1
|
Tran HT, Rodprasert W, Padeta I, Oontawee S, Purbantoro SD, Thongsit A, Siriarchavatana P, Srisuwatanasagul S, Egusa H, Osathanon T, Sawangmake C. Establishment of subcutaneous transplantation platform for delivering induced pluripotent stem cell-derived insulin-producing cells. PLoS One 2025; 20:e0318204. [PMID: 39883721 PMCID: PMC11781742 DOI: 10.1371/journal.pone.0318204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025] Open
Abstract
Potential trend of regenerative treatment for type I diabetes has been introduced for more than a decade. However, the technologies regarding insulin-producing cell (IPC) production and transplantation are still being developed. Here, we propose the potential IPC production protocol employing mouse gingival fibroblast-derived induced pluripotent stem cells (mGF-iPSCs) as a resource and the pre-clinical approved subcutaneous IPC transplantation platform for further clinical confirmation study. With a multi-step induction protocol, the functional and matured IPCs were generated by 13 days with a long-term survival capability. Further double encapsulation of mGF-iPSC-derived IPCs (mGF-iPSC-IPCs) could preserve the insulin secretion capacity and the transplantation potential of the generated IPCs. To address the potential on IPC transplantation, a 2-step subcutaneous transplantation procedure was established, comprising 1) vascularized subcutaneous pocket formation and 2) encapsulated IPC bead transplantation. The in vivo testing confirmed the safety and efficiency of the platform along with less inflammatory response which may help minimize tissue reaction and graft rejection. Further preliminary in vivo testing on subcutaneous IPC-bead transplantation in an induced type I diabetic mouse model showed beneficial trends on blood glucose control and survival rate sustainability of diabetic mice. Taken together, an established mGF-iPSC-IPC generation protocol in this study will be the potential backbone for developing the iPSC-derived IPC production employing human and animal cell resources. As well as the potential further development of IPC transplantation platform for diabetes treatment in human and veterinary practices using an established subcutaneous encapsulated IPC-bead transplantation platform presented in this study.
Collapse
Affiliation(s)
- Hong Thuan Tran
- Second Century Fund (C2F) Chulalongkorn University for Doctoral Scholarship, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, The International Graduate Program of Veterinary Science and Technology (VST), Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Watchareewan Rodprasert
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Irma Padeta
- Second Century Fund (C2F) Chulalongkorn University for Doctoral Scholarship, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, The International Graduate Program of Veterinary Science and Technology (VST), Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Saranyou Oontawee
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Steven dwi Purbantoro
- Second Century Fund (C2F) Chulalongkorn University for Doctoral Scholarship, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, The International Graduate Program of Veterinary Science and Technology (VST), Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Anatcha Thongsit
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
| | - Parkpoom Siriarchavatana
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Medicine, Western University, Kanchanaburi, Thailand
| | - Sayamon Srisuwatanasagul
- Faculty of Veterinary Science, Department of Anatomy, Chulalongkorn University, Bangkok, Thailand
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Thanaphum Osathanon
- Faculty of Dentistry, Dental Stem Cell Biology Research Unit and Department of Anatomy, Chulalongkorn University, Bangkok, Thailand
- Faculty of Dentistry, Center of Excellence in Regenerative Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chenphop Sawangmake
- Faculty of Veterinary Science, Veterinary Clinical Stem Cell and Bioengineering Research Unit, Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Chulalongkorn University, Bangkok, Thailand
- Faculty of Veterinary Science, Department of Pharmacology, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
2
|
Zhu Y, Guo S, Ravichandran D, Ramanathan A, Sobczak MT, Sacco AF, Patil D, Thummalapalli SV, Pulido TV, Lancaster JN, Yi J, Cornella JL, Lott DG, Chen X, Mei X, Zhang YS, Wang L, Wang X, Zhao Y, Hassan MK, Chambers LB, Theobald TG, Yang S, Liang L, Song K. 3D-Printed Polymeric Biomaterials for Health Applications. Adv Healthc Mater 2025; 14:e2402571. [PMID: 39498750 PMCID: PMC11694096 DOI: 10.1002/adhm.202402571] [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: 07/12/2024] [Revised: 09/17/2024] [Indexed: 11/07/2024]
Abstract
3D printing, also known as additive manufacturing, holds immense potential for rapid prototyping and customized production of functional health-related devices. With advancements in polymer chemistry and biomedical engineering, polymeric biomaterials have become integral to 3D-printed biomedical applications. However, there still exists a bottleneck in the compatibility of polymeric biomaterials with different 3D printing methods, as well as intrinsic challenges such as limited printing resolution and rates. Therefore, this review aims to introduce the current state-of-the-art in 3D-printed functional polymeric health-related devices. It begins with an overview of the landscape of 3D printing techniques, followed by an examination of commonly used polymeric biomaterials. Subsequently, examples of 3D-printed biomedical devices are provided and classified into categories such as biosensors, bioactuators, soft robotics, energy storage systems, self-powered devices, and data science in bioplotting. The emphasis is on exploring the current capabilities of 3D printing in manufacturing polymeric biomaterials into desired geometries that facilitate device functionality and studying the reasons for material choice. Finally, an outlook with challenges and possible improvements in the near future is presented, projecting the contribution of general 3D printing and polymeric biomaterials in the field of healthcare.
Collapse
Affiliation(s)
- Yuxiang Zhu
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
| | - Shenghan Guo
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
| | - Dharneedar Ravichandran
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
| | - Arunachalam Ramanathan
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - M. Taylor Sobczak
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Alaina F. Sacco
- School of Chemical, Materials and Biomedical Engineering (CMBE), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Dhanush Patil
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
| | - Sri Vaishnavi Thummalapalli
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Tiffany V. Pulido
- Department of ImmunologyMayo Clinic Arizona13400 E Shea BlvdScottsdaleAZ85259USA
| | - Jessica N. Lancaster
- Department of ImmunologyMayo Clinic Arizona13400 E Shea BlvdScottsdaleAZ85259USA
| | - Johnny Yi
- Department of Medical and Surgical GynecologyMayo Clinic Arizona5777 E Mayo BlvdPhoenixAZ85054USA
| | - Jeffrey L. Cornella
- Department of Medical and Surgical GynecologyMayo Clinic Arizona5777 E Mayo BlvdPhoenixAZ85054USA
| | - David G. Lott
- Division of Laryngology, Department of OtolaryngologyMayo Clinic ArizonaPhoenixAZUSA
| | - Xiangfan Chen
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
| | - Xuan Mei
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolCambridgeMA02139USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolCambridgeMA02139USA
| | - Linbing Wang
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Xianqiao Wang
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Yiping Zhao
- Physics, Franklin College of Arts and SciencesUniversity of GeorgiaAthensGA30602USA
| | | | - Lindsay B. Chambers
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Taylor G. Theobald
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Sui Yang
- Materials Science and Engineering, School for Engineering of MatterTransport and Energy (SEMTE) at Arizona State UniversityTempeAZ85287USA
| | | | - Kenan Song
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| |
Collapse
|
3
|
Abdalla MMI. Advancing diabetes management: Exploring pancreatic beta-cell restoration's potential and challenges. World J Gastroenterol 2024; 30:4339-4353. [PMID: 39494103 PMCID: PMC11525866 DOI: 10.3748/wjg.v30.i40.4339] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/05/2024] [Accepted: 09/24/2024] [Indexed: 10/16/2024] Open
Abstract
Diabetes mellitus, characterized by chronic hyperglycemia due to insulin deficiency or resistance, poses a significant global health burden. Central to its pathogenesis is the dysfunction or loss of pancreatic beta cells, which are res-ponsible for insulin production. Recent advances in beta-cell regeneration research offer promising strategies for diabetes treatment, aiming to restore endogenous insulin production and achieve glycemic control. This review explores the physiological basis of beta-cell function, recent scientific advan-cements, and the challenges in translating these findings into clinical applications. It highlights key developments in stem cell therapy, gene editing technologies, and the identification of novel regenerative molecules. Despite the potential, the field faces hurdles such as ensuring the safety and long-term efficacy of regen-erative therapies, ethical concerns around stem cell use, and the complexity of beta-cell differentiation and integration. The review highlights the importance of interdisciplinary collaboration, increased funding, the need for patient-centered approaches and the integration of new treatments into comprehensive care strategies to overcome these challenges. Through continued research and collaboration, beta-cell regeneration holds the potential to revolutionize diabetes care, turning a chronic condition into a manageable or even curable disease.
Collapse
Affiliation(s)
- Mona Mohamed Ibrahim Abdalla
- Department of Human Biology, School of Medicine, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Crist SB, Azzag K, Kiley J, Coleman I, Magli A, Perlingeiro RCR. The adult environment promotes the transcriptional maturation of human iPSC-derived muscle grafts. NPJ Regen Med 2024; 9:16. [PMID: 38575647 PMCID: PMC10994941 DOI: 10.1038/s41536-024-00360-4] [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: 10/30/2023] [Accepted: 03/26/2024] [Indexed: 04/06/2024] Open
Abstract
Pluripotent stem cell (PSC)-based cell therapy is an attractive option for the treatment of multiple human disorders, including muscular dystrophies. While in vitro differentiating PSCs can generate large numbers of human lineage-specific tissue, multiple studies evidenced that these cell populations mostly display embryonic/fetal features. We previously demonstrated that transplantation of PSC-derived myogenic progenitors provides long-term engraftment and functional improvement in several dystrophic mouse models, but it remained unknown whether donor-derived myofibers mature to match adult tissue. Here, we transplanted iPAX7 myogenic progenitors into muscles of non-dystrophic and dystrophic mice and compared the transcriptional landscape of human grafts with respective in vitro-differentiated iPAX7 myotubes as well as human skeletal muscle biospecimens. Pairing bulk RNA sequencing with computational deconvolution of human reads, we were able to pinpoint key myogenic changes that occur during the in vitro-to-in vivo transition, confirm developmental maturity, and consequently evaluate their applicability for cell-based therapies.
Collapse
Affiliation(s)
- Sarah B Crist
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Karim Azzag
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - James Kiley
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Ilsa Coleman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Alessandro Magli
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
- Sanofi, Genomic Medicine Unit, 225 2nd Ave, Waltham, MA, 02451, USA.
