1
|
Zhao Q, Li J, Lin Z, Tang Y, Yang D, Qin M, Ma X, Ji H, Chen H, Wang T, Chen M, Ju W, Wang D, Guo Z, Zhu X, Dan J, Hu A, He X. The First Case of Intra-portal Islet Implantation During Liver Machine Perfusion Allowing Simultaneous Islet-liver Transplantation in A Human: A New and Safe Treatment for End-stage Liver Disease Combined With Diabetes Mellitus. Ann Surg 2024; 281:00000658-990000000-01066. [PMID: 39247951 PMCID: PMC11723485 DOI: 10.1097/sla.0000000000006526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
OBJECTIVE Evaluating the safety and efficacy of implanting a liver with islet grafts into patients with end-stage liver disease and diabetes mellitus (DM). BACKGROUND DM and end-stage liver diseases are significant health concern worldwide, often coexisting and mutually influencing each other. Addressing both diseases simultaneously is paramount. METHODS We utilized the islet transplantation combined ischemia-free liver transplantation (ITIFLT) technique to treat a patient with hepatocellular carcinoma (HCC) and type 2 diabetes mellitus (T2DM). The liver was procured and preserved using the ischemia-free liver transplantation (IFLT) technique, and during normothermic machine perfusion (NMP), isolated and purified islet grafts were transplanted into the liver through the portal vein. Finally, the liver, incorporating with the transplant islet grafts, was implanted into the recipient without interruption of blood supply. RESULTS The patient received both liver and islet graft from the same donor. The patient achieved insulin-independence by post-transplant day (PTD) 9, and both liver and islet function remained robust. The patient was discharged on PTD 18 and experienced no surgical or transplantation-related complications during the follow-up period. Furthermore, islet grafts presence was observed in liver biopsies after islet transplantation. CONCLUSIONS This landmark case marks the inaugural application of ITIFLT in humans, signifying its potential as a promising treatment modality for end-stage liver disease with DM.
Collapse
Affiliation(s)
- Qiang Zhao
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Jiahao Li
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Zepeng Lin
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Yunhua Tang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Daopeng Yang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Meiting Qin
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Xue Ma
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Haibin Ji
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Honghui Chen
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Tielong Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Maogen Chen
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Weiqiang Ju
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Dongping Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Zhiyong Guo
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Xiaofeng Zhu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Jia Dan
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Anbin Hu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Xiaoshun He
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Medicine, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| |
Collapse
|
2
|
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
|
3
|
Purich K, Cai H, Yang B, Xu Z, Tessier AG, Black A, Hung RW, Boivin E, Xu B, Wu P, Zhang B, Xin D, Fallone BG, Rajotte RV, Wu Y, Rayat GR. MRI monitoring of transplanted neonatal porcine islets labeled with polyvinylpyrrolidone-coated superparamagnetic iron oxide nanoparticles in a mouse model. Xenotransplantation 2021; 29:e12720. [PMID: 34850455 DOI: 10.1111/xen.12720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/25/2021] [Accepted: 10/22/2021] [Indexed: 11/27/2022]
Abstract
Islet transplantation is a potential treatment option for certain patients with type 1 diabetes; however, it still faces barriers to widespread use, including the lack of tools to monitor islet grafts post-transplantation. This study investigates whether labeling neonatal porcine islets (NPI) with polyvinylpyrrolidone-coated superparamagnetic iron oxide nanoparticles (PVP-SPIO) affects their function, and whether this nanoparticle can be utilized to monitor NPI xenografts with magnetic resonance imaging (MRI) in a mouse model. In vitro, PVP-SPIO-labeled NPI in an agarose gel was visualized clearly by MRI. PVP-SPIO-labeled islets were then transplanted under the kidney capsules of immunodeficient nondiabetic and diabetic mice. All diabetic mice that received transplantation of PVP-SPIO-labeled islets reached normoglycemia. Grafts appeared as hypo-intense areas on MRI and were distinguishable from the surrounding tissues. Following injection of spleen cells from immunocompetent mice, normoglycemic recipient mice became diabetic and islet grafts showed an increase in volume, accompanied by a mixed signal on MRI. Overall, this study demonstrates that PVP-SPIO did not affect the function of NPI that PVP-SPIO-labeled islets were easily seen on MRI, and changes in MRI signals following rejection suggest a potential use of PVP-SPIO-labeled islets to monitor graft viability.
Collapse
Affiliation(s)
- Kieran Purich
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Haolei Cai
- Department of Surgery, 2nd Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Bin Yang
- Department of Surgery, 2nd Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Zhihao Xu
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anthony G Tessier
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Physics, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Adnan Black
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ryan W Hung
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
| | - Eric Boivin
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Baoyou Xu
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ping Wu
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Bo Zhang
- Department of Surgery, 2nd Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Dong Xin
- Department of Surgery, 2nd Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Biagio Gino Fallone
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Physics, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Ray V Rajotte
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Yulian Wu
- Department of Surgery, 2nd Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Gina R Rayat
- Department of Surgery, Ray Rajotte Surgical-Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
4
|
Akolpoglu MB, Inceoglu Y, Bozuyuk U, Sousa AR, Oliveira MB, Mano JF, Kizilel S. Recent advances in the design of implantable insulin secreting heterocellular islet organoids. Biomaterials 2020; 269:120627. [PMID: 33401104 DOI: 10.1016/j.biomaterials.2020.120627] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
Islet transplantation has proved one of the most remarkable transmissions from an experimental curiosity into a routine clinical application for the treatment of type I diabetes (T1D). Current efforts for taking this technology one-step further are now focusing on overcoming islet donor shortage, engraftment, prolonged islet availability, post-transplant vascularization, and coming up with new strategies to eliminate lifelong immunosuppression. To this end, insulin secreting 3D cell clusters composed of different types of cells, also referred as heterocellular islet organoids, spheroids, or pseudoislets, have been engineered to overcome the challenges encountered by the current islet transplantation protocols. β-cells or native islets are accompanied by helper cells, also referred to as accessory cells, to generate a cell cluster that is not only able to accurately secrete insulin in response to glucose, but also superior in terms of other key features (e.g. maintaining a vasculature, longer durability in vivo and not necessitating immunosuppression after transplantation). Over the past decade, numerous 3D cell culture techniques have been integrated to create an engineered heterocellular islet organoid that addresses current obstacles. Here, we first discuss the different cell types used to prepare heterocellular organoids for islet transplantation and their contribution to the organoids design. We then introduce various cell culture techniques that are incorporated to prepare a fully functional and insulin secreting organoids with select features. Finally, we discuss the challenges and present a future outlook for improving clinical outcomes of islet transplantation.
Collapse
Affiliation(s)
- M Birgul Akolpoglu
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey
| | - Yasemin Inceoglu
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey
| | - Ugur Bozuyuk
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey
| | - Ana Rita Sousa
- Department of Chemistry, CICECO - Aveiro Institute of Materials. University of Aveiro. Campus Universitário de Santiago. 3810-193 Aveiro. Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials. University of Aveiro. Campus Universitário de Santiago. 3810-193 Aveiro. Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials. University of Aveiro. Campus Universitário de Santiago. 3810-193 Aveiro. Portugal
| | - Seda Kizilel
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey.
| |
Collapse
|
5
|
Gastric submucosal alleviated pro-inflammation cytokines mediated initial dysfunction of islets allografts. Transpl Immunol 2020; 65:101292. [PMID: 32302641 DOI: 10.1016/j.trim.2020.101292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND The liver and renal capsule are the most common site for experimental pancreatic islet transplantation, but it is not optimal. Gastric submucosa space may be an ideal site for islet transplantation; however, whether pro-inflammation factors mediated islet dysfunction could be avoided or alleviated is still unclear. METHODS Islets of Sprague Dawley (SD) rat were transplanted into the streptozotocin-induced diabetic SD rats. Transplantation sites included gastric submucosa (GS), intraportal vein (PV) and kidney capsule (KC), and the efficiency of glycemic control and site-specific differences of islet grafts were compared. RESULTS With limited number of islets (800 IEQ) transplanted, improvement of recipient glycometabolism was superior in the GS group. When transplanted with 1200 IEQ islets, the survival of islet grafts were significantly prolonged in the GS group (25.87 ± 4.08 days, compared to 15.97 ± 0.83 days and 17.33 ± 1.41 days in PV and KC groups, respectively, P < .05). Compared with the PV group, the levels of IL-1β and TNF-α were significantly depressed in GS group after 12 h transplantation (15.5 ± 0.70 pg/mL and 13.28 ± 2.80 pg/mL vs. 262.26 ± 53.37 pg/mL and 138.51 ± 39.58 pg/mL, P < .05). CONCLUSIONS Gastric submucosal would be a potential ideal site for islet transplantation in rat. Gastric submucosal might alleviate the early islet dysfunction triggered by the IL-1β and TNF-α, and which requires a low number of transplanted islets and have a good glycemic control in return.
