1
|
Félix-Martínez GJ, Osorio-Londoño D, Godínez-Fernández JR. Impact of oxygen and glucose availability on the viability and connectivity of islet cells: A computational study of reconstructed avascular human islets. PLoS Comput Biol 2024; 20:e1012357. [PMID: 39137218 PMCID: PMC11343470 DOI: 10.1371/journal.pcbi.1012357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/23/2024] [Accepted: 07/22/2024] [Indexed: 08/15/2024] Open
Abstract
The experimental study and transplantation of pancreatic islets requires their isolation from the surrounding tissue, and therefore, from the vasculature. Under these conditions, avascular islets rely on the diffusion of peripheral oxygen and nutrients to comply with the requirements of islet cells while responding to changes in body glucose. As a complement to the experimental work, computational models have been widely used to estimate how avascular islets would be affected by the hypoxic conditions found both in culture and transplant sites. However, previous models have been based on simplified representations of pancreatic islets which has limited the reach of the simulations performed. Aiming to contribute with a more realistic model of avascular human islets, in this work we used architectures of human islets reconstructed from experimental data to simulate the availability of oxygen for α, β and δ-cells, emulating culture and transplant conditions at different glucose concentrations. The modeling approach proposed allowed us to quantitatively estimate how the loss of cells due to severe hypoxia would impact interactions between islet cells, ultimately segregating the islet into disconnected subnetworks. According to the simulations performed, islet encapsulation, by reducing the oxygen available within the islets, could severely compromise cell viability. Moreover, our model suggests that even without encapsulation, only microislets composed of less than 100 cells would remain viable in oxygenation conditions found in transplant sites. Overall, in this article we delineate a novel modeling methodology to simulate detailed avascular islets in experimental and transplant conditions with potential applications in the field of islet encapsulation.
Collapse
Affiliation(s)
- Gerardo J. Félix-Martínez
- Investigadoras e investigadores por México, Consejo Nacional de Humanidades, Ciencias y Tecnologías, México City, México
- Department of Electrical Engineering, Universidad Autónoma Metropolitana, Iztapalapa, México City, México
| | - Diana Osorio-Londoño
- Department of Electrical Engineering, Universidad Autónoma Metropolitana, Iztapalapa, México City, México
| | | |
Collapse
|
2
|
Thorngren J, Brboric A, Vasylovska S, Hjelmqvist D, Westermark GT, Saarimäki-Vire J, Kvist J, Balboa D, Otonkoski T, Carlsson PO, Lau J. Efficient Vascular and Neural Engraftment of Stem Cell-Derived Islets. Diabetes 2024; 73:1127-1139. [PMID: 38603470 PMCID: PMC11189832 DOI: 10.2337/db23-0123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/01/2024] [Indexed: 04/13/2024]
Abstract
Pluripotent stem cell-derived islets (SC-islets) have emerged as a new source for β-cell replacement therapy. The function of human islet transplants is hampered by excessive cell death posttransplantation; contributing factors include inflammatory reactions, insufficient revascularization, and islet amyloid formation. However, there is a gap in knowledge of the engraftment process of SC-islets. In this experimental study, we investigated the engraftment capability of SC-islets at 3 months posttransplantation and observed that cell apoptosis rates were lower but vascular density was similar in SC-islets compared with human islets. Whereas the human islet transplant vascular structures were a mixture of remnant donor endothelium and ingrowing blood vessels, the SC-islets contained ingrowing blood vessels only. Oxygenation in the SC-islet grafts was twice as high as that in the corresponding grafts of human islets, suggesting better vascular functionality. Similar to the blood vessel ingrowth, reinnervation of the SC-islets was four- to fivefold higher than that of the human islets. Both SC-islets and human islets contained amyloid at 1 and 3 months posttransplantation. We conclude that the vascular and neural engraftment of SC-islets are superior to those of human islets, but grafts of both origins develop amyloid, with potential long-term consequences. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Julia Thorngren
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Anja Brboric
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Daisy Hjelmqvist
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Jonna Saarimäki-Vire
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jouni Kvist
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Diego Balboa
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
3
|
French A, Hollister-Lock J, Sullivan BA, Stas E, Hwa AJ, Weir GC, Bonner-Weir S. Enhancement of Subcutaneous Islet Transplant Performance by Collagen 1 Gel. Cell Transplant 2024; 33:9636897241283728. [PMID: 39361612 PMCID: PMC11457190 DOI: 10.1177/09636897241283728] [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: 05/20/2024] [Revised: 07/14/2024] [Accepted: 08/14/2024] [Indexed: 10/05/2024] Open
Abstract
Human islets can be transplanted into the portal vein for T1 diabetes, and a similar procedure is being used in a clinical trial for stem cell-derived beta-like cells. Efforts have been underway to find an alternative transplant site that will foster better islet cell survival and function. Although conceptually attractive, the subcutaneous (SC) site has yielded disappointing results, in spite of some improvements resulting from more attention paid to vascularization and differentiation factors, including collagen. We developed a method to transplant rat islets in a disk of type 1 collagen gel and found improved efficacy of these transplants. Survival of islets following transplantation (tx) was determined by comparing insulin content of the graft to that of the pre-transplant islets from the same isolation. At 14 days after transplantation, grafts of the disks had more than double the recovered insulin than islets transplanted in ungelled collagen. SC grafts of disks had similar insulin content to grafts in a kidney site and in epididymal fat pads. In vivo disks underwent contraction to 10% of initial volume within 24 h but the islets remained healthy and well distributed. Whole mount imaging showed that residual donor vascular cells within the islets expanded and connected to ingrowing host blood vessels. Islets (400 rat islet equivalents (IEQ)) in the collagen disks transplanted into an SC site of NOD scid IL2R gammanull (NSG) mice reversed streptozotocin (STZ)-induced diabetes within 10 days as effectively as transplants in the kidney site. Thus, a simple change of placing islets into a gel of collagen 1 prior to transplantation allowed a prompt reversal of STZ-induced diabetes using SC site.
Collapse
Affiliation(s)
- Anna French
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Jennifer Hollister-Lock
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Brooke A. Sullivan
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Eline Stas
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Albert J. Hwa
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Gordon C. Weir
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Susan Bonner-Weir
- Harvard Medical School, Joslin Diabetes Center, Boston, MA, USA
- Section on Islet Cell & Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| |
Collapse
|
4
|
So WY, Liao Y, Liu WN, Rutter GA, Han W. Paired box 6 gene delivery preserves beta cells and improves islet transplantation efficacy. EMBO Mol Med 2023; 15:e17928. [PMID: 37933577 DOI: 10.15252/emmm.202317928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 11/08/2023] Open
Abstract
Loss of pancreatic beta cells is the central feature of all forms of diabetes. Current therapies fail to halt the declined beta cell mass. Thus, strategies to preserve beta cells are imperatively needed. In this study, we identified paired box 6 (PAX6) as a critical regulator of beta cell survival. Under diabetic conditions, the human beta cell line EndoC-βH1, db/db mouse and human islets displayed dampened insulin and incretin signalings and reduced beta cell survival, which were alleviated by PAX6 overexpression. Adeno-associated virus (AAV)-mediated PAX6 overexpression in beta cells of streptozotocin-induced diabetic mice and db/db mice led to a sustained maintenance of glucose homeostasis. AAV-PAX6 transduction in human islets reduced islet graft loss and improved glycemic control after transplantation into immunodeficient diabetic mice. Our study highlights a previously unappreciated role for PAX6 in beta cell survival and raises the possibility that ex vivo PAX6 gene transfer into islets prior to transplantation might enhance islet graft function and transplantation outcome.
Collapse
Affiliation(s)
- Wing Yan So
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yilie Liao
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, 528400, China
- Center for Neurometabolism and Regenerative Medicine, Bioland Laboratories, Guangzhou, Guangdong, 510530, China
| | - Wai Nam Liu
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Guy A Rutter
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore, Singapore
| | - Weiping Han
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
5
|
Einstein SA, Steyn LV, Weegman BP, Suszynski TM, Sambanis A, O'Brien TD, Avgoustiniatos ES, Firpo MT, Graham ML, Janecek J, Eberly LE, Garwood M, Putnam CW, Papas KK. Hypoxia within subcutaneously implanted macroencapsulation devices limits the viability and functionality of densely loaded islets. FRONTIERS IN TRANSPLANTATION 2023; 2:1257029. [PMID: 38993891 PMCID: PMC11235299 DOI: 10.3389/frtra.2023.1257029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/20/2023] [Indexed: 07/13/2024]
Abstract
Introduction Subcutaneous macroencapsulation devices circumvent disadvantages of intraportal islet therapy. However, a curative dose of islets within reasonably sized devices requires dense cell packing. We measured internal PO2 of implanted devices, mathematically modeled oxygen availability within devices and tested the predictions with implanted devices containing densely packed human islets. Methods Partial pressure of oxygen (PO2) within implanted empty devices was measured by noninvasive 19F-MRS. A mathematical model was constructed, predicting internal PO2, viability and functionality of densely packed islets as a function of external PO2. Finally, viability was measured by oxygen consumption rate (OCR) in day 7 explants loaded at various islet densities. Results In empty devices, PO2 was 12 mmHg or lower, despite successful external vascularization. Devices loaded with human islets implanted for 7 days, then explanted and assessed by OCR confirmed trends proffered by the model but viability was substantially lower than predicted. Co-localization of insulin and caspase-3 immunostaining suggested that apoptosis contributed to loss of beta cells. Discussion Measured PO2 within empty devices declined during the first few days post-transplant then modestly increased with neovascularization around the device. Viability of islets is inversely related to islet density within devices.
Collapse
Affiliation(s)
- Samuel A Einstein
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States
- Department of Radiology, The Pennsylvania State University, Hershey, PA, United States
| | - Leah V Steyn
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | - Bradley P Weegman
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States
- Sylvatica Biotech Inc., North Charleston, SC, United States
| | - Thomas M Suszynski
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Athanassios Sambanis
- Department of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Timothy D O'Brien
- Veterinary Population Medicine Department, University of Minnesota, Saint Paul, MN, United States
- Department of Medicine, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | | | - Meri T Firpo
- Department of Medicine, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | - Melanie L Graham
- Veterinary Population Medicine Department, University of Minnesota, Saint Paul, MN, United States
- Department of Surgery, Preclinical Research Center, University of Minnesota, Saint Paul, MN, United States
| | - Jody Janecek
- Department of Surgery, Preclinical Research Center, University of Minnesota, Saint Paul, MN, United States
| | - Lynn E Eberly
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, United States
| | - Michael Garwood
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States
| | - Charles W Putnam
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | - Klearchos K Papas
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
6
|
Wang X, Ji L, Wang J, Liu C. Matrix stiffness regulates osteoclast fate through integrin-dependent mechanotransduction. Bioact Mater 2023; 27:138-153. [PMID: 37064801 PMCID: PMC10090259 DOI: 10.1016/j.bioactmat.2023.03.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 04/01/2023] Open
Abstract
Osteoclasts ubiquitously participate in bone homeostasis, and their aberration leads to bone diseases, such as osteoporosis. Current clinical strategies by biochemical signaling molecules often perturb innate bone metabolism owing to the uncontrolled management of osteoclasts. Thus, an alternative strategy of precise regulation for osteoclast differentiation is urgently needed. To this end, this study proposed an assumption that mechanic stimulation might be a potential strategy. Here, a hydrogel was created to imitate the physiological bone microenvironment, with stiffnesses ranging from 2.43kPa to 68.2kPa. The impact of matrix stiffness on osteoclast behaviors was thoroughly investigated. Results showed that matrix stiffness could be harnessed for directing osteoclast fate in vitro and in vivo. In particular, increased matrix stiffness inhibited the integrin β3-responsive RhoA-ROCK2-YAP-related mechanotransduction and promoted osteoclastogenesis. Notably, preosteoclast development is facilitated by medium-stiffness hydrogel (M-gel) possessing the same stiffness as vessel ranging from 17.5 kPa to 44.6 kPa by partial suppression of mechanotransduction, which subsequently encouraged revascularization and bone regeneration in mice with bone defects. Our works provide an innovative approach for finely regulating osteoclast differentiation by selecting the optimum matrix stiffness and enable us further to develop a matrix stiffness-based strategy for bone tissue engineering.
Collapse
Affiliation(s)
- Xiaogang Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Luli Ji
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jing Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Corresponding author.
