1
|
Zhao X, Xue W, Ding W, Qiao Y, Chu X, Qiu Y, Tang M, Sun D, Fu X. A novel injectable sodium alginate/chitosan/sulfated bacterial cellulose hydrogel as biohybrid artificial pancreas for real-time glycaemic regulation. Carbohydr Polym 2025; 354:123323. [PMID: 39978905 DOI: 10.1016/j.carbpol.2025.123323] [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] [Received: 09/01/2024] [Revised: 01/19/2025] [Accepted: 01/26/2025] [Indexed: 02/22/2025]
Abstract
Type 1 diabetes mellitus (T1DM) are characterized by blood glucose elevation with pancreatic β cells deficiency. As a safe alternative to frequent subcutaneous insulin injection, pancreatic β cell transplantation provides a promising therapeutic option for blood glucose control in T1DM. However, pancreatic β cell transplantation faces intractable challenges of the poor viability and severe host immune rejection. Therefore, a novel approach capable of improving the poor oxygen/nutrients supply and severe host immune rejection is highly desired. Herein, a novel biohybrid artificial pancreas, presenting glucose-dependent insulin release behavior, is constructed via pancreatic β cells encapsulating in a hydrogel scaffold. The hydrogel scaffold is made of the commixture of sodium alginate (SA), chitosan (CS) and sulfated bacterial cellulose (SBC). The biocompatible three-dimensional (3D) hydrogels protected pancreatic β cells from immune response but also allowed the exchange of nutrients and insulin. As a result of the synergistic effect, the biohybrid artificial pancreas can reverse the hyperglycemia and achieve sustained glycemic control for at least 30 days in diabetic mice. Collectively, we consider that this biohybrid artificial pancreas with an elaborate structure could provide an effective option for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Xiang Zhao
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China; The Fourth Affiliated Hospital of Nanjing Medical University, China
| | - Wei Xue
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weixiao Ding
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, China
| | - Yalei Qiao
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, China
| | - Xuehui Chu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yudong Qiu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Min Tang
- Department of Imaging, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Dongping Sun
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, China.
| | - Xiao Fu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China; Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, Nanjing 210094, China.
| |
Collapse
|
2
|
Andreadi A, Lodeserto P, Todaro F, Meloni M, Romano M, Minasi A, Bellia A, Lauro D. Nanomedicine in the Treatment of Diabetes. Int J Mol Sci 2024; 25:7028. [PMID: 39000136 PMCID: PMC11241380 DOI: 10.3390/ijms25137028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Nanomedicine could improve the treatment of diabetes by exploiting various therapeutic mechanisms through the use of suitable nanoformulations. For example, glucose-sensitive nanoparticles can release insulin in response to high glucose levels, mimicking the physiological release of insulin. Oral nanoformulations for insulin uptake via the gut represent a long-sought alternative to subcutaneous injections, which cause pain, discomfort, and possible local infection. Nanoparticles containing oligonucleotides can be used in gene therapy and cell therapy to stimulate insulin production in β-cells or β-like cells and modulate the responses of T1DM-associated immune cells. In contrast, viral vectors do not induce immunogenicity. Finally, in diabetic wound healing, local delivery of nanoformulations containing regenerative molecules can stimulate tissue repair and thus provide a valuable tool to treat this diabetic complication. Here, we describe these different approaches to diabetes treatment with nanoformulations and their potential for clinical application.
Collapse
Affiliation(s)
- Aikaterini Andreadi
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Pietro Lodeserto
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Federica Todaro
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
| | - Marco Meloni
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Maria Romano
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Alessandro Minasi
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Alfonso Bellia
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| | - Davide Lauro
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (P.L.); (F.T.); (M.M.); (A.B.); (D.L.)
- Division of Endocrinology and Diabetology, Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (M.R.); (A.M.)
| |
Collapse
|
3
|
Sremac M, Luo H, Deng H, Parr MFE, Hutcheson J, Verde PS, Alagpulinsa DA, Kitzmann JM, Papas KK, Brauns T, Markmann JF, Lei J, Poznansky MC. Short-term function and immune-protection of microencapsulated adult porcine islets with alginate incorporating CXCL12 in healthy and diabetic non-human primates without systemic immune suppression: A pilot study. Xenotransplantation 2023; 30:e12826. [PMID: 37712342 DOI: 10.1111/xen.12826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/10/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023]
Abstract
Replacement of insulin-producing pancreatic beta-cells by islet transplantation offers a functional cure for type-1 diabetes (T1D). We recently demonstrated that a clinical grade alginate micro-encapsulant incorporating the immune-repellent chemokine and pro-survival factor CXCL12 could protect and sustain the integrity and function of autologous islets in healthy non-human primates (NHPs) without systemic immune suppression. In this pilot study, we examined the impact of the CXCL12 micro encapsulant on the function and inflammatory and immune responses of xenogeneic islets transplanted into the omental tissue bilayer sac (OB; n = 4) and diabetic (n = 1) NHPs. Changes in the expression of cytokines after implantation were limited to 2-6-fold changes in blood, most of which did not persist over the first 4 weeks after implantation. Flow cytometry of PBMCs following transplantation showed minimal changes in IFNγ or TNFα expression on xenoantigen-specific CD4+ or CD8+ T cells compared to unstimulated cells, and these occurred mainly in the first 4 weeks. Microbeads were readily retrievable for assessment at day 90 and day 180 and at retrieval were without microscopic signs of degradation or foreign body responses (FBR). In vitro and immunohistochemistry studies of explanted microbeads indicated the presence of functional xenogeneic islets at day 30 post transplantation in all biopsied NHPs. These results from a small pilot study revealed that CXCL12-microencapsulated xenogeneic islets abrogate inflammatory and adaptive immune responses to the xenograft. This work paves the way toward future larger scale studies of the transplantation of alginate microbeads with CXCL12 and porcine or human stem cell-derived beta cells or allogeneic islets into diabetic NHPs without systemic immunosuppression.
Collapse
Affiliation(s)
- Marinko Sremac
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hao Luo
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, China
| | - Hongping Deng
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Madeline F E Parr
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Pushkar S Verde
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David A Alagpulinsa
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jenna Miner Kitzmann
- Department of Surgery, Institute for Cellular Transplantation, University of Arizona, Tucson, Arizona, USA
| | - Klearchos K Papas
- Department of Surgery, Institute for Cellular Transplantation, University of Arizona, Tucson, Arizona, USA
| | - Timothy Brauns
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James F Markmann
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ji Lei
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Kaneko M, Moriguchi H, Futatsubashi R, Ayano S, Kobayashi G, Ito A. Transplantable cell-encapsulation device using a semipermeable ethylene-vinyl alcohol copolymer membrane in a mouse diabetic model. J Biosci Bioeng 2023; 136:415-422. [PMID: 37748982 DOI: 10.1016/j.jbiosc.2023.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/27/2023] [Accepted: 09/03/2023] [Indexed: 09/27/2023]
Abstract
Cell-based therapy is an attractive approach, and encapsulation of therapeutic cells is a promising strategy because it prevents immune responses and allows transplanted cells to be retrieved in case of dysfunction. Bioartificial pancreas, in which insulin-secreting cells are encapsulated in a semipermeable membrane bag, is a new class of medical device for treating type-I diabetes. In this study, we developed a macroencapsulation device in which the pancreatic beta cell line MIN6 was encapsulated in a semipermeable bag made of an ethylene-vinyl alcohol copolymer membrane. In vitro evaluation of ATP and insulin levels revealed that MIN6 cells grown in Matrigel within the device secreted insulin in response to glucose levels. Transplantation of the device lowered blood glucose levels for 30 days in diabetic mice. Histological observation revealed that MIN6 cells formed spheroids in Matrigel, and no host cells were detected within the device. Blood levels of inflammatory cytokines in the transplanted mice were similar to those in non-transplanted mice, and antibody levels in the device were lower than those in the intraperitoneal fluid. These results suggest that the semipermeable ethylene-vinyl alcohol copolymer membrane developed in this study is useful for cell encapsulation in cell-based therapies, including beta-cell macroencapsulation for type-1 diabetes.
Collapse
Affiliation(s)
- Masahiro Kaneko
- Department of Chemical Systems Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Hiroaki Moriguchi
- Department of Chemical Systems Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Ryo Futatsubashi
- Department of Chemical Systems Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Satoru Ayano
- Research and Development Division, Kuraray Co., Ltd., 41 Miyukigaoka, Tsukuba, Ibaraki 305-0841, Japan
| | - Goro Kobayashi
- Research and Development Division, Kuraray Co., Ltd., 41 Miyukigaoka, Tsukuba, Ibaraki 305-0841, Japan
| | - Akira Ito
- Department of Chemical Systems Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.
| |
Collapse
|
5
|
Johnson CD, Aranda-Espinoza H, Fisher JP. A Case for Material Stiffness as a Design Parameter in Encapsulated Islet Transplantation. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:334-346. [PMID: 36475851 PMCID: PMC10442690 DOI: 10.1089/ten.teb.2022.0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Diabetes is a disease that plagues over 463 million people globally. Approximately 40 million of these patients have type 1 diabetes mellitus (T1DM), and the global incidence is increasing by up to 5% per year. T1DM is where the body's immune system attacks the pancreas, specifically the pancreatic beta cells, with antibodies to prevent insulin production. Although current treatments such as exogenous insulin injections have been successful, exorbitant insulin costs and meticulous administration present the need for alternative long-term solutions to glucose dysregulation caused by diabetes. Encapsulated islet transplantation (EIT) is a tissue-engineered solution to diabetes. Donor islets are encapsulated in a semipermeable hydrogel, allowing the diffusion of oxygen, glucose, and insulin but preventing leukocyte infiltration and antibody access to the transplanted cells. Although successful in small animal models, EIT is still far from commercial use owing to necessary long-term systemic immunosuppressants and consistent immune rejection. Most published research has focused on tailoring the characteristics of the capsule material to promote clinical viability. However, most studies have been limited in scope to biochemical changes. Current mechanobiology studies on the effect of substrate stiffness on the function of leukocytes, especially macrophages-primary foreign body response (FBR) orchestrators, show promise in tailoring a favorable response to tissue-engineered therapies such as EIT. In this review, we explore strategies to improve the clinical viability of EIT. A brief overview of the immune system, the FBR, and current biochemical approaches will be elucidated throughout this exploration. Furthermore, an argument for using substrate stiffness as a capsule design parameter to increase EIT efficacy and clinical viability will be posed.
Collapse
Affiliation(s)
- Courtney D. Johnson
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Fischell Department of Bioengineering, Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - Helim Aranda-Espinoza
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - John P. Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Fischell Department of Bioengineering, Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
6
|
Chendke GS, Kharbikar BN, Ashe S, Faleo G, Sneddon JB, Tang Q, Hebrok M, Desai TA. Replenishable prevascularized cell encapsulation devices increase graft survival and function in the subcutaneous space. Bioeng Transl Med 2023; 8:e10520. [PMID: 37476069 PMCID: PMC10354771 DOI: 10.1002/btm2.10520] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/06/2023] [Accepted: 03/29/2023] [Indexed: 07/22/2023] Open
Abstract
Beta cell replacement therapy (BCRT) for patients with type 1 diabetes (T1D) improves blood glucose regulation by replenishing the endogenous beta cells destroyed by autoimmune attack. Several limitations, including immune isolation, prevent this therapy from reaching its full potential. Cell encapsulation devices used for BCRT provide a protective physical barrier for insulin-producing beta cells, thereby protecting transplanted cells from immune attack. However, poor device engraftment posttransplantation leads to nutrient deprivation and hypoxia, causing metabolic strain on transplanted beta cells. Prevascularization of encapsulation devices at the transplantation site can help establish a host vascular network around the implant, increasing solute transport to the encapsulated cells. Here, we present a replenishable prevascularized implantation methodology (RPVIM) that allows for the vascular integration of replenishable encapsulation devices in the subcutaneous space. Empty encapsulation devices were vascularized for 14 days, after which insulin-producing cells were inserted without disrupting the surrounding vasculature. The RPVIM devices were compared with nonprevascularized devices (Standard Implantation Methodology [SIM]) and previously established prevascularized devices (Standard Prevascularization Implantation Methodology [SPVIM]). Results show that over 75% of RPVIM devices containing stem cell-derived insulin-producing beta cell clusters showed a signal after 28 days of implantation in subcutaneous space. Notably, not only was the percent of RPVIM devices showing signal significantly greater than SIM and SPVIM devices, but the intraperitoneal glucose tolerance tests and histological analyses showed that encapsulated stem-cell derived insulin-producing beta cell clusters retained their function in the RPVIM devices, which is crucial for the successful management of T1D.
