuffield Department of Surgical Sciences, University of Oxford, Level 6, John Radcliffe Hospital, Oxford, UK.,2 Oxford Center for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, UK
| | - Helen L Brown
- 1 Islet Transplant Research Group, Nuffield Department of Surgical Sciences, University of Oxford, Level 6, John Radcliffe Hospital, Oxford, UK.,2 Oxford Center for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, UK
| | - Paul A Bateman
- 1 Islet Transplant Research Group, Nuffield Department of Surgical Sciences, University of Oxford, Level 6, John Radcliffe Hospital, Oxford, UK.,2 Oxford Center for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, UK
| | - Rebecca H Vaughan
- 1 Islet Transplant Research Group, Nuffield Department of Surgical Sciences, University of Oxford, Level 6, John Radcliffe Hospital, Oxford, UK.,2 Oxford Center for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, UK
| | - Stephen J Hughes
- 1 Islet Transplant Research Group, Nuffield Department of Surgical Sciences, University of Oxford, Level 6, John Radcliffe Hospital, Oxford, UK.,2 Oxford Center for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, UK
| | - Paul R V Johnson
- 1 Islet Transplant Research Group, Nuffield Department of Surgical Sciences, University of Oxford, Level 6, John Radcliffe Hospital, Oxford, UK.,2 Oxford Center for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, UK
| |
Collapse
|
32
|
Eich T, Ståhle M, Gustafsson B, Horneland R, Lempinen M, Lundgren T, Rafael E, Tufveson G, Zur-Mühlen BV, Olerud J, Scholz H, Korsgren O. Calcium: A Crucial Potentiator for Efficient Enzyme Digestion of the Human Pancreas. Cell Transplant 2018; 27:1031-1038. [PMID: 29945463 PMCID: PMC6158545 DOI: 10.1177/0963689718779350] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Effective digestive enzymes are crucial for successful islet isolation. Supplemental proteases are essential because they synergize with collagenase for effective pancreatic digestion. The activity of these enzymes is critically dependent on the presence of Ca2+ ions at a concentration of 5–10 mM. The present study aimed to determine the Ca2+ concentration during human islet isolation and to ascertain whether the addition of supplementary Ca2+ is required to maintain an optimal Ca2+ concentration during the various phases of the islet isolation process. Methods: Human islets were isolated according to standard methods and isolation parameters. Islet quality control and the number of isolations fulfilling standard transplantation criteria were evaluated. Ca2+ was determined by using standard clinical chemistry routines. Islet isolation was performed with or without addition of supplementary Ca2+ to reach a Ca2+ of 5 mM. Results: Ca2+ concentration was markedly reduced in bicarbonate-based buffers, especially if additional bicarbonate was used to adjust the pH as recommended by the Clinical Islet Transplantation Consortium. A major reduction in Ca2+ concentration was also observed during pancreatic enzyme perfusion, digestion, and harvest. Additional Ca2+ supplementation of media used for dissolving the enzymes and during digestion, perfusion, and harvest was necessary in order to obtain the concentration recommended for optimal enzyme activity and efficient liberation of a large number of islets from the human pancreas. Conclusions: Ca2+ is to a large extent consumed during clinical islet isolation, and in the absence of supplementation, the concentration fell below that recommended for optimal enzyme activity. Ca2+ supplementation of the media used during human pancreas digestion is necessary to maintain the concentration recommended for optimal enzyme activity. Addition of Ca2+ to the enzyme blend has been implemented in the standard isolation protocols in the Nordic Network for Clinical Islet Transplantation.
Collapse
Affiliation(s)
- Torsten Eich
- 1 Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Magnus Ståhle
- 1 Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bengt Gustafsson
- 2 Department of Transplantation, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rune Horneland
- 3 Department of Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Marko Lempinen
- 4 Department of Transplantation and Liver Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Torbjörn Lundgren
- 5 Division of Transplantation Surgery, CLINTEC, Karolinska University Hospital, Stockholm, Sweden
| | - Ehab Rafael
- 6 Transplantation Unit, Department of Surgery, Skåne University Hospital, Malmö, Sweden
| | - Gunnar Tufveson
- 7 Department of Surgical Sciences, Division of Transplantation Surgery, Uppsala University Hospital, Uppsala, Sweden
| | - Bengt von Zur-Mühlen
- 7 Department of Surgical Sciences, Division of Transplantation Surgery, Uppsala University Hospital, Uppsala, Sweden
| | - Johan Olerud
- 1 Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hanne Scholz
- 3 Department of Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Olle Korsgren
- 1 Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,8 Department of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
33
|
Rajasekaran SS, Kim J, Gaboardi GC, Gromada J, Shears SB, Dos Santos KT, Nolasco EL, Ferreira SDS, Illies C, Köhler M, Gu C, Ryu SH, Martins JO, Darè E, Barker CJ, Berggren PO. Inositol hexakisphosphate kinase 1 is a metabolic sensor in pancreatic β-cells. Cell Signal 2018. [PMID: 29522819 PMCID: PMC5899964 DOI: 10.1016/j.cellsig.2018.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Diphosphoinositol pentakisphosphate (IP7) is critical for the exocytotic capacity of the pancreatic β-cell, but its regulation by the primary instigator of β-cell exocytosis, glucose, is unknown. The high Km for ATP of the IP7-generating enzymes, the inositol hexakisphosphate kinases (IP6K1 and 2) suggests that these enzymes might serve as metabolic sensors in insulin secreting β-cells and act as translators of disrupted metabolism in diabetes. We investigated this hypothesis and now show that glucose stimulation, which increases the ATP/ADP ratio, leads to an early rise in IP7 concentration in β-cells. RNAi mediated knock down of the IP6K1 isoform inhibits both glucose-mediated increase in IP7 and first phase insulin secretion, demonstrating that IP6K1 integrates glucose metabolism and insulin exocytosis. In diabetic mouse islets the deranged ATP/ADP levels under both basal and glucose-stimulated conditions are mirrored in both disrupted IP7 generation and insulin release. Thus the unique metabolic sensing properties of IP6K1 guarantees appropriate concentrations of IP7 and thereby both correct basal insulin secretion and intact first phase insulin release. In addition, our data suggest that a specific cell signaling defect, namely, inappropriate IP7 generation may be an essential convergence point integrating multiple metabolic defects into the commonly observed phenotype in diabetes. Glucose increases IP7 levels transiently through IP6K1 in pancreatic β-cells. IP6K1 decodes glucose-driven increases in ATP/ADP ratio into 1st phase insulin release. IP7 production and insulin release mirror perturbed metabolism in diabetic islets. IP6K1 acts as a β-cell metabolic sensor under normal and pathological conditions.