Collapse
|
6
|
Peloso A, Citro A, Zoro T, Cobianchi L, Kahler-Quesada A, Bianchi CM, Andres A, Berishvili E, Piemonti L, Berney T, Toso C, Oldani G. Regenerative Medicine and Diabetes: Targeting the Extracellular Matrix Beyond the Stem Cell Approach and Encapsulation Technology. Front Endocrinol (Lausanne) 2018; 9:445. [PMID: 30233489 PMCID: PMC6127205 DOI: 10.3389/fendo.2018.00445] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/18/2018] [Indexed: 12/20/2022] Open
Abstract
According to the Juvenile Diabetes Research Foundation (JDRF), almost 1. 25 million people in the United States (US) have type 1 diabetes, which makes them dependent on insulin injections. Nationwide, type 2 diabetes rates have nearly doubled in the past 20 years resulting in more than 29 million American adults with diabetes and another 86 million in a pre-diabetic state. The International Diabetes Ferderation (IDF) has estimated that there will be almost 650 million adult diabetic patients worldwide at the end of the next 20 years (excluding patients over the age of 80). At this time, pancreas transplantation is the only available cure for selected patients, but it is offered only to a small percentage of them due to organ shortage and the risks linked to immunosuppressive regimes. Currently, exogenous insulin therapy is still considered to be the gold standard when managing diabetes, though stem cell biology is recognized as one of the most promising strategies for restoring endocrine pancreatic function. However, many issues remain to be solved, and there are currently no recognized treatments for diabetes based on stem cells. In addition to stem cell resesarch, several β-cell substitutive therapies have been explored in the recent era, including the use of acellular extracellular matrix scaffolding as a template for cellular seeding, thus providing an empty template to be repopulated with β-cells. Although this bioengineering approach still has to overcome important hurdles in regards to clinical application (including the origin of insulin producing cells as well as immune-related limitations), it could theoretically provide an inexhaustible source of bio-engineered pancreases.
Collapse
Affiliation(s)
- Andrea Peloso
- Division of Abdominal Surgery, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- HepatoPancreato-Biliary Centre, Geneva University Hospitals, Geneva, Switzerland
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Tamara Zoro
- Department of General Surgery, IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Lorenzo Cobianchi
- Department of General Surgery, IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Arianna Kahler-Quesada
- Division of Abdominal Surgery, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Carlo M. Bianchi
- Department of General Surgery, IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Axel Andres
- Division of Abdominal Surgery, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- HepatoPancreato-Biliary Centre, Geneva University Hospitals, Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, University of Geneva, Geneva, Switzerland
- Institute of Medical Research, Ilia State University, Tbilisi, Georgia
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Thierry Berney
- Cell Isolation and Transplantation Center, University of Geneva, Geneva, Switzerland
| | - Christian Toso
- Division of Abdominal Surgery, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- HepatoPancreato-Biliary Centre, Geneva University Hospitals, Geneva, Switzerland
| | - Graziano Oldani
- Division of Abdominal Surgery, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- HepatoPancreato-Biliary Centre, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
7
|
Lew B, Kim IY, Choi H, Kim K. Sustained exenatide delivery via intracapsular microspheres for improved survival and function of microencapsulated porcine islets. Drug Deliv Transl Res 2018; 8:857-862. [DOI: 10.1007/s13346-018-0484-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
8
|
Pileggi A, Molano RD, Berney T, Ichii H, Jose SS, Zahr E, Poggioli R, Linetsky E, Ricordi C, Inverardi L. Prolonged Allogeneic Islet Graft Survival by Protoporphyrins. Cell Transplant 2017; 14:85-96. [DOI: 10.3727/000000005783983160] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transplantation of islets of Langerhans in patients with type 1 diabetes allows for improved metabolic control and insulin independence. The need for chronic immunosuppression limits this procedure to selected patients with brittle diabetes. Definition of therapeutic strategies allowing permanent engraftment without the need for chronic immunosuppression could overcome such limitations. We tested the effect of the use of protoporphyrins (CoPP and FePP), powerful inducers of the cytoprotective protein hemeoxygenase 1 (HO-1), on allogeneic islet graft survival. Chemically induced diabetic C57BL/6 mice received DBA/2 islets. Treatment consisted in peritransplant administration of CoPP or saline. Islets were either cultured in the presence of FePP or vehicle before implant. Short-course administration of CoPP led to long-term islet allograft survival in a sizable proportion of recipients. Long-term graft-bearing animals rejected third-party islets while accepting a second set donor-specific graft permanently, without additional treatment. Preconditioning of islets with FePP by itself led to improved graft survival in untreated recipients, and provided additional advantage in CoPP-treated recipients, resulting in an increased proportion of long-term surviving grafts. Preconditioning of the graft with protoporphyrins prior to implant resulted in reduction of class II expression. Administration of protoporphyrins to the recipients of allogeneic islets also resulted in transient powerful immunosuppression with reduced lymphocyte proliferative responses, increased proportion of regulatory cells (CD4+CD25+), decreased mononuclear cell infiltrating the graft, paralleled by a systemic upregulation of HO-1 expression. All these mechanisms may have contributed to the induction of donor-specific hyporesponsiveness in a proportion of the protoporphyrintreated animals.
Collapse
Affiliation(s)
- Antonello Pileggi
- Cell Transplant Center, Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Avenue (R-134), Miami, FL 33136
| | - R. Damaris Molano
- Cell Transplant Center, Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Avenue (R-134), Miami, FL 33136
| | - Thierry Berney
- Cell Transplant Center, Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Avenue (R-134), Miami, FL 33136
| | - Hirohito Ichii
- Cell Transplant Center, Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Avenue (R-134), Miami, FL 33136
| | - Sergio San Jose
- Cell Transplant Center, Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Avenue (R-134), Miami, FL 33136
| | - Elsie Zahr
- Cell Transplant Center, Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Avenue (R-134), Miami, FL 33136
| | - Raffaella Poggioli
- Cell Transplant Center, Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Avenue (R-134), Miami, FL 33136
| | - Elina Linetsky
- Cell Transplant Center, Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Avenue (R-134), Miami, FL 33136
| | - Camillo Ricordi
- Cell Transplant Center, Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Avenue (R-134), Miami, FL 33136
| | - Luca Inverardi
- Cell Transplant Center, Diabetes Research Institute, University of Miami School of Medicine, 1450 NW 10th Avenue (R-134), Miami, FL 33136
| |
Collapse
|
9
|
von Seth E, Nyqvist D, Andersson A, Carlsson PO, Köhler M, Mattsson G, Nordin A, Berggren PO, Jansson L. Distribution of Intraportally Implanted Microspheres and Fluorescent Islets in Mice. Cell Transplant 2017; 16:621-7. [PMID: 17912953 DOI: 10.3727/000000007783465055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The aim of the study was to evaluate the distribution of intraportally transplanted islets in mice. We initially administered 2000 polystyrene microspheres with a diameter of 50 μm intraportally into normoglycemic C57BL/6 mice. In separate experiments other mice were injected similarly with 300 microspheres each with a diameter of 100 or 200 μm. One week later the animals were killed, and the lungs and livers were removed and divided into lobes. The number of microspheres in each individual liver lobe and in the lungs was counted using a stereomicroscope. In other experiments, athymic C57BL/6 mice were similarly implanted with 250 islets isolated from transgenic mice expressing the enhanced yellow fluorescent protein in the islet cells. The distribution of microspheres and islets was independent of size, and fairly homogenous within the liver, with the exception of the caudate lobe, which contained fewer microspheres and islets, respectively. Approximately one third of all microspheres and islets were present as aggregates. Eighty-five to 90% of the implanted microspheres were identified in the liver sections, whereas 60–65% of the implanted islets were recovered. Aggregates or single fluorescent cells were observed in the liver of islet-implanted mice. We conclude that islets and microspheres implanted into the liver distribute fairly homogenously and quite a few of them exist as aggregates or, with respect to islets, as fragments.
Collapse
Affiliation(s)
- Erik von Seth
- Department of Medical Cell Biology, Uppsala University, SE-75123 Uppsala, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Kosinová L, Cahová M, Fábryová E, Týcová I, Koblas T, Leontovyč I, Saudek F, Kříž J. Unstable Expression of Commonly Used Reference Genes in Rat Pancreatic Islets Early after Isolation Affects Results of Gene Expression Studies. PLoS One 2016; 11:e0152664. [PMID: 27035827 PMCID: PMC4817981 DOI: 10.1371/journal.pone.0152664] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 03/17/2016] [Indexed: 12/15/2022] Open
Abstract
The use of RT-qPCR provides a powerful tool for gene expression studies; however, the proper interpretation of the obtained data is crucially dependent on accurate normalization based on stable reference genes. Recently, strong evidence has been shown indicating that the expression of many commonly used reference genes may vary significantly due to diverse experimental conditions. The isolation of pancreatic islets is a complicated procedure which creates severe mechanical and metabolic stress leading possibly to cellular damage and alteration of gene expression. Despite of this, freshly isolated islets frequently serve as a control in various gene expression and intervention studies. The aim of our study was to determine expression of 16 candidate reference genes and one gene of interest (F3) in isolated rat pancreatic islets during short-term cultivation in order to find a suitable endogenous control for gene expression studies. We compared the expression stability of the most commonly used reference genes and evaluated the reliability of relative and absolute quantification using RT-qPCR during 0-120 hrs after isolation. In freshly isolated islets, the expression of all tested genes was markedly depressed and it increased several times throughout the first 48 hrs of cultivation. We observed significant variability among samples at 0 and 24 hrs but substantial stabilization from 48 hrs onwards. During the first 48 hrs, relative quantification failed to reflect the real changes in respective mRNA concentrations while in the interval 48-120 hrs, the relative expression generally paralleled the results determined by absolute quantification. Thus, our data call into question the suitability of relative quantification for gene expression analysis in pancreatic islets during the first 48 hrs of cultivation, as the results may be significantly affected by unstable expression of reference genes. However, this method could provide reliable information from 48 hrs onwards.