Collapse
Affiliation(s)
- Gauree S. Chendke
- UC Berkeley ‐ UCSF Graduate Program in BioengineeringSan FranciscoCaliforniaUSA
- Department of Bioengineering and Therapeutic SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Bhushan N. Kharbikar
- Department of Bioengineering and Therapeutic SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Sudipta Ashe
- Diabetes Center, University of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Gaetano Faleo
- Department of SurgeryUCSF Gladstone Institute of Genome ImmunologySan FranciscoCaliforniaUSA
| | - Julie B. Sneddon
- Diabetes Center, University of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Cell and Tissue BiologyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of AnatomyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchSan FranciscoCaliforniaUSA
| | - Qizhi Tang
- Diabetes Center, University of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of SurgeryUCSF Gladstone Institute of Genome ImmunologySan FranciscoCaliforniaUSA
| | - Matthias Hebrok
- Diabetes Center, University of California, San FranciscoSan FranciscoCaliforniaUSA
- Center for Organoid Systems, Technical University MunichGarchingGermany
- Institute for Diabetes Organoid Technology, Helmholtz Munich, Helmholtz Diabetes CenterNeuherbergGermany
| | - Tejal A. Desai
- UC Berkeley ‐ UCSF Graduate Program in BioengineeringSan FranciscoCaliforniaUSA
- Department of Bioengineering and Therapeutic SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Diabetes Center, University of California, San FranciscoSan FranciscoCaliforniaUSA
- School of Engineering, Brown UniversityProvidenceRhode IslandUSA
| |
Collapse
|
7
|
Tissue engineering of decellularized pancreas scaffolds for regenerative medicine in diabetes. Acta Biomater 2023; 157:49-66. [PMID: 36427686 DOI: 10.1016/j.actbio.2022.11.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
Diabetes mellitus is a global disease requiring long-term treatment and monitoring. At present, pancreas or islet transplantation is the only reliable treatment for achieving stable euglycemia in Type I diabetes patients. However, the shortage of viable pancreata for transplantation limits the use of this therapy for the majority of patients. Organ decellularization and recellularization is emerging as a promising solution to overcome the shortage of viable organs for transplantation by providing a potential alternative source of donor organs. Several studies on decellularization and recellularization of rodent, porcine, and human pancreata have been performed, and show promise for generating usable decellularized pancreas scaffolds for subsequent recellularization and transplantation. In this state-of-the-art review, we provide an overview of the latest advances in pancreas decellularization, recellularization, and revascularization. We also discuss clinical considerations such as potential transplantation sites, donor source, and immune considerations. We conclude with an outlook on the remaining work that needs to be done in order to realize the goal of using this technology to create bioengineered pancreata for transplantation in diabetes patients. STATEMENT OF SIGNIFICANCE: Pancreas or islet transplantation is a means of providing insulin-independence in diabetes patients. However, due to the shortage of viable pancreata, whole-organ decellularization and recellularization is emerging as a promising solution to overcome organ shortage for transplantation. Several studies on decellularization and recellularization of rodent, porcine, and human pancreata have shown promise for generating usable decellularized pancreas scaffolds for subsequent recellularization and transplantation. In this state-of-the-art review, we highlight the latest advances in pancreas decellularization, recellularization, and revascularization. We also discuss clinical considerations such as potential transplantation sites, donor source, and immune considerations. We conclude with future work that needs to be done in order to realize clinical translation of bioengineered pancreata for transplantation in diabetes patients.
Collapse
|
8
|
Yin J, Meng H, Lin J, Ji W, Xu T, Liu H. Pancreatic islet organoids-on-a-chip: how far have we gone? J Nanobiotechnology 2022; 20:308. [PMID: 35764957 PMCID: PMC9238112 DOI: 10.1186/s12951-022-01518-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/20/2022] [Indexed: 01/10/2023] Open
Abstract
Diabetes mellitus (DM) is a disease caused by dysfunction or disruption of pancreatic islets. The advent and development of microfluidic organoids-on-a-chip platforms have facilitated reproduce of complex and dynamic environment for tissue or organ development and complex disease processes. For the research and treatment of DM, the platforms have been widely used to investigate the physiology and pathophysiology of islets. In this review, we first highlight how pancreatic islet organoids-on-a-chip have improved the reproducibility of stem cell differentiation and organoid culture. We further discuss the efficiency of microfluidics in the functional evaluation of pancreatic islet organoids, such as single-islet-sensitivity detection, long-term real-time monitoring, and automatic glucose adjustment to provide relevant stimulation. Then, we present the applications of islet-on-a-chip technology in disease modeling, drug screening and cell replacement therapy. Finally, we summarize the development and challenges of islet-on-a-chip and discuss the prospects of future research.
Collapse
Affiliation(s)
- Jiaxiang Yin
- Bioland Laboratory, Guangzhou, Guangdong, China.,Guangzhou Laboratory, Guangzhou, Guangdong, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hao Meng
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | | | - Wei Ji
- Bioland Laboratory, Guangzhou, Guangdong, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Tao Xu
- Guangzhou Laboratory, Guangzhou, Guangdong, China. .,School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China.
| | - Huisheng Liu
- Bioland Laboratory, Guangzhou, Guangdong, China. .,Guangzhou Laboratory, Guangzhou, Guangdong, China. .,School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Lei J, Coronel MM, Yolcu ES, Deng H, Grimany-Nuno O, Hunckler MD, Ulker V, Yang Z, Lee KM, Zhang A, Luo H, Peters CW, Zou Z, Chen T, Wang Z, McCoy CS, Rosales IA, Markmann JF, Shirwan H, García AJ. FasL microgels induce immune acceptance of islet allografts in nonhuman primates. SCIENCE ADVANCES 2022; 8:eabm9881. [PMID: 35559682 PMCID: PMC9106299 DOI: 10.1126/sciadv.abm9881] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/30/2022] [Indexed: 05/23/2023]
Abstract
Islet transplantation to treat insulin-dependent diabetes is greatly limited by the need for maintenance immunosuppression. We report a strategy through which cotransplantation of allogeneic islets and streptavidin (SA)-FasL-presenting microgels to the omentum under transient rapamycin monotherapy resulted in robust glycemic control, sustained C-peptide levels, and graft survival in diabetic nonhuman primates for >6 months. Surgical extraction of the graft resulted in prompt hyperglycemia. In contrast, animals receiving microgels without SA-FasL under the same rapamycin regimen rejected islet grafts acutely. Graft survival was associated with increased number of FoxP3+ cells in the graft site with no significant changes in T cell systemic frequencies or responses to donor and third-party antigens, indicating localized tolerance. Recipients of SA-FasL microgels exhibited normal liver and kidney metabolic function, demonstrating safety. This localized immunomodulatory strategy succeeded with unmodified islets and does not require long-term immunosuppression, showing translational potential in β cell replacement for treating type 1 diabetes.
Collapse
Affiliation(s)
- Ji Lei
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - María M. Coronel
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Esma S. Yolcu
- Departments of Child Health and Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Department of Microbiology and Immunology, Institute for Cellular Therapeutics, University of Louisville, Louisville, KY, USA
| | - Hongping Deng
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Orlando Grimany-Nuno
- Department of Microbiology and Immunology, Institute for Cellular Therapeutics, University of Louisville, Louisville, KY, USA
| | - Michael D. Hunckler
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Vahap Ulker
- Departments of Child Health and Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Zhihong Yang
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kang M. Lee
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander Zhang
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hao Luo
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, China
| | - Cole W. Peters
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhongliang Zou
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tao Chen
- Cellular Therapy Department, Xiang’an Hospital, Xiamen University Medical School, Xiamen, China
| | - Zhenjuan Wang
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Colleen S. McCoy
- Division of Comparative Medicine, Massachusetts Institute of Technology, Boston, MA, USA
| | - Ivy A. Rosales
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James F. Markmann
- Center for Transplantation Science, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Haval Shirwan
- Departments of Child Health and Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Department of Microbiology and Immunology, Institute for Cellular Therapeutics, University of Louisville, Louisville, KY, USA
| | - Andrés J. García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
10
|
S. A. F, K. S. C, L. D, M. G, S. P, R. L. L, C. A. H. Engineering Vascularized Islet Macroencapsulation Devices: An in vitro Platform to Study Oxygen Transport in Perfused Immobilized Pancreatic Beta Cell Cultures. Front Bioeng Biotechnol 2022; 10:884071. [PMID: 35519615 PMCID: PMC9061948 DOI: 10.3389/fbioe.2022.884071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/01/2022] [Indexed: 02/01/2023] Open
Abstract
Islet encapsulation devices serve to deliver pancreatic beta cells to type 1 diabetic patients without the need for chronic immunosuppression. However, clinical translation is hampered by mass transport limitations causing graft hypoxia. This is exacerbated in devices relying only on passive diffusion for oxygenation. Here, we describe the application of a cylindrical in vitro perfusion system to study oxygen effects on islet-like clusters immobilized in alginate hydrogel. Mouse insulinoma 6 islet-like clusters were generated using microwell plates and characterized with respect to size distribution, viability, and oxygen consumption rate to determine an appropriate seeding density for perfusion studies. Immobilized clusters were perfused through a central channel at different oxygen tensions. Analysis of histological staining indicated the distribution of viable clusters was severely limited to near the perfusion channel at low oxygen tensions, while the distribution was broadest at normoxia. The results agreed with a 3D computational model designed to simulate the oxygen distribution within the perfusion device. Further simulations were generated to predict device performance with human islets under in vitro and in vivo conditions. The combination of experimental and computational findings suggest that a multichannel perfusion strategy could support in vivo viability and function of a therapeutic islet dose.
Collapse
Affiliation(s)
- Fernandez S. A.
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
| | - Champion K. S.
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
| | - Danielczak L.
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
| | - Gasparrini M.
- Human Islet Transplant Laboratory, McGill University Health Centre, Montréal, QC, Canada
| | - Paraskevas S.
- Human Islet Transplant Laboratory, McGill University Health Centre, Montréal, QC, Canada
- Department of Surgery, McGill University Health Centre, Montréal, QC, Canada
| | - Leask R. L.
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montréal, QC, Canada
| | - Hoesli C. A.
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montréal, QC, Canada
- *Correspondence: Hoesli C. A.,
| |
Collapse
|
11
|
Murray HE, Zafar A, Qureshi KM, Paget MB, Bailey CJ, Downing R. The potential role of multifunctional human amniotic epithelial cells in pancreatic islet transplantation. J Tissue Eng Regen Med 2021; 15:599-611. [PMID: 34216434 DOI: 10.1002/term.3214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/23/2021] [Indexed: 11/08/2022]
Abstract
Pancreatic islet cell transplantation has proven efficacy as a treatment for type 1 diabetes mellitus, chiefly in individuals who are refractory to conventional insulin replacement therapy. At present its clinical use is restricted, firstly by the limited access to suitable donor organs but also due to factors associated with the current clinical transplant procedure which inadvertently impair the long-term functionality of the islet graft. Of note, the physical, biochemical, inflammatory, and immunological stresses to which islets are subjected, either during pretransplant processing or following implantation are detrimental to their sustained viability, necessitating repeated islet infusions to attain adequate glucose control. Progressive decline in functional beta (β)-cell mass leads to graft failure and the eventual re-instatement of exogenous insulin treatment. Strategies which protect and/or preserve optimal islet function in the peri-transplant period would improve clinical outcomes. Human amniotic epithelial cells (HAEC) exhibit both pluripotency and immune-privilege and are ideally suited for use in replacement and regenerative therapies. The HAEC secretome exhibits trophic, anti-inflammatory, and immunomodulatory properties of relevance to islet graft survival. Facilitated by β-cell supportive 3D cell culture systems, HAEC may be integrated with islets bringing them into close spatial arrangement where they may exert paracrine influences that support β-cell function, reduce hypoxia-induced islet injury, and alter islet alloreactivity. The present review details the potential of multifunctional HAEC in the context of islet transplantation, with a focus on the innate capabilities that may counter adverse events associated with the current clinical transplant protocol to achieve long-term islet graft function.
