1
|
Tan WS, Lai Y, Chung Y, Adusumalli S, Lee XY, Tryggvason K, Tay HG. Retina-specific laminin-based generation of photoreceptor progenitors from human pluripotent stem cells under xeno-free and chemically defined conditions. Nat Protoc 2025:10.1038/s41596-025-01142-y. [PMID: 40000780 DOI: 10.1038/s41596-025-01142-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/07/2025] [Indexed: 02/27/2025]
Abstract
Photoreceptor cell replacement therapy for retinal degenerative diseases is a promising approach. Presently, most protocols aimed at generating clinically safe and functional cells for retinal diseases face challenges such as low efficiency, poor reproducibility, and time-consuming and complex procedures. These could be due to the dependency on animal-derived components in cell culture media and substrates that support the cell differentiation process. Such conditions are poorly defined chemically, which could affect the robustness of the method and hinder clinical translation of cell therapy in retinal diseases. Here, we describe a simple protocol that is xenogen free and chemically defined to differentiate human embryonic stem cells to photoreceptor progenitors. Human recombinant extracellular matrix laminin 523 and 521 isoforms were used to mimic the inter-photoreceptor matrix niche environment to promote the retinal cell differentiation process. This was also accomplished by the unique combination of two cell differentiation media that recapitulates the retinal development signaling processes. In comparison to other protocols, our protocol does not require any mechanical dissection, which can be technically subjective and tedious. Our directed differentiation method generates photoreceptor progenitors that express ~17% cone-rod homeobox (CRX) transcript based on single-cell transcriptomic analyses by day 32. These day 32 photoreceptor progenitors can be cryopreserved and still maintain high cell viability after thawing for cell transplantation. This protocol can be easily reproduced and performed by researchers with basic cell culture experience, which is particularly important for retinal research progress and clinical cell manufacturing in a Good Manufacturing Practice facility.
Collapse
Affiliation(s)
- Wei Sheng Tan
- Centre for Vision Research, Duke-NUS Medical School, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yixin Lai
- Centre for Vision Research, Duke-NUS Medical School, Singapore, Singapore
| | - Yingying Chung
- Centre for Vision Research, Duke-NUS Medical School, Singapore, Singapore
| | | | - Xin Yi Lee
- Centre for Vision Research, Duke-NUS Medical School, Singapore, Singapore
| | - Karl Tryggvason
- Centre for Vision Research, Duke-NUS Medical School, Singapore, Singapore
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
- Division of Nephrology, Department of Medicine, Duke University, Durham, NC, USA
| | - Hwee Goon Tay
- Centre for Vision Research, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
2
|
Sato K, Koyanagi-Aoi M, Uehara K, Yamashita Y, Shinohara M, Lee S, Reinhardt A, Woltjen K, Chiba K, Miyake H, Fujisawa M, Aoi T. Efficient differentiation of human iPSCs into Leydig-like cells capable of long-term stable secretion of testosterone. Stem Cell Reports 2025; 20:102392. [PMID: 39824187 PMCID: PMC11864132 DOI: 10.1016/j.stemcr.2024.102392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/20/2025] Open
Abstract
Late-onset hypogonadism (LOH) syndrome is characterized by age-related testosterone deficiency and negatively affects the quality of life of older men. A promising therapeutic approach for LOH syndrome is transplantation of testosterone-producing Leydig-like cells (LLCs) derived from human induced pluripotent stem cells (hiPSCs). However, previous studies have encountered obstacles, such as limited cell longevity, insufficient testosterone production, and inefficiency of differentiation. To address these issues, we developed a novel protocol that includes forced NR5A1 expression, a cytokine cocktail promoting mesoderm differentiation, and a transitional shift from 3D to 2D cultures. The resultant cells survived on culture dishes for over 16 weeks, produced 22-fold more testosterone than the conventional method, and constituted a homogeneous population of LLCs with a differentiation efficiency exceeding 99% without purification. Furthermore, these LLCs were successfully engrafted subcutaneously into mice, resulting in increased serum testosterone levels. Our study will facilitate innovative therapeutic strategies for LOH syndrome.
Collapse
Affiliation(s)
- Katsuya Sato
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan; Division of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Michiyo Koyanagi-Aoi
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan; Center for Human Resource Development for Regenerative Medicine, Kobe University Hospital, Kobe, Japan
| | - Keiichiro Uehara
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan; Department of Diagnostic Pathology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yosuke Yamashita
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan; Division of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Masakazu Shinohara
- The Integrated Center for Mass Spectrometry, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Molecular Epidemiology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Suji Lee
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Anika Reinhardt
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Knut Woltjen
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Koji Chiba
- Division of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Hideaki Miyake
- Division of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Masato Fujisawa
- Division of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Takashi Aoi
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan; Center for Human Resource Development for Regenerative Medicine, Kobe University Hospital, Kobe, Japan; Division of Signal Pathways, Biosignal Research Center, Kobe University, Kobe, Japan.
| |
Collapse
|
3
|
Liao YJ, Chen YS, Lin YC, Yang JR. Three-Dimensional Cell Culture Scaffold Supports Capillary-Like Network Formation by Endothelial Cells Derived from Porcine-Induced Pluripotent Stem Cells. Cells Tissues Organs 2024; 214:26-35. [PMID: 39008972 DOI: 10.1159/000539320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/03/2024] [Indexed: 07/17/2024] Open
Abstract
INTRODUCTION Endothelial cells (EC) can be generated from porcine-induced pluripotent stem cells (piPSC), but poor efficiency in driving EC differentiation hampers their application and efficacy. Additionally, the culture of piPSC-derived EC (piPSC-EC) on three-dimensional (3D) scaffolds has not been fully reported yet. Here, we report a method to improve the generation of EC differentiation from piPSC and to facilitate their culture on 3D scaffolds, providing a potential resource for in vitro drug testing and the generation of tissue-engineered vascular grafts. METHODS We initiated the differentiation of piPSC into EC by seeding them on laminin 411 and employing a three-stage protocol, which involved the use of distinct EC differentiation media supplemented with CHIR99021, BMP4, VEGF, and bFGF. RESULTS piPSC-EC not only expressed EC markers such as CD31, VE-cadherin, and von Willebrand factor (vWF) but also exhibited an upregulation of EC marker genes, including CD31, CD34, VEGFR2, VE-cadherin, and vWF. They exhibited functional characteristics similar to those of porcine coronary artery endothelial cells (PCAEC), such as tube formation and Dil-Ac-LDL uptake. Furthermore, when cultured on 3D scaffolds, piPSC-EC developed a 3D morphology and were capable of forming an endothelial layer and engineering capillary-like networks, though these lacked lumen structures. CONCLUSION Our study not only advances the generation of EC from piPSC through an inhibitor and growth factor cocktail but also provides a promising approach for constructing vascular network-like structures. Importantly, these findings open new avenues for drug discovery in vitro and tissue engineering in vivo.