Collapse
Affiliation(s)
- Subu Surendran Rajasekaran
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Jaeyoon Kim
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden; School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Gian-Carlo Gaboardi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | | | - Stephen B Shears
- Signal Transduction Laboratory/Inositol Signaling Group, NIEHS, Building 101, Room F239,111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Karen Tiago Dos Santos
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden; Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eduardo Lima Nolasco
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden; Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sabrina de Souza Ferreira
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden; Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Christopher Illies
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Martin Köhler
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Chunfang Gu
- Signal Transduction Laboratory/Inositol Signaling Group, NIEHS, Building 101, Room F239,111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Sung Ho Ryu
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Joilson O Martins
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Elisabetta Darè
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Christopher J Barker
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden.
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76 Stockholm, Sweden; School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea.
| |
Collapse
|
34
|
Lundberg M, Stenwall A, Tegehall A, Korsgren O, Skog O. Expression profiles of stress-related genes in islets from donors with progressively impaired glucose metabolism. Islets 2018; 10:69-79. [PMID: 29446696 PMCID: PMC5895176 DOI: 10.1080/19382014.2018.1433980] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
It is currently unknown how the islet transcriptional pattern changes as glucose metabolism deteriorates and progresses to fulminant type 2 diabetes (T2D). In this study, we hypothesized that islets from donors with elevated HbA1c levels, but not yet diagnosed with T2D, would show signs of cell stress on a transcriptional level. Laser capture microdissection and qPCR arrays including 330 genes related to mitochondria, oxidative stress, or the unfolded protein response were used to extract and analyze islets from organ donors with HbA1c <5.5% (37 mmol/mol), elevated HbA1c (6.0-6.5% (42-48 mmol/mol)), high HbA1c (>6.5% (48 mmol/mol)) or established T2D. Principal component analysis and hierarchical clustering based on the expression of all 330 genes displayed no obvious separation of the four different donor groups, indicating that the inter-donor variations were larger than the differences between groups. However, 44 genes were differentially expressed (P < 0.05, false discovery rate <30%) between islets from donors with HbA1c <5.5% (37 mmol/mol) compared with islets from T2D subjects. Twelve genes were differentially expressed compared to control islets in both donors with established T2D and donors with elevated HbA1c (6.0-6.5% (42-48 mmol/mol)). Overexpressed genes were related mainly to the unfolded protein response, whereas underexpressed genes were related to mitochondria. Our data on transcriptional changes in human islets retrieved by LCM from high-quality biopsies, as pre-diabetes progresses to established T2D, increase our understanding on how islet stress contributes to the disease development.
Collapse
Affiliation(s)
- Marcus Lundberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- CONTACT Marcus Lundberg Uppsala University, Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, 751 85 Uppsala, Sweden
| | - Anton Stenwall
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Angie Tegehall
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
35
|
Aloy-Reverté C, Moreno-Amador JL, Nacher M, Montanya E, Semino CE. Use of RGD-Functionalized Sandwich Cultures to Promote Redifferentiation of Human Pancreatic Beta Cells AfterIn VitroExpansion. Tissue Eng Part A 2018; 24:394-406. [DOI: 10.1089/ten.tea.2016.0493] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Caterina Aloy-Reverté
- Department of Bioengineering, Tissue Engineering Laboratory, IQS School of Engineering, Barcelona, Spain
| | - José L. Moreno-Amador
- Hospital Universitari Bellvitge-Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Diabetes and Metabolic Diseases (CIBERDEM), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Montserrat Nacher
- Hospital Universitari Bellvitge-Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Diabetes and Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Eduard Montanya
- Hospital Universitari Bellvitge-Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Diabetes and Metabolic Diseases (CIBERDEM), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Carlos E. Semino
- Department of Bioengineering, Tissue Engineering Laboratory, IQS School of Engineering, Barcelona, Spain
| |
Collapse
|
36
|
Gandasi NR, Yin P, Omar-Hmeadi M, Ottosson Laakso E, Vikman P, Barg S. Glucose-Dependent Granule Docking Limits Insulin Secretion and Is Decreased in Human Type 2 Diabetes. Cell Metab 2018; 27:470-478.e4. [PMID: 29414688 DOI: 10.1016/j.cmet.2017.12.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/26/2017] [Accepted: 12/23/2017] [Indexed: 01/19/2023]
Abstract
Glucose-stimulated insulin secretion is biphasic, with a rapid first phase and a slowly developing sustained second phase; both are disturbed in type 2 diabetes (T2D). Biphasic secretion results from vastly different release probabilities of individual insulin granules, but the morphological and molecular basis for this is unclear. Here, we show that human insulin secretion and exocytosis critically depend on the availability of membrane-docked granules and that T2D is associated with a strong reduction in granule docking. Glucose accelerated granule docking, and this effect was absent in T2D. Newly docked granules only slowly acquired release competence; this was regulated by major signaling pathways, but not glucose. Gene expression analysis indicated that key proteins involved in granule docking are downregulated in T2D, and overexpression of these proteins increased granule docking. The findings establish granule docking as an important glucose-dependent step in human insulin secretion that is dysregulated in T2D.
