1
|
Pham JA, Coronel MM. Unlocking Transplant Tolerance with Biomaterials. Adv Healthc Mater 2025; 14:e2400965. [PMID: 38843866 PMCID: PMC11834385 DOI: 10.1002/adhm.202400965] [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: 03/14/2024] [Revised: 05/31/2024] [Indexed: 07/04/2024]
Abstract
For patients suffering from organ failure due to injury or autoimmune disease, allogeneic organ transplantation with chronic immunosuppression is considered the god standard in terms of clinical treatment. However, the true "holy grail" of transplant immunology is operational tolerance, in which the recipient exhibits a sustained lack of alloreactivity toward unencountered antigen presented by the donor graft. This outcome is resultant from critical changes to the phenotype and genotype of the immune repertoire predicated by the activation of specific signaling pathways responsive to soluble and mechanosensitive cues. Biomaterials have emerged as a medium for interfacing with and reprogramming these endogenous pathways toward tolerance in precise, minimally invasive, and spatiotemporally defined manners. By viewing seminal and contemporary breakthroughs in transplant tolerance induction through the lens of biomaterials-mediated immunomodulation strategies-which include intrinsic material immunogenicity, the depot effect, graft coatings, induction and delivery of tolerogenic immune cells, biomimicry of tolerogenic immune cells, and in situ reprogramming-this review emphasizes the stunning diversity of approaches in the field and spotlights exciting future directions for research to come.
Collapse
Affiliation(s)
- John‐Paul A. Pham
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
- Elizabeth Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
| | - María M. Coronel
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
- Elizabeth Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
| |
Collapse
|
2
|
Grattoni A, Korbutt G, Tomei AA, García AJ, Pepper AR, Stabler C, Brehm M, Papas K, Citro A, Shirwan H, Millman JR, Melero-Martin J, Graham M, Sefton M, Ma M, Kenyon N, Veiseh O, Desai TA, Nostro MC, Marinac M, Sykes M, Russ HA, Odorico J, Tang Q, Ricordi C, Latres E, Mamrak NE, Giraldo J, Poznansky MC, de Vos P. Harnessing cellular therapeutics for type 1 diabetes mellitus: progress, challenges, and the road ahead. Nat Rev Endocrinol 2025; 21:14-30. [PMID: 39227741 PMCID: PMC11938328 DOI: 10.1038/s41574-024-01029-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 09/05/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is a growing global health concern that affects approximately 8.5 million individuals worldwide. T1DM is characterized by an autoimmune destruction of pancreatic β cells, leading to a disruption in glucose homeostasis. Therapeutic intervention for T1DM requires a complex regimen of glycaemic monitoring and the administration of exogenous insulin to regulate blood glucose levels. Advances in continuous glucose monitoring and algorithm-driven insulin delivery devices have improved the quality of life of patients. Despite this, mimicking islet function and complex physiological feedback remains challenging. Pancreatic islet transplantation represents a potential functional cure for T1DM but is hindered by donor scarcity, variability in harvested cells, aggressive immunosuppressive regimens and suboptimal clinical outcomes. Current research is directed towards generating alternative cell sources, improving transplantation methods, and enhancing cell survival without chronic immunosuppression. This Review maps the progress in cell replacement therapies for T1DM and outlines the remaining challenges and future directions. We explore the state-of-the-art strategies for generating replenishable β cells, cell delivery technologies and local targeted immune modulation. Finally, we highlight relevant animal models and the regulatory aspects for advancing these technologies towards clinical deployment.
Collapse
Affiliation(s)
- Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Surgery, Houston Methodist Hospital, Houston, TX, USA.
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| | - Gregory Korbutt
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrew R Pepper
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Cherie Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
- Diabetes Institute, University of Florida, Gainesville, FL, USA
| | - Michael Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Klearchos Papas
- Department of Surgery, The University of Arizona, Tucson, AZ, USA
| | - Antonio Citro
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Haval Shirwan
- Department of Pediatrics, Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Juan Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Melanie Graham
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA
| | - Michael Sefton
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Norma Kenyon
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Tejal A Desai
- University of California, San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, CA, USA
- Brown University, School of Engineering, Providence, RI, USA
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | - Megan Sykes
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
- Department of Surgery, Columbia University, New York, NY, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Jon Odorico
- UW Health Transplant Center, Madison, WI, USA
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Qizhi Tang
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
- Department of Surgery, University of California San Francisco, San Francisco, CA, US
- Gladstone Institute of Genomic Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Esther Latres
- Research Department, Breakthrough T1D, New York, NY, USA
| | | | - Jaime Giraldo
- Research Department, Breakthrough T1D, New York, NY, USA.
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands.
| |
Collapse
|
3
|
Gonzalez-Sanchez FA, Sanchez-Huerta TM, Huerta-Gonzalez A, Sepulveda-Villegas M, Altamirano J, Aguilar-Aleman JP, Garcia-Varela R. Diabetes current and future translatable therapies. Endocrine 2024; 86:865-881. [PMID: 38971945 DOI: 10.1007/s12020-024-03944-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/23/2024] [Indexed: 07/08/2024]
Abstract
Diabetes is one of the major diseases and concerns of public health systems that affects over 200 million patients worldwide. It is estimated that 90% of these patients suffer from diabetes type 2, while 10% present diabetes type 1. This type of diabetes and certain types of diabetes type 2, are characterized by dysregulation of blood glycemic levels due to the total or partial depletion of insulin-secreting pancreatic β-cells. Different approaches have been proposed for long-term treatment of insulin-dependent patients; amongst them, cell-based approaches have been the subject of basic and clinical research since they allow blood glucose level sensing and in situ insulin secretion. The current gold standard for insulin-dependent patients is on-demand exogenous insulin application; cell-based therapies aim to remove this burden from the patient and caregivers. In recent years, protocols to isolate and implant pancreatic islets from diseased donors have been developed and tested in clinical trials. Nevertheless, the shortage of donors, along with the need of immunosuppressive companion therapies, have pushed researchers to focus their attention and efforts to overcome these disadvantages and develop alternative strategies. This review discusses current tested clinical approaches and future potential alternatives for diabetes type 1, and some diabetes type 2, insulin-dependent patients. Additionally, advantages and disadvantages of these discussed methods.
Collapse
Affiliation(s)
- Fabio Antonio Gonzalez-Sanchez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Departamento de Bioingeniería y Biotecnología, Av. General Ramon Corona No 2514, Colonia Nuevo Mexico, CP 45201, Zapopan, Jalisco, México
| | - Triana Mayra Sanchez-Huerta
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Departamento de Bioingeniería y Biotecnología, Av. General Ramon Corona No 2514, Colonia Nuevo Mexico, CP 45201, Zapopan, Jalisco, México
| | - Alexandra Huerta-Gonzalez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Departamento de Bioingeniería y Biotecnología, Av. General Ramon Corona No 2514, Colonia Nuevo Mexico, CP 45201, Zapopan, Jalisco, México
| | - Maricruz Sepulveda-Villegas
- Departamento de Medicina Genómica y Hepatología, Hospital Civil de Guadalajara, "Fray Antonio Alcalde", Guadalajara, 44280, Jalisco, Mexico
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, 44100, Jalisco, Mexico
| | - Julio Altamirano
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Epigmenio González 500, San Pablo, 76130, Santiago de Queretaro, Qro, México
| | - Juan Pablo Aguilar-Aleman
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Departamento de Ingenieria Biomedica, Av. General Ramon Corona No 2514, Colonia Nuevo Mexico, CP 45201, Zapopan, Jalisco, México
| | - Rebeca Garcia-Varela
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Departamento de Bioingeniería y Biotecnología, Av. General Ramon Corona No 2514, Colonia Nuevo Mexico, CP 45201, Zapopan, Jalisco, México.
- Carbone Cancer Center, University of Wisconsin - Madison, 1111 Highland Ave, Wisconsin, 53705, Madison, USA.
| |
Collapse
|
4
|
Grimus S, Sarangova V, Welzel PB, Ludwig B, Seissler J, Kemter E, Wolf E, Ali A. Immunoprotection Strategies in β-Cell Replacement Therapy: A Closer Look at Porcine Islet Xenotransplantation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401385. [PMID: 38884159 PMCID: PMC11336975 DOI: 10.1002/advs.202401385] [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: 02/06/2024] [Revised: 05/28/2024] [Indexed: 06/18/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is characterized by absolute insulin deficiency primarily due to autoimmune destruction of pancreatic β-cells. The prevailing treatment for T1DM involves daily subcutaneous insulin injections, but a substantial proportion of patients face challenges such as severe hypoglycemic episodes and poorly controlled hyperglycemia. For T1DM patients, a more effective therapeutic option involves the replacement of β-cells through allogeneic transplantation of either the entire pancreas or isolated pancreatic islets. Unfortunately, the scarcity of transplantable human organs has led to a growing list of patients waiting for an islet transplant. One potential alternative is xenotransplantation of porcine pancreatic islets. However, due to inter-species molecular incompatibilities, porcine tissues trigger a robust immune response in humans, leading to xenograft rejection. Several promising strategies aim to overcome this challenge and enhance the long-term survival and functionality of xenogeneic islet grafts. These strategies include the use of islets derived from genetically modified pigs, immunoisolation of islets by encapsulation in biocompatible materials, and the creation of an immunomodulatory microenvironment by co-transplanting islets with accessory cells or utilizing immunomodulatory biomaterials. This review concentrates on delineating the primary obstacles in islet xenotransplantation and elucidates the fundamental principles and recent breakthroughs aimed at addressing these challenges.