Collapse
Affiliation(s)
- Yu-Jing Liao
- Genetics and Physiology Division, Taiwan Livestock Research Institute, Ministry of Agriculture, Tainan, Taiwan,
| | - Yi-Shiou Chen
- Genetics and Physiology Division, Taiwan Livestock Research Institute, Ministry of Agriculture, Tainan, Taiwan
| | - Yu-Ching Lin
- Genetics and Physiology Division, Taiwan Livestock Research Institute, Ministry of Agriculture, Tainan, Taiwan
| | - Jenn-Rong Yang
- Genetics and Physiology Division, Taiwan Livestock Research Institute, Ministry of Agriculture, Tainan, Taiwan
| |
Collapse
|
4
|
Zhao M, Taniguchi Y, Shimono C, Jonouchi T, Cheng Y, Shimizu Y, Nalbandian M, Yamamoto T, Nakagawa M, Sekiguchi K, Sakurai H. Heparan Sulfate Chain-Conjugated Laminin-E8 Fragments Advance Paraxial Mesodermal Differentiation Followed by High Myogenic Induction from hiPSCs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308306. [PMID: 38685581 PMCID: PMC11234437 DOI: 10.1002/advs.202308306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/26/2024] [Indexed: 05/02/2024]
Abstract
Human-induced pluripotent stem cells (hiPSCs) have great therapeutic potential. The cell source differentiated from hiPSCs requires xeno-free and robust methods for lineage-specific differentiation. Here, a system is described for differentiating hiPSCs on new generation laminin fragments (NGLFs), a recombinant form of a laminin E8 fragment conjugated to the heparan sulfate chains (HS) attachment domain of perlecan. Using NGLFs, hiPSCs are highly promoted to direct differentiation into a paraxial mesoderm state with high-efficiency muscle lineage generation. HS conjugation to the C-terminus of Laminin E8 fragments brings fibroblast growth factors (FGFs) bound to the HS close to the cell surface of hiPSCs, thereby facilitating stronger FGF signaling pathways stimulation and initiating HOX gene expression, which triggers the paraxial mesoderm differentiation of hiPSCs. This highly efficient differentiation system can provide a roadmap for paraxial mesoderm development and an infinite source of myocytes and muscle stem cells for disease modeling and regenerative medicine.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Clinical ApplicationCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
- Center for Medical EpigeneticsSchool of Basic Medical SciencesChongqing Medical University1 Yixueyuan Road, Yuzhong DistrictChongqing400016China
| | - Yukimasa Taniguchi
- Division of Matrixome Research and ApplicationInstitute for Protein ResearchOsaka University3‐2 Yamadaoka, SuitaOsaka565‐0871Japan
| | - Chisei Shimono
- Division of Matrixome Research and ApplicationInstitute for Protein ResearchOsaka University3‐2 Yamadaoka, SuitaOsaka565‐0871Japan
| | - Tatsuya Jonouchi
- Department of Clinical ApplicationCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| | - Yushen Cheng
- Department of Life Science FrontiersCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| | - Yasuhiro Shimizu
- Division of Matrixome Research and ApplicationInstitute for Protein ResearchOsaka University3‐2 Yamadaoka, SuitaOsaka565‐0871Japan
| | - Minas Nalbandian
- Department of Clinical ApplicationCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| | - Takuya Yamamoto
- Department of Life Science FrontiersCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| | - Masato Nakagawa
- Department of Life Science FrontiersCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and ApplicationInstitute for Protein ResearchOsaka University3‐2 Yamadaoka, SuitaOsaka565‐0871Japan
| | - Hidetoshi Sakurai
- Department of Clinical ApplicationCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| |
Collapse
|
5
|
Nakata T, Li C, Mayassi T, Lin H, Ghosh K, Segerstolpe Å, Diamond EL, Herbst P, Biancalani T, Gaddam S, Parkar S, Lu Z, Jaiswal A, Li B, Creasey EA, Lefkovith A, Daly MJ, Graham DB, Xavier RJ. Genetic vulnerability to Crohn's disease reveals a spatially resolved epithelial restitution program. Sci Transl Med 2023; 15:eadg5252. [PMID: 37878672 PMCID: PMC10798370 DOI: 10.1126/scitranslmed.adg5252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 10/06/2023] [Indexed: 10/27/2023]
Abstract
Effective tissue repair requires coordinated intercellular communication to sense damage, remodel the tissue, and restore function. Here, we dissected the healing response in the intestinal mucosa by mapping intercellular communication at single-cell resolution and integrating with spatial transcriptomics. We demonstrated that a risk variant for Crohn's disease, hepatocyte growth factor activator (HGFAC) Arg509His (R509H), disrupted a damage-sensing pathway connecting the coagulation cascade to growth factors that drive the differentiation of wound-associated epithelial (WAE) cells and production of a localized retinoic acid (RA) gradient to promote fibroblast-mediated tissue remodeling. Specifically, we showed that HGFAC R509H was activated by thrombin protease activity but exhibited impaired proteolytic activation of the growth factor macrophage-stimulating protein (MSP). In Hgfac R509H mice, reduced MSP activation in response to wounding of the colon resulted in impaired WAE cell induction and delayed healing. Through integration of single-cell transcriptomics and spatial transcriptomics, we demonstrated that WAE cells generated RA in a spatially restricted region of the wound site and that mucosal fibroblasts responded to this signal by producing extracellular matrix and growth factors. We further dissected this WAE cell-fibroblast signaling circuit in vitro using a genetically tractable organoid coculture model. Collectively, these studies exploited a genetic perturbation associated with human disease to disrupt a fundamental biological process and then reconstructed a spatially resolved mechanistic model of tissue healing.