Collapse
Affiliation(s)
- Hilary E Murray
- The Islet Research Laboratory, Worcester Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| | - Ali Zafar
- The Islet Research Laboratory, Worcester Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK.,Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Khalid M Qureshi
- The Islet Research Laboratory, Worcester Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK.,Bradford Royal Infirmary, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Michelle B Paget
- The Islet Research Laboratory, Worcester Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| | - Clifford J Bailey
- Diabetes Research, School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Richard Downing
- The Islet Research Laboratory, Worcester Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| |
Collapse
|
12
|
Huang L, Xiang J, Cheng Y, Xiao L, Wang Q, Zhang Y, Xu T, Chen Q, Xin H, Wang X. Regulation of Blood Glucose Using Islets Encapsulated in a Melanin-Modified Immune-Shielding Hydrogel. ACS APPLIED MATERIALS & INTERFACES 2021; 13:12877-12887. [PMID: 33689267 DOI: 10.1021/acsami.0c23010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Islet transplantation is currently a promising treatment for type 1 diabetes mellitus. However, the foreign body reaction and retrieval difficulty often lead to transplantation failure and hinder the clinical application. To address these two challenges, we propose a balanced charged sodium alginate-polyethyleneimine-melanin (SA-PEI-Melanin) threadlike hydrogel with immune shielding and retrievable properties. The attractiveness of this study lies in that the introduction of melanin can stimulate insulin secretion, especially under near-infrared (NIR) irradiation. After demonstrating a good immune-shielding effect, we performed an in vivo transplantation experiment. The results showed that the blood glucose level in the SA-PEI-Melanin group was stably controlled below the diabetic blood glucose criterion, and this blood glucose level could be further adjusted after NIR irradiation. In addition, the evaluation after retrieving the SA-PEI-Melanin hydrogel indicated that the islets still maintained a normal physiological function, further proving its excellent immunological protection. This study provides a new approach for the accurate regulation of blood glucose in patients with type 1 diabetes mellitus and contributes to developing a promising transplant system to reconcile real-time and precise light-defined insulin secretion regulation.
Collapse
Affiliation(s)
- Ling Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Jiajia Xiang
- College of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Yukai Cheng
- College of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Ling Xiao
- College of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Qingqing Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Yini Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Tieling Xu
- College of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Qianrui Chen
- College of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Hongbo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Xiaolei Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
- College of Chemistry, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| |
Collapse
|
13
|
Manay P, Turgeon N, Axelrod DA. Role of Whole Organ Pancreas Transplantation in the Day of Bioartificial and Artificial Pancreas. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00300-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
14
|
Gattás-Asfura KM, Abuid NJ, Labrada I, Stabler CL. Promoting Dendrimer Self-Assembly Enhances Covalent Layer-by-Layer Encapsulation of Pancreatic Islets. ACS Biomater Sci Eng 2020; 6:2641-2651. [PMID: 32587885 PMCID: PMC7316358 DOI: 10.1021/acsbiomaterials.9b01033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
For type 1 diabetics, islet transplantation can induce beneficial outcomes, including insulin independence and improved glycemic control. The long-term function of the grafted tissue, however, is challenged by host inflammatory and immune responses. Cell encapsulation can decrease detrimental host responses to the foreign implant, but standard microencapsulation imparts large transplant volumes and impaired metabolite and nutrient diffusion. To mitigate these effects, we developed an efficient covalent Layer-by-Layer (cLbL) approach for live-cell nanoencapsulation, based on oppositely charged hyperbranched polymers functionalized with complementary Staudinger ligation groups. Reliance on cationic polymers for cLbL, however, is problematic due to their poor biocompatibility. Herein, we incorporated the additional feature of supramolecular self-assembly of the dendritic polymers to enhance layer uniformity and decrease net polymer charge. Functionalization of poly (amino amide) (PAMAM) with triethoxysilane decreased polymer charge without compromising the uniformity and stability of resulting nanoscale islet coatings. Encapsulated pancreatic rat islets were viable and functional. The implantation of cLbL islets into diabetic mice resulted in stable normoglycemia, at equivalent dosage and efficiency as uncoated islets, with no observable alterations in cellular engraftment or foreign body responses. By balancing multi-functionality and self-assembly, nano-scale and stable covalent layer-by-layer polymeric coatings could be efficiently generated onto cellular organoids, presenting a highly adaptable platform for broad use in cellular transplantation.
Collapse
Affiliation(s)
- KM Gattás-Asfura
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - NJ Abuid
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - I Labrada
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - CL Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
- Interdisciplinary Program in Biomedical Sciences, University of Florida, Gainesville, FL, USA
- University of Florida Diabetes Institute, Gainesville, FL, USA
| |
Collapse
|
15
|
Zhang Y, Yang J, Zhang J, Li S, Zheng L, Zhang Y, Meng H, Zhang X, Wu Z. A bio-inspired injectable hydrogel as a cell platform for real-time glycaemic regulation. J Mater Chem B 2020; 8:4627-4641. [PMID: 32373901 DOI: 10.1039/d0tb00561d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Frequent subcutaneous insulin injection and islet transplantation are promising therapeutic options for type 1 diabetes mellitus. However, poor patient compliance, insufficient appropriate islet β cell donors and body immune rejection limit their clinical applications. The design of a platform capable of encapsulating insulin-secreting cells and achieving real-time blood glucose regulation, is a so far unmet need. Herein, inspired by the natural processes of regulating blood glucose in pancreatic islet β cells, we developed a poly(N-isopropylacrylamide-co-dextran-maleic acid-co-3-acrylamidophenylboronic acid) (P(AAPBA-Dex-NIPAM)) hydrogel as a cell platform with glucose responsiveness and thermo-responsiveness for the therapy of diabetes. This platform showed good biocompatibility against insulin-secreting cells and presented glucose-dependent insulin release behaviour. The bioinspired P(AAPBA6-Dex-NIPAM64) hydrogel had a positive effect on real-time glycaemic regulation, as observed by intraperitoneal glucose tolerance tests. The non-fasting blood glucose of diabetic rats was restored to a normal level during the period of treatment. Additionally, the inflammatory response did not occur after administration of the platform. Collectively, we expected that the bio-mimetic platform combined with an insulin-secreting capability could be a new diabetic treatment strategy.
Collapse
Affiliation(s)
- Yu Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Espona-Noguera A, Ciriza J, Cañibano-Hernández A, Orive G, Hernández RM, Saenz del Burgo L, Pedraz JL. Review of Advanced Hydrogel-Based Cell Encapsulation Systems for Insulin Delivery in Type 1 Diabetes Mellitus. Pharmaceutics 2019; 11:E597. [PMID: 31726670 PMCID: PMC6920807 DOI: 10.3390/pharmaceutics11110597] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/11/2022] Open
Abstract
: Type 1 Diabetes Mellitus (T1DM) is characterized by the autoimmune destruction of β-cells in the pancreatic islets. In this regard, islet transplantation aims for the replacement of the damaged β-cells through minimally invasive surgical procedures, thereby being the most suitable strategy to cure T1DM. Unfortunately, this procedure still has limitations for its widespread clinical application, including the need for long-term immunosuppression, the lack of pancreas donors and the loss of a large percentage of islets after transplantation. To overcome the aforementioned issues, islets can be encapsulated within hydrogel-like biomaterials to diminish the loss of islets, to protect the islets resulting in a reduction or elimination of immunosuppression and to enable the use of other insulin-producing cell sources. This review aims to provide an update on the different hydrogel-based encapsulation strategies of insulin-producing cells, highlighting the advantages and drawbacks for a successful clinical application.
Collapse
Affiliation(s)
- Albert Espona-Noguera
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Jesús Ciriza
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Alberto Cañibano-Hernández
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
- University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01006 Vitoria, Spain
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore
| | - Rosa María Hernández
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Laura Saenz del Burgo
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.E.-N.); (J.C.); (A.C.-H.); (R.M.H.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| |
Collapse
|
17
|
Chung H, Kim HJ, Kim JS, Yoon IH, Min BH, Shin JS, Kim JM, Lee WW, Park CG. CD4 + /CD8 + T-cell ratio correlates with the graft fate in pig-to-non-human primate islet xenotransplantation. Xenotransplantation 2019; 27:e12562. [PMID: 31642566 DOI: 10.1111/xen.12562] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/17/2019] [Accepted: 10/07/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND Xenogeneic islet transplantation using porcine pancreata has been a promising option for substituting human islet transplantation. Moreover, recent advances in pre-clinical results have put islet xenotransplantation closer to the possibility of clinical application. While preparing for the era of clinical xenotransplantation, developing non-invasive immune monitoring method which could predict the graft fate could benefit the patient. However, there are few reports showing predictive immune parameters associated with the fate of the graft in islet xenotransplantation. METHODS The absolute number and ratio of T-cell subsets have been measured via flow cytometry from the peripheral blood of 16 rhesus monkeys before and after porcine islet xenotransplantation. The correlation between the graft survival and the absolute number or ratio of T cells was retrospectively analyzed. RESULTS The ratio of CD4+ versus CD8+ T cells was significantly reduced due to CD8+ effector memory cells' increase. Correlation analyses revealed that CD4+ /CD8+ , CD4+ /CD8+ naïve, CD4+ naïve/CD8+ naïve, and CD4+ central memory/CD8+ naïve cell ratios negatively correlated with the duration of graft survival. Conversely, further analyses discovered strong, positive correlation of CD4+ /CD8+ cell ratios within the early graft-rejected monkeys (≤60 days). CONCLUSIONS This retrospective study demonstrated that CD4+ /CD8+ ratios correlated with graft survival, especially in recipients which rejected the graft in early post-transplantation periods. CD4+ /CD8+ ratios could be used as a surrogate marker to predict the graft fate in pig-to-NHP islet xenotransplantation.
Collapse
Affiliation(s)
- Hyunwoo Chung
- Seoul National University College of Medicine, Xenotransplantation Research Center, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Hyun-Je Kim
- Seoul National University College of Medicine, Xenotransplantation Research Center, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Jung-Sik Kim
- Seoul National University College of Medicine, Xenotransplantation Research Center, Seoul, Korea
| | - Il-Hee Yoon
- VHS Veterans Medical Research Institute, VHS Medical Center, Seoul, Korea
| | - Byoung-Hoon Min
- Seoul National University College of Medicine, Xenotransplantation Research Center, Seoul, Korea
| | - Jun-Seop Shin
- Seoul National University College of Medicine, Xenotransplantation Research Center, Seoul, Korea
| | - Jong-Min Kim
- Seoul National University College of Medicine, Xenotransplantation Research Center, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Korea
| | - Won-Woo Lee
- Seoul National University College of Medicine, Xenotransplantation Research Center, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Chung-Gyu Park
- Seoul National University College of Medicine, Xenotransplantation Research Center, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
18
|
Chendke GS, Faleo G, Juang C, Parent AV, Bernards DA, Hebrok M, Tang Q, Desai TA. Supporting Survival of Transplanted Stem-Cell-Derived Insulin-Producing Cells in an Encapsulation Device Augmented with Controlled Release of Amino Acids. ACTA ACUST UNITED AC 2019; 3. [PMID: 31633004 DOI: 10.1002/adbi.201900086] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Pancreatic islet transplantation is a promising treatment for type I diabetes, which is a chronic autoimmune disease in which the host immune cells attack insulin-producing beta cells. The impact of this therapy is limited due to tissue availability and dependence on immunosuppressive drugs that prevent immune rejection of the transplanted cells. These issues can be solved by encapsulating stem cell-derived insulin-producing cells in an immunoprotective device. However, encapsulation exacerbates ischemia, and the lack of vasculature at the implantation site post-transplantation worsens graft survival. Here, an encapsulation device that supplements nutrients to the cells is developed to improve the survival of encapsulated stem cell-derived insulin-producing cells in the poorly vascularized subcutaneous space. An internal compartment in the device is fabricated to provide zero-order release of alanine and glutamine for several weeks. The amino acid reservoir sustains viability of insulin-producing cells in nutrient limiting conditions in vitro. Moreover, the reservoir also increases cell survival by 30% after transplanting the graft in the subcutaneous space.
