1
|
Ahmadi F, Lotfi AS, Navaei-Nigjeh M, Kadivar M. Trimetazidine Preconditioning Potentiates the Effect of Mesenchymal Stem Cells Secretome on the Preservation of Rat Pancreatic Islet Survival and Function In Vitro. Appl Biochem Biotechnol 2023; 195:4796-4817. [PMID: 37184724 DOI: 10.1007/s12010-023-04532-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/16/2023]
Abstract
Islet transplantation offers improved glycemic control in individuals with type 1 diabetes mellitus. However, in vitro islet culture is associated with islet apoptosis and eventually will lose their functionality prior to transplantation. In this study, we examined the effects of mesenchymal stem cells (MSCs) secretome preconditioned with diazoxide (DZ) and trimetazidine (TMZ) on rat islet cells during pre-transplant culture. With and without preconditioned hAD-MSCs' concentrated conditioned media (CCM) were added to the culture medium containing rat islets every 12 h for 24 and 48 h, after testing for selected cytokine concentrations (interleukin (IL)-4, IL-6, IL-13). Insulin content, glucose-stimulated insulin secretion, islet cell apoptosis, and mRNA expression of pro-apoptotic (BAX, BAK-1, and PUMA) and anti-apoptotic factors (BCL-2, BCL-xL, and XIAP) in rat islets were assessed after 24 and 48 h of culture. The protein level of IL-6 and IL-4 was significantly higher in TMZ-MSC-CM compared to MSC-non-CM. In rat isolated islets, normalized secreted insulin in the presence of 16.7 mM glucose was significantly higher in treated islet groups compared to control islets at both 24 and 48 h cultivation. Also, the percentage of apoptotic islet cells TMZ-MSC-CCM-treated islets was significantly lower compared to MSC-CM and MSC-CCM-treated islets in both 24 and 48 h cultivation. Consistent with the number of apoptotic cells, after 24 h culture, the expression of BCL-2 and BCL-xL genes in the control islets was lower than all treatment islet groups and in 48 h was lower than only TMZ-MSC-CM-treated islets. Also, the expression of the XIAP gene in control islets was significantly lower compared to the TMZ-MSC-CCM-treated islets at both at 24 and 48 h. In addition, mRNA level of the BAX gene in TMZ-MSC-CCM-treated islets was significantly lower compared to other groups at 48 h. Our findings revealed that TMZ proved to be more effective than DZ and could enhance the potential of hAD-MSCs-CM to improve the function and viability of islets prior to transplantation.
Collapse
Affiliation(s)
- Fariborz Ahmadi
- Department of Clinical Biochemistry, Tarbiat Modares University, Tehran, Iran
| | | | - Mona Navaei-Nigjeh
- Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mehdi Kadivar
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
2
|
Teratani T, Kasahara N, Fujimoto Y, Sakuma Y, Miki A, Goto M, Sata N, Kitayama J. Mesenchymal Stem Cells Secretions Enhanced ATP Generation on Isolated Islets during Transplantation. Islets 2022; 14:69-81. [PMID: 35034568 PMCID: PMC8765074 DOI: 10.1080/19382014.2021.2022423] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The success of islet transplantation in both basic research and clinical settings has proven that cell therapy has the potential to cure diabetes. Islets intended for transplantation are inevitably subjected to damage from a number of sources, including ischemic injury during removal and delivery of the donor pancreas, enzymatic digestion during islet isolation, and reperfusion injury after transplantation in the recipient. Here, we found that protein factors secreted by porcine adipose-tissue mesenchymal stem cells (AT-MSCs) were capable of activating preserved porcine islets. A conditioned medium was prepared from the supernatant obtained by culturing porcine AT-MSCs for 2 days in serum-free medium. Islets were preserved at 4°C in University of Wisconsin solution during transportation and then incubated at 37°C in RPMI-1620 medium with fractions of various molecular weights prepared from the conditioned medium. After treatment with certain fractions of the AT-MSC secretions, the intracellular ATP levels of the activated islets had increased to over 160% of their initial values after 4 days of incubation. Our novel system may be able to restore the condition of isolated islets after transportation or preservation and may help to improve the long-term outcome of islet transplantation.Abbreviations: AT-MSC, adipose-tissue mesenchymal stem cell; Cas-3, caspase-3; DAPI, 4,6-diamidino-2-phenylindole; DTZ, dithizone; ES cell, embryonic stem cell; FITC, fluorescein isothiocyanate; IEQ, islet equivalent; INS, insulin; iPS cell, induced pluripotent stem cell; Luc-Tg rat, luciferase-transgenic rat; PCNA, proliferating cell nuclear antigen; PDX1, pancreatic and duodenal homeobox protein-1; UW, University of Wisconsin; ZO1, zona occludens 1.
Collapse
Affiliation(s)
- Takumi Teratani
- Division of Translational Research, Jichi Medical University, Tochigi, Japan
- Department of Surgery, Jichi Medical University, Tochigi, Japan
- CONTACT Takumi Teratani Division of Clinical Investigation, Jichi Medical University, 3311-1, Yakushiji, Shimotsukeshi, Tochigi329-0498, Japan
| | - Naoya Kasahara
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | | | - Yasunaru Sakuma
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Atsushi Miki
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Masafumi Goto
- New Industry Creation Hatchery Center, Tohoku University, Miyagi, Japan
| | - Naohiro Sata
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Joji Kitayama
- Division of Translational Research, Jichi Medical University, Tochigi, Japan
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
3
|
Dafoe TJ, Dos Santos T, Spigelman AF, Lyon J, Smith N, Bautista A, MacDonald PE, Manning Fox JE. Impacts of the COVID-19 pandemic on a human research islet program. Islets 2022; 14:101-113. [PMID: 35285768 PMCID: PMC8928860 DOI: 10.1080/19382014.2022.2047571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/23/2022] Open
Abstract
Designated a pandemic in March 2020, the spread of severe acute respiratory syndrome virus 2 (SARS-CoV2), the virus responsible for coronavirus disease 2019 (COVID-19), led to new guidelines and restrictions being implemented for individuals, businesses, and societies in efforts to limit the impacts of COVID-19 on personal health and healthcare systems. Here we report the impacts of the COVID-19 pandemic on pancreas processing and islet isolation/distribution outcomes at the Alberta Diabetes Institute IsletCore, a facility specializing in the processing and distribution of human pancreatic islets for research. While the number of organs processed was significantly reduced, organ quality and the function of cellular outputs were minimally impacted during the pandemic when compared to an equivalent period immediately prior. Despite the maintained quality of isolated islets, feedback from recipient groups was more negative. Our findings suggest this is likely due to disrupted distribution which led to increased transit times to recipient labs, particularly those overseas. Thus, to improve overall outcomes in a climate of limited research islet supply, prioritization of tissue recipients based on likely tissue transit times may be needed.
Collapse
Affiliation(s)
- Tina J. Dafoe
- Alberta Diabetes Institute IsletCore and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Theodore Dos Santos
- Alberta Diabetes Institute IsletCore and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Aliya F. Spigelman
- Alberta Diabetes Institute IsletCore and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - James Lyon
- Alberta Diabetes Institute IsletCore and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Nancy Smith
- Alberta Diabetes Institute IsletCore and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Austin Bautista
- Alberta Diabetes Institute IsletCore and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Patrick E. MacDonald
- Alberta Diabetes Institute IsletCore and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Jocelyn E. Manning Fox
- Alberta Diabetes Institute IsletCore and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
4
|
AP39, a Mitochondrial-Targeted H2S Donor, Improves Porcine Islet Survival in Culture. J Clin Med 2022; 11:jcm11185385. [PMID: 36143032 PMCID: PMC9504761 DOI: 10.3390/jcm11185385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/26/2022] [Accepted: 09/10/2022] [Indexed: 11/17/2022] Open
Abstract
The rapid deterioration of transplanted islets in culture is a well-established phenomenon. We recently reported that pancreas preservation with AP39 reduces reactive oxygen species (ROS) production and improves islet graft function. In this study, we investigated whether the addition of AP39 to the culture medium could reduce isolated islet deterioration and improve islet function. Isolated islets from porcine pancreata were cultured with 400 nM AP39 or without AP39 at 37 °C. After culturing for 6–72 h, the islet equivalents of porcine islets in the AP39(+) group were significantly higher than those in the AP39(−) group. The islets in the AP39(+) group exhibited significantly decreased levels of ROS production compared to the islets in the AP39(−) group. The islets in the AP39(+) group exhibited significantly increased mitochondrial membrane potential compared to the islets in the AP39(−) group. A marginal number (1500 IEs) of cultured islets from each group was then transplanted into streptozotocin-induced diabetic mice. Culturing isolated islets with AP39 improved islet transplantation outcomes in streptozotocin-induced diabetic mice. The addition of AP39 in culture medium reduces islet deterioration and furthers the advancements in β-cell replacement therapy.
Collapse
|
5
|
Nakashima Y, Iguchi H, Takakura K, Nakamura Y, Izumi K, Koba N, Haneda S, Tsukahara M. Adhesion Characteristics of Human Pancreatic Islets, Duct Epithelial Cells, and Acinar Cells to a Polymer Scaffold. Cell Transplant 2022; 31:9636897221120500. [PMID: 36062469 PMCID: PMC9449504 DOI: 10.1177/09636897221120500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
We reported in 2018 that among several extracellular matrices, fibronectin, type I collagen, type IV collagen, laminin I, fibrinogen, and bovine serum albumin, fibronectin is particularly useful for adhesion of porcine pancreatic tissue. Subsequently, we developed a technology that enables the chemical coating of the constituent motifs of fibronectin onto cell culture dishes. In this experiment, we used islets (purity ≥ 90%), duct epithelial cells (purity ≥ 60%), and acinar cells (purity ≥ 99%) isolated from human pancreas according to the Edmonton protocol published in 2000 and achieved adhesion to the constituent motifs of fibronectin. A solution including cGMP Prodo Islet Media was used as the assay solution. In islets, adhesion was enhanced with the constitutive motifs of fibronectin compared with uncoated islets. In the functional evaluation of islets, insulin mRNA expression and insulin secretion were enhanced by the constitutive motif of fibronectin compared with non-coated islets. The stimulation index was comparable between non-coated islets and fibronectin motifs. In duct epithelial cells, adhesion was mildly promoted by the fibronectin component compared with non-coated component, while in acinar cells, adhesion was inhibited by the fibronectin component compared with the non-coated component. These data suggest that the constitutive motifs of fibronectin are useful for the adhesion of islets and duct epithelial cells.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Center for iPS Cell Research and Application Foundation, Facility for iPS Cell Therapy, Kyoto University, Kyoto, Japan
| | - Hiroki Iguchi
- R&D Center Corporate Advanced Technology Institute Life Science Development Center, Sekisui Chemical Co., Ltd., Osaka, Japan
| | - Kenta Takakura
- R&D Center Corporate Advanced Technology Institute Life Science Development Center, Sekisui Chemical Co., Ltd., Osaka, Japan
| | - Yuta Nakamura
- R&D Center Corporate Advanced Technology Institute Life Science Development Center, Sekisui Chemical Co., Ltd., Osaka, Japan
| | | | | | - Satoshi Haneda
- R&D Center Corporate Advanced Technology Institute Life Science Development Center, Sekisui Chemical Co., Ltd., Osaka, Japan
| | - Masayoshi Tsukahara
- Center for iPS Cell Research and Application Foundation, Facility for iPS Cell Therapy, Kyoto University, Kyoto, Japan
| |
Collapse
|
6
|
Huang W, Wu T, Xie C, Rayner CK, Priest C, Ebendorff‐Heidepriem H, Zhao J(T. Sensing Intra‐ and Extra‐Cellular Ca 2+ in the Islet of Langerhans. ADVANCED FUNCTIONAL MATERIALS 2022; 32. [DOI: 10.1002/adfm.202106020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Indexed: 12/19/2024]
Abstract
AbstractCalcium ions (Ca2+) take part in intra‐ and inter‐cellular signaling to mediate cellular functions. Sensing this ubiquitous messenger is instrumental in disentangling the specific functions of cellular sub‐compartments and/or intercellular communications. In this review, the authors first describe intra‐ and inter‐cellular Ca2+ signaling in relation to insulin secretion from the pancreatic islets, and then outline the development of diverse sensors, for example, chemically synthesized indicators, genetically encoded proteins, and ion‐selective microelectrodes, for intra‐ and extra‐cellular sensing of Ca2+. Particular emphasis is placed on emerging approaches in this field, such as low‐affinity Ca2+ indicators and unique Ca2+‐responsive composite materials. The authors conclude by remarking on the challenges and opportunities for further developments in this field, which may facilitate a more comprehensive understanding of Ca2+ signaling within and outside the islets, and its relevance in health and disease.