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Kuo YC, Lin SY, De S, Rajesh R. Regeneration of Pancreatic Cells Using Optimized Nanoparticles and l-Glutamic Acid-Gelatin Scaffolds with Controlled Topography and Grafted Activin A/BMP4. ACS Biomater Sci Eng 2023; 9:6208-6224. [PMID: 37882705 DOI: 10.1021/acsbiomaterials.3c00791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Regeneration of insulin-producing cells (IPCs) from induced pluripotent stem cells (iPSCs) under controlled conditions has a lot of promise to emulate the pancreatic mechanism in vivo as a foundation of cell-based diabetic therapy. l-Glutamic acid-gelatin scaffolds with orderly pore sizes of 160 and 200 μm were grafted with activin A and bone morphogenic proteins 4 (BMP4) to differentiate iPSCs into definitive endoderm (DE) cells, which were then guided with fibroblast growth factor 7 (FGF7)-grafted retinoic acid (RA)-loaded solid lipid nanoparticles (FR-SLNs) to harvest IPCs. Response surface methodology was adopted to optimize the l-glutamic acid-to-gelatin ratio of scaffolds and to optimize surfactant concentration and lipid proportion in FR-SLNs. Experimental results of immunofluorescence, flow cytometry, and western blots revealed that activin A (100 ng/mL)-BMP4 (50 ng/mL)-l-glutamic acid (5%)-gelatin (95%) scaffolds provoked the largest number of SOX17-positive DE cells from iPSCs. Treatment with FGF7 (50 ng/mL)-RA (600 ng/mL)-SLNs elicited the highest number of PDX1-positive β-cells from differentiated DE cells. To imitate the natural pancreas, the scaffolds with controlled topography were appropriate for IPC production with sufficient insulin secretion. Hence, the current scheme using FR-SLNs and activin A-BMP4-l-glutamic acid-gelatin scaffolds in the two-stage differentiation of iPSCs can be promising for replacing impaired β-cells in diabetic management.
Collapse
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan 62102, ROC
- Advanced Institute of Manufacturing with High-tech Innovations, National Chung Cheng University, Chia-Yi, Taiwan 62102, ROC
| | - Sheng-Yuan Lin
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan 62102, ROC
| | - Sourav De
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan 62102, ROC
| | - Rajendiran Rajesh
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan 62102, ROC
| |
Collapse
|
6
|
Giuli L, Santopaolo F, Pallozzi M, Pellegrino A, Coppola G, Gasbarrini A, Ponziani FR. Cellular therapies in liver and pancreatic diseases. Dig Liver Dis 2023; 55:563-579. [PMID: 36543708 DOI: 10.1016/j.dld.2022.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/21/2022] [Accepted: 11/22/2022] [Indexed: 04/29/2023]
Abstract
Over the past two decades, developments in regenerative medicine in gastroenterology have been greatly enhanced by the application of stem cells, which can self-replicate and differentiate into any somatic cell. The discovery of induced pluripotent stem cells has opened remarkable perspectives on tissue regeneration, including their use as a bridge to transplantation or as supportive therapy in patients with organ failure. The improvements in DNA manipulation and gene editing strategies have also allowed to clarify the physiopathology and to correct the phenotype of several monogenic diseases, both in vivo and in vitro. Further progress has been made with the development of three-dimensional cultures, known as organoids, which have demonstrated morphological and functional complexity comparable to that of a miniature organ. Hence, owing to its protean applications and potential benefits, cell and organoid transplantation has become a hot topic for the management of gastrointestinal diseases. In this review, we describe current knowledge on cell therapies in hepatology and pancreatology, providing insight into their future applications in regenerative medicine.
Collapse
Affiliation(s)
- Lucia Giuli
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Santopaolo
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Pallozzi
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Pellegrino
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gaetano Coppola
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Francesca Romana Ponziani
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
7
|
Khazaei M, Khazaei F, Niromand E, Ghanbari E. Tissue engineering approaches and generation of insulin-producing cells to treat type 1 diabetes. J Drug Target 2023; 31:14-31. [PMID: 35896313 DOI: 10.1080/1061186x.2022.2107653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Tissue engineering (TE) has become a new effective solution to a variety of medical problems, including diabetes. Mesenchymal stem cells (MSCs), which have the ability to differentiate into endodermal and mesodermal cells, appear to be appropriate for this function. The purpose of this review was to evaluate the outcomes of various researches on the insulin-producing cells (IPCs) generation from MSCs with TE approaches to increase efficacy of type 1 diabetes treatments. The search was performed in PubMed/Medline, Scopus and Embase databases until 2021. Studies revealed that MSCs could also differentiate into IPCs under certain conditions. Therefore, a wide range of protocols have been used for this differentiation, but their effectiveness is very different. Scaffolds can provide a microenvironment that enhances the MSCs to IPCs differentiation, improves their metabolic activity and up-regulate pancreatic-specific transcription factors. They also preserve IPCs architecture and enhance insulin production as well as protect against cell death. This systematic review offers a framework for prospective research based on data. In vitro and in vivo evidence suggests that scaffold-based TE can improve the viability and function of IPCs.
Collapse
Affiliation(s)
- Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Khazaei
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Niromand
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Ghanbari
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
8
|
Zhou Z, Zhu X, Huang H, Xu Z, Jiang J, Chen B, Zhu H. Recent Progress of Research Regarding the Applications of Stem Cells for Treating Diabetes Mellitus. Stem Cells Dev 2022; 31:102-110. [PMID: 35072537 DOI: 10.1089/scd.2021.0083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
At present, the number of diabetes patients has exceeded 537 million worldwide and this number continues to increase. Stem cell therapy represents a new direction for the treatment of diabetes; the use of stem cells overcomes some shortcomings associated with traditional therapies. Functional β-cells play an important role in the pathogenesis of diabetes. As therapeutic targets, functional β-cells are restored by a variety of stem cells, including pluripotent stem cells, mesenchymal cells, and urine-derived stem cells. Although all types of stem cells have their own characteristics, they mainly promote the repair and regeneration of β-cells through directional differentiation, immunomodulation, and paracrine signaling after homing to the injured site. However, stem cell therapy still faces many obstacles, such as low long-term cell survival rate after transplantation, low maintenance time of blood glucose homeostasis, immune rejection, and tumorigenesis. Recently, genetically edited pluripotent stem cells and the co-transplantation of mesenchymal stem cells and islet cells have made significant progress in improving the efficacy of stem cell transplantation processes, also providing powerful tools for the study of the mechanisms underlying diabetes and disease modeling. In this review, we first focused on: (1) stem cells as a pool for the differentiation of insulin-producing cells; (2) stem cells as a source for regenerative repair of damaged islets and as a potential co-transplanted population with islets; (3) the potential of combining gene editing with stem cell therapy; and (4) selection of the stem cell transplantation approach. Based on these topics, we discuss the challenges within the field of adapting stem cell-supported and stem cell-derived transplantations, and the promising routes for overcoming these problems.
Collapse
Affiliation(s)
- Zijun Zhou
- The First Affiliated Hospital of Wenzhou Medical University, 89657, Endocrinology, Wenzhou, Zhejiang, China, 325000;
| | - Xiandong Zhu
- Wenzhou Medical University First Affiliated Hospital, 89657, Wenzhou, China, 325000;
| | - Hongjian Huang
- Wenzhou Medical College First Affiliated Hospital, 89657, Wenzhou, China, 325000;
| | - Zeru Xu
- The First Affiliated Hospital of Wenzhou Medical University, 89657, Wenzhou, China, 325000;
| | - Jiahong Jiang
- The First Affiliated Hospital of Wenzhou Medical University, 89657, endocrinology, Wenzhou, Zhejiang, China, 325000;
| | - Bicheng Chen
- Wenzhou Medical University First Affiliated Hospital, 89657, Wenzhou, China, 325000;
| | - Hong Zhu
- The First Affiliated Hospital of Wenzhou Medical University, 89657, Endocrinology, Wenzhou, Zhejiang, China, 325000;
| |
Collapse
|
9
|
Cell Transdifferentiation and Reprogramming in Disease Modeling: Insights into the Neuronal and Cardiac Disease Models and Current Translational Strategies. Cells 2021; 10:cells10102558. [PMID: 34685537 PMCID: PMC8533873 DOI: 10.3390/cells10102558] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
Cell transdifferentiation and reprogramming approaches in recent times have enabled the manipulation of cell fate by enrolling exogenous/artificial controls. The chemical/small molecule and regulatory components of transcription machinery serve as potential tools to execute cell transdifferentiation and have thereby uncovered new avenues for disease modeling and drug discovery. At the advanced stage, one can believe these methods can pave the way to develop efficient and sensitive gene therapy and regenerative medicine approaches. As we are beginning to learn about the utility of cell transdifferentiation and reprogramming, speculations about its applications in translational therapeutics are being largely anticipated. Although clinicians and researchers are endeavoring to scale these processes, we lack a comprehensive understanding of their mechanism(s), and the promises these offer for targeted and personalized therapeutics are scarce. In the present report, we endeavored to provide a detailed review of the original concept, methods and modalities enrolled in the field of cellular transdifferentiation and reprogramming. A special focus is given to the neuronal and cardiac systems/diseases towards scaling their utility in disease modeling and drug discovery.
