1
|
Kieffer TJ, Hoesli CA, Shapiro AMJ. Advances in Islet Transplantation and the Future of Stem Cell-Derived Islets to Treat Diabetes. Cold Spring Harb Perspect Med 2025; 15:a041624. [PMID: 39074874 PMCID: PMC12047745 DOI: 10.1101/cshperspect.a041624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
β-Cell replacement for type 1 diabetes (T1D) can restore normal glucose homeostasis, thereby eliminating the need for exogenous insulin and halting the progression of diabetes complications. Success in achieving insulin independence following transplantation of cadaveric islets fueled academic and industry efforts to develop techniques to mass produce β cells from human pluripotent stem cells, and these have now been clinically validated as an alternative source of regulated insulin production. Various encapsulation strategies are being pursued to contain implanted cells in a retrievable format, and different implant sites are being explored with some strategies reaching clinical studies. Stem cell lines, whether derived from embryonic sources or reprogrammed somatic cells, are being genetically modified for designer features, including immune evasiveness to enable implant without the use of chronic immunosuppression. Although hurdles remain in optimizing large-scale manufacturing, demonstrating efficacy, durability, and safety, products containing stem cell-derived β cells promise to provide a potent treatment for insulin-dependent diabetes.
Collapse
Affiliation(s)
- Timothy J Kieffer
- Department of Cellular and Physiological Sciences, Life Sciences Institute, School of Biomedical Engineering
- Department of Surgery, The University of British Columbia, Vancouver V6T1Z3, British Columbia, Canada
| | - Corinne A Hoesli
- Department of Chemical Engineering, Department of Biomedical Engineering, McGill University, Montreal H3A 0C5, Québec, Canada
- Associate Member, Department of Biomedical Engineering, McGill University, Montreal H3A 0C5, Québec, Canada
| | - A M James Shapiro
- Clinical Islet Transplant Program, University of Alberta, Edmonton T6G2E1, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton T6G2E1, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton T6G2E1, Alberta, Canada
| |
Collapse
|
2
|
Horikawa A, Michiue T. Controlling spheroid attachment improves pancreatic beta cell differentiation from human iPS cells. In Vitro Cell Dev Biol Anim 2024:10.1007/s11626-024-00991-3. [PMID: 39546193 DOI: 10.1007/s11626-024-00991-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/26/2024] [Indexed: 11/17/2024]
Abstract
Regenerative medicine using human induced pluripotent stem cells (hiPSCs) is available for treating type 1 diabetes; however, the efficiency and maturation of hiPSC differentiation into pancreatic beta cells requires improvement. Various protocols, including three-dimensional (3D) culture, have been developed to improve differentiation efficiency and maturation. Several methods for 3D culture have been reported; however, they require costly and complicated equipment, special materials, and complicated operations. To solve these problems, we developed a simple 3D culture method under static conditions using a cyclo-olefin polymer (COP) characterized by high moisture barrier properties, low surface energy, and hydrophobicity. Using this 3D method and our simple and low-cost protocol, we found that differentiation into the definitive endoderm (DE) was better when the spheroids were attached. Therefore, upon the addition of Y-27632, attached spheroids with unique shapes and cavities were formed, and the differentiation efficiency into DE increased. During DE differentiation, the attachment of spheroids to the substrate and their subsequent floating improved differentiation efficiency. We found that the amount of C-peptide in spheroids differentiated using COP dishes was greater than that in rotary culture. Furthermore, INSULIN was highly expressed in areas with low cell density, suggesting that the unique shape of the spheroids made from COP dishes improved differentiation efficiency. Our study suggests that a device-free, simple 3D culture method that controls spheroid attachment improves the efficiency of hiPSC differentiation into pancreatic beta cells.
Collapse
Affiliation(s)
- Ayumi Horikawa
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-Ku, Tokyo, 153-8902, Japan
| | - Tatsuo Michiue
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-Ku, Tokyo, 153-8902, Japan.
| |
Collapse
|
3
|
Diane A, Mohammed LI, Al-Siddiqi HH. Islets in the body are never flat: transitioning from two-dimensional (2D) monolayer culture to three-dimensional (3D) spheroid for better efficiency in the generation of functional hPSC-derived pancreatic β cells in vitro. Cell Commun Signal 2023; 21:151. [PMID: 37349801 PMCID: PMC10286450 DOI: 10.1186/s12964-023-01171-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/20/2023] [Indexed: 06/24/2023] Open
Abstract
Diabetes mellitus (DM), currently affecting more than 537 million people worldwide is a chronic disease characterized by impaired glucose metabolism resulting from a defect in insulin secretion, action, or both due to the loss or dysfunction of pancreatic β cells. Since cadaveric islet transplantation using Edmonton protocol has served as an effective intervention to restore normoglycaemia in T1D patients for months, stem cell-derived β cells have been explored for cell replacement therapy for diabetes. Thus, great effort has been concentrated by scientists on developing in vitro differentiation protocols to realize the therapeutic potential of hPSC-derived β cells. However, most of the 2D traditional monolayer culture could mainly generate insulin-producing β cells with immature phenotype. In the body, pancreatic islets are 3D cell arrangements with complex cell-cell and cell-ECM interactions. Therefore, it is important to consider the spatial organization of the cell in the culture environment. More recently, 3D cell culture platforms have emerged as powerful tools with huge translational potential, particularly for stem cell research. 3D protocols provide a better model to recapitulate not only the in vivo morphology, but also the cell connectivity, polarity, and gene expression mimicking more physiologically the in vivo cell niche. Therefore, the 3D culture constitutes a more relevant model that may help to fill the gap between in vitro and in vivo models. Interestingly, most of the 2D planar methodologies that successfully generated functional hPSC-derived β cells have switched to a 3D arrangement of cells from pancreatic progenitor stage either as suspension clusters or as aggregates, suggesting the effect of 3D on β cell functionality. In this review we highlight the role of dimensionality (2D vs 3D) on the differentiation efficiency for generation of hPSC-derived insulin-producing β cells in vitro. Consequently, how transitioning from 2D monolayer culture to 3D spheroid would provide a better model for an efficient generation of fully functional hPSC-derived β cells mimicking in vivo islet niche for diabetes therapy or drug screening. Video Abstract.
Collapse
Affiliation(s)
- Abdoulaye Diane
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.
| | - Layla Ibrahim Mohammed
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Heba H Al-Siddiqi
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
4
|
Goh SK, Bertera S, Richardson T, Banerjee I. Repopulation of decellularized organ scaffolds with human pluripotent stem cell-derived pancreatic progenitor cells. Biomed Mater 2023; 18. [PMID: 36720168 DOI: 10.1088/1748-605x/acb7bf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/31/2023] [Indexed: 02/02/2023]
Abstract
Diabetes is an emerging global epidemic that affects more that 285 million people worldwide. Engineering of endocrine pancreas tissue holds great promise for the future of diabetes therapy. Here we demonstrate the feasibility of re-engineering decellularized organ scaffolds using regenerative cell source. We differentiated human pluripotent stem cells (hPSC) toward pancreatic progenitor (PP) lineage and repopulated decellularized organ scaffolds with these hPSC-PP cells. We observed that hPSCs cultured and differentiated as aggregates are more suitable for organ repopulation than isolated single cell suspension. However, recellularization with hPSC-PP aggregates require a more extensive vascular support, which was found to be superior in decellularized liver over the decellularized pancreas scaffolds. Upon continued culture for nine days with chemical induction in the bioreactor, the seeded hPSC-PP aggregates demonstrated extensive and uniform cellular repopulation and viability throughout the thickness of the liver scaffolds. Furthermore, the decellularized liver scaffolds was supportive of the endocrine cell fate of the engrafted cells. Our novel strategy to engineer endocrine pancreas construct is expected to find potential applications in preclinical testing, drug discovery and diabetes therapy.
Collapse
Affiliation(s)
- Saik-Kia Goh
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Suzanne Bertera
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, United States of America
| | - Thomas Richardson
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ipsita Banerjee
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
5
|
Togo H, Terada K, Ujitsugu A, Hirose Y, Takeuchi H, Kusunoki M. Fabrication Scaffold with High Dimensional Control for Spheroids with Undifferentiated iPS Cell Properties. Cells 2023; 12:278. [PMID: 36672213 PMCID: PMC9857117 DOI: 10.3390/cells12020278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
Spheroids are expected to aid the establishment of an in vitro-based cell culture system that can realistically reproduce cellular dynamics in vivo. We developed a fluoropolymer scaffold with an extracellular matrix (ECM) dot array and confirmed the possibility of mass-producing spheroids with uniform dimensions. Controlling the quality of ECM dots is important as it ensures spheroid uniformity, but issues such as pattern deviation and ECM drying persist in the conventional microstamping method. In this study, these problems were overcome via ECM dot printing using a resin mask with dot-patterned holes. For dot diameters of φ 300 μm, 400 μm, and 600 μm, the average spheroid diameters of human iPS cells (hiPSCs) were φ 260.8 μm, 292.4 μm, and 330.7 μm, respectively. The standard deviation when each average was normalized to 100 was 14.1%. A high throughput of 89.9% for colony formation rate to the number of dots and 89.3% for spheroid collection rate was achieved. The cells proliferated on ECM dots, and the colonies could be naturally detached from the scaffold without the use of enzymes, so there was almost no stimulation of the cells. Thus, the undifferentiated nature of hiPSCs was maintained until day 4. Therefore, this method is expected to be useful in drug discovery and regenerative medicine.
Collapse
Affiliation(s)
- Hidetaka Togo
- Graduate School of Biology-Oriented-Science and Technology, Kindai University, 930 Nishimitani, Kinokawa 649-6493, Wakayama, Japan
| | - Kento Terada
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Akira Ujitsugu
- Faculty of Biology-Oriented-Science and Technology, Kindai University, 930 Nishimitani, Kinokawa 649-6493, Wakayama, Japan
| | - Yudai Hirose
- Graduate School of Biology-Oriented-Science and Technology, Kindai University, 930 Nishimitani, Kinokawa 649-6493, Wakayama, Japan
| | - Hiroki Takeuchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu 514-8507, Mie, Japan
| | - Masanobu Kusunoki
- Graduate School of Biology-Oriented-Science and Technology, Kindai University, 930 Nishimitani, Kinokawa 649-6493, Wakayama, Japan
- Faculty of Biology-Oriented-Science and Technology, Kindai University, 930 Nishimitani, Kinokawa 649-6493, Wakayama, Japan
| |
Collapse
|
6
|
Silva IBB, Kimura CH, Colantoni VP, Sogayar MC. Stem cells differentiation into insulin-producing cells (IPCs): recent advances and current challenges. Stem Cell Res Ther 2022; 13:309. [PMID: 35840987 PMCID: PMC9284809 DOI: 10.1186/s13287-022-02977-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 06/19/2022] [Indexed: 11/10/2022] Open
Abstract
Type 1 diabetes mellitus (T1D) is a chronic disease characterized by an autoimmune destruction of insulin-producing β-pancreatic cells. Although many advances have been achieved in T1D treatment, current therapy strategies are often unable to maintain perfect control of glycemic levels. Several studies are searching for new and improved methodologies for expansion of β-cell cultures in vitro to increase the supply of these cells for pancreatic islets replacement therapy. A promising approach consists of differentiation of stem cells into insulin-producing cells (IPCs) in sufficient number and functional status to be transplanted. Differentiation protocols have been designed using consecutive cytokines or signaling modulator treatments, at specific dosages, to activate or inhibit the main signaling pathways that control the differentiation of induced pluripotent stem cells (iPSCs) into pancreatic β-cells. Here, we provide an overview of the current approaches and achievements in obtaining stem cell-derived β-cells and the numerous challenges, which still need to be overcome to achieve this goal. Clinical translation of stem cells-derived β-cells for efficient maintenance of long-term euglycemia remains a major issue. Therefore, research efforts have been directed to the final steps of in vitro differentiation, aiming at production of functional and mature β-cells and integration of interdisciplinary fields to generate efficient cell therapy strategies capable of reversing the clinical outcome of T1D.