Collapse
Affiliation(s)
- Nikhil R Gandasi
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden
| | - Peng Yin
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden
| | - Muhmmad Omar-Hmeadi
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden
| | - Emilia Ottosson Laakso
- Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University Diabetes Centre, 20502 Malmö, Sweden
| | - Petter Vikman
- Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University Diabetes Centre, 20502 Malmö, Sweden
| | - Sebastian Barg
- Department of Medical Cell Biology, Uppsala University, BMC 571, 75123 Uppsala, Sweden.
| |
Collapse
|
37
|
Eriksson O, Johnström P, Cselenyi Z, Jahan M, Selvaraju RK, Jensen-Waern M, Takano A, Sörhede Winzell M, Halldin C, Skrtic S, Korsgren O. In Vivo Visualization of β-Cells by Targeting of GPR44. Diabetes 2018; 67:182-192. [PMID: 29208633 DOI: 10.2337/db17-0764] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/17/2017] [Indexed: 11/13/2022]
Abstract
GPR44 expression has recently been described as highly β-cell selective in the human pancreas and constitutes a tentative surrogate imaging biomarker in diabetes. A radiolabeled small-molecule GPR44 antagonist, [11C]AZ12204657, was evaluated for visualization of β-cells in pigs and nonhuman primates by positron emission tomography as well as in immunodeficient mice transplanted with human islets under the kidney capsule. In vitro autoradiography of human and animal pancreatic sections from subjects without and with diabetes, in combination with insulin staining, was performed to assess β-cell selectivity of the radiotracer. Proof of principle of in vivo targeting of human islets by [11C]AZ12204657 was shown in the immunodeficient mouse transplantation model. Furthermore, [11C]AZ12204657 bound by a GPR44-mediated mechanism in pancreatic sections from humans and pigs without diabetes, but not those with diabetes. In vivo [11C]AZ12204657 bound specifically to GPR44 in pancreas and spleen and could be competed away dose-dependently in nondiabetic pigs and nonhuman primates. [11C]AZ12204657 is a first-in-class surrogate imaging biomarker for pancreatic β-cells by targeting the protein GPR44.
Collapse
MESH Headings
- Animals
- Autoradiography
- Biomarkers/metabolism
- Biopsy
- Carbon Radioisotopes
- Diabetes Mellitus, Type 1/diagnostic imaging
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 2/diagnostic imaging
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Humans
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Intestinal Elimination
- Islets of Langerhans/diagnostic imaging
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Islets of Langerhans Transplantation/diagnostic imaging
- Islets of Langerhans Transplantation/pathology
- Ligands
- Macaca fascicularis
- Magnetic Resonance Imaging
- Mice, Nude
- Phenyl Ethers/administration & dosage
- Phenyl Ethers/pharmacokinetics
- Positron Emission Tomography Computed Tomography
- Proof of Concept Study
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/metabolism
- Receptors, Prostaglandin/antagonists & inhibitors
- Receptors, Prostaglandin/metabolism
- Sus scrofa
- Tissue Distribution
- Transplantation, Heterologous
- Transplantation, Heterotopic
Collapse
Affiliation(s)
- Olof Eriksson
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Peter Johnström
- Personalised Healthcare and Biomarkers, AstraZeneca PET Science Centre, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Zsolt Cselenyi
- Personalised Healthcare and Biomarkers, AstraZeneca PET Science Centre, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Mahabuba Jahan
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Ram K Selvaraju
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Marianne Jensen-Waern
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Akihiro Takano
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | | | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Stanko Skrtic
- AstraZeneca R&D, Mölndal, Sweden
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olle Korsgren
- Division of Immunology, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
38
|
Moreno-Amador JL, Téllez N, Marin S, Aloy-Reverté C, Semino C, Nacher M, Montanya E. Epithelial to mesenchymal transition in human endocrine islet cells. PLoS One 2018; 13:e0191104. [PMID: 29360826 PMCID: PMC5779658 DOI: 10.1371/journal.pone.0191104] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 12/28/2017] [Indexed: 12/13/2022] Open
Abstract
Background β-cells undergo an epithelial to mesenchymal transition (EMT) when expanded in monolayer culture and give rise to highly proliferative mesenchymal cells that retain the potential to re-differentiate into insulin-producing cells. Objective To investigate whether EMT takes place in the endocrine non-β cells of human islets. Methodology Human islets isolated from 12 multiorgan donors were dissociated into single cells, purified by magnetic cell sorting, and cultured in monolayer. Results Co-expression of insulin and the mesenchymal marker vimentin was identified within the first passage (p1) and increased subsequently (insulin+vimentin+ 7.2±6% at p1; 43±15% at p4). The endocrine non-β-cells did also co-express vimentin (glucagon+vimentin+ 59±1.5% and 93±6%, somatostatin+vimentin+ 16±9.4% and 90±10% at p1 and p4 respectively; PP+vimentin+ 74±14% at p1; 88±12% at p2). The percentage of cells expressing only endocrine markers was progressively reduced (0.6±0.2% insulin+, 0.2±0.1% glucagon+, and 0.3±0.2% somatostatin+ cells at p4, and 0.7±0.3% PP+ cells at p2. Changes in gene expression were also indicated of EMT, with reduced expression of endocrine markers and the epithelial marker CDH-1 (p<0.01), and increased expression of mesenchymal markers (CDH-2, SNAI2, ZEB1, ZEB2, VIM, NT5E and ACTA2; p<0.05). Treatment with the EMT inhibitor A83-01 significantly reduced the percentage of co-expressing cells and preserved the expression of endocrine markers. Conclusions In adult human islets, all four endocrine islet cell types undergo EMT when islet cells are expanded in monolayer conditions. The presence of EMT in all islet endocrine cells could be relevant to design of strategies aiming to re-differentiate the expanded islet cells towards a β-cell phenotype.