Collapse
Affiliation(s)
- Sarah Grimus
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
| | - Victoria Sarangova
- Leibniz‐Institut für Polymerforschung Dresden e.V.Max Bergmann Center of Biomaterials DresdenD‐01069DresdenGermany
| | - Petra B. Welzel
- Leibniz‐Institut für Polymerforschung Dresden e.V.Max Bergmann Center of Biomaterials DresdenD‐01069DresdenGermany
| | - Barbara Ludwig
- Department of Medicine IIIUniversity Hospital Carl Gustav CarusTechnische Universität DresdenD‐01307DresdenGermany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the Technische Universität DresdenD‐01307DresdenGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
- DFG‐Center for Regenerative Therapies DresdenTechnische Universität DresdenD‐01307DresdenGermany
| | - Jochen Seissler
- Medizinische Klinik und Poliklinik IVDiabetes Zentrum – Campus InnenstadtKlinikum der Ludwig‐Maximilians‐Universität MünchenD‐80336MunichGermany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
- German Center for Diabetes Research (DZD e.V.)D‐85764NeuherbergGermany
| | - Asghar Ali
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU MunichD‐81377MunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichD‐85764OberschleißheimGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichD‐81377MunichGermany
| |
Collapse
|
5
|
Maurya R, Misro L, Boini T, Radhakrishnan T, Nair PG, Gaidhani SN, Jain A. Transforming Medicinal Oil into Advanced Gel: An Update on Advancements. Gels 2024; 10:342. [PMID: 38786260 PMCID: PMC11121385 DOI: 10.3390/gels10050342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 05/25/2024] Open
Abstract
The present study delves into the evolution of traditional Ayurvedic oil preparations through innovative strategies to develop advanced gel formulations, aiming at amplifying their therapeutic efficacy. Ayurvedic oils have a rich historical context in healing practices, yet their conversion into contemporary gel-based formulations represents a revolutionary approach to augment their medicinal potential. The primary objective of this transformation is to leverage scientific advancements and modern pharmaceutical techniques to enhance the application, absorption, and overall therapeutic impact of these traditional remedies. By encapsulating the essential constituents of Ayurvedic oils within gel matrices, these novel strategies endeavor to improve their stability, bioavailability, and targeted delivery mechanisms. This review highlights the fusion of traditional Ayurvedic wisdom with cutting-edge pharmaceutical technology, paving the way for more effective and accessible utilization of these revered remedies in modern healthcare.
Collapse
Affiliation(s)
- Rahul Maurya
- National Ayurveda Research Institute for Panchakarma, CCRAS, Ministry of AYUSH, Government of India, Cheruthuruthy, Thrissur 679531, India; (L.M.); (T.B.); (T.R.); (P.G.N.); (S.N.G.)
| | - Lakshminarayana Misro
- National Ayurveda Research Institute for Panchakarma, CCRAS, Ministry of AYUSH, Government of India, Cheruthuruthy, Thrissur 679531, India; (L.M.); (T.B.); (T.R.); (P.G.N.); (S.N.G.)
| | - Thirupataiah Boini
- National Ayurveda Research Institute for Panchakarma, CCRAS, Ministry of AYUSH, Government of India, Cheruthuruthy, Thrissur 679531, India; (L.M.); (T.B.); (T.R.); (P.G.N.); (S.N.G.)
| | - Thulasi Radhakrishnan
- National Ayurveda Research Institute for Panchakarma, CCRAS, Ministry of AYUSH, Government of India, Cheruthuruthy, Thrissur 679531, India; (L.M.); (T.B.); (T.R.); (P.G.N.); (S.N.G.)
| | - Parvathy G. Nair
- National Ayurveda Research Institute for Panchakarma, CCRAS, Ministry of AYUSH, Government of India, Cheruthuruthy, Thrissur 679531, India; (L.M.); (T.B.); (T.R.); (P.G.N.); (S.N.G.)
| | - Sudesh N. Gaidhani
- National Ayurveda Research Institute for Panchakarma, CCRAS, Ministry of AYUSH, Government of India, Cheruthuruthy, Thrissur 679531, India; (L.M.); (T.B.); (T.R.); (P.G.N.); (S.N.G.)
| | - Ankit Jain
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani 333031, India
| |
Collapse
|
6
|
Han H, Zhan T, Guo N, Cui M, Xu Y. Cryopreservation of organoids: Strategies, innovation, and future prospects. Biotechnol J 2024; 19:e2300543. [PMID: 38403430 DOI: 10.1002/biot.202300543] [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: 10/09/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 02/27/2024]
Abstract
Organoid technology has demonstrated unique advantages in multidisciplinary fields such as disease research, tumor drug sensitivity, clinical immunity, drug toxicology, and regenerative medicine. It will become the most promising research tool in translational research. However, the long preparation time of organoids and the lack of high-quality cryopreservation methods limit the further application of organoids. Although the high-quality cryopreservation of small-volume biological samples such as cells and embryos has been successfully achieved, the existing cryopreservation methods for organoids still face many bottlenecks. In recent years, with the development of materials science, cryobiology, and interdisciplinary research, many new materials and methods have been applied to cryopreservation. Several new cryopreservation methods have emerged, such as cryoprotectants (CPAs) of natural origin, ice-controlled biomaterials, and rapid rewarming methods. The introduction of these technologies has expanded the research scope of cryopreservation of organoids, provided new approaches and methods for cryopreservation of organoids, and is expected to break through the current technical bottleneck of cryopreservation of organoids. This paper reviews the progress of cryopreservation of organoids in recent years from three aspects: damage factors of cryopreservation of organoids, new protective agents and loading methods, and new technologies of cryopreservation and rewarming.
Collapse
Affiliation(s)
- Hengxin Han
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China
| | - Taijie Zhan
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China
| | - Ning Guo
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China
| | - Mengdong Cui
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China
| | - Yi Xu
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, China
| |
Collapse
|
7
|
Li Y, He C, Liu R, Xiao Z, Sun B. Stem cells therapy for diabetes: from past to future. Cytotherapy 2023; 25:1125-1138. [PMID: 37256240 DOI: 10.1016/j.jcyt.2023.04.012] [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: 01/26/2023] [Revised: 04/05/2023] [Accepted: 04/24/2023] [Indexed: 06/01/2023]
Abstract
Diabetes mellitus is a chronic disease of carbohydrate metabolism characterized by uncontrolled hyperglycemia due to the body's impaired ability to produce or respond to insulin. Oral or injectable exogenous insulin and its analogs cannot mimic endogenous insulin secreted by healthy individuals, and pancreatic and islet transplants face a severe shortage of sources and transplant complications, all of which limit the widespread use of traditional strategies in diabetes treatment. We are now in the era of stem cells and their potential in ameliorating human disease. At the same time, the rapid development of gene editing and cell-encapsulation technologies has added to the wings of stem cell therapy. However, there are still many unanswered questions before stem cell therapy can be applied clinically to patients with diabetes. In this review, we discuss the progress of strategies to obtain insulin-producing cells from different types of stem cells, the application of gene editing in stem cell therapy for diabetes, as well as summarize the current advanced cell encapsulation technologies in diabetes therapy and look forward to the future development of stem cell therapy in diabetes.
Collapse
Affiliation(s)
- Yumin Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Cong He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China; Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital,The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Rui Liu
- Department of Genetic Engineering, College of Natural Science, University of Suwon, Kyunggi-Do, Republic of Korea
| | - Zhongdang Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China.
| | - Bo Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
8
|
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: 0] [Impact Index Per Article: 0] [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
|
9
|
Whulanza Y, Nathani RC, Adimillenva K, Irwansyah R, Wahyu Nurhayati R, Utomo MS, Abdullah AH. Effect of Flow Rate Modulation on Alginate Emulsification in Multistage Microfluidics. MICROMACHINES 2023; 14:1828. [PMID: 37893265 PMCID: PMC10609249 DOI: 10.3390/mi14101828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023]
Abstract
The encapsulation of stem cells into alginate microspheres is an important aspect of tissue engineering or bioprinting which ensures cell growth and development. We previously demonstrated the encapsulation of stem cells using the hanging drop method. However, this conventional process takes a relatively long time and only produces a small-volume droplet. Here, an experimental approach for alginate emulsification in multistage microfluidics is reported. By using the microfluidic method, the emulsification of alginate in oil can be manipulated by tuning the flow rate for both phases. Two-step droplet emulsification is conducted in a series of polycarbonate and polydimethylsiloxane microfluidic chips. Multistage emulsification of alginate for stem cell encapsulation has been successfully reported in this study under certain flow rates. Fundamental non-dimensional numbers such as Reynolds and capillary are used to evaluate the effect of flow rate on the emulsification process. Reynolds numbers of around 0.5-2.5 for alginate/water and 0.05-0.2 for oil phases were generated in the current study. The capillary number had a maximum value of 0.018 to ensure the formation of plug flow. By using the multistage emulsification system, the flow rates of each process can be tuned independently, offering a wider range of droplet sizes that can be produced. A final droplet size of 500-1000 µm can be produced using flow rates of 0.1-0.5 mL/h and 0.7-2.4 mL/h for the first stage and second stage, respectively.
Collapse
Affiliation(s)
- Yudan Whulanza
- Department of Mechanical Engineering, Faculty of Engineering, Universitas Indonesia, Depok 16424, Indonesia
- Research Center for Biomedical Engineering, Faculty of Engineering, Universitas Indonesia, Depok 16424, Indonesia
| | - Rithwik Chandur Nathani
- Department of Mechanical Engineering, Faculty of Engineering, Universitas Indonesia, Depok 16424, Indonesia
| | - Klaugusta Adimillenva
- Department of Mechanical Engineering, Faculty of Engineering, Universitas Indonesia, Depok 16424, Indonesia
| | - Ridho Irwansyah
- Department of Mechanical Engineering, Faculty of Engineering, Universitas Indonesia, Depok 16424, Indonesia
| | - Retno Wahyu Nurhayati
- Research Center for Biomedical Engineering, Faculty of Engineering, Universitas Indonesia, Depok 16424, Indonesia
- Department of Chemical Engineering, Faculty of Engineering, Universitas Indonesia, Depok 16424, Indonesia
- Stem Cell and Tissue Engineering Research Center, Indonesia Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | | | - Abdul Halim Abdullah
- Biomechanical & Clinical Engineering Research Group, School of Mechanical Engineering, College of Engineering, Universiti Teknologi MARA, Shah Alam 40450, Malaysia
| |
Collapse
|
10
|
Liao D, Liu C, Chen S, Liu F, Li W, Shangguan D, Shi Y. Recent advances in immune checkpoint inhibitor-induced type 1 diabetes mellitus. Int Immunopharmacol 2023; 122:110414. [PMID: 37390646 DOI: 10.1016/j.intimp.2023.110414] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 07/02/2023]
Abstract
As a new group of anticancer drugs, immune checkpoint inhibitors (ICIs) have exhibited favorable antitumor efficacy in numerous malignant tumors. Anti-cytotoxic T lymphocyte associated antigen-4 (CTLA-4), anti-programmed cell death-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) are three kinds of ICIs widely used in clinical practice. However, ICI therapy (monotherapy or combination therapy) is always accompanied by a unique toxicity profile known as immune-related adverse events (irAEs) affecting multiple organs. The endocrine glands are common targets of irAEs induced by ICIs, which cause type 1 diabetes mellitus (T1DM) when the pancreas is affected. Although the incidence rate of ICI-induced T1DM is rare, it will always lead to an irreversible impairment of β-cells and be potentially life-threatening. Hence, it is vital for endocrinologists and oncologists to obtain a comprehensive understanding of ICI-induced T1DM and its management. In our present manuscript, we have reviewed the epidemiology, pathology and mechanism, diagnosis, management, and treatments of ICI-induced T1DM.