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Corresponding author.
| |
Collapse
|
7
|
Cell Replacement Therapy for Type 1 Diabetes Patients: Potential Mechanisms Leading to Stem-Cell-Derived Pancreatic β-Cell Loss upon Transplant. Cells 2023; 12:cells12050698. [PMID: 36899834 PMCID: PMC10000642 DOI: 10.3390/cells12050698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Cell replacement therapy using stem-cell-derived insulin-producing β-like cells (sBCs) has been proposed as a practical cure for patients with type one diabetes (T1D). sBCs can correct diabetes in preclinical animal models, demonstrating the promise of this stem cell-based approach. However, in vivo studies have demonstrated that most sBCs, similarly to cadaveric human islets, are lost upon transplantation due to ischemia and other unknown mechanisms. Hence, there is a critical knowledge gap in the current field concerning the fate of sBCs upon engraftment. Here we review, discuss effects, and propose additional potential mechanisms that could contribute toward β-cell loss in vivo. We summarize and highlight some of the literature on phenotypic loss in β-cells under both steady, stressed, and diseased diabetic conditions. Specifically, we focus on β-cell death, dedifferentiation into progenitors, trans-differentiation into other hormone-expressing cells, and/or interconversion into less functional β-cell subtypes as potential mechanisms. While current cell replacement therapy efforts employing sBCs carry great promise as an abundant cell source, addressing the somewhat neglected aspect of β-cell loss in vivo will further accelerate sBC transplantation as a promising therapeutic modality that could significantly enhance the life quality of T1D patients.
Collapse
|
8
|
Berney T, Wassmer CH, Lebreton F, Bellofatto K, Fonseca LM, Bignard J, Hanna R, Peloso A, Berishvili E. From islet of Langerhans transplantation to the bioartificial pancreas. Presse Med 2022; 51:104139. [PMID: 36202182 DOI: 10.1016/j.lpm.2022.104139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
Type 1 diabetes is a disease resulting from autoimmune destruction of the insulin-producing beta cells in the pancreas. When type 1 diabetes develops into severe secondary complications, in particular end-stage nephropathy, or life-threatening severe hypoglycemia, the best therapeutic approach is pancreas transplantation, or more recently transplantation of the pancreatic islets of Langerhans. Islet transplantation is a cell therapy procedure, that is minimally invasive and has a low morbidity, but does not display the same rate of functional success as the more invasive pancreas transplantation because of suboptimal engraftment and survival. Another issue is that pancreas or islet transplantation (collectively known as beta cell replacement therapy) is limited by the shortage of organ donors and by the need for lifelong immunosuppression to prevent immune rejection and recurrence of autoimmunity. A bioartificial pancreas is a construct made of functional, insulin-producing tissue, embedded in an anti-inflammatory, immunomodulatory microenvironment and encapsulated in a perm-selective membrane allowing glucose sensing and insulin release, but isolating from attacks by cells of the immune system. A successful bioartificial pancreas would address the issues of engraftment, survival and rejection. Inclusion of unlimited sources of insulin-producing cells, such as xenogeneic porcine islets or stem cell-derived beta cells would further solve the problem of organ shortage. This article reviews the current status of clinical islet transplantation, the strategies aiming at developing a bioartificial pancreas, the clinical trials conducted in the field and the perspectives for further progress.
Collapse
Affiliation(s)
- Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland; Division of Transplantation, Department of Surgery, University of Geneva Hospitals, Geneva, Switzerland; Faculty Diabetes Center, University of Geneva School of Medicine, Geneva, Switzerland; Department of Surgery, School of Medicine and Natural Sciences, Ilia State University, Tbilisi, Georgia
| | - Charles H Wassmer
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland; Division of Transplantation, Department of Surgery, University of Geneva Hospitals, Geneva, Switzerland
| | - Fanny Lebreton
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland
| | - Kevin Bellofatto
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland
| | - Laura Mar Fonseca
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland; Division of Transplantation, Department of Surgery, University of Geneva Hospitals, Geneva, Switzerland
| | - Juliette Bignard
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland
| | - Reine Hanna
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland
| | - Andrea Peloso
- Division of Transplantation, Department of Surgery, University of Geneva Hospitals, Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva School of Medicine, Geneva, Switzerland; Faculty Diabetes Center, University of Geneva School of Medicine, Geneva, Switzerland; Institute of Medical and Public Health Research, Ilia State University, Tbilisi, Georgia.
| |
Collapse
|
9
|
Koehler N, Buhler L, Egger B, Gonelle-Gispert C. Multipotent Mesenchymal Stromal Cells Interact and Support Islet of Langerhans Viability and Function. Front Endocrinol (Lausanne) 2022; 13:822191. [PMID: 35222280 PMCID: PMC8864309 DOI: 10.3389/fendo.2022.822191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
Type 1 diabetes (T1D) is a widespread disease, affecting approximately 41.5 million people worldwide. It is generally treated with exogenous insulin, maintaining physiological blood glucose levels but also leading to long-term therapeutic complications. Pancreatic islet cell transplantation offers a potential alternative treatment to insulin injections. Shortage of human organ donors has raised the interest for porcine islet xenotransplantation. Neonatal porcine islets are highly available, can proliferate and mature in vitro as well as after transplantation in vivo. Despite promising preclinical results, delayed insulin secretion caused by immaturity and immunogenicity of the neonatal porcine islets remains a challenge for their clinical application. Multipotent mesenchymal stromal cells (MSCs) are known to have pro-angiogenic, anti-inflammatory and immunomodulatory effects. The current state of research emphasizes the great potential of co-culture and co-transplantation of islet cells with MSCs. Studies have shown enhanced islet proliferation and maturation, insulin secretion and graft survival, resulting in an improved graft outcome. This review summarizes the immunomodulatory and anti-inflammatory properties of MSC in the context of islet transplantation.
Collapse
Affiliation(s)
- Naomi Koehler
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Leo Buhler
- Department of Surgery, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | - Bernhard Egger
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Department of Surgery, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | - Carmen Gonelle-Gispert
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- *Correspondence: Carmen Gonelle-Gispert,
| |
Collapse
|
10
|
Arefanian H, Ramji Q, Gupta N, Spigelman AF, Grynoch D, MacDonald PE, Mueller TF, Gazda LS, Rajotte RV, Rayat GR. Yield, cell composition, and function of islets isolated from different ages of neonatal pigs. Front Endocrinol (Lausanne) 2022; 13:1032906. [PMID: 36619563 PMCID: PMC9811407 DOI: 10.3389/fendo.2022.1032906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/01/2022] [Indexed: 12/24/2022] Open
Abstract
The yield, cell composition, and function of islets isolated from various ages of neonatal pigs were characterized using in vitro and in vivo experimental models. Islets from 7- and 10-day-old pigs showed significantly better function both in vitro and in vivo compared to islets from 3- and 5-day-old pigs however, the islet yield from 10-day-old pigs were significantly less than those obtained from the other pigs. Since islets from 3-day-old pigs were used in our previous studies and islets from 7-day-old pigs reversed diabetes more efficiently than islets from other groups, we further evaluated the function of these islets post-transplantation. B6 rag-/- mouse recipients of various numbers of islets from 7-day-old pigs achieved normoglycemia faster and showed significantly improved response to glucose challenge compared to the recipients of the same numbers of islets from 3-day-old pigs. These results are in line with the findings that islets from 7-day-old pigs showed reduced voltage-dependent K+ (Kv) channel activity and their ability to recover from post-hypoxia/reoxygenation stress. Despite more resident immune cells and immunogenic characteristics detected in islets from 7-day-old pigs compared to islets from 3-day-old pigs, the combination of anti-LFA-1 and anti-CD154 monoclonal antibodies are equally effective at preventing the rejection of islets from both age groups of pigs. Collectively, these results suggest that islets from various ages of neonatal pigs vary in yield, cellular composition, and function. Such parameters may be considered when defining the optimal pancreas donor for islet xenotransplantation studies.
Collapse
Affiliation(s)
- Hossein Arefanian
- Alberta Diabetes Institute, Ray Rajotte Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Qahir Ramji
- Alberta Diabetes Institute, Ray Rajotte Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Nancy Gupta
- Alberta Diabetes Institute, Ray Rajotte Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Aliya F. Spigelman
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Donald Grynoch
- Alberta Precision Labs, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Patrick E. MacDonald
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Thomas F. Mueller
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | | | - Ray V. Rajotte
- Alberta Diabetes Institute, Ray Rajotte Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Gina R. Rayat, ; Ray V. Rajotte,
| | - Gina R. Rayat
- Alberta Diabetes Institute, Ray Rajotte Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Gina R. Rayat, ; Ray V. Rajotte,
| |
Collapse
|
11
|
Aghazadeh Y, Poon F, Sarangi F, Wong FTM, Khan ST, Sun X, Hatkar R, Cox BJ, Nunes SS, Nostro MC. Microvessels support engraftment and functionality of human islets and hESC-derived pancreatic progenitors in diabetes models. Cell Stem Cell 2021; 28:1936-1949.e8. [PMID: 34480863 DOI: 10.1016/j.stem.2021.08.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 04/27/2021] [Accepted: 08/04/2021] [Indexed: 12/19/2022]
Abstract
Islet transplantation is a promising treatment for type 1 diabetes (T1D), yet the low donor pool, poor islet engraftment, and life-long immunosuppression prevent it from becoming the standard of care. Human embryonic stem cell (hESC)-derived pancreatic cells could eliminate donor shortages, but interventions to improve graft survival are needed. Here, we enhanced subcutaneous engraftment by employing a unique vascularization strategy based on ready-made microvessels (MVs) isolated from the adipose tissue. This resulted in improved cell survival and effective glucose response of both human islets and hESC-derived pancreatic cells, which ameliorated preexisting diabetes in three mouse models of T1D.
Collapse
Affiliation(s)
- Yasaman Aghazadeh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Frankie Poon
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Deparment of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Farida Sarangi
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Frances T M Wong
- Deparment of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Safwat T Khan
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Xuetao Sun
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Rupal Hatkar
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Brian J Cox
- Deparment of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON M5G 1E2, Canada
| | - Sara S Nunes
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Heart & Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON M5S 3H2, Canada.
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Deparment of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
12
|
Sabbah S, Liew A, Brooks AM, Kundu R, Reading JL, Flatt A, Counter C, Choudhary P, Forbes S, Rosenthal MJ, Rutter MK, Cairns S, Johnson P, Casey J, Peakman M, Shaw JA, Tree TIM. Autoreactive T cell profiles are altered following allogeneic islet transplantation with alemtuzumab induction and re-emerging phenotype is associated with graft function. Am J Transplant 2021; 21:1027-1038. [PMID: 32865886 DOI: 10.1111/ajt.16285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/15/2020] [Accepted: 08/10/2020] [Indexed: 01/25/2023]
Abstract
Islet transplantation is an effective therapy for life-threatening hypoglycemia, but graft function gradually declines over time in many recipients. We characterized islet-specific T cells in recipients within an islet transplant program favoring alemtuzumab (ATZ) lymphodepleting induction and examined associations with graft function. Fifty-eight recipients were studied: 23 pretransplant and 40 posttransplant (including 5 with pretransplant phenotyping). The proportion with islet-specific T cell responses was not significantly different over time (pre-Tx: 59%; 1-6 m posttransplant: 38%; 7-12 m: 44%; 13-24 m: 47%; and >24 m: 45%). However, phenotype shifted significantly, with IFN-γ-dominated response in the pretransplant group replaced by IL-10-dominated response in the 1-6 m posttransplant group, reverting to predominantly IFN-γ-oriented response in the >24 m group. Clustering analysis of posttransplant responses revealed two main agglomerations, characterized by IFN-γ and IL-10 phenotypes, respectively. IL-10-oriented posttransplant response was associated with relatively low graft function. Recipients within the IL-10+ cluster had a significant decline in C-peptide levels in the period preceding the IL-10 response, but stable graft function following the response. In contrast, an IFN-γ response was associated with subsequently decreased C-peptide. Islet transplantation favoring ATZ induction is associated with an initial altered islet-specific T cell phenotype but reversion toward pretransplant profiles over time. Posttransplant autoreactive T cell phenotype may be a predictor of subsequent graft function.
Collapse
Affiliation(s)
- Shereen Sabbah
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King's College London, London, UK.,NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Aaron Liew
- Newcastle University Translational and Clinical Research Institute, Newcastle, UK
| | - Augustin M Brooks
- Newcastle University Translational and Clinical Research Institute, Newcastle, UK
| | - Rhiannon Kundu
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King's College London, London, UK.,NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - James L Reading
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King's College London, London, UK.,NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Anneliese Flatt
- Newcastle University Translational and Clinical Research Institute, Newcastle, UK
| | - Claire Counter
- Organ Donation and Transplantation, NHS Blood and Transplant, Bristol, UK
| | - Pratik Choudhary
- Diabetes Research Group, Guy's, King's and St. Thomas' School of Medicine, King's College London, London, UK
| | - Shareen Forbes
- Edinburgh Transplant Centre and Endocrinology Unit, University of Edinburgh, Edinburgh, UK
| | | | - Martin K Rutter
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester, UK.,Diabetes, Endocrinology and Metabolism Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Stephanie Cairns
- Clinical Immunology Department, Scottish National Blood Transfusion Service, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Paul Johnson
- Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - John Casey
- Edinburgh Transplant Centre and Endocrinology Unit, University of Edinburgh, Edinburgh, UK
| | - Mark Peakman
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King's College London, London, UK.,NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - James A Shaw
- Newcastle University Translational and Clinical Research Institute, Newcastle, UK.,Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Timothy I M Tree
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King's College London, London, UK.,NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
13
|
Bourgeois S, Sawatani T, Van Mulders A, De Leu N, Heremans Y, Heimberg H, Cnop M, Staels W. Towards a Functional Cure for Diabetes Using Stem Cell-Derived Beta Cells: Are We There Yet? Cells 2021; 10:cells10010191. [PMID: 33477961 PMCID: PMC7835995 DOI: 10.3390/cells10010191] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is a pandemic metabolic disorder that results from either the autoimmune destruction or the dysfunction of insulin-producing pancreatic beta cells. A promising cure is beta cell replacement through the transplantation of islets of Langerhans. However, donor shortage hinders the widespread implementation of this therapy. Human pluripotent stem cells, including embryonic stem cells and induced pluripotent stem cells, represent an attractive alternative beta cell source for transplantation. Although major advances over the past two decades have led to the generation of stem cell-derived beta-like cells that share many features with genuine beta cells, producing fully mature beta cells remains challenging. Here, we review the current status of beta cell differentiation protocols and highlight specific challenges that are associated with producing mature beta cells. We address the challenges and opportunities that are offered by monogenic forms of diabetes. Finally, we discuss the remaining hurdles for clinical application of stem cell-derived beta cells and the status of ongoing clinical trials.