Collapse
Affiliation(s)
- Lucie Kosinová
- Laboratory of Pancreatic Islets, Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Monika Cahová
- Department of Metabolism and Diabetes, Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Fábryová
- Laboratory of Pancreatic Islets, Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Irena Týcová
- Transplant Laboratory, Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Tomáš Koblas
- Laboratory of Pancreatic Islets, Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ivan Leontovyč
- Laboratory of Pancreatic Islets, Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - František Saudek
- Laboratory of Pancreatic Islets, Center of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Diabetes, Center of Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Kříž
- Department of Diabetes, Center of Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
11
|
Diabetes Is Reversed in a Murine Model by Marginal Mass Syngeneic Islet Transplantation Using a Subcutaneous Cell Pouch Device. Transplantation 2016; 99:2294-300. [PMID: 26308506 PMCID: PMC4623852 DOI: 10.1097/tp.0000000000000864] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Islet transplantation is a successful β-cell replacement therapy for selected patients with type 1 diabetes mellitus. Although high rates of early insulin independence are achieved routinely, long-term function wanes over time. Intraportal transplantation is associated with procedural risks, requires multiple donors, and does not afford routine biopsy. Stem cell technologies may require potential for retrievability, and graft removal by hepatectomy is impractical. There is a clear clinical need for an alternative, optimized transplantation site. The subcutaneous space is a potential substitute, but transplantation of islets into this site has routinely failed to reverse diabetes. However, an implanted device, which becomes prevascularized before transplantation, may alter this equation. The authors apply a new implanted subcutaneous cell pouch (CP) device in a mouse diabetes model. Mouse islets transplanted into the CP restore glycemic control with well respond to glucose challenge. CP may serve as a potential alternative to clinical intraportal islet transplantation.
Collapse
|
12
|
Hawthorne WJ, Williams L, Chew YV. Clinical Islet Isolation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 938:89-122. [PMID: 27586424 DOI: 10.1007/978-3-319-39824-2_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The overarching success of islet transplantation relies on the success in the laboratory to isolate the islets. This chapter focuses on the processes of human islet cell isolation and the ways to optimally provide islet cells for transplantation. The major improvements in regards to the choice of enzyme type, way the digested pancreas tissue is handled to best separate islets from the acinar and surrounding tissues, the various methods of purification of the islets, their subsequent culture and quality assurance to improve outcomes to culminate in safe and effective islet transplantation will be discussed. After decades of improvements, islet cell isolation and transplantation now clearly offer a safe, effective and feasible therapeutic treatment option for an increasing number of patients suffering from type 1 diabetes specifically for those with severe hypoglycaemic unawareness.
Collapse
Affiliation(s)
- Wayne J Hawthorne
- National Pancreas and Islet Transplant Laboratories, The Westmead Institute for Medical Research, Westmead, NSW, 2145, Australia. .,Department of Surgery, Westmead Clinical School, Westmead Hospital, University of Sydney, Westmead, NSW, 2145, Australia.
| | - Lindy Williams
- National Pancreas and Islet Transplant Laboratories, The Westmead Institute for Medical Research, Westmead, NSW, 2145, Australia
| | - Yi Vee Chew
- National Pancreas and Islet Transplant Laboratories, The Westmead Institute for Medical Research, Westmead, NSW, 2145, Australia
| |
Collapse
|
13
|
Pepper AR, Gala-Lopez B, Pawlick R, Merani S, Kin T, Shapiro AMJ. A prevascularized subcutaneous device-less site for islet and cellular transplantation. Nat Biotechnol 2015; 33:518-23. [PMID: 25893782 DOI: 10.1038/nbt.3211] [Citation(s) in RCA: 269] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 03/12/2015] [Indexed: 02/07/2023]
Abstract
Transplantation of donor-derived islets into the liver is a successful cellular replacement therapy for individuals with diabetes. However, the hepatic vasculature is not an optimal transplant site for several reasons, including graft attrition and the inability to retrieve or image the islets. Here we describe islet transplantation into a prevascularized, subcutaneous site created by temporary placement of a medically approved vascular access catheter. In mice with streptozotocin (STZ)-induced diabetes, transplantation of ∼500 syngeneic islets into the resulting 'device-less' space reversed diabetes in 91% of mice and maintained normoglycemia for >100 days. The approach was also effective in mice with pre-existing diabetes, in another mouse strain that mounts a more vigorous inflammatory response, and across an allogeneic barrier. These results demonstrate that transient priming of a subcutaneous site supports diabetes-reversing islet transplantation in mouse models without the need for a permanent cell-encapsulation device.
Collapse
Affiliation(s)
- Andrew R Pepper
- Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Boris Gala-Lopez
- Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Rena Pawlick
- Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Shaheed Merani
- Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Tatsuya Kin
- 1] Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada. [2] Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - A M James Shapiro
- 1] Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada. [2] Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
14
|
He S, Wang D, Wei L. Practical and critical instruction for nonhuman primate diabetic models. Transplant Proc 2014; 45:1856-65. [PMID: 23769058 DOI: 10.1016/j.transproceed.2012.11.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 11/20/2012] [Indexed: 02/05/2023]
Abstract
Diabetes mellitus, a disease of metabolic dysregulation, is characterized by inappropriate hyperglycemia resulting from progressive loss of insulin secretion or action. The potential of nonhuman primate (NHP) models in diabetes research has been well understood. NHPs have long been regarded as the "gold standard" for preclinical studies. However, there are persistent, severe obstacles to the development and application of these models. At present, a consensus for standardized strategies of diabetic induction has not been achieved. The different modeling methods of diabetes has led to various characterizations of the pathology of the disease; however, there are deficiencies of systemic evaluation programs for nonhuman primate diabetes models. In this scenario, experimental systemic programs provide the highly required guidelines for NHP diabetic models. Moreover, given the expensive and relatively small population of primates and the fatal diabetic complications, it is imperative to carefully manage the care and use of these animals in biomedical research studies. This article briefly reviews the technical and managerial aspects of NHP diabetes models providing practical and critical instruction on housing and care, routine management, development strategy, modeling diagnosis, evaluation, and disease control, as well as guidelines for model selection for various purposes. The present article sought to provide guidelines for NHP models of diabetes in their development and application. It is not intended to outline mandatory requirements for clinical accreditation.
Collapse
Affiliation(s)
- S He
- Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, and the Center for Cell Transplantation (Seventh Unit of General Surgery Department), Institute of Organ Transplantation, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, PR China
| | | | | |
Collapse
|
15
|
Hypoxia as a target for tissue specific gene therapy. J Control Release 2013; 172:484-94. [DOI: 10.1016/j.jconrel.2013.05.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 05/13/2013] [Accepted: 05/24/2013] [Indexed: 12/28/2022]
|
16
|
Zacharovová K, Berková Z, Jirák D, Herynek V, Vancová M, Dovolilová E, Saudek F. Processing of superparamagnetic iron contrast agent ferucarbotran in transplanted pancreatic islets. CONTRAST MEDIA & MOLECULAR IMAGING 2013; 7:485-93. [PMID: 22991314 DOI: 10.1002/cmmi.1477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Labeling of pancreatic islets with superparamagnetic iron oxide (SPIO) nanoparticles enables their post-transplant monitoring by magnetic resonance imaging (MRI). Although the nanoparticles are incorporated into islet cells in culture, little is known about their fate in vivo. We studied the morphology of labeled islets after transplantation, aiming to identify the MRI contrast particles and their relationship to transplantation outcomes. Rat islets labeled with the ferucarbotran were transplanted into the liver or under the kidney capsule of syngeneic and allogeneic rats. After in vivo MRI, morphology was studied by light, fluorescence and transmission electron microscopy. Morphology of syngeneic islets transplanted beneath the kidney capsule vs into the liver was similar. Iron particles were almost completely eliminated from the endocrine cells and remained located in host-derived macrophages surrounding the vital islets for the entire study period. In the allogeneic model, islets lost their function and were completely rejected within nine days following transplantation in both transplant models. However, intercellular transport of the SPIO particles and subsequent MRI findings was different in the liver and kidney. In the liver, the decreasing number of islet-related MRI spots corresponded with clearance of iron particles in rejected islets; in contrast, with renal transplants extensive iron deposits with a high effect on MRI signal persisted in phagocytic cells beneath the capsule. We conclude that MRI detection of the iron contrast agent correlates with islet survival and function in islet transplantation into the liver, while it does not correlate in the case of transplantation beneath the renal capsule.
Collapse
Affiliation(s)
- Klára Zacharovová
- Laboratory of Langerhans Islets, Centre of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
17
|
Padmasekar M, Lingwal N, Samikannu B, Chen C, Sauer H, Linn T. Exendin-4 protects hypoxic islets from oxidative stress and improves islet transplantation outcome. Endocrinology 2013; 154:1424-33. [PMID: 23471218 DOI: 10.1210/en.2012-1983] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Oxidative stress produced during pancreatic islet isolation leads to significant β-cell damage. Homeostatic cytokines secreted subsequently to islet transplantation damage β-cells by generating oxygen free radicals. In this study, exendin-4, a glucagon-like peptide-1 analog improved islet transplantation outcome by increasing the survival of diabetic recipient mice from 58% to 100%. We hypothesized that this beneficial effect was due to the ability of exendin-4 to reduce oxidative stress. Further experiments showed that it significantly reduced the apoptotic rate of cultured β-cells subjected to hypoxia or to IL-1β. Reduction of apoptotic events was confirmed in pancreatic islet grafts of exendin-4-treated mice. Exendin-4 enhanced Akt phosphorylation of β-cells and insulin released from them. It even augmented insulin secretion from islets cultivated at hypoxic conditions. Exposure to hypoxia led to a decrease in the activation of Akt, which was reversed when β-cells were pretreated with exendin-4. Moreover, exendin-4 increased the activity of redox enzymes in a hypoxia-treated β-cell line and reduced reactive oxygen species production in isolated pancreatic islets. Recovery from diabetes in mice transplanted with hypoxic islets was more efficient when they received exendin-4. In conclusion, exendin-4 rescued islets from oxidative stress caused by hypoxia or due to cytokine exposure. It improved the outcome of syngenic and xenogenic islet transplantation.