Collapse
Affiliation(s)
- Dehua Liao
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China
| | - Chaoyi Liu
- Department of Information, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China
| | - Shanshan Chen
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China
| | - Fen Liu
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China
| | - Wei Li
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China
| | - Dangang Shangguan
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China.
| | - Yingrui Shi
- Department of Radiation Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China.
| |
Collapse
|
11
|
Han H, Zhan T, Cui M, Guo N, Dang H, Yang G, Shu S, He W, Xu Y. Investigation of Rapid Rewarming Chips for Cryopreservation by Joule Heating. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:11048-11062. [PMID: 37497679 DOI: 10.1021/acs.langmuir.3c01364] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Rapid and uniform rewarming is critical to cryopreservation. Current rapid rewarming methods require complex physical field application devices (such as lasers or radio frequencies) and the addition of nanoparticles as heating media. These complex devices and nanoparticles limit the promotion of the rapid rewarming method and pose potential biosafety concerns. In this work, a joule heating-based rapid electric heating chip (EHC) was designed for cryopreservation. Uniform and rapid rewarming of biological samples in different volumes can be achieved through simple operations. EHC loaded with 0.28 mL of CPA solution can achieve a rewarming rate of 3.2 × 105 °C/min (2.8 mL with 2.3 × 103 °C/min), approximately 2 orders of magnitude greater than the rewarming rates observed with an equal capacity straw when combined with laser nanowarming or magnetic induction heating. In addition, the degree of supercooling can be significantly reduced without manual nucleation during the cooling of the EHC. Subsequently, the results of cryopreservation validation of cells and spheroids showed that the cell viability and spheroid structural integrity were significantly improved after cryopreservation. The viability of human lung adenocarcinoma (A549) cells postcryopreservation was 97.2%, which was significantly higher than 93% in the cryogenic vials (CV) group. Similar results were seen in human mesenchymal stem cells (MSCs), with 93.18% cell survival in the EHC group, significantly higher than 86.83% in the CV group, and cells in the EHC group were also significantly better than those in the CV group for further apoptosis and necrosis assays. This work provides an efficient rewarming protocol for the cryopreservation of biological samples, significantly improving the quantity and quality of cells and spheroids postcryopreservation.
Collapse
Affiliation(s)
- Hengxin Han
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai 200093, China
| | - Taijie Zhan
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai 200093, China
| | - Mengdong Cui
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai 200093, China
| | - Ning Guo
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai 200093, China
| | - Hangyu Dang
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai 200093, China
| | - Guoliang Yang
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai 200093, China
| | - Shuang Shu
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai 200093, China
| | - Wei He
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai 200093, China
| | - Yi Xu
- Institute of Biothermal Science & Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Co-innovation Center for Energy Therapy of Tumors, Shanghai 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai 200093, China
| |
Collapse
|
12
|
Okcu A, Yazir Y, Şimşek T, Mert S, Duruksu G, Öztürk A, Kiliç KC, Akpinar G, Kasap M. Investigation of the effect of pancreatic decellularized matrix on encapsulated Islets of Langerhans with mesenchymal stem cells. Tissue Cell 2023; 82:102110. [PMID: 37235912 DOI: 10.1016/j.tice.2023.102110] [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: 02/22/2023] [Revised: 05/02/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023]
Abstract
OBJECTIVE In this study, it was aimed to provide a therapeutic approach for T1DM by encapsulating the pancreatic islets with mesenchymal stem cells and decellularized pancreatic extracellular matrix to support the survival of islets while maintaining their cellular activity. METHOD Pancreatic extracellular matrix was decellularized using different concentrations of detergent series. After the preparation of the protein-based tissue extracellular matrix was shown to be free of cells or any genetic material by molecular, immunofluorescence and histochemical techniques. Following the homogenization of the decellularized pancreatic extracellular matrix and the analysis of its protein composition by LC-MS, the matrix proteins were incorporated with pancreatic islets and rat adipose tissue-derived MSCs (rAT-MSCs) in alginate microcapsules. Glucose-stimulated insulin secretion property of the islet cells in the microbeads was evaluated by insulin ELISA. The gene expression profile of the encapsulated cells was analyzed by Real-Time PCR. RESULTS Unlike the protein composition of whole pancreatic tissue, the decellularized pancreas matrix was free of histone proteins or proteins originated from mitochondria. The protein matrix derived from pancreatic tissue was shown to support the growth and maintenance of the islet cells. When compared to the non-encapsulated pancreatic islet, the encapsulated cells demonstrate to be more efficient in terms of insulin expression. CONCLUSION The extracellular pancreatic matrix obtained in this study was directly used as supplementary in the alginate-based microcapsule enhancing the cell survival. The tissue matrix protein and alginate had a synergistic effect on total insulin secretion, which might have the potential to overcome the insulin deficiency. Despite the improvement in the cell viability and the number, the efficiency of the insulin secretion in response to glucose stimulation from the alginate microcapsules did not meet the expectation when compared with the non-encapsulated pancreatic islets.
Collapse
Affiliation(s)
- Alparslan Okcu
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey
| | - Yusufhan Yazir
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Department of Histology and Embryology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey.
| | - Turgay Şimşek
- Department of General Surgery, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Serap Mert
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Department of Chemistry and Chemical Processing Technology, Kocaeli University, Turkey; Department of Polymer Science and Technology, Kocaeli University, Kocaeli, Turkey
| | - Gökhan Duruksu
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey
| | - Ahmet Öztürk
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey
| | - Kamil Can Kiliç
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey
| | - Gürler Akpinar
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Murat Kasap
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
13
|
Challenges with Cell-based Therapies for Type 1 Diabetes Mellitus. Stem Cell Rev Rep 2022; 19:601-624. [PMID: 36434300 DOI: 10.1007/s12015-022-10482-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2022] [Indexed: 11/27/2022]
Abstract
Type 1 diabetes (T1D) is a chronic, lifelong metabolic disease. It is characterised by the autoimmune-mediated loss of insulin-producing pancreatic β cells in the islets of Langerhans (β-islets), resulting in disrupted glucose homeostasis. Administration of exogenous insulin is the most common management method for T1D, but this requires lifelong reliance on insulin injections and invasive blood glucose monitoring. Replacement therapies with beta cells are being developed as an advanced curative treatment for T1D. Unfortunately, this approach is limited by the lack of donated pancreatic tissue, the difficulties in beta cell isolation and viability maintenance, the longevity of the transplanted cells in vivo, and consequently high costs. Emerging approaches to address these limitations are under intensive investigations, including the production of insulin-producing beta cells from various stem cells, and the development of bioengineered devices including nanotechnologies for improving islet transplantation efficacy without the need for recipients taking toxic anti-rejection drugs. These emerging approaches present promising prospects, while the challenges with the new techniques need to be tackled for ultimately clinical treatment of T1D. This review discussed the benefits and limitations of the cell-based therapies for beta cell replacement as potential curative treatment for T1D, and the applications of bioengineered devices including nanotechnology to overcome the challenges associated with beta cell transplantation.
Collapse
|
14
|
Alkhursani SA, Ghobashy MM, Al-Gahtany SA, Meganid AS, Abd El-Halim SM, Ahmad Z, Khan FS, Atia GAN, Cavalu S. Application of Nano-Inspired Scaffolds-Based Biopolymer Hydrogel for Bone and Periodontal Tissue Regeneration. Polymers (Basel) 2022; 14:3791. [PMID: 36145936 PMCID: PMC9504130 DOI: 10.3390/polym14183791] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
This review's objectives are to provide an overview of the various kinds of biopolymer hydrogels that are currently used for bone tissue and periodontal tissue regeneration, to list the advantages and disadvantages of using them, to assess how well they might be used for nanoscale fabrication and biofunctionalization, and to describe their production processes and processes for functionalization with active biomolecules. They are applied in conjunction with other materials (such as microparticles (MPs) and nanoparticles (NPs)) and other novel techniques to replicate physiological bone generation more faithfully. Enhancing the biocompatibility of hydrogels created from blends of natural and synthetic biopolymers can result in the creation of the best scaffold match to the extracellular matrix (ECM) for bone and periodontal tissue regeneration. Additionally, adding various nanoparticles can increase the scaffold hydrogel stability and provide a number of biological effects. In this review, the research study of polysaccharide hydrogel as a scaffold will be critical in creating valuable materials for effective bone tissue regeneration, with a future impact predicted in repairing bone defects.