Collapse
Affiliation(s)
- Stephanie Bourgeois
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
| | - Toshiaki Sawatani
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, 1070 Brussels, Belgium; (T.S.); (M.C.)
| | - Annelore Van Mulders
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
| | - Nico De Leu
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
- Department of Endocrinology, University Hospital Brussels, 1090 Brussels, Belgium
- Department of Endocrinology, ASZ Aalst, 9300 Aalst, Belgium
| | - Yves Heremans
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
| | - Harry Heimberg
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
| | - Miriam Cnop
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, 1070 Brussels, Belgium; (T.S.); (M.C.)
- Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Willem Staels
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
- Service of Pediatric Endocrinology, Department of Pediatrics, KidZ Health Castle, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium
- Correspondence: ; Tel.: +32-0-24774473
| |
Collapse
|
14
|
Weitz J, Menegaz D, Caicedo A. Deciphering the Complex Communication Networks That Orchestrate Pancreatic Islet Function. Diabetes 2021; 70:17-26. [PMID: 33355306 PMCID: PMC7881851 DOI: 10.2337/dbi19-0033] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022]
Abstract
Pancreatic islets are clusters of hormone-secreting endocrine cells that rely on intricate cell-cell communication mechanisms for proper function. The importance of multicellular cooperation in islet cell physiology was first noted nearly 30 years ago in seminal studies showing that hormone secretion from endocrine cell types is diminished when these cells are dispersed. These studies showed that reestablishing cellular contacts in so-called pseudoislets caused endocrine cells to regain hormone secretory function. This not only demonstrated that cooperation between islet cells is highly synergistic but also gave birth to the field of pancreatic islet organoids. Here we review recent advances related to the mechanisms of islet cell cross talk. We first describe new developments that revise current notions about purinergic and GABA signaling in islets. Then we comment on novel multicellular imaging studies that are revealing emergent properties of islet communication networks. We finish by highlighting and discussing recent synthetic approaches that use islet organoids of varied cellular composition to interrogate intraislet signaling mechanisms. This reverse engineering of islets not only will shed light on the mechanisms of intraislet signaling and define communication networks but also may guide efforts aimed at restoring islet function and β-cell mass in diabetes.
Collapse
Affiliation(s)
- Jonathan Weitz
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL
| | - Danusa Menegaz
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL
| | - Alejandro Caicedo
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Program in Neuroscience, University of Miami Leonard M. Miller School of Medicine, Miami, FL
| |
Collapse
|
15
|
Abstract
OBJECTIVES Type 2 diabetes (T2D) is histopathologically characterized by islet amyloid and is closely connected with vascular complications. Here, we explore the presence of pancreatic angiopathy (PA) associated with islet amyloid and T2D. METHODS From a total of 172 autopsy cases who had a history of T2D diagnosis, we randomly selected 30 T2D autopsy cases with islet amyloid (DA+) in comparison with islet amyloid-free (DA-) 30 T2D cases and 60 nondiabetic (ND) controls. Amyloid deposits and PA including atherosclerosis of pancreatic interlobar arteries, arterial calcification, atheroembolism, hyaline arteriosclerosis of small arterioles, and islet capillary density were detected in all groups. RESULTS Pancreatic angiopathy was found in 91.7% of patients with T2D and in 68.3% of ND controls (P < 0.01). Furthermore, 100% of DA+ patients and 83.3% of DA- subjects showed PA. The intraislet capillary density was significantly lower in DA+ subjects than DA- subjects (mean [standard deviation], DA+: 205 [82] count/mm; DA-: 344 [76] count/mm; ND: 291 [94] count/mm; P < 0.01). Finally, interlobar arteriosclerosis (R = 0.603, P < 0.01) was linearly correlated with the severity of islet amyloid deposits. CONCLUSIONS Pancreatic angiopathy might be both a cause and a consequence of islet amyloid and T2D.
Collapse
|
16
|
Wassmer CH, Lebreton F, Bellofatto K, Bosco D, Berney T, Berishvili E. Generation of insulin-secreting organoids: a step toward engineering and transplanting the bioartificial pancreas. Transpl Int 2020; 33:1577-1588. [PMID: 32852858 PMCID: PMC7756715 DOI: 10.1111/tri.13721] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/06/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
Diabetes is a major health issue of increasing prevalence. ß‐cell replacement, by pancreas or islet transplantation, is the only long‐term curative option for patients with insulin‐dependent diabetes. Despite good functional results, pancreas transplantation remains a major surgery with potentially severe complications. Islet transplantation is a minimally invasive alternative that can widen the indications in view of its lower morbidity. However, the islet isolation procedure disrupts their vasculature and connection to the surrounding extracellular matrix, exposing them to ischemia and anoikis. Implanted islets are also the target of innate and adaptive immune attacks, thus preventing robust engraftment and prolonged full function. Generation of organoids, defined as functional 3D structures assembled with cell types from different sources, is a strategy increasingly used in regenerative medicine for tissue replacement or repair, in a variety of inflammatory or degenerative disorders. Applied to ß‐cell replacement, it offers the possibility to control the size and composition of islet‐like structures (pseudo‐islets), and to include cells with anti‐inflammatory or immunomodulatory properties. In this review, we will present approaches to generate islet cell organoids and discuss how these strategies can be applied to the generation of a bioartificial pancreas for the treatment of type 1 diabetes.
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.,Division of Transplantation, Department of Surgery, University of Geneva Hospitals, 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
| | - 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
| | - 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.,Division of Transplantation, Department of Surgery, University of Geneva Hospitals, 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 and Public Health Research, Ilia State University, Tbilisi, Georgia
| |
Collapse
|
17
|
Gurlin RE, Giraldo JA, Latres E. 3D Bioprinting and Translation of Beta Cell Replacement Therapies for Type 1 Diabetes. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:238-252. [PMID: 32907514 DOI: 10.1089/ten.teb.2020.0192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disorder in which the body's own immune system selectively attacks beta cells within pancreatic islets resulting in insufficient insulin production and loss of the ability to regulate blood glucose (BG) levels. Currently, the standard of care consists of BG level monitoring and insulin administration, which are essential to avoid the consequences of dysglycemia and long-term complications. Although recent advances in continuous glucose monitoring and automated insulin delivery systems have resulted in improved clinical outcomes for users, nearly 80% of people with T1D fail to achieve their target hemoglobin A1c (HbA1c) levels defined by the American Diabetes Association. Intraportal islet transplantation into immunosuppressed individuals with T1D suffering from impaired awareness of hypoglycemia has resulted in lower HbA1c, elimination of severe hypoglycemic events, and insulin independence, demonstrating the unique potential of beta cell replacement therapy (BCRT) in providing optimal glycemic control and a functional cure for T1D. BCRTs need to maximize cell engraftment, long-term survival, and function in the absence of immunosuppression to provide meaningful clinical outcomes to all people living with T1D. One innovative technology that could enable widespread translation of this approach into the clinic is three-dimensional (3D) bioprinting. Herein, we review how bioprinting could facilitate translation of BCRTs as well as the current and forthcoming techniques used for bioprinting of a BCRT product. We discuss the strengths and weaknesses of 3D bioprinting in this context in addition to the road ahead for the development of BCRTs. Impact statement Significant research developments in beta cell replacement therapies show its promise in providing a functional cure for type 1 diabetes (T1D); yet, their widespread clinical use has been difficult to achieve. This review provides a brief overview of the requirements for a beta cell replacement product followed by a discussion on both the promise and limitations of three-dimensional bioprinting in facilitating the fabrication of such products to enable translation into the clinic. Advancements in this area could be a key component to unlocking the safety and effectiveness of beta cell therapy for T1D.
Collapse
Affiliation(s)
- Rachel E Gurlin
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | | | | |
Collapse
|
18
|
Figueiredo H, Figueroa ALC, Garcia A, Fernandez-Ruiz R, Broca C, Wojtusciszyn A, Malpique R, Gasa R, Gomis R. Targeting pancreatic islet PTP1B improves islet graft revascularization and transplant outcomes. Sci Transl Med 2020; 11:11/497/eaar6294. [PMID: 31217339 DOI: 10.1126/scitranslmed.aar6294] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/16/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
Deficient vascularization is a major driver of early islet graft loss and one of the primary reasons for the failure of islet transplantation as a viable treatment for type 1 diabetes. This study identifies the protein tyrosine phosphatase 1B (PTP1B) as a potential modulator of islet graft revascularization. We demonstrate that grafts of pancreatic islets lacking PTP1B exhibit increased revascularization, which is accompanied by improved graft survival and function, and recovery of normoglycemia and glucose tolerance in diabetic mice transplanted with PTP1B-deficient islets. Mechanistically, we show that the absence of PTP1B leads to activation of hypoxia-inducible factor 1α-independent peroxisome proliferator-activated receptor γ coactivator 1α/estrogen-related receptor α signaling and enhanced expression and production of vascular endothelial growth factor A (VEGF-A) by β cells. These observations were reproduced in human islets. Together, these findings reveal that PTP1B regulates islet VEGF-A production and suggest that this phosphatase could be targeted to improve islet transplantation outcomes.
Collapse
Affiliation(s)
- Hugo Figueiredo
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,University of Barcelona, 08036 Barcelona, Spain.,Escuela de Medicina y Ciencias de la Salud, Dept. Medicina Cardiovascular y Metabolómica, Tecnológico de Monterrey, 66278 San Pedro Garza García, Nuevo León, Mexico
| | - Ana Lucia C Figueroa
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,University of Barcelona, 08036 Barcelona, Spain
| | - Ainhoa Garcia
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Rebeca Fernandez-Ruiz
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Christophe Broca
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, 34295 Montpellier, France
| | - Anne Wojtusciszyn
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, 34295 Montpellier, France.,Department of Endocrinology, Diabetes and Nutrition, University Hospital of Montpellier, Lapeyronie Hospital, 34295 Montpellier, France.,Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Rita Malpique
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Rosa Gasa
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Ramon Gomis
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain. .,University of Barcelona, 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.,Universitat Oberta de Catalunya (UOC), 08018 Barcelona, Spain.,Department of Endocrinology and Nutrition, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
19
|
Bikmulina PY, Kosheleva NV, Shpichka AI, Efremov YM, Yusupov VI, Timashev PS, Rochev YA. Beyond 2D: effects of photobiomodulation in 3D tissue-like systems. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:1-16. [PMID: 32351077 PMCID: PMC7189416 DOI: 10.1117/1.jbo.25.4.048001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 04/09/2020] [Indexed: 05/02/2023]
Abstract
SIGNIFICANCE Currently, various scaffolds with immobilized cells are widely used in tissue engineering and regenerative medicine. However, the physiological activity and cell viability in such constructs might be impaired due to a lack of oxygen and nutrients. Photobiomodulation (PBM) is a promising method of preconditioning cells to increase their metabolic activity and to activate proliferation or differentiation. AIM Investigation of the potential of PBM for stimulation of cell activities in hydrogels. APPROACH Mesenchymal stromal cells (MSCs) isolated from human gingival mucosa were encapsulated in modified fibrin hydrogels with different thicknesses and concentrations. Constructs with cells were subjected to a single-time exposure to red (630 nm) and near-infrared (IR) (840 nm) low-intensity irradiation. After 3 days of cultivation, the viability and physiological activity of the cells were analyzed using confocal microscopy and a set of classical tests for cytotoxicity. RESULTS The cell viability in fibrin hydrogels depended both on the thickness of the hydrogels and the concentration of gel-forming proteins. The PBM was able to improve cell viability in hydrogels. The most pronounced effect was achieved with near-IR irradiation at the 840-nm wavelength. CONCLUSIONS PBM using near-IR light can be applied for stimulation of MSCs metabolism and proliferation in hydrogel-based constructs with thicknesses up to 3 mm.