Collapse
Affiliation(s)
- Toru Nakata
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Chenhao Li
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Toufic Mayassi
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Helen Lin
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Koushik Ghosh
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Åsa Segerstolpe
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Emma L. Diamond
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Paula Herbst
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | - Ziqing Lu
- Genentech, South San Francisco, CA 94080, USA
| | - Alok Jaiswal
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Bihua Li
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Elizabeth A. Creasey
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ariel Lefkovith
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Mark J. Daly
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Analytical and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Daniel B. Graham
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ramnik J. Xavier
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
6
|
Sugiyama-Nakagiri Y, Yamashita S, Taniguchi Y, Shimono C, Sekiguchi K. Laminin fragments conjugated with perlecan's growth factor-binding domain differentiate human induced pluripotent stem cells into skin-derived precursor cells. Sci Rep 2023; 13:14556. [PMID: 37666868 PMCID: PMC10477235 DOI: 10.1038/s41598-023-41701-5] [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: 12/11/2022] [Accepted: 08/30/2023] [Indexed: 09/06/2023] Open
Abstract
Deriving stem cells to regenerate full-thickness human skin is important for treating skin disorders without invasive surgical procedures. Our previous protocol to differentiate human induced pluripotent stem cells (iPSCs) into skin-derived precursor cells (SKPs) as a source of dermal stem cells employs mouse fibroblasts as feeder cells and is therefore unsuitable for clinical use. Herein, we report a feeder-free method for differentiating iPSCs into SKPs by customising culture substrates. We immunohistochemically screened for laminins expressed in dermal papillae (DP) and explored the conditions for inducing the differentiation of iPSCs into SKPs on recombinant laminin E8 (LM-E8) fragments with or without conjugation to domain I of perlecan (PDI), which binds to growth factors through heparan sulphate chains. Several LM-E8 fragments, including those of LM111, 121, 332, 421, 511, and 521, supported iPSC differentiation into SKPs without PDI conjugation. However, the SKP yield was significantly enhanced on PDI-conjugated LM-E8 fragments. SKPs induced on PDI-conjugated LM111-E8 fragments retained the gene expression patterns characteristic of SKPs, as well as the ability to differentiate into adipocytes, osteocytes, and Schwann cells. Thus, PDI-conjugated LM-E8 fragments are promising agents for inducing iPSC differentiation into SKPs in clinical settings.
Collapse
Affiliation(s)
| | - Shiho Yamashita
- Kao Corporation, 2602, Akabane Ichikai-Machi, Haga-gun, Tochigi, 321-3497, Japan
| | - Yukimasa Taniguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Chisei Shimono
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
7
|
Ho WJ, Kobayashi M, Murata K, Hashimoto Y, Izumi K, Kimura T, Kanemitsu H, Yamazaki K, Ikeda T, Minatoya K, Kishida A, Masumoto H. A novel approach for the endothelialization of xenogeneic decellularized vascular tissues by human cells utilizing surface modification and dynamic culture. Sci Rep 2022; 12:22294. [PMID: 36566330 PMCID: PMC9789980 DOI: 10.1038/s41598-022-26792-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Decellularized xenogeneic vascular grafts can be used in revascularization surgeries. We have developed decellularization methods using high hydrostatic pressure (HHP), which preserves the extracellular structure. Here, we attempted ex vivo endothelialization of HHP-decellularized xenogeneic tissues using human endothelial cells (ECs) to prevent clot formation against human blood. Slices of porcine aortic endothelium were decellularized using HHP and coated with gelatin. Human umbilical vein ECs were directly seeded and cultured under dynamic flow or static conditions for 14 days. Dynamic flow cultures tend to demonstrate higher cell coverage. We then coated the tissues with the E8 fragment of human laminin-411 (hL411), which has high affinity for ECs, and found that Dynamic/hL411showed high area coverage, almost reaching 100% (Dynamic/Gelatin vs Dynamic/hL411; 58.7 ± 11.4 vs 97.5 ± 1.9%, P = 0.0017). Immunostaining revealed sufficient endothelial cell coverage as a single cell layer in Dynamic/hL411. A clot formation assay using human whole blood showed low clot formation in Dynamic/hL411, almost similar to that in the negative control, polytetrafluoroethylene. Surface modification of HHP-decellularized xenogeneic endothelial tissues combined with dynamic culture achieved sufficient ex vivo endothelialization along with prevention of clot formation, indicating their potential for clinical use as vascular grafts in the future.
Collapse
Affiliation(s)
- Wen-Jin Ho
- grid.258799.80000 0004 0372 2033Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Mako Kobayashi
- grid.265073.50000 0001 1014 9130Department of Material-Based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan ,grid.69566.3a0000 0001 2248 6943Present Address: Department of Materials Processing, Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Kozue Murata
- grid.258799.80000 0004 0372 2033Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan ,grid.508743.dClinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan ,grid.411217.00000 0004 0531 2775Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Yoshihide Hashimoto
- grid.265073.50000 0001 1014 9130Department of Material-Based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Tsuyoshi Kimura
- grid.265073.50000 0001 1014 9130Department of Material-Based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hideo Kanemitsu
- grid.258799.80000 0004 0372 2033Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan ,grid.415392.80000 0004 0378 7849Present Address: Department of Cardiovascular Surgery, Kitano Hospital, Osaka, Japan
| | - Kazuhiro Yamazaki
- grid.258799.80000 0004 0372 2033Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Tadashi Ikeda
- grid.258799.80000 0004 0372 2033Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Kenji Minatoya
- grid.258799.80000 0004 0372 2033Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Akio Kishida
- grid.265073.50000 0001 1014 9130Department of Material-Based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hidetoshi Masumoto
- grid.258799.80000 0004 0372 2033Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507 Japan ,grid.508743.dClinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
8
|
Kamishibahara Y, Okamoto S, Ohkuma T, Taniguchi H. Stabilized generation of human iPSC-derived liver organoids using a modified coating approach. Biol Methods Protoc 2022; 8:bpac034. [PMID: 36694573 PMCID: PMC9869720 DOI: 10.1093/biomethods/bpac034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
Human-induced pluripotent stem cell (hiPSC)-derived hepatic cells are useful tools for regenerative medicine, and various culture substrates are currently used for their differentiation. We differentiated hiPSC-derived hepatic endoderm (HE), endothelial cells (ECs), and mesenchymal cells (MCs) using Laminin-511 (LN) coating to generate liver organoids, hiPSC-liver buds (hiPSC-LBs), which exhibited therapeutic effects when transplanted into disease model animals. Stably producing significant amounts of hiPSC-LBs is necessary for sufficient therapeutic effects. However, general precoating (standard coating) requires quick manipulation, often causing failure for inexperienced cell cultures, we thus tested direct LN addition to the culture medium (Direct coating). Using quantitative gene expression, flow cytometry, albumin secretion, and ammonia metabolism, we demonstrated that Standard and Direct coating similarly induce hiPSC-derived hepatocyte, mesodermal cell, EC, and MC differentiation. Standard and Direct coating-differentiated cells generated iPSC-LBs with equivalent hepatic functions. Furthermore, Direct coating enabled stable induction of differentiation independent of individual culture skills and reduced total amount of LN use as the same differentiated cell quality can be obtained upon LN supplementation at lower concentrations. In summary, the results of this study suggest that Direct coating could enable stable hiPSC-LB production at a low cost, thereby yielding mass cell production using hiPSCs.