Collapse
Affiliation(s)
- Weikun Huang
- Adelaide Medical School Centre of Research Excellence in Translating Nutritional Science to Good Health The University of Adelaide Adelaide South Australia 5005 Australia
- Institute for Photonics and Advanced Sensing School of Physical Sciences ARC Centre of Excellence for Nanoscale BioPhotonics University of Adelaide Adelaide South Australia 5005 Australia
| | - Tongzhi Wu
- Adelaide Medical School Centre of Research Excellence in Translating Nutritional Science to Good Health The University of Adelaide Adelaide South Australia 5005 Australia
| | - Cong Xie
- Adelaide Medical School Centre of Research Excellence in Translating Nutritional Science to Good Health The University of Adelaide Adelaide South Australia 5005 Australia
| | - Christopher K. Rayner
- Adelaide Medical School Centre of Research Excellence in Translating Nutritional Science to Good Health The University of Adelaide Adelaide South Australia 5005 Australia
| | - Craig Priest
- Australian National Fabrication Facility and Future Industries Institute UniSA STEM University of South Australia Mawson Lakes South Australia 5095 Australia
| | - Heike Ebendorff‐Heidepriem
- Institute for Photonics and Advanced Sensing School of Physical Sciences ARC Centre of Excellence for Nanoscale BioPhotonics University of Adelaide Adelaide South Australia 5005 Australia
| | - Jiangbo (Tim) Zhao
- Institute for Photonics and Advanced Sensing School of Physical Sciences ARC Centre of Excellence for Nanoscale BioPhotonics University of Adelaide Adelaide South Australia 5005 Australia
- Department of Engineering Faculty of Science and Engineering University of Hull Hull HU6 7RX UK
| |
Collapse
|
7
|
Olack BJ, Alexander M, Swanson CJ, Kilburn J, Corrales N, Flores A, Heng J, Arulmoli J, Omori K, Chlebeck PJ, Zitur L, Salgado M, Lakey JRT, Niland JC. Optimal Time to Ship Human Islets Post Tissue Culture to Maximize Islet. Cell Transplant 2021; 29:963689720974582. [PMID: 33231091 PMCID: PMC7885128 DOI: 10.1177/0963689720974582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Access to functional high-quality pancreatic human islets is critical to advance diabetes research. The Integrated Islet Distribution Program (IIDP), a major source for human islet distribution for over 15 years, conducted a study to evaluate the most advantageous times to ship islets postisolation to maximize islet recovery. For the evaluation, three experienced IIDP Islet Isolation Centers each provided samples from five human islet isolations, shipping 10,000 islet equivalents (IEQ) at four different time periods postislet isolation (no 37°C culture and shipped within 0 to 18 hours; or held in 37°C culture for 18 to 42, 48 to 96, or 144 to 192 hours). A central evaluation center compared samples for islet quantity, quality, and viability for each experimental condition preshipment and postshipment, as well as post 37°C culture 18 to 24 hours after shipment receipt. Additional evaluations included measures of functional potency by static glucose-stimulated insulin release (GSIR), represented as a stimulation index. Comparing the results of the four preshipment holding periods, the greatest IEQ loss postshipment occurred with the shortest preshipment times. Similar patterns emerged when comparing preshipment to postculture losses. In vitro islet function (GSIR) was not adversely impacted by increased tissue culture time. These data indicate that allowing time for islet recovery postisolation, prior to shipping, yields less islet loss during shipment without decreasing islet function.
Collapse
Affiliation(s)
- Barbara J Olack
- Integrated Islet Distribution Program, Department of Diabetes & Cancer Discovery Science, City of Hope, Duarte, CA, USA
| | - Michael Alexander
- Department of Surgery, University of California Irvine, Orange, CA, USA
| | - Carol J Swanson
- Integrated Islet Distribution Program, Department of Diabetes & Cancer Discovery Science, City of Hope, Duarte, CA, USA
| | - Julie Kilburn
- Integrated Islet Distribution Program, Department of Diabetes & Cancer Discovery Science, City of Hope, Duarte, CA, USA
| | - Nicole Corrales
- Department of Surgery, University of California Irvine, Orange, CA, USA
| | - Antonio Flores
- Department of Surgery, University of California Irvine, Orange, CA, USA
| | - Jennifer Heng
- Department of Surgery, University of California Irvine, Orange, CA, USA
| | | | - Keiko Omori
- Department of Translational Research and Cellular Therapeutics, City of Hope, Duarte, CA, USA
| | - Peter J Chlebeck
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Laura Zitur
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Mayra Salgado
- Department of Translational Research and Cellular Therapeutics, City of Hope, Duarte, CA, USA
| | - Jonathan R T Lakey
- Department of Surgery, University of California Irvine, Orange, CA, USA.,Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Joyce C Niland
- Integrated Islet Distribution Program, Department of Diabetes & Cancer Discovery Science, City of Hope, Duarte, CA, USA
| |
Collapse
|
8
|
Verhoeff K, Henschke SJ, Marfil-Garza BA, Dadheech N, Shapiro AMJ. Inducible Pluripotent Stem Cells as a Potential Cure for Diabetes. Cells 2021; 10:cells10020278. [PMID: 33573247 PMCID: PMC7911560 DOI: 10.3390/cells10020278] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 02/07/2023] Open
Abstract
Over the last century, diabetes has been treated with subcutaneous insulin, a discovery that enabled patients to forego death from hyperglycemia. Despite novel insulin formulations, patients with diabetes continue to suffer morbidity and mortality with unsustainable costs to the health care system. Continuous glucose monitoring, wearable insulin pumps, and closed-loop artificial pancreas systems represent an advance, but still fail to recreate physiologic euglycemia and are not universally available. Islet cell transplantation has evolved into a successful modality for treating a subset of patients with ‘brittle’ diabetes but is limited by organ donor supply and immunosuppression requirements. A novel approach involves generating autologous or immune-protected islet cells for transplant from inducible pluripotent stem cells to eliminate detrimental immune responses and organ supply limitations. In this review, we briefly discuss novel mechanisms for subcutaneous insulin delivery and define their shortfalls. We describe embryological development and physiology of islets to better understand their role in glycemic control and, finally, discuss cell-based therapies for diabetes and barriers to widespread use. In response to these barriers, we present the promise of stem cell therapy, and review the current gaps requiring solutions to enable widespread use of stem cells as a potential cure for diabetes.
Collapse
Affiliation(s)
- Kevin Verhoeff
- Department of Surgery, University of Alberta, Edmonton, AB T6G 2B7, Canada;
- Correspondence: ; Tel.: +1-780-984-1836
| | - Sarah J. Henschke
- Department of Emergency Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada;
| | | | - Nidheesh Dadheech
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada;
| | - Andrew Mark James Shapiro
- FRCS (Eng) FRCSC MSM FCAHS, Clinical Islet Transplant Program, Alberta Diabetes Institute, Department of Surgery, Canadian National Transplant Research Program, Edmonton, AB T6G 2B7, Canada;
| |
Collapse
|
9
|
Yang Z, Li X, Zhang C, Sun N, Guo T, Lin J, Li F, Zhang J. Amniotic Membrane Extract Protects Islets From Serum-Deprivation Induced Impairments and Improves Islet Transplantation Outcome. Front Endocrinol (Lausanne) 2020; 11:587450. [PMID: 33363516 PMCID: PMC7753361 DOI: 10.3389/fendo.2020.587450] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/04/2020] [Indexed: 11/25/2022] Open
Abstract
Islet culture prior to transplantation is a standard practice in many transplantation centers. Nevertheless, the abundant islet mass loss and function impairment during this serum-deprivation culture period restrain the success of islet transplantation. In the present study, we used a natural biomaterial derived product, amniotic membrane extract (AME), as medium supplementation of islet pretransplant cultivation to investigate its protective effect on islet survival and function and its underlying mechanisms, as well as the engraftment outcome of islets following AME treatment. Results showed that AME supplementation improved islet viability and function, and decreased islet apoptosis and islet loss during serum-deprived culture. This was associated with the increased phosphorylation of PI3K/Akt and MAPK/ERK signaling pathway. Moreover, transplantation of serum-deprivation stressed islets that were pre-treated with AME into diabetic mice revealed better blood glucose control and improved islet graft survival. In conclusion, AME could improve islet survival and function in vivo and in vitro, and was at least partially through increasing phosphorylation of PI3K/Akt and MAPK/ERK signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jialin Zhang
- Department of Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Misun PM, Yesildag B, Forschler F, Neelakandhan A, Rousset N, Biernath A, Hierlemann A, Frey O. In Vitro Platform for Studying Human Insulin Release Dynamics of Single Pancreatic Islet Microtissues at High Resolution. ADVANCED BIOSYSTEMS 2020; 4:e1900291. [PMID: 32293140 PMCID: PMC7610574 DOI: 10.1002/adbi.201900291] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Indexed: 01/18/2023]
Abstract
Insulin is released from pancreatic islets in a biphasic and pulsatile manner in response to elevated glucose levels. This highly dynamic insulin release can be studied in vitro with islet perifusion assays. Herein, a novel platform to perform glucose-stimulated insulin secretion (GSIS) assays with single islets is presented for studying the dynamics of insulin release at high temporal resolution. A standardized human islet model is developed and a microfluidic hanging-drop-based perifusion system is engineered, which facilitates rapid glucose switching, minimal sample dilution, low analyte dispersion, and short sampling intervals. Human islet microtissues feature robust and long-term glucose responsiveness and demonstrate reproducible dynamic GSIS with a prominent first phase and a sustained, pulsatile second phase. Perifusion of single islet microtissues produces a higher peak secretion rate, higher secretion during the first and second phases of insulin release, as well as more defined pulsations during the second phase in comparison to perifusion of pooled islets. The developed platform enables to study compound effects on both phases of insulin secretion as shown with two classes of insulin secretagogs. It provides a new tool for studying physiologically relevant dynamic insulin secretion at comparably low sample-to-sample variation and high temporal resolution.
Collapse
Affiliation(s)
- Patrick M. Misun
- Bio Engineering Laboratory Department of Biosystems Science and Engineering ETH Zürich Mattenstrasse 26, 4058 Basel, Switzerland
| | | | - Felix Forschler
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| | | | - Nassim Rousset
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| | | | - Andreas Hierlemann
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Olivier Frey
- InSphero AG Wagistrasse 27, 8952 Schlieren, Switzerland
| |
Collapse
|
11
|
Brandhorst H, Brandhorst D, Abraham A, Acreman S, Schive SW, Scholz H, Johnson PR. Proteomic Profiling Reveals the Ambivalent Character of the Mesenchymal Stem Cell Secretome: Assessing the Effect of Preconditioned Media on Isolated Human Islets. Cell Transplant 2020; 29:963689720952332. [PMID: 33150790 PMCID: PMC7784517 DOI: 10.1177/0963689720952332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 07/23/2020] [Accepted: 08/03/2020] [Indexed: 12/23/2022] Open
Abstract
Previous studies in rodents have indicated that function and survival of transplanted islets can be substantially improved by mesenchymal stem cells (MSC). The few human islet studies to date have confirmed these findings but have not determined whether physical contact between MSC and islets is required or whether the benefit to islets results from MSC-secreted proteins. This study aimed to investigate the protective capacity of MSC-preconditioned media for human islets. MSC were cultured for 2 or 5 days in normoxia or hypoxia before harvesting the cell-depleted media for human islet culture in normoxia or hypoxia for 6-8 or 3-4 days, respectively. To characterize MSC-preconditioned media, proteomic secretome profiling was performed to identify angiogenesis- and inflammation-related proteins. A protective effect of MSC-preconditioned media on survival and in vitro function of hypoxic human islets was observed irrespective of the atmosphere used for MSC preconditioning. Islet morphology changed markedly when media from hypoxic MSC were used for culture. However, PDX-1 and insulin gene expression did not confirm a change in the genetic phenotype of these islets. Proteomic profiling of preconditioned media revealed the heterogenicity of the secretome comprising angiogenic and antiapoptotic as well as angiostatic or proinflammatory mediators released at an identical pattern regardless whether MSC had been cultured in normoxic or hypoxic atmosphere. These findings do not allow a clear discrimination between normoxia and hypoxia as stimulus for protective MSC capabilities but indicate an ambivalent character of the MSC angiogenesis- and inflammation-related secretome. Nevertheless, culture of human islets in acellular MSC-preconditioned media resulted in improved morphological and functional islet integrity suggesting a disbalance in favor of protective factors. Further approaches should aim to eliminate potentially detrimental factors to enable the production of advanced clinical grade islet culture media with higher protective qualities.