Collapse
Affiliation(s)
- Polina Y. Bikmulina
- Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, Moscow, Russia
| | - Nastasia V. Kosheleva
- Lomonosov Moscow State University, Faculty of Biology, Moscow, Russia
- FSBSI “Institute of General Pathology and Pathophysiology,” Moscow, Russia
- FSBEI FPE “Russian Medical Academy of Continuous Professional Education,” Ministry of Healthcare of Russia, Moscow, Russia
| | - Anastasia I. Shpichka
- Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, Moscow, Russia
- Lomonosov Moscow State University, Chemistry Department, Moscow, Russia
| | - Yuri M. Efremov
- Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, Moscow, Russia
| | - Vladimir I. Yusupov
- Institute of Photon Technologies of FSRC “Crystallography and Photonics” RAS, Troitsk, Moscow, Russia
| | - Peter S. Timashev
- Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, Moscow, Russia
- Lomonosov Moscow State University, Chemistry Department, Moscow, Russia
- Institute of Photon Technologies of FSRC “Crystallography and Photonics” RAS, Troitsk, Moscow, Russia
- N.N. Semenov Institute of Chemical Physics, Department of Polymers and Composites, Moscow, Russia
| | - Yury A. Rochev
- Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, Moscow, Russia
- National University of Ireland, National Centre for Biomedical Engineering Science, Galway, Ireland
| |
Collapse
|
20
|
Brboric A, Vasylovska S, Saarimäki-Vire J, Espes D, Caballero-Corbalan J, Larfors G, Otonkoski T, Lau J. Characterization of neural crest-derived stem cells isolated from human bone marrow for improvement of transplanted islet function. Ups J Med Sci 2019; 124:228-237. [PMID: 31623497 PMCID: PMC6968573 DOI: 10.1080/03009734.2019.1658661] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Murine boundary cap-derived neural crest stem cells (NCSCs) are capable of enhancing islet function by stimulating beta cell proliferation as well as increasing the neural and vascular density in the islets both in vitro and in vivo. This study aimed to isolate NCSC-like cells from human bone marrow.Methods: CD271 magnetic cell separation and culture techniques were used to purify a NCSC-enriched population of human bone marrow. Analyses of the CD271+ and CD271- fractions in terms of protein expression were performed, and the capacity of the CD271+ bone marrow cells to form 3-dimensional spheres when grown under non-adherent conditions was also investigated. Moreover, the NCSC characteristics of the CD271+ cells were evaluated by their ability to migrate toward human islets as well as human islet-like cell clusters (ICC) derived from pluripotent stem cells.Results: The CD271+ bone marrow population fulfilled the criterion of being multipotent stem cells, having the potential to differentiate into glial cells, neurons as well as myofibroblasts in vitro. They had the capacity to form 3-dimensional spheres as well as an ability to migrate toward human islets, further supporting their NCSC identity. Additionally, we demonstrated similar migration features toward stem cell-derived ICC.Conclusion: The results support the NCSC identity of the CD271-enriched human bone marrow population. It remains to investigate whether the human bone marrow-derived NCSCs have the ability to improve transplantation efficacy of not only human islets but stem cell-derived ICC as well.
Collapse
Affiliation(s)
- Anja Brboric
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Jonna Saarimäki-Vire
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Daniel Espes
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Gunnar Larfors
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Timo Otonkoski
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- CONTACT Joey Lau Department of Medical Cell Biology, Uppsala University, Husargatan 3, Box 571, SE-751 23 Uppsala, Sweden
| |
Collapse
|
21
|
Staels W, Heremans Y, Heimberg H, De Leu N. VEGF-A and blood vessels: a beta cell perspective. Diabetologia 2019; 62:1961-1968. [PMID: 31414144 DOI: 10.1007/s00125-019-4969-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/11/2019] [Indexed: 02/07/2023]
Abstract
Reciprocal signalling between the endothelium and the pancreatic epithelium is crucial for coordinated differentiation of the embryonic endocrine and exocrine pancreas. In the adult pancreas, islets depend on their dense capillary network to adequately respond to changes in plasma glucose levels. Vascular changes contribute to the onset and progression of both type 1 and type 2 diabetes. Impaired revascularisation of islets transplanted in individuals with type 1 diabetes is linked to islet graft failure and graft loss. This review summarises our understanding of the role of vascular endothelial growth factor-A (VEGF-A) and endothelial cells in beta cell development, physiology and disease. In addition, the therapeutic potential of modulating VEGF-A levels in beta and beta-like cells for transplantation is discussed.
Collapse
Affiliation(s)
- Willem Staels
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Institut Cochin, CNRS, INSERM, Université de Paris, F-75014, Paris, France
| | - Yves Heremans
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Harry Heimberg
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Nico De Leu
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
- Department of Endocrinology, UZ Brussel, Brussels, Belgium.
- Department of Endocrinology, ASZ Aalst, Aalst, Belgium.
| |
Collapse
|
22
|
Biofabrication of a vascularized islet organ for type 1 diabetes. Biomaterials 2019; 199:40-51. [PMID: 30735895 DOI: 10.1016/j.biomaterials.2019.01.035] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/27/2018] [Accepted: 01/18/2019] [Indexed: 12/17/2022]
Abstract
Islet transplantation is superior to extrinsic insulin supplementation in the treating severe Type 1 diabetes. However, its efficiency and longevity are limited by substantial islet loss post-transplantation due to lack of engraftment and vascular supply. To overcome these limitations, we developed a novel approach to bio-fabricate functional, vascularized islet organs (VIOs) ex vivo. We endothelialized acellular lung matrixes to provide a biocompatible multicompartment scaffold with an intact hierarchical vascular tree as a backbone for islet engraftment. Over seven days of culture, islets anatomically and functionally integrated into the surrounding bio-engineered vasculature, generating a functional perfusable endocrine organ. When exposed to supra-physiologic arterial glucose levels in vivo and ex vivo, mature VIOs responded with a physiologic insulin release from the vein and provided more efficient reduction of hyperglycemia compared to intraportally transplanted fresh islets. In long-term transplants in diabetic mice, subcutaneously implanted VIOs achieved normoglycemia significantly faster and more efficiently compared to islets that were transplanted in deviceless fashion. We conclude that ex vivo bio-fabrication of VIOs enables islet engraftment and vascularization before transplantation, and thereby helps to overcome limited islet survival and function observed in conventional islet transplantation. Given recent progress in stem cells, this technology may enable assembly of functional personalized endocrine organs.
Collapse
|
23
|
Yu Y, Gamble A, Pawlick R, Pepper AR, Salama B, Toms D, Razian G, Ellis C, Bruni A, Gala-Lopez B, Lu JL, Vovko H, Chiu C, Abdo S, Kin T, Korbutt G, Shapiro AMJ, Ungrin M. Bioengineered human pseudoislets form efficiently from donated tissue, compare favourably with native islets in vitro and restore normoglycaemia in mice. Diabetologia 2018; 61:2016-2029. [PMID: 29971529 PMCID: PMC6096633 DOI: 10.1007/s00125-018-4672-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/23/2018] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Islet transplantation is a treatment option that can help individuals with type 1 diabetes become insulin independent, but inefficient oxygen and nutrient delivery can hamper islet survival and engraftment due to the size of the islets and loss of the native microvasculature. We hypothesised that size-controlled pseudoislets engineered via centrifugal-forced-aggregation (CFA-PI) in a platform we previously developed would compare favourably with native islets, even after taking into account cell loss during the process. METHODS Human islets were dissociated and reaggregated into uniform, size-controlled CFA-PI in our microwell system. Their performance was assessed in vitro and in vivo over a range of sizes, and compared with that of unmodified native islets, as well as islet cell clusters formed by a conventional spontaneous aggregation approach (in which dissociated islet cells are cultured on ultra-low-attachment plates). In vitro studies included assays for membrane integrity, apoptosis, glucose-stimulated insulin secretion assay and total DNA content. In vivo efficacy was determined by transplantation under the kidney capsule of streptozotocin-treated Rag1-/- mice, with non-fasting blood glucose monitoring three times per week and IPGTT at day 60 for glucose response. A recovery nephrectomy, removing the graft, was conducted to confirm efficacy after completing the IPGTT. Architecture and composition were analysed by histological assessment via insulin, glucagon, pancreatic polypeptide, somatostatin, CD31 and von Willebrand factor staining. RESULTS CFA-PI exhibit markedly increased uniformity over native islets, as well as substantially improved glucose-stimulated insulin secretion (8.8-fold to 11.1-fold, even after taking cell loss into account) and hypoxia tolerance. In vivo, CFA-PI function similarly to (and potentially better than) native islets in reversing hyperglycaemia (55.6% for CFA-PI vs 20.0% for native islets at 500 islet equivalents [IEQ], and 77.8% for CFA-PI vs 55.6% for native islets at 1000 IEQ), and significantly better than spontaneously aggregated control cells (55.6% for CFA-PI vs 0% for spontaneous aggregation at 500 IEQ, and 77.8% CFA-PI vs 33.4% for spontaneous aggregation at 1000 IEQ; p < 0.05). Glucose clearance in the CFA-PI groups was improved over that in the native islet groups (CFA-PI 18.1 mmol/l vs native islets 29.7 mmol/l at 60 min; p < 0.05) to the point where they were comparable with the non-transplanted naive normoglycaemic control mice at a low IEQ of 500 IEQ (17.2 mmol/l at 60 min). CONCLUSIONS/INTERPRETATION The ability to efficiently reformat dissociated islet cells into engineered pseudoislets with improved properties has high potential for both research and therapeutic applications.
Collapse
Affiliation(s)
- Yang Yu
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Anissa Gamble
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Rena Pawlick
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Canadian National Transplant Research Program, Edmonton, AB, Canada
| | - Andrew R Pepper
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Canadian National Transplant Research Program, Edmonton, AB, Canada
| | - Bassem Salama
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Canadian National Transplant Research Program, Edmonton, AB, Canada
| | - Derek Toms
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Heritage Medical Research Building Room 412, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Golsa Razian
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Heritage Medical Research Building Room 412, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Cara Ellis
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Antonio Bruni
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Canadian National Transplant Research Program, Edmonton, AB, Canada
| | - Boris Gala-Lopez
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Canadian National Transplant Research Program, Edmonton, AB, Canada
| | - Jia Lulu Lu
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Heritage Medical Research Building Room 412, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Heather Vovko
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Heritage Medical Research Building Room 412, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Cecilia Chiu
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Heritage Medical Research Building Room 412, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Shaaban Abdo
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Heritage Medical Research Building Room 412, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Tatsuya Kin
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Greg Korbutt
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - A M James Shapiro
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Canadian National Transplant Research Program, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Mark Ungrin
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada.
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Heritage Medical Research Building Room 412, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
24
|
Staels W, Verdonck Y, Heremans Y, Leuckx G, De Groef S, Heirman C, de Koning E, Gysemans C, Thielemans K, Baeyens L, Heimberg H, De Leu N. Vegf-A mRNA transfection as a novel approach to improve mouse and human islet graft revascularisation. Diabetologia 2018; 61:1804-1810. [PMID: 29789879 DOI: 10.1007/s00125-018-4646-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/23/2018] [Indexed: 12/28/2022]
Abstract
AIMS/HYPOTHESIS The initial avascular period following islet transplantation seriously compromises graft function and survival. Enhancing graft revascularisation to improve engraftment has been attempted through virus-based delivery of angiogenic triggers, but risks associated with viral vectors have hampered clinical translation. In vitro transcribed mRNA transfection circumvents these risks and may be used for improving islet engraftment. METHODS Mouse and human pancreatic islet cells were transfected with mRNA encoding the angiogenic growth factor vascular endothelial growth factor A (VEGF-A) before transplantation under the kidney capsule in mice. RESULTS At day 7 post transplantation, revascularisation of grafts transfected with Vegf-A (also known as Vegfa) mRNA was significantly higher compared with non-transfected or Gfp mRNA-transfected controls in mouse islet grafts (2.11- and 1.87-fold, respectively) (vessel area/graft area, mean ± SEM: 0.118 ± 0.01 [n = 3] in Vegf-A mRNA transfected group (VEGF) vs 0.056 ± 0.01 [n = 3] in no RNA [p < 0.05] vs 0.063 ± 0.02 [n = 4] in Gfp mRNA transfected group (GFP) [p < 0.05]); EndoC-bH3 grafts (2.85- and 2.48-fold. respectively) (0.085 ± 0.02 [n = 4] in VEGF vs 0.030 ± 0.004 [n = 4] in no RNA [p < 0.05] vs 0.034 ± 0.01 [n = 5] in GFP [p < 0.05]); and human islet grafts (3.17- and 3.80-fold, respectively) (0.048 ± 0.013 [n = 3] in VEGF vs 0.015 ± 0.0051 [n = 4] in no RNA [p < 0.01] vs 0.013 ± 0.0046 [n = 4] in GFP [p < 0.01]). At day 30 post transplantation, human islet grafts maintained a vascularisation benefit (1.70- and 1.82-fold, respectively) (0.049 ± 0.0042 [n = 8] in VEGF vs 0.029 ± 0.0052 [n = 5] in no RNA [p < 0.05] vs 0.027 ± 0.0056 [n = 4] in GFP [p < 0.05]) and a higher beta cell volume (1.64- and 2.26-fold, respectively) (0.0292 ± 0.0032 μl [n = 7] in VEGF vs 0.0178 ± 0.0021 μl [n = 5] in no RNA [p < 0.01] vs 0.0129 ± 0.0012 μl [n = 4] in GFP [p < 0.001]). CONCLUSIONS/INTERPRETATION Vegf-A mRNA transfection before transplantation provides a promising and safe strategy to improve engraftment of islets and other cell-based implants.