Collapse
Affiliation(s)
- M Padmasekar
- Medical Clinic and Policlinic 3, Justus Liebig University, D-35392 Giessen, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Wang P, Yigit MV, Ran C, Ross A, Wei L, Dai G, Medarova Z, Moore A. A theranostic small interfering RNA nanoprobe protects pancreatic islet grafts from adoptively transferred immune rejection. Diabetes 2012; 61:3247-54. [PMID: 22923469 PMCID: PMC3501867 DOI: 10.2337/db12-0441] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Islet transplantation has recently emerged as an acceptable clinical modality for restoring normoglycemia in patients with type 1 diabetes mellitus (T1DM). The long-term survival and function of islet grafts is compromised by immune rejection-related factors. Downregulation of factors that mediate immune rejection using RNA interference holds promise for improving islet graft resistance to damaging factors after transplantation. Here, we used a dual-purpose therapy/imaging small interfering (si)RNA magnetic nanoparticle (MN) probe that targets β(2) microglobulin (B2M), a key component of the major histocompatibility class I complex (MHC I). In addition to serving as a siRNA carrier, this MN-siB2M probe enables monitoring of graft persistence noninvasively using magnetic resonance imaging (MRI). Human islets labeled with these MNs before transplantation into B2M (null) NOD/scid mice showed significantly improved preservation of graft volume starting at 2 weeks, as determined by longitudinal MRI in an adoptive transfer model (P < 0.05). Furthermore, animals transplanted with MN-siB2M-labeled islets demonstrated a significant delay of up to 23.8 ± 4.8 days in diabetes onset after the adoptive transfer of T cells relative to 6.5 ± 4.5 days in controls. This study demonstrated that our approach could protect pancreatic islet grafts from immune rejection and could potentially be applied to allotransplantation and prevention of the autoimmune recurrence of T1DM in islet transplantation or endogenous islets.
Collapse
Affiliation(s)
- Ping Wang
- From the Molecular Imaging Laboratory, (MGH)/(MIT)/(HMS) Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and the
| | - Mehmet V. Yigit
- From the Molecular Imaging Laboratory, (MGH)/(MIT)/(HMS) Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and the
| | - Chongzhao Ran
- From the Molecular Imaging Laboratory, (MGH)/(MIT)/(HMS) Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and the
| | - Alana Ross
- From the Molecular Imaging Laboratory, (MGH)/(MIT)/(HMS) Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and the
| | - Lingling Wei
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Guangping Dai
- From the Molecular Imaging Laboratory, (MGH)/(MIT)/(HMS) Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and the
| | - Zdravka Medarova
- From the Molecular Imaging Laboratory, (MGH)/(MIT)/(HMS) Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and the
| | - Anna Moore
- From the Molecular Imaging Laboratory, (MGH)/(MIT)/(HMS) Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and the
- Corresponding author: Anna Moore,
| |
Collapse
|
19
|
Molecular imaging: a promising tool to monitor islet transplantation. J Transplant 2011; 2011:202915. [PMID: 22013504 PMCID: PMC3195545 DOI: 10.1155/2011/202915] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 07/29/2011] [Indexed: 12/18/2022] Open
Abstract
Replacement of insulin production by pancreatic islet transplantation has great potential as a therapy for type 1 diabetes mellitus. At present, the lack of an effective approach to islet grafts assessment limits the success of this treatment. The development of molecular imaging techniques has the potential to fulfill the goal of real-time noninvasive monitoring of the functional status and viability of the islet grafts. We review the application of a variety of imaging modalities for detecting endogenous and transplanted beta-cell mass. The review also explores the various molecular imaging strategies for assessing islet delivery, the metabolic effects on the islet grafts as well as detection of immunorejection. Here, we highlight the use of combined imaging and therapeutic interventions in islet transplantation and the in vivo monitoring of stem cells differentiation into insulin-producing cells.
Collapse
|
20
|
Jalili RB, Moeen Rezakhanlou A, Hosseini-Tabatabaei A, Ao Z, Warnock GL, Ghahary A. Fibroblast populated collagen matrix promotes islet survival and reduces the number of islets required for diabetes reversal. J Cell Physiol 2011; 226:1813-9. [PMID: 21506112 DOI: 10.1002/jcp.22515] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Islet transplantation represents a viable treatment for type 1 diabetes. However, due to loss of substantial mass of islets early after transplantation, islets from two or more donors are required to achieve insulin independence. Islet-extracellular matrix disengagement, which occurs during islet isolation process, leads to subsequent islet cell apoptosis and is an important contributing factor to early islet loss. In this study, we developed a fibroblast populated collagen matrix (FPCM) as a novel scaffold to improve islet cell viability and function post-transplantation. FPCM was developed by embedding fibroblasts within type-I collagen and used as scaffold for islet grafts. Viability and insulin secretory function of islets embedded within FPCM was evaluated in vitro and in a syngeneic murine islet transplantation model. Islets embedded within acellular matrix or naked islets were used as control. Islet cell survival and function was markedly improved particularly after embedding within FPCM. The composite scaffold significantly promoted islet isograft survival and reduced the critical islet mass required for diabetes reversal by half (from 200 to 100 islets per recipient). Fibroblast embedded within FPCM produced fibronectin and growth factors and induced islet cell proliferation. No evidence of fibroblast over-growth within composite grafts was noticed. These results confirm that FPCM significantly promotes islet viability and functionality, enhances engraftment of islet grafts and decreases the critical islet mass needed to reverse hyperglycemia. This promising finding offers a new approach to reducing the number of islet donors per recipient and improving islet transplant outcome.
Collapse
Affiliation(s)
- Reza B Jalili
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | |
Collapse
|
21
|
Testicular Sertoli cells influence the proliferation and immunogenicity of co-cultured endothelial cells. Biochem Biophys Res Commun 2011; 404:829-33. [DOI: 10.1016/j.bbrc.2010.12.068] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Accepted: 12/15/2010] [Indexed: 11/22/2022]
|
22
|
Li F, Mahato RI. RNA interference for improving the outcome of islet transplantation. Adv Drug Deliv Rev 2011; 63:47-68. [PMID: 21156190 DOI: 10.1016/j.addr.2010.11.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 11/19/2010] [Accepted: 11/25/2010] [Indexed: 01/06/2023]
Abstract
Islet transplantation has the potential to cure type 1 diabetes. Despite recent therapeutic success, it is still not common because a large number of transplanted islets get damaged by multiple challenges including instant blood mediated inflammatory reaction, hypoxia/reperfusion injury, inflammatory cytokines, and immune rejection. RNA interference (RNAi) is a novel strategy to selectively degrade target mRNA. The use of RNAi technologies to downregulate the expression of harmful genes has the potential to improve the outcome of islet transplantation. The aim of this review is to gain a thorough understanding of biological obstacles to islet transplantation and discuss how to overcome these barriers using different RNAi technologies. This eventually will help improve islet survival and function post transplantation. Chemically synthesized small interferring RNA (siRNA), vector based short hairpin RNA (shRNA), and their critical design elements (such as sequences, promoters, and backbone) are discussed. The application of combinatorial RNAi in islet transplantation is also discussed. Last but not the least, several delivery strategies for enhanced gene silencing are discussed, including chemical modification of siRNA, complex formation, bioconjugation, and viral vectors.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | | |
Collapse
|
23
|
Vaithilingam V, Oberholzer J, Guillemin GJ, Tuch BE. Beneficial effects of desferrioxamine on encapsulated human islets--in vitro and in vivo study. Am J Transplant 2010; 10:1961-9. [PMID: 20645943 DOI: 10.1111/j.1600-6143.2010.03209.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
As many as 2000 IEQs (islet equivalent) of encapsulated human islets are required to normalize glucose levels in diabetic mice. To reduce this number, encapsulated islets were exposed to 100 μM desferrioxamine (DFO) prior to transplantation. Cell viability, glucose-induced insulin secretion, VEGF (Vascular endothelial growth factor), HIF-1α (Hypoxia inducible factor-1 alpha), caspase-3 and caspase-8 levels were assessed after exposure to DFO for 12, 24 or 72 h. Subsequently, 1000, 750 or 500 encapsulated IEQs were infused into peritoneal cavity of diabetic mice after 24 h exposure to DFO. Neither viability nor function in vitro was affected by DFO, and levels of caspase-3 and caspase-8 were unchanged. DFO significantly enhanced VEGF secretion by 1.6- and 2.5-fold at 24 and 72 h, respectively, with a concomitant increase in HIF-1α levels. Euglycemia was achieved in 100% mice receiving 1000 preconditioned IEQs, as compared to only 36% receiving unconditioned IEQs (p < 0.001). Similarly, with 750 IEQ, euglycemia was achieved in 50% mice receiving preconditioned islets as compared to 10% receiving unconditioned islets (p = 0.049). Mice receiving preconditioned islets had lower glucose levels than those receiving unconditioned islets. In summary, DFO treatment enhances HIF-1α and VEGF expression in encapsulated human islets and improves their ability to function when transplanted.