Collapse
Affiliation(s)
- Heide Brandhorst
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Daniel Brandhorst
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Anju Abraham
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Samuel Acreman
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Simen W. Schive
- Department of Transplantation Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
| | - Hanne Scholz
- Department of Transplantation Medicine and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub, Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Paul R.V. Johnson
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Powell-Palm MJ, Zhang Y, Aruda J, Rubinsky B. Isochoric conditions enable high subfreezing temperature pancreatic islet preservation without osmotic cryoprotective agents. Cryobiology 2019; 86:130-133. [DOI: 10.1016/j.cryobiol.2019.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/06/2018] [Accepted: 01/04/2019] [Indexed: 11/30/2022]
|
13
|
Miki A, Ricordi C, Sakuma Y, Yamamoto T, Misawa R, Mita A, Molano RD, Vaziri ND, Pileggi A, Ichii H. Divergent antioxidant capacity of human islet cell subsets: A potential cause of beta-cell vulnerability in diabetes and islet transplantation. PLoS One 2018; 13:e0196570. [PMID: 29723228 PMCID: PMC5933778 DOI: 10.1371/journal.pone.0196570] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 04/16/2018] [Indexed: 01/09/2023] Open
Abstract
Background Type 1 and Type 2 diabetes mellitus (T1DM and T2DM) are caused by beta(β)-cell loss and functional impairment. Identification of mechanisms of β-cell death and therapeutic interventions to enhance β-cell survival are essential for prevention and treatment of diabetes. Oxidative stress is a common feature of both T1DM and T2DM; elevated biomarkers of oxidative stress are detected in blood, urine and tissues including pancreas of patients with DM. Islet transplantation is a promising treatment for diabetes. However, exposure to stress (chemical and mechanical) and ischemia-reperfusion during isolation and transplantation causes islet loss by generation of reactive oxygen species (ROS). Human intracellular antioxidant enzymes and related molecules are essential defenses against ROS. Antioxidant enzyme levels including superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPX) have been shown to be low in islet cells. However, little is known about the expression and function of antioxidant enzymes within islet cell subsets. We evaluated the expression of the key antioxidant enzymes in β- and alpha(α)-cell and accessed effects of oxidative stress, islet isolation and transplantation on β/α-cell ratio and viability in human islets. Methods Human pancreata from T1DM, T2DM and non-diabetic deceased donors were obtained and analyzed by confocal microscopy. Isolated islets were (I) transplanted in the renal sub-capsular space of streptozotocin-induced diabetic nude mice (in vivo bioassay), or (II) exposed to oxidative (H2O2) and nitrosative (NO donor) stress for 24 hrs in vitro. The ratio, % viability and death of β- and α-cells, and DNA damage (8OHdG) were measured. Results and conclusions Catalase and GPX expression was much lower in β- than α-cells. The β/α-cell ratio fells significantly following islet isolation and transplantation. Exposure to oxidative stress caused a significantly lower survival and viability, with higher DNA damage in β- than α-cells. These findings identified the weakness of β-cell antioxidant capacity as a main cause of vulnerability to oxidative stress. Potential strategies to enhance β-cell antioxidant capacity might be effective in prevention/treatment of diabetes.
Collapse
Affiliation(s)
- Atsushi Miki
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Camillo Ricordi
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Yasunaru Sakuma
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Toshiyuki Yamamoto
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Ryosuke Misawa
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Atsuyoshi Mita
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Ruth D Molano
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Nosratola D Vaziri
- Department of Medicine, University of California, Irvine, United States of America
| | - Antonello Pileggi
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America
| | - Hirohito Ichii
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, Florida, United States of America.,Department of Surgery, University of California, Irvine, United States of America
| |
Collapse
|
14
|
Rojas-Canales DM, Waibel M, Forget A, Penko D, Nitschke J, Harding FJ, Delalat B, Blencowe A, Loudovaris T, Grey ST, Thomas HE, Kay TWH, Drogemuller CJ, Voelcker NH, Coates PT. Oxygen-permeable microwell device maintains islet mass and integrity during shipping. Endocr Connect 2018; 7:490-503. [PMID: 29483160 PMCID: PMC5861371 DOI: 10.1530/ec-17-0349] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 02/26/2018] [Indexed: 01/05/2023]
Abstract
Islet transplantation is currently the only minimally invasive therapy available for patients with type 1 diabetes that can lead to insulin independence; however, it is limited to only a small number of patients. Although clinical procedures have improved in the isolation and culture of islets, a large number of islets are still lost in the pre-transplant period, limiting the success of this treatment. Moreover, current practice includes islets being prepared at specialized centers, which are sometimes remote to the transplant location. Thus, a critical point of intervention to maintain the quality and quantity of isolated islets is during transportation between isolation centers and the transplanting hospitals, during which 20-40% of functional islets can be lost. The current study investigated the use of an oxygen-permeable PDMS microwell device for long-distance transportation of isolated islets. We demonstrate that the microwell device protected islets from aggregation during transport, maintaining viability and average islet size during shipping.
Collapse
Affiliation(s)
- Darling M Rojas-Canales
- The Centre for Clinical and Experimental Transplantation (CCET) The Royal Adelaide HospitalAdelaide, South Australia, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- Department of MedicineFaculty of Health and Medical Sciences, University of Adelaide, South Australia, Australia
| | - Michaela Waibel
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- St Vincent's Institute of Medical ResearchFitzroy, Victoria, Australia
- The University of MelbourneDepartment of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Aurelien Forget
- Science and Engineering FacultyQueensland University of Technology, Brisbane, Queensland, Australia
| | - Daniella Penko
- The Centre for Clinical and Experimental Transplantation (CCET) The Royal Adelaide HospitalAdelaide, South Australia, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- Department of MedicineFaculty of Health and Medical Sciences, University of Adelaide, South Australia, Australia
| | - Jodie Nitschke
- The Centre for Clinical and Experimental Transplantation (CCET) The Royal Adelaide HospitalAdelaide, South Australia, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- Department of MedicineFaculty of Health and Medical Sciences, University of Adelaide, South Australia, Australia
| | - Fran J Harding
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- Future Industries InstituteUniversity of South Australia, Mawson Lakes, South Australia, Australia
| | - Bahman Delalat
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- Future Industries InstituteUniversity of South Australia, Mawson Lakes, South Australia, Australia
| | - Anton Blencowe
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- Future Industries InstituteUniversity of South Australia, Mawson Lakes, South Australia, Australia
- School of Pharmacy and Medical SciencesUniversity of South Australia, Adelaide, South Australia, Australia
| | - Thomas Loudovaris
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- St Vincent's Institute of Medical ResearchFitzroy, Victoria, Australia
| | - Shane T Grey
- The Centre for Clinical and Experimental Transplantation (CCET) The Royal Adelaide HospitalAdelaide, South Australia, Australia
- Transplantation Immunology GroupGarvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Helen E Thomas
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- St Vincent's Institute of Medical ResearchFitzroy, Victoria, Australia
- The University of MelbourneDepartment of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Thomas W H Kay
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- St Vincent's Institute of Medical ResearchFitzroy, Victoria, Australia
- The University of MelbourneDepartment of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Chris J Drogemuller
- The Centre for Clinical and Experimental Transplantation (CCET) The Royal Adelaide HospitalAdelaide, South Australia, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- Department of MedicineFaculty of Health and Medical Sciences, University of Adelaide, South Australia, Australia
| | - Nicolas H Voelcker
- Future Industries InstituteUniversity of South Australia, Mawson Lakes, South Australia, Australia
- Monash Institute of Pharmaceutical SciencesMonash University, Parkville, Victoria, Australia
| | - Patrick T Coates
- The Centre for Clinical and Experimental Transplantation (CCET) The Royal Adelaide HospitalAdelaide, South Australia, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing (CRC-CTM)Adelaide, South Australia, Australia
- Department of MedicineFaculty of Health and Medical Sciences, University of Adelaide, South Australia, Australia
| |
Collapse
|
15
|
Smith KE, Kelly AC, Min CG, Weber CS, McCarthy FM, Steyn LV, Badarinarayana V, Stanton JB, Kitzmann JP, Strop P, Gruessner AC, Lynch RM, Limesand SW, Papas KK. Acute Ischemia Induced by High-Density Culture Increases Cytokine Expression and Diminishes the Function and Viability of Highly Purified Human Islets of Langerhans. Transplantation 2017; 101:2705-2712. [PMID: 28263224 PMCID: PMC6319561 DOI: 10.1097/tp.0000000000001714] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/24/2017] [Accepted: 02/16/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND Encapsulation devices have the potential to enable cell-based insulin replacement therapies (such as human islet or stem cell-derived β cell transplantation) without immunosuppression. However, reasonably sized encapsulation devices promote ischemia due to high β cell densities creating prohibitively large diffusional distances for nutrients. It is hypothesized that even acute ischemic exposure will compromise the therapeutic potential of cell-based insulin replacement. In this study, the acute effects of high-density ischemia were investigated in human islets to develop a detailed profile of early ischemia induced changes and targets for intervention. METHODS Human islets were exposed in a pairwise model simulating high-density encapsulation to normoxic or ischemic culture for 12 hours, after which viability and function were measured. RNA sequencing was conducted to assess transcriptome-wide changes in gene expression. RESULTS Islet viability after acute ischemic exposure was reduced compared to normoxic culture conditions (P < 0.01). Insulin secretion was also diminished, with ischemic β cells losing their insulin secretory response to stimulatory glucose levels (P < 0.01). RNA sequencing revealed 657 differentially expressed genes following ischemia, with many that are associated with increased inflammatory and hypoxia-response signaling and decreased nutrient transport and metabolism. CONCLUSIONS In order for cell-based insulin replacement to be applied as a treatment for type 1 diabetes, oxygen and nutrient delivery to β cells will need to be maintained. We demonstrate that even brief ischemic exposure such as would be experienced in encapsulation devices damages islet viability and β cell function and leads to increased inflammatory signaling.
Collapse
Affiliation(s)
- Kate E. Smith
- Department of Surgery, University of Arizona, Tucson, AZ
- Department of Physiological Sciences GIDP, University of Arizona, Tucson, AZ
| | - Amy C. Kelly
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ
| | - Catherine G. Min
- Department of Surgery, University of Arizona, Tucson, AZ
- Department of Physiological Sciences GIDP, University of Arizona, Tucson, AZ
| | - Craig S. Weber
- Department of Physiology, University of Arizona, Tucson, AZ
| | - Fiona M. McCarthy
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ
| | - Leah V. Steyn
- Department of Surgery, University of Arizona, Tucson, AZ
| | | | | | | | - Peter Strop
- Sanofi-Aventis Group, Tucson, AZ
- Icagen, Inc., Tucson, AZ
| | | | | | - Sean W. Limesand
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ
| | | |
Collapse
|
16
|
Abstract
Clinical pancreatic islet transplantation can be considered one of the safest and least invasive transplant procedures. Remarkable progress has occurred in both the technical aspects of islet cell processing and the outcomes of clinical islet transplantation. With >1,500 patients treated since 2000, this therapeutic strategy has moved from a curiosity to a realistic treatment option for selected patients with type 1 diabetes mellitus (that is, those with hypoglycaemia unawareness, severe hypoglycaemic episodes and glycaemic lability). This Review outlines the techniques required for human islet isolation, in vitro culture before the transplant and clinical islet transplantation, and discusses indications, optimization of recipient immunosuppression and management of adjunctive immunomodulatory and anti-inflammatory strategies. The potential risks, long-term outcomes and advances in treatment after the transplant are also discussed to further move this treatment towards becoming a more widely available option for patients with type 1 diabetes mellitus and eventually a potential cure.