Collapse
Affiliation(s)
- Isaura Beatriz Borges Silva
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil.,Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Camila Harumi Kimura
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil
| | - Vitor Prado Colantoni
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil.,Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Mari Cleide Sogayar
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil. .,Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
7
|
Sandilya S, Singh S. Development of islet organoids from human induced pluripotent stem cells in a cross-linked collagen scaffold. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:38. [PMID: 34850295 PMCID: PMC8633270 DOI: 10.1186/s13619-021-00099-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 09/03/2021] [Indexed: 12/14/2022]
Abstract
Islets organoids would have value in the cell replacement therapy for diabetes apart from usual personalized drug screening routes. Generation of a large number of Islets like clusters, with ability to respond to glucose stimulation appears to be an ideal choice. In this study we have generated islet organoids with the ability to respond to glucose stimulation by insulin release. The source of the cells was an iPSC cell line differentiated into the pancreatic progenitors. These cells were assembled in matrigel or cross-linked collagen scaffold and compared for their efficacy to release insulin upon stimulation with glucose. The assembled organoids were examined by immunohistochemistry and expression of the relevant marker genes. The organoids showed expression of islet like markers in both - matrigel and crosslinked collagen scaffold. The islet organoids in both the cases showed release of insulin upon stimulation with glucose. The crosslinked collagen scaffold is quite stable and supports islet cells growth and function.
Collapse
Affiliation(s)
- Shruti Sandilya
- CSIR- Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
| | - Shashi Singh
- CSIR- Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India.
| |
Collapse
|
8
|
Nihad M, Shenoy P S, Bose B. Cell therapy research for Diabetes: Pancreatic β cell differentiation from pluripotent stem cells. Diabetes Res Clin Pract 2021; 181:109084. [PMID: 34673084 DOI: 10.1016/j.diabres.2021.109084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022]
Abstract
Human pluripotent stem cells (PSCs), both embryonic and induced pluripotent stem cells (iPSCs), have been differentiated into pancreatic β isletsin vitrofor more than a decade. The idea is to get enough β cells for cell transplantation for diabetics. Finding a standard cell therapy for diabetes is essential because of the logarithmic increase in the global population of people with diabetes and the insufficient availability of the human cadaveric pancreas. Moreover, with better insights into developmental biology, thein vitroβ cell differentiation protocols have depended on thein vivoβ cell organogenesis. Various protocols for pancreatic β cell differentiation have been developed. Such protocols are based on the modulation of cell signalling pathways with growth factors, small molecules, RNAi approaches, directed differentiation using transcription factors, genome editing. Growth factor free differentiation protocols, epigenetic modulations, 3D differentiation approaches, and encapsulation strategies have also been reported for better glycemic control and endocrine modulations. Here, we have reviewed various aforementionedin vitroβ cell differentiation protocols from human PSCs, their respective comparisons, challenges, past, present, and future. The literature has been reviewed primarily from PubMed from the year 2000 till date using the mentioned keywords.
Collapse
Affiliation(s)
- Muhammad Nihad
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Pincode-575 018, Karnataka, India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Pincode-575 018, Karnataka, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Pincode-575 018, Karnataka, India.
| |
Collapse
|
9
|
Kuo YC, Tsao CW, Rajesh R. Dual-sized inverted colloidal crystal scaffolds grafted with GDF-8 and Wnt3a for enhancing differentiation of iPS cells toward islet β-cells. J Taiwan Inst Chem Eng 2021. [DOI: 10.1016/j.jtice.2021.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
10
|
Choi H, Shinohara M, Ibuki M, Nishikawa M, Sakai Y. Differentiation of Human-Induced Pluripotent Stem Cell-Derived Endocrine Progenitors to Islet-like Cells Using a Dialysis Suspension Culture System. Cells 2021; 10:cells10082017. [PMID: 34440786 PMCID: PMC8392085 DOI: 10.3390/cells10082017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 12/01/2022] Open
Abstract
The production of functional islet-like cells from human-induced pluripotent stem cells (hiPSCs) is a promising strategy for the therapeutic use and disease modeling for type 1 diabetes. However, the production cost of islet-like cells is extremely high due to the use of expensive growth factors for differentiation. In a conventional culture method, growth factors and beneficial autocrine factors remaining in the culture medium are removed along with toxic metabolites during the medium change, and it limits the efficient utilization of those factors. In this study, we demonstrated that the dialysis suspension culture system is possible to reduce the usage of growth factors to one-third in the differentiation of hiPSC-derived endocrine progenitor cells to islet-like cells by reducing the medium change frequency with the refinement of the culture medium. Furthermore, the expression levels of hormone-secretion-related genes and the efficiency of differentiation were improved with the dialysis suspension culture system, possibly due to the retaining of autocrine factors. In addition, we confirmed several improvements required for the further study of the dialysis culture system. These findings showed the promising possibility of the dialysis suspension culture system for the low-cost production of islet-like cells.
Collapse
Affiliation(s)
- Hyunjin Choi
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
- Correspondence:
| | - Marie Shinohara
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan; (M.S.); (M.I.)
| | - Masato Ibuki
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan; (M.S.); (M.I.)
- Kaneka Corporation, Osaka 530-0005, Japan
| | - Masaki Nishikawa
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan; (M.N.); (Y.S.)
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan; (M.N.); (Y.S.)
| |
Collapse
|
11
|
Golchin A, Farzaneh S, Porjabbar B, Sadegian F, Estaji M, Ranjbarvan P, Kanafimahbob M, Ranjbari J, Salehi-Nik N, Hosseinzadeh S. Regenerative Medicine Under the Control of 3D Scaffolds: Current State and Progress of Tissue Scaffolds. Curr Stem Cell Res Ther 2021; 16:209-229. [DOI: 10.2174/1574888x15666200720115519] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 11/22/2022]
Abstract
Currently, combining stem cells (SCs) with biomaterial scaffolds provides a promising strategy
for the future of biomedicine and regenerative medicine (RG). The cells need similar substrates of
the extracellular matrix (ECM) for normal tissue development, which signifies the importance of
three dimensional (3D) scaffolds to determine cell fate. Herein, the importance and positive contributions
of corresponding 3D scaffolds on cell functions, including cell interactions, cell migrations,
and nutrient delivery, are presented. Furthermore, the synthesis techniques which are recruited to
fabricate the 3D scaffolds are discussed, and the related studies of 3D scaffold for different tissues
are also reported in this paper. This review focuses on 3D scaffolds that have been used for tissue
engineering purposes and directing stem cell fate as a means of producing replacements for biomedical
applications.
Collapse
Affiliation(s)
- Ali Golchin
- Department of Clinical Biochemistry and Applied Cell Science, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Sina Farzaneh
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh Porjabbar
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sadegian
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Estaji
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parviz Ranjbarvan
- Department of Clinical Biochemistry and Applied Cell Science, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Kanafimahbob
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Ranjbari
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Nasim Salehi-Nik
- Department of Biomechanical Engineering, University of Twente, Enschede, Netherlands
| | - Simzar Hosseinzadeh
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
McMinn P, Guckenberger DJ, Beebe DJ. Induced Pluripotent Stem Cells on a Chip: A Self-Contained, Accessible, Pipette-less iPSC Culturing and Differentiation Kit. SLAS Technol 2021; 26:80-91. [PMID: 32552316 PMCID: PMC10843275 DOI: 10.1177/2472630320921173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Over the past decade, induced pluripotent stem cells (iPSCs) have become a major focus of stem cell and developmental biology research, offering researchers a clinically relevant source of cells that are amenable to genetic engineering approaches. Though stem cells are promising for both research and commercial endeavors, iPSC-based assays require tedious protocols that include complex treatments, expensive reagents, and specialized equipment that limit their integration into academic curricula and cell biology research groups. Expanding on existing Kit-On-A-Lid-Assay (KOALA) technologies, we have developed a self-contained, injection molded, pipette-less iPSC culture and differentiation platform that significantly reduces associated costs and labor of stem cell maintenance and differentiation. The KOALA kit offers users the full range of iPSC culture necessities, including cell cryopreservation, media exchanges, differentiation, endpoint analysis, and a new capability, cell passaging. Using the KOALA kit, we were able to culture ~20,000 iPSCs per microchannel for at least 7 days, while maintaining stable expression of stemness markers (SSEA4 and Oct4) and normal iPSC phenotype. We also adapted protocols for differentiating iPSCs into neuroepithelial cells, cardiomyocytes, and definitive endodermal cells, a cell type from each germ layer of human development.
Collapse
Affiliation(s)
- Patrick McMinn
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, Madison, WI, USA
| | - David J Guckenberger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
13
|
Enhanced differentiation of human pluripotent stem cells into pancreatic endocrine cells in 3D culture by inhibition of focal adhesion kinase. Stem Cell Res Ther 2020; 11:488. [PMID: 33198821 PMCID: PMC7667734 DOI: 10.1186/s13287-020-02003-z] [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: 06/04/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022] Open
Abstract
Background Generation of insulin-producing cells from human pluripotent stem cells (hPSCs) in vitro would be useful for drug discovery and cell therapy in diabetes. Three-dimensional (3D) culture is important for the acquisition of mature insulin-producing cells from hPSCs, but the mechanism by which it promotes β cell maturation is poorly understood. Methods We established a stepwise method to induce high-efficiency differentiation of human embryonic stem cells (hESCs) into mature monohormonal pancreatic endocrine cells (PECs), with the last maturation stage in 3D culture. To comprehensively compare two-dimensional (2D) and 3D cultures, we examined gene expression, pancreas-specific markers, and functional characteristics in 2D culture-induced PECs and 3D culture-induced PECs. The mechanisms were considered from the perspectives of cell–cell and cell–extracellular matrix interactions which are fundamentally different between 2D and 3D cultures. Results The expression of the pancreatic endocrine-specific transcription factors PDX1, NKX6.1, NGN3, ISL1, and PAX6 and the hormones INS, GCG, and SST was significantly increased in 3D culture-induced PECs. 3D culture yielded monohormonal endocrine cells, while 2D culture-induced PECs co-expressed INS and GCG or INS and SST or even expressed all three hormones. We found that focal adhesion kinase (FAK) phosphorylation was significantly downregulated in 3D culture-induced PECs, and treatment with the selective FAK inhibitor PF-228 improved the expression of β cell-specific transcription factors in 2D culture-induced PECs. We further demonstrated that 3D culture may promote endocrine commitment by limiting FAK-dependent activation of the SMAD2/3 pathway. Moreover, the expression of the gap junction protein Connexin 36 was much higher in 3D culture-induced PECs than in 2D culture-induced PECs, and inhibition of the FAK pathway in 2D culture increased Connexin 36 expression. Conclusion We developed a strategy to induce differentiation of monohormonal mature PECs from hPSCs and found limited FAK-dependent activation of the SMAD2/3 pathway and unregulated expression of Connexin 36 in 3D culture-induced PECs. This study has important implications for the generation of mature, functional β cells for drug discovery and cell transplantation therapy for diabetes and sheds new light on the signaling events that regulate endocrine specification. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-020-02003-z.