Collapse
Affiliation(s)
- Gauree S Chendke
- Department of Bioengineering and Therapeutic Sciences, University of California, 1700 4th Street, Byers Hall, Box 2520, San Francisco, CA 94158, USA
| | - Gaetano Faleo
- Department of Surgery, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Charity Juang
- UCSF Diabetes Center, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Audrey V Parent
- UCSF Diabetes Center, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Daniel A Bernards
- Department of Bioengineering and Therapeutic Sciences, University of California, 1700 4th Street, Byers Hall, Box 2520, San Francisco, CA 94158, USA
| | - Matthias Hebrok
- UCSF Diabetes Center, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Qizhi Tang
- Department of Surgery, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, 1700 4th Street, Byers Hall, Box 2520, San Francisco, CA 94158, USA
| |
Collapse
|
19
|
Preliminary Studies of the Impact of CXCL12 on the Foreign Body Reaction to Pancreatic Islets Microencapsulated in Alginate in Nonhuman Primates. Transplant Direct 2019; 5:e447. [PMID: 31165082 PMCID: PMC6511446 DOI: 10.1097/txd.0000000000000890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/04/2019] [Indexed: 12/18/2022] Open
Abstract
Supplemental Digital Content is available in the text. Background. We previously demonstrated that the incorporation of the chemokine CXCL12 into alginate microbeads supported long-term survival of microencapsulated auto-, allo-, and xenogeneic islets in murine models of diabetes without systemic immune suppression. The purpose of this study was to test whether CXCL12 could abrogate foreign body responses (FBRs) against alginate microbeads which were empty or contained autologous islets in healthy nonhuman primates (NHPs; n = 4). Methods. Two NHPs received intraperitoneal implants of 400 000 alginate microbeads with or without CXCL12, and postimplantation immunological and histopathological changes were evaluated up to 6 months postimplantation. A similar evaluation of autologous islets in CXCL12-containing alginate microbeads was performed in NHPs (n = 2). Results. CXCL12-containing alginate microbeads were associated with a markedly reduced FBR to microbeads. Host responses to microbead implants were minimal, as assessed by clinical observations, blood counts, and chemistry. Evaluation of encapsulated islets was limited by the development of necrotizing pancreatitis after hemipancreatectomy in 1 NHP. A limited number of functioning islets were detectable at 6 months posttransplantation in the second NHP. In general, empty microbeads or islet-containing beads were found to be evenly distributed through the intraperitoneal cavity and did not accumulate in the Pouch of Douglas. Conclusions. Inclusion of CXCL12 in alginate microbeads minimized localized FBR. The NHP autologous islet implant model had limited utility for excluding inflammatory/immune responses to implanted islets because of the complexity of pancreatic surgery (hemipancreatectomy) before transplantation and the need to microencapsulate and transplant encapsulated autologous islets immediately after pancreatectomy and islet isolation.
Collapse
|
20
|
Zafar A, Lee J, Yesmin S, Paget MB, Bailey CJ, Murray HE, Downing R. Rotational culture and integration with amniotic stem cells reduce porcine islet immunoreactivity in vitro and slow xeno-rejection in a murine model of islet transplantation. Xenotransplantation 2019; 26:e12508. [PMID: 30963627 DOI: 10.1111/xen.12508] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/04/2019] [Accepted: 03/07/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Pre-transplant modification of porcine islets may improve their suitability for clinical use in diabetes management by supporting graft function and reducing the potential for xeno-rejection. The present study investigates intra-graft incorporation of stem cells that secrete beta (β)-cell trophic and immunomodulatory factors to preserve function and alter immune cell responsiveness to porcine islets. METHODS Isolated porcine islets were maintained in a three-dimensional rotational cell culture system (RCCS) to facilitate aggregation with human amniotic epithelial cells (AECs). Assembled islet constructs were assessed for functional integrity and ability to avoid xeno-recognition by CD4+ T-cells using mixed islet:lymphocyte reaction assays. To determine whether stem cell-mediated modification of porcine islets provided a survival advantage over native islets, structural integrity was examined in a pig-to-mouse islet transplant model. RESULTS Rotational cell culture system supported the formation of porcine islet:AEC aggregates with improved insulin-secretory capacity compared to unmodified islets, whilst the xeno-response of purified CD4+ T-cells to AEC-bearing grafts was significantly (P < 0.05) attenuated. Transplanted AEC-bearing grafts demonstrated slower rejection in immune-competent recipients compared to unmodified islets. CONCLUSIONS/INTERPRETATION Rotational culture enables pre-transplant modification of porcine islets by integration with immunomodulatory stem cells capable of subduing xeno-reactivity to CD4+ T-cells. This reduces islet rejection and offers translational potential to widen availability and improve the clinical effectiveness of islet transplantation.
Collapse
Affiliation(s)
- Ali Zafar
- The Islet Research Laboratory, Worcestershire Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| | - Jou Lee
- The Islet Research Laboratory, Worcestershire Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| | - Shameema Yesmin
- The Islet Research Laboratory, Worcestershire Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| | - Michelle B Paget
- The Islet Research Laboratory, Worcestershire Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| | - Clifford J Bailey
- Diabetes Research, School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Hilary E Murray
- The Islet Research Laboratory, Worcestershire Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| | - Richard Downing
- The Islet Research Laboratory, Worcestershire Clinical Research Unit, Worcestershire Acute Hospitals NHS Trust, Worcester, UK
| |
Collapse
|
21
|
Espona-Noguera A, Etxebarria-Elezgarai J, Saenz Del Burgo L, Cañibano-Hernández A, Gurruchaga H, Blanco FJ, Orive G, Hernández RM, Benito-Lopez F, Ciriza J, Basabe-Desmonts L, Pedraz JL. Type 1 Diabetes Mellitus reversal via implantation of magnetically purified microencapsulated pseudoislets. Int J Pharm 2019; 560:65-77. [PMID: 30742984 DOI: 10.1016/j.ijpharm.2019.01.058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 01/13/2023]
Abstract
Microencapsulation of pancreatic islets for the treatment of Type I Diabetes Mellitus (T1DM) generates a high quantity of empty microcapsules, resulting in high therapeutic graft volumes that can enhance the host's immune response. We report a 3D printed microfluidic magnetic sorting device for microcapsules purification with the objective to reduce the number of empty microcapsules prior transplantation. In this study, INS1E pseudoislets were microencapsulated within alginate (A) and alginate-poly-L-lysine-alginate (APA) microcapsules and purified through the microfluidic device. APA microcapsules demonstrated higher mechanical integrity and stability than A microcapsules, showing better pseudoislets viability and biological function. Importantly, we obtained a reduction of the graft volume of 77.5% for A microcapsules and 78.6% for APA microcapsules. After subcutaneous implantation of induced diabetic Wistar rats with magnetically purified APA microencapsulated pseudoislets, blood glucose levels were restored into normoglycemia (<200 mg/dL) for almost 17 weeks. In conclusion, our described microfluidic magnetic sorting device represents a great alternative approach for the graft volume reduction of microencapsulated pseudoislets and its application in T1DM disease.
Collapse
Affiliation(s)
- A Espona-Noguera
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - J Etxebarria-Elezgarai
- BIOMICs-microfluidics Research Group, Microfluidics Cluster UPV/EHU, University of the Basque Country, Spain
| | - L Saenz Del Burgo
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - A Cañibano-Hernández
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - H Gurruchaga
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - F J Blanco
- INIBIC-Hospital Universitario La Coruña, La Coruña, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), La Coruña, Spain
| | - G Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), BTI Biotechnology Institute, Vitoria-Gasteiz, Spain
| | - Rosa M Hernández
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - F Benito-Lopez
- AMMa LOAC Research Group, Microfluidics Cluster UPV/EHU, University of the Basque Country, Spain
| | - J Ciriza
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - L Basabe-Desmonts
- BIOMICs-microfluidics Research Group, Microfluidics Cluster UPV/EHU, University of the Basque Country, Spain; Basque Foundation of Science, IKERBASQUE, Spain.
| | - J L Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain.
| |
Collapse
|
22
|
Stephens CH, Orr KS, Acton AJ, Tersey SA, Mirmira RG, Considine RV, Voytik-Harbin SL. In situ type I oligomeric collagen macroencapsulation promotes islet longevity and function in vitro and in vivo. Am J Physiol Endocrinol Metab 2018; 315:E650-E661. [PMID: 29894201 PMCID: PMC6230705 DOI: 10.1152/ajpendo.00073.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Widespread use of pancreatic islet transplantation for treatment of type 1 diabetes (T1D) is currently limited by requirements for long-term immunosuppression, limited donor supply, and poor long-term engraftment and function. Upon isolation from their native microenvironment, islets undergo rapid apoptosis, which is further exacerbated by poor oxygen and nutrient supply following infusion into the portal vein. Identifying alternative strategies to restore critical microenvironmental cues, while maximizing islet health and function, is needed to advance this cellular therapy. We hypothesized that biophysical properties provided through type I oligomeric collagen macroencapsulation are important considerations when designing strategies to improve islet survival, phenotype, and function. Mouse islets were encapsulated at various Oligomer concentrations (0.5 -3.0 mg/ml) or suspended in media and cultured for 14 days, after which viability, protein expression, and function were assessed. Oligomer-encapsulated islets showed a density-dependent improvement in in vitro viability, cytoarchitecture, and insulin secretion, with 3 mg/ml yielding values comparable to freshly isolated islets. For transplantation into streptozotocin-induced diabetic mice, 500 islets were mixed in Oligomer and injected subcutaneously, where rapid in situ macroencapsulation occurred, or injected with saline. Mice treated with Oligomer-encapsulated islets exhibited rapid (within 24 h) diabetes reversal and maintenance of normoglycemia for 14 (immunocompromised), 90 (syngeneic), and 40 days (allogeneic). Histological analysis showed Oligomer-islet engraftment with maintenance of islet cytoarchitecture, revascularization, and no foreign body response. Oligomer-islet macroencapsulation may provide a useful strategy for prolonging the health and function of cultured islets and has potential as a subcutaneous injectable islet transplantation strategy for treatment of T1D.
Collapse
Affiliation(s)
| | - Kara S Orr
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Pediatrics, Indiana University School of Medicine , Indianapolis, Indiana
| | - Anthony J Acton
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Sarah A Tersey
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Pediatrics, Indiana University School of Medicine , Indianapolis, Indiana
| | - Raghavendra G Mirmira
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Pediatrics, Indiana University School of Medicine , Indianapolis, Indiana
| | - Robert V Considine
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine , Indianapolis, Indiana
- Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University , West Lafayette, Indiana
- Department of Basic Medical Sciences, Purdue University , West Lafayette, Indiana
| |
Collapse
|
23
|
Rios PD, Skoumal M, Liu J, Youngblood R, Kniazeva E, Garcia AJ, Shea LD. Evaluation of encapsulating and microporous nondegradable hydrogel scaffold designs on islet engraftment in rodent models of diabetes. Biotechnol Bioeng 2018; 115:2356-2364. [PMID: 29873059 PMCID: PMC6131066 DOI: 10.1002/bit.26741] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/11/2018] [Accepted: 05/22/2018] [Indexed: 02/06/2023]
Abstract
Islet transplantation is a promising therapeutic option for type 1 diabetes mellitus, yet the current delivery into the hepatic portal vasculature is limited by poor engraftment. Biomaterials have been used as a means to promote engraftment and function at extrahepatic sites, with strategies being categorized as encapsulation or microporous scaffolds that can either isolate or integrate islets with the host tissue, respectively. Although these approaches are typically studied separately using distinct material platforms, herein, we developed nondegradable polyethylene glycol (PEG)-based hydrogels for islet encapsulation or as microporous scaffolds for islet seeding to compare the initial engraftment and function of islets in syngeneic diabetic mice. Normoglycemia was restored with transplantation of islets within either encapsulating or microporous hydrogels containing 700 islet equivalents (IEQ), with transplantation on microporous hydrogels producing lower blood glucose levels at earlier times. A glucose challenge test at 1 month after transplant indicated that encapsulated islets had a delay in glucose-stimulated insulin secretion, whereas microporous hydrogels restored normoglycemia in times consistent with native pancreata. Encapsulated islets remained isolated from the host tissue, whereas the microporous scaffolds allowed for revascularization of the islets after transplant. Finally, we compared the inflammatory response after transplantation for the two systems and noted that microporous hydrogels had a substantially increased presence of neutrophils. Collectively, these findings suggest that both encapsulation and microporous PEG scaffold designs allow for stable engraftment of syngeneic islets and the ability to restore normoglycemia, yet the architecture influences islet function and responsiveness after transplantation.