Collapse
Affiliation(s)
- A M James Shapiro
- Clinical Islet Transplant Program, University of Alberta, 2000 College Plaza, 8215 112th Street, Edmonton, Alberta T6G 2C8, Canada
- The Diabetes Research Institute Federation, 1450 NW 10 Avenue, Miami, Florida 33136, USA
- The Cure Alliance, 550 Bay Point Road, Miami, Florida 33137, USA
| | - Marta Pokrywczynska
- The Diabetes Research Institute Federation, 1450 NW 10 Avenue, Miami, Florida 33136, USA
- The Cure Alliance, 550 Bay Point Road, Miami, Florida 33137, USA
- Department of Regenerative Medicine, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz, Karlowicza 24 Street, 85-092 Bydgoszcz, Poland
| | - Camillo Ricordi
- The Diabetes Research Institute Federation, 1450 NW 10 Avenue, Miami, Florida 33136, USA
- The Cure Alliance, 550 Bay Point Road, Miami, Florida 33137, USA
- Diabetes Research Institute and Cell Transplant Program, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida 33136, USA
| |
Collapse
|
17
|
Kesseli SJ, Wagar M, Jung MK, Smith KD, Lin YK, Walsh RM, Hatipoglu B, Freeman ML, Pruett TL, Beilman GJ, Sutherland DER, Dunn TB, Axelrod DA, Chaidarun SS, Stevens TK, Bellin M, Gardner TB. Long-Term Glycemic Control in Adult Patients Undergoing Remote vs. Local Total Pancreatectomy With Islet Autotransplantation. Am J Gastroenterol 2017; 112:643-649. [PMID: 28169284 DOI: 10.1038/ajg.2017.14] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Total pancreatectomy with islet autotransplantation (TPIAT) is increasingly performed with remote islet cell processing and preparation, i.e., with islet cell isolation performed remotely from the primary surgical site at an appropriately equipped islet isolation facility. We aimed to determine whether TPIAT using remote islet isolation results in comparable long-term glycemic outcomes compared with TPIAT performed with standard local isolation. METHODS We performed a retrospective cohort study of adult patients who underwent TPIAT at three tertiary care centers from 2010 to 2013. Two centers performed remote isolation and one performed local isolation. Explanted pancreata in the remote cohort were transported ∼130 miles to and from islet isolation facilities. The primary outcome was insulin independence 1 year following transplant. RESULTS Baseline characteristics were similar between groups except the remote cohort had higher preoperative hemoglobin A1c (HbA1c; 5.43 vs. 5.25, P=0.02) and there were more females in the local cohort (58% vs. 76%, P=0.049). At 1 year, 27% of remote and 32% of local patients were insulin independent (P=0.48). Remote patients experienced a greater drop in fasting c-peptide (-1.66 vs. -0.64, P=0.006) and a greater rise in HbA1c (1.65 vs. 0.99, P=0.014) at 1-year follow-up. A preoperative c-peptide >2.7 (odds ratio (OR) 4.4, 95% confidence interval (CI) 1.6-14.3) and >3,000 islet equivalents/kg (OR 11.0, 95% CI 3.2-37.3) were associated with one-year insulin independence in the local group. CONCLUSIONS At 1 year after TPIAT, patients undergoing remote surgery have equivalent rates of long-term insulin independence compared with patients undergoing TPIAT locally, but metabolic control is superior with local isolation.
Collapse
Affiliation(s)
- Samuel J Kesseli
- Section of Gastroenterology and Hepatology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Matthew Wagar
- Section of Endocrinology, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - Min K Jung
- Section of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Kerrington D Smith
- Section of General Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Yu Kuei Lin
- Department of Endocrinology, Endocrinology and Metabolism Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - R Matthew Walsh
- Department of General Surgery, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Betul Hatipoglu
- Section of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Martin L Freeman
- Section of Gastroenterology, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - Timothy L Pruett
- Deparment of Surgery, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - Gregory J Beilman
- Deparment of Surgery, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - David E R Sutherland
- Deparment of Surgery, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - Ty B Dunn
- Deparment of Surgery, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - David A Axelrod
- Section of Transplant Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Sushela S Chaidarun
- Section of Endocrinology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Tyler K Stevens
- Section of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Melena Bellin
- Section of Endocrinology, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - Timothy B Gardner
- Section of Gastroenterology and Hepatology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| |
Collapse
|
18
|
Islet Cell Yield Following Remote Total Pancreatectomy With Islet Autotransplant is Independent of Cold Ischemia Time. Pancreas 2017; 46:380-384. [PMID: 28129232 PMCID: PMC5308539 DOI: 10.1097/mpa.0000000000000792] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Total pancreatectomy with islet autotransplantation is increasingly being performed remotely, that is, removing the pancreas in 1 location, isolating the islet cells in another location, then returning the islets to the original location for reimplantation into the patient. We determined the influence of extended cold ischemia time on key clinical outcomes in remote islet autotransplantation. METHODS We evaluated patients who underwent remote islet autotransplantation at 2 centers from 2011 to 2014. Patients were divided into 2 groups: those with and those without a decrease in C-peptide greater than 50% from baseline. The primary clinical outcome was the quantity of isolated islet equivalents per kilogram body weight (IEQs/kg). RESULTS Twenty-five patients met inclusion criteria; 15 had a decrease in C-peptide greater than 50% from baseline and had lower corresponding IEQs/kg compared with those without a decrease greater than 50% (4045 vs 6654 IEQs/kg, P = 0.01). There was no difference in cold ischemia time between the 2 groups (664 vs 600 minutes, P = 0.25). Daily insulin use at 1 year nearly met statistical significance (25.3 vs 8 U, P = 0.06), as did glycated hemoglobin (8.07 vs 6.69 mmol/L, P = 0.06). CONCLUSIONS Cold ischemia time does not influence islet yield in patients undergoing pancreatectomy with remote isolation.
Collapse
|
19
|
Otsuki K, Ito T, Kenmochi T, Maruyama M, Akutsu N, Saigo K, Hasegawa M, Aoyama H, Matsumoto I, Uchino Y. Positron Emission Tomography and Autoradiography of (18)F-Fluorodeoxyglucose Labeled Islets With or Without Warm Ischemic Stress in Portal Transplanted Rats. Transplant Proc 2016; 48:229-33. [PMID: 26915873 DOI: 10.1016/j.transproceed.2015.12.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 12/22/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The use of positron-emission tomography (PET) with (18)F-fluorodeoxyglucose (FDG) -labeled islets has been considered to be a potential modality to visualize and quantify early engraftment of islet transplantation. The objective of this study was to evaluate the early islets' survival of the FDG-labeled islets with or without warm ischemic stress in portal transplanted rats using PET and autoradiography. METHODS Islets were isolated from Lewis rat pancreata with or without 30-minute warm ischemia times (WITs). For islets' labeling, 300 islets were incubated with 3 MBq FDG for 60 minutes. FDG-labeled islets were transplanted into the liver via portal vein. In in vivo study, a PET study was scanned for 90 minutes and the FDG uptake was expressed as percentage of liver injection dose (ID). In ex vivo study, the liver was exposed for 30 minutes with single fluorescence autoradiography. RESULTS In the PET study, the percentage of liver ID of the islets without WIT was 27.8 and that of the WIT islets was 20.1 at the end of islet transplantation. At 90 minutes after transplantation, the percentage of liver ID was decreased to 14.7 in the islets without WIT and 10.1 in the WIT islets. In the autoradiogram, the number of hot spots was more obviously visualized in the liver transplanted without WIT islets than in the liver transplanted with WIT islets. CONCLUSION Almost 50% of the islets were immediately lost in both the islets without WIT and those with WIT transplantation in the early period. However, islet survival was 1.4 times higher in the islets without WIT than that in those with WIT in the early engraftment phase.
Collapse
Affiliation(s)
- K Otsuki
- Department of Surgery, National Chiba-East Hospital, Chiba, Japan.
| | - T Ito
- Department of Transplantation Surgery, National Chiba-East Hospital, Chiba, Japan
| | - T Kenmochi
- Department of Transplantation Surgery, National Chiba-East Hospital, Chiba, Japan
| | - M Maruyama
- Department of Surgery, National Chiba-East Hospital, Chiba, Japan
| | - N Akutsu
- Department of Surgery, National Chiba-East Hospital, Chiba, Japan
| | - K Saigo
- Department of Surgery, National Chiba-East Hospital, Chiba, Japan
| | - M Hasegawa
- Department of Surgery, National Chiba-East Hospital, Chiba, Japan
| | - H Aoyama
- Department of Surgery, National Chiba-East Hospital, Chiba, Japan
| | - I Matsumoto
- Department of Surgery, National Chiba-East Hospital, Chiba, Japan
| | - Y Uchino
- Chiba Ryogo Center, PET Imaging Division, Chiba, Japan
| |
Collapse
|
20
|
Brandhorst D, Brandhorst H, Mullooly N, Acreman S, Johnson PRV. High Seeding Density Induces Local Hypoxia and Triggers a Proinflammatory Response in Isolated Human Islets. Cell Transplant 2015; 25:1539-46. [PMID: 26628048 DOI: 10.3727/096368915x689929] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Hypoxia is the main threat to morphological and functional integrity of isolated pancreatic islets. Lack of oxygen seems to be of particular importance for functionality of encapsulated islets. The present study was initiated as an experimental model for the environment experienced by human islets in a confined space present during culture, shipment, and in an implanted macrodevice. Quadruplicate aliquots of isolated human islets (n = 12) were cultured for 24 h at 37°C under normoxic conditions using 24-well plates equipped with 8-µm pore size filter inserts and filled with islet aliquots adjusted to obtain a seeding density of 75, 150, 300, or 600 IEQ/cm(2). After culture viability, glucose-stimulated insulin release, DNA content as well as Bax and Bcl-2 gene expression were measured. Culture supernatants were collected to determine production of VEGF and MCP-1. Viability correlated inversely with IEQ seeding density (r = -0.71, p < 0.001), while the correlation of VEGF and MCP-1 secretion with seeding density was positive (r = 0.78, p < 0.001; r = 0.54, p < 0.001). Decreased viability corresponded with a significant increase in the Bax/Bcl-2 mRNA ratio at 300 and 600 IEQ/cm(2) and with a sigificantly reduced glucose-stimulated insulin secretion and insulin content compared to 75 or 150 IEQ/cm(2) (p < 0.01). The present study demonstrates that the seeding density is inversely correlated with islet viability and in vitro function. This is associated with a significant increase in VEGF and MCP-1 release suggesting a hypoxic and proinflammatory islet microenvironment.
Collapse
Affiliation(s)
| | - Heide Brandhorst
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | | | | | | |
Collapse
|
21
|
Tse HM, Kozlovskaya V, Kharlampieva E, Hunter CS. Minireview: Directed Differentiation and Encapsulation of Islet β-Cells-Recent Advances and Future Considerations. Mol Endocrinol 2015; 29:1388-99. [PMID: 26340406 DOI: 10.1210/me.2015-1085] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Diabetes mellitus has rapidly become a 21st century epidemic with the promise to create vast economic and health burdens, if left unchecked. The 2 major forms of diabetes arise from unique causes, with outcomes being an absolute (type 1) or relative (type 2) loss of functional pancreatic islet β-cell mass. Currently, patients rely on exogenous insulin and/or other pharmacologies that restore glucose homeostasis. Although these therapies have prolonged countless lives over the decades, the striking increases in both type 1 and type 2 diabetic diagnoses worldwide suggest a need for improved treatments. To this end, islet biologists are developing cell-based therapies by which a patient's lost insulin-producing β-cell mass is replenished. Pancreatic or islet transplantation from cadaveric donors into diabetic patients has been successful, yet the functional islet demand far surpasses supply. Thus, the field has been striving toward transplantation of renewable in vitro-derived β-cells that can restore euglycemia. Challenges have been numerous, but progress over the past decade has generated much excitement. In this review we will summarize recent findings that have placed us closer than ever to β-cell replacement therapies. With the promise of cell-based diabetes therapies on the horizon, we will also provide an overview of cellular encapsulation technologies that will deliver critical protection of newly implanted cells.
Collapse
Affiliation(s)
- Hubert M Tse
- Department of Microbiology and the Comprehensive Diabetes Center (H.M.T.) and Departments of Chemistry (V.K., E.K.) and Medicine, Division of Endocrinology Diabetes and Metabolism, and Comprehensive Diabetes Center (C.S.H.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Veronika Kozlovskaya
- Department of Microbiology and the Comprehensive Diabetes Center (H.M.T.) and Departments of Chemistry (V.K., E.K.) and Medicine, Division of Endocrinology Diabetes and Metabolism, and Comprehensive Diabetes Center (C.S.H.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Eugenia Kharlampieva
- Department of Microbiology and the Comprehensive Diabetes Center (H.M.T.) and Departments of Chemistry (V.K., E.K.) and Medicine, Division of Endocrinology Diabetes and Metabolism, and Comprehensive Diabetes Center (C.S.H.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Chad S Hunter
- Department of Microbiology and the Comprehensive Diabetes Center (H.M.T.) and Departments of Chemistry (V.K., E.K.) and Medicine, Division of Endocrinology Diabetes and Metabolism, and Comprehensive Diabetes Center (C.S.H.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
22
|
Noguchi H, Miyagi-Shiohira C, Kurima K, Kobayashi N, Saitoh I, Watanabe M, Noguchi Y, Matsushita M. Islet Culture/Preservation Before Islet Transplantation. CELL MEDICINE 2015; 8:25-9. [PMID: 26858905 DOI: 10.3727/215517915x689047] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Although islet culture prior to transplantation provides flexibility for the evaluation of isolated islets and the pretreatment of patients, it is well known that isolated islets deteriorate rapidly in culture. Human serum albumin (HSA) is used for medium supplementation instead of fetal bovine serum (FBS), which is typically used for islet culture research, to avoid the introduction of xenogeneic materials. However, FBS contains several factors that are beneficial to islet viability and which also neutralize the endogenous pancreatic enzymes or exogenous enzymes left over from the isolation process. Several groups have reported the comparison of cultures at 22°C and 37°C. Recent studies have demonstrated the superiority of 4°C preservation to 22°C and 37°C cultures. We herein review the current research on islet culture/preservation for clinical islet transplantation.