Collapse
Affiliation(s)
- José Luis Moreno-Amador
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Noèlia Téllez
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Sandra Marin
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | | | - Carlos Semino
- IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain
| | - Montserrat Nacher
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Hospital Universitari Bellvitge, Hospitalet de Llobregat, Spain
- * E-mail: (MN); (ED)
| | - Eduard Montanya
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Hospital Universitari Bellvitge, Hospitalet de Llobregat, Spain
- University of Barcelona, Barcelona, Spain
- * E-mail: (MN); (ED)
| |
Collapse
|
39
|
Abstract
OBJECTIVES The aims of this study were to investigate the presence of human herpesvirus 6 (HHV6) A and B in human pancreata and to search for signs of active infection in this organ of subjects with and without type 1 diabetes (T1D). METHODS Pancreata from brain-dead organ donors with and without T1D were examined for the presence of HHV6 genomic sequences by polymerase chain reaction (PCR), transcripts by reverse transcriptase-PCR, and protein by immunohistochemistry. Quantitative PCR of isolated pancreatic islets and exocrine cell clusters was used to determine the intrapancreatic location of HHV6 DNA. RESULTS Human herpesvirus 6B genomic sequences were present in 1 of 2 donors who died of acute-onset T1D, 4 of 6 donors with long-standing T1D, and 9 of 12 nondiabetic donors. Higher copy numbers of HHV6B DNA were present in isolated islets than in exocrine tissue from the same donors. No signs of active HHV6 transcription were found. Human herpesvirus 6A was not present in any tested pancreas. CONCLUSIONS The herein presented data demonstrate, for the first time, the presence of a latent HHV6B infection in the pancreas and islets of Langerhans. Whether this virus can contribute to disease in the pancreas remains to be determined.
Collapse
|
40
|
Hals I, Ohki T, Singh R, Ma Z, Björklund A, Balasuriya C, Scholz H, Grill V. Hyperoxia reduces insulin release and induces mitochondrial dysfunction with possible implications for hyperoxic treatment of neonates. Physiol Rep 2017; 5:5/19/e13447. [PMID: 29038359 PMCID: PMC5641934 DOI: 10.14814/phy2.13447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 08/18/2017] [Indexed: 12/20/2022] Open
Abstract
We previously showed that hyperoxia in vitro negatively affects beta cells of the rat. Here, we tested for possible clinical significance as well as mitochondrial interactions by hyperoxia, using human islets (function and viability), INS‐1 832/13 cells (mitochondrial metabolism), and mouse neonates (effects in vivo). Lastly, we assessed relevant parameters in a cohort of individuals born preterm and then exposed to hyperoxia. Human islets and INS‐1 832/13 cells were exposed to 24 h of hyperoxia (90–92% oxygen). Mouse neonates were subjected to 5 days of continuous hyperoxia. Individuals born preterm were evaluated in terms of glucose homeostasis and beta cell function by HbA1c and the HOMA2 formula. In human islets, hyperoxia significantly reduced glucose‐stimulated insulin secretion by 42.2 ± 5.3% and viability assessed by MTT by 22.5 ± 5.4%. Hyperoxia down‐regulated mitochondrial complex II by 21 ± 5% and upregulated complex III by 26 ± 10.1% and complex IV by 37 ± 10.6%. Partly similar effects on mitochondrial complexes were found in hyperoxia‐exposed INS‐1 832/13 cells. Exposure to hyperoxia swiftly reduced oxygen consumption in these cells and increased mitochondrial uncoupling. Hyperoxia transiently but significantly reduced insulin release in mouse neonates. Individuals born preterm displayed higher HbA1c versus controls, as well as insulin resistance. Thus, hyperoxia exerts negative effects in vitro on human beta cells and results indicate inhibitory effects on insulin secretion in vivo in mouse neonates. Negative effects may be lessened by the demonstrated swift and profound mitochondrial adaptability. Our findings open the possibility that hyperoxia could negatively affect beta cells of preterm human neonates.
Collapse
Affiliation(s)
- Ingrid Hals
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tsuyoshi Ohki
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Rinku Singh
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Zuheng Ma
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Anneli Björklund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Chandima Balasuriya
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Endocrinology, St Olavs University Hospital, Trondheim, Norway
| | - Hanne Scholz
- Department of Transplantation Medicine, Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
| | - Valdemar Grill
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Endocrinology, St Olavs University Hospital, Trondheim, Norway
| |
Collapse
|
41
|
The Optimization of Short-Term Hepatocyte Preservation Before Transplantation. Transplant Direct 2017; 3:e176. [PMID: 28706979 PMCID: PMC5498017 DOI: 10.1097/txd.0000000000000687] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/10/2017] [Indexed: 01/15/2023] Open
Abstract
Background No optimal methods for short-term hepatocyte preservation have been established. We have recently developed a prominent oxygen-permeable bag (Tohoku Device [TD]) for pancreatic islet culture and transplantation. In this study, we investigated whether TD is also effective for hepatocyte preservation and tried to optimize other conditions. Methods Hepatocytes were preserved in the following conditions, and their outcomes were observed. First, the effectiveness of TD was investigated. Second, hepatocyte medium (HM) and organ preservation solutions with or without fetal bovine serum (FBS) were compared. Third, as supplementations, FBS and human serum albumin (HSA) were compared. Fourth, low, room and high temperature were compared. And finally, hepatocytes preserved in various conditions were transplanted into the subrenal capsule space of nonalbumin rats and engrafted areas were assessed. Results The survival rate of hepatocytes preserved in TD tended to be higher and their viability and function were maintained significantly greater than those of non-TD group. Irrespective of FBS supplementation, the survival rate of HM group was significantly higher than those of organ preservation solution group while viabilities and plating efficiency were similar among them. Although survival rates of groups without FBS were extremely low, results of HSA supplemented group were not inferior to FBS supplemented group. Hepatocytes preserved at high temperature had the worst results. The engrafted area of TD group tended to be higher than those of other groups. Conclusions TD is effective for short-term hepatocyte preservation. HSA is a useful substitute for FBS, and preserving in HM at low temperature is recommended.