Collapse
Affiliation(s)
- Peter D Rios
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois
| | - Michael Skoumal
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Jeffrey Liu
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois
| | - Richard Youngblood
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Ekaterina Kniazeva
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, Illinois
| | - Andrés J Garcia
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
24
|
Kopan C, Tucker T, Alexander M, Mohammadi MR, Pone EJ, Lakey JRT. Approaches in Immunotherapy, Regenerative Medicine, and Bioengineering for Type 1 Diabetes. Front Immunol 2018; 9:1354. [PMID: 29963051 PMCID: PMC6011033 DOI: 10.3389/fimmu.2018.01354] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
Recent advances on using immune and stem cells as two-pronged approaches for type 1 diabetes mellitus (T1DM) treatment show promise for advancement into clinical practice. As T1DM is thought to arise from autoimmune attack destroying pancreatic β-cells, increasing treatments that use biologics and cells to manipulate the immune system are achieving better results in pre-clinical and clinical studies. Increasingly, focus has shifted from small molecule drugs that suppress the immune system nonspecifically to more complex biologics that show enhanced efficacy due to their selectivity for specific types of immune cells. Approaches that seek to inhibit only autoreactive effector T cells or enhance the suppressive regulatory T cell subset are showing remarkable promise. These modern immune interventions are also enabling the transplantation of pancreatic islets or β-like cells derived from stem cells. While complete immune tolerance and body acceptance of grafted islets and cells is still challenging, bioengineering approaches that shield the implanted cells are also advancing. Integrating immunotherapy, stem cell-mediated β-cell or islet production and bioengineering to interface with the patient is expected to lead to a durable cure or pave the way for a clinical solution for T1DM.
Collapse
Affiliation(s)
- Christopher Kopan
- Department of Surgery, University of California Irvine, Irvine, CA, United States
| | - Tori Tucker
- Department of Cell and Molecular Biosciences, University of California Irvine, Irvine, CA, United States
| | - Michael Alexander
- Department of Surgery, University of California Irvine, Irvine, CA, United States
| | - M. Rezaa Mohammadi
- Department of Chemical Engineering and Materials Science, University of California Irvine, Irvine, CA, United States
| | - Egest J. Pone
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, United States
| | - Jonathan Robert Todd Lakey
- Department of Surgery, University of California Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, United States
| |
Collapse
|
25
|
Yang KC, Yanai G, Yang SY, Canning P, Satou Y, Kawagoe M, Sumi S. Low-adhesive ethylene vinyl alcohol-based packaging to xenogeneic islet encapsulation for type 1 diabetes treatment. Biotechnol Bioeng 2018; 115:2341-2355. [PMID: 29777589 DOI: 10.1002/bit.26730] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/17/2018] [Accepted: 05/16/2018] [Indexed: 01/16/2023]
Abstract
Transplantation of encapsulated porcine islets is proposed to treat type 1 diabetes. However, the envelopment of fibrous tissue and the infiltration of immune cells impair islet function and eventually cause implant failure. It is known that hemodialysis using an ethylene vinyl alcohol (EVOH) membrane results in minor tissue responses. Therefore, we hypothesized that using a low-adhesive EVOH membrane for encapsulation may prevent host cell accumulation and fibrous capsule formation. In this study, rat islets suspended in chitosan gel were encapsulated in bags made from highly porous EVOH membranes, and their in vitro insulin secretion function as well as in vivo performance was evaluated. The results showed that the EVOH bag did not affect islet survival or glucose-stimulated insulin secretion. Whereas naked islets were dysfunctional after 7 days of culture in vitro, islets within the EVOH bag produced insulin continuously for 30 days. Streptozotocin-induced diabetic mice were given islets-chitosan gel-EVOH implants intraperitoneally (650-800 islets equivalent) and exhibited lower blood glucose levels and regained body weight during a 4-week observation period. The transplanted mice had higher levels of serum insulin and C-peptide, with an improved blood glucose disappearance rate. Retrieved implants had minor tissue adhesion, and histology showed a limited number of mononuclear cells and fibroblasts surrounding the implants. No invasion of host cells into the EVOH bags was noticed, and the encapsulated islets were intact and positive for insulin-glucagon immunostaining. In conclusion, an EVOH bag can protect encapsulated islets, limit fibrous capsule formation, and extend graft function.
Collapse
Affiliation(s)
- Kai-Chiang Yang
- Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Goichi Yanai
- Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Sin-Yu Yang
- Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Priyadarshini Canning
- Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yoshio Satou
- Molding Component Business Department, Kuraray Co., Ltd, Tokyo, Japan
| | - Masako Kawagoe
- Molding Component Business Department, Kuraray Co., Ltd, Tokyo, Japan
| | - Shoichiro Sumi
- Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
26
|
Transcutaneously refillable, 3D-printed biopolymeric encapsulation system for the transplantation of endocrine cells. Biomaterials 2018; 177:125-138. [PMID: 29886385 DOI: 10.1016/j.biomaterials.2018.05.047] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/17/2018] [Accepted: 05/27/2018] [Indexed: 12/17/2022]
Abstract
Autologous cell transplantation holds enormous promise to restore organ and tissue functions in the treatment of various pathologies including endocrine, cardiovascular, and neurological diseases among others. Even though immune rejection is circumvented with autologous transplantation, clinical adoption remains limited due to poor cell retention and survival. Cell transplant success requires homing to vascularized environment, cell engraftment and importantly, maintenance of inherent cell function. To address this need, we developed a three dimensional (3D) printed cell encapsulation device created with polylactic acid (PLA), termed neovascularized implantable cell homing and encapsulation (NICHE). In this paper, we present the development and systematic evaluation of the NICHE in vitro, and the in vivo validation with encapsulated testosterone-secreting Leydig cells in Rag1-/- castrated mice. Enhanced subcutaneous vascularization of NICHE via platelet-rich plasma (PRP) hydrogel coating and filling was demonstrated in vivo via a chorioallantoic membrane (CAM) assay as well as in mice. After establishment of a pre-vascularized bed within the NICHE, transcutaneously transplanted Leydig cells, maintained viability and robust testosterone secretion for the duration of the study. Immunohistochemical analysis revealed extensive Leydig cell colonization in the NICHE. Furthermore, transplanted cells achieved physiologic testosterone levels in castrated mice. The promising results provide a proof of concept for the NICHE as a viable platform technology for autologous cell transplantation for the treatment of a variety of diseases.
Collapse
|
27
|
Lee SH, Park HS, Yang Y, Lee EY, Kim JW, Khang G, Yoon KH. Improvement of islet function and survival by integration of perfluorodecalin into microcapsules in vivo and in vitro. J Tissue Eng Regen Med 2018; 12:e2110-e2122. [PMID: 29330944 DOI: 10.1002/term.2643] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/04/2017] [Accepted: 01/02/2018] [Indexed: 12/24/2022]
Abstract
Hypoxic injury of islets is a major obstacle for encapsulated islet transplantation into the peritoneal cavity. To improve oxygen delivery to encapsulated islets, we integrated 20% of the oxygen carrier material, perfluorodecalin (PFD), in alginate capsules mixed with islets (PFD-alginate). Integration of PFD clearly improved islet viability and decreased reactive oxygen species production compared to islets encapsulated with alginate only (alginate) and naked islets exposed to hypoxia in vitro. In PFD-alginate capsules, HIF-1α expression was minimal, and insulin expression was well maintained. Furthermore, the best islet function represented by glucose-stimulated insulin secretion was observed for the PFD-alginate capsules in hypoxic condition. For the in vivo study, the same number of naked islets and encapsulated islets (alginate and PFD-alginate) was transplanted into streptozotocin-induced diabetic mice. Nonfasting blood glucose levels and the area under the curve for glucose based on intraperitoneal glucose tolerance tests in the PFD-alginate group were lower than in the alginate group. The harvested islets stained positive for insulin in all groups, but the ratio of dead cell area was 4 times higher in the alginate group than in the PFD-alginate group. In conclusion, integration of PFD in alginate microcapsules improved islet function and survival by minimizing the hypoxic damage of islets after intraperitoneal transplantation.
Collapse
Affiliation(s)
- Sang-Ho Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Heon-Seok Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yeoree Yang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji-Won Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Gilson Khang
- Department of Polymer Nano Science and Technology, Department of BIN Fusion Technology and BK-21 Polymer BIN Fusion Research Team, Chonbuk National University, Jeonju, South Korea
| | - Kun-Ho Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
28
|
Solez K, Fung KC, Saliba KA, Sheldon VLC, Petrosyan A, Perin L, Burdick JF, Fissell WH, Demetris AJ, Cornell LD. The bridge between transplantation and regenerative medicine: Beginning a new Banff classification of tissue engineering pathology. Am J Transplant 2018; 18:321-327. [PMID: 29194964 PMCID: PMC5817246 DOI: 10.1111/ajt.14610] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 11/21/2017] [Accepted: 11/24/2017] [Indexed: 01/25/2023]
Abstract
The science of regenerative medicine is arguably older than transplantation-the first major textbook was published in 1901-and a major regenerative medicine meeting took place in 1988, three years before the first Banff transplant pathology meeting. However, the subject of regenerative medicine/tissue engineering pathology has never received focused attention. Defining and classifying tissue engineering pathology is long overdue. In the next decades, the field of transplantation will enlarge at least tenfold, through a hybrid of tissue engineering combined with existing approaches to lessening the organ shortage. Gradually, transplantation pathologists will become tissue-(re-) engineering pathologists with enhanced skill sets to address concerns involving the use of bioengineered organs. We outline ways of categorizing abnormalities in tissue-engineered organs through traditional light microscopy or other modalities including biomarkers. We propose creating a new Banff classification of tissue engineering pathology to standardize and assess de novo bioengineered solid organs transplantable success in vivo. We recommend constructing a framework for a classification of tissue engineering pathology now with interdisciplinary consensus discussions to further develop and finalize the classification at future Banff Transplant Pathology meetings, in collaboration with the human cell atlas project. A possible nosology of pathologic abnormalities in tissue-engineered organs is suggested.
Collapse
Affiliation(s)
- K. Solez
- Department of Laboratory Medicine and PathologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - K. C. Fung
- Department of Laboratory Medicine and PathologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - K. A. Saliba
- Department of Laboratory Medicine and PathologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - V. L. C. Sheldon
- Medical Anthropology ProgramDepartment of AnthropologyFaculty of Arts and SciencesUniversity of TorontoTorontoOntarioCanada
| | - A. Petrosyan
- Division of Urology GOFARR Laboratory for Organ Regenerative Research and Cell TherapeuticsChildren's Hospital Los AngelesSaban Research InstituteUniversity of Southern CaliforniaLos AngelesCAUSA
| | - L. Perin
- Division of Urology GOFARR Laboratory for Organ Regenerative Research and Cell TherapeuticsChildren's Hospital Los AngelesSaban Research InstituteUniversity of Southern CaliforniaLos AngelesCAUSA
| | - J. F. Burdick
- Department of SurgeryJohns Hopkins School of MedicineBaltimoreMDUSA
| | - W. H. Fissell
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - A. J. Demetris
- Department of PathologyUniversity of PittsburghUPMC‐MontefiorePittsburghPAUSA
| | - L. D. Cornell
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| |
Collapse
|
29
|
Zhu H, Li W, Liu Z, Li W, Chen N, Lu L, Zhang W, Wang Z, Wang B, Pan K, Zhang X, Chen G. Selection of Implantation Sites for Transplantation of Encapsulated Pancreatic Islets. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:191-214. [PMID: 29048258 DOI: 10.1089/ten.teb.2017.0311] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic islet transplantation has been validated as a valuable therapy for type 1 diabetes mellitus patients with exhausted insulin treatment. However, this therapy remains limited by the shortage of donor and the requirement of lifelong immunosuppression. Islet encapsulation, as an available bioartificial pancreas (BAP), represents a promising approach to enable protecting islet grafts without or with minimal immunosuppression and possibly expanding the donor pool. To develop a clinically implantable BAP, some key aspects need to be taken into account: encapsulation material, capsule design, and implant site. Among them, the implant site exerts an important influence on the engraftment, stability, and biocompatibility of implanted BAP. Currently, an optimal site for encapsulated islet transplantation may include sufficient capacity to host large graft volumes, portal drainage, ease of access using safe and reproducible procedure, adequate blood/oxygen supply, minimal immune/inflammatory reaction, pliable for noninvasive imaging and biopsy, and potential of local microenvironment manipulation or bioengineering. Varying degrees of success have been confirmed with the utilization of liver or extrahepatic sites in an experimental or preclinical setting. However, the ideal implant site remains to be further engineered or selected for the widespread application of encapsulated islet transplantation.