Collapse
Affiliation(s)
- V Vaithilingam
- Department of Pharmacology, University of New South Wales, Sydney, Australia
| | | | | | | |
Collapse
|
24
|
Calcium phosphate cement chamber as an immunoisolative device for bioartificial pancreas: in vitro and preliminary in vivo study. Pancreas 2010; 39:444-51. [PMID: 20084047 DOI: 10.1097/mpa.0b013e3181be2f95] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES This study examined a calcium phosphate cement (CPC) chamber as an immunoisolative device to facilitate the use of xenogeneic cell sources without immunosuppression for the bioartificial pancreas (BAP). METHODS Mouse insulinoma cells were encapsulated in agarose gel and then enclosed in a CPC chamber to create a BAP. Bioartificial pancreas were evaluated by cell viability, live-dead cell ratio, and cytokine-mediated cytotoxicity assay and implanted into the peritoneal cavity of diabetic rats. Nonfasting blood glucose and serum insulin levels were analyzed perioperatively; BAPs were also retrieved for histological examination. RESULTS Insulinoma cells enclosed in the CPC chamber had normal viability, cell survival, and insulin secretion that was even cultured in media with cytokines. The nonfasting blood glucose level of rats was decreased from 460 +/- 50 to 132 +/- 43 mg/dL and maintained euglycemia for 22 days; serum insulin level was increased from 0.34 +/- 0.11 to 1.43 +/- 0.30 microg/dL after operation. Histological examination revealed the fibrous tissue envelopment, and immune-related cells that competed for oxygen resulting in hypoxia could be attributed to the dysfunction of BAPs. CONCLUSIONS This study proved the feasibility for using a CPC chamber as an immunoisolative device for the BAP. An alternative implanted site should be considered to extend the functional longevity of BAPs in further study.
Collapse
|
25
|
Sakata N, Chan NK, Chrisler J, Obenaus A, Hathout E. Bone marrow cells produce nerve growth factor and promote angiogenesis around transplanted islets. World J Gastroenterol 2010; 16:1215-20. [PMID: 20222164 PMCID: PMC2839173 DOI: 10.3748/wjg.v16.i10.1215] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To clarify the mechanism by which bone marrow cells promote angiogenesis around transplanted islets.
METHODS: Streptozotocin induced diabetic BALB/c mice were transplanted syngeneically under the kidney capsule with the following: (1) 200 islets (islet group: n = 12), (2) 1-5 × 106 bone marrow cells (bone marrow group: n = 11), (3) 200 islets and 1-5 × 106 bone marrow cells (islet + bone marrow group: n = 13), or (4) no cells (sham group: n = 5). All mice were evaluated for blood glucose, serum insulin, serum nerve growth factor (NGF) and glucose tolerance (GTT) up to postoperative day (POD) 14. Histological assessment for insulin, von Willebrand factor (vWF) and NGF was performed at POD 3, 7 and 14.
RESULTS: Blood glucose level was lowest and serum insulin was highest in the islet + bone marrow group. Serum NGF increased in islet, bone marrow, and islet + bone marrow groups after transplantation, and there was a significant difference (P = 0.0496, ANOVA) between the bone marrow and sham groups. The number of vessels within the graft area was significantly increased in both the bone marrow and islet + bone marrow groups at POD 14 as compared to the islet alone group (21.2 ± 3.6 in bone marrow, P = 0.01, vs islet group, 22.6 ± 1.9 in islet + bone marrow, P = 0.0003, vs islet group, 5.3 ± 1.6 in islet-alone transplants). NGF was more strongly expressed in bone marrow cells compared with islets.
CONCLUSION: Bone marrow cells produce NGF and promote angiogenesis. Islet co-transplantation with bone marrow is associated with improvement of islet graft function.
Collapse
|
26
|
Lyle DB, Shallcross JC, Langone JJ. Sensitivity of insulin production from encapsulated islets to endotoxin-stimulated macrophage inflammatory mediators. J Biomed Mater Res A 2009; 91:1221-38. [DOI: 10.1002/jbm.a.32351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
27
|
Kim SK, Kim JY, Choi YS, Lee MK, Lee BW, Lee HC, Lee EJ. A dual-reporter system for specific tracing of pancreatic ß-cell lines that non-invasively measures viable in vivo islet cells. Biotechnol Lett 2009; 32:53-7. [DOI: 10.1007/s10529-009-0113-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 08/14/2009] [Accepted: 08/14/2009] [Indexed: 11/28/2022]
|
28
|
Abstract
Sensitivity of pancreatic islets to hypoxia is one of the most important of the obstacles responsible for their failure to survive within the recipients. The aim of this study was to compare the in vitro hypoxia tolerance of neonatal and adult rat islet cells and to study the glucose metabolism in these cells after exposure to hypoxia. Islet cells from both age categories were cultured in different hypoxic levels for 24 h and insulin secretion and some metabolites of glucose metabolism were analysed. Glucose-stimulated insulin secretion decreased dramatically in both cell preparations in response to the decrease in oxygen level. The reduction of insulin secretion was more detectable in adult cells and started at 5% O(2), while a significant reduction was obtained at 1% O(2) in neonatal cells. Moreover, basal insulin release of neonatal cells showed an adaptation to hypoxia after a 4-day culture in hypoxia. Intracellular pyruvate was higher in neonatal cells than in adult ones, while no difference in lactate level was observed between them. Similar results to that of pyruvate were observed for adenosine triphosphate (ATP) and the second messenger cyclic adenosine monophosphate (cAMP). The study reveals that neonatal rat islet cells are more hypoxia-tolerant than the adult ones. The most obvious metabolic observation was that both pyruvate and lactate were actively produced in neonatal cells, while adult cells depended mainly on lactate production as an end-product of glycolysis, indicating a more enhanced metabolic flexibility of neonatal cells to utilize the available oxygen and, at the same time, maintain metabolism anaerobically.
Collapse
Affiliation(s)
- Ayman Hyder
- Department of Physiology and Biochemistry of Nutrition, Max Rubner Institute, and Clinical Research Center, Innovation and Technology centre, Kiel, Germany
| | | | | |
Collapse
|
29
|
Abstract
The development of new methods for noninvasive imaging is an area of biotechnology that is of great relevance for the diagnosis and characterization of diabetes mellitus. Noninvasive imaging can be used to study the dynamics of beta-cell mass and function; beta-cell death; vascularity, innervation and autoimmune attack of pancreatic islets; and the efficacy of islet transplantation to remedy beta-cell loss in patients with diabetes mellitus. In this Review, we focus on the application of MRI for monitoring islet transplantation and on the potential causes of islet graft failure, which are still poorly understood. Questions that have been addressed by MRI studies encompass graft longevity, and the effects of immune rejection, glucose toxic effects, and the transplanted islets' purity on graft fate. We also highlight novel technologies for simultaneous imaging and delivery of experimental therapies that aim to extend the lifespan and functionality of islet grafts. On the basis of this evidence, MRI represents a valuable platform for a thorough investigation of beta-cell function in the context of islet transplantation. State-of-the-art multimodality approaches, such as PET-MRI, can extend our current capabilities and help answer the critical questions that currently inhibit the prevention and cure of diabetes mellitus.
Collapse
Affiliation(s)
- Zdravka Medarova
- Molecular Imaging Laboratory, Massachusetts General Hospital-Massachusetts Institute of Technology-Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, 13th Street, Charlestown, MA 02129, USA
| | | |
Collapse
|
30
|
Perez-Basterrechea M, Briones RM, Alvarez-Viejo M, Garcia-Perez E, Esteban MM, Garcia V, Obaya AJ, Barneo L, Meana A, Otero J. Plasma-fibroblast gel as scaffold for islet transplantation. Tissue Eng Part A 2009; 15:569-77. [PMID: 18694292 DOI: 10.1089/ten.tea.2008.0088] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The transplant of pancreatic islets into the liver can restore normal blood glucose levels in patients with type I diabetes. However, long-term results have indicated that the site and method of transplantation still need to be optimized to improve islet engraftment. This study was designed to assess the efficiency of the use of clotted blood plasma containing fibroblasts ("plasma-fibroblast gel") as a scaffold for subcutaneous islet transplantation in diabetic athymic mice. Islets embedded in the plasma-fibroblast gel were able to resolve hyperglycemia in transplanted mice, restoring normoglycemia over a 60-day period and allowing gradual body weight recovery. Glucose clearances were significantly improved when compared to those recorded in diabetic animals and similar to those observed in the control group (free islets transplanted beneath the kidney capsule). Histological evaluation revealed functional islets within a subcutaneous tissue rich in collagen fibers that was well vascularized, with blood vessels observed around and inside the islets. These findings suggest that this approach could be used as an alternative option for the treatment of type I diabetes in human clinical practice.
Collapse
|
31
|
Eliaschewitz FG, Franco DR, Mares-Guia TR, Noronha IL, Labriola L, Sogayar MC. Transplante de ilhotas na prática clínica: estado atual e perspectivas. ACTA ACUST UNITED AC 2009; 53:15-23. [DOI: 10.1590/s0004-27302009000100004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Accepted: 11/19/2008] [Indexed: 02/08/2023]
Abstract
O transplante de ilhotas é um procedimento em desenvolvimento, como alternativa para o tratamento do diabetes tipo 1 que está na fronteira entre o experimental e o clínico. É uma terapia celular na qual as células são implantadas em território diferente do fisiológico em que apenas determinado número incerto conseguirá se adaptar. Aperfeiçoar este processo para obter os mesmos resultados que no transplante de pâncreas, representa um desafio para o qual convergem contribuições da biologia celular, da imunologia e das técnicas de laboratório que se entrelaçam de maneira extremamente complexa. Este trabalho revisa a literatura expondo a evolução do procedimento, a sua metodologia atual e os resultados clínicos obtidos. As perspectivas futuras do transplante diante dos recentes avanços também são discutidas.