Collapse
|
42
|
Lundberg M, Seiron P, Ingvast S, Korsgren O, Skog O. Insulitis in human diabetes: a histological evaluation of donor pancreases. Diabetologia 2017; 60:346-353. [PMID: 27796420 PMCID: PMC6518093 DOI: 10.1007/s00125-016-4140-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/27/2016] [Indexed: 01/05/2023]
Abstract
AIMS/HYPOTHESIS According to the consensus criteria developed for type 1 diabetes, an individual can be diagnosed with insulitis when ≥ 15 CD45+ cells are found within the parenchyma or in the islet-exocrine interface in ≥ 3 islets. The aim of this study was to determine the frequency of individuals with type 2 diabetes fulfilling these criteria with reference to non-diabetic and type 1 diabetic individuals. METHODS Insulitis was determined by examining CD45+ cells in the pancreases of 50, 13 and 44 organ donors with type 2 diabetes, type 1 diabetes and no diabetes, respectively. CD3+ cells (T cells) infiltrating the islets were evaluated in insulitic donors. In insulitic donors with type 2 diabetes, the pancreases were characterised according to the presence of CD68 (macrophages), myeloperoxidase (MPO; neutrophils), CD3, CD20 (B cells) and HLA class I hyperstained islets. In all type 2 diabetic donors, potential correlations of insulitis with dynamic glucose-stimulated insulin secretion in vitro or age, BMI, HbA1c or autoantibody positivity were examined. RESULTS Overall, 28% of the type 2 diabetic donors fulfilled the consensus criteria for insulitis developed for type 1 diabetes. Of the type 1 diabetic donors, 31% fulfilled the criteria. None of the non-diabetic donors met the criteria. Only type 1 diabetic donors had ≥ 15 CD3+ cells in ≥ 3 islets. Type 2 diabetic donors with insulitis also had a substantial number of CD45+ cells in the exocrine parenchyma. Macrophages constituted the largest fraction of CD45+ cells, followed by neutrophils and T cells. Of type 2 diabetic pancreases with insulitis, 36% contained islets that hyperstained for HLA class I. Isolated islets from type 2 diabetic donors secreted less insulin than controls, although with preserved dynamics. Insulitis in the type 2 diabetic donors did not correlate with glucose-stimulated insulin secretion, the presence of autoantibodies, BMI or HbA1c. CONCLUSIONS/INTERPRETATION The current definition of insulitis cannot be used to distinguish pancreases retrieved from individuals with type 1 diabetes from those with type 2 diabetes. On the basis of our findings, we propose a revised definition of insulitis, with a positive diagnosis when ≥ 15 CD3+ cells, not CD45+ cells, are found in ≥ 3 islets.
Collapse
Affiliation(s)
- Marcus Lundberg
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, Clinical Immunology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden.
| | - Peter Seiron
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, Clinical Immunology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| | - Sofie Ingvast
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, Clinical Immunology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, Clinical Immunology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, Clinical Immunology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| |
Collapse
|
43
|
Echovirus 6 Infects Human Exocrine and Endocrine Pancreatic Cells and Induces Pro-Inflammatory Innate Immune Response. Viruses 2017; 9:v9020025. [PMID: 28146100 PMCID: PMC5332944 DOI: 10.3390/v9020025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/16/2017] [Indexed: 12/22/2022] Open
Abstract
Human enteroviruses (HEV), especially coxsackievirus serotype B (CVB) and echovirus (E), have been associated with diseases of both the exocrine and endocrine pancreas, but so far evidence on HEV infection in human pancreas has been reported only in islets and ductal cells. This study aimed to investigate the capability of echovirus strains to infect human exocrine and endocrine pancreatic cells. Infection of explanted human islets and exocrine cells with seven field strains of E6 caused cytopathic effect, virus titer increase and production of HEV protein VP1 in both cell types. Virus particles were found in islets and acinar cells infected with E6. No cytopathic effect or infectious progeny production was observed in exocrine cells exposed to the beta cell-tropic strains of E16 and E30. Endocrine cells responded to E6, E16 and E30 by upregulating the transcription of interferon-induced with helicase C domain 1 (IF1H1), 2′-5′-oligoadenylate synthetase 1 (OAS1), interferon-β (IFN-β), chemokine (C–X–C motif) ligand 10 (CXCL10) and chemokine (C–C motif) ligand 5 (CCL5). Echovirus 6, but not E16 or E30, led to increased transcription of these genes in exocrine cells. These data demonstrate for the first time that human exocrine cells represent a target for E6 infection and suggest that certain HEV serotypes can replicate in human pancreatic exocrine cells, while the pancreatic endocrine cells are permissive to a wider range of HEV.