Collapse
Affiliation(s)
- Yu Kamishibahara
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Satoshi Okamoto
- Correspondence address. (S.O.) Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan. Tel/: +81 45 787 8963; E-mail: . (H.T.) Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan. Tel: +81 3 5449 5698; E-mail:
| | - Takuya Ohkuma
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Hideki Taniguchi
- Correspondence address. (S.O.) Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan. Tel/: +81 45 787 8963; E-mail: . (H.T.) Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan. Tel: +81 3 5449 5698; E-mail:
| |
Collapse
|
9
|
Abstract
Formation of the vasculature is a critical step within the developing embryo and its disruption causes early embryonic lethality. This complex process is driven by a cascade of signaling events that controls differentiation of mesodermal progenitors into primordial endothelial cells and their further specification into distinct subtypes (arterial, venous, hemogenic) that are needed to generate a blood circulatory network. Hemogenic endothelial cells give rise to hematopoietic stem and progenitor cells that generate all blood cells in the body during embryogenesis and postnatally. We focus our discussion on the regulation of endothelial cell differentiation, and subsequent hemogenic specification, and highlight many of the signaling pathways involved in these processes, which are conserved across vertebrates. Gaining a better understanding of the regulation of these processes will yield insights needed to optimize the treatment of vascular and hematopoietic disease and generate human stem cell-derived vascular and hematopoietic cells for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Jordon W Aragon
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
- Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
- Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
- Departments of Medicine and Genetics, Yale University School of Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut 06520, USA
| |
Collapse
|
10
|
Redox signaling induces laminin receptor ribosomal protein-SA expression to improve cell adhesion following radiofrequency glow discharge treatments. Sci Rep 2022; 12:7742. [PMID: 35546602 PMCID: PMC9095671 DOI: 10.1038/s41598-022-11766-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/20/2022] [Indexed: 11/19/2022] Open
Abstract
Current biomaterials effectively replace biological structures but are limited by infections and long-term material failures. This study examined the molecular mechanisms of radio frequency glow discharge treatments (RFGDT) in mediating the disinfection of biomaterial surfaces and concurrently promoting cell attachment and proliferation. Dental biomaterials were subjected to RFGDT, and viability of oral microbial species, namely Streptococcus mutants (SM), Streptococcus gordonii (SG), Moraxella catarrhalis (MC), and Porphyromonas gingivalis (PG), were assessed. Cell attachment and survival of a pre-odontoblast cell line, MDPC-23, was examined. Finally, mechanistic investigations into redox generation and biological signaling were investigated. Based on their compositions, dental biomaterials induced reactive oxygen species (ROS) following dose-dependent RFGDT. Reduced microbial viability was evident following RFGDT in the catalase-negative (SM and SG) species more prominently than catalase-positive (MC and PG) species. Cell adhesion assays noted improved MDPC-23 attachment and survival. Pretreatments with N-acetylcysteine (NAC) and catalase abrogated these responses. Immunoassays noted redox-induced downstream expression of a laminin receptor, Ribosomal Protein SA, following RFGDT. Thus, RFGDT-induced redox mediates antimicrobial and improves cell responses such as adhesion and proliferation. These observations together provide a mechanistic rationale for the clinical utility of RFGDT with dental biomaterials for regenerative clinical applications.
Collapse
|
11
|
Engineering-Induced Pluripotent Stem Cells for Cancer Immunotherapy. Cancers (Basel) 2022; 14:cancers14092266. [PMID: 35565395 PMCID: PMC9100203 DOI: 10.3390/cancers14092266] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/23/2022] [Accepted: 04/29/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Induced pluripotent stem cells (iPSCs) that can be genetically engineered and differentiated into different types of immune cells, providing an unlimited resource for developing off-the-shelf cell therapies. Here, we present a comprehensive review that describes the current stages of iPSC-based cell therapies, including iPSC-derived T, nature killer (NK), invariant natural killer T (iNKT), gamma delta T (γδ T), mucosal-associated invariant T (MAIT) cells, and macrophages (Mφs). Abstract Cell-based immunotherapy, such as chimeric antigen receptor (CAR) T cell therapy, has revolutionized the treatment of hematological malignancies, especially in patients who are refractory to other therapies. However, there are critical obstacles that hinder the widespread clinical applications of current autologous therapies, such as high cost, challenging large-scale manufacturing, and inaccessibility to the therapy for lymphopenia patients. Therefore, it is in great demand to generate the universal off-the-shelf cell products with significant scalability. Human induced pluripotent stem cells (iPSCs) provide an “unlimited supply” for cell therapy because of their unique self-renewal properties and the capacity to be genetically engineered. iPSCs can be differentiated into different immune cells, such as T cells, natural killer (NK) cells, invariant natural killer T (iNKT) cells, gamma delta T (γδ T), mucosal-associated invariant T (MAIT) cells, and macrophages (Mφs). In this review, we describe iPSC-based allogeneic cell therapy, the different culture methods of generating iPSC-derived immune cells (e.g., iPSC-T, iPSC-NK, iPSC-iNKT, iPSC-γδT, iPSC-MAIT and iPSC-Mφ), as well as the recent advances in iPSC-T and iPSC-NK cell therapies, particularly in combinations with CAR-engineering. We also discuss the current challenges and the future perspectives in this field towards the foreseeable applications of iPSC-based immune therapy.
Collapse
|
12
|
Hall ML, Givens S, Santosh N, Iacovino M, Kyba M, Ogle BM. Laminin 411 mediates endothelial specification via multiple signaling axes that converge on β-catenin. Stem Cell Reports 2022; 17:569-583. [PMID: 35120622 PMCID: PMC9039757 DOI: 10.1016/j.stemcr.2022.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 11/24/2022] Open
Abstract
The extracellular matrix (ECM) provides essential cues to promote endothelial specification during tissue development in vivo; correspondingly, ECM is considered essential for endothelial differentiation outside of the body. However, systematic studies to assess the precise contribution of individual ECM proteins to endothelial differentiation have not been conducted. Further, the multi-component nature of differentiation protocols makes it challenging to study the underlying mechanisms by which the ECM contributes to cell fate. In this study, we determined that Laminin 411 alone increases endothelial differentiation of induced pluripotent stem cells over collagen I or Matrigel. The effect of ECM was shown to be independent of vascular endothelial growth factor (VEGF) binding capacity. We also show that ECM-guided endothelial differentiation is dependent on activation of focal adhesion kinase (FAK), integrin-linked kinase (ILK), Notch, and β-catenin pathways. Our results indicate that ECM contributes to endothelial differentiation through multiple avenues, which converge at the expression of active β-catenin.