Collapse
Affiliation(s)
- Willem Staels
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Department of Paediatrics, Division of Paediatric Endocrinology, Ghent University, Ghent, Belgium
| | - Yannick Verdonck
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Yves Heremans
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Gunter Leuckx
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Sofie De Groef
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eelco de Koning
- Department of Medicine, Section of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Conny Gysemans
- Laboratory of Clinical and Experimental Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Luc Baeyens
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Harry Heimberg
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Nico De Leu
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
- Department of Endocrinology, UZ Brussel, Brussels, Belgium.
- Department of Endocrinology, ASZ Aalst, Aalst, Belgium.
| |
Collapse
|
25
|
Garcia Ribeiro RS, Gysemans C, da Cunha JPMCM, Manshian BB, Jirak D, Kriz J, Gallo J, Bañobre-López M, Struys T, De Cuyper M, Mathieu C, Soenen SJ, Gsell W, Himmelreich U. Magnetoliposomes as Contrast Agents for Longitudinal in vivo Assessment of Transplanted Pancreatic Islets in a Diabetic Rat Model. Sci Rep 2018; 8:11487. [PMID: 30065302 PMCID: PMC6068133 DOI: 10.1038/s41598-018-29136-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 06/12/2018] [Indexed: 01/07/2023] Open
Abstract
Magnetoliposomes (MLs) were synthesized and tested for longitudinal monitoring of transplanted pancreatic islets using magnetic resonance imaging (MRI) in rat models. The rat insulinoma cell line INS-1E and isolated pancreatic islets from outbred and inbred rats were used to optimize labeling conditions in vitro. Strong MRI contrast was generated by islets exposed to 50 µg Fe/ml for 24 hours without any increased cell death, loss of function or other signs of toxicity. In vivo experiments showed that pancreatic islets (50-1000 units) labeled with MLs were detectable for up to 6 weeks post-transplantation in the kidney subcapsular space. Islets were also monitored for two weeks following transplantation through the portal vein of the liver. Hereby, islets labeled with MLs and transplanted under the left kidney capsule were able to correct hyperglycemia and had stable MRI signals until nephrectomy. Interestingly, in vivo MRI of streptozotocin induced diabetic rats transplanted with allogeneic islets demonstrated loss of MRI contrast between 7-16 days, indicative of loss of islet structure. MLs used in this study were not only beneficial for monitoring the location of transplanted islets in vivo with high sensitivity but also reported on islet integrity and hereby indirectly on islet function and rejection.
Collapse
Affiliation(s)
- Rita Sofia Garcia Ribeiro
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Biomedical Sciences Group, KU LEUVEN, Herestraat 49, 3000, Leuven, Belgium
| | - Conny Gysemans
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU LEUVEN, Herestraat 49, 3000, Leuven, Belgium
| | | | - Bella B Manshian
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Biomedical Sciences Group, KU LEUVEN, Herestraat 49, 3000, Leuven, Belgium
| | - Daniel Jirak
- MR Spectroscopy Unit, Institute for Clinical and Experimental Medicine (IKEM), Videnska 1958/9, 140 21, Prague, Czech Republic
- Department of Biophysics, Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovska 1, 120 00, Prague 2, Czech Republic
| | - Jan Kriz
- Diabetes Center, Institute for Clinical and Experimental Medicine (IKEM), Videnska 1958/9, 140 21, Prague, Czech Republic
| | - Juan Gallo
- Diagnostic Tools & Methods/Advanced (magnetic) Theranostic Nanostructures Lab, International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330, Braga, Portugal
| | - Manuel Bañobre-López
- Diagnostic Tools & Methods/Advanced (magnetic) Theranostic Nanostructures Lab, International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330, Braga, Portugal
| | - Tom Struys
- Lab of Histology, Biomedical Research Institute, Hasselt University, Campus Diepenbeek, Agoralaan, B3590, Diepenbeek, Belgium
| | - Marcel De Cuyper
- Laboratory of BioNanoColloids, Interdisciplinary Research Centre, KULAK/KU LEUVEN, Etienne Sabbelaan 53, 8500, Kortrijk, Belgium
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU LEUVEN, Herestraat 49, 3000, Leuven, Belgium
| | - Stefaan J Soenen
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Biomedical Sciences Group, KU LEUVEN, Herestraat 49, 3000, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Biomedical Sciences Group, KU LEUVEN, Herestraat 49, 3000, Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Biomedical Sciences Group, KU LEUVEN, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
26
|
Grapensparr L, Christoffersson G, Carlsson PO. Bioengineering with Endothelial Progenitor Cells Improves the Vascular Engraftment of Transplanted Human Islets. Cell Transplant 2018; 27:948-956. [PMID: 29862837 PMCID: PMC6050913 DOI: 10.1177/0963689718759474] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 11/16/2022] Open
Abstract
Pancreatic islets isolated for transplantation are disconnected from their vascular supply and need to establish a new functional network posttransplantation. Due to poor revascularization, prevailing hypoxia with correlating increased apoptosis rates in experimental studies can be observed for months posttransplantation. Endothelial progenitor cells (EPCs) are bone marrow-derived cells that promote neovascularization. The present study tested the hypothesis that EPCs, isolated from human umbilical cord blood, could be coated to human islet surfaces and be used to promote islet vascular engraftment. Control or EPC bioengineered human islets were transplanted into the renal subcapsular space of nonobese diabetic/severe combined immunodeficiency mice. Four weeks posttransplantation, graft blood perfusion and oxygen tension were measured using laser Doppler flowmetry and Clark microelectrodes, respectively. Vessel functionality was also assessed by in vivo confocal imaging. The vascular density and the respective contribution of human and recipient endothelium were assessed immunohistochemically by staining for human and mouse CD31. Islet grafts with EPCs had substantially higher blood perfusion and oxygen tension than control transplants. Furthermore, analysis of the vascular network of the grafts revealed that grafts containing EPC bioengineered islets had a superior vascular density compared with control grafts, with functional chimeric blood vessels. We conclude that a simple procedure of surface coating with EPCs provides a possibility to improve the vascular engraftment of transplanted human islets. Established protocols are also easily applicable for intraportal islet transplantation in order to obtain a novel directed cellular therapy at the site of implantation in the liver.
Collapse
Affiliation(s)
- Liza Grapensparr
- Department of Medical Cell Biology, Uppsala University, Uppsala,
Sweden
| | | | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala,
Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
27
|
Canzano JS, Nasif LH, Butterworth EA, Fu DA, Atkinson MA, Campbell-Thompson M. Islet Microvasculature Alterations With Loss of Beta-cells in Patients With Type 1 Diabetes. J Histochem Cytochem 2018; 67:41-52. [PMID: 29771178 DOI: 10.1369/0022155418778546] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Islet microvasculature provides key architectural and functional roles, yet the morphological features of islets from patients with type 1 diabetes are poorly defined. We examined islet and exocrine microvasculature networks by multiplex immunofluorescence imaging of pancreases from organ donors with and without type 1 diabetes (n=17 and n=16, respectively) and determined vessel diameter, density, and area. We also analyzed these variables in insulin-positive and insulin-negative islets of 7 type 1 diabetes donors. Control islet vessel diameter was significantly larger (7.6 ± 1.1 μm) compared with vessels in diabetic islets (6.2 ± 0.8 μm; p<0.001). Control islet vessel density (number/islet) was significantly lower (5.3 ± 0.6) versus diabetic islets (9.3 ± 0.2; p<0.001). Exocrine vessel variables were not significantly different between groups. Islets with residual beta-cells were comparable to control islets for both vessel diameter and density and were significantly different from insulin-negative islets within diabetic donors (p<0.05). Islet smooth muscle actin area had a significant positive correlation with age in both groups (p<0.05), which could negatively impact islet transplantation efficiency from older donors. These data underscore the critical relationship of islet beta-cells and islet vessel morphology in type 1 diabetes. These studies provide new knowledge of the islet microvasculature in diabetes and aging.
Collapse
Affiliation(s)
- Joseph S Canzano
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Lith H Nasif
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Elizabeth A Butterworth
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Dongtao A Fu
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida.,Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
28
|
Hospodiuk M, Dey M, Ayan B, Sosnoski D, Moncal KK, Wu Y, Ozbolat IT. Sprouting angiogenesis in engineered pseudo islets. Biofabrication 2018; 10:035003. [PMID: 29451122 DOI: 10.1088/1758-5090/aab002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Despite the recent achievements in cell-based therapies for curing type-1 diabetes (T1D), capillarization in beta (β)-cell clusters is still a major roadblock as it is essential for long-term viability and function of β-cells in vivo. In this research, we report sprouting angiogenesis in engineered pseudo islets (EPIs) made of mouse insulinoma βTC3 cells and rat heart microvascular endothelial cells (RHMVECs). Upon culturing in three-dimensional (3D) constructs under angiogenic conditions, EPIs sprouted extensive capillaries into the surrounding matrix. Ultra-morphological analysis through histological sections also revealed presence of capillarization within EPIs. EPIs cultured in 3D constructs maintained their viability and functionality over time while non-vascularized EPIs, without the presence of RHMVECs, could not retain their viability nor functionality. Here we demonstrate angiogenesis in engineered islets, where patient specific stem cell-derived human beta cells can be combined with microvascular endothelial cells in the near future for long-term graft survival in T1D patients.
Collapse
Affiliation(s)
- Monika Hospodiuk
- The Huck Institutes of the Life Sciences, Penn State University, State College, PA 16801, United States of America. Department of Agriculture and Biological Engineering, Penn State University, State College, PA 16801, United States of America
| | | | | | | | | | | | | |
Collapse
|
29
|
Min BH, Shin JS, Kim JM, Kang SJ, Kim HJ, Yoon IH, Park SK, Choi JW, Lee MS, Park CG. Delayed revascularization of islets after transplantation by IL-6 blockade in pig to non-human primate islet xenotransplantation model. Xenotransplantation 2017; 25. [PMID: 29210476 DOI: 10.1111/xen.12374] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/12/2017] [Accepted: 11/10/2017] [Indexed: 01/16/2023]
Abstract
BACKGROUND Pancreatic islet transplantation is currently proven as a promising treatment for type 1 diabetes patients with labile glycemic control and severe hypoglycemia unawareness. Upon islet transplantation, revascularization is essential for proper functioning of the transplanted islets. As IL-6 is important for endothelial cell survival and systemic inflammation related to xenograft, the effect of IL-6 receptor antagonist, tocilizumab, on revascularization of the transplanted islets was examined in pig to non-human primate islet xenotransplantation model. Also, the endothelial cell origin in a new vessel of the transplanted pig islets was determined. METHODS Pig islets were isolated from designated pathogen-free (DPF) SNU miniature pigs and transplanted via portal vein into five streptozotocin-induced diabetic monkeys. One group (n = 2, basal group) was treated with anti-thymoglobulin (ATG), anti-CD40 antibody (2C10R4), sirolimus, and tacrolimus, and the other group was additionally given tocilizumab on top of basal immunosuppression (n = 3, Tocilizumab group). To confirm IL-6 blocking effect, C-reactive protein (CRP) levels and serum IL-6 concentration were measured. Scheduled biopsy of the margin of the posterior segment right lobe inferior of the liver was performed at 3 weeks after transplantation to assess the degree of revascularization of the transplanted islets. Immunohistochemical staining using anti-insulin, anti-CD31 antibodies, and lectin IB4 was conducted to find the origin of endothelial cells in the islet graft. RESULTS CRP significantly increased at 1~2 days after transplantation in Basal group, but not in Tocilizumab group, and higher serum IL-6 concentration was measured in latter group, showing the biological potency of tocilizumab. In Basal group, well-developed endothelial cells were observed on the peri- and intraislet area, whereas the number of CD31+ cells in the intraislet space was significantly reduced in Tocilizumab group. Finally, new endothelial cells in the pig islet graft were positive for CD31, but not for lectin IB4, suggesting that they are originated from the recipient monkey. CONCLUSIONS Our results demonstrated that tocilizumab can delay revascularization of the transplanted islet, although this effect had no significant correlation to the overall islet graft survival. In the pig to NHP islet xenotransplantation model, the endothelial cells from recipient monkey form new blood vessels in and around pig islets.