Collapse
Affiliation(s)
- Haitao Zhu
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China .,2 Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University , Xi'an, China
| | - Wenjing Li
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Zhongwei Liu
- 3 Department of Cardiology, Shaanxi Provincial People's Hospital , Xi'an, China
| | - Wenliang Li
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Niuniu Chen
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Linlin Lu
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Wei Zhang
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Zhen Wang
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Bo Wang
- 2 Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University , Xi'an, China .,4 Institute of Advanced Surgical Technology and Engineering, Xi'an Jiaotong University , Xi'an, China
| | - Kaili Pan
- 5 Department of Pediatrics (No. 2 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Xiaoge Zhang
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Guoqiang Chen
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| |
Collapse
|
30
|
Abstract
BACKGROUND New methods of beta-cell replacement have been developed to maintain excellent glycemic control, improve quality of life, and even eliminate insulin injections in patients with type 1 diabetes mellitus (T1DM). Previously, we demonstrated that being insulin-free is the strongest motivation for accepting a newly developed therapy. Multiple allogeneic islet transplantations with immunosuppression using a human donor is the best option to be insulin-free, but the necessity for immunosuppression and donor shortage are major issues. However, these issues have been improved with scientific progress. The aim of this study was to investigate the opinions of patients and their families about the current progress. METHODS We conducted a questionnaire survey of T1DM patients (n = 47) and their family members (n = 49) about newly developed therapies: single and multiple allogeneic islet transplantation, single and multiple encapsulated allogeneic islet transplantation, single and multiple xenogeneic islet transplantation, and induced pluripotent stem cell therapy. RESULTS More than 90% of respondents wished to be insulin-free and have stable glycemic control. More than 90% of respondents accepted at least one of the new therapies. The current standard treatment multiple allogeneic islet transplantation was not well accepted or favored. CONCLUSIONS The next generation of treatments, including xenotransplantation and induced pluripotent stem cell therapy, were more acceptable and favorable. Even though the majority of patients wish to become insulin-free, it is not sufficiently strong motivation for accepting newly developed treatments.
Collapse
Affiliation(s)
- Akitsu Kawabe
- Islet Cell Transplantation Project, Diabetes Research Center, Research Institute of National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinichi Matsumoto
- Islet Cell Transplantation Project, Diabetes Research Center, Research Institute of National Center for Global Health and Medicine, Tokyo, Japan
- Research and Development Center, Otsuka Pharmaceutical Factory Inc., Naruto, Japan
| | - Masayuki Shimoda
- Islet Cell Transplantation Project, Diabetes Research Center, Research Institute of National Center for Global Health and Medicine, Tokyo, Japan
- CONTACT Masayuki Shimoda Islet Cell Transplantation Project, Diabetes Research Center, Research Institute of National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| |
Collapse
|
31
|
Lee SJ, Lee JB, Park YW, Lee DY. 3D Bioprinting for Artificial Pancreas Organ. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:355-374. [PMID: 30471043 DOI: 10.1007/978-981-13-0445-3_21] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Type 1 diabetes mellitus (T1DM) results from an autoimmune destruction of insulin-producing beta cells in the islet of the endocrine pancreas. Although islet transplantation has been regarded as an ideal strategy for T1D, transplanted islets are rejected from host immune system. To immunologically protect them, islet encapsulation technology with biocompatible materials is emerged as an immuno-barrier. However, this technology has been limited for clinical trial such as hypoxia in the central core of islet bead, impurity of islet bead and retrievability from the body. Recently, 3D bioprinting has been emerged as an alternative approach to make the artificial pancreas. It can be used to position live cells in a desired location with real scale of human organ. Furthermore, constructing a vascularization of the artificial pancreas is actualized with 3D bioprinting. Therefore, it is possible to create real pancreas-mimic artificial organ for clinical application. In conclusion, 3D bioprinting can become a new leader in the development of the artificial pancreas to overcome the existed islet.
Collapse
Affiliation(s)
- Seon Jae Lee
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea
| | - Jae Bin Lee
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea
| | - Young-Woo Park
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea. .,Institute of Nano Science & Technology (INST), Hanyang University, Seoul, South Korea.
| |
Collapse
|
32
|
Fung RKF, Kerridge IH. Gene editing advance re-ignites debate on the merits and risks of animal to human transplantation. Intern Med J 2017; 46:1017-22. [PMID: 27633468 DOI: 10.1111/imj.13183] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 12/21/2022]
Abstract
In Australia, and internationally, the shortage of organ and tissue donors significantly limits the number of patients with critical organ or tissue failure who are able to receive a transplant each year. The rationale for xenotransplantation - the transplantation of living cells, tissues or organs from one species to another - is to meet this shortfall in human donor material. While early clinical trials showed promise, particularly in patients with type I diabetes whose insulin dependence could be temporarily reversed by the transplantation of porcine islet cells, these benefits have been balanced with scientific, clinical and ethical concerns revolving around the risks of immune rejection and the potential transmission of porcine endogenous retroviruses or other infectious agents from porcine grafts to human recipients. However, the advent of CRISPR/Cas9, a revolutionary gene editing technology, has reignited interest in the field with the possibility of genetically engineering porcine organs and tissues that are less immunogenic and have virtually no risk of transmission of porcine endogenous retroviruses. At the same time, CRISPR/Cas9 may also open up a myriad of possibilities for tissue engineering and stem cell research, which may complement xenotransplantation research by providing an additional source of donor cells, tissues and organs for transplantation into patients. The recent international symposium on gene editing, organised by the US National Academy of Sciences, highlights both the enormous therapeutic potential of CRISPR/Cas9 and the raft of ethical and regulatory challenges that may follow its utilisation in transplantation and in medicine more generally.
Collapse
Affiliation(s)
- R K F Fung
- Centre for Values, Ethics and the Law in Medicine, School of Public Health, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia. .,Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.
| | - I H Kerridge
- Centre for Values, Ethics and the Law in Medicine, School of Public Health, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
33
|
Liu Z, Hu W, He T, Dai Y, Hara H, Bottino R, Cooper DKC, Cai Z, Mou L. Pig-to-Primate Islet Xenotransplantation: Past, Present, and Future. Cell Transplant 2017; 26:925-947. [PMID: 28155815 PMCID: PMC5657750 DOI: 10.3727/096368917x694859] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 03/21/2017] [Indexed: 12/17/2022] Open
Abstract
Islet allotransplantation results in increasing success in treating type 1 diabetes, but the shortage of deceased human donor pancreata limits progress. Islet xenotransplantation, using pigs as a source of islets, is a promising approach to overcome this limitation. The greatest obstacle is the primate immune/inflammatory response to the porcine (pig) islets, which may take the form of rapid early graft rejection (the instant blood-mediated inflammatory reaction) or T-cell-mediated rejection. These problems are being resolved by the genetic engineering of the source pigs combined with improved immunosuppressive therapy. The results of pig-to-diabetic nonhuman primate islet xenotransplantation are steadily improving, with insulin independence being achieved for periods >1 year. An alternative approach is to isolate islets within a micro- or macroencapsulation device aimed at protecting them from the human recipient's immune response. Clinical trials using this approach are currently underway. This review focuses on the major aspects of pig-to-primate islet xenotransplantation and its potential for treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Zhengzhao Liu
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Wenbao Hu
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Tian He
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Yifan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Hidetaka Hara
- Xenotransplantation Program/Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rita Bottino
- Institute for Cellular Therapeutics, Allegheny-Singer Research Institute, Pittsburgh, PA, USA
| | - David K. C. Cooper
- Xenotransplantation Program/Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhiming Cai
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| | - Lisha Mou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, P.R. China
| |
Collapse
|
34
|
Chan KH, Krishnan R, Alexander M, Lakey JRT. Developing a Rapid Algorithm to Enable Rapid Characterization of Alginate Microcapsules. Cell Transplant 2017; 26:765-772. [PMID: 27729095 DOI: 10.3727/096368916x693446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The islets of Langerhans are endocrine tissue clusters that secrete hormones that regulate the body's glucose, carbohydrate, and fat metabolism, the most important of which is insulin, a hormone secreted by β-cells within the islets. In certain instances, a person's own immune system attacks and destroys them, leading to the development of type 1 diabetes (T1D), a life-long condition that needs daily insulin administration to maintain health and prolong survival. Islet transplantation is a surgical procedure that has demonstrated the ability to normalize blood sugar levels for up to a few years, but the need for chronic immunosuppression relegates it to a last resort that is often only used sparingly and in seriously ill patients. Islet microencapsulation is a biomedical innovation designed to protect islets from the immune system by coating them with a biocompatible polymer, and this new technology has demonstrated various degrees of success in small- and large-animal studies. This success is significantly impacted by microcapsule morphology and encapsulation efficiency. Since hundreds of thousands of microcapsules are generated during the process, characterization of encapsulated islets without the help of some degree of automation would be difficult, time-consuming, and error prone due to inherent observer bias. We have developed an image analysis algorithm that can analyze hundreds of microencapsulated islets and characterize their size, shape, circularity, and distortion with minimal observer bias. This algorithm can be easily adapted to similar nano- or microencapsulation technologies to implement stricter quality control and improve biomaterial device design and success.
Collapse
|
35
|
Harrington S, Williams J, Rawal S, Ramachandran K, Stehno-Bittel L. Hyaluronic Acid/Collagen Hydrogel as an Alternative to Alginate for Long-Term Immunoprotected Islet Transplantation<sup/>. Tissue Eng Part A 2017; 23:1088-1099. [PMID: 28142500 DOI: 10.1089/ten.tea.2016.0477] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Alginate has long been the material of choice for immunoprotection of islets due to its low cost and ability to easily form microspheres. Unfortunately, this seaweed-derived material is notoriously prone to fibrotic overgrowth in vivo, resulting in premature graft failure. The purpose of this study was to test an alternative, hyaluronic acid (HA-COL), for in vitro function, viability, and allogeneic islet transplant outcomes in diabetic rats. In vitro studies indicated that the HA-COL gel had diffusion characteristics that would allow small molecules such as glucose and insulin to enter and exit the gel, whereas larger molecules (70 and 500 kDa dextrans) were impeded from diffusing past the gel edge in 24 h. Islets encapsulated in HA-COL hydrogel showed significantly improved in vitro viability over unencapsulated islets and retained their morphology and glucose sensitivity for 28 days. When unencapsulated allogeneic islet transplants were administered to the omentum of outbred rats, they initially were normoglycemic, but by 11 days returned to hyperglycemia. Immunohistological examination of the grafts and surrounding tissue indicated strong graft rejection. By comparison, when using the same outbred strain of rats, allogeneic transplantation of islets within the HA-COL gel reversed long-term diabetes and prevented graft rejection in all animals. Animals were sacrificed at 40, 52, 64, and 80 weeks for evaluation, and all were non-diabetic at sacrifice. Explanted grafts revealed viable islets in the transplant site as well as intact hydrogel, with little or no evidence of fibrotic overgrowth or cellular rejection. The results of these studies demonstrate great potential for HA-COL hydrogel as an alternative to sodium alginate for long-term immunoprotected islet transplantation.
Collapse
Affiliation(s)
- Stephen Harrington
- 1 School of Engineering, University of Kansas , Lawrence, Kansas.,2 University of Kansas Medical Center , Kansas City, Kansas.,3 Likarda, LLC, Kansas City, Kansas
| | - Janette Williams
- 2 University of Kansas Medical Center , Kansas City, Kansas.,3 Likarda, LLC, Kansas City, Kansas
| | - Sonia Rawal
- 2 University of Kansas Medical Center , Kansas City, Kansas
| | | | - Lisa Stehno-Bittel
- 1 School of Engineering, University of Kansas , Lawrence, Kansas.,2 University of Kansas Medical Center , Kansas City, Kansas.,3 Likarda, LLC, Kansas City, Kansas
| |
Collapse
|
36
|
Smole A, Lainšček D, Bezeljak U, Horvat S, Jerala R. A Synthetic Mammalian Therapeutic Gene Circuit for Sensing and Suppressing Inflammation. Mol Ther 2017; 25:102-119. [PMID: 28129106 DOI: 10.1016/j.ymthe.2016.10.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 10/20/2016] [Accepted: 10/21/2016] [Indexed: 12/20/2022] Open
Abstract
Inflammation, which is a highly regulated host response against danger signals, may be harmful if it is excessive and deregulated. Ideally, anti-inflammatory therapy should autonomously commence as soon as possible after the onset of inflammation, should be controllable by a physician, and should not systemically block beneficial immune response in the long term. We describe a genetically encoded anti-inflammatory mammalian cell device based on a modular engineered genetic circuit comprising a sensor, an amplifier, a "thresholder" to restrict activation of a positive-feedback loop, a combination of advanced clinically used biopharmaceutical proteins, and orthogonal regulatory elements that linked modules into the functional device. This genetic circuit was autonomously activated by inflammatory signals, including endogenous cecal ligation and puncture (CLP)-induced inflammation in mice and serum from a systemic juvenile idiopathic arthritis (sIJA) patient, and could be reset externally by a chemical signal. The microencapsulated anti-inflammatory device significantly reduced the pathology in dextran sodium sulfate (DSS)-induced acute murine colitis, demonstrating a synthetic immunological approach for autonomous anti-inflammatory therapy.