Collapse
Affiliation(s)
- Sheikha A. Alkhursani
- Faculty of Science and Humanities-Jubail, Imam Abdulrahman Bin Faisal University, Jubail 31441, Saudi Arabia
| | - Mohamed Mohamady Ghobashy
- Radiation Research of Polymer Chemistry Department, National Center for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo 11787, Egypt
| | | | - Abeer S. Meganid
- Faculty of Science and Humanities-Jubail, Imam Abdulrahman Bin Faisal University, Jubail 31441, Saudi Arabia
| | - Shady M. Abd El-Halim
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, October 6 University, 6th of October City, Giza 12585, Egypt
| | - Zubair Ahmad
- Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia
- Biology Department, College of Arts and Sciences, Dehran Al-Junub, King Khalid University, Abha 61413, Saudi Arabia
| | - Farhat S. Khan
- Biology Department, College of Arts and Sciences, Dehran Al-Junub, King Khalid University, Abha 61413, Saudi Arabia
| | - Gamal Abdel Nasser Atia
- Department of Oral Medicine, Periodontology and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia 41522, Egypt
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|
15
|
Huang H, Shang Y, Li H, Feng Q, Liu Y, Chen J, Dong H. Co-transplantation of Islets-Laden Microgels and Biodegradable O 2-Generating Microspheres for Diabetes Treatment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:38448-38458. [PMID: 35980755 DOI: 10.1021/acsami.2c07215] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Pancreatic islets transplantation is an optimal alternative to exogenous insulin injection for long-term effective type 1 diabetes treatment. However, direct islets transplantation without any protection can induce cell necrosis due to severe host immune rejection. Insufficient O2 supply induced by the lack of capillary network at the early stage of islets transplantation is another critical constraint limiting islets survival and insulin-secretion function. In this paper, we design a novel co-transplantation system composed of islets-laden nanocomposite microgels and O2-generating microspheres. In particular, nanocomposite microgels confer the encapsulated islets with simultaneous physical protection and chemical anti-inflammation/immunosuppression by covalently anchoring rapamycin-loaded cyclodextrin nanoparticles to microgel network. Meanwhile, O2-generating microspheres prepared by blending inorganic peroxides in biodegradable polycaprolactone and polylactic acid can generate in situ O2 gas and thus avoid hypoxia environment around transplanted islets. In vivo therapeutic effect of diabetic mice proves the reversion of the high blood glucose level back to normoglycemia and superior glucose tolerance for at least 90 days post co-transplantation. In brief, the localized drug and oxygen codelivery, as well as physical protection provided by our co-transplantation system, has the potential to overcome to a large extent the inflammatory, hypoxia, and host immune rejection after islets transplantation. This new strategy may have wider application in other cell replacement therapies.
Collapse
Affiliation(s)
- Hanhao Huang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| | - Yulian Shang
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Haofei Li
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510641, China
| | - Qi Feng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| | - Yang Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| | - Junlin Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| | - Hua Dong
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510641, China
| |
Collapse
|
16
|
Stock AA, Gonzalez GC, Pete SI, De Toni T, Berman DM, Rabassa A, Diaz W, Geary JC, Willman M, Jackson JM, DeHaseth NH, Ziebarth NM, Hogan AR, Ricordi C, Kenyon NS, Tomei AA. Performance of islets of Langerhans conformally coated via an emulsion cross-linking method in diabetic rodents and nonhuman primates. SCIENCE ADVANCES 2022; 8:eabm3145. [PMID: 35767620 PMCID: PMC9242596 DOI: 10.1126/sciadv.abm3145] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
Polyethylene glycol (PEG)-based conformal coating (CC) encapsulation of transplanted islets is a promising β cell replacement therapy for the treatment of type 1 diabetes without chronic immunosuppression because it minimizes capsule thickness, graft volume, and insulin secretion delay. However, we show here that our original CC method, the direct method, requiring exposure of islets to low pH levels and inclusion of viscosity enhancers during coating, severely affected the viability, scalability, and biocompatibility of CC islets in nonhuman primate preclinical models of type 1 diabetes. We therefore developed and validated in vitro and in vivo, in several small- and large-animal models of type 1 diabetes, an augmented CC method-emulsion method-that achieves hydrogel CCs around islets at physiological pH for improved cytocompatibility, with PEG hydrogels for increased biocompatibility and with fivefold increase in encapsulation throughput for enhanced scalability.
Collapse
Affiliation(s)
- Aaron A. Stock
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Grisell C. Gonzalez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sophia I. Pete
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Teresa De Toni
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Dora M. Berman
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alexander Rabassa
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Waldo Diaz
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - James C. Geary
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Melissa Willman
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joy M. Jackson
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Noa H. DeHaseth
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Noel M. Ziebarth
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Anthony R. Hogan
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Norma S. Kenyon
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alice A. Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
17
|
Domingo-Lopez DA, Lattanzi G, H. J. Schreiber L, Wallace EJ, Wylie R, O'Sullivan J, Dolan EB, Duffy GP. Medical devices, smart drug delivery, wearables and technology for the treatment of Diabetes Mellitus. Adv Drug Deliv Rev 2022; 185:114280. [PMID: 35405298 DOI: 10.1016/j.addr.2022.114280] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/21/2022] [Accepted: 04/05/2022] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus refers to a group of metabolic disorders which affect how the body uses glucose impacting approximately 9% of the population worldwide. This review covers the most recent technological advances envisioned to control and/or reverse Type 1 diabetes mellitus (T1DM), many of which will also prove effective in treating the other forms of diabetes mellitus. Current standard therapy for T1DM involves multiple daily glucose measurements and insulin injections. Advances in glucose monitors, hormone delivery systems, and control algorithms generate more autonomous and personalised treatments through hybrid and fully automated closed-loop systems, which significantly reduce hypo- and hyperglycaemic episodes and their subsequent complications. Bi-hormonal systems that co-deliver glucagon or amylin with insulin aim to reduce hypoglycaemic events or increase time spent in target glycaemic range, respectively. Stimuli responsive materials for the controlled delivery of insulin or glucagon are a promising alternative to glucose monitors and insulin pumps. By their self-regulated mechanism, these "smart" drugs modulate their potency, pharmacokinetics and dosing depending on patients' glucose levels. Islet transplantation is a potential cure for T1DM as it restores endogenous insulin and glucagon production, but its use is not yet widespread due to limited islet sources and risks of chronic immunosuppression. New encapsulation strategies that promote angiogenesis and oxygen delivery while protecting islets from recipients' immune response may overcome current limiting factors.
Collapse
|
18
|
Du S, Li Y, Geng Z, Zhang Q, Buhler LH, Gonelle-Gispert C, Wang Y. Engineering Islets From Stem Cells: The Optimal Solution for the Treatment of Diabetes? Front Immunol 2022; 13:869514. [PMID: 35572568 PMCID: PMC9092457 DOI: 10.3389/fimmu.2022.869514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetes is a metabolic disease characterized by insulin deficiency. Bioengineering of stem cells with the aim to restore insulin production and glucose regulation has the potential to cure diabetic patients. In this review, we focus on the recent developments for bioengineering of induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), and pancreatic progenitor cells in view of generating insulin producing and glucose regulating cells for β-cell replacement therapies. Recent clinical trials using islet cells derived from stem cells have been initiated for the transplantation into diabetic patients, with crucial bottlenecks of tumorigenesis, post-transplant survival, genetic instability, and immunogenicity that should be further optimized. As a new approach given high expectations, bioengineered islets from stem cells occupies considerable potential for the future clinical application and addressing the treatment dilemma of diabetes.
Collapse
Affiliation(s)
- Suya Du
- Department of Clinical Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanjiao Li
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhen Geng
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, China.,Institute of Organ Transplantation, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences, Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Qi Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Leo H Buhler
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, China.,Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | | | - Yi Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, China.,Institute of Organ Transplantation, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences, Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
19
|
De Toni T, Stock AA, Devaux F, Gonzalez GC, Nunez K, Rubanich JC, Safley SA, Weber CJ, Ziebarth NM, Buchwald P, Tomei AA. Parallel Evaluation of Polyethylene Glycol Conformal Coating and Alginate Microencapsulation as Immunoisolation Strategies for Pancreatic Islet Transplantation. Front Bioeng Biotechnol 2022; 10:886483. [PMID: 35651551 PMCID: PMC9149081 DOI: 10.3389/fbioe.2022.886483] [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: 02/28/2022] [Accepted: 04/12/2022] [Indexed: 01/21/2023] Open
Abstract
Pancreatic islet transplantation improves metabolic control and prevents complications in patients with brittle type 1 diabetes (T1D). However, chronic immunosuppression is required to prevent allograft rejection and recurrence of autoimmunity. Islet encapsulation may eliminate the need for immunosuppression. Here, we analyzed in parallel two microencapsulation platforms that provided long-term diabetes reversal in preclinical T1D models, alginate single and double capsules versus polyethylene glycol conformal coating, to identify benefits and weaknesses that could inform the design of future clinical trials with microencapsulated islets. We performed in vitro and in vivo functionality assays with human islets and analyzed the explanted grafts by immunofluorescence. We quantified the size of islets and capsules, measured capsule permeability, and used these data for in silico simulations of islet functionality in COMSOL Multiphysics. We demonstrated that insulin response to glucose stimulation is dependent on capsule size, and the presence of permselective materials augments delays in insulin secretion. Non-coated and conformally coated islets could be transplanted into the fat pad of diabetic mice, resulting in comparable functionality and metabolic control. Mac-2+ cells were found in conformally coated grafts, indicating possible host reactivity. Due to their larger volume, alginate capsules were transplanted in the peritoneal cavity. Despite achieving diabetes reversal, changes in islet composition were found in retrieved capsules, and recipient mice experienced hypoglycemia indicative of hyperinsulinemia induced by glucose retention in large capsules as the in silico model predicted. We concluded that minimal capsule size is critical for physiological insulin secretion, and anti-inflammatory modulation may be beneficial for small conformal capsules.
Collapse
Affiliation(s)
- Teresa De Toni
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States
| | - Aaron A. Stock
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States
| | - Floriane Devaux
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States
| | - Grisell C. Gonzalez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kailyn Nunez
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States
| | - Jessica C. Rubanich
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States
| | - Susan A. Safley
- Department of Surgery, Emory University, Atlanta, GA, United States
| | - Collin J. Weber
- Department of Surgery, Emory University, Atlanta, GA, United States
| | - Noel M. Ziebarth
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States
| | - Peter Buchwald
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Alice A. Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
- *Correspondence: Alice A. Tomei,
| |
Collapse
|
20
|
Burke JA, Zhang X, Bobbala S, Frey MA, Bohorquez Fuentes C, Freire Haddad H, Allen SD, Richardson RAK, Ameer GA, Scott EA. Subcutaneous nanotherapy repurposes the immunosuppressive mechanism of rapamycin to enhance allogeneic islet graft viability. NATURE NANOTECHNOLOGY 2022; 17:319-330. [PMID: 35039683 PMCID: PMC8934301 DOI: 10.1038/s41565-021-01048-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/09/2021] [Indexed: 05/03/2023]
Abstract
Standard oral rapamycin (that is, Rapamune) administration is plagued by poor bioavailability and broad biodistribution. Thus, this pleotropic mammalian target of rapamycin (mTOR) inhibitor has a narrow therapeutic window and numerous side effects and provides inadequate protection to transplanted cells and tissues. Furthermore, the hydrophobicity of rapamycin limits its use in parenteral formulations. Here, we demonstrate that subcutaneous delivery via poly(ethylene glycol)-b-poly(propylene sulfide) polymersome nanocarriers significantly alters rapamycin's cellular biodistribution to repurpose its mechanism of action for tolerance, instead of immunosuppression, and minimize side effects. While oral rapamycin inhibits T cell proliferation directly, subcutaneously administered rapamycin-loaded polymersomes modulate antigen presenting cells in lieu of T cells, significantly improving maintenance of normoglycemia in a clinically relevant, major histocompatibility complex-mismatched, allogeneic, intraportal (liver) islet transplantation model. These results demonstrate the ability of a rationally designed nanocarrier to re-engineer the immunosuppressive mechanism of a drug by controlling cellular biodistribution.