Collapse
|
10
|
Agrawal A, Narayan G, Gogoi R, Thummer RP. Recent Advances in the Generation of β-Cells from Induced Pluripotent Stem Cells as a Potential Cure for Diabetes Mellitus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:1-27. [PMID: 34426962 DOI: 10.1007/5584_2021_653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Diabetes mellitus (DM) is a group of metabolic disorders characterized by high blood glucose levels due to insufficient insulin secretion, insulin action, or both. The present-day solution to diabetes mellitus includes regular administration of insulin, which brings about many medical complications in diabetic patients. Although islet transplantation from cadaveric subjects was proposed to be a permanent cure, the increased risk of infections, the need for immunosuppressive drugs, and their unavailability had restricted its use. To overcome this, the generation of renewable and transplantable β-cells derived from autologous induced pluripotent stem cells (iPSCs) has gained enormous interest as a potential therapeutic strategy to treat diabetes mellitus permanently. To date, extensive research has been undertaken to derive transplantable insulin-producing β-cells (iβ-cells) from iPSCs in vitro by recapitulating the in vivo developmental process of the pancreas. This in vivo developmental process relies on transcription factors, signaling molecules, growth factors, and culture microenvironment. This review highlights the various factors facilitating the generation of mature β-cells from iPSCs. Moreover, this review also describes the generation of pancreatic progenitors and β-cells from diabetic patient-specific iPSCs, exploring the potential of the diabetes disease model and drug discovery. In addition, the applications of genome editing strategies have also been discussed to achieve patient-specific diabetes cell therapy. Last, we have discussed the current challenges and prospects of iPSC-derived β-cells to improve the relative efficacy of the available treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Akriti Agrawal
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Gloria Narayan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Ranadeep Gogoi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Guwahati, Changsari, Guwahati, Assam, India
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.
| |
Collapse
|
11
|
Tailored generation of insulin producing cells from canine mesenchymal stem cells derived from bone marrow and adipose tissue. Sci Rep 2021; 11:12409. [PMID: 34117315 PMCID: PMC8196068 DOI: 10.1038/s41598-021-91774-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 06/01/2021] [Indexed: 12/30/2022] Open
Abstract
The trend of regenerative therapy for diabetes in human and veterinary practices has conceptually been proven according to the Edmonton protocol and animal models. Establishing an alternative insulin-producing cell (IPC) resource for further clinical application is a challenging task. This study investigated IPC generation from two practical canine mesenchymal stem cells (cMSCs), canine bone marrow-derived MSCs (cBM-MSCs) and canine adipose-derived MSCs (cAD-MSCs). The results illustrated that cBM-MSCs and cAD-MSCs contain distinct pancreatic differentiation potential and require the tailor-made induction protocols. The effective generation of cBM-MSC-derived IPCs needs the integration of genetic and microenvironment manipulation using a hanging-drop culture of PDX1-transfected cBM-MSCs under a three-step pancreatic induction protocol. However, this protocol is resource- and time-consuming. Another study on cAD-MSC-derived IPC generation found that IPC colonies could be obtained by a low attachment culture under the three-step induction protocol. Further, Notch signaling inhibition during pancreatic endoderm/progenitor induction yielded IPC colonies through the trend of glucose-responsive C-peptide secretion. Thus, this study showed that IPCs could be obtained from cBM-MSCs and cAD-MSCs through different induction techniques. Also, further signaling manipulation studies should be conducted to maximize the protocol’s efficiency.
Collapse
|
12
|
Pushp P, Nogueira DES, Rodrigues CAV, Ferreira FC, Cabral JMS, Gupta MK. A Concise Review on Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Personalized Regenerative Medicine. Stem Cell Rev Rep 2021; 17:748-776. [PMID: 33098306 DOI: 10.1007/s12015-020-10061-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 02/07/2023]
Abstract
The induced pluripotent stem cells (iPSCs) are derived from somatic cells by using reprogramming factors such as Oct4, Sox2, Klf4, and c-Myc (OSKM) or Oct4, Sox2, Nanog and Lin28 (OSNL). They resemble embryonic stem cells (ESCs) and have the ability to differentiate into cell lineage of all three germ-layer, including cardiomyocytes (CMs). The CMs can be generated from iPSCs by inducing embryoid bodies (EBs) formation and treatment with activin A, bone morphogenic protein 4 (BMP4), and inhibitors of Wnt signaling. However, these iPSC-derived CMs are a heterogeneous population of cells and require purification and maturation to mimic the in vivo CMs. The matured CMs can be used for various therapeutic purposes in regenerative medicine by cardiomyoplasty or through the development of tissue-engineered cardiac patches. In recent years, significant advancements have been made in the isolation of iPSC and their differentiation, purification, and maturation into clinically usable CMs. Newer small molecules have also been identified to substitute the reprogramming factors for iPSC generation as well as for direct differentiation of somatic cells into CMs without an intermediary pluripotent state. This review provides a concise update on the generation of iPSC-derived CMs and their application in personalized cardiac regenerative medicine. It also discusses the current limitations and challenges in the application of iPSC-derived CMs. Graphical abstract.
Collapse
Affiliation(s)
- Pallavi Pushp
- Department of Biotechnology, Institute of Engineering and Technology (IET), Bundelkhand University, Jhansi, Uttar Pradesh, 284128, India
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, 769 008, India
| | - Diogo E S Nogueira
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Frederico C Ferreira
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering, and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| | - Mukesh Kumar Gupta
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Odisha, 769 008, India.
| |
Collapse
|
13
|
Abstract
Type 1 diabetes mellitus is a common and highly morbid disease for which there is no cure. Treatment primarily involves exogenous insulin administration, and, under specific circumstances, islet or pancreas transplantation. However, insulin replacement alone fails to replicate the endocrine function of the pancreas and does not provide durable euglycemia. In addition, transplantation requires lifelong use of immunosuppressive medications, which has deleterious side effects, is expensive, and is inappropriate for use in adolescents. A bioartificial pancreas that provides total endocrine pancreatic function without immunosuppression is a potential therapy for treatment of type 1 diabetes. Numerous models are in development and take different approaches to cell source, encapsulation method, and device implantation location. We review current therapies for type 1 diabetes mellitus, the requirements for a bioartificial pancreas, and quantitatively compare device function.
Collapse
Affiliation(s)
- Sara J. Photiadis
- From the Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, CA
| | - Rebecca C. Gologorsky
- From the Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, CA
| | - Deepika Sarode
- From the Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, CA
| |
Collapse
|
14
|
Generation of high yield insulin-producing cells (IPCs) from various sources of stem cells. VITAMINS AND HORMONES 2021; 116:235-268. [PMID: 33752820 DOI: 10.1016/bs.vh.2021.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Type 1 diabetes mellitus occurs when beta cell mass is reduced to less than 20% of the normal level due to immune system destruction of beta cell resulting in an inability to secrete enough insulin. The prevalence of diabetes is expanding according to the American Diabetes Association and the World Health Organization (WHO), foretold to exceed 350 million by 2030. The current treatment does not cure many of the serious complications associated with the disease such as neuropathy, nephropathy, dyslipidemia, retinopathy and cardiovascular disease. Whole pancreas or isolated pancreatic islet transplantation as an alternative therapy can prevent or reduce some of the complications of diabetes. However, the shortage of matched organ or islets cells donor and alloimmune responses limit this therapeutic strategy. Recently, several reports have raised extremely promising results to use different sources of stem cells to differentiate insulin-producing cells and focus on the expansion of these alternative sources. Stem cells, due to their potential for multiple differentiation and self-renewal can differentiate into all cell types, including insulin-producing cells (IPCs). Generation of new beta cells can be achieved from various stem cell sources, including embryonic stem cells (ESCs), adult stem cells, such as mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs). Thus, this chapter discusses on the assistance of cellular reprogramming of various stem cells as candidates for the generation of IPCs using transcription factors/miRNA, cytokines/small molecules and tissue engineering.
Collapse
|
15
|
Tootee A, Nikbin B, Ghahary A, Esfahani EN, Arjmand B, Aghayan H, Qorbani M, Larijani B. Immunopathology of Type 1 Diabetes and Immunomodulatory Effects of Stem Cells: A Narrative Review of the Literature. Endocr Metab Immune Disord Drug Targets 2021; 22:169-197. [PMID: 33538679 DOI: 10.2174/1871530321666210203212809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/11/2020] [Accepted: 10/27/2020] [Indexed: 11/22/2022]
Abstract
Type 1 Diabetes (T1D) is a complex autoimmune disorder which occurs as a result of an intricate series of pathologic interactions between pancreatic β-cells and a wide range of components of both the innate and the adaptive immune systems. Stem-cell therapy, a recently-emerged potentially therapeutic option for curative treatment of diabetes, is demonstrated to cause significant alternations to both different immune cells such as macrophages, natural killer (NK) cells, dendritic cells, T cells, and B cells and non-cellular elements including serum cytokines and different components of the complement system. Although there exists overwhelming evidence indicating that the documented therapeutic effects of stem cells on patients with T1D is primarily due to their potential for immune regulation rather than pancreatic tissue regeneration, to date, the precise underlying mechanisms remain obscure. On the other hand, immune-mediated rejection of stem cells remains one of the main obstacles to regenerative medicine. Moreover, the consequences of efferocytosis of stem-cells by the recipients' lung-resident macrophages have recently emerged as a responsible mechanism for some immune-mediated therapeutic effects of stem-cells. This review focuses on the nature of the interactions amongst different compartments of the immune systems which are involved in the pathogenesis of T1D and provides explanation as to how stem cell-based interventions can influence immune system and maintain the physiologic equilibrium.
Collapse
Affiliation(s)
- Ali Tootee
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Behrouz Nikbin
- Research Center of Molecular Immunology, Tehran University of Medical Sciences, Tehran, . Iran
| | - Aziz Ghahary
- British Columbia Professional Firefighters' Burn and Wound Healing Research Laboratory, Department of Surgery, Plastic Surgery, University of British Columbia, Vancouver, . Canada
| | - Ensieh Nasli Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Babak Arjmand
- Cell therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Hamidreza Aghayan
- Cell therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, . Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| |
Collapse
|
16
|
Guo Q, Lu Y, Huang Y, Guo Y, Zhu S, Zhang Q, Zhu D, Wang Z, Luo J. Exosomes from β-Cells Promote Differentiation of Induced Pluripotent Stem Cells into Insulin-Producing Cells Through microRNA-Dependent Mechanisms. Diabetes Metab Syndr Obes 2021; 14:4767-4782. [PMID: 34934332 PMCID: PMC8678630 DOI: 10.2147/dmso.s342647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Exosomes have emerged as potential tools for the differentiation of induced pluripotent stem cells (iPSCs) into insulin-producing cells (IPCs). Exosomal microRNAs are receiving increasing attention in this process. Here, we aimed at investigating the role of exosomes derived from a murine pancreatic β-cell line and identifying signature exosomal miRNAs on iPSCs differentiation. METHODS Exosomes were isolated from MIN6 cells and identified with TEM, NTA and Western blot. PKH67 tracer and transwell assay were used to confirm exosome delivery into iPSCs. qRT-PCR was applied to detect key pancreatic transcription gene expression and exosome-derived miRNA expression. Insulin secretion was determined using FCM and immunofluorescence. The specific exosomal miRNAs were determined via RNA-interference of Ago2. The therapeutic effect of 21 day-exosome-induced IPCs was validated in T1D mice induced by STZ. RESULTS iPSCs cultured in medium containing exosomes showed sustained higher expression of MAFA, Insulin1, Insulin2, Isl1, Neuroud1, Nkx6.1 and NGN3 compared to control iPSCs. In FCM analysis, approximately 52.7% of the differentiated cells displayed insulin expression at the middle stage. Consistent with the gene expression data, immunofluorescence assays showed that Nkx6.1 and insulin expression in iPSCs were significantly upregulated. Intriguingly, the expression of pancreatic markers and insulin was significantly decreased in iPSCs cultured with siAgo2 exosomes. Transplantation of 21 day-induced IPCs intoT1D mice efficiently enhanced glucose tolerance and partially controlled hyperglycemia. The therapeutic effect was significantly attenuated in T1D mice that received iPSCs cultured with siAgo2 exosomes. Of the seven exosomal microRNAs selected for validation, miR-706, miR-709, miR-466c-5p, and miR-423-5p showed dynamic expression during 21 days in culture. CONCLUSION These data indicate that differentiation of exosome-induced iPSCs into functional cells is crucially dependent on the specific miRNAs encased within exosomes, whose functional analysis is likely to provide insight into novel regulatory mechanisms governing iPSCs differentiation into IPCs.