Collapse
Affiliation(s)
- Mikayla L Hall
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, 7-130 Nils Hasselmo Hall, 312 Church St. SE, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Sophie Givens
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, 7-130 Nils Hasselmo Hall, 312 Church St. SE, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Natasha Santosh
- Stem Cell Institute, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Michelina Iacovino
- Stem Cell Institute, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Michael Kyba
- Stem Cell Institute, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA; Lillehei Heart Institute, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, 7-130 Nils Hasselmo Hall, 312 Church St. SE, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA; Lillehei Heart Institute, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA; Institute for Engineering in Medicine, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA.
| |
Collapse
|
13
|
Lau S, Gossen M, Lendlein A. Designing Cardiovascular Implants Taking in View the Endothelial Basement Membrane. Int J Mol Sci 2021; 22:ijms222313120. [PMID: 34884923 PMCID: PMC8658568 DOI: 10.3390/ijms222313120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 12/28/2022] Open
Abstract
Insufficient endothelialization of cardiovascular grafts is a major hurdle in vascular surgery and regenerative medicine, bearing a risk for early graft thrombosis. Neither of the numerous strategies pursued to solve these problems were conclusive. Endothelialization is regulated by the endothelial basement membrane (EBM), a highly specialized part of the vascular extracellular matrix. Thus, a detailed understanding of the structure–function interrelations of the EBM components is fundamental for designing biomimetic materials aiming to mimic EBM functions. In this review, a detailed description of the structure and functions of the EBM are provided, including the luminal and abluminal interactions with adjacent cell types, such as vascular smooth muscle cells. Moreover, in vivo as well as in vitro strategies to build or renew EBM are summarized and critically discussed. The spectrum of methods includes vessel decellularization and implant biofunctionalization strategies as well as tissue engineering-based approaches and bioprinting. Finally, the limitations of these methods are highlighted, and future directions are suggested to help improve future design strategies for EBM-inspired materials in the cardiovascular field.
Collapse
Affiliation(s)
- Skadi Lau
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, Kantstraße 55, 14513 Teltow, Germany; (S.L.); (M.G.)
| | - Manfred Gossen
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, Kantstraße 55, 14513 Teltow, Germany; (S.L.); (M.G.)
| | - Andreas Lendlein
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, Kantstraße 55, 14513 Teltow, Germany; (S.L.); (M.G.)
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Straße 25, 14476 Potsdam, Germany
- Correspondence:
| |
Collapse
|
14
|
Assessment of the Hematopoietic Differentiation Potential of Human Pluripotent Stem Cells in 2D and 3D Culture Systems. Cells 2021; 10:cells10112858. [PMID: 34831080 PMCID: PMC8616232 DOI: 10.3390/cells10112858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND In vitro methods for hematopoietic differentiation of human pluripotent stem cells (hPSC) are a matter of priority for the in-depth research into the mechanisms of early embryogenesis. So-far, published results regarding the generation of hematopoietic cells come from studies using either 2D or 3D culture formats, hence, it is difficult to discern their particular contribution to the development of the concept of a unique in vitro model in close resemblance to in vivo hematopoiesis. AIM OF THE STUDY To assess using the same culture conditions and the same time course, the potential of each of these two formats to support differentiation of human pluripotent stem cells to primitive hematopoiesis without exogenous activation of Wnt signaling. METHODS We used in parallel 2D and 3D formats, the same culture environment and assay methods (flow cytometry, IF, qPCR) to investigate stages of commitment and specification of mesodermal, and hemogenic endothelial cells to CD34 hematopoietic cells and evaluated their clonogenic capacity in a CFU system. RESULTS We show an adequate formation of mesoderm, an efficient commitment to hemogenic endothelium, a higher number of CD34 hematopoietic cells, and colony-forming capacity potential only in the 3D format-supported differentiation. CONCLUSIONS This study shows that the 3D but not the 2D format ensures the induction and realization by endogenous mechanisms of human pluripotent stem cells' intrinsic differentiation program to primitive hematopoietic cells. We propose that the 3D format provides an adequate level of upregulation of the endogenous Wnt/β-catenin signaling.
Collapse
|
15
|
Ewald ML, Chen YH, Lee AP, Hughes CCW. The vascular niche in next generation microphysiological systems. LAB ON A CHIP 2021; 21:3244-3262. [PMID: 34396383 PMCID: PMC8635227 DOI: 10.1039/d1lc00530h] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
In recent years, microphysiological system (MPS, also known as, organ-on-a-chip or tissue chip) platforms have emerged with great promise to improve the predictive capacity of preclinical modeling thereby reducing the high attrition rates when drugs move into trials. While their designs can vary quite significantly, in general MPS are bioengineered in vitro microenvironments that recapitulate key functional units of human organs, and that have broad applications in human physiology, pathophysiology, and clinical pharmacology. A critical next step in the evolution of MPS devices is the widespread incorporation of functional vasculature within tissues. The vasculature itself is a major organ that carries nutrients, immune cells, signaling molecules and therapeutics to all other organs. It also plays critical roles in inducing and maintaining tissue identity through expression of angiocrine factors, and in providing tissue-specific milieus (i.e., the vascular niche) that can support the survival and function of stem cells. Thus, organs are patterned, maintained and supported by the vasculature, which in turn receives signals that drive tissue specific gene expression. In this review, we will discuss published vascularized MPS platforms and present considerations for next-generation devices looking to incorporate this critical constituent. Finally, we will highlight the organ-patterning processes governed by the vasculature, and how the incorporation of a vascular niche within MPS platforms will establish a unique opportunity to study stem cell development.
Collapse
Affiliation(s)
- Makena L Ewald
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
16
|
Matsuo S, Nishinaka-Arai Y, Kazuki Y, Oshimura M, Nakahata T, Niwa A, Saito MK. Pluripotent stem cell model of early hematopoiesis in Down syndrome reveals quantitative effects of short-form GATA1 protein on lineage specification. PLoS One 2021; 16:e0247595. [PMID: 33780474 PMCID: PMC8007000 DOI: 10.1371/journal.pone.0247595] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Children with Down syndrome (DS) are susceptible to two blood disorders, transient abnormal myelopoiesis (TAM) and Down syndrome-associated acute megakaryocytic leukemia (DS-AMKL). Mutations in GATA binding protein 1 (GATA1) have been identified as the cause of these diseases, and the expression levels of the resulting protein, short-form GATA1 (GATA1s), are known to correlate with the severity of TAM. On the other hand, despite the presence of GATA1 mutations in almost all cases of DS-AMKL, the incidence of DS-AMKL in TAM patients is inversely correlated with the expression of GATA1s. This discovery has required the need to clarify the role of GATA1s in generating the cells of origin linked to the risk of both diseases. Focusing on this point, we examined the characteristics of GATA1 mutant trisomy-21 pluripotent stem cells transfected with a doxycycline (Dox)-inducible GATA1s expression cassette in a stepwise hematopoietic differentiation protocol. We found that higher GATA1s expression significantly reduced commitment into the megakaryocytic lineage at the early hematopoietic progenitor cell (HPC) stage, but once committed, the effect was reversed in progenitor cells and acted to maintain the progenitors. These differentiation stage-dependent reversal effects were in contrast to the results of myeloid lineage, where GATA1s simply sustained and increased the number of immature myeloid cells. These results suggest that although GATA1 mutant cells cause the increase in myeloid and megakaryocytic progenitors regardless of the intensity of GATA1s expression, the pathways vary with the expression level. This study provides experimental support for the paradoxical clinical features of GATA1 mutations in the two diseases.