Collapse
|
14
|
Insulin/Glucose-Responsive Cells Derived from Induced Pluripotent Stem Cells: Disease Modeling and Treatment of Diabetes. Cells 2020; 9:cells9112465. [PMID: 33198288 PMCID: PMC7696367 DOI: 10.3390/cells9112465] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes, characterized by dysfunction of pancreatic β-cells and insulin resistance in peripheral organs, accounts for more than 90% of all diabetes. Despite current developments of new drugs and strategies to prevent/treat diabetes, there is no ideal therapy targeting all aspects of the disease. Restoration, however, of insulin-producing β-cells, as well as insulin-responsive cells, would be a logical strategy for the treatment of diabetes. In recent years, generation of transplantable cells derived from stem cells in vitro has emerged as an important research area. Pluripotent stem cells, either embryonic or induced, are alternative and feasible sources of insulin-secreting and glucose-responsive cells. This notwithstanding, consistent generation of robust glucose/insulin-responsive cells remains challenging. In this review, we describe basic concepts of the generation of induced pluripotent stem cells and subsequent differentiation of these into pancreatic β-like cells, myotubes, as well as adipocyte- and hepatocyte-like cells. Use of these for modeling of human disease is now feasible, while development of replacement therapies requires continued efforts.
Collapse
|
15
|
Li L, Tan D, Liu S, Jiao R, Yang X, Li F, Wu H, Huang W. Optimization of Factor Combinations for Stem Cell Differentiations on a Design-of-Experiment Microfluidic Chip. Anal Chem 2020; 92:14228-14235. [PMID: 33017151 DOI: 10.1021/acs.analchem.0c03488] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Directed differentiation of stem cells plays a vital role in cell replacement therapy. Many activators and inhibitors targeting different signaling pathways have been identified to contribute to each step of differentiation. Most studies relied on empirically optimizing the combinations of the aforementioned factors for each step to optimize the efficiency of differentiation, which are time-consuming and nonsystematic. Design-of-experiment (DOE) is a powerful strategy to identify the critical combinations from multiple factors systematically. However, it is prohibitively complicated for typical laboratories, given a large number of potential combinations. Here, we develop a multilayer polymethyl methacrylate-based, reusable microfluidic chip to directly facilitate the DOE in the differentiation of stem cells. The chip consists of an inlet layer and multiple disperse layers. Different solutions are injected simultaneously to the chip through the inlet layer. Subsequently, the channels in the disperse layers split and recombine the flow streams to generate solution combinations based on hard-wired DOE designs. We demonstrated that it is in quantitative agreement with the designs using fluorescent dyes. Moreover, we constructed a human-induced pluripotent stem reporter cell line to improve the consistency of the cellular state measurements and use the chip to identify critical factors for cell differentiation to definitive endoderm (DE). We found that the differentiation efficiencies under various factor combinations are significantly different, and CHIR99201 and GDF8 are the most critical factors for differentiation to DE. Our method is potentially applicable to the optimization of factor combinations for multi-step stem cell differentiation and combinatorial drug screening.
Collapse
Affiliation(s)
- Lijun Li
- Department of Biology, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055 Guangdong, China.,Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, 999077 Hong Kong, China
| | - Deng Tan
- Department of Biology, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055 Guangdong, China.,Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, 999077 Hong Kong, China
| | - Shuqin Liu
- Department of Biology, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055 Guangdong, China
| | - Ruifeng Jiao
- Department of Biology, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055 Guangdong, China
| | - Xiaofei Yang
- Translational Medicine Collaborative Innovation Center, The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, 1017 Dongmen North Road, Luohu District, Shenzhen, 518020 Guangdong, China
| | - Furong Li
- Translational Medicine Collaborative Innovation Center, The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, 1017 Dongmen North Road, Luohu District, Shenzhen, 518020 Guangdong, China
| | - Hongkai Wu
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, 999077 Hong Kong, China.,Guangzhou First People's Hospital, 1 Panfu Rd, Yuexiu District, Guangzhou, 510180 Guangdong, China
| | - Wei Huang
- Department of Biology, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055 Guangdong, China
| |
Collapse
|
16
|
Goh SK, Halfter W, Richardson T, Bertera S, Vaidya V, Candiello J, Bradford M, Banerjee I. Organ-specific ECM arrays for investigating Cell-ECM interactions during stem cell differentiation. Biofabrication 2020; 13. [PMID: 33045682 DOI: 10.1088/1758-5090/abc05f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/12/2020] [Indexed: 12/22/2022]
Abstract
Pluripotent stem cells are promising source of cells for tissue engineering, regenerative medicine and drug discovery applications. The process of stem cell differentiation is regulated by multi-parametric cues from the surrounding microenvironment, one of the critical one being cell interaction with extracellular matrix (ECM). The ECM is a complex tissue-specific structure which are important physiological regulators of stem cell function and fate. Recapitulating this native ECM microenvironment niche is best facilitated by decellularized tissue/ organ derived ECM, which can faithfully reproduce the physiological environment with high fidelity to in vivo condition and promote tissue-specific cellular development and maturation. Recognizing the need for organ specific ECM in a 3D culture environment in driving phenotypic differentiation and maturation of hPSCs, we fabricated an ECM array platform using native-mimicry ECM from decellularized organs (namely pancreas, liver and heart), which allows cell-ECM interactions in both 2D and 3D configuration. The ECM array was integrated with rapid quantitative imaging for a systematic investigation of matrix protein profiles and sensitive measurement of cell-ECM interaction during hPSC differentiation. We tested our platform by elucidating the role of the three different organ-specific ECM in supporting induced pancreatic differentiation of hPSCs. While the focus of this report is on pancreatic differentiation, the developed platform is versatile to be applied to characterize any lineage specific differentiation.
Collapse
Affiliation(s)
- Saik Kia Goh
- University of Pittsburgh, Pittsburgh, 15261, UNITED STATES
| | - Willi Halfter
- University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Thomas Richardson
- Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Suzanne Bertera
- Allegheny Health Network, Pittsburgh, Pennsylvania, UNITED STATES
| | - Vimal Vaidya
- University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Joe Candiello
- University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Mahalia Bradford
- Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Ipsita Banerjee
- Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, UNITED STATES
| |
Collapse
|
17
|
Jamwal VS, Vishnu VV, Domreddy A, Parekh Y, Kumar BK, Chandra Shekar P, Singh S. Generation of iPSC from fetal fibroblast cells obtained from an abortus with type-I tri-allelic variants. Stem Cell Res 2020; 48:101963. [PMID: 32916634 DOI: 10.1016/j.scr.2020.101963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 08/07/2020] [Accepted: 08/23/2020] [Indexed: 12/16/2022] Open
Abstract
An integration free iPSC line was generated from fibroblast obtained from the skin of an aborted fetus in feeder free conditions using episomal based vectors expressing the pluripotency factors. The cell line generated was characterized and tested for pluripotency both in vitro and in vivo by teratoma formation and differentiation into defined lineages and brain organoids. Cell line reported here is shown to be mycoplasma free.
Collapse
Affiliation(s)
| | - Vijay V Vishnu
- Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | - Yash Parekh
- Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | | - Shashi Singh
- Centre for Cellular and Molecular Biology, Hyderabad, India.
| |
Collapse
|
18
|
Huang H, Bader TN, Jin S. Signaling Molecules Regulating Pancreatic Endocrine Development from Pluripotent Stem Cell Differentiation. Int J Mol Sci 2020; 21:E5867. [PMID: 32824212 PMCID: PMC7461594 DOI: 10.3390/ijms21165867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/08/2020] [Accepted: 08/09/2020] [Indexed: 12/24/2022] Open
Abstract
Diabetes is one of the leading causes of death globally. Currently, the donor pancreas is the only source of human islets, placing extreme constraints on supply. Hence, it is imperative to develop renewable islets for diabetes research and treatment. To date, extensive efforts have been made to derive insulin-secreting cells from human pluripotent stem cells with substantial success. However, the in vitro generation of functional islet organoids remains a challenge due in part to our poor understanding of the signaling molecules indispensable for controlling differentiation pathways towards the self-assembly of functional islets from stem cells. Since this process relies on a variety of signaling molecules to guide the differentiation pathways, as well as the culture microenvironments that mimic in vivo physiological conditions, this review highlights extracellular matrix proteins, growth factors, signaling molecules, and microenvironments facilitating the generation of biologically functional pancreatic endocrine cells from human pluripotent stem cells. Signaling pathways involved in stepwise differentiation that guide the progression of stem cells into the endocrine lineage are also discussed. The development of protocols enabling the generation of islet organoids with hormone release capacities equivalent to native adult islets for clinical applications, disease modeling, and diabetes research are anticipated.
Collapse
Affiliation(s)
- Hui Huang
- Department of Biomedical Engineering, Thomas J. Watson School of Engineering and Applied Sciences, State University of New York at Binghamton, Binghamton, NY 13902, USA; (H.H.); (T.N.B.)
| | - Taylor N. Bader
- Department of Biomedical Engineering, Thomas J. Watson School of Engineering and Applied Sciences, State University of New York at Binghamton, Binghamton, NY 13902, USA; (H.H.); (T.N.B.)
| | - Sha Jin
- Department of Biomedical Engineering, Thomas J. Watson School of Engineering and Applied Sciences, State University of New York at Binghamton, Binghamton, NY 13902, USA; (H.H.); (T.N.B.)