Collapse
Affiliation(s)
- Qingsong Guo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Yuhua Lu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Yibing Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Shajun Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Qiuqiang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Donghui Zhu
- Nantong University Medical School, Nantong, 226001, People’s Republic of China
| | - Zhiwei Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
- Zhiwei Wang Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Xi Si Road, Nantong, 226001, People’s Republic of China Email
| | - Jia Luo
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
- Correspondence: Jia Luo Department of Pharmacy, Affiliated Hospital of Nantong University, Xi Si Road, Nantong, 226001, People’s Republic of China Email
| |
Collapse
|
17
|
Exosome-Mediated Differentiation of Mouse Embryonic Fibroblasts and Exocrine Cells into β-Like Cells and the Identification of Key miRNAs for Differentiation. Biomedicines 2020; 8:biomedicines8110485. [PMID: 33182285 PMCID: PMC7695333 DOI: 10.3390/biomedicines8110485] [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] [Received: 10/14/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 01/08/2023] Open
Abstract
Diabetes is a concerning health malady worldwide. Islet or pancreas transplantation is the only long-term treatment available; however, the scarcity of transplantable tissues hampers this approach. Therefore, new cell sources and differentiation approaches are required. Apart from the genetic- and small molecule-based approaches, exosomes could induce cellular differentiation by means of their cargo, including miRNA. We developed a chemical-based protocol to differentiate mouse embryonic fibroblasts (MEFs) into β-like cells and employed mouse insulinoma (MIN6)-derived exosomes in the presence or absence of specific small molecules to encourage their differentiation into β-like cells. The differentiated β-like cells were functional and expressed pancreatic genes such as Pdx1, Nkx6.1, and insulin 1 and 2. We found that the exosome plus small molecule combination differentiated the MEFs most efficiently. Using miRNA-sequencing, we identified miR-127 and miR-709, and found that individually and in combination, the miRNAs differentiated MEFs into β-like cells similar to the exosome treatment. We also confirmed that exocrine cells can be differentiated into β-like cells by exosomes and the exosome-identified miRNAs. A new differentiation approach based on the use of exosome-identified miRNAs could help people afflicted with diabetes
Collapse
|
18
|
Manay P, Turgeon N, Axelrod DA. Role of Whole Organ Pancreas Transplantation in the Day of Bioartificial and Artificial Pancreas. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00300-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
19
|
Induced Pluripotent Stem Cells: Hope in the Treatment of Diseases, including Muscular Dystrophies. Int J Mol Sci 2020; 21:ijms21155467. [PMID: 32751747 PMCID: PMC7432218 DOI: 10.3390/ijms21155467] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem (iPS) cells are laboratory-produced cells that combine the biological advantages of somatic adult and stem cells for cell-based therapy. The reprogramming of cells, such as fibroblasts, to an embryonic stem cell-like state is done by the ectopic expression of transcription factors responsible for generating embryonic stem cell properties. These primary factors are octamer-binding transcription factor 4 (Oct3/4), sex-determining region Y-box 2 (Sox2), Krüppel-like factor 4 (Klf4), and the proto-oncogene protein homolog of avian myelocytomatosis (c-Myc). The somatic cells can be easily obtained from the patient who will be subjected to cellular therapy and be reprogrammed to acquire the necessary high plasticity of embryonic stem cells. These cells have no ethical limitations involved, as in the case of embryonic stem cells, and display minimal immunological rejection risks after transplant. Currently, several clinical trials are in progress, most of them in phase I or II. Still, some inherent risks, such as chromosomal instability, insertional tumors, and teratoma formation, must be overcome to reach full clinical translation. However, with the clinical trials and extensive basic research studying the biology of these cells, a promising future for human cell-based therapies using iPS cells seems to be increasingly clear and close.
Collapse
|
20
|
Sharma A, Sances S, Workman MJ, Svendsen CN. Multi-lineage Human iPSC-Derived Platforms for Disease Modeling and Drug Discovery. Cell Stem Cell 2020; 26:309-329. [PMID: 32142662 PMCID: PMC7159985 DOI: 10.1016/j.stem.2020.02.011] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) provide a powerful platform for disease modeling and have unlocked new possibilities for understanding the mechanisms governing human biology, physiology, and genetics. However, hiPSC-derivatives have traditionally been utilized in two-dimensional monocultures, in contrast to the multi-systemic interactions that influence cells in the body. We will discuss recent advances in generating more complex hiPSC-based systems using three-dimensional organoids, tissue-engineering, microfluidic organ-chips, and humanized animal systems. While hiPSC differentiation still requires optimization, these next-generation multi-lineage technologies can augment the biomedical researcher's toolkit and enable more realistic models of human tissue function.
Collapse
Affiliation(s)
- Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Samuel Sances
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Michael J Workman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
21
|
Jara C, Oyarzun-Ampuero F, Carrión F, González-Echeverría E, Cappelli C, Caviedes P. Microencapsulation of cellular aggregates composed of differentiated insulin and glucagon-producing cells from human mesenchymal stem cells derived from adipose tissue. Diabetol Metab Syndr 2020; 12:66. [PMID: 32774470 PMCID: PMC7409404 DOI: 10.1186/s13098-020-00573-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/21/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND In type I diabetes mellitus (T1DM) pancreatic β cells are destroyed. Treatment entails exogenous insulin administration and strict diet control, yet optimal glycemic control is hardly attainable. Islet transplant could be an alternative in patients with poor glycemic control, but inefficient islet purification and autoimmune response of patients is still a challenge. For these reasons, it is necessary to explore new cellular sources and immunological isolation methods oriented to develop T1DM cell-based therapies. AIMS We postulate human adipose-derived stem cell (hASC) as an adequate source to generate pancreatic islet cells in vitro, and to produce islet-like structures. Furthermore, we propose microencapsulation of these aggregates as an immunological isolation strategy. METHODS hASC obtained from lipoaspirated fat tissue from human donors were differentiated in vitro to insulin (Ins) and glucagon (Gcg) producing cells. Then, insulin producing cells (IPC) and glucagon producing cells (GPC) were cocultured in low adhesion conditions to form cellular aggregates, and later encapsulated in a sodium alginate polymer. Expression of pancreatic lineage markers and secretion of insulin or glucagon in vitro were analyzed. RESULTS The results show that multipotent hASC efficiently differentiate to IPC and GPC, and express pancreatic markers, including insulin or glucagon hormones which they secrete upon stimulation (fivefold for insulin in IPC, and fourfold for glucagon, compared to undifferentiated cells). In turn, calculation of the Feret diameter and area of cellular aggregates revealed mean diameters of ~ 80 µm, and 65% of the aggregates reached 4000 µm2 at 72 h of formation. IPC/GPC aggregates were then microencapsulated in sodium-alginate polymer microgels, which were found to be more stable when stabilized with Ba2+, yielding average diameters of ~ 300 µm. Interestingly, Ba2+-microencapsulated aggregates respond to high external glucose with insulin secretion. CONCLUSIONS The IPC/GPC differentiation process from hASC, followed by the generation of cellular aggregates that are later microencapsulated, could represent a possible treatment for T1DM.
Collapse
Affiliation(s)
- Claudia Jara
- Programa de Farmacología Molecular y Clínica, ICBM, Facultad de Medicina, Universidad de Chile, Independencia 1027., Casilla 7, Clasificador Nº 7, 8389100 Santiago, Chile
| | - Felipe Oyarzun-Ampuero
- Advanced Center of Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
- Depto. de Ciencias y Tecnología Farmacéuticas, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Flavio Carrión
- Programa de Inmunología Traslacional, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Esteban González-Echeverría
- Programa de Farmacología Molecular y Clínica, ICBM, Facultad de Medicina, Universidad de Chile, Independencia 1027., Casilla 7, Clasificador Nº 7, 8389100 Santiago, Chile
| | - Claudio Cappelli
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Pablo Caviedes
- Programa de Farmacología Molecular y Clínica, ICBM, Facultad de Medicina, Universidad de Chile, Independencia 1027., Casilla 7, Clasificador Nº 7, 8389100 Santiago, Chile
- Centro de Biotecnología y Bioingeniería (CeBiB), Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
22
|
Hoveizi E, Tavakol S, Shirian S, Sanamiri K. Electrospun Nanofibers for Diabetes: Tissue Engineering and Cell-Based Therapies. Curr Stem Cell Res Ther 2019; 14:152-168. [PMID: 30338744 DOI: 10.2174/1574888x13666181018150107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/11/2018] [Accepted: 09/05/2018] [Indexed: 02/08/2023]
Abstract
Diabetes mellitus is an autoimmune disease which causes loss of insulin secretion producing hyperglycemia by promoting progressive destruction of pancreatic β cells. An ideal therapeutic approach to manage diabetes mellitus is pancreatic β cells replacement. The aim of this review article was to evaluate the role of nanofibrous scaffolds and stem cells in the treatment of diabetes mellitus. Various studies have pointed out that application of electrospun biomaterials has considerably attracted researchers in the field of tissue engineering. The principles of cell therapy for diabetes have been reviewed in the first part of this article, while the usability of tissue engineering as a new therapeutic approach is discussed in the second part.