Collapse
Affiliation(s)
- Shiori Matsuo
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yoko Nishinaka-Arai
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail: (YNA); (AN); (MKS)
| | - Yasuhiro Kazuki
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Mitsuo Oshimura
- Chromosome Engineering Research Center, Tottori University, Tottori, Japan
| | - Tatsutoshi Nakahata
- Drug Discovery Technology Development Office, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Akira Niwa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- * E-mail: (YNA); (AN); (MKS)
| | - Megumu K. Saito
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- * E-mail: (YNA); (AN); (MKS)
| |
Collapse
|
17
|
Matsuo M, Nadanaka S, Soga M, Sugiyama T, Serigano S, Shimano K, Ichinose F, Nakamura T, Maeda T, Houkin K, Era T, Kitagawa H. Vulnerability to shear stress caused by altered peri-endothelial matrix is a key feature of Moyamoya disease. Sci Rep 2021; 11:1552. [PMID: 33452387 PMCID: PMC7810726 DOI: 10.1038/s41598-021-81282-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 12/28/2020] [Indexed: 02/06/2023] Open
Abstract
Moyamoya disease (MMD) is characterized by progressive bilateral stenotic changes in the terminal portion of the internal carotid arteries. Although RNF213 was identified as a susceptibility gene for MMD, the exact pathogenesis remains unknown. Immunohistochemical analysis of autopsy specimens from a patient with MMD revealed marked accumulation of hyaluronan and chondroitin sulfate (CS) in the thickened intima of occlusive lesions of MMD. Hyaluronan synthase 2 was strongly expressed in endothelial progenitor cells in the thickened intima. Furthermore, MMD lesions showed minimal staining for CS and hyaluronan in the endothelium, in contrast to control endothelium showing positive staining for both. Glycosaminoglycans of endothelial cells derived from MMD and control induced pluripotent stem cells demonstrated a decreased amount of CS, especially sulfated CS, in MMD. A computational fluid dynamics model showed highest wall shear stress values in the terminal portion of the internal carotid artery, which is the predisposing region in MMD. Because the peri-endothelial extracellular matrix plays an important role in protection, cell adhesion and migration, an altered peri-endothelial matrix in MMD may contribute to endothelial vulnerability to wall shear stress. Invading endothelial progenitor cells repairing endothelial injury would produce excessive hyaluronan and CS in the intima, and cause vascular stenosis.
Collapse
Affiliation(s)
- Muneaki Matsuo
- Department of Pediatrics, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| | - Satomi Nadanaka
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Minami Soga
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Taku Sugiyama
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shota Serigano
- Department of Mechanical Systems Engineering, Faculty of Engineering, Tokyo City University, Setagaya, Japan
| | - Kenjiro Shimano
- Department of Mechanical Systems Engineering, Faculty of Engineering, Tokyo City University, Setagaya, Japan
| | - Fumio Ichinose
- Department of Pediatrics, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Takuji Nakamura
- Department of Pediatrics, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Toshiyuki Maeda
- Department of Pediatrics, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Kiyohiro Houkin
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| |
Collapse
|
18
|
Motallebnejad P, Azarin SM. Chemically defined human vascular laminins for biologically relevant culture of hiPSC-derived brain microvascular endothelial cells. Fluids Barriers CNS 2020; 17:54. [PMID: 32912242 PMCID: PMC7488267 DOI: 10.1186/s12987-020-00215-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/25/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND In recent years, differentiation of human induced pluripotent stem cells (hiPSCs) into brain-specific microvascular endothelial cells (iBMECs) has frequently been used to model the blood-brain barrier (BBB). However, there are limitations in the use of iBMECs for in vitro studies, such as transendothelial electrical resistance (TEER) instability, weak junctional expression of VE-cadherin, and lack of proper fluid shear stress response. In vivo, the basement membrane (BM) composition of the BBB evolves throughout development, and laminins become the dominant component of the mature vascular BM. However, laminin isoforms of the endothelial BM have not been used for culture of differentiated iBMECs. The main goal of this study is to investigate the effect of different laminin isoforms of the endothelial BM on iBMEC functionality and to determine whether better recapitulation of the physiological BM in vitro can address the aforementioned limitations of iBMECs. METHODS Using a previously reported method, hiPSCs were differentiated into iBMECs. The influence of main laminins of the endothelial BM, LN 411 and LN 511, on iBMEC functionality was studied and compared to a collagen IV and fibronectin mixture (CN IV-FN). Quantitative RT-PCR, immunocytochemistry, and TEER measurement were utilized to assess gene and protein expression and barrier properties of iBMECs on different extracellular matrices. Single-channel microfluidic devices were used to study the effect of shear stress on iBMECs. RESULTS LN 511, but not LN 411, improved iBMEC barrier properties and resulted in more sustained TEER stability. Immunocytochemistry showed improved junctional protein expression compared to iBMECs cultured on CN IV-FN. iBMECs cultured on LN 511 showed a reduction of stress fibers, indicating resting endothelial phenotype, whereas gene expression analysis revealed upregulation of multiple genes involved in endothelial activation in iBMECs on CN IV-FN. Finally, culturing iBMECs on LN 511 enhanced physiological responses to shear stress, including morphological changes and enhanced junctional protein association. CONCLUSION LN 511 improves the functionality and long-term barrier stability of iBMECs. Our findings suggest that incorporation of physiologically relevant LN 511 in iBMEC culture would be beneficial for disease modeling applications and BBB-on-a-chip platforms that accommodate fluid flow.
Collapse
Affiliation(s)
- Pedram Motallebnejad
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
19
|
Paik DT, Cho S, Tian L, Chang HY, Wu JC. Single-cell RNA sequencing in cardiovascular development, disease and medicine. Nat Rev Cardiol 2020; 17:457-473. [PMID: 32231331 PMCID: PMC7528042 DOI: 10.1038/s41569-020-0359-y] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/24/2020] [Indexed: 02/08/2023]
Abstract
Advances in single-cell RNA sequencing (scRNA-seq) technologies in the past 10 years have had a transformative effect on biomedical research, enabling the profiling and analysis of the transcriptomes of single cells at unprecedented resolution and throughput. Specifically, scRNA-seq has facilitated the identification of novel or rare cell types, the analysis of single-cell trajectory construction and stem or progenitor cell differentiation, and the comparison of healthy and disease-related tissues at single-cell resolution. These applications have been critical in advances in cardiovascular research in the past decade as evidenced by the generation of cell atlases of mammalian heart and blood vessels and the elucidation of mechanisms involved in cardiovascular development and stem or progenitor cell differentiation. In this Review, we summarize the currently available scRNA-seq technologies and analytical tools and discuss the latest findings using scRNA-seq that have substantially improved our knowledge on the development of the cardiovascular system and the mechanisms underlying cardiovascular diseases. Furthermore, we examine emerging strategies that integrate multimodal single-cell platforms, focusing on future applications in cardiovascular precision medicine that use single-cell omics approaches to characterize cell-specific responses to drugs or environmental stimuli and to develop effective patient-specific therapeutics.