Collapse
|
44
|
Radenkovic M, Uvebrant K, Skog O, Sarmiento L, Avartsson J, Storm P, Vickman P, Bertilsson PA, Fex M, Korgsgren O, Cilio CM. Characterization of resident lymphocytes in human pancreatic islets. Clin Exp Immunol 2016; 187:418-427. [PMID: 27783386 DOI: 10.1111/cei.12892] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2016] [Indexed: 12/25/2022] Open
Abstract
The current view of type 1 diabetes (T1D) is that it is an immune-mediated disease where lymphocytes infiltrate the pancreatic islets, promote killing of beta cells and cause overt diabetes. Although tissue resident immune cells have been demonstrated in several organs, the composition of lymphocytes in human healthy pancreatic islets have been scarcely studied. Here we aimed to investigate the phenotype of immune cells associated with human islets of non-diabetic organ donors. A flow cytometry analysis of isolated islets from perfused pancreases (n = 38) was employed to identify alpha, beta, T, natural killer (NK) and B cells. Moreover, the expression of insulin and glucagon transcripts was evaluated by RNA sequencing. Up to 80% of the lymphocytes were CD3+ T cells with a remarkable bias towards CD8+ cells. Central memory and effector memory phenotypes dominated within the CD8+ and CD4+ T cells and most CD8+ T cells were positive for CD69 and up to 50-70% for CD103, both markers of resident memory cells. The frequency of B and NK cells was low in most islet preparations (12 and 3% of CD45+ cells, respectively), and the frequency of alpha and beta cells varied between donors and correlated clearly with insulin and glucagon mRNA expression. In conclusion, we demonstrated the predominance of canonical tissue resident memory CD8+ T cells associated with human islets. We believe that these results are important to understand more clearly the immunobiology of human islets and the disease-related phenotypes observed in diabetes.
Collapse
Affiliation(s)
- M Radenkovic
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - K Uvebrant
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - O Skog
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University Hospital, Uppsala, Sweden
| | - L Sarmiento
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - J Avartsson
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - P Storm
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - P Vickman
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - P-A Bertilsson
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - M Fex
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - O Korgsgren
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University Hospital, Uppsala, Sweden
| | - C M Cilio
- Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| |
Collapse
|
45
|
Ståhle M, Foss A, Gustafsson B, Lempinen M, Lundgren T, Rafael E, Tufveson G, Theisinger B, Brandhorst D, Korsgren O, Friberg A. Evaluation of Perfluorohexyloctane/Polydimethylsiloxane for Pancreas Preservation for Clinical Islet Isolation and Transplantation. Cell Transplant 2016; 25:2269-2276. [DOI: 10.3727/096368916x691709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This study aimed to evaluate a 50:50 mix of perfluorohexyloctane/polydimethylsiloxane 5 (F6H8S5) preservation of pancreases in a clinical setting compared with standard solutions for 1) cold ischemia time (CIT) <10 h and 2) an extended CIT >20 h. Procured clinical-grade pancreases were shipped in either F6H8S5 or in standard preservation solutions, that is, University of Wisconsin (UW) or Custodiol. F6H5S5 was preoxygenated for at least 15 min. Included clinical-grade pancreases were procured in UW or Custodiol. Upon arrival at the islet isolation laboratory, the duodenum was removed followed by rough trimming while F6H8S5 was oxygenated for 15-20 min. Trimmed pancreases were immersed into oxygenated F6H8S5 and stored at 4°C overnight followed by subsequent islet isolation. Pancreas preservation using F6H8S5 proved as effective as UW and Custadiol when used within CIT up to 10 h, in terms of both isolation outcome and islet functionality. Preservation in F6H8S5 of pancreases with extended CIT gave results similar to controls with CIT <10 h for both isolated islet functionality and isolation outcome. This study of clinically obtained pancreases indicates a clear benefit of using F6H8S5 on pancreases with extended CIT as it seems to allow extended cold ischemic time without affecting islet function and islet numbers.
Collapse
Affiliation(s)
- Magnus Ståhle
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Aksel Foss
- Department of Transplantation Surgery, Rikshospitalet-Radiumhospitalet Medical Center, Oslo, Norway
| | - Bengt Gustafsson
- Department of Transplantation, University Hospital, Gothenburg, Sweden
| | - Marko Lempinen
- Division of Transplantation, Surgical Hospital, Helsinki University, Helsinki, Finland
| | - Torbjörn Lundgren
- Division of Transplantation Surgery, CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Ehab Rafael
- Department of Nephrology and Transplantation, University Hospital, Malmö, Sweden
| | - Gunnar Tufveson
- Department of Surgical Sciences, Division of Transplantation Surgery, Uppsala University Hospital, Uppsala, Sweden
| | | | - Daniel Brandhorst
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Andrew Friberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
46
|
Hals IK, Singh R, Ma Z, Scholz H, Björklund A, Grill V. Culture at low glucose up-regulates mitochondrial function in pancreatic β cells with accompanying effects on viability. Islets 2016; 8:165-176. [PMID: 27763807 PMCID: PMC5161144 DOI: 10.1080/19382014.2016.1246637] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
We tested whether exposure of β cells at reduced glucose leads to mitochondrial adaptions and whether such adaptions modulate effects of hypoxia. Rat islets, human islets and INS-1 832/13 cells were pre-cultured short term at half standard glucose concentrations (5.5 mM for rat islets and cells, 2.75 mM for human islets) without overtly negative effects on subsequently measured function (insulin secretion and cellular insulin contents) or on viability. Culture at half standard glucose upregulated complex I and tended to upregulate complex II in islets and INS-1 cells alike. An increased release of lactate dehydrogenase that followed exposure to hypoxia was attenuated in rat islets which had been pre-cultured at half standard glucose. In INS-1 cells exposure to half standard glucose attenuated hypoxia-induced effects on several viability parameters (MTT, cell number and incremental apoptotic DNA). Thus culture at reduced glucose of pancreatic islets and clonal β cells leads to mitochondrial adaptions which possibly lessen the negative impact of hypoxia on β cell viability. These findings appear relevant in the search for optimization of pre-transplant conditions in a clinical setting.