Collapse
Affiliation(s)
- Jacqueline A Burke
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Xiaomin Zhang
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Molly A Frey
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Carolina Bohorquez Fuentes
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Helena Freire Haddad
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Sean D Allen
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Reese A K Richardson
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Guillermo A Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA.
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA.
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
21
|
Fanaropoulou NM. Hope Injections: The Promises of Regenerative Medicine in Curing Type 1 Diabetes Mellitus. EJIFCC 2021; 32:392-397. [PMID: 34819828 PMCID: PMC8592635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
22
|
Samojlik MM, Stabler CL. Designing biomaterials for the modulation of allogeneic and autoimmune responses to cellular implants in Type 1 Diabetes. Acta Biomater 2021; 133:87-101. [PMID: 34102338 PMCID: PMC9148663 DOI: 10.1016/j.actbio.2021.05.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
The effective suppression of adaptive immune responses is essential for the success of allogeneic cell therapies. In islet transplantation for Type 1 Diabetes, pre-existing autoimmunity provides an additional hurdle, as memory autoimmune T cells mediate both an autoantigen-specific attack on the donor beta cells and an alloantigen-specific attack on the donor graft cells. Immunosuppressive agents used for islet transplantation are generally successful in suppressing alloimmune responses, but dramatically hinder the widespread adoption of this therapeutic approach and fail to control memory T cell populations, which leaves the graft vulnerable to destruction. In this review, we highlight the capacity of biomaterials to provide local and nuanced instruction to suppress or alter immune pathways activated in response to an allogeneic islet transplant. Biomaterial immunoisolation is a common approach employed to block direct antigen recognition and downstream cell-mediated graft destruction; however, immunoisolation alone still permits shed donor antigens to escape into the host environment, resulting in indirect antigen recognition, immune cell activation, and the creation of a toxic graft site. Designing materials to decrease antigen escape, improve cell viability, and increase material compatibility are all approaches that can decrease the local release of antigen and danger signals into the implant microenvironment. Implant materials can be further enhanced through the local delivery of anti-inflammatory, suppressive, chemotactic, and/or tolerogenic agents, which serve to control both the innate and adaptive immune responses to the implant with a benefit of reduced systemic effects. Lessons learned from understanding how to manipulate allogeneic and autogenic immune responses to pancreatic islets can also be applied to other cell therapies to improve their efficacy and duration. STATEMENT OF SIGNIFICANCE: This review explores key immunologic concepts and critical pathways mediating graft rejection in Type 1 Diabetes, which can instruct the future purposeful design of immunomodulatory biomaterials for cell therapy. A summary of immunological pathways initiated following cellular implantation, as well as current systemic immunomodulatory agents used, is provided. We then outline the potential of biomaterials to modulate these responses. The capacity of polymeric encapsulation to block some powerful rejection pathways is covered. We also highlight the role of cellular health and biocompatibility in mitigating immune responses. Finally, we review the use of bioactive materials to proactively modulate local immune responses, focusing on key concepts of anti-inflammatory, suppressive, and tolerogenic agents.
Collapse
Affiliation(s)
- Magdalena M Samojlik
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Cherie L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; University of Florida Diabetes Institute, Gainesville, FL, USA; Graduate Program in Biomedical Sciences, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
23
|
Xiang D, Liu Y, Zhou E, Wang Y. Advances in the applications of polymer biomaterials for in vitro follicle culture. Biomed Pharmacother 2021; 140:111422. [PMID: 34098195 DOI: 10.1016/j.biopha.2021.111422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/05/2021] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
The ovarian reserve (OR) indicates ovarian function by representing the quantity and quality of ovarian follicles, and it gradually decreases with increasing age. With the prolongation of women's lives, the protection provided by estrogen is lost for decades in postmenopausal women, and the related cardiovascular and cerebrovascular diseases, osteoporosis, and decreased immunity are the main risk factors affecting women's quality of life and longevity. Pharmacologic hormone replacement therapy (PHRT) has been controversial, and the construction of artificial ovary (AO) has attracted increasing attention. The most critical step of AO generation is the establishment of an in vitro culture (IVC) system to support the development of isolated follicles. This article mainly compares the advantages and disadvantages of different polymer biomaterials for use in follicle IVC, provides theoretical support for the development and construction of the follicle IVC system using natural biological materials, and provides a theoretical basis for establishing mature AO technology.
Collapse
Affiliation(s)
- Du Xiang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Yang Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Encheng Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China.
| |
Collapse
|
24
|
Kharbikar BN, Chendke GS, Desai TA. Modulating the foreign body response of implants for diabetes treatment. Adv Drug Deliv Rev 2021; 174:87-113. [PMID: 33484736 PMCID: PMC8217111 DOI: 10.1016/j.addr.2021.01.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/30/2020] [Accepted: 01/10/2021] [Indexed: 02/06/2023]
Abstract
Diabetes Mellitus is a group of diseases characterized by high blood glucose levels due to patients' inability to produce sufficient insulin. Current interventions often require implants that can detect and correct high blood glucose levels with minimal patient intervention. However, these implantable technologies have not reached their full potential in vivo due to the foreign body response and subsequent development of fibrosis. Therefore, for long-term function of implants, modulating the initial immune response is crucial in preventing the activation and progression of the immune cascade. This review discusses the different molecular mechanisms and cellular interactions involved in the activation and progression of foreign body response (FBR) and fibrosis, specifically for implants used in diabetes. We also highlight the various strategies and techniques that have been used for immunomodulation and prevention of fibrosis. We investigate how these general strategies have been applied to implants used for the treatment of diabetes, offering insights on how these devices can be further modified to circumvent FBR and fibrosis.
Collapse
Affiliation(s)
- Bhushan N Kharbikar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gauree S Chendke
- University of California Berkeley - University of California San Francisco Graduate Program in Bioengineering, San Francisco, CA 94143, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; University of California Berkeley - University of California San Francisco Graduate Program in Bioengineering, San Francisco, CA 94143, USA; Department of Bioengineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
25
|
Lopez-Mendez TB, Santos-Vizcaino E, Pedraz JL, Orive G, Hernandez RM. Cell microencapsulation technologies for sustained drug delivery: Latest advances in efficacy and biosafety. J Control Release 2021; 335:619-636. [PMID: 34116135 DOI: 10.1016/j.jconrel.2021.06.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 06/04/2021] [Accepted: 06/06/2021] [Indexed: 10/21/2022]
Abstract
The development of cell microencapsulation systems began several decades ago. However, today few systems have been tested in clinical trials. For this reason, in the last years, researchers have directed efforts towards trying to solve some of the key aspects that still limit efficacy and biosafety, the two major criteria that must be satisfied to reach the clinical practice. Regarding the efficacy, which is closely related to biocompatibility, substantial improvements have been made, such as the purification or chemical modification of the alginates that normally form the microspheres. Each of the components that make up the microcapsules has been carefully selected to avoid toxicities that can damage the encapsulated cells or generate an immune response leading to pericapsular fibrosis. As for the biosafety, researchers have developed biological circuits capable of actively responding to the needs of the patients to precisely and accurately release the demanded drug dose. Furthermore, the structure of the devices has been subject of study to adequately protect the encapsulated cells and prevent their spread in the body. The objective of this review is to describe the latest advances made by scientist to improve the efficacy and biosafety of cell microencapsulation systems for sustained drug delivery, also highlighting those points that still need to be optimized.
Collapse
Affiliation(s)
- Tania B Lopez-Mendez
- NanoBioCel Research Group, 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), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, 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), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Jose Luis Pedraz
- NanoBioCel Research Group, 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), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Gorka Orive
- NanoBioCel Research Group, 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), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), BTI Biotechnology Institute, Vitoria-Gasteiz, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, 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), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| |
Collapse
|
26
|
Jeon SI, Jeong JH, Kim JE, Haque MR, Kim J, Byun Y, Ahn CH. Synthesis of PEG-dendron for surface modification of pancreatic islets and suppression of the immune response. J Mater Chem B 2021; 9:2631-2640. [PMID: 33683280 DOI: 10.1039/d1tb00069a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Islet cell transplantation has been an effective method for the treatment of type 1 diabetes. The transplanted islets release insulin in response to changes in blood glucose levels. The clinical application of islet transplantation, however, has been hindered because of some critical problems including immune responses to grafted islets and side effects caused by overdosed immunosuppressive drugs. Herein, surface modification technology using poly(ethylene glycol) (PEG)-dendron was proposed to safeguard islets from the host immune system. PEG-dendron was synthesized by a divergent polymerization method and utilized to cover the islet antigen surface. Successful conjugation of PEG-dendron on the islet surface was achieved without affecting islet morphology, viability, and functionality at a concentration of 1.00%. Surface modification using PEG-dendron effectively prevented protein absorption and immune activation. Foremost, it improved the survival rate of islet grafts in vivo when combined with a low dose of immunosuppressive drugs. In conclusion, PEG-dendron is a potential candidate for the surface modification of pancreatic islets to mitigate immune responses after transplantation.