Collapse
Affiliation(s)
- Byoung-Hoon Min
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Korea
| | - Jun-Seop Shin
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Min Kim
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Korea
| | - Seong-Jun Kang
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Je Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Il-Hee Yoon
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Korea
| | - Su-Kyoung Park
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Won Choi
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Min-Suk Lee
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Chung-Gyu Park
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Desai CS, Khan KM, Ma X, Li H, Wang J, Fan L, Chen G, Smith JP, Cui W. Effect of liver histopathology on islet cell engraftment in the model mimicking autologous islet cell transplantation. Islets 2017; 9:140-149. [PMID: 28902579 PMCID: PMC5710696 DOI: 10.1080/19382014.2017.1356558] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The inflammatory milieu in the liver as determined by histopathology is different in individual patients undergoing autologous islet cell transplantation. We hypothesized that inflammation related to fatty-liver adversely impacts islet survival. To test this hypothesis, we used a mouse model of fatty-liver to determine the outcome of syngeneic islet transplantation after chemical pancreatectomy. METHODS Mice (C57BL/6) were fed a high-fat-diet from 6 weeks of age until attaining a weight of ≥28 grams (6-8 weeks) to produce a fatty liver (histologically > 30% fat);steatosis was confirmed with lipidomic profile of liver tissue. Islets were infused via the intra-portal route in fatty-liver and control mice after streptozotocin induction of diabetes. Outcomes were assessed by the rate of euglycemia, liver histopathology, evaluation of liver inflammation by measuring tissue cytokines IL-1β and TNF-α by RT-PCR and CD31 expression by immunohistochemistry. RESULTS The difference in the euglycemic fraction between the normal liver group (90%, 9/10) and the fatty-liver group (37.5%, 3/8) was statistically significant at the 18th day post- transplant and was maintained to the end of the study (day 28) (p = 0.019, X2 = 5.51). Levels of TNF-α and IL-1β were elevated in fatty-liver mice (p = 0.042, p = 0.037). Compared to controls cytokine levels were elevated after islet cell transplantation and in transplanted fatty-liver mice as compared to either fatty- or islet transplant group alone (p = NS). A difference in the histochemical pattern of CD31 could not be determined. CONCLUSION Fatty-liver creates an inflammatory state which adversely affects the outcome of autologous islet cell transplantation.
Collapse
Affiliation(s)
- Chirag S. Desai
- Department of Surgery, University of North Carolina, Chapel Hill, NC, USA
- CONTACT Chirag S. Desai Department of Surgery, University of North Carolina4021 Burnett Womack Building, Campus Box 7211, Chapel Hill, NC 27599, USA
| | - Khalid M. Khan
- Medstar Georgetown Transplant Institute, Washington DC, USA
| | - Xiaobo Ma
- Islet Cell Laboratory, Medstar Georgetown University Hospital, Washington DC, USA
| | - Henghong Li
- Department of Medicine, Georgetown University Medical Center, Washington DC, USA
| | - Juan Wang
- Department of Medicine, Georgetown University Medical Center, Washington DC, USA
| | - Lijuan Fan
- Department of Medicine, Georgetown University Medical Center, Washington DC, USA
| | - Guoling Chen
- Islet Cell Laboratory, Medstar Georgetown University Hospital, Washington DC, USA
| | - Jill P. Smith
- Department of Medicine, Georgetown University Medical Center, Washington DC, USA
| | - Wanxing Cui
- Islet Cell Laboratory, Medstar Georgetown University Hospital, Washington DC, USA
| |
Collapse
|
31
|
Tootoonchi MH, Hashempour M, Blackwelder PL, Fraker CA. Manganese oxide particles as cytoprotective, oxygen generating agents. Acta Biomater 2017; 59:327-337. [PMID: 28688986 DOI: 10.1016/j.actbio.2017.07.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 02/05/2023]
Abstract
Cell culture and cellular transplant therapies are adversely affected by oxidative species and radicals. Herein, we present the production of bioactive manganese oxide nanoparticles for the purpose of radical scavenging and cytoprotection. Manganese comprises the core active structure of somatic enzymes that perform the same function, in vivo. Formulated nanoparticles were characterized structurally and surveyed for maximal activity (superoxide scavenging, hydrogen peroxide scavenging with resultant oxygen generation) and minimal cytotoxicity (48-h direct exposure to titrated manganese oxide concentrations). Cytoprotective capacity was tested using cell exposure to hydrogen peroxide in the presence or absence of the nanoparticles. Several ideal compounds were manufactured and utilized that showed complete disproportionation of superoxide produced by the xanthine/xanthine oxidase reaction. Further, the nanoparticles showed catalase-like activity by completely converting hydrogen peroxide into the corresponding concentration of oxygen. Finally, the particles protected cells (murine β-cell insulinoma) against insult from hydrogen peroxide exposure. Based on these observed properties, these particles could be utilized to combat oxidative stress and inflammatory response in a variety of cell therapy applications. STATEMENT OF SIGNIFICANCE Maintaining viability once cells have been removed from their physiological niche, e.g. culture and transplant, demands proper control of critical variables such as oxygenation and removal of harmful substances e.g. reactive oxygen species. Limited catalysts can transform reactive oxygen species into molecular oxygen and, thereby, have the potential to maintain cell viability and function. Among these are manganese oxide particles which are the subject of this study.
Collapse
|
32
|
Kumar S, Marriott CE, Alhasawi NF, Bone AJ, Macfarlane WM. The role of tumour suppressor PDCD4 in beta cell death in hypoxia. PLoS One 2017; 12:e0181235. [PMID: 28750063 PMCID: PMC5531437 DOI: 10.1371/journal.pone.0181235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 06/28/2017] [Indexed: 12/31/2022] Open
Abstract
Objective Hypoxia is known to induce pancreatic beta cell dysfunction and apoptosis. Changes in Programmed Cell Death Gene 4 (PDCD4) expression have previously been linked with beta cell neogenesis and function. Our aim was to investigate the effects of hypoxia on cell viability, PDCD4 expression and subcellular localisation. Methods MIN6 beta cells and ARIP ductal cells were exposed to 1% (hypoxia) or 21% O2 (normoxia) for 12 or 24 hours. MTT assay, HPI staining, scanning electron microscopy, western blotting and immunocytochemistry analyses were performed to determine the effect of hypoxia on cell viability, morphology and PDCD4 expression. Results 24 hour exposure to hypoxia resulted in ~70% loss of beta cell viability (P<0.001) compared to normoxia. Both HPI staining and SEM analysis demonstrated beta cell apoptosis and necrosis after 12 hours exposure to hypoxia. ARIP cells also displayed hypoxia-induced apoptosis and altered surface morphology after 24 hours, but no significant growth difference (p>0.05) was observed between hypoxic and normoxic conditions. Significantly higher expression of PDCD4 was observed in both beta cells (P<0.001) and ductal (P<0.01) cells under hypoxic conditions compared to controls. PDCD4 expression was localised to the cytoplasm of both beta cells and ductal cells, with no observed effects of hypoxia, normoxia or serum free conditions on intracellular shuttling of PDCD4. Conclusion These findings indicate that hypoxia-induced expression of PDCD4 is associated with increased beta cell death and suggests that PDCD4 may be an important factor in regulating beta cell survival during hypoxic stress.
Collapse
Affiliation(s)
- Sandeep Kumar
- Diabetes Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Claire E. Marriott
- Diabetes Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Nouf F. Alhasawi
- Diabetes Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Adrian J. Bone
- Diabetes Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Wendy M. Macfarlane
- Diabetes Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Jones GL, Juszczak MT, Hughes SJ, Kooner P, Powis SH, Press M. Time Course and Quantification of Pancreatic Islet Revasculariztion following Intraportal Transplantation. Cell Transplant 2017; 16:505-16. [PMID: 17708340 DOI: 10.3727/000000007783464993] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
A large proportion of islets are lost after transplantation partly due to a lack of functional vasculature. Islets revascularize from host tissue but the process takes up to 2 weeks and has been suggested to result in reduced vascular density in engrafted islets. We describe a method for observing and quantifying the revascularization of intraportally transplanted islets that includes number, density, and branching of islet capillaries. Syngeneic islets were transplanted selectively into the two right posterior lobes of the liver of adult Lewis rats. Sections of the livers were dual stained for insulin and Bandeiraea simplicifolia and analyzed for islet morphology, area, and vascular density from day 0 to day 14 posttransplant and compared to native islets. Vascular density was 1431 ± 75.7 vessels/mm2 in native islets and fell to 325.3 ± 30.8 vessels/mm2 (p < 0.001) by day 1 posttransplant and subsequently increased until day 14 when it was significantly higher than in native islets (2612.5 ± 107.8 vessels/mm2, p < 0.001). The percentage of islet area occupied by vascular space was 9.1 ± 0.9% in native islets. After falling to 2.3 ± 0.3% (p < 0.001) 1 day posttransplant this rose to supranormal levels (21.5 ± 0.8%, p < 0.001) by day 14. The index of capillary branching was 0.771 ± 0.017 in native islets and fell to 0.465 ± 0.02 (p = 0.001) by day 3 but returned to native values by day 7 posttransplantation (0.726 ± 0.03). This technique provides a robust method for tracking and quantifying the revascularization of intraportally transplanted islets, which should enable the comparison of different strategies aimed at accelerating islet revascularization.
Collapse
Affiliation(s)
- Gareth L Jones
- Centre for Nephrology, Royal Free Campus, Royal Free and University College Medical School, London, NW3 2PF, UK
| | | | | | | | | | | |
Collapse
|
34
|
Johansson A, Sandvik D, Carlsson PO. Inhibition of p38 MAP Kinase in the Early Posttransplantation Phase Redistributes Blood Vessels from the Surrounding Stroma into the Transplanted Endocrine Tissue. Cell Transplant 2017; 15:483-8. [PMID: 17121159 DOI: 10.3727/000000006783981729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transplanted pancreatic islets attain a chronically decreased vascular density following transplantation, despite the increased concentrations of vascular endothelial growth factor (VEGF) secreted from beta-cells in response to hypoxia during culture and in the immediate posttransplantation phase. VEGF, however, exerts dual effects on endothelial cells, and in islet endothelial cells of the adult, the vascular permeability-inducing effects of VEGF seem normally more pronounced than those to induce angiogenesis. p38 MAP kinase activity has recently been shown to serve as a switch to separate these properties of VEGF; inhibition of p38 MAP kinase activity enhances VEGF-induced angiogenesis and, at the same time, abrogates VEGF-induced vascular permeability. We hypothesized that the revascularization of transplanted islets may be hampered by a predisposition of adult islet endothelial cells to react to VEGF by forming fenestrae rather than migrating and proliferating. We therefore administered the p38 MAP kinase inhibitor SB203580 by daily IP injections for the first 14 days following transplantation, and then studied the influence of this treatment on the oxygen tension, blood perfusion, and vascular density of the islet grafts 1 month posttransplantation. SB203580 treatment redistributed islet graft blood vessels from the stroma into the endocrine tissue, and this redistribution of blood vessels into the endocrine tissue was accompanied by an increased oxygenation of the islet cells. However, the total number of blood vessels in the tissue was not affected. The blood perfusion of the islet grafts was also similar in control and SB203580-treated animals. Our results suggest that effects of VEGF to preferentially induce vascular permeability may partially contribute to, but is not the main cause of, low revascularization of transplanted islets.
Collapse
Affiliation(s)
- Asa Johansson
- Department of Medical Cell Biology, Uppsala University, SE-751 23 Uppsala, Sweden
| | | | | |
Collapse
|
35
|
Kidszun A, Schneider D, Erb D, Hertl G, Schmidt V, Eckhard M, Preissner KT, Breier G, Bretzel RG, Linn T. Isolated Pancreatic Islets in Three-Dimensional Matrices are Responsive to Stimulators and Inhibitors of Angiogenesis. Cell Transplant 2017; 15:489-97. [PMID: 17121160 DOI: 10.3727/000000006783981774] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The formation of a new microvasculature is essential for the long-term survival and function of the islet graft. In this study we examined endothelium of isolated pancreatic islets by stimulation with growth factors, different culture conditions, and genetic modification. We also inspected the effect of immunosuppressives used in human transplantation on angiogenesis. Isolated islets were embedded in a three-dimensional fibrin or Matrigel matrix. The effect of hyperglycemia, hypoxia, and the addition of VEGF and bFGF was investigated. We exposed islets from transgenic mice expressing the VEGF gene (RIP1VEGF-A) to high glucose (16.7 mmol/L) medium and tested the immunosuppressive agents rapamycin (100 ng/ml) and FK506 (100 ng/ml). To quantify angiogenesis the percentage of sprouting islets was determined. New endothelial capillary-like structures protruded from isolated pancreatic islets. Addition of VEGF to the islets and transgenic RIP-VEGF islets showed a two- to threefold increase of sprouting islets compared to control. Hypoxic culture conditions stimulated angiogenesis, resulting in a twofold increase of capillary sprouting. Rapamycin and FK506 proved to be potent inhibitors of angiogenesis in this system, because a decrease of sprouting islets of more than 20% by both agents was observed. Isolated pancreatic islets are capable of forming new capillary structures and are susceptible to pro- and antiangiogenic stimuli.