Collapse
Affiliation(s)
- Freddy Goldberg Eliaschewitz
- Universidade de São Paulo; Centro de Pesquisa Clínica; Hospital Heliópolis; notre-Dame intermédica Sistema de Saúde
| | - Denise Reis Franco
- Universidade de São Paulo; Centro de Pesquisa Clínica; Hospital Heliópolis
| | | | | | | | | |
Collapse
|
32
|
Jiao Y, Peng ZH, Xing TH, Qin J, Zhong CP. Assessment of islet graft survival using a 3.0-Tesla magnetic resonance scanner. Anat Rec (Hoboken) 2009; 291:1684-92. [PMID: 18951510 DOI: 10.1002/ar.20770] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Some studies have recently described a magnetic resonance (MR) method for detection of iron-labeled islets transplanted into the liver. The aim of this work was to assess the survival of islet graft using a clinical 3.0-T scanner. Islets from Lewis rats were cultured in the presence of iron oxide nanoparticles. One thousand iron-labeled islets were transplanted into the portal vein of diabetic rats. Blood glucose levels were measured daily through day 14 post-transplantation. MR imaging of the same section of the liver was performed on 1, 3, 7, 10, and 14 days post-transplantation. The labeled islets were visualized by MR as distinct hypointensive spots distributed in the liver. There was a linear correlation between the relative value of delta R2* relaxometry multiplied by the cubic diameter (relative value of the iron volume, Ir) and blood glucose level on 14 days post-transplantation in allograft and isograft (P<0.05). The relative value of delta R2* relaxometry, diameter, and number of hypointensive spots could be calculated to assess the survival of the iron-labeled islet grafts. Assessment of iron-labeled islet grafts using a clinical 3.0-T magnetic resonance scanner represents a useful method that has potential for clinical use.
Collapse
Affiliation(s)
- Yang Jiao
- Shanghai Organ Transplantation Clinical Medical Center, the First People's Hospital affiliated Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | |
Collapse
|
33
|
Fenjves ES, Ochoa MS, Cechin S, Gay-Rabinstein C, Pérez-Alvarez I, Ichii H, Mendez A, Ricordi C, Curran MA. Protection of human pancreatic islets using a lentiviral vector expressing two genes: cFLIP and GFP. Cell Transplant 2008; 17:793-802. [PMID: 19044206 DOI: 10.3727/096368908786516828] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pancreatic islet transplantation can provide insulin independence to diabetic patients. However, apoptosis of islets often leads to early graft failure. Genetic engineering with protective gene(s) can improve the viability of these cells. Here we show successful transduction of human islets with a feline immunodeficiency virus (FIV) vector expressing both a cytoprotective (cFLIP) gene and the green fluorescent protein (GFP). Despite using low virus titers to maximize safety, transduced islets expressed both genes, resulting in improved beta-cell metabolic activity and viability. Although only approximately 10% of total islet cells were transduced, the significant viability advantages suggest a "barrier" effect in which protecting the periphery of the islet shields the core. These results provide the first demonstration that a lentiviral vector can express two genes in islets. Furthermore, the engineered islets are resistant to a variety of apoptotic stimuli, suggesting the potential of this approach in enhancing the viability of transplanted cells.
Collapse
Affiliation(s)
- Elizabeth S Fenjves
- Diabetes Research Institute, University of Miami School of Medicine, Miami, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yuan Y, Cong C, Zhang J, Wei L, Li S, Chen Y, Tan W, Cheng J, Li Y, Zhao X, Lu Y. Self-assembling peptide nanofiber as potential substrates in islet transplantation. Transplant Proc 2008; 40:2571-4. [PMID: 18929804 DOI: 10.1016/j.transproceed.2008.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hypoxia and reoxygenation (H/R)-induced damage often happens soon after islets are transplantation. The process of islet isolation and purification causes the rapid onset of hypoxia. We sought to develop a functional scaffold to sustain the structure and function of islets as well as to recover some of the surface molecules damaged during isolation, seeking to improve islet transplantation outcomes. Self-assembling peptide nanofiber (SAPNF), a new type of substrate has been shown to be an excellent biological material for neuronal cell culture and tissue engineering in animals. In this study, we investigated the protective effect of SAPNF on damage to rat islets. Freshly prepared rat islets from male Sprague-Dawley rats were seeded in plates coated with (SAPNF-treated group) or without (control group) SAPNF. The islets were then divided into two groups culture under normoxia for 7 days versus exposure to hypoxia (< 1% O2) for 6 hours followed by reoxygenation for 24 hours. The results showed that SAPNF exhibited improving effects on viability and function of cultured islets, protecting the one from H/R-induced damage. In both groups, the stimulation index of SAPNF-treated groups were about two times the controls. SAPNF treatment decreased apoptotic rates of islet cells. These results suggested the usefulness of SAPNF to maintain the viability and function of rat pancreatic islets. SAPNF may be a potential scaffold for clinical islet transplantation.
Collapse
Affiliation(s)
- Y Yuan
- Key Lab of Transplant Engineering and Immunology, Ministry of Health, P R China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Type 1 diabetes (insulin-dependent, IDDM) results in immune-mediated destruction of pancreatic beta cells, which leads to a deficiency in insulin secretion and as a result, to hyperglycaemia. Keeping blood glucose levels under tight control represents the most effective way either to prevent the onset or to reduce the progression of the chronic complications of IDDM. At present, pancreatic islet transplantation is emerging as the most promising clinical modality, which can stop diabetes progression without increasing the incidence of hypoglycaemic events. Although early results of clinical trials using the Edmonton Protocol and its variations are very encouraging, it is still unclear how long the islets will survive and how often the transplantation procedure will be successful. In order to monitor transplantation efficiency and graft survival, reliable non-invasive imaging methods are critically needed. If such methods are introduced clinically, essential information regarding the location, function and viability of transplanted islets can be obtained repeatedly and non-invasively. This review will focus on the latest advancements in the field of in vivo imaging of islet transplantation and describe various islet labelling and imaging techniques. In addition, we will critically look into limitations and obstacles currently present on the way to successful clinical implementation of this approach.
Collapse
Affiliation(s)
- Z Medarova
- Molecular Imaging Program, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
| | | |
Collapse
|
36
|
Caiazzo R, Gmyr V, Hubert T, Delalleau N, Lamberts R, Moerman E, Kerr-Conte J, Pattou F. Evaluation of alternative sites for islet transplantation in the minipig: interest and limits of the gastric submucosa. Transplant Proc 2007; 39:2620-3. [PMID: 17954193 DOI: 10.1016/j.transproceed.2007.08.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Since the introduction of glucocorticoid-free immunosuppressive regimens, islet transplantation offers a less invasive alternative to pancreas transplantation. However, complications associated with intraportal islet injection and the progressive functional decline of intrahepatic islets encourage the exploration of alternative sites. Herein we evaluated, in the minipig, the use of the gastric submucosa (GS; group 1, n = 5) for islet transplantation compared with the kidney capsule (KC; group 2, n = 5). Subsequently we attempted to improve the vascularization of the submucosal graft (group 3, n = 5) by the addition of an extracellular matrix rich in growth factors (Matrigel). One month after grafting, we evaluated transplanted islet function in vivo and in vitro. Our study showed better function of islets engrafted in the GS than in the KC (P < .05). Despite the growth factors, Matrigel did not offer a more suitable environment to further improve engraftment (group 3, P < .05). Thus, even if the liver remains the gold standard, the GS represents a potential islet engraftment site, confirming the data obtained in vitro and in the rodent. Offering easy access by endoscopy, this site could constitute an interesting alternative for experimental studies in large mammals and, eventually, for clinical application.
Collapse
Affiliation(s)
- R Caiazzo
- INSERM UNIT-M 859, Diabetes Cell Therapy, Faculty of Medicine, Lille 2 University, 1 Place de Verdun, 59045 Lille, France.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Immunologic and nonimmunologic events lead to significant graft loss after islet transplantation. Unfortunately, current metabolic testing methods are inadequate to detect many of these changes, leading to a critical need for noninvasive monitoring of islet rejection. However, their small size and distribution after transplantation pose specific problems for direct islet imaging. This article reviews the relative merits of several imaging modalities for the noninvasive monitoring of islet transplantation and rejection.
Collapse
Affiliation(s)
- Jason L Gaglia
- Department of Medicine, Harvard Medical School, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA.
| |
Collapse
|
38
|
Medarova Z, Evgenov NV, Dai G, Bonner-Weir S, Moore A. In vivo multimodal imaging of transplanted pancreatic islets. Nat Protoc 2007; 1:429-35. [PMID: 17406265 DOI: 10.1038/nprot.2006.63] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Interest is increasing in the transplantation of pancreatic islets as a means to achieve insulin independence in individuals with type I diabetes. The success of this approach is hampered by the absence of methods to follow the fate of transplanted islets non-invasively. In vivo imaging seems to be the most appropriate technique to achieve this goal in small animals and eventually in humans. Here we describe a protocol for labeling and subsequent imaging of transplanted islets in vivo using magnetic resonance imaging (MRI) and optical imaging. The whole series of experiments can be carried out in roughly 48 h. We believe that our approach can significantly advance the current ability to determine islet distribution, and possibly survival, after transplantation. This information would be essential not only for the long-term monitoring of graft function but also for the design of improved transplantation and immunomodulatory methods.
Collapse
Affiliation(s)
- Zdravka Medarova
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, USA
| | | | | | | | | |
Collapse
|
39
|
Emamaullee JA, Shapiro AMJ. Factors influencing the loss of beta-cell mass in islet transplantation. Cell Transplant 2007; 16:1-8. [PMID: 17436849 DOI: 10.3727/000000007783464461] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent advances in clinical islet transplantation have clearly demonstrated that this procedure can provide excellent glycemic control and often insulin independence in a population of patients with type 1 diabetes. A key limitation in the widespread application of clinical islet transplantation is the requirement of 10,000 islet equivalents/kg in most recipients, generally derived from two or more cadaveric donors. It has been determined that a majority of the transplanted islets fail to engraft and become fully functional. In this review article, the factors that contribute to this early loss of islets following transplantation are discussed in depth.