- Center of Biomanufacturing for Regenerative Medicine, State University of New York at Binghamton, Binghamton, NY 13902, USA
| |
Collapse
|
19
|
Bi H, Karanth SS, Ye K, Stein R, Jin S. Decellularized Tissue Matrix Enhances Self-Assembly of Islet Organoids from Pluripotent Stem Cell Differentiation. ACS Biomater Sci Eng 2020; 6:4155-4165. [PMID: 33463310 DOI: 10.1021/acsbiomaterials.0c00088] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regenerating human islet organoids from stem cells remains a significant challenge because of our limited knowledge on cues essential for developing the endocrine organoids in vitro. In this study, we discovered that a natural material prepared from a decellularized rat pancreatic extracellular matrix (dpECM) induces the self-assembly of human islet organoids during induced pluripotent stem cell (iPSC) pancreatic differentiation. For the first time, we demonstrated that the iPSC-derived islet organoids formed in the presence of the dpECM are capable of glucose-responsive secretion of both insulin and glucagon, two major hormones that maintain blood glucose homeostasis. The characterization of the organoids revealed that the organoids consisted of all major endocrine cell types, including α, β, δ, and pancreatic polypeptide cells, that were assembled into a tissue architecture similar to that of human islets. The exposure of iPSCs to the dpECM during differentiation resulted in considerably elevated expression of key pancreatic transcription factors such as PDX-1, MAFA, and NKX6.1 and the production of all major hormones, including insulin, glucagon, somatostatin, and pancreatic polypeptide from stem cell-derived organoids. This study highlights the importance of natural, bioactive biomaterials for building microenvironments crucial to regenerating islet organoids from stem cells.
Collapse
Affiliation(s)
- Huanjing Bi
- Department of Biomedical Engineering, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, United States
| | - Soujanya S Karanth
- Department of Biomedical Engineering, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, United States
| | - Kaiming Ye
- Department of Biomedical Engineering, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, United States.,Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, United States
| | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Sha Jin
- Department of Biomedical Engineering, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, United States.,Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, United States
| |
Collapse
|
20
|
Thakur G, Lee HJ, Jeon RH, Lee SL, Rho GJ. Small Molecule-Induced Pancreatic β-Like Cell Development: Mechanistic Approaches and Available Strategies. Int J Mol Sci 2020; 21:E2388. [PMID: 32235681 PMCID: PMC7178115 DOI: 10.3390/ijms21072388] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes is a metabolic disease which affects not only glucose metabolism but also lipid and protein metabolism. It encompasses two major types: type 1 and 2 diabetes. Despite the different etiologies of type 1 and 2 diabetes mellitus (T1DM and T2DM, respectively), the defining features of the two forms are insulin deficiency and resistance, respectively. Stem cell therapy is an efficient method for the treatment of diabetes, which can be achieved by differentiating pancreatic β-like cells. The consistent generation of glucose-responsive insulin releasing cells remains challenging. In this review article, we present basic concepts of pancreatic organogenesis, which intermittently provides a basis for engineering differentiation procedures, mainly based on the use of small molecules. Small molecules are more auspicious than any other growth factors, as they have unique, valuable properties like cell-permeability, as well as a nonimmunogenic nature; furthermore, they offer immense benefits in terms of generating efficient functional beta-like cells. We also summarize advances in the generation of stem cell-derived pancreatic cell lineages, especially endocrine β-like cells or islet organoids. The successful induction of stem cells depends on the quantity and quality of available stem cells and the efficient use of small molecules.
Collapse
Affiliation(s)
- Gitika Thakur
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Hyeon-Jeong Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Ryoung-Hoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Sung-Lim Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Gyu-Jin Rho
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| |
Collapse
|
21
|
Rahimi-Sherbaf F, Nadri S, Rahmani A, Dabiri Oskoei A. Placenta mesenchymal stem cells differentiation toward neuronal-like cells on nanofibrous scaffold. ACTA ACUST UNITED AC 2020; 10:117-122. [PMID: 32363155 PMCID: PMC7186541 DOI: 10.34172/bi.2020.14] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/06/2019] [Accepted: 09/07/2019] [Indexed: 12/20/2022]
Abstract
Introduction: Transplantation of stem cells with a nanofibrous scaffold is a promising approach for spinal cord injury therapy. The aim of this work was to differentiate neural-like cells from placenta-derived mesenchymal stem cells (PDMSCs) using suitable induction reagents in three (3D) and two dimensional (2D) culture systems. Methods: After isolation and characterization of PDMSCs, the cells were cultivated on poly-L-lactide acid (PLLA)/poly caprolactone (PCL) nanofibrous scaffold and treated with a neuronal medium for 7 days. Electron microscopy, qPCR, and immunostaining were used to examine the differentiation of PDMSCs (on scaffold and tissue culture polystyrene [TCPS]) and the expression rate of neuronal markers (beta-tubulin, nestin, GFAP, and MAP-2). Results: qPCR analysis showed that beta-tubulin (1.672 fold; P ≤ 0.0001), nestin (11.145 fold; P ≤ 0.0001), and GFAP (80.171; P ≤ 0.0001) gene expressions were higher on scaffolds compared with TCPS. Immunofluorescence analysis showed that nestin and beta-tubulin proteins were recognized in the PDMSCs differentiated on TCPS and scaffold after 7 days in the neuroinductive differentiation medium. Conclusion: Taken together, these results delegated that PDMSCs differentiated on PLLA/PCL scaffolds are more likely to differentiate towards diversity lineages of neural cells. It proposed that PDMSCs have cell subpopulations that have the capability to be differentiated into neurogenic cells.
Collapse
Affiliation(s)
- Fatemeh Rahimi-Sherbaf
- Department of Obstetrics and Gynecology, School of Medicine, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Samad Nadri
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Rahmani
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Atousa Dabiri Oskoei
- Department of Obstetrics and Gynecology, Mousavi Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
22
|
Kuo YC, Lee IH, Rajesh R. Self-assembled ternary poly(vinyl alcohol)-alginate-gelatin hydrogel with controlled-release nanoparticles for pancreatic differentiation of iPS cells. J Taiwan Inst Chem Eng 2019. [DOI: 10.1016/j.jtice.2019.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
23
|
Kim JH, Park BG, Kim SK, Lee DH, Lee GG, Kim DH, Choi BO, Lee KB, Kim JH. Nanotopographical regulation of pancreatic islet-like cluster formation from human pluripotent stem cells using a gradient-pattern chip. Acta Biomater 2019; 95:337-347. [PMID: 30529081 DOI: 10.1016/j.actbio.2018.12.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022]
Abstract
Bioengineering approaches to regulate stem cell fates aim to recapitulate the in vivo microenvironment. In recent years, manipulating the micro- and nano-scale topography of the stem cell niche has gained considerable interest for the purposes of controlling extrinsic mechanical cues to regulate stem cell fate and behavior in vitro. Here, we established an optimal nanotopographical system to improve 3-dimensional (3D) differentiation of pancreatic cells from human pluripotent stem cells (hPSCs) by testing gradient-pattern chips of nano-scale polystyrene surface structures with varying sizes and shapes. The optimal conditions for 3D differentiation of pancreatic cells were identified by assessing the expression of developmental regulators that are required for pancreatic islet development and maturation. Our results showed that the gradient chip of pore-part 2 (Po-2, 200-300 nm diameter) pattern was the most efficient setting to generate clusters of pancreatic endocrine progenitors (PDX1+ and NGN3+) compared to those of other pore diameters (Po-1, 100-200 or Po-3, 300-400 nm) tested across a range of pillar patterns and flat surfaces. Furthermore, the Po-2 gradient pattern-derived clusters generated islet-like 3D spheroids and tested positive for the zinc-chelating dye dithizone. The spheroids consisted of more than 30% CD200 + endocrine cells and also expressed NKX6.1 and NKX2.2. In addition, pancreatic β- cells expressing insulin and polyhormonal cells expressing both insulin and glucagon were obtained at the final stage of pancreatic differentiation. In conclusion, our data suggest that an optimal topographical structure for differentiation to specific cell types from hPSCs can be tested efficiently by using gradient-pattern chips designed with varying sizes and surfaces. STATEMENT OF SIGNIFICANCE: Our study provides demonstrates of using gradient nanopatterned chips for differentiation of pancreatic islet-like clusters. Gradient nanopatterned chips are consisted of two different shapes (nanopillar and nanopore) in three different ranges of nano sizes (100-200, 200-300, 300-400 nm). We found that optimal nanostructures for differentiation of pancreatic islet-like clusters were 200-300 nm nano pores. Cell transplantation is one of the major therapeutic option for type 1 diabetes mellitus (DM) using stem cell-derived β-like cells. We generated 50 um pancreatic islet-like clusters in size, which would be an optimal size for cell transplantation. Futuremore, the small clusters provide a powerful source for cell therapy. Our findings suggest gradient nanopatterned chip provides a powerful tool to generate specific functional cell types of a high purity for potential uses in cell therapy development.
Collapse
|
24
|
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
|
25
|
Reyes‐Martínez JE, Ruiz‐Pacheco JA, Flores‐Valdéz MA, Elsawy MA, Vallejo‐Cardona AA, Castillo‐Díaz LA. Advanced hydrogels for treatment of diabetes. J Tissue Eng Regen Med 2019; 13:1375-1393. [DOI: 10.1002/term.2880] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 03/31/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Juana E. Reyes‐Martínez
- Departamento de Biología. División de Ciencias Naturales y ExactasUniversidad de Guanajuato Guanajuato México
| | | | - Mario A. Flores‐Valdéz
- Biotecnología Médica y FarmacéuticaCentro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ) Guadalajara México
| | - Mohamed A. Elsawy
- School of Pharmacy and Biomedical SciencesUniversity of Central Lancashire Preston UK
| | - Alba A. Vallejo‐Cardona
- Biotecnología Médica y FarmacéuticaCentro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ) Guadalajara México
| | - Luis A. Castillo‐Díaz
- Departamento de Medicina y Ciencias de la Salud, División de Ciencias Biológicas y de la SaludUniversidad de Sonora Hermosillo México
| |
Collapse
|
26
|
A simple method for the generation of insulin producing cells from bone marrow mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2019; 55:462-471. [PMID: 31111346 DOI: 10.1007/s11626-019-00358-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 04/11/2019] [Indexed: 10/26/2022]
Abstract
To produce insulin-producing cells (IPCs) from bone marrow mesenchymal stem cells (BM-MSCs) using a simple and cost effective method. During the initial 7 days of three-dimensional (3D) culture, BM-MSCs were cultured on 1% agar or agarose to form multicellular spheroids. Spheroids and spheroid-derived single cells (SS and SSC, respectively) were cultured in the absence of any proteinaceous growth factor in a simple specific medium for a further 7 d. The insulin content of the differentiated cells was evaluated at the mRNA and protein levels. Furthermore, the expression of pancreatic beta cells-related genes other than INS as well as the in vitro responses of IPCs to different glucose concentrations were investigated. Cellular clusters generated on agar and SS conditions (agar+SS-IPCs) stained better with beta cell specific stains and were more reactive to serum-containing insulin reactive antibodies compared with agarose-SS-IPCs. Gene expression analysis revealed that in comparison to agarose + SS-IPCs, agar+SS-IPCs expressed significantly higher levels of INS-1, INS-2, PDX-1, NKX6.1, and XBP-1. Of interest, agar+SS-IPCs expressed 2215.3 ± 120.8-fold more INS-1 gene compared to BM-MSCs. The expression of β-cell associated genes was also higher in agar+SS-IPCs compared to the agar+SSC-IPCs. Moreover, the expression of INS-1 gene was significantly higher in agar+SS-IPCs compared with agar+SSC-IPCs after culture in media with high concentration of glucose. Compared to the most expensive and time-consuming protocols, 3D culture of MSCs on agar followed by 2D culture of cellular clusters in a minimally supplemented high glucose media produced highly potent IPCs which may pay the way to the treatment of diabetic patients.