Collapse
Affiliation(s)
- Seong Ik Jeon
- Research Institute of Advanced Materials (RIAM), Department of Materials Science and Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | | | | | | | | | | | | |
Collapse
|
27
|
Fuchs S, Ernst AU, Wang LH, Shariati K, Wang X, Liu Q, Ma M. Hydrogels in Emerging Technologies for Type 1 Diabetes. Chem Rev 2020; 121:11458-11526. [DOI: 10.1021/acs.chemrev.0c01062] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Stephanie Fuchs
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Alexander U. Ernst
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Long-Hai Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Kaavian Shariati
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Xi Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Qingsheng Liu
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Minglin Ma
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
28
|
Mohammadi MR, Dehkordi-Vakil F, Ricks-Oddie J, Mansfield R, Kashimiri H, Daniels M, Zhao W, Lakey JR. Preferences of Type 1 Diabetic Patients on Devices for Islet Transplantation. Cell Transplant 2020; 29:963689720952343. [PMID: 33023311 PMCID: PMC7784499 DOI: 10.1177/0963689720952343] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transplantation of pancreatic islets within a biomaterial device is currently
under investigation in clinical trials for the treatment of patients with type 1
diabetes (T1D). Patients’ preferences on such implants could guide the designs
of next-generation implantable devices; however, such information is not
currently available. We surveyed the preferences of 482 patients with T1D on the
size, shape, visibility, and transplantation site of islet containing implants.
More than 83% of participants were willing to receive autologous stem cells, and
there was no significant association between implant fabricated by one’s own
stem cell with gender (χ2 (1, n = 468) = 0.28; P = 0.6) or
with age (χ2 (4, n = 468) = 2.92; P = 0.6).
Preferred location for islet transplantation within devices was under the skin
(52.7%). 48.3% preferred microscopic disks, and 32.3% preferred a thin device
(like a credit card). Moreover, 58.4% preferred the implant to be as small as
possible, 25.4% did not care about visibility, and 16.2% preferred their
implants not to be visible. Among female participants, 81% cared about the
implant visibility, whereas this number was 64% for male respondents
(χ2 test (1, n = 468) = 16.34; P <
0.0001). 22% of those younger than 50 years of age and 30% of those older than
50 did not care about the visibility of implant (χ2 test (4, n = 468) = 23.69; P <
0.0001). These results suggest that subcutaneous sites and micron-sized devices
are preferred choices among patients with T1D who participated in our
survey.
Collapse
Affiliation(s)
- M Rezaa Mohammadi
- Department of Materials Science and Engineering, 8788University of California, Irvine, CA, USA.,Sue and Bill Gross Stem Cell Research Center, 8788University of California, Irvine, CA, USA
| | - Farideh Dehkordi-Vakil
- Center for Statistical Consulting, Department of Statistics, 8788University of California, Irvine, CA, USA
| | - Joni Ricks-Oddie
- Center for Statistical Consulting, Department of Statistics, 8788University of California, Irvine, CA, USA
| | - Robert Mansfield
- 369679Juvenile Diabetes Research Foundation Orange County Chapter, Irvine, CA, USA
| | | | - Mark Daniels
- CHOC Children's Endocrine & Diabetes Center, Orange, CA, USA
| | - Weian Zhao
- Sue and Bill Gross Stem Cell Research Center, 8788University of California, Irvine, CA, USA.,Department of Pharmaceutical Sciences, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, 8788University of California, Irvine, Irvine, CA, USA.,Department of Biomedical Engineering, 8788University of California, Irvine, Irvine, CA, USA.,Department of Biological Chemistry, 8788University of California, Irvine, Irvine, CA, USA
| | - Jonathan Rt Lakey
- Sue and Bill Gross Stem Cell Research Center, 8788University of California, Irvine, CA, USA.,Department of Surgery and Biomedical Engineering, 8788University of California Irvine, Orange, CA, USA
| |
Collapse
|
29
|
Transplant Islets Into the Pinna of the Ear: A Mouse Islet Transplant Model. Transplant Proc 2020; 53:450-456. [PMID: 32928554 DOI: 10.1016/j.transproceed.2020.06.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/29/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Islets transplanted under the ear skin would allow easy observation of the graft response and survival in vivo. This research was designed to establish an efficient mouse islet transplant model to probe the dynamic cellular interplay in vivo. METHODS Green fluorescent protein transgenic mice and BALB/c mice were used as donors and recipients. All recipients were divided into 6 groups of 6 mice each. First, we treated the transplant recipients, including diabetes induction, autologous epididymal fat pad, and MATRIGEL transplant to the ears. Then, 1. we transplanted isolated islets to the ear/ear with fat/ear with MATRIGEL; and 2. transplanted islets with collagen + basic fibroblast growth factor or islets with collagen + vascular endothelial growth factor. Mice in the control group received a sham transplantation with phosphate buffer saline. All recipients were then observed for 30 days with blood glucose (BG) monitoring. Finally, ears were removed with graft on day 28 for histologic examination. RESULTS It was suggested that transplant of islets alone could not correct hyperglycemia. Fat, MATRIGEL, collagen, and growth factors have the similar function to form a microenvironment conducive to islet survival. The effect of islet transplantation for correcting hyperglycemia of the fat modification group was better than other groups (P < .05). BG could be normalized, and living islets were detected by anti-insulin immunohistochemistry. CONCLUSIONS Transplant islets into the ear with transplanted autologous fat is the optimal way which can be used to analyze the allograft response in vivo and track cell population and migration using labels by confocal microscopy.
Collapse
|
30
|
White Adipose Tissue as a Site for Islet Transplantation. TRANSPLANTOLOGY 2020. [DOI: 10.3390/transplantology1010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Although islet transplantation is recognized as a useful cellular replacement therapy for severe diabetes, surgeons face difficulties in islet engraftment. The transplant site is a pivotal factor that influences the engraftment. Although the liver is the current representative site for clinical islet transplantation, it is not the best site because of limitations in immunity, inflammation, and hypoxia. White adipose tissue, including omentum, is recognized as a useful candidate site for islet transplantation. Its effectiveness has been evaluated in not only various basic and translational studies using small and large animals but also in some recent clinical trials. In this review, we attempt to shed light on the characteristics and usefulness of white adipose tissue as a transplant site for islets.
Collapse
|
31
|
White Adipose Tissue as a Site for Islet Transplantation. TRANSPLANTOLOGY 2020. [DOI: 10.3390/transplantology1020006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although islet transplantation is recognized as a useful cellular replacement therapy for severe diabetes, surgeons face difficulties in islet engraftment. The transplant site is a pivotal factor that influences the engraftment. Although the liver is the current representative site for clinical islet transplantation, it is not the best site because of limitations in immunity, inflammation, and hypoxia. White adipose tissue, including omentum, is recognized as a useful candidate site for islet transplantation. Its effectiveness has been evaluated in not only various basic and translational studies using small and large animals but also in some recent clinical trials. In this review, we attempt to shed light on the characteristics and usefulness of white adipose tissue as a transplant site for islets.
Collapse
|
32
|
Krol S, Baronti W, Marchetti P. Nanoencapsulated human pancreatic islets for β-cell replacement in Type 1 diabetes. Nanomedicine (Lond) 2020; 15:1735-1738. [PMID: 32669019 DOI: 10.2217/nnm-2020-0166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Silke Krol
- Laboratory for Personalized Medicine, IRCCS Ospedale Specializzato in Gastroenterologia 'Saverio de Bellis', Castellana Grotte (BA), Italy
| | - Walter Baronti
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
33
|
Facklam AL, Volpatti LR, Anderson DG. Biomaterials for Personalized Cell Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1902005. [PMID: 31495970 DOI: 10.1002/adma.201902005] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/26/2019] [Indexed: 05/13/2023]
Abstract
Cell therapy has already had an important impact on healthcare and provided new treatments for previously intractable diseases. Notable examples include mesenchymal stem cells for tissue regeneration, islet transplantation for diabetes treatment, and T cell delivery for cancer immunotherapy. Biomaterials have the potential to extend the therapeutic impact of cell therapies by serving as carriers that provide 3D organization and support cell viability and function. With the growing emphasis on personalized medicine, cell therapies hold great potential for their ability to sense and respond to the biology of an individual patient. These therapies can be further personalized through the use of patient-specific cells or with precision biomaterials to guide cellular activity in response to the needs of each patient. Here, the role of biomaterials for applications in tissue regeneration, therapeutic protein delivery, and cancer immunotherapy is reviewed, with a focus on progress in engineering material properties and functionalities for personalized cell therapies.
Collapse
Affiliation(s)
- Amanda L Facklam
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Lisa R Volpatti
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Daniel G Anderson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
34
|
Sibilla S, Manenti S, Cazzato T, Colombo F, Tomei AA, Redaelli A, Manzoli V, Consolo F. Smoothed Particle Hydrodynamics multiphase modelling of an experimental microfluidic device for conformal coating of pancreatic islets. Med Eng Phys 2020; 77:19-30. [PMID: 32008936 DOI: 10.1016/j.medengphy.2020.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 12/13/2019] [Accepted: 01/05/2020] [Indexed: 11/29/2022]
Abstract
The paper discusses a Smoothed Particle Hydrodynamics (SPH) model for the analysis of the multiphase flow occurring in an experimental microfluidic device for conformal coating of pancreatic islets with a biocompatible and permeable polymer. The proposed numerical model, based on a weakly-compressible SPH approach, accurately mimics the encapsulation process while assuring phase conservation, thus overcoming potential limitations of grid-based models. The proposed SPH model is a triphasic multi-phase model that allows one: (i) to reproduce the physics of islet conformal coating, including the effects of surface tension at the interface of the involved fluids and of the islet diameter; and (ii) to evaluate how modulation of process parameters influences the fluid dynamics within the microfluidic device and the resulting coating characteristics. This model can represent a valuable, time- and cost-effective tool for the definition of optimized encapsulation conditions through in silico screening of novel combinations of conformal coating parameters, including polymeric coating blends, size range of insulin-secreting cell clusters, utilized chemical reagents, device geometry and scale.