Collapse
Affiliation(s)
- Elham Hoveizi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran.,Stem Cells and Transgenic Technology Research Center (STTRC), Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,Shiraz Molecular Research Center, Dr. Daneshbod Pathology Lab, Shiraz, Iran
| | - Khadije Sanamiri
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| |
Collapse
|
23
|
Gharravi AM, Jafar A, Ebrahimi M, Mahmodi A, Pourhashemi E, Haseli N, Talaie N, Hajiasgarli P. Current status of stem cell therapy, scaffolds for the treatment of diabetes mellitus. Diabetes Metab Syndr 2018; 12:1133-1139. [PMID: 30168429 DOI: 10.1016/j.dsx.2018.06.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 06/25/2018] [Indexed: 12/24/2022]
Abstract
Diabetes mellitus (DM) remains the 7th leading cause of death in the world. Daily insulin injection is one component of a treatment plan for people with Diabetes mellitus type 1 (T1DM) that restores normal or near-normal blood sugar levels. However, Insulin treatment depends upon a variety of individual factors and leads to poor and drastic glycemic control. The need for an effective cell replacement strategy will be the aim of future clinical trials. Therefore, the aim of this systematic review is to outline the latest advances in scaffolding and stem cell therapy as a non-pharmacologic treatment for T1DM. It also emphasizes on some pancreas differentiation protocols and the clinical trials associated with stem cell therapy regarding T1DM in vitro and in vivo.
Collapse
Affiliation(s)
- Anneh Mohammad Gharravi
- Stem Cells and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Alireza Jafar
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mehrdad Ebrahimi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ahmad Mahmodi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Erfan Pourhashemi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Nasrin Haseli
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Niloofar Talaie
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Parinaz Hajiasgarli
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
24
|
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
|
25
|
Zhu H, Zhang X, He Y, Yu L, Lü Y, Pan K, Wang B, Chen G. [Research progress on the donor cell sources of pancreatic islet transplantation for treatment of diabetes mellitus]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:104-111. [PMID: 29806374 PMCID: PMC8414200 DOI: 10.7507/1002-1892.201707049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 12/13/2017] [Indexed: 11/03/2022]
Abstract
Objective To summarize the research progress on the source and selection of donor cells in the field of islet replacement therapy for diabetes mellitus. Methods Domestic and abroad literature concerning islet replacement therapy for diabetes mellitus, as well as donor source and donor selection was reviewed and analyzed thoroughly. Results The shortage of donor supply is still a major obstacle for the widely clinical application of pancreatic islet transplantation (PIT). Currently, in addition to the progress on the allogeneic/autologous donor islet supply, some remarkable achievements have been also attained in the application of xenogeneic islet (from pig donor), as well as islet like cells derived from stem cells and islet cell line, potentially enlarging the source of implantable cells. Conclusion Adequate and suitable donor cell supply is an essential prerequisite for widely clinical application of PIT therapy for type 1 diabetes mellitus (T1DM). Further perfection of organ donation system, together with development of immune-tolerance induction, gene and bioengineering technology etc. will possibly solve the problem of donor cell shortage and provide a basis for clinical application of cellular replacement therapy for T1DM.
Collapse
Affiliation(s)
- Haitao Zhu
- Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital, Xi'an Shaanxi, 710061, P.R.China;Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China
| | - Xiaoge Zhang
- Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital, Xi'an Shaanxi, 710061, P.R.China
| | - Yayi He
- Department of Endocrinology, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China
| | - Liang Yu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China
| | - Yi Lü
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China;Research Institute of Advanced Surgical Technology and Engineering, Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China
| | - Kaili Pan
- Department of Pediatrics (No. 2 Ward), Northwest Women's and Children's Hospital, Xi'an Shaanxi, 710061, P.R.China
| | - Bo Wang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061, P.R.China;Department of Endocrinology, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an Shaanxi, 710061,
| | - Guoqiang Chen
- Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital, Xi'an Shaanxi, 710061,
| |
Collapse
|
26
|
Human Induced Pluripotent Stem Cells in the Curative Treatment of Diabetes and Potential Impediments Ahead. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1144:25-35. [PMID: 30569414 DOI: 10.1007/5584_2018_305] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The successful landmark discovery of mouse and human inducible pluripotential stem cells (iPSC's) by Takahashi and Yamanaka in 2006 and 2007 has triggered a revolution in the potential generation of self-compatible cells for regenerative medicine, and further opened up a new avenue for "disease in dish" drug screening of self-target cells (Neofytou et al. 2015). The introduction of four 'Yamanaka' transcription factors through viral or other transfection of mature cells can induce pluripotency and acquired plasticity. These factors include transduction with octamer-binding transcription factor-4 (Oct-4), nanog homeobox (Nanog), sex-determining region Y-box-2 (Sox-2) and MYC protooncogene (cMyc). Such cells become iPSC's (Takahashi and Yamanaka 2006). These reprogrammed cells exhibit increased telomerase activity and have a hypomethylated gene promotor region similar to embryonic stem cells (ESC's). These milestone discoveries have generated immense hope that diseases such as diabetes could be treated and effectively cured by transplantation of self-compatible, personalized autologous stem cell transplantation of β-cells that release physiological insulin under glycemic control (Maehr et al. 2009; Park et al. 2008) (Fig. 1). Diabetes is a profligate disease of disordered glucose metabolism resulting from an absolute or relative deficiency of insulin, the consequences of which lead to immense socio-economic societal burden. While there are many different types of diabetes, the two major types (type 1 diabetes (T1DM) and type 2 diabetes (T2DM) are caused respectively by immune-mediated destruction (T1DM) or malfunctioning (T2DM) insulin-producing β-cells within the endocrine pancreas, the islets of Langerhans (Atkinson et al. 2011; Holman et al. 2015; You and Henneberg 2016). Almost 425 million people are affected by the global burden of diabetes, and this is predicted to increase by 48% (629 million) by 2045 (International Diabetes Federation Atlas 8th Ed 2018). Whole pancreas or islet cell transplantation offer an effective alternative to injected insulin, but both require lifelong potent immunosuppression to control both allo-and autoimmunity. Whole pancreas transplantation involves invasive complex surgery and is associated with greater morbidity and occasional mortality, while islet transplantation involves a minimally invasive intraportal hepatic infusion. Generally, whole pancreas transplantation provides greater metabolic reserve, but this may be matched by cumulative multiple islet infusions to achieve insulin independence. An additional challenge of islet transplantation is progressive loss of complete insulin independence over time, which may be multifactorial, the dominant factor however being ineffective control of autoimmunity. Both whole pancreas and islet transplantation are restricted to patients at risk of severe hypoglycemia that cannot be stabilized by alternate means, or in recipients that are already immunosuppressed in order to sustain a kidney or other solid organ transplant. The risks of chronic immunosuppression and the scarcity of human organ donors mean that both of these transplantation therapies cannot presently be extended to the broader diabetic population (Shapiro 2011; Shapiro et al. 2006). Recent progress in xenotransplantation of multiple knock-out 'humanized' pig islets could offer one potential solution, perhaps aided by clustered regularly interspaced short palindromic repeats/CRISPR associated-9 (CRISPR/Cas-9) gene editing approaches, but this remains to be proven in practice. Human stem cell derived new β-cell products could effectively address the global supply challenge for broad application across all forms of diabetes, but recurrent autoimmunity may still remain an insurmountable challenge. Considerable progress in the generation of human stem cell derived SC-β cells from ESC, iPS and other adult cell sources such as mesenchymal stem cells (MSCs) offer huge hope that a personalized, 'syngeneic' cell could be transplanted without risk of alloimmunity, thereby securing sufficient supply to meet future global demand (Cito et al. 2018).
Collapse
|
27
|
Al-Khawaga S, Memon B, Butler AE, Taheri S, Abou-Samra AB, Abdelalim EM. Pathways governing development of stem cell-derived pancreatic β cells: lessons from embryogenesis. Biol Rev Camb Philos Soc 2017. [DOI: 10.1111/brv.12349] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sara Al-Khawaga
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| | - Bushra Memon
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| | - Alexandra E. Butler
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine; University of California; Los Angeles CA 90095 U.S.A
| | - Shahrad Taheri
- Department of Medicine; Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, PO BOX 24144; Doha Qatar
- Department of Medicine; Qatar Metabolic Institute, Hamad Medical Corporation; Doha Qatar
| | - Abdul B. Abou-Samra
- Department of Medicine; Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, PO BOX 24144; Doha Qatar
- Department of Medicine; Qatar Metabolic Institute, Hamad Medical Corporation; Doha Qatar
| | - Essam M. Abdelalim
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| |
Collapse
|
28
|
Vieira A, Druelle N, Avolio F, Napolitano T, Navarro-Sanz S, Silvano S, Collombat P. β-Cell Replacement Strategies: The Increasing Need for a "β-Cell Dogma". Front Genet 2017. [PMID: 28634486 PMCID: PMC5459879 DOI: 10.3389/fgene.2017.00075] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes is an auto-immune disease resulting in the loss of pancreatic β-cells and, consequently, in chronic hyperglycemia. Insulin supplementation allows diabetic patients to control their glycaemia quite efficiently, but treated patients still display an overall shortened life expectancy and an altered quality of life as compared to their healthy counterparts. In this context and due to the ever increasing number of diabetics, establishing alternative therapies has become a crucial research goal. Most current efforts therefore aim at generating fully functional insulin-secreting β-like cells using multiple approaches. In this review, we screened the literature published since 2011 and inventoried the selected markers used to characterize insulin-secreting cells generated by in vitro differentiation of stem/precursor cells or by means of in vivo transdifferentiation. By listing these features, we noted important discrepancies when comparing the different approaches for the initial characterization of insulin-producing cells as true β-cells. Considering the recent advances achieved in this field of research, the necessity to establish strict guidelines has become a subject of crucial importance, especially should one contemplate the next step, which is the transplantation of in vitro or ex vivo generated insulin-secreting cells in type 1 diabetic patients.