Collapse
Affiliation(s)
- André Kidszun
- Medical Clinic and Policlinic 3, Justus Liebig University, Rodthohl 6, 35392 Giessen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hogan MF, Hull RL. The islet endothelial cell: a novel contributor to beta cell secretory dysfunction in diabetes. Diabetologia 2017; 60:952-959. [PMID: 28396983 PMCID: PMC5505567 DOI: 10.1007/s00125-017-4272-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/02/2017] [Indexed: 11/25/2022]
Abstract
The pancreatic islet is highly vascularised, with an extensive capillary network. In addition to providing nutrients and oxygen to islet endocrine cells and transporting hormones to the peripheral circulation, islet capillaries (comprised primarily of islet endothelial cells) are an important source of signals that enhance survival and function of the islet beta cell. In type 2 diabetes, and animal models thereof, evidence exists of morphological and functional abnormalities in these islet endothelial cells. In diabetes, islet capillaries are thickened, dilated and fragmented, and islet endothelial cells express markers of inflammation and activation. In vitro data suggest that this dysfunctional islet endothelial phenotype may contribute to impaired insulin release from the beta cell. This review examines potential candidate molecules that may mediate the positive effects of islet endothelial cells on beta cell survival and function under normal conditions. Further, it explores possible mechanisms underlying the development of islet endothelial dysfunction in diabetes and reviews therapeutic options for ameliorating this aspect of the islet lesion in type 2 diabetes. Finally, considerations regarding differences between human and rodent islet vasculature and the potentially unforeseen negative consequences of strategies to expand the islet vasculature, particularly under diabetic conditions, are discussed.
Collapse
Affiliation(s)
- Meghan F Hogan
- Division of Metabolism, Endocrinology and Nutrition, VA Puget Sound Health Care System (151), 1660 South Columbian Way, Seattle, WA, 98108, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Rebecca L Hull
- Division of Metabolism, Endocrinology and Nutrition, VA Puget Sound Health Care System (151), 1660 South Columbian Way, Seattle, WA, 98108, USA.
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
37
|
Cohrs CM, Chen C, Jahn SR, Stertmann J, Chmelova H, Weitz J, Bähr A, Klymiuk N, Steffen A, Ludwig B, Kamvissi V, Wolf E, Bornstein SR, Solimena M, Speier S. Vessel Network Architecture of Adult Human Islets Promotes Distinct Cell-Cell Interactions In Situ and Is Altered After Transplantation. Endocrinology 2017; 158:1373-1385. [PMID: 28324008 DOI: 10.1210/en.2016-1184] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 01/24/2017] [Indexed: 11/19/2022]
Abstract
Islet-cell hormone release is modulated by signals from endothelial and endocrine cells within the islet. However, models of intraislet vascularization and paracrine cell signaling are mostly based on the rodent pancreas. We assessed the architecture and endocrine cell interaction of the vascular network in unperturbed human islets in situ and their potential to re-establish their endogenous vascular network after transplantation in vivo. We prepared slices of fresh pancreas tissue obtained from nondiabetic patients undergoing partial pancreatectomy. In addition, we transplanted human donor islets into the anterior chamber of the mouse eye. Next, we performed three-dimensional in situ and in vivo imaging of islet cell and vessel architecture at cellular resolution and compared our findings with mouse and porcine islets. Our data reveal a significantly different vascular architecture with decreased vessel diameter, reduced vessel branching, and shortened total vessel network in human compared with mouse islets. Together with the distinct cellular arrangement in human islets, this limits β to endothelial cell interactions, facilitates connection of α and β cells, and promotes the formation of independent β-cell clusters within islets. Furthermore, our results show that the endogenous vascular network of islets is significantly altered after transplantation in a donor age-related mechanism. Thus, our study provides insight into the vascular architecture and cellular arrangement of human islets with apparent consequences for intercellular islet signaling. Moreover, our findings suggest that human islet engraftment after transplantation can be improved by using alternative, less mature islet-cell sources.
Collapse
Affiliation(s)
- Christian M Cohrs
- Paul Langerhans Institute Dresden of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 München-Neuherberg, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- DFG-Center for Regenerative Therapies Dresden, Faculty of Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Chunguang Chen
- Paul Langerhans Institute Dresden of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 München-Neuherberg, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- DFG-Center for Regenerative Therapies Dresden, Faculty of Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Stephan R Jahn
- Paul Langerhans Institute Dresden of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 München-Neuherberg, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- DFG-Center for Regenerative Therapies Dresden, Faculty of Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Julia Stertmann
- Paul Langerhans Institute Dresden of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 München-Neuherberg, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- DFG-Center for Regenerative Therapies Dresden, Faculty of Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Helena Chmelova
- Paul Langerhans Institute Dresden of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 München-Neuherberg, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- DFG-Center for Regenerative Therapies Dresden, Faculty of Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Jürgen Weitz
- Department of GI, Thoracic and Vascular Surgery, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Andrea Bähr
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, 81377 Oberschleißheim, Germany
| | - Nikolai Klymiuk
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, 81377 Oberschleißheim, Germany
| | - Anja Steffen
- Paul Langerhans Institute Dresden of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 München-Neuherberg, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- Department of Medicine III, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Barbara Ludwig
- Paul Langerhans Institute Dresden of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 München-Neuherberg, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- Department of Medicine III, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Virginia Kamvissi
- Department of Medicine III, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Division of Diabetes and Nutritional Sciences, King's College London, SE19NH London, United Kingdom
| | - Eckhard Wolf
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, 81377 Oberschleißheim, Germany
| | - Stefan R Bornstein
- Paul Langerhans Institute Dresden of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 München-Neuherberg, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- Department of Medicine III, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Division of Diabetes and Nutritional Sciences, King's College London, SE19NH London, United Kingdom
| | - Michele Solimena
- Paul Langerhans Institute Dresden of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 München-Neuherberg, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Stephan Speier
- Paul Langerhans Institute Dresden of Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 München-Neuherberg, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
- DFG-Center for Regenerative Therapies Dresden, Faculty of Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
38
|
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
|
39
|
Espes D, Lau J, Quach M, Ullsten S, Christoffersson G, Carlsson PO. Rapid Restoration of Vascularity and Oxygenation in Mouse and Human Islets Transplanted to Omentum May Contribute to Their Superior Function Compared to Intraportally Transplanted Islets. Am J Transplant 2016; 16:3246-3254. [PMID: 27321369 DOI: 10.1111/ajt.13927] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 06/08/2016] [Accepted: 06/12/2016] [Indexed: 01/25/2023]
Abstract
Transplantation of islets into the liver confers several site-specific challenges, including a delayed vascularization and prevailing hypoxia. The greater omentum has in several experimental studies been suggested as an alternative implantation site for clinical use, but there has been no direct functional comparison to the liver. In this experimental study in mice, we characterized the engraftment of mouse and human islets in the omentum and compared engraftment and functional outcome with those in the intraportal site. The vascularization and innervation of the islets transplanted into the omentum were restored within the first month by paralleled ingrowth of capillaries and nerves. The hypoxic conditions in the islets early posttransplantation were transient and restricted to the first days. Newly formed blood vessels were fully functional, and the blood perfusion and oxygenation of the islets became similar to that of endogenous islets. Furthermore, islet grafts in the omentum showed at 1 month posttransplantation functional superiority to intraportally transplanted grafts. We conclude that in contrast to the liver the omentum provides excellent engraftment conditions for transplanted islets. Future studies in humans will be of great interest to investigate the capability of this site to also harbor larger grafts without interfering with islet functionality.
Collapse
Affiliation(s)
- D Espes
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden. .,Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| | - J Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - M Quach
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - S Ullsten
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - G Christoffersson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - P O Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
40
|
Publisher's note. Regen Ther 2016. [DOI: 10.1016/j.reth.2016.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
41
|
Tennant BR, Vanderkruk B, Dhillon J, Dai D, Verchere CB, Hoffman BG. Myt3 suppression sensitizes islet cells to high glucose-induced cell death via Bim induction. Cell Death Dis 2016; 7:e2233. [PMID: 27195679 PMCID: PMC4917670 DOI: 10.1038/cddis.2016.141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/11/2016] [Accepted: 04/22/2016] [Indexed: 12/13/2022]
Abstract
Diabetes is a chronic disease that results from the body's inability to properly control circulating blood glucose levels. The loss of glucose homoeostasis can arise from a loss of β-cell mass because of immune-cell-mediated attack, as in type 1 diabetes, and/or from dysfunction of individual β-cells (in conjunction with target organ insulin resistance), as in type 2 diabetes. A better understanding of the transcriptional pathways regulating islet-cell survival is of great importance for the development of therapeutic strategies that target β-cells for diabetes. To this end, we previously identified the transcription factor Myt3 as a pro-survival factor in islets following acute suppression of Myt3 in vitro. To determine the effects of Myt3 suppression on islet-cell survival in vivo, we used an adenovirus to express an shRNA targeting Myt3 in syngeneic optimal and marginal mass islet transplants, and demonstrate that suppression of Myt3 impairs the function of marginal mass grafts. Analysis of grafts 5 weeks post-transplant revealed that grafts transduced with the shMyt3 adenovirus contained ~20% the number of transduced cells as grafts transduced with a control adenovirus. In fact, increased apoptosis and significant cell loss in the shMyt3-transduced grafts was evident after only 5 days, suggesting that Myt3 suppression sensitizes islet cells to stresses present in the early post-transplant period. Specifically, we find that Myt3 suppression sensitizes islet cells to high glucose-induced cell death via upregulation of the pro-apoptotic Bcl2 family member Bim. Taken together these data suggest that Myt3 may be an important link between glucotoxic and immune signalling pathways.
Collapse
Affiliation(s)
- B R Tennant
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4
| | - B Vanderkruk
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4
| | - J Dhillon
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4
| | - D Dai
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4
| | - C B Verchere
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4.,Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada V5Z 4E3.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada V6T 2B5
| | - B G Hoffman
- Child and Family Research Institute, British Columbia Children's Hospital, 950 W28th Avenue, Vancouver, British Columbia, Canada V5Z 4H4.,Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada V5Z 4E3
| |
Collapse
|
42
|
Su K, Wang CF, Zhang Y, Cai YJ, Zhang YY, Zhao Q. The inhibitory effects of carnosic acid on cervical cancer cells growth by promoting apoptosis via ROS-regulated signaling pathway. Biomed Pharmacother 2016; 82:180-91. [PMID: 27470354 DOI: 10.1016/j.biopha.2016.04.056] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 04/26/2016] [Accepted: 04/26/2016] [Indexed: 12/20/2022] Open
Abstract
Cervical cancer has been the fourth most common cancer killing many women across the world. Carnosic acid (CA), as a phenolic diterpene, has been suggested to against cancer, exerting protective effects associated with inflammatory cytokines. It is aimed to demonstrate the therapeutic role of carnosic acid against cervical cancer and indicate its underlying molecular mechanisms. 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) was performed to assess the possible anti-proliferative effects of carnosic acid. And also, colony formation was used to further estimate carnosic acid's ability in suppressing cervical cancer cells proliferation. Flow cytometry assays were performed here to indicate the alterations of cervical cancer cells cycle and the development of apoptosis. Western blot assays and RT-PCR were also applied to clarify the apoptosis-associated signaling pathways affected by reactive oxygen species (ROS) generation. And immunofluorescence was used to detect ROS-positive cells. In vivo experiments, CaSki xenograft model samples of nude mice were involved to further elucidate the effects of carnosic acid. In our results, we found that carnosic acid exerted anti-tumor ability in vitro supported by up-regulation of apoptosis and ROS production in cervical cancer cells. Also, acceleration of ROS led to the phospharylation of (c-Jun N-terminal kinase (JNK) and its-related signals, as well as activation of Endoplasmic Reticulum (ER) stress, promoting the progression of apoptosis via stimulating Caspase3 expression. The development and growth of xenograft tumors in nude mice were found to be inhibited by the administration of carnosic acid for five weeks. And the suppressed role of carnosic acid in proliferation of cervical cancer cells and apoptosis of nude mice with tumor tissues were observed in our study. Taken together, our data indicated that carnosic acid resulted in apoptosis both in vitro and vivo experiments via promoting ROS and activating JNK signaling pathways in human cervical cancer cells, which supplied a potential therapy for the application of carnosic acid in clinical treatment.