Collapse
Affiliation(s)
- Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus , Okinawa , Japan
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus , Okinawa , Japan
| | - Kiyoto Kurima
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus , Okinawa , Japan
| | | | - Issei Saitoh
- ‡ Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University , Niigata , Japan
| | - Masami Watanabe
- § Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| | - Yasufumi Noguchi
- ¶ Department of Socio-environmental Design, Hiroshima International University , Hiroshima , Japan
| | - Masayuki Matsushita
- # Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus , Okinawa , Japan
| |
Collapse
|
23
|
Nacher M, Estil Les E, Garcia A, Nadal B, Pairó M, Garcia C, Secanella L, Novials A, Montanya E. Human Serum Versus Human Serum Albumin Supplementation in Human Islet Pretransplantation Culture: In Vitro and In Vivo Assessment. Cell Transplant 2015; 25:343-52. [PMID: 25955150 DOI: 10.3727/096368915x688119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
There is conflicting evidence favoring both the use of human serum (HS) and of human serum albumin (HSA) in human islet culture. We evaluated the effects of HS versus HSA supplementation on 1) in vitro β-cell viability and function and 2) in vivo islet graft revascularization, islet viability, β-cell death, and metabolic outcome after transplantation. Islets isolated from 14 cadaveric organ donors were cultured for 3 days in CMRL 1066 medium supplemented with HS or HSA. After 3 days in culture, β-cell apoptosis was lower in HS group (1.41 ± 0.27 vs. 2.38 ± 0.39%, p = 0.029), and the recovery of islets was 77 ± 11% and 54 ± 1% in HS- and HSA-cultured groups, respectively. Glucose-stimulated insulin secretion (GSIS) was higher in HS group (29.4, range 10.4-99.9, vs. 22.3, range 8.7-70.6, p = 0.031). In vivo viability and revascularization was determined in HS- and HSA-cultured islets transplanted into the anterior chamber of the eye of Balb/c mice (n = 14), and β-cell apoptosis in paraffin-embedded mouse eyes. Islet viability and β-cell apoptosis were similar in both groups. Revascularization was observed in one graft (HS group) on day 10 after transplantation. Islet function was determined in streptozotocin (STZ)-diabetic nude mice (n = 33) transplanted with 2,000 IEQs cultured with HS or HSA that showed similar blood glucose levels and percentage of normoglycemic animals over time. In conclusion, human islets cultured in medium supplemented with HS showed higher survival in vitro, as well as islet viability and function. The higher in vitro survival increased the number of islets available for transplantation. However, the beneficial effect on viability and function did not translate into an improved metabolic evolution when a similar number of HSA- and HS-cultured islets was transplanted.
Collapse
Affiliation(s)
- Montserrat Nacher
- Hospital Universitari Bellvitge-IDIBELLL, Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lee S, Takahashi Y, Lee K, Mizuno M, Nemeno J, Takebe T, Lee J. Viability and Functional Assessment of Murine Pancreatic Islets After Transportation Between Korea and Japan. Transplant Proc 2015; 47:738-41. [DOI: 10.1016/j.transproceed.2014.12.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/31/2014] [Indexed: 11/25/2022]
|
25
|
Li N, Zhang Y, Xiu Z, Wang Y, Chen L, Wang S, Li S, Guo X, Ma X. The preservation of islet with alginate encapsulation in the process of transportation. Biotechnol Appl Biochem 2015; 62:530-6. [PMID: 25223970 DOI: 10.1002/bab.1295] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/10/2014] [Indexed: 12/12/2022]
Abstract
Restoration of insulin secretion by transplantation of isolated islets is a treatment for type Ι diabetes mellitus. One of the major issues with clinical treatment of islet transplantation is how to maintain islet viability during transportation from the donor to the patient. We developed a method that uses alginate encapsulation to protect islets from mechanical damage during shipment. We tested several variables for their impact on islet viability during transportation and used the significant variable to build a response surface methodology (RSM) model by the Box-Behnken design method. This type of model is a mathematical and statistical technique that we used to optimize the conditions for islet viability during shipment. In this study, the factors that significantly affected islet survival rate were incubation time, serum concentration, and preservation time. Then, an empirical model was built to optimize conditions of the islets for shipping according to the responses of the effect factors with RSM. This model can be used to predict the islet survival rate and can serve as a guide for optimizing the transportation method of islets and increasing the success rate of the transplant procedure.
Collapse
Affiliation(s)
- Na Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China.,Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Ying Zhang
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China
| | - Zhilong Xiu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China
| | - Yu Wang
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Li Chen
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Shujun Wang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China.,Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China
| | - Shen Li
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China.,Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China
| | - Xin Guo
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China
| | - Xiaojun Ma
- Laboratory of Biomedical Material Engineering, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, People's Republic of China
| |
Collapse
|
26
|
Ramnath RD, Maillard E, Jones K, Bateman PA, Hughes SSJ, Gralla J, Johnson PR, Gray DWR. In Vitro Assessment of Human Islet Vulnerability to Instant Blood-Mediated Inflammatory Reaction (IBMIR) and Its Use to Demonstrate a Beneficial Effect of Tissue Culture. Cell Transplant 2014; 24:2505-12. [PMID: 25375416 DOI: 10.3727/096368914x685320] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Culture of human pancreatic islets is now routinely carried out prior to clinical islet allotransplantation, using conditions that have been developed empirically. One of the major causes of early islet destruction after transplantation is the process termed instant blood-mediated inflammatory reaction (IBMIR). The aim of this study was to develop in vitro methods to investigate IBMIR and apply them to the culture conditions used routinely in our human islet isolation laboratory. Freshly isolated or precultured (24 h, 48 h) human islets were incubated in either ABO-compatible allogeneic human blood or Hank's buffered salt solution (HBSS) for 1 h at 37°C. Tissue factor (TF) expression and leukocyte migration were assessed by light microscopy. TF was also quantified by ELISA. To assess β-cell function, glucose-stimulated insulin secretion (GSIS) assay was carried out. The extent of islet β-cell damage was quantified using a proinsulin assay. Islets cultured for 24 h had higher GSIS when compared to freshly isolated or 48-h precultured islets. Freshly isolated islets had significantly higher TF content than 24-h and 48-h precultured islets. Incubation of freshly isolated human islets in allogeneic human blood released 6.5-fold higher level of proinsulin in comparison to freshly isolated human islets in HBSS. The high level of proinsulin released was significantly attenuated when precultured islets (24 h or 48 h) were exposed to fresh blood. Histological examination of fresh islets in blood clot showed that some islets were fragmented, showing signs of extraislet insulin leakage and extensive neutrophil infiltration and necrosis. These features were markedly reduced when the islets were cultured for 24 h. These results suggest that our standard 24-h islet culture is markedly beneficial in attenuating IBMIR, as evidenced by increased GSIS, lower content of TF, decrease islet fragmentation, and proinsulin release.
Collapse
Affiliation(s)
- Raina D Ramnath
- University of Oxford, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, Headington, Oxford, England, UK
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
OBJECTIVES The effects of glucocorticoid during culture on human islet cells have been controversial. Exendin-4 (EX) enhances the insulin secretion and significantly improves clinical outcomes in islet cell transplantation. In this study, we examined the effects of glucocorticoids and EX on human islet cells during pretransplant culture. METHODS Methylprednisolone (MP) and/or EX were added to the standard culture medium for clinical islet cell transplantation. Islets were cultured for 24 hours with 3 different conditions (control, no additives; MP alone; and MP + EX). β-Cell fractional viability, cellular composition, multiple cytokine/chemokine production, multiple phosphorylation proteins, and glucose-induced insulin secretion were evaluated. RESULTS Viable β-cell survival in MP and MP + EX group was significantly higher than in the control group. Exendin-4 prevented MP-induced reduction of insulin secretion. Methylprednisolone supplementation to the culture medium decreased cytokine and chemokine production. Moreover, extracellular signal-regulated kinase 1/2 phosphorylation was significantly increased by MP and MP + EX. CONCLUSIONS Glucocorticoid supplementation into culture media significantly decreased the cytokine/chemokine production and increased the extracellular signal-regulated kinase 1/2 phosphorylation, resulting in the improvement of human β-cell survival. In addition, EX maintained the insulin secretion suppressed by MP. The supplementation of MP and EX together could be a useful strategy to create suitable human islets for transplantation.
Collapse
|
28
|
Kitzmann JP, Pepper AR, Lopez BG, Pawlick R, Kin T, O’Gorman D, Mueller KR, Gruessner AC, Avgoustiniatos ES, Karatzas T, Szot GL, Posselt AM, Stock PG, Wilson JR, Shapiro AM, Papas KK. Human islet viability and function is maintained during high-density shipment in silicone rubber membrane vessels. Transplant Proc 2014; 46:1989-1991. [PMID: 25131090 PMCID: PMC4169700 DOI: 10.1016/j.transproceed.2014.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The shipment of human islets (IE) from processing centers to distant laboratories is beneficial for both research and clinical applications. The maintenance of islet viability and function in transit is critically important. Gas-permeable silicone rubber membrane (SRM) vessels reduce the risk of hypoxia-induced death or dysfunction during high-density islet culture or shipment. SRM vessels may offer additional advantages: they are cost-effective (fewer flasks, less labor needed), safer (lower contamination risk), and simpler (culture vessel can also be used for shipment). METHOD IE were isolated from two manufacturing centers and shipped in 10-cm(2) surface area SRM vessels in temperature- and pressure-controlled containers to a distant center after at least 2 days of culture (n = 6). Three conditions were examined: low density (LD), high density (HD), and a microcentrifuge tube negative control (NC). LD was designed to mimic the standard culture density for IE preparations (200 IE/cm(2)), while HD was designed to have a 20-fold higher tissue density, which would enable the culture of an entire human isolation in 1-3 vessels. Upon receipt, islets were assessed for viability (measured by oxygen consumption rate normalized to DNA content [OCR/DNA)]), quantity (measured by DNA), and, when possible, potency and function (measured by dynamic glucose-stimulated insulin secretion measurements and transplants in immunodeficient B6 Rag(+/-) mice). Postshipment OCR/DNA was not reduced in HD vs LD and was substantially reduced in the NC condition. HD islets exhibited normal function postshipment. Based on the data, we conclude that entire islet isolations (up to 400,000 IE) may be shipped using a single, larger SRM vessel with no negative effect on viability and ex vivo and in vivo function.
Collapse
Affiliation(s)
| | - Andrew R Pepper
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Boris G Lopez
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Rena Pawlick
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Tatsuya Kin
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Doug O’Gorman
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Kathryn R Mueller
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | | | | | - Theodore Karatzas
- Department of Surgery, University of Arizona, Tucson, AZ, United States
- Second Department of Propedeutic Surgery University of Athens, School of Medicine, Athens, Greece
| | - Greg L Szot
- Diabetes Center, University of California, San Francisco, California, United States
| | - Andrew M Posselt
- Diabetes Center, University of California, San Francisco, California, United States
| | - Peter G Stock
- Diabetes Center, University of California, San Francisco, California, United States
| | - John R Wilson
- Wilson Wolf Manufacturing Corporation, New Brighton, Minnesota, United States
| | - AM Shapiro
- Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Klearchos K Papas
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
29
|
Hughes A, Rojas-Canales D, Drogemuller C, Voelcker NH, Grey ST, Coates PTH. IGF2: an endocrine hormone to improve islet transplant survival. J Endocrinol 2014; 221:R41-8. [PMID: 24883437 DOI: 10.1530/joe-13-0557] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the week following pancreatic islet transplantation, up to 50% of transplanted islets are lost due to apoptotic cell death triggered by hypoxic and pro-inflammatory cytokine-mediated cell stress. Thus, therapeutic approaches designed to protect islet cells from apoptosis could significantly improve islet transplant success. IGF2 is an anti-apoptotic endocrine protein that inhibits apoptotic cell death through the mitochondrial (intrinsic pathway) or via antagonising activation of pro-inflammatory cytokine signalling (extrinsic pathway), in doing so IGF2 has emerged as a promising therapeutic molecule to improve islet survival in the immediate post-transplant period. The development of novel biomaterials coated with IGF2 is a promising strategy to achieve this. This review examines the mechanisms mediating islet cell apoptosis in the peri- and post-transplant period and aims to identify the utility of IGF2 to promote islet survival and enhance long-term insulin independence rates within the setting of clinical islet transplantation.