Collapse
Affiliation(s)
- David T Paik
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Sangkyun Cho
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lei Tian
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
20
|
Ohata K, Ott HC. Human-scale lung regeneration based on decellularized matrix scaffolds as a biologic platform. Surg Today 2020; 50:633-643. [PMID: 32363425 PMCID: PMC7305261 DOI: 10.1007/s00595-020-02000-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/06/2020] [Indexed: 12/25/2022]
Abstract
Lung transplantation is currently the only curative treatment for patients with end-stage lung disease; however, donor organ shortage and the need for intense immunosuppression limit its broad clinical application. Bioartificial lungs created by combining native matrix scaffolds with patient-derived cells might overcome these problems. Decellularization involves stripping away cells while leaving behind the extracellular matrix scaffold. Cadaveric lungs are decellularized by detergent perfusion, and histologic examination confirms the absence of cellular components but the preservation of matrix proteins. The resulting lung scaffolds are recellularized in a bioreactor that provides biomimetic conditions, including vascular perfusion and liquid ventilation. Cell seeding, engraftment, and tissue maturation are achieved in whole-organ culture. Bioartificial lungs are transplantable, similarly to donor lungs, because the scaffolds preserve the vascular and airway architecture. In rat and porcine transplantation models, successful anastomoses of the vasculature and the airway were achieved, and gas exchange was evident after reperfusion. However, long-term function has not been achieved because of the immaturity of the vascular bed and distal lung epithelia. The goal of this strategy is to create patient-specific transplantable lungs using induced pluripotent stem cell (iPSC)-derived cells. The repopulation of decellularized scaffolds to create transplantable organs is one of possible future clinical applications of iPSCs.
Collapse
Affiliation(s)
- Keiji Ohata
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, CPZN 4800, Boston, MA, 02114, USA
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Harald C Ott
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, CPZN 4800, Boston, MA, 02114, USA.
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
McCracken IR, Taylor RS, Kok FO, de la Cuesta F, Dobie R, Henderson BEP, Mountford JC, Caudrillier A, Henderson NC, Ponting CP, Baker AH. Transcriptional dynamics of pluripotent stem cell-derived endothelial cell differentiation revealed by single-cell RNA sequencing. Eur Heart J 2020; 41:1024-1036. [PMID: 31242503 PMCID: PMC9597329 DOI: 10.1093/eurheartj/ehz351] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 03/13/2019] [Accepted: 05/14/2019] [Indexed: 12/20/2022] Open
Abstract
AIMS Pluripotent stem cell-derived endothelial cell products possess therapeutic potential in ischaemic vascular disease. However, the factors that drive endothelial differentiation from pluripotency and cellular specification are largely unknown. The aims of this study were to use single-cell RNA sequencing (scRNA-seq) to map the transcriptional landscape and cellular dynamics of directed differentiation of human embryonic stem cell-derived endothelial cells (hESC-EC) and to compare these cells to mature endothelial cells from diverse vascular beds. METHODS AND RESULTS A highly efficient directed 8-day differentiation protocol was used to generate a hESC-derived endothelial cell product (hESC-ECP), in which 66% of cells co-expressed CD31 and CD144. We observed largely homogeneous hESC and mesodermal populations at Days 0 and 4, respectively, followed by a rapid emergence of distinct endothelial and mesenchymal populations. Pseudotime trajectory identified transcriptional signatures of endothelial commitment and maturation during the differentiation process. Concordance in transcriptional signatures was verified by scRNA-seq analysis using both a second hESC line RC11, and an alternative hESC-EC differentiation protocol. In total, 105 727 cells were subjected to scRNA-seq analysis. Global transcriptional comparison revealed a transcriptional architecture of hESC-EC that differs from freshly isolated and cultured human endothelial cells and from organ-specific endothelial cells. CONCLUSION A transcriptional bifurcation into endothelial and mesenchymal lineages was identified, as well as novel transcriptional signatures underpinning commitment and maturation. The transcriptional architecture of hESC-ECP was distinct from mature and foetal human EC.
Collapse
Affiliation(s)
- Ian R McCracken
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Richard S Taylor
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Fatma O Kok
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Fernando de la Cuesta
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Ross Dobie
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Beth E P Henderson
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | | - Axelle Caudrillier
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Neil C Henderson
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Chris P Ponting
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
22
|
Yap L, Tay HG, Nguyen MT, Tjin MS, Tryggvason K. Laminins in Cellular Differentiation. Trends Cell Biol 2019; 29:987-1000. [DOI: 10.1016/j.tcb.2019.10.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/01/2019] [Accepted: 10/04/2019] [Indexed: 12/21/2022]
|
23
|
Genome-wide microhomologies enable precise template-free editing of biologically relevant deletion mutations. Nat Commun 2019; 10:4856. [PMID: 31649251 PMCID: PMC6813315 DOI: 10.1038/s41467-019-12829-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023] Open
Abstract
The functional effect of a gene edit by designer nucleases depends on the DNA repair outcome at the targeted locus. While non-homologous end joining (NHEJ) repair results in various mutations, microhomology-mediated end joining (MMEJ) creates precise deletions based on the alignment of flanking microhomologies (µHs). Recently, the sequence context surrounding nuclease-induced double strand breaks (DSBs) has been shown to predict repair outcomes, for which µH plays an important role. Here, we survey naturally occurring human deletion variants and identify that 11 million or 57% are flanked by µHs, covering 88% of protein-coding genes. These biologically relevant mutations are candidates for precise creation in a template-free manner by MMEJ repair. Using CRISPR-Cas9 in human induced pluripotent stem cells (hiPSCs), we efficiently create pathogenic deletion mutations for demonstrable disease models with both gain- and loss-of-function phenotypes. We anticipate this dataset and gene editing strategy to enable functional genetic studies and drug screening.