Collapse
Affiliation(s)
- Andhira Vieira
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Noémie Druelle
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Fabio Avolio
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Tiziana Napolitano
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Sergi Navarro-Sanz
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Serena Silvano
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Patrick Collombat
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| |
Collapse
|
29
|
Langhans MT, Yu S, Tuan RS. Stem Cells in Skeletal Tissue Engineering: Technologies and Models. Curr Stem Cell Res Ther 2016; 11:453-474. [PMID: 26423296 DOI: 10.2174/1574888x10666151001115248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/01/2015] [Accepted: 04/01/2015] [Indexed: 12/14/2022]
Abstract
This review surveys the use of pluripotent and multipotent stem cells in skeletal tissue engineering. Specific emphasis is focused on evaluating the function and activities of these cells in the context of development in vivo, and how technologies and methods of stem cell-based tissue engineering for stem cells must draw inspiration from developmental biology. Information on the embryonic origin and in vivo differentiation of skeletal tissues is first reviewed, to shed light on the persistence and activities of adult stem cells that remain in skeletal tissues after embryogenesis. Next, the development and differentiation of pluripotent stem cells is discussed, and some of their advantages and disadvantages in the context of tissue engineering are presented. The final section highlights current use of multipotent adult mesenchymal stem cells, reviewing their origin, differentiation capacity, and potential applications to tissue engineering.
Collapse
Affiliation(s)
| | | | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA 15219, USA.
| |
Collapse
|
30
|
Baker K. Comparison of bioartificial and artificial pancreatic transplantation as promising therapies for Type I Diabetes Mellitus. ACTA ACUST UNITED AC 2016. [DOI: 10.1093/biohorizons/hzw002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
31
|
Takahashi Y, Takebe T, Taniguchi H. Engineering pancreatic tissues from stem cells towards therapy. Regen Ther 2016; 3:15-23. [PMID: 31245468 PMCID: PMC6581807 DOI: 10.1016/j.reth.2016.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/17/2015] [Accepted: 01/20/2016] [Indexed: 12/20/2022] Open
Abstract
Pancreatic islet transplantation is performed as a potential treatment for type 1 diabetes mellitus. However, this approach is significantly limited due to the critical shortage of islet sources. Recently, a number of publications have developed protocols for directed β-cell differentiation of pluripotent cells, such as embryonic stem (ES) or induced pluripotent stem (iPS) cells. Decades of studies have led to the development of modified protocols that recapitulate molecular developmental cues by combining various growth factors and small molecules with improved efficiency. However, the later step of pancreatic differentiation into functional β-cells has yet to be satisfactory in vitro, highlighting alternative approach by recapitulating spatiotemporal multicellular interaction in three-dimensional (3D) culture. Here, we summarize recent progress in the directed differentiation into pancreatic β-cells with a focus on both two-dimensional (2D) and 3D differentiation settings. We also discuss the potential transplantation strategies in combination with current bioengineering approaches towards diabetes therapy. Transplantation of stem cell derived pancreatic progenitors is a possible approach for generating mature β-cell in vivo. Promise of 3-D (or 4-D) culture has started to be explored by reconstituting pancreatic tissue structures. Self-condensation culture is a basic technique of integrating multiple heterotypic lineages including vasculatures. Bioengineering approach has been combined for developing effective transplant strategies.
Collapse
Key Words
- 2D, two-dimensional
- 3D, three-dimensional
- BMP, bone morphogenic protein
- Diabetes
- ES, embryonic stem
- FGF, fibroblast growth factors
- Heterotypic cellular interaction
- IBMIR, instant blood-mediated reaction
- ILV, indolactam V
- Ngn3, neurogenin 3
- PEG, polyethylene glycol
- PI3K, phosphatidylinositol-3 kinase
- PIPAAm, poly-N-isopropylacrylamide
- PVA, polyvinyl alcohol
- Pancreas
- Pdx1, pancreatic and duodenal homeobox 1
- Ptf1a, pancreatic transcription factor 1a
- Regenerative medicine
- VEGF, vascular endothelial growth factor
- Vascularization
- iPS, induced pluripotent stem
- iPS/ES cell
Collapse
Affiliation(s)
- Yoshinobu Takahashi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan
| | - Takanori Takebe
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan.,Advanced Medical Research Center, Yokohama City University, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan.,PRESTO, Japan Science and Technology Agency, 4-1-8, Honcho, Kawaguchi, Saitama, 332-0012, Japan.,Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH, 45229- 3039, USA
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan.,Advanced Medical Research Center, Yokohama City University, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan
| |
Collapse
|
32
|
Hayek A, King CC. Brief review: cell replacement therapies to treat type 1 diabetes mellitus. Clin Diabetes Endocrinol 2016; 2:4. [PMID: 28702240 PMCID: PMC5471705 DOI: 10.1186/s40842-016-0023-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/19/2016] [Indexed: 12/22/2022] Open
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent cells (iPSCs) have the potential to differentiate into any somatic cell, making them ideal candidates for cell replacement therapies to treat a number of human diseases and regenerate damaged or non-functional tissues and organs. Key to the promise of regenerative medicine is developing standardized protocols that can safely be applied in patients. Progress towards this goal has occurred in a number of fields, including type 1 diabetes mellitus (T1D). During the past 10 years, significant technological advances in hESC/iPSC biochemistry have provided a roadmap to generate sufficient quantities of glucose-responsive, insulin-producing cells capable of eliminating diabetes in rodents. Although many of the molecular mechanisms underlying the genesis of these cells remain to be elucidated, the field of cell-based therapeutics to treat T1D has advanced to the point where the first Phase I/II trials in humans have begun. Here, we provide a concise review of the history of cell replacement therapies to treat T1D from islet transplantations and xenotranplantation, to current work in hESC/iPSC. We also highlight the latest advances in efforts to employ insulin-producing, glucose-responsive β-like cells derived from hESC as therapeutics.
Collapse
Affiliation(s)
- Alberto Hayek
- Scripps Whittier Diabetes Institute, La Jolla, CA 92037 USA
| | - Charles C King
- Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093 USA
| |
Collapse
|
33
|
Kawser Hossain M, Abdal Dayem A, Han J, Kumar Saha S, Yang GM, Choi HY, Cho SG. Recent Advances in Disease Modeling and Drug Discovery for Diabetes Mellitus Using Induced Pluripotent Stem Cells. Int J Mol Sci 2016; 17:256. [PMID: 26907255 PMCID: PMC4783985 DOI: 10.3390/ijms17020256] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 02/05/2016] [Accepted: 02/15/2016] [Indexed: 02/07/2023] Open
Abstract
Diabetes mellitus (DM) is a widespread metabolic disease with a progressive incidence of morbidity and mortality worldwide. Despite extensive research, treatment options for diabetic patients remains limited. Although significant challenges remain, induced pluripotent stem cells (iPSCs) have the capacity to differentiate into any cell type, including insulin-secreting pancreatic β cells, highlighting its potential as a treatment option for DM. Several iPSC lines have recently been derived from both diabetic and healthy donors. Using different reprogramming techniques, iPSCs were differentiated into insulin-secreting pancreatic βcells. Furthermore, diabetes patient-derived iPSCs (DiPSCs) are increasingly being used as a platform to perform cell-based drug screening in order to develop DiPSC-based cell therapies against DM. Toxicity and teratogenicity assays based on iPSC-derived cells can also provide additional information on safety before advancing drugs to clinical trials. In this review, we summarize recent advances in the development of techniques for differentiation of iPSCs or DiPSCs into insulin-secreting pancreatic β cells, their applications in drug screening, and their role in complementing and replacing animal testing in clinical use. Advances in iPSC technologies will provide new knowledge needed to develop patient-specific iPSC-based diabetic therapies.
Collapse
Affiliation(s)
- Mohammed Kawser Hossain
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Gwangjin-gu, Seoul 05029, Korea.
| | - Ahmed Abdal Dayem
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Gwangjin-gu, Seoul 05029, Korea.
| | - Jihae Han
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Gwangjin-gu, Seoul 05029, Korea.
| | - Subbroto Kumar Saha
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Gwangjin-gu, Seoul 05029, Korea.
| | - Gwang-Mo Yang
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Gwangjin-gu, Seoul 05029, Korea.
| | - Hye Yeon Choi
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Gwangjin-gu, Seoul 05029, Korea.
| | - Ssang-Goo Cho
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Gwangjin-gu, Seoul 05029, Korea.
| |
Collapse
|
34
|
Abdelalim EM, Emara MM. Pluripotent Stem Cell-Derived Pancreatic β Cells: From In Vitro Maturation to Clinical Application. RECENT ADVANCES IN STEM CELLS 2016. [DOI: 10.1007/978-3-319-33270-3_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
35
|
Pellegrini S, Ungaro F, Mercalli A, Melzi R, Sebastiani G, Dotta F, Broccoli V, Piemonti L, Sordi V. Human induced pluripotent stem cells differentiate into insulin-producing cells able to engraft in vivo. Acta Diabetol 2015; 52:1025-35. [PMID: 25733399 DOI: 10.1007/s00592-015-0726-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 02/12/2015] [Indexed: 12/31/2022]
Abstract
AIMS New sources of insulin-secreting cells are strongly required for the cure of diabetes. Recent successes in differentiating embryonic stem cells, in combination with the discovery that it is possible to derive human induced pluripotent stem cells (iPSCs) from somatic cells, have raised the possibility that patient-specific beta cells might be derived from patients through cell reprogramming and differentiation. In this study, we aimed to obtain insulin-producing cells from human iPSCs and test their ability to secrete insulin in vivo. METHODS Human iPSCs, derived from both fetal and adult fibroblasts, were differentiated in vitro into pancreas-committed cells and then transplanted into immunodeficient mice at two different stages of differentiation (posterior foregut and endocrine cells). RESULTS IPSCs were shown to differentiate in insulin-producing cells in vitro, following the stages of pancreatic organogenesis. At the end of the differentiation, the production of INSULIN mRNA was highly increased and 5 ± 2.9 % of the cell population became insulin-positive. Terminally differentiated cells also produced C-peptide in vitro in both basal and stimulated conditions. In vivo, mice transplanted with pancreatic cells secreted human C-peptide in response to glucose stimulus, but transplanted cells were observed to lose insulin secretion capacity during the time. At histological evaluation, the grafts resulted to be composed of a mixed population of cells containing mature pancreatic cells, but also pluripotent and some neuronal cells. CONCLUSION These data overall suggest that human iPSCs have the potential to generate insulin-producing cells and that these differentiated cells can engraft and secrete insulin in vivo.