Collapse
Affiliation(s)
- Ke Su
- Department of gynecology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan 450052, PR China
| | - Chun-Fang Wang
- Department of gynecology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan 450052, PR China
| | - Ying Zhang
- Department of gynecology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan 450052, PR China
| | - Yu-Jie Cai
- Department of gynecology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan 450052, PR China
| | - Yan-Yan Zhang
- Department of gynecology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan 450052, PR China
| | - Qian Zhao
- Department of gynecology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan 450052, PR China.
| |
Collapse
|
43
|
Jansson L, Barbu A, Bodin B, Drott CJ, Espes D, Gao X, Grapensparr L, Källskog Ö, Lau J, Liljebäck H, Palm F, Quach M, Sandberg M, Strömberg V, Ullsten S, Carlsson PO. Pancreatic islet blood flow and its measurement. Ups J Med Sci 2016; 121:81-95. [PMID: 27124642 PMCID: PMC4900068 DOI: 10.3109/03009734.2016.1164769] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pancreatic islets are richly vascularized, and islet blood vessels are uniquely adapted to maintain and support the internal milieu of the islets favoring normal endocrine function. Islet blood flow is normally very high compared with that to the exocrine pancreas and is autonomously regulated through complex interactions between the nervous system, metabolites from insulin secreting β-cells, endothelium-derived mediators, and hormones. The islet blood flow is normally coupled to the needs for insulin release and is usually disturbed during glucose intolerance and overt diabetes. The present review provides a brief background on islet vascular function and especially focuses on available techniques to measure islet blood perfusion. The gold standard for islet blood flow measurements in experimental animals is the microsphere technique, and its advantages and disadvantages will be discussed. In humans there are still no methods to measure islet blood flow selectively, but new developments in radiological techniques hold great hopes for the future.
Collapse
Affiliation(s)
- Leif Jansson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- CONTACT Leif Jansson, Department of Medical Cell Biology, Biomedical Centre, Box 571, Husargatan 3, SE-75123 Uppsala, Sweden
| | - Andreea Barbu
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Birgitta Bodin
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Carl Johan Drott
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Daniel Espes
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Xiang Gao
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Liza Grapensparr
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Örjan Källskog
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Hanna Liljebäck
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Fredrik Palm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - My Quach
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Monica Sandberg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Sara Ullsten
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
44
|
Barkai U, Rotem A, de Vos P. Survival of encapsulated islets: More than a membrane story. World J Transplant 2016; 6:69-90. [PMID: 27011906 PMCID: PMC4801806 DOI: 10.5500/wjt.v6.i1.69] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/02/2015] [Accepted: 12/20/2015] [Indexed: 02/05/2023] Open
Abstract
At present, proven clinical treatments but no cures are available for diabetes, a global epidemic with a huge economic burden. Transplantation of islets of Langerhans by their infusion into vascularized organs is an experimental clinical protocol, the first approach to attain cure. However, it is associated with lifelong use of immunosuppressants. To overcome the need for immunosuppression, islets are encapsulated and separated from the host immune system by a permselective membrane. The lead material for this application is alginate which was tested in many animal models and a few clinical trials. This review discusses all aspects related to the function of transplanted encapsulated islets such as the basic requirements from a permselective membrane (e.g., allowable hydrodynamic radii, implications of the thickness of the membrane and relative electrical charge). Another aspect involves adequate oxygen supply, which is essential for survival/performance of transplanted islets, especially when using large retrievable macro-capsules implanted in poorly oxygenated sites like the subcutis. Notably, islets can survive under low oxygen tension and are physiologically active at > 40 Torr. Surprisingly, when densely crowded, islets are fully functional under hyperoxic pressure of up to 500 Torr (> 300% of atmospheric oxygen tension). The review also addresses an additional category of requirements for optimal performance of transplanted islets, named auxiliary technologies. These include control of inflammation, apoptosis, angiogenesis, and the intra-capsular environment. The review highlights that curing diabetes with a functional bio-artificial pancreas requires optimizing all of these aspects, and that significant advances have already been made in many of them.
Collapse
|
45
|
A hybrid of cells and pancreatic islets toward a new bioartificial pancreas. Regen Ther 2016; 3:68-74. [PMID: 31245475 PMCID: PMC6581840 DOI: 10.1016/j.reth.2016.03.004] [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: 11/30/2015] [Revised: 01/31/2016] [Accepted: 02/12/2016] [Indexed: 01/30/2023] Open
Abstract
Cell surface engineering using single-stranded DNA-poly(ethylene glycol)-conjugated phospholipid (ssDNA-PEG-lipid) is useful for inducing cell-cell attachment two and three dimensionally. In this review, we summarize our recent techniques for cell surface engineering and their applications to islet transplantation. Because any DNA sequence can be immobilized onto the cell surface by hydrophobic interactions between ssDNA-PEG-lipid and the cellular membrane without impairing cell function, a cell-cell hybrid can be formed through the DNA hybridization. With this technique, it would be possible to create three-dimensional hybrid structures of pancreatic islets coated with various accessory cells, such as patients' own cells, mesenchymal and adipose-derived stem cells, endothelial progenitor cells, neural crest stem cells or regulatory T cells, which might significantly improve the outcome of islet transplantation in diabetic patients.
Collapse
|
46
|
Staels W, De Groef S, Heremans Y, Coppens V, Van Gassen N, Leuckx G, Van de Casteele M, Van Riet I, Luttun A, Heimberg H, De Leu N. Accessory cells for β-cell transplantation. Diabetes Obes Metab 2016; 18:115-24. [PMID: 26289770 DOI: 10.1111/dom.12556] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/22/2015] [Accepted: 08/13/2015] [Indexed: 12/16/2022]
Abstract
Despite recent advances, insulin therapy remains a treatment, not a cure, for diabetes mellitus with persistent risk of glycaemic alterations and life-threatening complications. Restoration of the endogenous β-cell mass through regeneration or transplantation offers an attractive alternative. Unfortunately, signals that drive β-cell regeneration remain enigmatic and β-cell replacement therapy still faces major hurdles that prevent its widespread application. Co-transplantation of accessory non-islet cells with islet cells has been shown to improve the outcome of experimental islet transplantation. This review will highlight current travails in β-cell therapy and focuses on the potential benefits of accessory cells for islet transplantation in diabetes.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Cell Separation/trends
- Cells, Cultured
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/surgery
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/surgery
- Endothelial Progenitor Cells/cytology
- Endothelial Progenitor Cells/immunology
- Endothelial Progenitor Cells/pathology
- Endothelial Progenitor Cells/transplantation
- Graft Rejection/immunology
- Graft Rejection/metabolism
- Graft Rejection/prevention & control
- Graft Survival
- Humans
- Immune Tolerance
- Insulin-Secreting Cells/cytology
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/transplantation
- Islets of Langerhans Transplantation/adverse effects
- Islets of Langerhans Transplantation/immunology
- Mesenchymal Stem Cell Transplantation/adverse effects
- Mesenchymal Stem Cell Transplantation/trends
- Neural Crest/cytology
- Neural Crest/immunology
- Neural Crest/pathology
- Neural Crest/transplantation
- Stem Cell Transplantation/adverse effects
- Stem Cell Transplantation/trends
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- T-Lymphocytes, Regulatory/transplantation
- Transplantation, Autologous/adverse effects
- Transplantation, Autologous/trends
- Transplantation, Heterotopic/adverse effects
- Transplantation, Heterotopic/trends
- Transplantation, Homologous/adverse effects
- Transplantation, Homologous/trends
Collapse
Affiliation(s)
- W Staels
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
- Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium
| | - S De Groef
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Y Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - V Coppens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - N Van Gassen
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - G Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - M Van de Casteele
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - I Van Riet
- Department Hematology Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - A Luttun
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - H Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - N De Leu
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Endocrinology, UZ Brussel, Brussels, Belgium
- Department of Endocrinology, ASZ Aalst, Aalst, Belgium
| |
Collapse
|
47
|
Suszynski TM, Avgoustiniatos ES, Papas KK. Oxygenation of the Intraportally Transplanted Pancreatic Islet. J Diabetes Res 2016; 2016:7625947. [PMID: 27872862 PMCID: PMC5107248 DOI: 10.1155/2016/7625947] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 04/27/2016] [Indexed: 12/04/2022] Open
Abstract
Intraportal islet transplantation (IT) is not widely utilized as a treatment for type 1 diabetes. Oxygenation of the intraportally transplanted islet has not been studied extensively. We present a diffusion-reaction model that predicts the presence of an anoxic core and a larger partly functional core within intraportally transplanted islets. Four variables were studied: islet diameter, islet fractional viability, external oxygen partial pressure (P) (in surrounding portal blood), and presence or absence of a thrombus on the islet surface. Results indicate that an islet with average size and fractional viability exhibits an anoxic volume fraction (AVF) of 14% and a function loss of 72% at a low external P. Thrombus formation increased AVF to 30% and function loss to 92%, suggesting that the effect of thrombosis may be substantial. External P and islet diameter accounted for the greatest overall impact on AVF and loss of function. At our institutions, large human alloislets (>200 μm diameter) account for ~20% of total islet number but ~70% of total islet volume; since most of the total transplanted islet volume is accounted for by large islets, most of the intraportal islet cells are likely to be anoxic and not fully functional.
Collapse
Affiliation(s)
| | | | - Klearchos K. Papas
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Cellular Transplantation, Department of Surgery, University of Arizona, Tucson, AZ 85724, USA
- *Klearchos K. Papas:
| |
Collapse
|
48
|
Moore SJ, Gala-Lopez BL, Pepper AR, Pawlick RL, Shapiro AMJ. Bioengineered stem cells as an alternative for islet cell transplantation. World J Transplant 2015; 5:1-10. [PMID: 25815266 PMCID: PMC4371156 DOI: 10.5500/wjt.v5.i1.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 02/18/2014] [Accepted: 10/29/2014] [Indexed: 02/05/2023] Open
Abstract
Type 1 diabetes is an autoimmune and increasingly prevalent condition caused by immunological destruction of beta cells. Insulin remains the mainstay of therapy. Endeavours in islet transplantation have clearly demonstrated that type 1 diabetes is treatable by cellular replacement. Many challenges remain with this approach. The opportunity to use bioengineered embryonic or adult pluripotential stem cells, or islets derived from porcine xenograft sources could address future demands, but are still associated with considerable challenges. This detailed review outlines current progress in clinical islet transplantation, and places this in perspective for the remarkable scientific advances now occurring in stem cell and regenerative medicine approaches in the treatment of future curative treatment of diabetes.
Collapse
|
49
|
Abstract
BACKGROUND The islet size distribution in a preparation may contribute to islet transplant outcomes. At the same islet equivalent (IE) dose, larger islets may exhibit poorer therapeutic value and this may be because of oxygen diffusion limitations that worsen in proportion to islet size. METHODS To test this hypothesis, we studied the impact of islet size index (ISI) and other islet product characteristics on outcomes after islet autotransplant (IAT) in recipients receiving a marginal islet dose (2000-4999 IEs per kg body weight) from January 1, 2009 to June 11, 2012, at the University of Minnesota (n=58). ISI was defined as the number of IE divided by the number of islet particles (IPs) in a preparation; an ISI less than 1 indicates a mean islet diameter that is less than 150 μm. The primary post-IAT outcome was 6-month insulin use status. RESULTS Logistic regression analysis indicate that IPs/kg (P=0.001), IEs/kg (P=0.019), total IPs transplanted (P=0.040), and ISI (P=0.074) were most strongly correlated with the primary outcome. The ISI (mean±standard error) was lower for recipients achieving insulin independence at 6 months (0.71±0.05) versus those partially (0.83±0.05) or completely (1.00±0.07) insulin dependent. The combination of islet dose (expressed as units IPs/kg) and ISI exhibited a sensitivity of 75% and specificity of 74% in predicting insulin independence in this population of patients. CONCLUSION Islet autotransplant recipients of a marginal islet doses were more likely to achieve insulin independence when transplanted with a greater number of smaller islets.
Collapse
|
50
|
Abstract
Islet transplantation (IT) is a promising therapy for the treatment of diabetes. The large number of islets required to achieve insulin independence limit its cost-effectiveness and the number of patients who can be treated. It is believed that >50% of islets are lost in the immediate post-IT period. Poor oxygenation in the early post-IT period is recognized as a possible reason for islet loss and dysfunction but has not been extensively studied. Several key variables affect oxygenation in this setting, including (1) local oxygen partial pressure (pO(2)), (2) islet oxygen consumption, (3) islet size (diameter, D), and (4) presence or absence of thrombosis on the islet surface. We discuss implications of oxygen-limiting conditions on intraportal islet viability and function. Of the 4 key variables, the islet size appears to be the most important determinant of the anoxic and nonfunctional islet volume fractions. Similarly, the effect of thrombus formation on the islet surface may be substantial. At the University of Minnesota, average size distribution data from clinical alloislet preparations (n = 10) indicate that >150-µm D islets account for only ~30% of the total islet number, but >85% of the total islet volume. This suggests that improved oxygen supply to the islets may have a profound impact on islet survivability and function since most of the β-cell volume is within large islets which are most susceptible to oxygen-limiting conditions. The assumption that the liver is a suitable islet transplant site from the standpoint of oxygenation should be reconsidered.
Collapse
|