Collapse
|
27
|
Small molecules and extrinsic factors promoting differentiation of stem cells into insulin-producing cells. ANNALES D'ENDOCRINOLOGIE 2019; 80:128-133. [DOI: 10.1016/j.ando.2018.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/14/2018] [Accepted: 11/05/2018] [Indexed: 12/26/2022]
|
28
|
Chemically defined and xenogeneic-free differentiation of human pluripotent stem cells into definitive endoderm in 3D culture. Sci Rep 2019; 9:996. [PMID: 30700818 PMCID: PMC6353891 DOI: 10.1038/s41598-018-37650-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
In vitro differentiation of human pluripotent stem cells (hPSCs) into definitive endoderm (DE) represents a key step towards somatic cells of lung, liver and pancreas. For future clinical applications, mass production of differentiated cells at chemically defined conditions and free of xenogeneic substances is envisioned. In this study we adapted our previously published two-dimensional (2D) DE induction protocol to three-dimensional (3D) static suspension culture in the absence of the xenogeneic extracellular matrix Matrigel. Next, fetal calf serum and bovine serum albumin present in the standard medium were replaced by a custom-made and xeno-free B-27. This yielded in a chemically defined and xenogeneic-free 3D culture protocol for differentiation of hPSCs into DE at efficiencies similar to standard 2D conditions. This novel protocol successfully worked with different hPSC lines including hESCs and hiPSCs maintained in two different stem cell media prior to differentiation. DE cells obtained by our novel BSA-free 3D protocol could be further differentiated into PDX1- or NKX6.1-expressing pancreatic progenitor cells. Notably, upon DE differentiation, we also identified a CXCR4+/NCAM+/EpCAMlow cell population with reduced DE marker gene expression. These CXCR4+/NCAM+/EpCAMlow cells emerge as a result of Wnt/beta-catenin hyperactivation via elevated CHIR-99021 concentrations and likely represent misspecified DE.
Collapse
|
29
|
Yabe SG, Fukuda S, Nishida J, Takeda F, Nashiro K, Okochi H. Induction of functional islet-like cells from human iPS cells by suspension culture. Regen Ther 2019; 10:69-76. [PMID: 30623004 PMCID: PMC6317273 DOI: 10.1016/j.reth.2018.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/06/2018] [Accepted: 11/27/2018] [Indexed: 02/06/2023] Open
Abstract
Introduction To complement islet transplantation for type1 diabetic patients, cell-based therapy using pluripotent stem cells such as ES cells and iPS cells is promising. Many papers have already reported the induction of pancreatic β cells from these cell types, but a suspension culture system has not usually been employed. The aim of this study is to establish a suspension culture method for inducing functional islet-like cells from human iPS cells. Methods We used 30 ml spinner type culture vessels for human iPS cells throughout the differentiation process. Differentiated cells were analyzed by immunostaining and C-peptide secretion. Cell transplantation experiments were performed with STZ-induced diabetic NOD/SCID mice. Blood human C-peptide and glucagon levels were measured serially in mice, and grafts were analyzed histologically. Results We obtained spherical pancreatic beta-like cells from human iPS cells and detected verifiable amounts of C-peptide secretion in vitro. We demonstrated reversal of hyperglycemia in diabetic model mice after transplantation of these cells, maintaining non-fasting blood glucose levels along with the human glycemic set point. We confirmed the secretion of human insulin and glucagon dependent on the blood glucose level in vivo. Immunohistological analysis revealed that grafted cells became α, β and δ cells in vivo. Conclusions These results suggest that differentiated cells derived from human iPS cells grown in suspension culture mature and function like pancreatic islets in vivo. Functional islet-like cells were induced from human iPS cells by suspension culture. They ameliorated hyperglycemia in diabetic mice and secreted human insulin and glucagon. Grafted cells became α, β and δ cells in vivo.
Collapse
Affiliation(s)
- Shigeharu G Yabe
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Satsuki Fukuda
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Junko Nishida
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Fujie Takeda
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Kiyoko Nashiro
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Hitoshi Okochi
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| |
Collapse
|
30
|
Aydin S, Sağraç D, Şahin F. Differentiation Potential of Mesenchymal Stem Cells into Pancreatic β-Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1247:135-156. [PMID: 32002800 DOI: 10.1007/5584_2019_476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stem cells having the capability to differentiate into other type of cells and renewing themselves, gained so much importance in recent years. Investigations in stem cells revealed that mesenchymal stem cells can successfully differentiate into other type of cells like adipocytes, hepatocytes, osteocytes, neurocytes and chondrocytes. In addition, these cells can also differentiate into insulin-producing beta cells. Insulin is a crucial hormone for glucose balance of the body. Insufficiency or unavailability of insulin is called diabetes. External insulin intake, as well as pancreas or islet transplantation, is the most basic treatment of diabetes. In vivo and in vitro studies demonstrate that stem cell therapy is also used in the cure of diabetes. Differentiation process of stem cells into beta cells releasing insulin is quite complicated. There are many different reports for the differentiation of stem cells in the literature. The success of differentiation of stem cells into beta cells depends on several factors like the source of stem cells, chemicals added into the differentiation medium and the duration of differentiation protocol. Distinct studies for the differentiation of stem cells into insulin-secreting cells are available in the literature. Moreover, thanks to the superior differentiation capacity of stem cells, they are being preferred in clinical studies. Stem cells were clinically used to heal diabetic ulcer, to increase c-peptide level and insulin secretion in both type 1 and type 2 diabetes. Mesenchymal stem cells having high differentiation potential to insulin-secreting cells are encouraging vehicles for both in vivo and in vitro studies together with clinical trials for diabetes mellitus.
Collapse
Affiliation(s)
- Safa Aydin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey.
| | - Derya Sağraç
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| |
Collapse
|
31
|
Lee S, Moon S, Oh JY, Seo EH, Kim YH, Jun E, Shim IK, Kim SC. Enhanced insulin production and reprogramming efficiency of mesenchymal stem cells derived from porcine pancreas using suitable induction medium. Xenotransplantation 2019; 26:e12451. [PMID: 30252163 DOI: 10.1111/xen.12451] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/23/2018] [Accepted: 06/26/2018] [Indexed: 01/21/2023]
Abstract
BACKGROUND Genetic reprogramming is a powerful method for altering cell properties and inducing differentiation. However, even if the same gene is reprogrammed, the results vary among cells. Therefore, a better possible strategy involves treating cells with factors that further stimulate differentiation while using stem cells with the same tissue origin. This study aimed to increase induction efficiency and insulin production in reprogrammed cells using a combination of factors that promote cell differentiation. METHODS Porcine pancreatic cells were cultured to obtain mesenchymal stem cells expressing pancreatic cell-specific markers through sequential passages. The characteristics of these cells were identified, and the M3 gene (Pdx1, Ngn3, MafA) was reprogrammed to induce differentiation into insulin-producing cells. Additionally, the differentiation efficiency of insulin-producing cells was compared by treating reprogrammed cells with a differentiation-promoting factor. RESULTS Mesenchymal stem cells isolated from porcine pancreatic tissues expressed exocrine cell markers, including amylase and cytokeratin 18, and most cells continuously expressed the beta cell transcription factors Ngn3 and NeuroD. Reprogramming of the M3 gene resulted in differentiation into insulin-producing cells. Moreover, significantly increased insulin and glucagon expressions were observed in the suitable induction medium, and the characteristic beta cell transcription factors Pdx1, Ngn3, and MafA were expressed at levels as high as those in pancreatic islet cells. CONCLUSIONS Differentiation into insulin-producing cells represents an alternative therapy for insufficient pancreatic islet cells when treating diabetes. Therefore, cells with the characteristics of the target cell should be used to improve differentiation efficiency by creating an environment that promotes reprogramming and differentiation.
Collapse
Affiliation(s)
- Song Lee
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soobin Moon
- Department of Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ju Yun Oh
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun Ha Seo
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yang Hee Kim
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eunsung Jun
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - In Kyoung Shim
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Song Cheol Kim
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
32
|
Hashemitabar M, Heidari E. Redefining the signaling pathways from pluripotency to pancreas development: In vitro β-cell differentiation. J Cell Physiol 2018; 234:7811-7827. [PMID: 30480819 DOI: 10.1002/jcp.27736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 10/22/2018] [Indexed: 02/06/2023]
Abstract
Pancreatic β-cells are destroyed by the immune system, in type 1 diabetes (T1D) and are impaired by glucose insensitivity in type 2 diabetes (T2D). Islet-cells transplantation is a promising therapeutic approach based on in vitro differentiation of pluripotent stem cells (PSCs) to insulin-producing cells (IPCs). According to evolutionary stages in β-cell development, there are several distinct checkpoints; each one has a unique characteristic, including definitive endoderm (DE), primitive gut (PG), posterior foregut (PF), pancreatic epithelium (PE), endocrine precursor (EP), and immature β-cells up to functional β-cells. A better understanding of the gene regulatory networks (GRN) and associated transcription factors in each specific developmental stage, guarantees the achievement of the next successful checkpoints and ensures an efficient β-cell differentiation procedure. The new findings in signaling pathways, related to the development of the pancreas are discussed here, including Wnt, Activin/Nodal, FGF, BMP, retinoic acid (RA), sonic hedgehog (Shh), Notch, and downstream regulators, required for β-cell commitment. We also summarized different approaches in the IPCs protocol to conceptually define a standardized system, leading to the creation of a reproducible method for β-cell differentiation. To normalize blood glucose level in diabetic mice, the replacement therapy in the early differentiation stage, such as EP stages was associated with better outcome when compared with the fully differentiated β-cells' graft.
Collapse
Affiliation(s)
- Mahmoud Hashemitabar
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomy and Embryology, Faculty of Medicine, Joundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elham Heidari
- Department of Anatomy and Embryology, Faculty of Medicine, Joundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
33
|
Astro V, Adamo A. Epigenetic Control of Endocrine Pancreas Differentiation in vitro: Current Knowledge and Future Perspectives. Front Cell Dev Biol 2018; 6:141. [PMID: 30410880 PMCID: PMC6209628 DOI: 10.3389/fcell.2018.00141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/04/2018] [Indexed: 12/11/2022] Open
Abstract
The raising worldwide prevalence of Type 1 and Type 2 diabetes mellitus (T1DM and T2DM) solicits the derivation of in vitro methods yielding mature and fully functional β-cells to be used in regenerative medicine. Several protocols to differentiate human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) into human pancreatic β-like cells have recently been developed. These methods, coupled with a bioengineering approach using biocompatible encapsulating devices, have recently led to experimental clinical trials showing great promises to ultimately end the battle of diabetic patients for managing hyperglycemia. However, in vitro differentiation protocols face the challenge of achieving homogenous population of mono-hormonal insulin-secreting mature β-cells. Major epigenetic events such as DNA methylation, post-translational modification of histones and non-coding RNAs expression, orchestrate physiological endocrine pancreas specification into α-, β-, γ-, and δ-cells, both in vivo and in vitro. The dysregulation of such epigenetic processes is associated to multiple pancreatic disorders including diabetes. Understanding the epigenomic and transcriptomic landscape underlying endocrine pancreas development could, therefore, improve in vitro differentiation methods. In this review, we summarize the most effective protocols for in vitro differentiation of hESCs/hiPSCs toward pancreatic β-cells and we discuss the current limitations in the derivation of functional glucose-responsive, insulin-releasing β-cells. Moreover, we focus on the main transcriptional and epigenetic events leading to pancreatic specification and on the applicative potential of novel epigenetic drugs for the establishment of innovative pharmacological therapeutic approaches.