Collapse
Affiliation(s)
- Anže Smole
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Urban Bezeljak
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Simon Horvat
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; EN-FIST Centre of Excellence, 1000 Ljubljana, Slovenia; Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; EN-FIST Centre of Excellence, 1000 Ljubljana, Slovenia.
| |
Collapse
|
37
|
Rios PD, Zhang X, Luo X, Shea LD. Mold-casted non-degradable, islet macro-encapsulating hydrogel devices for restoration of normoglycemia in diabetic mice. Biotechnol Bioeng 2016; 113:2485-95. [PMID: 27159557 PMCID: PMC11287382 DOI: 10.1002/bit.26005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/01/2016] [Accepted: 05/02/2016] [Indexed: 12/14/2022]
Abstract
Islet transplantation is a potential cure for diabetic patients, however this procedure is not widely adopted due to the high rate of graft failure. Islet encapsulation within hydrogels is employed to provide a three-dimensional microenvironment conducive to survival of transplanted islets to extend graft function. Herein, we present a novel macroencapsulation device, composed of PEG hydrogel, that combines encapsulation with lithography techniques to generate polydimethylsiloxane (PDMS) molds. PEG solutions are mixed with islets, which are then cast into PDMS molds for subsequent crosslinking. The molds can also be employed to provide complex architectures, such as microchannels that may allow vascular ingrowth through pre-defined regions of the hydrogel. PDMS molds allowed for the formation of stable gels with encapsulation of islets, and in complex architectures. Hydrogel devices with a thickness of 600 μm containing 500 islets promoted normoglycemia within 12 days following transplantation into the epididymal fat pad, which was sustained over the two-month period of study until removal of the device. The inclusion of microchannels, which had a similar minimum distance between islets and the hydrogel surface, similarly promoted normoglycemia. A glucose challenge test indicated hydrogel devices achieved normoglycemia 90 min post-dextrose injections, similar to control mice with native pancreata. Histochemical staining revealed that transplanted islets, identified as insulin positive, were viable and isolated from host tissue at 8 weeks post-transplantation, yet devices with microchannels had tissue and vascular ingrowth within the channels. Taken together, these results demonstrate a system for creating non-degradable hydrogels with complex geometries for encapsulating islets capable of restoring normoglycemia, which may expand islet transplantation as a treatment option for diabetic patients. Biotechnol. Bioeng. 2016;113: 2485-2495. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Peter Daniel Rios
- Simpson Querrey Institute for BioNanotechnology in Medicine, Northwestern University, 303 East Superior Street, Chicago, Illinois, 60611
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois
| | - Xiaomin Zhang
- Division of Transplantation, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xunrong Luo
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Lonnie D Shea
- Simpson Querrey Institute for BioNanotechnology in Medicine, Northwestern University, 303 East Superior Street, Chicago, Illinois, 60611.
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Rd, Evanston, Illinois, 60208.
- Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, Illinois, 60208.
- The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 303 East Superior Street, Chicago, Illinois, 60611.
- Department of Obstetrics and Gynecology, Northwestern University, 205 East Superior Street, Chicago, Illinois, 60611.
- Department of Biomedical Engineering, University of Michigan, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, Michigan, 48109-2099.
| |
Collapse
|
38
|
Appel AA, Ibarra V, Somo SI, Larson JC, Garson AB, Guan H, McQuilling JP, Zhong Z, Anastasio MA, Opara EC, Brey EM. Imaging of Hydrogel Microsphere Structure and Foreign Body Response Based on Endogenous X-Ray Phase Contrast. Tissue Eng Part C Methods 2016; 22:1038-1048. [PMID: 27796159 PMCID: PMC5116683 DOI: 10.1089/ten.tec.2016.0253] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/28/2016] [Indexed: 12/22/2022] Open
Abstract
Transplantation of functional islets encapsulated in stable biomaterials has the potential to cure Type I diabetes. However, the success of these materials requires the ability to quantitatively evaluate their stability. Imaging techniques that enable monitoring of biomaterial performance are critical to further development in the field. X-ray phase-contrast (XPC) imaging is an emerging class of X-ray techniques that have shown significant promise for imaging biomaterial and soft tissue structures. In this study, XPC imaging techniques are shown to enable three dimensional (3D) imaging and evaluation of islet volume, alginate hydrogel structure, and local soft tissue features ex vivo. Rat islets were encapsulated in sterile ultrapurified alginate systems produced using a high-throughput microfluidic system. The encapsulated islets were implanted in omentum pouches created in a rodent model of type 1 diabetes. Microbeads were imaged with XPC imaging before implantation and as whole tissue samples after explantation from the animals. XPC microcomputed tomography (μCT) was performed with systems using tube-based and synchrotron X-ray sources. Islets could be identified within alginate beads and the islet volume was quantified in the synchrotron-based μCT volumes. Omental adipose tissue could be distinguished from inflammatory regions resulting from implanted beads in harvested samples with both XPC imaging techniques. Individual beads and the local encapsulation response were observed and quantified using quantitative measurements, which showed good agreement with histology. The 3D structure of the microbeads could be characterized with XPC imaging and failed beads could also be identified. These results point to the substantial potential of XPC imaging as a tool for imaging biomaterials in small animal models and deliver a critical step toward in vivo imaging.
Collapse
Affiliation(s)
- Alyssa A. Appel
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
- Research Services, Edward Hines Jr. VA Hospital, Chicago, Illinois
| | - Veronica Ibarra
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
| | - Sami I. Somo
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
| | - Jeffery C. Larson
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
- Research Services, Edward Hines Jr. VA Hospital, Chicago, Illinois
| | - Alfred B. Garson
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Huifeng Guan
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | | | - Zhong Zhong
- National Synchrotron Light Source, Brookhaven National Laboratory, Upton, New York
| | - Mark A. Anastasio
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Emmanuel C. Opara
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, North Carolina
| | - Eric M. Brey
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
- Research Services, Edward Hines Jr. VA Hospital, Chicago, Illinois
| |
Collapse
|
39
|
Zhu HT, Lu L, Liu XY, Yu L, Lyu Y, Wang B. Treatment of diabetes with encapsulated pig islets: an update on current developments. J Zhejiang Univ Sci B 2016; 16:329-43. [PMID: 25990050 DOI: 10.1631/jzus.b1400310] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The potential use of allogeneic islet transplantation in curing type 1 diabetes mellitus has been adequately demonstrated, but its large-scale application is limited by the short supply of donor islets and the need for sustained and heavy immunosuppressive therapy. Encapsulation of pig islets was therefore suggested with a view to providing a possible alternative source of islet grafts and avoiding chronic immunosuppression and associated adverse or toxic effects. Nevertheless, several vital elements should be taken into account before this therapy becomes a clinical reality, including cell sources, encapsulation approaches, and implantation sites. This paper provides a comprehensive review of xenotransplantation of encapsulated pig islets for the treatment of type 1 diabetes mellitus, including current research findings and suggestions for future studies.
Collapse
Affiliation(s)
- Hai-tao Zhu
- Heart Center, Northwest Women's and Children's Hospital, Xi'an 710061, China; Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an 710061, China; Department of Hand Surgery, China-Japan Union Hospital, Norman Bethune Health Science Center, Jilin University, Changchun 130033, China; Institute of Advanced Surgical Technology and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | | | | | | | | | | |
Collapse
|
40
|
Baker K. Comparison of bioartificial and artificial pancreatic transplantation as promising therapies for Type I Diabetes Mellitus. ACTA ACUST UNITED AC 2016. [DOI: 10.1093/biohorizons/hzw002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
41
|
He Y, Xu Z, Fu H, Chen B, Wang S, Chen B, Zhou M, Cai Y. Combined Microencapsulated Islet Transplantation and Revascularization of Aortorenal Bypass in a Diabetic Nephropathy Rat Model. J Diabetes Res 2016; 2016:9706321. [PMID: 27119088 PMCID: PMC4826923 DOI: 10.1155/2016/9706321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/08/2016] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE Revascularization of aortorenal bypass is a preferred technique for renal artery stenosis (RAS) in diabetic nephropathy (DN) patients. Restenosis of graft vessels also should be considered in patients lacking good control of blood glucose. In this study, we explored a combined strategy to prevent the recurrence of RAS in the DN rat model. METHODS A model of DN was established by intraperitoneal injection of streptozotocin. Rats were divided into 4 groups: SR group, MIT group, Com group, and the untreated group. The levels of blood glucose and urine protein were measured, and changes in renal pathology were observed. The expression of monocyte chemoattractant protein-1 (MCP-1) in graft vessels was assessed by immunohistochemical staining. Histopathological staining was performed to assess the pathological changes of glomeruli and tubules. RESULTS The levels of urine protein and the expression of MCP-1 in graft vessels were decreased after islet transplantation. The injury of glomerular basement membrane and podocytes was significantly ameliorated. CONCLUSIONS The combined strategy of revascularization and microencapsulated islet transplantation had multiple protective effects on diabetic nephropathy, including preventing atherosclerosis in the graft vessels and alleviating injury to the glomerular filtration barrier. This combined strategy may be helpful for DN patients with RAS.
Collapse
Affiliation(s)
- Yunqiang He
- Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Ziqiang Xu
- Department of Transplantation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hongxing Fu
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Bin Chen
- Department of B-Mode Ultrasound, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Silu Wang
- Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Bicheng Chen
- Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Mengtao Zhou
- Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Department of Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- *Mengtao Zhou: and
| | - Yong Cai
- Department of Transplantation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- *Yong Cai:
| |
Collapse
|
42
|
Köllmer M, Appel AA, Somo SI, Brey EM. Long-Term Function of Alginate-Encapsulated Islets. TISSUE ENGINEERING PART B-REVIEWS 2015; 22:34-46. [PMID: 26414084 DOI: 10.1089/ten.teb.2015.0140] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Human trials have demonstrated the feasibility of alginate-encapsulated islet cells for the treatment of type 1 diabetes. Encapsulated islets can be protected from the host's immune system and remain viable and functional following transplantation. However, the long-term success of these therapies requires that alginate microcapsules maintain their immunoprotective capacity and stability in vivo for sustained periods. In part, as a consequence of different encapsulation strategies, islet encapsulation studies have produced inconsistent results in regard to graft functioning time, stability, and overall metabolic benefits. Alginate composition (proportion of M- and G-blocks), alginate purity, the cross-linking ions (calcium or barium), and the presence or absence of additional polymer coating layers influence the success of cell encapsulation. This review summarizes the outcomes of long-term studies of alginate-encapsulated islet transplants in animals and humans and provides a critical discussion of the graft failure mechanisms, including issues with graft biocompatibility, transplantation site, and integrity of the encapsulated islet grafts. Strategies to improve the mechanical stability of alginate capsules and methods for monitoring graft survival and function in vivo are presented.
Collapse
Affiliation(s)
- Melanie Köllmer
- 1 Department of Biomedical Engineering, Illinois Institute of Technology , Chicago, Illinois
| | - Alyssa A Appel
- 1 Department of Biomedical Engineering, Illinois Institute of Technology , Chicago, Illinois.,2 Research Service, Hines Veterans Administration Hospital , Hines, Illinois
| | - Sami I Somo
- 1 Department of Biomedical Engineering, Illinois Institute of Technology , Chicago, Illinois.,2 Research Service, Hines Veterans Administration Hospital , Hines, Illinois
| | - Eric M Brey
- 1 Department of Biomedical Engineering, Illinois Institute of Technology , Chicago, Illinois.,2 Research Service, Hines Veterans Administration Hospital , Hines, Illinois
| |
Collapse
|
43
|
Collins DJ, Neild A, deMello A, Liu AQ, Ai Y. The Poisson distribution and beyond: methods for microfluidic droplet production and single cell encapsulation. LAB ON A CHIP 2015; 15:3439-59. [PMID: 26226550 DOI: 10.1039/c5lc00614g] [Citation(s) in RCA: 345] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
There is a recognized and growing need for rapid and efficient cell assays, where the size of microfluidic devices lend themselves to the manipulation of cellular populations down to the single cell level. An exceptional way to analyze cells independently is to encapsulate them within aqueous droplets surrounded by an immiscible fluid, so that reagents and reaction products are contained within a controlled microenvironment. Most cell encapsulation work has focused on the development and use of passive methods, where droplets are produced continuously at high rates by pumping fluids from external pressure-driven reservoirs through defined microfluidic geometries. With limited exceptions, the number of cells encapsulated per droplet in these systems is dictated by Poisson statistics, reducing the proportion of droplets that contain the desired number of cells and thus the effective rate at which single cells can be encapsulated. Nevertheless, a number of recently developed actively-controlled droplet production methods present an alternative route to the production of droplets at similar rates and with the potential to improve the efficiency of single-cell encapsulation. In this critical review, we examine both passive and active methods for droplet production and explore how these can be used to deterministically and non-deterministically encapsulate cells.