Collapse
Affiliation(s)
- Silvia Pellegrini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Federica Ungaro
- Stem Cells and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Alessia Mercalli
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Raffaella Melzi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, 53100, Siena, Italy
- Fondazione Umberto Di Mario ONLUS, c/o Toscana Life Sciences, 53100, Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, 53100, Siena, Italy
- Fondazione Umberto Di Mario ONLUS, c/o Toscana Life Sciences, 53100, Siena, Italy
| | - Vania Broccoli
- Stem Cells and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Valeria Sordi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
36
|
Abstract
Regenerative medicine using patient's own stem cells (SCs) to repair dysfunctional tissues is an attractive approach to complement surgical and pharmacological treatments for aging and degenerative disorders. Recently, dental SCs have drawn much attention owing to their accessibility, plasticity and applicability for regenerative use not only for dental, but also other body tissues. In ophthalmology, there has been increasing interest to differentiate dental pulp SC and periodontal ligament SC (PDLSC) towards ocular lineage. Both can commit to retinal fate expressing eye field transcription factors and generate rhodopsin-positive photoreceptor-like cells. This proposes a novel therapeutic alternative for retinal degeneration diseases. Moreover, as PDLSC shares similar cranial neural crest origin and proteoglycan secretion with corneal stromal keratoctyes and corneal endothelial cells, this offers the possibility of differentiating PDLSC to these corneal cell types. The advance could lead to a shift in the medical management of corneal opacities and endothelial disorders from highly invasive corneal transplantation using limited donor tissue to cell therapy utilizing autologous cells. This article provides an overview of dental SC research and the perspective of utilizing dental SCs for ocular regenerative medicine.
Collapse
|
37
|
Archibald PRT, Williams DJ. Using the cost–effectiveness of allogeneic islet transplantation to inform induced pluripotent stem cell-derived β-cell therapy reimbursement. Regen Med 2015; 10:959-73. [DOI: 10.2217/rme.15.59] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aims: In the present study a cost–effectiveness analysis of allogeneic islet transplantation was performed and the financial feasibility of a human induced pluripotent stem cell-derived β-cell therapy was explored. Methods: Previously published cost and health benefit data for islet transplantation were utilized to perform the cost–effectiveness and sensitivity analyses. Results & conclusion: It was determined that, over a 9-year time horizon, islet transplantation would become cost saving and ‘dominate’ the comparator. Over a 20-year time horizon, islet transplantation would incur significant cost savings over the comparator (GB£59,000). Finally, assuming a similar cost of goods to islet transplantation and a lack of requirement for immunosuppression, a human induced pluripotent stem cell-derived β-cell therapy would dominate the comparator over an 8-year time horizon.
Collapse
Affiliation(s)
- Peter RT Archibald
- Centre for Biological Engineering, Loughborough University, Loughborough, UK
| | - David J Williams
- Centre for Biological Engineering, Loughborough University, Loughborough, UK
| |
Collapse
|
38
|
Abstract
There is an increasing worldwide shortage of organs and cells for transplantation in patients with end-stage organ failure or cellular dysfunction. This shortage could be resolved by the transplantation of organs or cells from pigs into humans. What competing approaches might provide support for the patient with end-stage organ or cell failure? Four main approaches are receiving increasing attention - (i) implantable mechanical devices, although these are currently limited almost entirely to devices aimed at supporting or replacing the heart, (ii) stem cell technology, at present directed mainly to replace absent or failing cells, but which is also fundamental to progress in (iii) tissue engineering and regenerative medicine, in which the ultimate aim is to replace an entire organ. A final novel potential approach is (iv) blastocyst complementation. These potential alternative approaches are briefly reviewed, and comments added on their current status and whether they are now (or will soon become) realistic alternative therapies to xenotransplantation.
Collapse
|
39
|
Calafiore R, Basta G. Stem cells for the cell and molecular therapy of type 1 diabetes mellitus (T1D): the gap between dream and reality. AMERICAN JOURNAL OF STEM CELLS 2015; 4:22-31. [PMID: 25973328 PMCID: PMC4396156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 11/14/2014] [Indexed: 06/04/2023]
Abstract
In spite of intense research, over the past 2-3 decades, targeted to validating methods for the cure of T1D, based on cell substitution therapy in the place of exogenously administered insulin injections, achievement of the final goal continues to remain out of reach. In fact, aside of very limited clinical success of the few clinical trials of pancreatic islet cell transplantation in totally immunosuppressed patients with T1D, the vast majority of these diabetic patients invariably is insulin-dependent. New advances for cell and molecular therapy for T1D, including use of stem cells, are reviewed and discussed in an attempt to clearly establish where we are and where are we may go for the final cure for T1DM.
Collapse
Affiliation(s)
- Riccardo Calafiore
- Department of Medicine, Section of Clinical Cardiovascular, Endocrine and Metabolic Physiology, University of Perugia School of Medicine at Terni Terni, Italy
| | - Giuseppe Basta
- Department of Medicine, Section of Clinical Cardiovascular, Endocrine and Metabolic Physiology, University of Perugia School of Medicine at Terni Terni, Italy
| |
Collapse
|
40
|
Stepniewski J, Kachamakova-Trojanowska N, Ogrocki D, Szopa M, Matlok M, Beilharz M, Dyduch G, Malecki MT, Jozkowicz A, Dulak J. Induced pluripotent stem cells as a model for diabetes investigation. Sci Rep 2015; 5:8597. [PMID: 25716801 PMCID: PMC4341212 DOI: 10.1038/srep08597] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 01/27/2015] [Indexed: 12/15/2022] Open
Abstract
Mouse and human induced pluripotent stem cells (iPSCs) may represent a novel approach for modeling diabetes. Taking this into consideration, the aim of this study was to generate and evaluate differentiation potential of iPSCs from lepdb/db (db/db) mice, the model of diabetes type 2 as well as from patients with Maturity Onset Diabetes of the Young 3 (HNF1A MODY). Murine iPSC colonies from both wild type and db/db mice were positive for markers of pluripotency: Oct3/4A, Nanog, SSEA1, CDy1 and alkaline phosphatase and differentiated in vitro and in vivo into cells originating from three germ layers. However, our results suggest impaired differentiation of db/db cells into endothelial progenitor-like cells expressing CD34 and Tie2 markers and their reduced angiogenic potential. Human control and HNF1A MODY reprogrammed cells also expressed pluripotency markers: OCT3/4A, SSEA4, TRA-1–60, TRA-1-81, formed embryoid bodies (EBs) and differentiated into cells of three germ layers. Additionally, insulin expressing cells were obtained from those partially reprogrammed cells with direct as well as EB-mediated differentiation method. Our findings indicate that disease-specific iPSCs may help to better understand the mechanisms responsible for defective insulin production or vascular dysfunction upon differentiation toward cell types affected by diabetes.
Collapse
Affiliation(s)
- J Stepniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - N Kachamakova-Trojanowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - D Ogrocki
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - M Szopa
- 1] Clinic of Metabolic Diseases, Jagiellonian University Medical College, Krakow, Poland [2] University Hospital Krakow Poland
| | - M Matlok
- II Clinic of General Surgery, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - M Beilharz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - G Dyduch
- Department of Clinical and Experimental Pathomorphology, Jagiellonian University Medical College, Krakow, Poland
| | - M T Malecki
- 1] Clinic of Metabolic Diseases, Jagiellonian University Medical College, Krakow, Poland [2] University Hospital Krakow Poland
| | - A Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - J Dulak
- 1] Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland [2] Malopolska Centre of Biotechnology, Jagiellonian University, Poland
| |
Collapse
|
41
|
Abstract
The discovery of hypocretins (orexins) and their causal implication in narcolepsy is the most important advance in sleep research and sleep medicine since the discovery of rapid eye movement sleep. Narcolepsy with cataplexy is caused by hypocretin deficiency owing to destruction of most of the hypocretin-producing neurons in the hypothalamus. Ablation of hypocretin or hypocretin receptors also leads to narcolepsy phenotypes in animal models. Although the exact mechanism of hypocretin deficiency is unknown, evidence from the past 20 years strongly favours an immune-mediated or autoimmune attack, targeting specifically hypocretin neurons in genetically predisposed individuals. These neurons form an extensive network of projections throughout the brain and show activity linked to motivational behaviours. The hypothesis that a targeted immune-mediated or autoimmune attack causes the specific degeneration of hypocretin neurons arose mainly through the discovery of genetic associations, first with the HLA-DQB1*06:02 allele and then with the T-cell receptor α locus. Guided by these genetic findings and now awaiting experimental testing are models of the possible immune mechanisms by which a specific and localised brain cell population could become targeted by T-cell subsets. Great hopes for the identification of new targets for therapeutic intervention in narcolepsy also reside in the development of patient-derived induced pluripotent stem cell systems.
Collapse
|
42
|
Abdelalim EM, Emara MM. Advances and challenges in the differentiation of pluripotent stem cells into pancreatic β cells. World J Stem Cells 2015; 7:174-181. [PMID: 25621117 PMCID: PMC4300928 DOI: 10.4252/wjsc.v7.i1.174] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/16/2014] [Accepted: 09/19/2014] [Indexed: 02/06/2023] Open
Abstract
Pluripotent stem cells (PSCs) are able to differentiate into several cell types, including pancreatic β cells. Differentiation of pancreatic β cells depends on certain transcription factors, which function in a coordinated way during pancreas development. The existing protocols for in vitro differentiation produce pancreatic β cells, which are not highly responsive to glucose stimulation except after their transplantation into immune-compromised mice and allowing several weeks for further differentiation to ensure the maturation of these cells in vivo. Thus, although the substantial improvement that has been made for the differentiation of induced PSCs and embryonic stem cells toward pancreatic β cells, several challenges still hindering their full generation. Here, we summarize recent advances in the differentiation of PSCs into pancreatic β cells and discuss the challenges facing their differentiation as well as the different applications of these potential PSC-derived β cells.