Collapse
Affiliation(s)
- Ingrid K. Hals
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- CONTACT Ingrid K. Hals Department of Cancer Research and Molecular Medicine, NTNU, Gastrosenter, St Olavs Hospital, Prinsesse Kristinas gate 1, 7006 Trondheim, Norway
| | - Rinku Singh
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Zuheng Ma
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Hanne Scholz
- Department of Transplantation Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
| | - Anneli Björklund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Valdemar Grill
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
47
|
Halvorsen B, Santilli F, Scholz H, Sahraoui A, Gulseth HL, Wium C, Lattanzio S, Formoso G, Di Fulvio P, Otterdal K, Retterstøl K, Holven KB, Gregersen I, Stavik B, Bjerkeli V, Michelsen AE, Ueland T, Liani R, Davi G, Aukrust P. LIGHT/TNFSF14 is increased in patients with type 2 diabetes mellitus and promotes islet cell dysfunction and endothelial cell inflammation in vitro. Diabetologia 2016; 59:2134-44. [PMID: 27421726 PMCID: PMC5016561 DOI: 10.1007/s00125-016-4036-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 06/08/2016] [Indexed: 12/02/2022]
Abstract
AIMS/HYPOTHESIS Activation of inflammatory pathways is involved in the pathogenesis of type 2 diabetes mellitus. On the basis of its role in vascular inflammation and in metabolic disorders, we hypothesised that the TNF superfamily (TNFSF) member 14 (LIGHT/TNFSF14) could be involved in the pathogenesis of type 2 diabetes mellitus. METHODS Plasma levels of LIGHT were measured in two cohorts of type 2 diabetes mellitus patients (191 Italian and 40 Norwegian). Human pancreatic islet cells and arterial endothelial cells were used to explore regulation and relevant effects of LIGHT in vitro. RESULTS Our major findings were: (1) in both diabetic cohorts, plasma levels of LIGHT were significantly raised compared with sex- and age-matched healthy controls (n = 32); (2) enhanced release from activated platelets seems to be an important contributor to the raised LIGHT levels in type 2 diabetes mellitus; (3) in human pancreatic islet cells, inflammatory cytokines increased the release of LIGHT and upregulated mRNA and protein levels of the LIGHT receptors lymphotoxin β receptor (LTβR) and TNF receptor superfamily member 14 (HVEM/TNFRSF14); (4) in these cells, LIGHT attenuated the insulin release in response to high glucose at least partly via pro-apoptotic effects; and (5) in human arterial endothelial cells, glucose boosted inflammatory response to LIGHT, accompanied by an upregulation of mRNA levels of HVEM (also known as TNFRSF14) and LTβR (also known as LTBR). CONCLUSIONS/INTERPRETATION Our findings show that patients with type 2 diabetes mellitus are characterised by increased plasma LIGHT levels. Our in vitro findings suggest that LIGHT may contribute to the progression of type 2 diabetes mellitus by attenuating insulin secretion in pancreatic islet cells and by contributing to vascular inflammation.
Collapse
Affiliation(s)
- Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway.
- K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | | | - Hanne Scholz
- Institute for Surgical Research, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section for Transplantation Surgery, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Afaf Sahraoui
- Institute for Surgical Research, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section for Transplantation Surgery, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Hanne L Gulseth
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Cecilie Wium
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Lipid Clinic, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Gloria Formoso
- Center of Excellence on Aging, University of Chieti, Chieti, Italy
| | | | - Kari Otterdal
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kjetil Retterstøl
- Lipid Clinic, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kirsten B Holven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Benedicte Stavik
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
| | - Vigdis Bjerkeli
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Rossella Liani
- Center of Excellence on Aging, University of Chieti, Chieti, Italy
| | - Giovanni Davi
- Center of Excellence on Aging, University of Chieti, Chieti, Italy
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- K.G. Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
- K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
| |
Collapse
|
48
|
Eguchi H, Kawamura T, Kashiyama N, Matsuura R, Sakai R, Nakahata K, Lo PC, Asada M, Maeda A, Goto M, Toyoda M, Okuyama H, Miyagawa S. Supplemental Analysis for N-linked Sugars in Adult Pig Islets. Transplant Proc 2016; 48:1302-3. [PMID: 27320609 DOI: 10.1016/j.transproceed.2015.10.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/03/2015] [Indexed: 10/21/2022]
Abstract
The pig pancreas is considered to be one of the most suitable sources of islets for clinical xenotransplantation. However, after producing α1-3galactosyltransferase knockout pigs, most of the organs of these pigs showed less antigenicity to the human body. Wild-type adult pig islets (APIs) that originally produced negligible levels of α-Gal, different from neonatal porcine islet-like cell clusters, showed a clear antigenicity to human serum. Concerning the so-called non-Gal epitopes, many studies related to glycoproteins and glycolipids are ongoing in efforts to identify them. However, our knowledge of non-Gal glycoantigens remains incomplete. In our previous study, N-glycans were isolated from APIs, and the structures of 28 of the N-glycans were detected. In this study, to identify additional structures, further analyses were performed by liquid chromatography-mass spectrometry (LC-MS). N-glycans were isolated from APIs by the method described by O'Neil et al with minor modifications and LC-MS-based structural analyses were then performed. The detected N-glycan peaks in the LC-MS spectra were selected using the FLexAnalysis software program and the structures of the glycans were predicted using the GlyocoMod Tool. The API preparation contained 11 peaks and 16 structures were then nominated as containing N-linked sugars. Among them, 5 sulfated glycans were estimated, confirming the existence of sulfate structures in N-glycans in API. In addition, these data may supplement several N-glycan structures that contain two deoxyhexose units, such as fucose, to our previous report. The data herein will be helpful for future studies of antigenicity associated with API.