Collapse
|
30
|
Wang X, Chang F, Bai Y, Chen F, Zhang J, Chen L. Bisphenol A enhances kisspeptin neurons in anteroventral periventricular nucleus of female mice. J Endocrinol 2014; 221:201-13. [PMID: 24532816 DOI: 10.1530/joe-13-0475] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bisphenol-A (BPA), an environmental estrogen, adversely affects female reproductive health. However, the underlying mechanisms remain largely unknown. We found that oral administration (p.o.) of BPA (20 μg/kg) to adult female mice at proestrus, but not at estrus or diestrus, significantly increased the levels of plasma E₂, LH and FSH, and Gnrh mRNA within 6 h. The administration of BPA at proestrus, but not at diestrus, could elevate the levels of Kiss1 mRNA and kisspeptin protein in anteroventral periventricular nucleus (AVPV) within 6 h. In contrast, the level of Kiss1 mRNA in arcuate nucleus (ARC) was hardly altered by BPA administration. In addition, at proestrus, a single injection (i.c.v.) of BPA dose-dependently enhanced the AVPV-kisspeptin expression within 6 h, this was sensitive to E₂ depletion by ovariectomy and an estrogen receptor α (ERα) antagonist. Similarly, the injection of BPA (i.c.v.) at proestrus could elevate the levels of plasma E₂, LH, and Gnrh mRNA within 6 h in a dose-dependent manner, which was blocked by antagonists of GPR54 or ERα. Injection of BPA (i.c.v.) at proestrus failed to alter the timing and peak concentration of LH-surge generation. In ovariectomized mice, the application of E₂ induced a dose-dependent increase in the AVPV-Kiss1 mRNA level, indicating 'E₂-induced positive feedback', which was enhanced by BPA injection (i.c.v.). The levels of Erα (Esr1) and Erβ (Esr2) mRNAs in AVPV and ARC did not differ significantly between vehicle-and BPA-treated groups. This study provides in vivo evidence that exposure of adult female mice to a low dose of BPA disrupts the hypothalamic-pituitary-gonadal reproductive endocrine system through enhancing AVPV-kisspeptin expression and release.
Collapse
Affiliation(s)
- Xiaoli Wang
- State Key Laboratory of Reproductive Medicine Department of Physiology, Nanjing Medical University, Hanzhong Road 140, Nanjing 210029, China MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | | | | | | | | | | |
Collapse
|
31
|
Rotational transport of islets: the best way for islets to get around? BIOMED RESEARCH INTERNATIONAL 2013; 2013:975608. [PMID: 24324977 PMCID: PMC3845626 DOI: 10.1155/2013/975608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 10/21/2013] [Indexed: 11/18/2022]
Abstract
Islet transplantation is a valid treatment option for patients suffering from type 1 diabetes mellitus. To assure optimal islet cell quality, specialized islet isolation facilities have been developed. Utilization of such facilities necessitates transportation of islet cells to distant institutions for transplantation. Despite its importance, a clinically feasible solution for the transport of islets has still not been established. We here compare the functionality of isolated islets from C57BL/6 mice directly after the isolation procedure as well as after two simulated transport conditions, static versus rotation. Islet cell quality was assessed using real-time live confocal microscopy. In vivo islet function after syngeneic transplantation was determined by weight and blood sugar measurements as well as by intraperitoneal glucose tolerance tests. Vascularization of islets was documented by fluorescence microscopy and immunohistochemistry. All viability parameters documented comparable cell viability in the rotary group and the group transplanted immediately after isolation. Functional parameters assessed in vivo displayed no significant difference between these two groups. Moreover, vascularization of islets was similar in both groups. In conclusion, rotary culture conditions allows the maintenance of highest islet quality for at least 15 h, which is comparable to that of freshly isolated islets.
Collapse
|
32
|
Kim TS, Lee HS, Oh SH, Moon H, Lee S, Song S, Shin M, Park JB, Kim SJ, Joh JW, Lee SK. Optimal device and method for transportation of isolated porcine islet. Transplant Proc 2013; 45:3097-101. [PMID: 24157043 DOI: 10.1016/j.transproceed.2013.08.083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
INTRODUCTION We investigated the optimal method for transportation of isolated porcine islets from an isolation facility to a transplant hospital or research center in terms of temperature, oxygen supply, and shaking effect. METHODS Commercially available insulator boxes with thermoregulators exposed for 5 hours under two external temperatures (4°C and 37°C) were monitored using HOBO temperature loggers. To find the optimal transport device, we compared islet counts, viability, quality, and function in conical tubes, gas-permeable bags (GPB) and gas-permeable flasks (GPF) after 1, 3 and 5 hours. To evaluate the effects of shaking on islets, we also analyzed the difference between a control and a shaking group in each device with time. RESULTS Commercially available Styrofoam insulators with thermoregulators maintained the internal temperature near the target. Islet recovery rate for GPF, which was higher than other devices, was maintained, while those decreased with time for conical tube and GPB containers adenosine diphosphate/adenosine triphosphate (ADP/ATP) ratio for GPF was lower than other devices, albeit not significantly fluoroscein acrimide/propidium iodide (AO/PI) ratio for GPF was higher than other devices after 5 hours. Glucose stimulated index was not different among the devices. In comparison with the control group, shaking yielded comparable islet survival, viability and function. CONCLUSION Our study demonstrated that the use of commercially available insulator boxes with thermoregulators maintained internal temperature close to the target value and that GPF was more favorable for islet oxygenation during transportation. This study also suggested negligible impact of shaking on isolated porcine islets during transportation.
Collapse
Affiliation(s)
- T-S Kim
- Department of Surgery, Samsung Medical Center, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Yamashita S, Ohashi K, Utoh R, Kin T, Shapiro AMJ, Yamamoto M, Gotoh M, Okano T. Quality of Air-Transported Human Islets for Single Islet Cell Preparations. CELL MEDICINE 2013; 6:33-8. [PMID: 26858878 DOI: 10.3727/215517913x674243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In new generation medical therapies for type 1 diabetes mellitus (DM), cell-based approaches using pancreatic islets have attracted significant attention worldwide. In particular, dispersed islet cells obtained from isolated pancreatic islets have been a valuable source in the cell biology and tissue engineering fields. Our experimental approach to the development of new islet-based DM therapies consisted of creating a monolithic islet cell sheet format using dispersed islet cells. In this experiment, we explored the potential of internationally transporting human islets from Alberta, Canada to Tokyo, Japan and obtaining viable dispersed islet cells. A total of 34 batches of isolated and purified human islets were transported using a commercial air courier service. Prior to shipping, the human islets had been in culture for 0-108 h at the University of Alberta. The transportation period from Alberta to Tokyo was 2-5 days. The transported human islet cells were enzymatically dispersed as single cells in Tokyo. The number of single islet cells decreased as the number of transportation days increased. In contrast, cell viability was maintained regardless of the number of transportation days. The preshipment culture time had no effect on the number or viability of single cells dispersed in Tokyo. When dispersed single islet cells were plated on laminin-5-coated temperature-responsive polymer-grafted culture dishes, the cells showed favorable attachment followed by extension as a monolithic format. The present study demonstrated that long-distance transported human islets are a viable cell source for experiments utilizing dispersed human islet cells.
Collapse
Affiliation(s)
- Shingo Yamashita
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University , Tokyo , Japan
| | - Kazuo Ohashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan; †Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Rie Utoh
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University , Tokyo , Japan
| | - Tatsuya Kin
- ‡ Clinical Islet Transplant Program, University of Alberta , Edmonton, Alberta , Canada
| | - A M James Shapiro
- ‡ Clinical Islet Transplant Program, University of Alberta , Edmonton, Alberta , Canada
| | - Masakazu Yamamoto
- † Department of Surgery, Institute of Gastroenterology, Tokyo Women's Medical University , Tokyo , Japan
| | - Mitsukazu Gotoh
- § Department of Regenerative Surgery, Fukushima Medical University , Fukushima , Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University , Tokyo , Japan
| |
Collapse
|
34
|
Yamada S, Shimada M, Utsunomiya T, Ikemoto T, Saito Y, Morine Y, Imura S, Mori H, Arakawa Y, Kanamoto M, Iwahashi S. Trophic effect of adipose tissue-derived stem cells on porcine islet cells. J Surg Res 2013; 187:667-72. [PMID: 24238974 DOI: 10.1016/j.jss.2013.10.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 10/11/2013] [Accepted: 10/17/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Adipose tissue-derived stem cells (ADSCs), which are widely known as multipotent progenitor cells, release several cytokines that support cell survival and repair. The aim of this study was to investigate whether ADSC-secreted molecules could induce a trophic effect in pancreatic islet culture conditions in vitro. MATERIALS AND METHODS We cocultured porcine islet cells with ADSCs using a transwell system for 48 h and evaluated the viability of islet cells. We also determined the concentration levels of cytokines and insulin in the supernatant of the culture medium. We used anti-vascular endothelial growth factor (VEGF) and anti-interleukin (IL)-6 receptor antibodies to investigate the effect of VEGF and IL-6 on islet cells. RESULTS ADSCs improved the viability of islet cells in the absence of cell-cell contact (P < 0.05). VEGF and IL-6 levels in the culture medium increased when islet cells were cocultured with ADSCs (P < 0.05). Furthermore, inhibition of VEGF decreased the viability of islet cells (P < 0.05); however, inhibition of IL-6 did not affect islet cell viability. CONCLUSIONS These results suggested that trophic factors, particularly VEGF, secreted by human ADSCs enhanced the survival and function of porcine islet cells.
Collapse
Affiliation(s)
- Shinichiro Yamada
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan
| | - Mitsuo Shimada
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan.
| | - Tohru Utsunomiya
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan
| | - Tetsuya Ikemoto
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan
| | - Yu Saito
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan
| | - Yuji Morine
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan
| | - Satoru Imura
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan
| | - Hiroki Mori
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan
| | - Yusuke Arakawa
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan
| | - Mami Kanamoto
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan
| | - Shuichi Iwahashi
- Department of Digestive and Transplant Surgery, The University of Tokushima, Tokushima City, Tokushima, Japan
| |
Collapse
|
35
|
Hogan AR, Doni M, Molano RD, Ribeiro MM, Szeto A, Cobianchi L, Zahr-Akrawi E, Molina J, Fornoni A, Mendez AJ, Ricordi C, Pastori RL, Pileggi A. Beneficial effects of ischemic preconditioning on pancreas cold preservation. Cell Transplant 2013; 21:1349-60. [PMID: 22305457 DOI: 10.3727/096368911x623853] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ischemic preconditioning (IPC) confers tissue resistance to subsequent ischemia in several organs. The protective effects are obtained by applying short periods of warm ischemia followed by reperfusion prior to extended ischemic insults to the organs. In the present study, we evaluated whether IPC can reduce pancreatic tissue injury following cold ischemic preservation. Rat pancreata were exposed to IPC (10 min of warm ischemia followed by 10 min of reperfusion) prior to ~18 h of cold preservation before assessment of organ injury or islet isolation. Pancreas IPC improved islet yields (964 ± 336 vs. 711 ± 204 IEQ/pancreas; p = 0.004) and lowered islet loss after culture (33 ± 10% vs. 51 ± 14%; p = 0.0005). Islet potency in vivo was well preserved with diabetes reversal and improved glucose clearance. Pancreas IPC reduced levels of NADPH-dependent oxidase, a source of reactive oxygen species, in pancreas homogenates versus controls (78.4 ± 45.9 vs. 216.2 ± 53.8 RLU/μg; p = 0.002). Microarray genomic analysis of pancreata revealed upregulation of 81 genes and downregulation of 454 genes (greater than twofold change) when comparing IPC-treated glands to controls, respectively, and showing a decrease in markers of apoptosis and oxidative stress. Collectively, our study demonstrates beneficial effects of IPC of the pancreas prior to cold organ preservation and provides evidence of the key role of IPC-mediated modulation of oxidative stress pathways. The use of IPC of the pancreas may contribute to increasing the quality of donor pancreas for transplantation and to improving organ utilization.
Collapse
Affiliation(s)
- Anthony R Hogan
- Diabetes Research Institute, University of Miami, Miami, FL 33136, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kasahara N, Teratani T, Doi J, Iijima Y, Maeda M, Uemoto S, Fujimoto Y, Sata N, Yasuda Y, Kobayashi E. Use of Mesenchymal Stem Cell-Conditioned Medium to Activate Islets in Preservation Solution. CELL MEDICINE 2013; 5:75-81. [PMID: 26858869 DOI: 10.3727/215517913x666477] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Pancreatic islet transplantation has received widespread attention as a promising treatment for type 1 diabetes. However, islets for transplantation are subject to damage from a number of sources, including ischemic injury during removal and delivery of the donor pancreas, enzymatic digestion during islet isolation, and reperfusion injury after transplantation in the recipient. Here we found that protein fractions secreted by mesenchymal stem cells (MSCs) were capable of activating preserved islets. A conditioned medium from the supernatant obtained by culturing adipose tissue MSCs (derived from wild-type Lewis rats) was prepared for 2 days in serum-free medium. Luc-Tg rat islets to which an organ preservation solution was added were then incubated at 4°C with fractions of various molecular weights prepared from the conditioned medium. Under the treatment with some of the fractions, by 4 days the relative luminescence intensities (representative of the ATP levels of the cold-preserved islets) had increased to over 150% of their initial values. Our novel system may be able to restore isolated islets to the condition they were in before transport, culture, and transplantation.