Collapse
Affiliation(s)
- Veronica Astro
- Biological and Environmental Science and Engineering Division, KAUST Environmental Epigenetics Program, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Antonio Adamo
- Biological and Environmental Science and Engineering Division, KAUST Environmental Epigenetics Program, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| |
Collapse
|
34
|
Yagi Mendoza H, Yokoyama T, Tanaka T, Ii H, Yaegaki K. Regeneration of insulin-producing islets from dental pulp stem cells using a 3D culture system. Regen Med 2018; 13:673-687. [PMID: 30028236 DOI: 10.2217/rme-2018-0074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM In this study, we aimed to establish the differentiation protocol of dental pulp stem cells (DPSCs) into pancreatic islets using a 3D structure. MATERIALS & METHODS DPSCs were differentiated in a 3D culture system using a stepwise protocol. Expression of β-cell markers, glucose-stimulated insulin secretion, and PI3K/AKT and WNT pathways were compared between monolayer-cultured pancreatic cells and islets. RESULTS Islet formation increased insulin and C-peptide production, and enhanced the expression of pancreatic markers. Glucose-dependent secretion of insulin was increased by islets. Pancreatic endocrine markers, transcriptional factors, and the PI3K/AKT and WNT pathways were also upregulated. CONCLUSION Pancreatic islets were generated from DPSCs in a 3D culture system. This system could provide novel strategies for controlling diabetes through regenerative medicine.
Collapse
Affiliation(s)
- Hiromi Yagi Mendoza
- Department of Oral Health, School of Life Dentistry at Tokyo, Nippon Dental University, 1-9-20, Fujimi, Chiyoda ku, 102-8159 Tokyo, Japan
| | - Tomomi Yokoyama
- Department of Oral Health, School of Life Dentistry at Tokyo, Nippon Dental University, 1-9-20, Fujimi, Chiyoda ku, 102-8159 Tokyo, Japan
| | - Tomoko Tanaka
- Department of Oral Health, School of Life Dentistry at Tokyo, Nippon Dental University, 1-9-20, Fujimi, Chiyoda ku, 102-8159 Tokyo, Japan
| | - Hisataka Ii
- Department of Oral Health, School of Life Dentistry at Tokyo, Nippon Dental University, 1-9-20, Fujimi, Chiyoda ku, 102-8159 Tokyo, Japan
| | - Ken Yaegaki
- Department of Oral Health, School of Life Dentistry at Tokyo, Nippon Dental University, 1-9-20, Fujimi, Chiyoda ku, 102-8159 Tokyo, Japan
| |
Collapse
|
35
|
Ghazalli N, Wu X, Walker S, Trieu N, Hsin LY, Choe J, Chen C, Hsu J, LeBon J, Kozlowski MT, Rawson J, Tirrell DA, Yip MLR, Ku HT. Glucocorticoid Signaling Enhances Expression of Glucose-Sensing Molecules in Immature Pancreatic Beta-Like Cells Derived from Murine Embryonic Stem Cells In Vitro. Stem Cells Dev 2018; 27:898-909. [PMID: 29717618 DOI: 10.1089/scd.2017.0160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Pluripotent stem cells may serve as an alternative source of beta-like cells for replacement therapy of type 1 diabetes; however, the beta-like cells generated in many differentiation protocols are immature. The maturation of endogenous beta cells involves an increase in insulin expression starting in late gestation and a gradual acquisition of the abilities to sense glucose and secrete insulin by week 2 after birth in mice; however, what molecules regulate these maturation processes are incompletely known. In this study, we aim to identify small molecules that affect immature beta cells. A cell-based assay, using pancreatic beta-like cells derived from murine embryonic stem (ES) cells harboring a transgene containing an insulin 1-promoter driven enhanced green fluorescent protein reporter, was used to screen a compound library (NIH Clinical Collection-003). Cortisone, a glucocorticoid, was among five positive hit compounds. Quantitative reverse transcription-polymerase chain reaction analysis revealed that glucocorticoids enhance the gene expression of not only insulin 1 but also glucose transporter-2 (Glut2; Slc2a2) and glucokinase (Gck), two molecules important for glucose sensing. Mifepristone, a pharmacological inhibitor of glucocorticoid receptor (GR) signaling, reduced the effects of glucocorticoids on Glut2 and Gck expression. The effects of glucocorticoids on ES-derived cells were further validated in immature primary islets. Isolated islets from 1-week-old mice had an increased Glut2 and Gck expression in response to a 4-day treatment of exogenous hydrocortisone in vitro. Gene deletion of GR in beta cells using rat insulin 2 promoter-driven Cre crossed with GRflox/flox mice resulted in a reduced gene expression of Glut2, but not Gck, and an abrogation of insulin secretion when islets were incubated in 0.5 mM d-glucose and stimulated by 17 mM d-glucose in vitro. These results demonstrate that glucocorticoids positively regulate glucose sensors in immature murine beta-like cells.
Collapse
Affiliation(s)
- Nadiah Ghazalli
- 1 Department of Translational Research and Cellular Therapeutics, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
- 2 The Irell and Manella Graduate School of Biological Sciences, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
- 3 Faculty of Medicine and Health Sciences, Genetics and Regenerative Medicine Research Center, Universiti Putra Malaysia , Serdang, Malaysia
| | - Xiaoxing Wu
- 1 Department of Translational Research and Cellular Therapeutics, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| | - Stephanie Walker
- 1 Department of Translational Research and Cellular Therapeutics, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| | - Nancy Trieu
- 1 Department of Translational Research and Cellular Therapeutics, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| | - Li-Yu Hsin
- 4 High Throughput Screening Core, Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| | - Justin Choe
- 1 Department of Translational Research and Cellular Therapeutics, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| | - Chialin Chen
- 1 Department of Translational Research and Cellular Therapeutics, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| | - Jasper Hsu
- 1 Department of Translational Research and Cellular Therapeutics, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| | - Jeanne LeBon
- 1 Department of Translational Research and Cellular Therapeutics, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| | - Mark T Kozlowski
- 5 Division of Chemistry and Chemical Engineering, California Institute of Technology , Pasadena, California
| | - Jeffrey Rawson
- 1 Department of Translational Research and Cellular Therapeutics, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| | - David A Tirrell
- 5 Division of Chemistry and Chemical Engineering, California Institute of Technology , Pasadena, California
| | - M L Richard Yip
- 4 High Throughput Screening Core, Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| | - Hsun Teresa Ku
- 1 Department of Translational Research and Cellular Therapeutics, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
- 2 The Irell and Manella Graduate School of Biological Sciences, and Diabetes and Metabolism Research Institute and Beckman Research Institute of City of Hope , Duarte, California
| |
Collapse
|
36
|
Aigha II, Memon B, Elsayed AK, Abdelalim EM. Differentiation of human pluripotent stem cells into two distinct NKX6.1 populations of pancreatic progenitors. Stem Cell Res Ther 2018; 9:83. [PMID: 29615106 PMCID: PMC5883581 DOI: 10.1186/s13287-018-0834-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 03/09/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The expression of a specific combination of transcription factors (TFs) in the multipotent progenitor cells (MPCs) is critical for determining pancreatic cell fate. NKX6.1 expression in PDX1+ MPCs is required for functional β cell generation. We have recently demonstrated the generation of a novel population of human pluripotent stem cell (hPSC)-derived MPCs that exclusively express NKX6.1, independently of PDX1 (PDX1-/NKX6.1+). Therefore, the aim of this study was to characterize this novel population to elucidate its role in pancreatic development. METHODS The hPSCs were exposed to two differentiation protocols to generate MPCs that were analyzed using different techniques. RESULTS Based on the expression of PDX1 and NKX6.1, we generated three different populations of MPCs, two of them were NKX6.1+. One of these NKX6.1 populations coexpressed PDX1 (PDX1+/NKX6.1+) which is known to mature into functional β cells, and an additional novel population did not express PDX1 (PDX1-/NKX6.1+) with an undefined role in pancreatic cell fate. This novel population was enriched using our recently established protocol, allowing their reorganization in three-dimensional (3D) structures. Since NKX6.1 induction in MPCs can direct them to endocrine and/or ductal cells in humans, we examined the coexpression of endocrine and ductal markers. We found that the expression of the pancreatic endocrine progenitor markers chromogranin A (CHGA) and neurogenin 3 (NGN3) was not detected in the NKX6.1+ 3D structures, while few structures were positive for NKX2.2, another endocrine progenitor marker, thereby shedding light on the origin of this novel population and its role in pancreatic endocrine development. Furthermore, SOX9 was highly expressed in the 3D structures, but cytokeratin 19, a main ductal marker, was not detected in these structures. CONCLUSIONS These data support the existence of two independent NKX6.1+ MPC populations during human pancreatic development and the novel PDX1-/NKX6.1+ population may be involved in a unique trajectory to generate β cells in humans.
Collapse
Affiliation(s)
- Idil I Aigha
- 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
| | - Ahmed K Elsayed
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Education City, Doha, Qatar.,Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Essam M Abdelalim
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Education City, Doha, Qatar.
| |
Collapse
|
37
|
Loo LSW, Lau HH, Jasmen JB, Lim CS, Teo AKK. An arduous journey from human pluripotent stem cells to functional pancreatic β cells. Diabetes Obes Metab 2018; 20:3-13. [PMID: 28474496 DOI: 10.1111/dom.12996] [Citation(s) in RCA: 22] [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: 03/02/2017] [Revised: 04/29/2017] [Accepted: 05/01/2017] [Indexed: 12/11/2022]
Abstract
Type 1 and type 2 diabetes are caused by a destruction and decrease in the number of functional insulin-producing β cells, respectively; therefore, the generation of functional β cells from human embryonic stem cells and human induced pluripotent stem cells, collectively known as human pluripotent stem cells (hPSCs), for potential cell replacement therapy and disease modelling is an intensely investigated area. Recent scientific breakthroughs enabled derivation of large quantities of human pancreatic β-like cells in vitro, although with varied glucose-stimulated insulin secretion kinetics. In the present review, we comprehensively summarize, compare and critically analyze the intricacies of these developing technologies, including differentiation platforms, robustness of protocols, and methodologies used to characterize hPSC-derived β-like cells. We also discuss experimental issues that need to be resolved before these β-like cells can be used clinically.