Collapse
|
40
|
Biancone L, Crich SG, Cantaluppi V, Romanazzi GM, Russo S, Scalabrino E, Esposito G, Figliolini F, Beltramo S, Perin PC, Segoloni GP, Aime S, Camussi G. Magnetic resonance imaging of gadolinium-labeled pancreatic islets for experimental transplantation. NMR IN BIOMEDICINE 2007; 20:40-8. [PMID: 16986104 DOI: 10.1002/nbm.1088] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
New imaging techniques that couple anatomical resolution to sensitivity may greatly contribute to improving islet transplantation. In the present work, a report is given of the direct detection of islets by magnetic resonance imaging (MRI) after ex vivo cell labeling with the MRI T(1) contrast agent GdHPDO3A. Experiments on mouse and human islets demonstrated well-tolerated uptake of GdHPDO3A, based on morphology, viability, glucose-dependent insulin response and apoptosis/toxicity gene array profile. GdHPDO3A loading was sufficient for in vitro MRI cell detection. In vivo isotransplanted mouse islets into the kidney capsule and xenotransplanted human islets within the mouse liver were detected. Imaging specificity was supported by the absence of signal in unlabeled islet transplants, its persistence upon using fat-suppression MRI protocols and the colocalization with the transplanted islets. In conclusion, direct islet imaging with high spatial and contrast resolution after labeling with GdHPDO3A is demonstrated, allowing visualization of kidney subcapsular mouse islet grafts and intrahepatic human islet xenografts.
Collapse
Affiliation(s)
- Luigi Biancone
- Department of Internal Medicine and Research Center for Experimental Medicine (CeRMS), University of Turin, Corso Dogliotti 14, Turin 10126, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The Edmonton trials have brought about a marked improvement in the short-term rate of success of islet transplantation with rates of insulin-independence of 80% at 1-year being reported by several institutions worldwide. Unfortunately, this rate consistently decreases to 10-15% by 5 years post-transplantation. Several mechanisms have been proposed to explain this apparent 'islet exhaustion', but are difficult to pinpoint in a given patient. Understanding the reasons for islet graft exhaustion and its kinetics is a prerequisite for the improvement of islet transplantation outcome. In this regard, efficient monitoring tools for the islet graft have been conspicuously lacking and are required to detect islet damage and diagnose its mechanisms in a timely fashion, so as to initiate salvage therapy such as antirejection treatment. Tools for the monitoring of the islet graft include follow-up of metabolic function but mostly indicate dysfunction when it is too late to take action. Progress is likely to arise in the fields of immune monitoring, molecular monitoring and islet imaging, notably thanks to magnetic resonance (MR) or positron emission tomography (PET) technologies.
Collapse
Affiliation(s)
- T Berney
- Cell Isolation and Transplantation Center, Division of Visceral and Transplant Surgery, Geneva University Hospitals, Geneva, Switzerland.
| | | |
Collapse
|
42
|
Emamaullee JA, Shapiro AMJ, Rajotte RV, Korbutt G, Elliott JF. Neonatal porcine islets exhibit natural resistance to hypoxia-induced apoptosis. Transplantation 2006; 82:945-52. [PMID: 17038911 DOI: 10.1097/01.tp.0000238677.00750.32] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Despite the success of the Edmonton protocol for human islet transplantation, an alternate source of islet tissue must be developed if beta-cell replacement therapy is to see widespread application. Neonatal porcine islets (NPI) represent one potential source of tissue. When human or rodent islets are transplanted, the majority of cells undergo hypoxia-induce apoptosis soon after the grafts are placed in the recipient. In the present study, we investigated whether NPI were similarly sensitive to hypoxia. METHODS NPI were exposed to hypoxia and hypoxia/reoxygenation using an in vitro hypoxic chamber. Afterwards, viability, frequency of apoptosis, and beta-cell function were evaluated. NPI and adult porcine islets were transplanted into chemically diabetic, immunodeficient mice and graft apoptosis was assessed 24 hours and seven days posttransplant. RESULTS NPI demonstrated a remarkable capacity to resist apoptosis and maintain insulin secretion despite severe stresses such as hypoxia/reoxygenation. One day after transplantation, NPI grafts showed limited apoptosis, confined to rare strongly insulin positive cells. In contrast, adult porcine islet grafts underwent widespread apoptosis. Western blotting revealed that NPI express high levels of at least one potent endogenous antiapoptotic protein (XIAP). CONCLUSIONS The majority of cells within transplanted human islets undergo apoptosis soon after portal infusion. In contrast, NPI have the capacity to resist this early posttransplant apoptosis, with likely reduced antigen release and diminished immune stimulation. NPI appear to contain a population of insulin-low to insulin-negative pre-beta-cells, which are resistant to hypoxia-induced apoptosis and still capable of differentiating into mature beta-cells.
Collapse
Affiliation(s)
- Juliet A Emamaullee
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
43
|
Miao G, Ostrowski RP, Mace J, Hough J, Hopper A, Peverini R, Chinnock R, Zhang J, Hathout E. Dynamic production of hypoxia-inducible factor-1alpha in early transplanted islets. Am J Transplant 2006; 6:2636-43. [PMID: 17049056 DOI: 10.1111/j.1600-6143.2006.01541.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
More than half of transplanted beta-cells undergo apoptotic cell death triggered by nonimmunological factors within a few days after transplantation. To investigate the dynamic hypoxic responses in early transplanted islets, syngeneic islets were transplanted under the kidney capsule of balb/c mice. Hypoxia-inducible factor-1alpha (HIF-1alpha) was strongly expressed at post-transplant day (POD) 1, increased on POD 3, and gradually diminished on POD 14. Insulin secretion decreased on POD 3 in association with a significant increase of HIF-1alpha-related beta-cell death, which can be suppressed by short-term hyperbaric oxygen therapy. On POD 7, apoptosis was not further activated by continually produced HIF-1alpha. In contrast, improvement of nerve growth factor and duodenal homeobox factor-1 (PDx-1) production resulted in islet graft recovery and remodeling. In addition, significant activation of vascular endothelial growth factor in islet grafts on POD 7 correlated with development of massive newly formed microvessels, whose maturation is advanced on POD 14 with gradual diminution of HIF-1alpha. We conclude that (1) transplanted islets strongly express HIF-1alpha in association with beta-cell death and decreased insulin production until adequate revascularization is established and (2) early suppression of HIF-1alpha results in less beta-cell death thereby minimizing early graft failure.
Collapse
Affiliation(s)
- G Miao
- Islet Transplant Laboratory, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Cantaluppi V, Biancone L, Romanazzi GM, Figliolini F, Beltramo S, Ninniri MS, Galimi F, Romagnoli R, Franchello A, Salizzoni M, Perin PC, Ricordi C, Segoloni GP, Camussi G. Antiangiogenic and immunomodulatory effects of rapamycin on islet endothelium: relevance for islet transplantation. Am J Transplant 2006; 6:2601-11. [PMID: 16981910 DOI: 10.1111/j.1600-6143.2006.01534.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Donor intra-islet endothelial cells contribute to neovascularization after transplantation. Several factors may interfere with this process and ultimately influence islet engraftment. Rapamycin, a central immunosuppressant in islet transplantation, is an mTOR inhibitor that has been shown to inhibit cancer angiogenesis. The aim of this study was to evaluate the effects of rapamycin on islet endothelium. Rapamycin inhibited the outgrowth of endothelial cells from freshly purified human islets and the formation of capillary-like structures in vitro and in vivo after subcutaneous injection within Matrigel plugs into SCID mice. Rapamycin decreased migration, proliferation and angiogenic properties of human and mouse islet-derived endothelial cell lines with appearance of apoptosis. The expression of angiogenesis-related factors VEGF, alphaVbeta3 integrin and thrombospondin-1 on islet endothelium was altered in the presence of rapamycin. On the other hand, rapamycin decreased the surface expression of molecules involved in immune processes such as ICAM-1 and CD40 and reduced the adhesion of T cells to islet endothelium. Our results suggest that rapamycin exerts dual effects on islet endothelium inducing a simultaneous inhibition of angiogenesis and a down-regulation of receptors involved in lymphocyte adhesion and activation.
Collapse
Affiliation(s)
- V Cantaluppi
- Department of Internal Medicine and Research Centre for Experimental Medicine (CeRMS), University of Torino, Corso Dogliotti 14, Torino 10126, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
DM (diabetes mellitus) is a metabolic disorder of either absolute or relative insulin deficiency. Optimized insulin injections remain the mainstay life-sustaining therapy for patients with T1DM (Type I DM) in 2006; however, a small subset of patients with T1DM (approx. 10%) are exquisitely sensitive to insulin and lack counter-regulatory measures, putting them at higher risk of neuroglycopenia. One alternative strategy to injected insulin therapy is pancreatic islet transplantation. Islet transplantation came of age when Paul E. Lacy successfully reversed chemical diabetes in rodent models in 1972. In a landmark study published in 2000, Shapiro et al. [A. M. Shapiro, J. R. Lakey, E. A. Ryan, G. S. Korbutt, E. Toth, G. L. Warnock, N. M. Kneteman and R. V. Rajotte (2000) N. Engl. J. Med. 343, 230-238] reported seven consecutive patients treated with islet transplants under the Edmonton protocol, all of whom maintained insulin independence out to 1 year. Substantial progress has occurred in aspects of pancreas procurement, transportation (using the oxygenated two-layer method) and in islet isolation (with controlled enzymatic perfusion and subsequent digestion in the Ricordi chamber). Clinical protocols to optimize islet survival and function post-transplantation improved dramatically with the introduction of the Edmonton protocol, but it is clear that this approach still has potential limitations. Newer pharmacotherapies and interventions designed to promote islet survival, prevent apoptosis, to promote islet growth and to protect islets in the long run from immunological injury are rapidly approaching clinical trials, and it seems likely that clinical outcomes of islet transplantation will continue to improve at the current exponential pace.