Collapse
Affiliation(s)
- Naoya Kasahara
- Division of Development of Advanced Therapy, Center for Development of Advanced Medical Technology, Jichi Medical University, Shimotsukeshi, Tochigi, Japan; †Department of Surgery, Jichi Medical University, Shimotsukeshi, Tochigi, Japan
| | - Takumi Teratani
- Division of Development of Advanced Therapy, Center for Development of Advanced Medical Technology, Jichi Medical University , Shimotsukeshi, Tochigi , Japan
| | - Junshi Doi
- Division of Development of Advanced Therapy, Center for Development of Advanced Medical Technology, Jichi Medical University , Shimotsukeshi, Tochigi , Japan
| | - Yuki Iijima
- Division of Development of Advanced Therapy, Center for Development of Advanced Medical Technology, Jichi Medical University , Shimotsukeshi, Tochigi , Japan
| | - Masashi Maeda
- Division of Development of Advanced Therapy, Center for Development of Advanced Medical Technology, Jichi Medical University , Shimotsukeshi, Tochigi , Japan
| | - Shinji Uemoto
- ‡ Division of Hepato-Pancreato-Biliary Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine , Syougoin, Sakyoku, Kyotoshi, Kyoto , Japan
| | - Yasuhiro Fujimoto
- Division of Development of Advanced Therapy, Center for Development of Advanced Medical Technology, Jichi Medical University, Shimotsukeshi, Tochigi, Japan; ‡Division of Hepato-Pancreato-Biliary Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine, Syougoin, Sakyoku, Kyotoshi, Kyoto, Japan
| | - Naohiro Sata
- † Department of Surgery, Jichi Medical University , Shimotsukeshi, Tochigi , Japan
| | - Yoshikazu Yasuda
- † Department of Surgery, Jichi Medical University , Shimotsukeshi, Tochigi , Japan
| | - Eiji Kobayashi
- Division of Development of Advanced Therapy, Center for Development of Advanced Medical Technology, Jichi Medical University , Shimotsukeshi, Tochigi , Japan
| |
Collapse
|
37
|
Takita M, Matsumoto S, Shimoda M, Chujo D, Itoh T, Sorelle JA, Purcell K, Onaca N, Naziruddin B, Levy MF. Safety and tolerability of the T-cell depletion protocol coupled with anakinra and etanercept for clinical islet cell transplantation. Clin Transplant 2013; 26:E471-84. [PMID: 23061757 DOI: 10.1111/ctr.12011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Islet cell transplantation (ICT) is a promising approach to cure patients with type 1 diabetes. We have implemented a new immunosuppression protocol with antithymoglobulin plus anti-inflammatory agents of anakinra and eternacept for induction and tacrolimus plus mycophenolate mofetil for maintenance [T-cell depletion with anti-inflammatory (TCD-AI) protocol], resulting in successful single-donor ICT. METHODS Eight islet recipients with type 1 diabetes reported adverse events (AEs) monthly. AEs were compared between three groups: first infusion with the TCD-AI protocol (TCD-AI-1st) and first and second infusion with the Edmonton-type protocol (Edmonton-1st and Edmonton-2nd). RESULTS The incidence of symptomatic AEs within the initial three months in the TCD-AI-1st group was less than in the Edmonton-1st and Edmonton-2nd groups, with a marginally significant difference (mean ± SE: 5.5 ± 0.3, 7.5 ± 0.5, and 8.3 ± 1.3, respectively; p = 0.07). A significant reduction in liver enzyme elevation after ICT was found in the TCD-AI-1st group compared with the Edmonton-1st and Edmonton-2nd groups (p < 0.05). Because of AEs, all patients in the Edmonton protocol eventually converted to the TCD-AI protocol, whereas all patients tolerated the TCD-AI protocol. CONCLUSIONS TCD-AI protocol can be tolerated for successful ICT, although this study includes small cohort, and large population trial should be taken.
Collapse
Affiliation(s)
- Morihito Takita
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Combined strategy of endothelial cells coating, Sertoli cells coculture and infusion improves vascularization and rejection protection of islet graft. PLoS One 2013; 8:e56696. [PMID: 23437215 PMCID: PMC3577699 DOI: 10.1371/journal.pone.0056696] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 01/14/2013] [Indexed: 11/19/2022] Open
Abstract
Improving islet graft revascularization and inhibiting rejection become crucial tasks for prolonging islet graft survival. Endothelial cells (ECs) are the basis of islet vascularization and Sertoli cells (SCs) have the talent to provide nutritional support and exert immunosuppressive effects. We construct a combined strategy of ECs coating in the presence of nutritious and immune factors supplied by SCs in a co-culture system to investigate the effect of vascularization and rejection inhibition for islet graft. In vivo, the combined strategy improved the survival and vascularization as well as inhibited lymphocytes and inflammatory cytokines. In vitro, we found the combinatorial strategy improved the function of islets and the effect of ECs-coating on islets. Combined strategy treated islets revealed higher levels of anti-apoptotic signal molecules (Bcl-2 and HSP-32), survival and function related molecules (PDX-1, Ki-67, ERK1/2 and Akt) and demonstrated increased vascular endothelial growth factor receptor 2 (KDR) and angiogenesis signal molecules (FAk and PLC-γ). SCs effectively inhibited the activation of lymphocyte stimulated by islets and ECs. Predominantly immunosuppressive cytokines could be detected in culture supernatants of the SCs coculture group. These results suggest that ECs-coating and Sertoli cells co-culture or infusion synergistically enhance islet survival and function after transplantation.
Collapse
|
39
|
Buchwald P, Cechin SR. Glucose-stimulated insulin secretion in isolated pancreatic islets: Multiphysics FEM model calculations compared to results of perifusion experiments with human islets. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/jbise.2013.65a006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
40
|
Shapiro AMJ. Islet transplantation in type 1 diabetes: ongoing challenges, refined procedures, and long-term outcome. Rev Diabet Stud 2012; 9:385-406. [PMID: 23804275 DOI: 10.1900/rds.2012.9.385] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Remarkable progress has been made in islet transplantation over a span of 40 years. Once just an experimental curiosity in mice, this therapy has moved forward, and can now provide robust therapy for highly selected patients with type 1 diabetes (T1D), refractory to stabilization by other means. This progress could not have occurred without extensive dynamic international collaboration. Currently, 1,085 patients have undergone islet transplantation at 40 international sites since the Edmonton Protocol was reported in 2000 (752 allografts, 333 autografts), according to the Collaborative Islet Transplant Registry. The long-term results of islet transplantation in selected centers now match registry data of pancreas-alone transplantation, with 6 sites reporting five-year insulin independence rates ≥50%. Islet transplantation has been criticized for the use of multiple donor pancreas organs, but progress has also occurred in single-donor success, with 10 sites reporting increased single-donor engraftment. The next wave of innovative clinical trial interventions will address instant blood-mediated inflammatory reaction (IBMIR), apoptosis, and inflammation, and will translate into further marked improvements in single-donor success. Effective control of auto- and alloimmunity is the key to long-term islet function, and high-resolution cellular and antibody-based assays will add considerable precision to this process. Advances in immunosuppression, with new antibody-based targeting of costimulatory blockade and other T-B cellular signaling, will have further profound impact on the safety record of immunotherapy. Clinical trials will move forward shortly to test out new human stem cell derived islets, and in parallel trials will move forward, testing pig islets for compatibility in patients. Induction of immunological tolerance to self-islet antigens and to allografts is a difficult challenge, but potentially within our grasp.
Collapse
Affiliation(s)
- A M James Shapiro
- Clinical Islet Transplant Program, University of Alberta, 2000 College Plaza, 8215 112th Street, Edmonton AB Canada T6G 2C8.
| |
Collapse
|
41
|
Noguchi H, Naziruddin B, Jackson A, Shimoda M, Ikemoto T, Fujita Y, Chujo D, Takita M, Peng H, Sugimoto K, Itoh T, Kobayashi N, Onaca N, Levy MF, Matsumoto S. Fresh islets are more effective for islet transplantation than cultured islets. Cell Transplant 2012; 21:517-23. [PMID: 22793060 DOI: 10.3727/096368911x605439] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
For clinical islet transplantation, isolated islets deteriorate rapidly in culture, although culturing islets prior to transplantation provides flexibility for evaluation of isolated islets and pretreatment of patients. In the present study, we compared human fresh islets to cultured islets with in vitro and in vivo assays. After culture for 24, 48, and 72 h, islet yield significantly decreased from 2,000 to 1,738 ± 26 (13% loss), 1,525 ± 30 (24% loss), or 1,298 ± 18 IEQ (35% loss), respectively. The ATP contents were significantly higher in the 6-h cultured group (near fresh group) than in 48-h culture groups. The stimulation index was relatively higher in the 6-h cultured group than in 48-h cultured group. Human islets with or without culture were transplanted into diabetic nude mice. The attainability of posttransplantation normoglycemia was significantly higher in fresh group than in the culture groups. Intraperitoneal glucose tolerance testing (IPGTT) showed that the blood glucose levels of mice transplanted with fresh islets were significantly lower than with cultured islets at 30, 60, 90, and 120 min after injection. These data suggest that human islet transplantation without culture could avoid the deterioration of islets during culture and improve the outcome of islet transplantation. Based on these data, we have transplanted fresh islets without culture for our current clinical islet transplantation protocol.
Collapse
Affiliation(s)
- Hirofumi Noguchi
- Baylor All Saints Medical Center, Baylor Research Institute, Fort Worth, TX, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kaddis JS, Hanson MS, Cravens J, Qian D, Olack B, Antler M, Papas KK, Iglesias I, Barbaro B, Fernandez L, Powers AC, Niland JC. Standardized transportation of human islets: an islet cell resource center study of more than 2,000 shipments. Cell Transplant 2012; 22:1101-11. [PMID: 22889479 DOI: 10.3727/096368912x653219] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Preservation of cell quality during shipment of human pancreatic islets for use in laboratory research is a crucial, but neglected, topic. Mammalian cells, including islets, have been shown to be adversely affected by temperature changes in vitro and in vivo, yet protocols that control for thermal fluctuations during cell transport are lacking. To evaluate an optimal method of shipping human islets, an initial assessment of transportation conditions was conducted using standardized materials and operating procedures in 48 shipments sent to a central location by eight pancreas-processing laboratories using a single commercial airline transporter. Optimization of preliminary conditions was conducted, and human islet quality was then evaluated in 2,338 shipments pre- and postimplementation of a finalized transportation container and standard operating procedures. The initial assessment revealed that the outside temperature ranged from a mean of -4.6 ± 10.3°C to 20.9 ± 4.8°C. Within-container temperature drops to or below 15°C occurred in 16 shipments (36%), while the temperature was found to be stabilized between 15°C and 29°C in 29 shipments (64%). Implementation of an optimized transportation container and operating procedure reduced the number of within-container temperature drops (≤ 15°C) to 13% (n = 37 of 289 winter shipments), improved the number desirably maintained between 15°C and 29°C to 86% (n = 250), but also increased the number reaching or exceeding 29°C to 1% (n = 2; overall p < 0.0001). Additionally, postreceipt quality ratings of excellent to good improved pre- versus postimplantation of the standardized protocol, adjusting for preshipment purity/viability levels (p < 0.0001). Our results show that extreme temperature fluctuations during transport of human islets, occurring when using a commercial airline transporter for long distance shipping, can be controlled using standardized containers, materials, and operating procedures. This cost-effective and pragmatic standardized protocol for the transportation of human islets can potentially be adapted for use with other mammalian cell systems and is available online at http://iidp.coh.org/sops.aspx.