Collapse
Affiliation(s)
- David J Collins
- Engineering Product Design pillar, Singapore University of Technology and Design, Singapore.
| | | | | | | | | |
Collapse
|
44
|
Boettler T, Schneider D, Cheng Y, Kadoya K, Brandon EP, Martinson L, von Herrath M. Pancreatic Tissue Transplanted in TheraCyte Encapsulation Devices Is Protected and Prevents Hyperglycemia in a Mouse Model of Immune-Mediated Diabetes. Cell Transplant 2015; 25:609-14. [PMID: 26300527 DOI: 10.3727/096368915x688939] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Type 1 diabetes (T1D) is characterized by destruction of glucose-responsive insulin-producing pancreatic β-cells and exhibits immune infiltration of pancreatic islets, where CD8 lymphocytes are most prominent. Curative transplantation of pancreatic islets is seriously hampered by the persistence of autoreactive immune cells that require high doses of immunosuppressive drugs. An elegant approach to confer graft protection while obviating the need for immunosuppression is the use of encapsulation devices that allow for the transfer of oxygen and nutrients, yet prevent immune cells from making direct contact with the islet grafts. Here we demonstrate that macroencapsulation devices (TheraCyte) loaded with neonatal pancreatic tissue and transplanted into RIP-LCMV.GP mice prevented disease onset in a model of virus-induced diabetes mellitus. Histological analyses revealed that insulin-producing cells survived within the device in animal models of diabetes. Our results demonstrate that these encapsulation devices can protect from an immune-mediated attack and can contain a sufficient amount of insulin-producing cells to prevent overt hyperglycemia.
Collapse
Affiliation(s)
- Tobias Boettler
- Type 1 Diabetes Center at the La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Nyitray CE, Chang R, Faleo G, Lance KD, Bernards DA, Tang Q, Desai T. Polycaprolactone Thin-Film Micro- and Nanoporous Cell-Encapsulation Devices. ACS NANO 2015; 9:5675-82. [PMID: 25950860 PMCID: PMC4628825 DOI: 10.1021/acsnano.5b00679] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Cell-encapsulating devices can play an important role in advancing the types of tissue available for transplantation and further improving transplant success rates. To have an effective device, encapsulated cells must remain viable, respond to external stimulus, and be protected from immune responses, and the device itself must elicit a minimal foreign body response. To address these challenges, we developed a micro- and a nanoporous thin-film cell encapsulation device from polycaprolactone (PCL), a material previously used in FDA-approved biomedical devices. The thin-film device construct allows long-term bioluminescent transfer imaging, which can be used for monitoring cell viability and device tracking. The ability to tune the microporous and nanoporous membrane allows selective protection from immune cell invasion and cytokine-mediated cell death in vitro, all while maintaining typical cell function, as demonstrated by encapsulated cells' insulin production in response to glucose stimulation. To demonstrate the ability to track, visualize, and monitor the viability of cells encapsulated in implanted thin-film devices, we encapsulated and implanted luciferase-positive MIN6 cells in allogeneic mouse models for up to 90 days. Lack of foreign body response in combination with rapid neovascularization around the device shows promise in using this technology for cell encapsulation. These devices can help elucidate the metrics required for cell encapsulation success and direct future immune-isolation therapies.
Collapse
Affiliation(s)
- Crystal E. Nyitray
- Program in Chemistry and Chemical Biology, University of California, San Francisco, 1700 4th Street, Byers Hall, Box 2520, San Francisco, California 94158, United States
| | - Ryan Chang
- UCB/UCSF Joint Program in Bioengineering, University of California, San Francisco, 1700 4th Street, Byers Hall, Box 2520, San Francisco, California 94158, United States
| | - Gaetano Faleo
- Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue HSE520 Box 0780, San Francisco, California 94143, United States
| | - Kevin D. Lance
- UCB/UCSF Joint Program in Bioengineering, University of California, San Francisco, 1700 4th Street, Byers Hall, Box 2520, San Francisco, California 94158, United States
| | - Daniel A. Bernards
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1700 4th Street, Byers Hall, Box 2520, San Francisco, California 94158, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue HSE520 Box 0780, San Francisco, California 94143, United States
| | - TejalA Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1700 4th Street, Byers Hall, Box 2520, San Francisco, California 94158, United States
| |
Collapse
|
46
|
Abstract
Stem cells have great potential in basic research and are being slowly integrated into toxicological research. This symposium provided an overview of the state of the field, stem cell models, described allogenic stem cell treatments and issues of immunogenicity associated with protein therapeutics, and tehn concentrated on stem cell uses in regenerative medicine focusing on lung and testing strategies on engineered tissues from a pathologist's perspective.
Collapse
Affiliation(s)
- Alan Trounson
- Monash University, Hudson Institute for Medical Research, Clayton, Victoria, Australia
| | | | - Thomas Petersen
- United Therapeutics Corporation, Regenerative Medicine, Research Triangle Park, NC, USA
| | | | - Maralee McVean
- Pre-Clinical Research Services, Inc, Ft Collins, CO, USA
| | | |
Collapse
|
47
|
Veiseh O, Tang BC, Whitehead KA, Anderson DG, Langer R. Managing diabetes with nanomedicine: challenges and opportunities. Nat Rev Drug Discov 2015; 14:45-57. [PMID: 25430866 PMCID: PMC4751590 DOI: 10.1038/nrd4477] [Citation(s) in RCA: 375] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nanotechnology-based approaches hold substantial potential for improving the care of patients with diabetes. Nanoparticles are being developed as imaging contrast agents to assist in the early diagnosis of type 1 diabetes. Glucose nanosensors are being incorporated in implantable devices that enable more accurate and patient-friendly real-time tracking of blood glucose levels, and are also providing the basis for glucose-responsive nanoparticles that better mimic the body's physiological needs for insulin. Finally, nanotechnology is being used in non-invasive approaches to insulin delivery and to engineer more effective vaccine, cell and gene therapies for type 1 diabetes. Here, we analyse the current state of these approaches and discuss key issues for their translation to clinical practice.
Collapse
Affiliation(s)
- Omid Veiseh
- 1] Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [2] David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [3] Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave., Boston, Massachusetts 02115, USA. [4]
| | - Benjamin C Tang
- 1] David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [2] Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave., Boston, Massachusetts 02115, USA. [3]
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, 5000 Forbes Ave., Pittsburgh, Pennsylvania 15213, USA
| | - Daniel G Anderson
- 1] Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [2] David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [3] Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave., Boston, Massachusetts 02115, USA. [4] Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA. [5] Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Robert Langer
- 1] Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [2] David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA. [3] Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave., Boston, Massachusetts 02115, USA. [4] Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA. [5] Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
48
|
Salvadori M, Bertoni E. What's new in clinical solid organ transplantation by 2013. World J Transplant 2014; 4:243-266. [PMID: 25540734 PMCID: PMC4274595 DOI: 10.5500/wjt.v4.i4.243] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/11/2014] [Accepted: 07/27/2014] [Indexed: 02/05/2023] Open
Abstract
Innovative and exciting advances in the clinical science in solid organ transplantation continuously realize as the results of studies, clinical trials, international conferences, consensus conferences, new technologies and discoveries. This review will address to the full spectrum of news in transplantation, that verified by 2013. The key areas covered are the transplantation activity, with particular regards to the donors, the news for solid organs such as kidney, pancreas, liver, heart and lung, the news in immunosuppressive therapies, the news in the field of tolerance and some of the main complications following transplantation as infections and cancers. The period of time covered by the study starts from the international meetings held in 2012, whose results were published in 2013, up to the 2013 meetings, conferences and consensus published in the first months of 2014. In particular for every organ, the trends in numbers and survival have been reviewed as well as the most relevant problems such as organ preservation, ischemia reperfusion injuries, and rejections with particular regards to the antibody mediated rejection that involves all solid organs. The new drugs and strategies applied in organ transplantation have been divided into new way of using old drugs or strategies and drugs new not yet on the market, but on phase Ito III of clinical studies and trials.
Collapse
|
49
|
Kontturi LS, Collin EC, Murtomäki L, Pandit AS, Yliperttula M, Urtti A. Encapsulated cells for long-term secretion of soluble VEGF receptor 1: Material optimization and simulation of ocular drug response. Eur J Pharm Biopharm 2014; 95:387-97. [PMID: 25460143 DOI: 10.1016/j.ejpb.2014.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 10/14/2014] [Accepted: 10/14/2014] [Indexed: 12/26/2022]
Abstract
Anti-angiogenic therapies with vascular endothelial growth factor (VEGF) inhibiting factors are effective treatment options for neovascular diseases of the retina, but these proteins can only be delivered as intravitreal (IVT) injections. To sustain a therapeutic drug level in the retina, VEGF inhibitors have to be delivered frequently, every 4-8weeks, causing inconvenience for the patients and expenses for the healthcare system. The aim of this study was to investigate cell encapsulation as a delivery system for prolonged anti-angiogenic treatment of retinal neovascularization. Genetically engineered ARPE-19 cells secreting soluble vascular endothelial growth factor receptor 1 (sVEGFR1) were encapsulated in a hydrogel of cross-linked collagen and interpenetrating hyaluronic acid (HA). The system was optimized in terms of matrix composition and cell density, and long-term cell viability and protein secretion measurements were performed. sVEGFR1 ARPE-19 cells in the optimized hydrogel remained viable and secreted sVEGFR1 at a constant rate for at least 50days. Based on pharmacokinetic/pharmacodynamic (PK/PD) modeling, delivery of sVEGFR1 from this cell encapsulation system is expected to lead only to modest VEGF inhibition, but improvements of the protein structure and/or secretion rate should result in strong and prolonged therapeutic effect. In conclusion, the hydrogel matrix herein supported the survival and protein secretion from the encapsulated cells. The PK/PD simulation is a convenient approach to predict the efficiency of the cell encapsulation system before in vivo experiments.
Collapse
Affiliation(s)
- Leena-Stiina Kontturi
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland.
| | - Estelle C Collin
- Network of Excellence for Functional Biomaterials, National University of Ireland, Galway, Ireland
| | | | - Abhay S Pandit
- Network of Excellence for Functional Biomaterials, National University of Ireland, Galway, Ireland
| | - Marjo Yliperttula
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Arto Urtti
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
50
|
Sartipy P, Björquist P. Employment of the Triple Helix concept for development of regenerative medicine applications based on human pluripotent stem cells. Clin Transl Med 2014; 3:9. [PMID: 24872863 PMCID: PMC4018621 DOI: 10.1186/2001-1326-3-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/30/2014] [Indexed: 11/29/2022] Open
Abstract
Using human pluripotent stem cells as a source to generate differentiated progenies for regenerative medicine applications has attracted substantial interest during recent years. Having the capability to produce large quantities of human cells that can replace damaged tissue due to disease or injury opens novel avenues for relieving symptoms and also potentially offers cures for many severe human diseases. Although tremendous advancements have been made, there is still much research and development left before human pluripotent stem cell derived products can be made available for cell therapy applications. In order to speed up the development processes, we argue strongly in favor of cross-disciplinary collaborative efforts which have many advantages, especially in a relatively new field such as regenerative medicine based on human pluripotent stem cells. In this review, we aim to illustrate how some of the hurdles for bringing human pluripotent stem cell derivatives from bench-to-bed can be effectively addressed through the establishment of collaborative programs involving academic institutions, biotech industries, and pharmaceutical companies. By taking advantage of the strengths from each organization, innovation and productivity can be maximized from a resource perspective and thus, the chances of successfully bringing novel regenerative medicine treatment options to patients increase.
Collapse
Affiliation(s)
- Peter Sartipy
- Cellectis AB, Arvid Wallgrens Backe 20, 413 46 Göteborg, Sweden ; Systems Biology Research Center, School of Bioscience, University of Skövde, 541 28 Skövde, Sweden
| | - Petter Björquist
- Cellectis AB, Arvid Wallgrens Backe 20, 413 46 Göteborg, Sweden ; Current address: NovaHep AB, Arvid Wallgrens Backe 20, 413 46 Göteborg, Sweden
| |
Collapse
|