Collapse
Affiliation(s)
- Shaheed Merani
- Clinical Islet Transplant Program, University of Alberta, Roberts Centre, 2000 College Plaza, Edmonton, Alberta, Canada T6G 2C8
| | | |
Collapse
|
46
|
Berney T, Mamin A, James Shapiro AM, Ritz-Laser B, Brulhart MC, Toso C, Demuylder-Mischler S, Armanet M, Baertschiger R, Wojtusciszyn A, Benhamou PY, Bosco D, Morel P, Philippe J. Detection of insulin mRNA in the peripheral blood after human islet transplantion predicts deterioration of metabolic control. Am J Transplant 2006; 6:1704-11. [PMID: 16827874 DOI: 10.1111/j.1600-6143.2006.01373.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Recent updates of the Edmonton trial have shown that insulin independence is progressively lost in approximately 90% of islet transplant recipients over the first 5 years. Early prediction of islet graft injury could prompt the implementation of strategies attempting to salvage the transplanted islets. We hypothesize that islet damage is associated with the release and detection of insulin mRNA in the circulating blood. Whole blood samples were prospectively taken from 19 patients with type 1 diabetes receiving 31 islet transplants, immediately prior to transplantation and at regular time-points thereafter. After RNA extraction, levels of insulin mRNA were determined by quantitative reverse tran-scriptase-polymerase chain reaction. All patients exhibited a primary peak of insulin mRNA immediately after transplantation, without correlation of duration and amplitude with graft size or outcome. Twenty-five subsequent peaks were observed during the follow-up of 17 transplantations. Fourteen secondary peaks (56%) were closely followed by events related to islet graft function. Duration and amplitude of peaks were higher when they heralded occurrence of an adverse event. Peaks of insulin mRNA can be detected and are often associated with alterations of islet graft function. These data suggest that insulin mRNA detection in the peripheral blood is a promising method for the prediction of islet graft damage.
Collapse
Affiliation(s)
- T Berney
- Division of Visceral and Transplant Surgery, Geneva University Hospitals, Geneva, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Berkova Z, Kriz J, Girman P, Zacharovova K, Koblas T, Dovolilova E, Saudek F. Vitality of pancreatic islets labeled for magnetic resonance imaging with iron particles. Transplant Proc 2006; 37:3496-8. [PMID: 16298640 DOI: 10.1016/j.transproceed.2005.09.052] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We previously described an in vivo method for pancreatic islet visualization using magnetic resonance imaging with the aid of superparamagnetic nanoparticles of iron oxide (Resovist) or by magnetic beads precoated with antibodies (Dynabeads). The aim of this study was to investigate the in vitro effect of islet labeling on their quality. Isolated rat islets were cultivated for 48 hours with a contrast agent or, in the case of magnetic antibody-coated beads, for only 2 hours. The ability to secrete insulin was tested by a static insulin release assay and the results were expressed as a stimulation index. Staining with propidium iodide and acridine orange was performed to determine the ratio of live to dead cells. Stimulation indices in the Resovist islets (n = 23) vs controls (n = 14) were 15.3 and 15.0, respectively, and in the Dynabeads islets (n = 15) vs controls (n = 12) 21.3 and 19.9, respectively. The vitality of the Resovist islets vs controls determined by live/dead cells ratio was 90.8% and 91.1%, respectively (n = 20), and in the Dynabeads islets vs controls was 89.4% and 91.8%, respectively (n = 11). Islet labeling with the contrast agent as well as with specific antibodies with iron beads did not change the vitality and insulin-secreting capacity assessed in vitro (P > .05). Magnetic resonance using iron nanoparticles represents the only method for in-vivo visualization of transplanted islets so far. Our data represent an important contribution for its clinical use.
Collapse
Affiliation(s)
- Z Berkova
- Laboratory of Pancreatic Islets, Institute for Clinical and Experimental Medicine, Center for Cell Therapy and Tissue Repair, Videnska 1958/9, 140-21 Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
48
|
Biancone L, Cantaluppi V, Romanazzi GM, Russo S, Figliolini F, Beltramo S, Scalabrino E, Deregibus MC, Romagnoli R, Franchello A, Salizzoni M, Perin PC, Ricordi C, Segoloni GP, Camussi G. Platelet-Activating Factor Synthesis and Response on Pancreatic Islet Endothelial Cells: Relevance for Islet Transplantation. Transplantation 2006; 81:511-8. [PMID: 16495796 DOI: 10.1097/01.tp.0000200306.51689.f2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Recent data suggest that donor intraislet endothelial cells may survive islet transplantation and participate to the events that influence islet engraftment. However, the mechanisms that regulate islet endothelial behavior in this setting are poorly known. METHODS We obtained immortalized human (hIECs) and mouse (mIECs) islet endothelial cells by transfection with SV40-T-large antigen and studied the synthesis and response to Platelet-activating factor (PAF), a multipotent phospholipid that acts as endothelial mediator of both inflammation and angiogenesis. RESULTS HIECs showed typical endothelial markers such as expression of vWF, CD31, and CD105, uptake of acetylated-LDL and binding to ULE-A lectin. Moreover, they expressed nestin, the PAF-receptor and possess surface fenestrations and in vitro angiogenic ability of forming tubular structures on Matrigel. Likewise, mIECs showed expression of vWF, CD31, nestin, PAF-receptor and CD105, and uptake of acetylated-LDL. HIECs and mIECs rapidly produced PAF under stimulation with thrombin in a dose-dependent way. Exogenous PAF or thrombin-induced PAF synthesis increased leukocyte adhesion to hIECS and mIECs and cell motility of both endothelial cell lines. Moreover, PAF or thrombin-induced PAF synthesis accelerated in vitro formation of vessel-like tubular structures when hIECs are seeded on Matrigel. Notably, gene-microarray analysis detected up-regulation of beta3 integrin gene on hIECs stimulated with PAF, that was confirmed at the protein level. CONCLUSIONS Based on the novel development of immortalized islet endothelium, these results suggest that PAF may have a dual role that links inflammation to angiogenesis in the early events of islet transplantation.
Collapse
Affiliation(s)
- Luigi Biancone
- Department of Internal Medicine and Research Center for Experimental Medicine (CeRMS), University of Torino, Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Emamaullee JA, Rajotte RV, Liston P, Korneluk RG, Lakey JRT, Shapiro AMJ, Elliott JF. XIAP overexpression in human islets prevents early posttransplant apoptosis and reduces the islet mass needed to treat diabetes. Diabetes 2005; 54:2541-8. [PMID: 16123341 DOI: 10.2337/diabetes.54.9.2541] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Edmonton Protocol for treatment of type 1 diabetes requires islets from two or more donors to achieve euglycemia in a single recipient, primarily because soon after portal infusion, the majority of the transplanted cells undergo apoptosis due to hypoxia and hypoxia reperfusion injury. X-linked inhibitor of apoptosis protein (XIAP) is a potent endogenous inhibitor of apoptosis that is capable of blocking the activation of multiple downstream caspases, and XIAP overexpression has previously been shown to enhance engraftment of a murine beta-cell line. In this study, human islets transduced with a XIAP-expressing recombinant adenovirus were resistant to apoptosis and functionally recovered following in vitro stresses of hypoxia and hypoxia with reoxygenation (models reperfusion injury). Furthermore Ad-XIAP transduction dramatically reduced the number of human islets required to reverse hyperglycemia in chemically diabetic immunodeficient mice. These results suggest that by transiently overexpressing XIAP in the immediate posttransplant period, human islets from a single donor might be used to effectively treat two diabetic recipients.
Collapse
Affiliation(s)
- Juliet A Emamaullee
- 1-21 Medical Sciences Building, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada.
| | | | | | | | | | | | | |
Collapse
|
50
|
Johansson H, Lukinius A, Moberg L, Lundgren T, Berne C, Foss A, Felldin M, Källen R, Salmela K, Tibell A, Tufveson G, Ekdahl KN, Elgue G, Korsgren O, Nilsson B. Tissue factor produced by the endocrine cells of the islets of Langerhans is associated with a negative outcome of clinical islet transplantation. Diabetes 2005; 54:1755-62. [PMID: 15919797 DOI: 10.2337/diabetes.54.6.1755] [Citation(s) in RCA: 238] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
There are strong indications that only a small fraction of grafts successfully engraft in clinical islet transplantation. One explanation may be the instant blood-mediated inflammatory reaction (IBMIR) elicited by tissue factor, which is produced by the endocrine cells. In the present study, we show that islets intended for islet transplantation produce tissue factor in both the transmembrane and the alternatively spliced form and that the membrane-bound form is released as microparticles often associated with both insulin and glucagon granules. A low-molecular mass factor VIIa (FVIIa) inhibitor that indirectly blocks both forms of tissue factor was shown in vitro to be a promising drug to eliminate the IBMIR. Thrombin-antithrombin complex (TAT) and FVIIa-antithrombin complex (FVIIa-AT) were measured in nine patients who together received 20 infusions of isolated human islets. Both the TAT and FVIIa-AT complexes increased rapidly within 15-60 min after infusion. When the initial TAT and FVIIa-AT levels were plotted against the increase in C-peptide concentration after 7 days, patients with an initially strong IBMIR showed no significant increase in insulin synthesis after 7 days. In conclusion, tissue factor present in both the islets and the culture medium and elicits IBMIR, which affects the function of the transplanted islets.
Collapse
Affiliation(s)
- Helena Johansson
- The Rudbeck Laboratory, University Hospital, 751 85 Uppsala, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|