Collapse
Affiliation(s)
- John S Kaddis
- Department of Information Sciences, City of Hope, Duarte, CA 91010-3000, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Williams SJ, Huang HH, Kover K, Moore W, Berkland C, Singh M, Smirnova IV, MacGregor R, Stehno-Bittel L. Reduction of diffusion barriers in isolated rat islets improves survival, but not insulin secretion or transplantation outcome. Organogenesis 2012; 6:115-24. [PMID: 20885858 DOI: 10.4161/org.6.2.10373] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Accepted: 10/20/2009] [Indexed: 01/05/2023] Open
Abstract
For people with type 1 diabetes and severe hypoglycemic unawareness, islet transplants offer hope for improving the quality of life. However, islet cell death occurs quickly during or after transplantation, requiring large quantities of islets per transplant. The purpose of this study was to determine whether poor function demonstrated in large islets was a result of diffusion barriers and if removing those barriers could improve function and transplantation outcomes. Islets were isolated from male DA rats and measured for cell viability, islet survival, glucose diffusion and insulin secretion. Modeling of diffusion barriers was completed using dynamic partial differential equations for a sphere. Core cell death occurred in 100% of the large islets (diameter >150 µm), resulting in poor survival within 7 days after isolation. In contrast, small islets (diameter <100 µm) exhibited good survival rates in culture (91%). Glucose diffusion into islets was tracked with 2-NBDG; 4.2 µm/min in small islets and 2.8 µm/min in large islets. 2-NBDG never permeated to the core cells of islets larger than 150 µm diameter. Reducing the diffusion barrier in large islets improved their immediate and long-term viability in culture. However, reduction of the diffusion barrier in large islets failed to improve their inferior in vitro insulin secretion compared to small islets, and did not return glucose control to diabetic animals following transplantation. Thus, diffusion barriers lead to low viability and poor survival for large islets, but are not solely responsible for the inferior insulin secretion or poor transplantation outcomes of large versus small islets.
Collapse
Affiliation(s)
- S Janette Williams
- Department of Physical Therapy and Rehabilitation Science, University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Hermann M, Wurm M, Lubei V, Pirkebner D, Draxl A, Margreiter R, Hengster P. Keep on rolling: optimizing human islet transport conditions using a perfused rotary system. Islets 2012; 4:152-7. [PMID: 22627172 DOI: 10.4161/isl.19753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The setup of an islet isolation facility designed along the rules of good manufacturing practice (GMP) is a technically challenging, cost and time intensive process. ( 1) Consequently, several institutions have decided to perform transplantation of islets isolated at another center with an already standing expertise. Such a solution includes the necessity to transport the isolated islets from the isolation to the transplantation facility. In spite of its importance, an ideal solution for the transport of the isolated human islets has still not been established. Here, we present an islet transport device suited to transport human islet cells under reproducible conditions and minimized cell stress. The transport simulation of the human islets was performed in a transfused "rotary transport system for islets" termed "ROTi." Besides measuring standard metabolic (LDH, lactate, glucose) and physical parameters (pH, dissolved oxygen and temperature), we used five different live stains in combination with real time live confocal microscopy to document islet quality parameters. As live stains we added tetramethylrhodamine methyl ester, cell permeant acetoxymethylester, propidium iodide, annexin-fitc and fluorescent wheat germ agglutinin, and assessed mitochondrial membrane potentials, calcium levels, cell death, apoptosis or cell morphology, respectively. We compared the viability of human islets after 24 h incubation in the ROTi device to an incubation simulating "standard" shipment of islets in 50 ml tubes. All cell viability parameters studied (mitochondrial membrane potentials, calcium content, apoptosis, cell death as well as cell morphology) documented a significantly better cell viability in the ROTi fraction compared with the simulated "standard" shipment fraction. Besides maintaining islet cell viability, the ROTi bears the advantage of a better reproducibility of islet transport conditions.
Collapse
Affiliation(s)
- Martin Hermann
- KMT Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Innsbruck Medical University, Innsbruck, Tyrol, Austria.
| | | | | | | | | | | | | |
Collapse
|
45
|
Kuise T, Noguchi H. Recent progress in pancreatic islet transplantation. World J Transplant 2011; 1:13-8. [PMID: 24175188 PMCID: PMC3782227 DOI: 10.5500/wjt.v1.i1.13] [Citation(s) in RCA: 8] [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] [Received: 07/14/2011] [Revised: 10/26/2011] [Accepted: 12/19/2011] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus remains a major burden. More than 200 million people are affected worldwide, which represents 6% of the world’s population. Type 1 diabetes mellitus is an autoimmune disease, which induces the permanent destruction of the β-cells of the pancreatic islets of Langerhans. Although intensive insulin therapy has proven effective to delay and sometimes prevent the progression of complications such as nephropathy, neuropathy or retinopathy, it is difficult to achieve and maintain long term in most subjects. The successes achieved over the last few decades by the transplantation of whole pancreas and isolated islets suggest that diabetes can be cured by the replenishment of deficient β cells. However, islet transplantation efforts have various limitations, including the limited supply of donor pancreata, the paucity of experienced islet isolation teams, side effects of immunosuppressants and poor long term results. The purpose of this article is to review the recent progress in clinical islet transplantation for the treatment of diabetes and to describe the recent progress on pancreatic stem/progenitor cell research, which has opened up several possibilities for the development of new treatments for diabetes.
Collapse
Affiliation(s)
- Takashi Kuise
- Takashi Kuise, Hirofumi Noguchi, Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | | |
Collapse
|
46
|
Aldibbiat A, Huang GC, Zhao M, Holliman GN, Ferguson L, Hughes S, Brigham K, Wardle J, Williams R, Dickinson A, White SA, Johnson PRV, Manas D, Amiel SA, Shaw JAM. Validation of Islet Transport From a Geographically Distant Isolation Center Enabling Equitable Access and National Health Service Funding of a Clinical Islet Transplant Program for England. CELL MEDICINE 2011; 2:97-104. [PMID: 27004135 DOI: 10.3727/215517911x617905] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Islet transplantation has become established as a successful treatment for type 1 diabetes complicated by recurrent severe hypoglycemia. In the UK access has been limited to a few centrally located units. Our goal was to validate a quality-assured system for safe/effective transport of human islets in the UK and to successfully undertake the first transplants with transported islets. Pancreases were retrieved from deceased donors in the north of England and transported to King's College London using two-layer method (TLM) or University of Wisconsin solution alone. Islets were isolated and transported back to Newcastle in standard blood transfusion or gas-permeable bags with detailed evaluation pre- and posttransport. In the preclinical phase, islets were isolated from 10 pancreases with mean yield of 258,000 islet equivalents. No significant differences were seen between TLM and University of Wisconsin solution organ preservation. A significant loss of integrity was demonstrated in islets shipped in gas-permeable bags, whereas sterility, number, purity, and viability were maintained in blood transfusion bags. Maintenance of secretory granules and glucose-stimulated insulin secretion was confirmed following transport. A Standard Operating Procedure enabling final pretransplant quality control from a simple side-arm sample was validated. Moreover, levels of insulin and cytokines in transport medium were low, enabling transplant without centrifugation/resuspension at the recipient site. Six clinical transplants of transported islets were undertaken in five recipients with 100% primary graft function and resolution of severe hypoglycemia. Safe and clinically effective islet transport has been established facilitating sustainable NHS funding of a clinical islet transplant program for the UK.
Collapse
Affiliation(s)
- Ali Aldibbiat
- Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Guo Cai Huang
- † Division of Diabetes and Nutritional Sciences, King's College London , London , UK
| | - Min Zhao
- † Division of Diabetes and Nutritional Sciences, King's College London , London , UK
| | - Graham N Holliman
- Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Linda Ferguson
- Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Stephen Hughes
- ‡ Nuffield Department of Surgical Sciences, University of Oxford , Oxford , UK
| | - Ken Brigham
- § Department of Haematology, Newcastle University , Newcastle upon Tyne , UK
| | - Julie Wardle
- ¶ Institute of Transplantation, Freeman Hospital , Newcastle upon Tyne , UK
| | - Rob Williams
- ¶ Institute of Transplantation, Freeman Hospital , Newcastle upon Tyne , UK
| | - Anne Dickinson
- § Department of Haematology, Newcastle University , Newcastle upon Tyne , UK
| | - Steven A White
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK; ¶Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Paul R V Johnson
- ‡ Nuffield Department of Surgical Sciences, University of Oxford , Oxford , UK
| | - Derek Manas
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK; ¶Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, UK
| | - Stephanie A Amiel
- † Division of Diabetes and Nutritional Sciences, King's College London , London , UK
| | - James A M Shaw
- Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| |
Collapse
|
47
|
Abstract
Autologous islet transplantation (AIT) is performed to prevent surgical diabetes after total or semi-total pancreatectomy for the treatment of chronic pancreatitis with severe abdominal pain. In addition, AIT is used in cases of benign pancreatic tumors and pancreatic trauma. It has been shown that AIT results in better outcomes in terms of glycemic control compared with allogeneic islet transplantation. The reasons for the favorable outcomes of AIT are thought to be: (i) patients have no autoimmune diseases; (ii) the transplanted islets do not suffer allogeneic rejection; (iii) diabetogenic antirejection drugs are not required; (iv) pancreata do not undergo a cytokine storm as a result of periods of brain death; (v) the period of cold preservation of retrieved pancreata is short; (vi) the isolated islets are immediately transplanted without culture; and (vii) pancreata with pancreatitis may contain more progenitor cells. Further research into AIT would help improve the results of allogeneic islet transplantation. Conversely, the technical difficulties associated with islet isolation appear to be the largest hurdle for AIT; therefore, remote center islet isolation may prove to be key in the promotion of this treatment.
Collapse
Affiliation(s)
- Shinichi Matsumoto
- Baylor All Saints Islet Cell Laboratory, Baylor Research Institute Fort Worth Campus, Fort Worth, Texas 76104, USA.
| |
Collapse
|
48
|
Association Between the Secretory Unit of Islet Transplant Objects Index and Satisfaction With Insulin Therapy Among Insulin-Dependent Islet Recipients. Transplant Proc 2011; 43:3250-5. [DOI: 10.1016/j.transproceed.2011.10.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
49
|
Jahansouz C, Jahansouz C, Kumer SC, Brayman KL. Evolution of β-Cell Replacement Therapy in Diabetes Mellitus: Islet Cell Transplantation. J Transplant 2011; 2011:247959. [PMID: 22013505 PMCID: PMC3195999 DOI: 10.1155/2011/247959] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Accepted: 08/08/2011] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus remains one of the leading causes of morbidity and mortality worldwide. According to the Centers for Disease Control and Prevention, approximately 23.6 million people in the United States are affected. Of these individuals, 5 to 10% have been diagnosed with Type 1 diabetes mellitus (T1DM), an autoimmune disease. Although it often appears in childhood, T1DM may manifest at any age, leading to significant morbidity and decreased quality of life. Since the 1960s, the surgical treatment for diabetes mellitus has evolved to become a viable alternative to insulin administration, beginning with pancreatic transplantation. While islet cell transplantation has emerged as another potential alternative, its role in the treatment of T1DM remains to be solidified as research continues to establish it as a truly viable alternative for achieving insulin independence. In this paper, the historical evolution, procurement, current status, benefits, risks, and ongoing research of islet cell transplantation are explored.
Collapse
Affiliation(s)
- Cyrus Jahansouz
- School of Medicine, University of Virginia, Charlottesville, VA 22102, USA
| | | | | | | |
Collapse
|
50
|
Khan A, Jindal RM, Shriver C, Guy SR, Vertrees AE, Wang X, Xu X, Szust J, Ricordi C. Remote processing of pancreas can restore normal glucose homeostasis in autologous islet transplantation after traumatic whipple pancreatectomy: technical considerations. Cell Transplant 2011; 21:1261-7. [PMID: 21944862 DOI: 10.3727/096368911x600984] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
An emergency autologous islet transplant after a traumatic Whipple operation and subsequent total pancreatectomy was performed for a 21-year-old patient who was wounded with multiple abdominal gunshot wounds. After Whipple pancreatectomy, the remnant pancreas (63.5 g), along with other damaged organs, was removed by the surgeons at Walter Reed Army Medical Center (WRAMC) and shipped to Diabetes Research Institute (DRI) for islet isolation. The pancreas was preserved in UW solution for 9.25 h prior to islet isolation. Upon arrival, the organ was visually inspected; the pancreatic head was missing, the rest of the pancreas was damaged and full of blood; the tail looked normal. A 16-gauge catheter was inserted into the main duct and directed towards tail of the pancreas after the dissection of main duct in the midbody of the pancreas. The pancreas was distended with collagenase solution (Roche MTF) through the catheter. During 10 min of intraductal delivery of enzyme, the gland was distended uniformly. No leakage of the solution was observed. The pancreas was transferred to a Ricordi chamber for automated mechanical and enzymatic digestion. Islets were purified using a COBE 2991 cell processor. Islet equivalents (IEQ; 221,250) of 40% purity and 90% viability were recovered during the isolation, which were shipped back to WRAMC and infused by intraportal injection into the patient. Immediate islet function was demonstrated by the rapid elevation of serum C peptide followed by insulin independence with near normal oral glucose tolerance test (OGTT) 1 and 2 months later. It is possible to restore near normal glucose tolerance with autologous islet transplantation after total pancreatectomy even with suboptimal number of islets while confirming that islets processed at a remote site are suitable for transplantation.
Collapse
Affiliation(s)
- Aisha Khan
- Diabetes Research Institute and Cell Transplant Center, University of Miami, Miami, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|