Collapse
|
43
|
Abdelalim EM, Bonnefond A, Bennaceur-Griscelli A, Froguel P. Pluripotent stem cells as a potential tool for disease modelling and cell therapy in diabetes. Stem Cell Rev Rep 2014; 10:327-37. [PMID: 24577791 DOI: 10.1007/s12015-014-9503-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diabetes mellitus is the most prevailing disease with progressive incidence worldwide. To date, the pathogenesis of diabetes is far to be understood, and there is no permanent treatment available for diabetes. One of the promising approaches to understand and cure diabetes is to use pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced PCSs (iPSCs). ESCs and iPSCs have a great potential to differentiate into all cell types, and they have a high ability to differentiate into insulin-secreting β cells. Obtaining PSCs genetically identical to the patient presenting with diabetes has been a longstanding dream for the in vitro modeling of disease and ultimately cell therapy. For several years, somatic cell nuclear transfer (SCNT) was the method of choice to generate patient-specific ESC lines. However, this technology faces ethical and practical concerns. Interestingly, the recently established iPSC technology overcomes the major problems of other stem cell types including the lack of ethical concern and no risk of immune rejection. Several iPSC lines have been recently generated from patients with different types of diabetes, and most of these cell lines are able to differentiate into insulin-secreting β cells. In this review, we summarize recent advances in the differentiation of pancreatic β cells from PSCs, and describe the challenges for their clinical use in diabetes cell therapy. Furthermore, we discuss the potential use of patient-specific PSCs as an in vitro model, providing new insights into the pathophysiology of diabetes.
Collapse
Affiliation(s)
- Essam M Abdelalim
- Qatar Biomedical Research Institute, Qatar Foundation, Education City, 5825, Doha, Qatar,
| | | | | | | |
Collapse
|
44
|
Kumar SS, Alarfaj AA, Munusamy MA, Singh AJAR, Peng IC, Priya SP, Hamat RA, Higuchi A. Recent developments in β-cell differentiation of pluripotent stem cells induced by small and large molecules. Int J Mol Sci 2014; 15:23418-47. [PMID: 25526563 PMCID: PMC4284775 DOI: 10.3390/ijms151223418] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/03/2014] [Accepted: 12/08/2014] [Indexed: 12/21/2022] Open
Abstract
Human pluripotent stem cells, including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), hold promise as novel therapeutic tools for diabetes treatment because of their self-renewal capacity and ability to differentiate into beta (β)-cells. Small and large molecules play important roles in each stage of β-cell differentiation from both hESCs and hiPSCs. The small and large molecules that are described in this review have significantly advanced efforts to cure diabetic disease. Lately, effective protocols have been implemented to induce hESCs and human mesenchymal stem cells (hMSCs) to differentiate into functional β-cells. Several small molecules, proteins, and growth factors promote pancreatic differentiation from hESCs and hMSCs. These small molecules (e.g., cyclopamine, wortmannin, retinoic acid, and sodium butyrate) and large molecules (e.g. activin A, betacellulin, bone morphogentic protein (BMP4), epidermal growth factor (EGF), fibroblast growth factor (FGF), keratinocyte growth factor (KGF), hepatocyte growth factor (HGF), noggin, transforming growth factor (TGF-α), and WNT3A) are thought to contribute from the initial stages of definitive endoderm formation to the final stages of maturation of functional endocrine cells. We discuss the importance of such small and large molecules in uniquely optimized protocols of β-cell differentiation from stem cells. A global understanding of various small and large molecules and their functions will help to establish an efficient protocol for β-cell differentiation.
Collapse
Affiliation(s)
- S Suresh Kumar
- Department of Medical Microbiology and Parasitology, Universities Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Abdullah A Alarfaj
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Murugan A Munusamy
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - A J A Ranjith Singh
- Department of Bioscience, Jacintha Peter College of Arts and Sciences, Ayakudi, Tenkasi, Tamilnadu 627852, India.
| | - I-Chia Peng
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan 32001, Taiwan.
| | - Sivan Padma Priya
- Department of Basic Science and Department of Surgical Sciences, Ajman University of Science and Technology-Fujairah Campus, P.O. Box 9520, Al Fujairah, United Arab Emirates.
| | - Rukman Awang Hamat
- Department of Medical Microbiology and Parasitology, Universities Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Akon Higuchi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
45
|
Kanherkar RR, Bhatia-Dey N, Makarev E, Csoka AB. Cellular reprogramming for understanding and treating human disease. Front Cell Dev Biol 2014; 2:67. [PMID: 25429365 PMCID: PMC4228919 DOI: 10.3389/fcell.2014.00067] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/27/2014] [Indexed: 12/15/2022] Open
Abstract
In the last two decades we have witnessed a paradigm shift in our understanding of cells so radical that it has rewritten the rules of biology. The study of cellular reprogramming has gone from little more than a hypothesis, to applied bioengineering, with the creation of a variety of important cell types. By way of metaphor, we can compare the discovery of reprogramming with the archeological discovery of the Rosetta stone. This stone slab made possible the initial decipherment of Egyptian hieroglyphics because it allowed us to see this language in a way that was previously impossible. We propose that cellular reprogramming will have an equally profound impact on understanding and curing human disease, because it allows us to perceive and study molecular biological processes such as differentiation, epigenetics, and chromatin in ways that were likewise previously impossible. Stem cells could be called “cellular Rosetta stones” because they allow also us to perceive the connections between development, disease, cancer, aging, and regeneration in novel ways. Here we present a comprehensive historical review of stem cells and cellular reprogramming, and illustrate the developing synergy between many previously unconnected fields. We show how stem cells can be used to create in vitro models of human disease and provide examples of how reprogramming is being used to study and treat such diverse diseases as cancer, aging, and accelerated aging syndromes, infectious diseases such as AIDS, and epigenetic diseases such as polycystic ovary syndrome. While the technology of reprogramming is being developed and refined there have also been significant ongoing developments in other complementary technologies such as gene editing, progenitor cell production, and tissue engineering. These technologies are the foundations of what is becoming a fully-functional field of regenerative medicine and are converging to a point that will allow us to treat almost any disease.
Collapse
Affiliation(s)
- Riya R Kanherkar
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| | - Naina Bhatia-Dey
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| | - Evgeny Makarev
- InSilico Medicine, Emerging Technology Center, Johns Hopkins University Eastern Baltimore, MD, USA
| | - Antonei B Csoka
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| |
Collapse
|
46
|
Minie M, Chopra G, Sethi G, Horst J, White G, Roy A, Hatti K, Samudrala R. CANDO and the infinite drug discovery frontier. Drug Discov Today 2014; 19:1353-63. [PMID: 24980786 DOI: 10.1016/j.drudis.2014.06.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 12/21/2022]
Abstract
The Computational Analysis of Novel Drug Opportunities (CANDO) platform (http://protinfo.org/cando) uses similarity of compound-proteome interaction signatures to infer homology of compound/drug behavior. We constructed interaction signatures for 3733 human ingestible compounds covering 48,278 protein structures mapping to 2030 indications based on basic science methodologies to predict and analyze protein structure, function, and interactions developed by us and others. Our signature comparison and ranking approach yielded benchmarking accuracies of 12-25% for 1439 indications with at least two approved compounds. We prospectively validated 49/82 'high value' predictions from nine studies covering seven indications, with comparable or better activity to existing drugs, which serve as novel repurposed therapeutics. Our approach may be generalized to compounds beyond those approved by the FDA, and can also consider mutations in protein structures to enable personalization. Our platform provides a holistic multiscale modeling framework of complex atomic, molecular, and physiological systems with broader applications in medicine and engineering.
Collapse
Affiliation(s)
- Mark Minie
- University of Washington, Department of Bioengineering, Seattle, WA 98109, United States
| | - Gaurav Chopra
- University of Washington, Department of Microbiology, Seattle, WA 98109, United States; University of California, San Francisco, Diabetes Center, San Francisco, CA 94143, United States
| | - Geetika Sethi
- University of Washington, Department of Microbiology, Seattle, WA 98109, United States
| | - Jeremy Horst
- University of California, School of Medicine, San Francisco, CA 94143, United States
| | - George White
- University of Washington, Department of Microbiology, Seattle, WA 98109, United States
| | - Ambrish Roy
- Georgia Institute of Technology, Center for the Study of Systems Biology, Atlanta, GA 30318, United States
| | - Kaushik Hatti
- Molecular Biophysics Unit, Indian Institute of Science Bangalore, 560012, India
| | - Ram Samudrala
- University of Washington, Department of Microbiology, Seattle, WA 98109, United States.
| |
Collapse
|
47
|
Van Pham P, Thi-My Nguyen P, Thai-Quynh Nguyen A, Minh Pham V, Nguyen-Tu Bui A, Thi-Tung Dang L, Gia Nguyen K, Kim Phan N. Improved differentiation of umbilical cord blood-derived mesenchymal stem cells into insulin-producing cells by PDX-1 mRNA transfection. Differentiation 2014; 87:200-8. [DOI: 10.1016/j.diff.2014.08.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 08/04/2014] [Accepted: 08/18/2014] [Indexed: 02/08/2023]
|
48
|
Amer LD, Mahoney MJ, Bryant SJ. Tissue engineering approaches to cell-based type 1 diabetes therapy. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:455-67. [PMID: 24417705 DOI: 10.1089/ten.teb.2013.0462] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Type 1 diabetes mellitus is an autoimmune disease resulting from the destruction of insulin-producing pancreatic β-cells. Cell-based therapies, involving the transplantation of functional β-cells into diabetic patients, have been explored as a potential long-term treatment for this condition; however, success is limited. A tissue engineering approach of culturing insulin-producing cells with extracellular matrix (ECM) molecules in three-dimensional (3D) constructs has the potential to enhance the efficacy of cell-based therapies for diabetes. When cultured in 3D environments, insulin-producing cells are often more viable and secrete more insulin than those in two dimensions. The addition of ECM molecules to the culture environments, depending on the specific type of molecule, can further enhance the viability and insulin secretion. This review addresses the different cell sources that can be utilized as β-cell replacements, the essential ECM molecules for the survival of these cells, and the 3D culture techniques that have been used to benefit cell function.
Collapse
Affiliation(s)
- Luke D Amer
- 1 Department of Chemical and Biological Engineering, University of Colorado , Boulder, Colorado
| | | | | |
Collapse
|
49
|
Wang P, Moore A. Theranostic MRI: the future for Type 1 diabetes management? ACTA ACUST UNITED AC 2014. [DOI: 10.2217/iim.13.67] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
50
|
Lahmy R, Soleimani M, Sanati MH, Behmanesh M, Kouhkan F, Mobarra N. miRNA-375 promotes beta pancreatic differentiation in human induced pluripotent stem (hiPS) cells. Mol Biol Rep 2014; 41:2055-66. [DOI: 10.1007/s11033-014-3054-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 01/04/2014] [Indexed: 01/28/2023]
|