Collapse
Affiliation(s)
- Larry Sai Weng Loo
- Programme in Stem Cell, Regenerative Medicine and Ageing, Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Hwee Hui Lau
- Programme in Stem Cell, Regenerative Medicine and Ageing, Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Joanita Binte Jasmen
- Programme in Stem Cell, Regenerative Medicine and Ageing, Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Chang Siang Lim
- Programme in Stem Cell, Regenerative Medicine and Ageing, Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Adrian Kee Keong Teo
- Programme in Stem Cell, Regenerative Medicine and Ageing, Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
38
|
Nadri S, Barati G, Mostafavi H, Esmaeilzadeh A, Enderami SE. Differentiation of conjunctiva mesenchymal stem cells into secreting islet beta cells on plasma treated electrospun nanofibrous scaffold. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:178-187. [DOI: 10.1080/21691401.2017.1416391] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Samad Nadri
- Metabolic Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ghasem Barati
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Mostafavi
- Department of Physiology and Pharmacology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology & Cancer Gene therapy Research Center, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Ehsan Enderami
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
39
|
Tu C, Xu R, Koleti M, Zoldan J. Glycogen synthase kinase-3 inhibition sensitizes human induced pluripotent stem cells to thiol-containing antioxidants induced apoptosis. Stem Cell Res 2017; 23:182-187. [DOI: 10.1016/j.scr.2017.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/20/2017] [Accepted: 07/19/2017] [Indexed: 01/22/2023] Open
|
40
|
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
|
41
|
Mao GH, Lu P, Wang YN, Tian CG, Huang XH, Feng ZG, Zhang JL, Chang HY. Role of PI3K p110β in the differentiation of human embryonic stem cells into islet-like cells. Biochem Biophys Res Commun 2017; 488:109-115. [PMID: 28479244 DOI: 10.1016/j.bbrc.2017.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 05/03/2017] [Indexed: 12/16/2022]
Abstract
To investigate the effects of the PI3K inhibitors on the differentiation of insulin-producing cells derived from human embryonic stem cells. Here, we report that human embryonic stem cells induced by phosphatidylinositol-3-kinase (PI3K) p110β inhibitors could produce more mature islet-like cells. Findings were validated by immunofluorescence analysis, quantitative real-time PCR, insulin secretion in vitro and cell transplantation for the diabetic SCID mice. Immunofluorescence analysis revealed that unihormonal insulin-positive cells were predominant in cultures with rare polyhormonal cells. Real-time PCR data showed that islet-like cells expressed key markers of pancreatic endocrine hormones and mature pancreatic β cells including MAFA. Furthermore, this study showed that the expression of most pancreatic endocrine hormones was similar between groups treated with the LY294002 (nonselective PI3K inhibitor) and TGX-221 (PI3K isoform selective inhibitors of class 1β) derivatives. However, the level of insulin mRNA in TGX-221-treated cells was significantly higher than that in LY294002-treated cells. In addition, islet-like cells displayed glucose-stimulated insulin secretion in vitro. After transplantation, islet-like cells improved glycaemic control and ameliorated the survival outcome in diabetic mice. This study demonstrated an important role for PI3K p110β in regulating the differentiation and maturation of islet-like cells derived from human embryonic stem cells.
Collapse
Affiliation(s)
- Gen-Hong Mao
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China.
| | - Ping Lu
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Ya-Nan Wang
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Chen-Guang Tian
- Department of Endocrinology and Metabolic Diseases, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Xiao-Hui Huang
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Zong-Gang Feng
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Jin-Lan Zhang
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Hong-Yang Chang
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| |
Collapse
|
42
|
Mihara Y, Matsuura K, Sakamoto Y, Okano T, Kokudo N, Shimizu T. Production of pancreatic progenitor cells from human induced pluripotent stem cells using a three-dimensional suspension bioreactor system. J Tissue Eng Regen Med 2017; 11:3193-3201. [DOI: 10.1002/term.2228] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 04/11/2016] [Accepted: 04/25/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Yuichiro Mihara
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; Tokyo Japan
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; Tokyo Japan
| | - Yoshihiro Sakamoto
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; Tokyo Japan
| | - Norihiro Kokudo
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine; The University of Tokyo; Tokyo Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; Tokyo Japan
| |
Collapse
|
43
|
Yabe SG, Fukuda S, Takeda F, Nashiro K, Shimoda M, Okochi H. Efficient generation of functional pancreatic β-cells from human induced pluripotent stem cells. J Diabetes 2017; 9:168-179. [PMID: 27038181 DOI: 10.1111/1753-0407.12400] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 03/13/2016] [Accepted: 03/25/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Insulin-secreting cells have been generated from human embryonic or induced pluripotent stem cells (iPSCs) by mimicking developmental processes. However, these cells do not always secrete glucose-responsive insulin, one of the most important characteristics of pancreatic β-cells. We focused on the importance of endodermal differentiation from human iPSCs in order to obtain functional pancreatic β-cells. METHODS A six-stage protocol was established for the differentiation of human iPSCs to pancreatic β-cells using defined culture media without feeders or serum. The effects of CHIR99021, a selective glycogen synthase kinase-3β inhibitor, were examined in the presence of fibroblast growth factor 2, activin, and bone morphogenetic protein 4 (FAB) during definitive endodermal induction by immunostaining for SRY (sex determining region Y)-box 17 (SOX17) and Forkhead box protein A2 (FOXA2). Insulin secretion was compared between the last stage of monolayer culture and spheroid culture conditions. Cultured cells were transplanted under kidney capsules of streptozotocin-diabetic non-obese diabetic-severe combined immunodeficiency mice, and blood glucose levels were measured once a week. Immunohistochemical analyses were performed 4 and 12 weeks after transplantation. RESULTS Addition of CHIR99021 (3 μmol/L) in the presence of FAB for 2 days improved endodermal cell viability, maintaining the high SOX17-positive rate. Spheroid formation after the endocrine progenitor stage showed more efficient insulin secretion than did monolayer culture. After cell transplantation, diabetic mice had lower blood glucose levels, and islet-like structures were detected in vivo. CONCLUSION Functional pancreatic β-cells were generated from human iPSCs. Induction of definitive endoderm and spheroid formation may be key steps for producing these cells.
Collapse
MESH Headings
- Activins/pharmacology
- Animals
- Bone Morphogenetic Protein 4/pharmacology
- Cell Culture Techniques/methods
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cells, Cultured
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/therapy
- Endoderm/cytology
- Endoderm/drug effects
- Endoderm/metabolism
- Fibroblast Growth Factor 2/pharmacology
- Gene Expression/drug effects
- Hepatocyte Nuclear Factor 3-beta/genetics
- Hepatocyte Nuclear Factor 3-beta/metabolism
- Humans
- Immunohistochemistry
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/metabolism
- Induced Pluripotent Stem Cells/transplantation
- Insulin/genetics
- Insulin/metabolism
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Male
- Mice, Inbred NOD
- Mice, SCID
- Pyridines/pharmacology
- Pyrimidines/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- SOXF Transcription Factors/genetics
- SOXF Transcription Factors/metabolism
- Spheroids, Cellular/cytology
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/metabolism
- Stem Cell Transplantation/methods
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Shigeharu G Yabe
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Satsuki Fukuda
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Fujie Takeda
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kiyoko Nashiro
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Pancreatic Islet Transplantation Project, Department of Diabetes Research Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hitoshi Okochi
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
44
|
Al Madhoun A, Ali H, AlKandari S, Atizado VL, Akhter N, Al-Mulla F, Atari M. Defined three-dimensional culture conditions mediate efficient induction of definitive endoderm lineage from human umbilical cord Wharton's jelly mesenchymal stem cells. Stem Cell Res Ther 2016; 7:165. [PMID: 27852316 PMCID: PMC5111269 DOI: 10.1186/s13287-016-0426-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 10/18/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) are gaining increasing interest as an alternative source of stem cells for regenerative medicine applications. Definitive endoderm (DE) specification is a prerequisite for the development of vital organs such as liver and pancreas. Hence, efficient induction of the DE lineage from stem cells is crucial for subsequent generation of clinically relevant cell types. Here we present a defined 3D differentiation protocol of WJ-MSCs into DE cells. METHODS WJ-MSCs were cultured in suspension to generate spheroids, about 1500 cells each, for 7 days. The serum-free differentiation media contained specific growth factors, cytokines, and small molecules that specifically regulate signaling pathways including sonic hedgehog, bone morphogenetic protein, Activin/Wnt, and Notch. RESULTS We obtained more than 85 % DE cells as shown with FACS analysis using antibodies directed against the DE marker CXCR4. In addition, biochemical and molecular analysis of bona-fide DE markers revealed a time-course induction of Sox17, CXCR4, and FoxA2. Focused PCR-based array also indicated a specific induction into the DE lineage. CONCLUSIONS In this study, we report an efficient serum-free protocol to differentiate WJ-MSCs into DE cells utilizing 3D spheroid formation. Our approach might aid in the development of new protocols to obtain DE-derivative lineages including liver-like and pancreatic insulin-producing cells.
Collapse
Affiliation(s)
| | - Hamad Ali
- Research Division, Dasman Diabetes Institute, 1180 Dasman, Kuwait
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center, Kuwait University, Al-Jabriya, Kuwait
| | - Sarah AlKandari
- Research Division, Dasman Diabetes Institute, 1180 Dasman, Kuwait
| | | | - Nadeem Akhter
- Research Division, Dasman Diabetes Institute, 1180 Dasman, Kuwait
| | - Fahd Al-Mulla
- Department of Pathology, Molecular Pathology Unit, Faculty of Medicine, Health Sciences Center, Kuwait University, Al-Jabriya, Kuwait
| | - Maher Atari
- UIC Regenerative Medicine Research Institute, International University of Catalonia, Barcelona, Spain
| |
Collapse
|
45
|
Johnson JD. The quest to make fully functional human pancreatic beta cells from embryonic stem cells: climbing a mountain in the clouds. Diabetologia 2016; 59:2047-57. [PMID: 27473069 DOI: 10.1007/s00125-016-4059-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/23/2016] [Indexed: 01/10/2023]
Abstract
The production of fully functional insulin-secreting cells to treat diabetes is a major goal of regenerative medicine. In this article, I review progress towards this goal over the last 15 years from the perspective of a beta cell biologist. I describe the current state-of-the-art, and speculate on the general approaches that will be required to identify and achieve our ultimate goal of producing functional beta cells. The need for deeper phenotyping of heterogeneous cultures of stem cell derived islet-like cells in parallel with a better understanding of the heterogeneity of the target cell type(s) is emphasised. This deep phenotyping should include high-throughput single-cell analysis, as well as comprehensive 'omics technologies to provide unbiased characterisation of cell products and human beta cells. There are justified calls for more detailed and well-powered studies of primary human pancreatic beta cell physiology, and I propose online databases of standardised human beta cell responses to physiological stimuli, including both functional and metabolomic/proteomic/transcriptomic profiles. With a concerted, community-wide effort, including both basic and applied scientists, beta cell replacement will become a clinical reality for patients with diabetes.