Collapse
Affiliation(s)
- H Eguchi
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - T Kawamura
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - N Kashiyama
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - R Matsuura
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - R Sakai
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - K Nakahata
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - P-C Lo
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - M Asada
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - A Maeda
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - M Goto
- International Advanced Research and Education Organization, Tohoku University, Miyagi, Japan
| | - M Toyoda
- Research Department, Sumitomo Bakelite Co. Ltd, Japan
| | - H Okuyama
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - S Miyagawa
- Department of Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
49
|
Hellström-Lindahl E, Danielsson A, Ponten F, Czernichow P, Korsgren O, Johansson L, Eriksson O. GPR44 is a pancreatic protein restricted to the human beta cell. Acta Diabetol 2016; 53:413-21. [PMID: 26467464 DOI: 10.1007/s00592-015-0811-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/21/2015] [Indexed: 10/22/2022]
Abstract
AIMS To address questions regarding onset and progression of types 1 and 2 diabetes (T1D/T2D), surrogate imaging biomarkers for beta cell function and mass are needed. Here, we assess the potential of GPR44 as a surrogate marker for beta cells, in a direct comparison with clinically used biomarker VMAT2. METHODS GPR44 surface availability was assessed by flow cytometry of human beta cells. RNA transcription levels in different pancreas compartments were evaluated. The density of GPR44 receptor in endocrine and exocrine tissues was assessed by the radiolabeled GPR44 ligand [(3)H]AZD 3825. A direct comparison with the established beta cell marker VMAT2 was performed by radiolabeled [(3)H]DTBZ. RESULTS GPR44 was available on the cell surface, and pancreatic RNA levels were restricted to the islets of Langerhans. [(3)H]AZD 3825 had nanomolar affinity for GPR44 in human islets and EndoC-βH1 beta cells, and the specific binding to human beta cells was close to 50 times higher than in exocrine preparations. The endocrine-to-exocrine binding ratio was approximately 10 times higher for [(3)H]AZD 3825 than for [(3)H]DTBZ. CONCLUSION GPR44 is a highly beta cell-specific target, which potentially offers improved imaging contrast between the human beta cell and the exocrine pancreas.
Collapse
Affiliation(s)
- Ewa Hellström-Lindahl
- Department of Medicinal Chemistry, Preclinical PET Platform, Uppsala University, 751 83, Uppsala, Sweden
| | - Angelika Danielsson
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 83, Uppsala, Sweden
- Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, 751 85, Uppsala, Sweden
| | - Fredrik Ponten
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 83, Uppsala, Sweden
| | | | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 83, Uppsala, Sweden
| | - Lars Johansson
- Department of Radiology, Oncology and Radiation Sciences, Uppsala University, 751 83, Uppsala, Sweden
| | - Olof Eriksson
- Department of Medicinal Chemistry, Preclinical PET Platform, Uppsala University, 751 83, Uppsala, Sweden.
| |
Collapse
|
50
|
Yokoi S, Murakami M, Morikawa M, Goi T, Yamaguchi A, Terada S. Sericin in the isolating solution improves the yield of islets isolated from the pancreas. Cytotechnology 2016; 68:2491-2502. [PMID: 27126060 PMCID: PMC5101320 DOI: 10.1007/s10616-016-9970-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/15/2016] [Indexed: 11/26/2022] Open
Abstract
Approximately half of the transplantable pancreatic islet tissue is lost during isolation, including the digestion and purification steps. Modifying the isolation method could increase the yield. This would enable the one donor-one recipient concept and improve the therapeutic effects of islet transplantation. This study aims to improve islet transplantation by increasing the yield of islets from the pancreas, both the number of islets and their size. Therefore, we used a sericin-containing isolating solution. Rat pancreatic islets were isolated by collagenase digestion and hand picking. We refer to islets isolated with or without sericin in the isolation solution as the sericin and control group, respectively. Volume yield, endocrine function, and islet morphology were compared between the groups. Histological distribution of sericin was evaluated by immunofluorescence staining to examine its mechanism of action in pancreatic islets. The pancreatic islet yield in the sericin group was significantly higher than that in the control group. The endocrine function of islets in the sericin group was comparable to that of islets isolated by conventional methods. Sericin adhered to the surface of isolated pancreatic islets and colocalized with E-cadherin, a cell membrane protein, which might explain the cytoprotective effects of sericin. The islet morphology tended to be better preserved in the sericin group. Sericin could prevent cytoarchitectural damage during the isolation and purification process, resulting in increased pancreatic islet yield. This suggests that sericin could contribute to islet therapy by enhancing the stability of islets.
Collapse
Affiliation(s)
- Shigehiro Yokoi
- First Department of Surgery, School of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuokashimoaizuki, eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan.
| | - Makoto Murakami
- First Department of Surgery, School of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuokashimoaizuki, eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Mitsuhiro Morikawa
- First Department of Surgery, School of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuokashimoaizuki, eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Takanori Goi
- First Department of Surgery, School of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuokashimoaizuki, eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Akio Yamaguchi
- First Department of Surgery, School of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuokashimoaizuki, eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Satoshi Terada
- Department of Applied Chemistry and Biotechnology, University of Fukui, 3-9-1 Bunkyou, Fukui, Fukui, 910-8507, Japan
| |
Collapse
|