Collapse
Affiliation(s)
- Stefano Sibilla
- Dipartimento di Ingegneria Civile e Architettura, Università di Pavia, via Ferrata 3, 27100 Pavia, Italy.
| | - Sauro Manenti
- Dipartimento di Ingegneria Civile e Architettura, Università di Pavia, via Ferrata 3, 27100 Pavia, Italy
| | - Tommaso Cazzato
- Dipartimento di Elettronica, Informazione e Bioingegneria Politecnico di Milano, via Ponzio 34/5, 20133 Milano, Italy
| | - Federica Colombo
- Dipartimento di Elettronica, Informazione e Bioingegneria Politecnico di Milano, via Ponzio 34/5, 20133 Milano, Italy
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136-1011, USA; Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, McArthur Engineering Building, Coral Gables, FL 33146, USA
| | - Alberto Redaelli
- Dipartimento di Elettronica, Informazione e Bioingegneria Politecnico di Milano, via Ponzio 34/5, 20133 Milano, Italy
| | - Vita Manzoli
- Dipartimento di Elettronica, Informazione e Bioingegneria Politecnico di Milano, via Ponzio 34/5, 20133 Milano, Italy; Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136-1011, USA
| | - Filippo Consolo
- Dipartimento di Elettronica, Informazione e Bioingegneria Politecnico di Milano, via Ponzio 34/5, 20133 Milano, Italy; Università Vita Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| |
Collapse
|
35
|
Stock AA, Manzoli V, De Toni T, Abreu MM, Poh YC, Ye L, Roose A, Pagliuca FW, Thanos C, Ricordi C, Tomei AA. Conformal Coating of Stem Cell-Derived Islets for β Cell Replacement in Type 1 Diabetes. Stem Cell Reports 2020; 14:91-104. [PMID: 31839542 PMCID: PMC6962554 DOI: 10.1016/j.stemcr.2019.11.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 11/08/2019] [Accepted: 11/15/2019] [Indexed: 12/29/2022] Open
Abstract
The scarcity of donors and need for immunosuppression limit pancreatic islet transplantation to a few patients with labile type 1 diabetes. Transplantation of encapsulated stem cell-derived islets (SC islets) might extend the applicability of islet transplantation to a larger cohort of patients. Transplantation of conformal-coated islets into a confined well-vascularized site allows long-term diabetes reversal in fully MHC-mismatched diabetic mice without immunosuppression. Here, we demonstrated that human SC islets reaggregated from cryopreserved cells display glucose-stimulated insulin secretion in vitro. Importantly, we showed that conformally coated SC islets displayed comparable in vitro function with unencapsulated SC islets, with conformal coating permitting physiological insulin secretion. Transplantation of SC islets into the gonadal fat pad of diabetic NOD-scid mice revealed that both unencapsulated and conformal-coated SC islets could reverse diabetes and maintain human-level euglycemia for more than 80 days. Overall, these results provide support for further evaluation of safety and efficacy of conformal-coated SC islets in larger species.
Collapse
Affiliation(s)
- Aaron A Stock
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Vita Manzoli
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA
| | - Teresa De Toni
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Maria M Abreu
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA
| | | | - Lillian Ye
- Semma Therapeutics, Inc., Cambridge, MA 02142, USA
| | - Adam Roose
- Semma Therapeutics, Inc., Cambridge, MA 02142, USA
| | | | - Chris Thanos
- Semma Therapeutics, Inc., Cambridge, MA 02142, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10(th) Avenue, Miami, FL 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
36
|
White AM, Shamul JG, Xu J, Stewart S, Bromberg JS, He X. Engineering Strategies to Improve Islet Transplantation for Type 1 Diabetes Therapy. ACS Biomater Sci Eng 2019; 6:2543-2562. [PMID: 33299929 DOI: 10.1021/acsbiomaterials.9b01406] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Type 1 diabetes is an autoimmune disease in which the immune system attacks insulin-producing beta cells of pancreatic islets. Type 1 diabetes can be treated with islet transplantation; however, patients must be administered immunosuppressants to prevent immune rejection of the transplanted islets if they are not autologous or not engineered with immune protection/isolation. To overcome biological barriers of islet transplantation, encapsulation strategies have been developed and robustly investigated. While islet encapsulation can prevent the need for immunosuppressants, these approaches have not shown much success in clinical trials due to a lack of long-term insulin production. Multiple engineering strategies have been used to improve encapsulation and post-transplantation islet survival. In addition, more efficient islet cryopreservation methods have been designed to facilitate the scaling-up of islet transplantation. Other islet sources have been identified including porcine islets and stem cell-derived islet-like aggregates. Overall, islet-laden capsule transplantation has greatly improved over the past 30 years and is moving towards becoming a clinically feasible treatment for type 1 diabetes.
Collapse
Affiliation(s)
- Alisa M White
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Jiangsheng Xu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Samantha Stewart
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201.,Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA, Baltimore, MD 21201, USA
| |
Collapse
|
37
|
Gonzalez Badillo FE, Zisi Tegou F, Abreu MM, Masina R, Sha D, Najjar M, Wright SH, Bayer AL, Korpos É, Pugliese A, Molano RD, Tomei AA. CCL21 Expression in β-Cells Induces Antigen-Expressing Stromal Cell Networks in the Pancreas and Prevents Autoimmune Diabetes in Mice. Diabetes 2019; 68:1990-2003. [PMID: 31371518 PMCID: PMC6754241 DOI: 10.2337/db19-0239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/21/2019] [Indexed: 12/31/2022]
Abstract
Tumors induce tolerance toward their antigens by producing the chemokine CCL21, leading to the formation of tertiary lymphoid organs (TLOs). Ins2-CCL21 transgenic, nonobese diabetic (NOD) mice express CCL21 in pancreatic β-cells and do not develop autoimmune diabetes. We investigated by which mechanisms CCL21 expression prevented diabetes. Ins2-CCL21 mice develop TLOs by 4 weeks of age, consisting of naive CD4+ T cells compartmentalized within networks of CD45-gp38+CD31- fibroblastic reticular cell (FRC)-like cells. Importantly, 12-week-old Ins2-CCL21 TLOs contained FRC-like cells with higher contractility, regulatory, and anti-inflammatory properties and enhanced expression of β-cell autoantigens compared with nontransgenic NOD TLOs found in inflamed islets. Consistently, transgenic mice harbored fewer autoreactive T cells and a higher proportion of regulatory T cells in the islets. Using adoptive transfer and islet transplantation models, we demonstrate that TLO formation in Ins2-CCL21 transgenic islets is critical for the regulation of autoimmunity, and although the effect is systemic, the induction is mediated locally likely by lymphocyte trafficking through TLOs. Overall, our findings suggest that CCL21 promotes TLOs that differ from inflammatory TLOs found in type 1 diabetic islets in that they resemble lymph nodes, contain FRC-like cells expressing β-cell autoantigens, and are able to induce systemic and antigen-specific tolerance leading to diabetes prevention.
Collapse
Affiliation(s)
- Freddy E Gonzalez Badillo
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Biomedical Engineering, University of Miami, Miami, FL
| | - Flavia Zisi Tegou
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Biomedical Engineering, University of Miami, Miami, FL
| | - Maria M Abreu
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Riccardo Masina
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Divya Sha
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Mejdi Najjar
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Shane H Wright
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Allison L Bayer
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
| | - Éva Korpos
- Institute of Physiological Chemistry and Pathobiochemistry and Cells in Motion, Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Alberto Pugliese
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - R Damaris Molano
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Biomedical Engineering, University of Miami, Miami, FL
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
38
|
Pathak S, Pham TT, Jeong JH, Byun Y. Immunoisolation of pancreatic islets via thin-layer surface modification. J Control Release 2019; 305:176-193. [DOI: 10.1016/j.jconrel.2019.04.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/15/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
|
39
|
Hu S, de Vos P. Polymeric Approaches to Reduce Tissue Responses Against Devices Applied for Islet-Cell Encapsulation. Front Bioeng Biotechnol 2019; 7:134. [PMID: 31214587 PMCID: PMC6558039 DOI: 10.3389/fbioe.2019.00134] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022] Open
Abstract
Immunoisolation of pancreatic islets is a technology in which islets are encapsulated in semipermeable but immunoprotective polymeric membranes. The technology allows for successful transplantation of insulin-producing cells in the absence of immunosuppression. Different approaches of immunoisolation are currently under development. These approaches involve intravascular devices that are connected to the bloodstream and extravascular devices that can be distinguished in micro- and macrocapsules and are usually implanted in the peritoneal cavity or under the skin. The technology has been subject of intense fundamental research in the past decade. It has co-evolved with novel replenishable cell sources for cure of diseases such as Type 1 Diabetes Mellitus that need to be protected for the host immune system. Although the devices have shown significant success in animal models and even in human safety studies most technologies still suffer from undesired tissue responses in the host. Here we review the past and current approaches to modulate and reduce tissue responses against extravascular cell-containing micro- and macrocapsules with a focus on rational choices for polymer (combinations). Choices for polymers but also choices for crosslinking agents that induce more stable and biocompatible capsules are discussed. Combining beneficial properties of molecules in diblock polymers or application of these molecules or other anti-biofouling molecules have been reviewed. Emerging are also the principles of polymer brushes that prevent protein and cell-adhesion. Recently also immunomodulating biomaterials that bind to specific immune receptors have entered the field. Several natural and synthetic polymers and even combinations of these polymers have demonstrated significant improvement in outcomes of encapsulated grafts. Adequate polymeric surface properties have been shown to be essential but how the surface should be composed to avoid host responses remains to be identified. Current insight is that optimal biocompatible devices can be created which raises optimism that immunoisolating devices can be created that allows for long term survival of encapsulated replenishable insulin-producing cell sources for treatment of Type 1 Diabetes Mellitus.
Collapse
Affiliation(s)
- Shuixan Hu
- Division of Medical Biology, Department of Pathology and Medical Biology, Immunoendocrinology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | | |
Collapse
|
40
|
|
41
|
Paving the way for successful islet encapsulation. Drug Discov Today 2019; 24:737-748. [PMID: 30738185 DOI: 10.1016/j.drudis.2019.01.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/13/2018] [Accepted: 01/29/2019] [Indexed: 01/02/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is a disorder that decimates pancreatic β-cells which produce insulin. Direct pancreatic islet transplantation cannot serve as a widespread therapeutic modality owing to the need for lifelong immunosuppression and donor shortage. Therefore, several encapsulation techniques have been developed to enclose the islets in semipermeable vehicles that will allow oxygen and nutrient input as well as insulin, other metabolites and waste output, while accomplishing immunoisolation. Although encapsulation technology continues to face significant obstacles, recent advances in material science, stem cell biology and immunology potentially serve as pathways to success. This review summarizes the accomplishments of the past 5 years.