Collapse
Affiliation(s)
- James D Johnson
- Diabetes Research Group, Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, 5358-2350 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3.
| |
Collapse
|
46
|
Borisov MA, Petrakova OS, Gvazava IG, Kalistratova EN, Vasiliev AV. Stem Cells in the Treatment of Insulin-Dependent Diabetes Mellitus. Acta Naturae 2016; 8:31-43. [PMID: 27795842 PMCID: PMC5081704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Indexed: 11/02/2022] Open
Abstract
Diabetes affects over 350 million people worldwide, with the figure projected to rise to nearly 500 million over the next 20 years, according to the World Health Organization. Insulin-dependent diabetes mellitus (type 1 diabetes) is an endocrine disorder caused by an autoimmune reaction that destroys insulin-producing β-cells in the pancreas, which leads to insulin deficiency. Administration of exogenous insulin remains at the moment the treatment mainstay. This approach helps to regulate blood glucose levels and significantly increases the life expectancy of patients. However, type 1 diabetes is accompanied by long-term complications associated with the systemic nature of the disease and metabolic abnormalities having a profound impact on health. Of greater impact would be a therapeutic approach which would overcome these limitations by better control of blood glucose levels and prevention of acute and chronic complications. The current efforts in the field of regenerative medicine are aimed at finding such an approach. In this review, we discuss the time-honored technique of donor islets of Langerhans transplantation. We also focus on the use of pluripotent stem and committed cells and cellular reprogramming. The molecular mechanisms of pancreatic differentiation are highlighted. Much attention is devoted to the methods of grafts delivery and to the materials used during its creation.
Collapse
Affiliation(s)
- M. A. Borisov
- Pirogov Russian National Research Medical University, Ostrovitianov str. 1, Moscow, 117997, Russia
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova str. 26, Moscow, 119334, Russia
| | - O. S. Petrakova
- Pirogov Russian National Research Medical University, Ostrovitianov str. 1, Moscow, 117997, Russia
- Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bld. 12, Moscow, 119991 , Russia
| | - I. G. Gvazava
- Pirogov Russian National Research Medical University, Ostrovitianov str. 1, Moscow, 117997, Russia
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova str. 26, Moscow, 119334, Russia
| | - E. N. Kalistratova
- Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bld. 12, Moscow, 119991 , Russia
| | - A. V. Vasiliev
- Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, bld. 12, Moscow, 119991 , Russia
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova str. 26, Moscow, 119334, Russia
| |
Collapse
|
47
|
Kim JH, Kim HW, Cha KJ, Han J, Jang YJ, Kim DS, Kim JH. Nanotopography Promotes Pancreatic Differentiation of Human Embryonic Stem Cells and Induced Pluripotent Stem Cells. ACS NANO 2016; 10:3342-55. [PMID: 26900863 DOI: 10.1021/acsnano.5b06985] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Although previous studies suggest that nanotopographical features influence properties and behaviors of stem cells, only a few studies have attempted to derive clinically useful somatic cells from human pluripotent stem cells using nanopatterned surfaces. In the present study, we report that polystyrene nanopore-patterned surfaces significantly promote the pancreatic differentiation of human embryonic and induced pluripotent stem cells. We compared different diameters of nanopores and showed that 200 nm nanopore-patterned surfaces highly upregulated the expression of PDX1, a critical transcription factor for pancreatic development, leading to an approximately 3-fold increase in the percentage of differentiating PDX1(+) pancreatic progenitors compared with control flat surfaces. Furthermore, in the presence of biochemical factors, 200 nm nanopore-patterned surfaces profoundly enhanced the derivation of pancreatic endocrine cells producing insulin, glucagon, or somatostatin. We also demonstrate that nanopore-patterned surface-induced upregulation of PDX1 is associated with downregulation of TAZ, suggesting the potential role of TAZ in nanopore-patterned surface-mediated mechanotransduction. Our study suggests that appropriate cytokine treatments combined with nanotopographical stimulation could be a powerful tool for deriving a high purity of desired cells from human pluripotent stem cells.
Collapse
Affiliation(s)
- Jong Hyun Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Science Campus, Korea University , 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| | - Hyung Woo Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) , 77 Cheongam-Ro, Nam-gu, Pohang, Gyeongbuk 37673, Korea
| | - Kyoung Je Cha
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) , 77 Cheongam-Ro, Nam-gu, Pohang, Gyeongbuk 37673, Korea
| | - Jiyou Han
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Science Campus, Korea University , 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| | - Yu Jin Jang
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Science Campus, Korea University , 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) , 77 Cheongam-Ro, Nam-gu, Pohang, Gyeongbuk 37673, Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Science Campus, Korea University , 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| |
Collapse
|
48
|
Bartlett ST, Markmann JF, Johnson P, Korsgren O, Hering BJ, Scharp D, Kay TWH, Bromberg J, Odorico JS, Weir GC, Bridges N, Kandaswamy R, Stock P, Friend P, Gotoh M, Cooper DKC, Park CG, O'Connell P, Stabler C, Matsumoto S, Ludwig B, Choudhary P, Kovatchev B, Rickels MR, Sykes M, Wood K, Kraemer K, Hwa A, Stanley E, Ricordi C, Zimmerman M, Greenstein J, Montanya E, Otonkoski T. Report from IPITA-TTS Opinion Leaders Meeting on the Future of β-Cell Replacement. Transplantation 2016; 100 Suppl 2:S1-44. [PMID: 26840096 PMCID: PMC4741413 DOI: 10.1097/tp.0000000000001055] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/07/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Stephen T. Bartlett
- Department of Surgery, University of Maryland School of Medicine, Baltimore MD
| | - James F. Markmann
- Division of Transplantation, Massachusetts General Hospital, Boston MA
| | - Paul Johnson
- Nuffield Department of Surgical Sciences and Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bernhard J. Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - David Scharp
- Prodo Laboratories, LLC, Irvine, CA
- The Scharp-Lacy Research Institute, Irvine, CA
| | - Thomas W. H. Kay
- Department of Medicine, St. Vincent’s Hospital, St. Vincent's Institute of Medical Research and The University of Melbourne Victoria, Australia
| | - Jonathan Bromberg
- Division of Transplantation, Massachusetts General Hospital, Boston MA
| | - Jon S. Odorico
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI
| | - Gordon C. Weir
- Joslin Diabetes Center and Harvard Medical School, Boston, MA
| | - Nancy Bridges
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Raja Kandaswamy
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Peter Stock
- Division of Transplantation, University of San Francisco Medical Center, San Francisco, CA
| | - Peter Friend
- Nuffield Department of Surgical Sciences and Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Mitsukazu Gotoh
- Department of Surgery, Fukushima Medical University, Fukushima, Japan
| | - David K. C. Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
| | - Chung-Gyu Park
- Xenotransplantation Research Center, Department of Microbiology and Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Phillip O'Connell
- The Center for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Westmead, NSW, Australia
| | - Cherie Stabler
- Diabetes Research Institute, School of Medicine, University of Miami, Coral Gables, FL
| | - Shinichi Matsumoto
- National Center for Global Health and Medicine, Tokyo, Japan
- Otsuka Pharmaceutical Factory inc, Naruto Japan
| | - Barbara Ludwig
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden and DZD-German Centre for Diabetes Research, Dresden, Germany
| | - Pratik Choudhary
- Diabetes Research Group, King's College London, Weston Education Centre, London, United Kingdom
| | - Boris Kovatchev
- University of Virginia, Center for Diabetes Technology, Charlottesville, VA
| | - Michael R. Rickels
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Coulmbia University Medical Center, New York, NY
| | - Kathryn Wood
- Nuffield Department of Surgical Sciences and Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Kristy Kraemer
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Albert Hwa
- Juvenile Diabetes Research Foundation, New York, NY
| | - Edward Stanley
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Monash University, Melbourne, VIC, Australia
| | - Camillo Ricordi
- Diabetes Research Institute, School of Medicine, University of Miami, Coral Gables, FL
| | - Mark Zimmerman
- BetaLogics, a business unit in Janssen Research and Development LLC, Raritan, NJ
| | - Julia Greenstein
- Discovery Research, Juvenile Diabetes Research Foundation New York, NY
| | - Eduard Montanya
- Bellvitge Biomedical Research Institute (IDIBELL), Hospital Universitari Bellvitge, CIBER of Diabetes and Metabolic Diseases (CIBERDEM), University of Barcelona, Barcelona, Spain
| | - Timo Otonkoski
- Children's Hospital and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| |
Collapse
|
49
|
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]
|
50
|
Tasnim F, Phan D, Toh YC, Yu H. Cost-effective differentiation of hepatocyte-like cells from human pluripotent stem cells using small molecules. Biomaterials 2015; 70:115-25. [PMID: 26310107 DOI: 10.1016/j.biomaterials.2015.08.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 08/01/2015] [Indexed: 12/20/2022]
Abstract
Significant efforts have been invested into the differentiation of stem cells into functional hepatocyte-like cells that can be used for cell therapy, disease modeling and drug screening. Most of these efforts have been concentrated on the use of growth factors to recapitulate developmental signals under in vitro conditions. Using small molecules instead of growth factors would provide an attractive alternative since small molecules are cell-permeable and cheaper than growth factors. We have developed a protocol for the differentiation of human embryonic stem cells into hepatocyte-like cells using a predominantly small molecule-based approach (SM-Hep). This 3 step differentiation strategy involves the use of optimized concentrations of LY294002 and bromo-indirubin-3'-oxime (BIO) for the generation of definitive endoderm; sodium butyrate and dimethyl sulfoxide (DMSO) for the generation of hepatoblasts and SB431542 for differentiation into hepatocyte-like cells. Activin A is the only growth factor required in this protocol. Our results showed that SM-Hep were morphologically and functionally similar or better compared to the hepatocytes derived from the growth-factor induced differentiation (GF-Hep) in terms of expression of hepatic markers, urea and albumin production and cytochrome P450 (CYP1A2 and CYP3A4) activities. Cell viability assays following treatment with paradigm hepatotoxicants Acetaminophen, Chlorpromazine, Diclofenac, Digoxin, Quinidine and Troglitazone showed that their sensitivity to these drugs was similar to human primary hepatocytes (PHHs). Using SM-Hep would result in 67% and 81% cost reduction compared to GF-Hep and PHHs respectively. Therefore, SM-Hep can serve as a robust and cost effective replacement for PHHs for drug screening and development.
Collapse
Affiliation(s)
- Farah Tasnim
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Derek Phan
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Yi-Chin Toh
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Hanry Yu
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, MD9-03-03, 2 Medical Drive, Singapore 117597, Singapore; NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, #05-01, 28 Medical Drive, Singapore 117576, Singapore; Mechanobiology Institute, T-Labs, #05-01, 5A Engineering Drive 1, Singapore 117411, Singapore; Singapore-MIT Alliance for Research and Technology, 3 Science Drive 2, S16-05-08, Singapore 117543, Singapore; Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA.
| |
Collapse
|