Collapse
|
42
|
Ernst AU, Bowers DT, Wang LH, Shariati K, Plesser MD, Brown NK, Mehrabyan T, Ma M. Nanotechnology in cell replacement therapies for type 1 diabetes. Adv Drug Deliv Rev 2019; 139:116-138. [PMID: 30716349 PMCID: PMC6677642 DOI: 10.1016/j.addr.2019.01.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/17/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022]
Abstract
Islet transplantation is a promising long-term, compliance-free, complication-preventing treatment for type 1 diabetes. However, islet transplantation is currently limited to a narrow set of patients due to the shortage of donor islets and side effects from immunosuppression. Encapsulating cells in an immunoisolating membrane can allow for their transplantation without the need for immunosuppression. Alternatively, "open" systems may improve islet health and function by allowing vascular ingrowth at clinically attractive sites. Many processes that enable graft success in both approaches occur at the nanoscale level-in this review we thus consider nanotechnology in cell replacement therapies for type 1 diabetes. A variety of biomaterial-based strategies at the nanometer range have emerged to promote immune-isolation or modulation, proangiogenic, or insulinotropic effects. Additionally, coating islets with nano-thin polymer films has burgeoned as an islet protection modality. Materials approaches that utilize nanoscale features manipulate biology at the molecular scale, offering unique solutions to the enduring challenges of islet transplantation.
Collapse
Affiliation(s)
- Alexander U Ernst
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Daniel T Bowers
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Long-Hai Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Kaavian Shariati
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Mitchell D Plesser
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Natalie K Brown
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Tigran Mehrabyan
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
43
|
Farina M, Alexander JF, Thekkedath U, Ferrari M, Grattoni A. Cell encapsulation: Overcoming barriers in cell transplantation in diabetes and beyond. Adv Drug Deliv Rev 2019; 139:92-115. [PMID: 29719210 DOI: 10.1016/j.addr.2018.04.018] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/19/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023]
Abstract
Cell-based therapy is emerging as a promising strategy for treating a wide range of human diseases, such as diabetes, blood disorders, acute liver failure, spinal cord injury, and several types of cancer. Pancreatic islets, blood cells, hepatocytes, and stem cells are among the many cell types currently used for this strategy. The encapsulation of these "therapeutic" cells is under intense investigation to not only prevent immune rejection but also provide a controlled and supportive environment so they can function effectively. Some of the advanced encapsulation systems provide active agents to the cells and enable a complete retrieval of the graft in the case of an adverse body reaction. Here, we review various encapsulation strategies developed in academic and industrial settings, including the state-of-the-art technologies in advanced preclinical phases as well as those undergoing clinical trials, and assess their advantages and challenges. We also emphasize the importance of stimulus-responsive encapsulated cell systems that provide a "smart and live" therapeutic delivery to overcome barriers in cell transplantation as well as their use in patients.
Collapse
|
44
|
Perez-Basterrechea M, Esteban MM, Vega JA, Obaya AJ. Tissue-engineering approaches in pancreatic islet transplantation. Biotechnol Bioeng 2018; 115:3009-3029. [PMID: 30144310 DOI: 10.1002/bit.26821] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 12/15/2022]
Abstract
Pancreatic islet transplantation is a promising alternative to whole-pancreas transplantation as a treatment of type 1 diabetes mellitus. This technique has been extensively developed during the past few years, with the main purpose of minimizing the complications arising from the standard protocols used in organ transplantation. By using a variety of strategies used in tissue engineering and regenerative medicine, pancreatic islets have been successfully introduced in host patients with different outcomes in terms of islet survival and functionality, as well as the desired normoglycemic control. Here, we describe and discuss those strategies to transplant islets together with different scaffolds, in combination with various cell types and diffusible factors, and always with the aim of reducing host immune response and achieving islet survival, regardless of the site of transplantation.
Collapse
Affiliation(s)
- Marcos Perez-Basterrechea
- Unidad de Terapia Celular y Medicina Regenerativa, Servicio de Hematología y Hemoterapia, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain.,Plataforma de Terapias Avanzadas, Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Manuel M Esteban
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| | - Jose A Vega
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain.,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Alvaro J Obaya
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
45
|
Youngblood RL, Truong NF, Segura T, Shea LD. It's All in the Delivery: Designing Hydrogels for Cell and Non-viral Gene Therapies. Mol Ther 2018; 26:2087-2106. [PMID: 30107997 PMCID: PMC6127639 DOI: 10.1016/j.ymthe.2018.07.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 01/08/2023] Open
Abstract
Hydrogels provide a regenerative medicine platform with their ability to create an environment that supports transplanted or endogenous infiltrating cells and enables these cells to restore or replace the function of tissues lost to disease or trauma. Furthermore, these systems have been employed as delivery vehicles for therapeutic genes, which can direct and/or enhance the function of the transplanted or endogenous cells. Herein, we review recent advances in the development of hydrogels for cell and non-viral gene delivery through understanding the design parameters, including both physical and biological components, on promoting transgene expression, cell engraftment, and ultimately cell function. Furthermore, this review identifies emerging opportunities for combining cell and gene delivery approaches to overcome challenges to the field.
Collapse
Affiliation(s)
- Richard L Youngblood
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Norman F Truong
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tatiana Segura
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
46
|
Pellegrini S, Piemonti L, Sordi V. Pluripotent stem cell replacement approaches to treat type 1 diabetes. Curr Opin Pharmacol 2018; 43:20-26. [PMID: 30071348 DOI: 10.1016/j.coph.2018.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/06/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022]
Abstract
Stem cells represent a potential candidate for β cell replacement in type 1 diabetes. Pluripotent stem cells are able to differentiate in vitro into functional insulin producing cells, that can restore normoglycemia in diabetic mice. Clinical trials with embryonic stem cell-derived pancreatic progenitors are ongoing. Besides, induced pluripotent stem cells offer the chance of personalized cell therapy. So far, transition to the clinic still needs to face critical issues, such as immunogenicity and safety of stem cell derived β cells. To this purpose, new strategies for immunoprotection, including micro and macro-encapsulation, but also gene editing approaches, are being explored.
Collapse
Affiliation(s)
- Silvia Pellegrini
- Diabetes Research Institute - IRCCS San Raffaele Scientific Institute, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute - IRCCS San Raffaele Scientific Institute, Italy; Vita-Salute San Raffaele University, Italy
| | - Valeria Sordi
- Diabetes Research Institute - IRCCS San Raffaele Scientific Institute, Italy.
| |
Collapse
|
47
|
Lee JK, Choi IS, Oh TI, Lee E. Cell-Surface Engineering for Advanced Cell Therapy. Chemistry 2018; 24:15725-15743. [PMID: 29791047 DOI: 10.1002/chem.201801710] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/22/2018] [Indexed: 12/16/2022]
Abstract
Stem cells opened great opportunity to overcome diseases that conventional therapy had only limited success. Use of scaffolds made from biomaterials not only helps handling of stem cells for delivery or transplantation but also supports enhanced cell survival. Likewise, cell encapsulation can provide stability for living animal cells even in a state of separateness. Although various chemical reactions were tried to encapsulate stolid microbial cells such as yeasts, a culture environment for the growth of animal cells allows only highly biocompatible reactions. Therefore, the animal cells were mostly encapsulated in hydrogels, which resulted in enhanced cell survival. Interestingly, major findings of chemistry on biological interfaces demonstrate that cell encapsulation in hydrogels have a further a competence for modulating cell characteristics that can go beyond just enhancing the cell survival. In this review, we present a comprehensive overview on the chemical reactions applied to hydrogel-based cell encapsulation and their effects on the characteristics and behavior of living animal cells.
Collapse
Affiliation(s)
- Jungkyu K Lee
- Department of Chemistry and Green-Nano Materials Research Center, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, Korea
| | - Insung S Choi
- Department of Chemistry and Center for Cell-Encapsulation Research, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Korea
| | - Tong In Oh
- Department of Biomedical Engineering, Kyung Hee University, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - EunAh Lee
- Impedance Imaging Research Center (IIRC), Kyung Hee University, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| |
Collapse
|
48
|
Duffy C, Prugue C, Glew R, Smith T, Howell C, Choi G, Cook AD. Feasibility of Induced Pluripotent Stem Cell Therapies for Treatment of Type 1 Diabetes. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:482-492. [PMID: 29947303 DOI: 10.1089/ten.teb.2018.0124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
IMPACT STATEMENT This review of iPSCs to treat T1D provides a current assessment of the challenges and potential for this proposed new therapy.
Collapse
Affiliation(s)
- Caden Duffy
- Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | - Cesar Prugue
- Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | - Rachel Glew
- Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | - Taryn Smith
- Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | - Calvin Howell
- Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | - Gina Choi
- Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | - Alonzo D Cook
- Department of Chemical Engineering, Brigham Young University , Provo, Utah
| |
Collapse
|
49
|
Abstract
The pancreas is made from two distinct components: the exocrine pancreas, a reservoir of digestive enzymes, and the endocrine islets, the source of the vital metabolic hormone insulin. Human islets possess limited regenerative ability; loss of islet β-cells in diseases such as type 1 diabetes requires therapeutic intervention. The leading strategy for restoration of β-cell mass is through the generation and transplantation of new β-cells derived from human pluripotent stem cells. Other approaches include stimulating endogenous β-cell proliferation, reprogramming non-β-cells to β-like cells, and harvesting islets from genetically engineered animals. Together these approaches form a rich pipeline of therapeutic development for pancreatic regeneration.
Collapse
|
50
|
Proceedings of the signature series event of the international society for cellular therapy: "Advancements in cellular therapies and regenerative medicine in digestive diseases," London, United Kingdom, May 3, 2017. Cytotherapy 2018; 20:461-476. [PMID: 29398624 DOI: 10.1016/j.jcyt.2017.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 12/01/2017] [Indexed: 12/18/2022]
Abstract
A summary of the First Signature Series Event, "Advancements in Cellular Therapies and Regenerative Medicine for Digestive Diseases," held on May 3, 2017, in London, United Kingdom, is presented. Twelve speakers from three continents covered major topics in the areas of cellular therapy and regenerative medicine applied to liver and gastrointestinal medicine as well as to diabetes mellitus. Highlights from their presentations, together with an overview of the global impact of digestive diseases and a proposal for a shared online collection and data-monitoring platform tool, are included in this proceedings. Although growing evidence demonstrate the feasibility and safety of exploiting cell-based technologies for the treatment of digestive diseases, regulatory and methodological obstacles will need to be overcome before the successful implementation in the clinic of these novel attractive therapeutic strategies.
Collapse
|