Collapse
|
24
|
Matsubara H, Niwa A, Nakahata T, Saito MK. Induction of human pluripotent stem cell-derived natural killer cells for immunotherapy under chemically defined conditions. Biochem Biophys Res Commun 2019; 515:1-8. [DOI: 10.1016/j.bbrc.2019.03.085] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 10/27/2022]
|
25
|
Novikova OA, Nazarkina ZK, Cherepanova AV, Laktionov PP, Chelobanov BP, Murashov IS, Deev RV, Pokushalov EA, Karpenko AA, Laktionov PP. Isolation, culturing and gene expression profiling of inner mass cells from stable and vulnerable carotid atherosclerotic plaques. PLoS One 2019; 14:e0218892. [PMID: 31242269 PMCID: PMC6594632 DOI: 10.1371/journal.pone.0218892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
The connective tissue components that form the atherosclerotic plaque body are produced by the plaque inner mass cells (PIMC), located inside the plaque. We report an approach to isolate and culture cells from the connective tissue of stable and vulnerable human atherosclerotic plaques based on elimination of non-connective tissue cells such as blood and non-plaque intima cells with a lysis buffer. The resulting plaque cells were characterized by growth capacity, morphology, transcriptome profiling and specific protein expression. Plaque cells slowly proliferated for up to three passages unaffected by the use of proliferation stimulants or changes of culture media composition. Stable plaques yielded more cells than vulnerable ones. Plaque cell cultures also contained several morphological cellular types. RNA-seq profiles of plaque cells were different from any of the cell types known to be involved in atherogenesis. The expression of the following proteins was observed in cultured plaque cells: smooth muscle cells marker α-SMA, macrophage marker CD14, extracellular matrix proteins aggrecan, fibronectin, neovascularisation markers VEGF-A, CD105, cellular adhesion receptor CD31 and progenitor/dedifferentiation receptor CD34. Differential expression of several notable transcripts in cells from stable and vulnerable plaques suggests the value of plaque cell culture studies for the search of plaque vulnerability markers.
Collapse
Affiliation(s)
- Olga A. Novikova
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Zhanna K. Nazarkina
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | - Anna V. Cherepanova
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
- * E-mail:
| | - Petr P. Laktionov
- Laboratory of Genomics, SB RAS Institute of Molecular and Cellular Biology, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Boris P. Chelobanov
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Ivan S. Murashov
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | | | - Evgeny A. Pokushalov
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | - Andrey A. Karpenko
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Pavel P. Laktionov
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| |
Collapse
|
26
|
|
27
|
Hall ML, Ogle BM. Cardiac Extracellular Matrix Modification as a Therapeutic Approach. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1098:131-150. [PMID: 30238369 PMCID: PMC6584040 DOI: 10.1007/978-3-319-97421-7_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The cardiac extracellular matrix (cECM) is comprised of proteins and polysaccharides secreted by cardiac cell types, which provide structural and biochemical support to cardiovascular tissue. The roles of cECM proteins and the associated family of cell surface receptor, integrins, have been explored in vivo via the generation of knockout experimental animal models. However, the complexity of tissues makes it difficult to isolate the effects of individual cECM proteins on a particular cell process or disease state. The desire to further dissect the role of cECM has led to the development of a variety of in vitro model systems, which are now being used not only for basic studies but also for testing drug efficacy and toxicity and for generating therapeutic scaffolds. These systems began with 2D coatings of cECM derived from tissue and have developed to include recombinant ECM proteins, ECM fragments, and ECM mimics. Most recently 3D model systems have emerged, made possible by several developing technologies including, and most notably, 3D bioprinting. This chapter will attempt to track the evolution of our understanding of the relationship between cECM and cell behavior from in vivo model to in vitro control systems. We end the chapter with a summary of how basic studies such as these have informed the use of cECM as a direct therapy.
Collapse
Affiliation(s)
- Mikayla L Hall
- Department of Biomedical Engineering, University of Minnesota - Twin Cities, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota - Twin Cities, Minneapolis, MN, USA
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Stem Cell Institute, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Lillehei Heart Institute, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Institute for Engineering in Medicine, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
| |
Collapse
|
28
|
Masuda S, Matsuura K, Shimizu T. Preparation of iPS cell-derived CD31 + endothelial cells using three-dimensional suspension culture. Regen Ther 2018; 9:1-9. [PMID: 30525069 PMCID: PMC6222294 DOI: 10.1016/j.reth.2018.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/24/2018] [Accepted: 06/22/2018] [Indexed: 12/03/2022] Open
Abstract
A well-organised vascular network is essential for metabolic exchange to maintain homoeostasis in the body. Therefore, for progress in regenerative medicine, it is particularly important to establish methods of vascularization in bioengineered three-dimensional (3D) functional tissues. In addition, it is necessary to develop methods to supply a large number of iPS cell-derived endothelial cells for fabricating the vascular network structure. There are already many reports on the method of inducing the differentiation of endothelial cells from iPS cells using 2D culture. However, there are few reports on methods for preparing a large number of iPS cell-derived endothelial cells. Therefore, we developed methods for inducing vascular endothelial cells from human inducible pluripotent stem (hiPS) cells using 3D suspension culture. hiPS cell-derived CD31+ cells expressed several endothelial marker genes and formed endothelial cell network structures, similar to human umbilical vein endothelial cells. These results indicate that hiPS cell-derived CD31+ cells may be a useful cell source for pre-vascularised network structures in 3D functional tissues, and it is important to develop 3D mass culture system for preparing a large number of cells to fabricate bioengineered tissues.
Collapse
Affiliation(s)
- Shinako Masuda
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
- Department of Cardiology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| |
Collapse
|
29
|
Shibata S, Hayashi R, Okubo T, Kudo Y, Katayama T, Ishikawa Y, Toga J, Yagi E, Honma Y, Quantock AJ, Sekiguchi K, Nishida K. Selective Laminin-Directed Differentiation of Human Induced Pluripotent Stem Cells into Distinct Ocular Lineages. Cell Rep 2018; 25:1668-1679.e5. [DOI: 10.1016/j.celrep.2018.10.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 09/10/2018] [Accepted: 10/05/2018] [Indexed: 12/22/2022] Open
|
30
|
Abstract
Disease-associated induced pluripotent stem cells (iPSCs) established from patients are now widely used for disease modeling. They can provide an unlimited source of hematopoietic cells that carry the patients' genetic background, making them advantageous for modeling immunological disorders. To obtain functional immune cells from human iPSCs, we have developed a differentiation system that generates immortalized myeloid cells including neutrophils and monocytic cells. By using this strategy, we have established in vitro models of many immunological disorders. In this review, we focus on autoinflammatory disorders. These models have proven useful for genetic diagnosis and elucidation of the disease mechanism.
Collapse
Affiliation(s)
- Megumu K Saito
- a Department of Clinical Application, CiRA , Kyoto University , Kyoto , Japan
| |
Collapse
|
31
|
Paik DT, Tian L, Lee J, Sayed N, Chen IY, Rhee S, Rhee JW, Kim Y, Wirka RC, Buikema JW, Wu SM, Red-Horse K, Quertermous T, Wu JC. Large-Scale Single-Cell RNA-Seq Reveals Molecular Signatures of Heterogeneous Populations of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells. Circ Res 2018; 123:443-450. [PMID: 29986945 PMCID: PMC6202208 DOI: 10.1161/circresaha.118.312913] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RATIONALE Human-induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) have risen as a useful tool in cardiovascular research, offering a wide gamut of translational and clinical applications. However, inefficiency of the currently available iPSC-EC differentiation protocol and underlying heterogeneity of derived iPSC-ECs remain as major limitations of iPSC-EC technology. OBJECTIVE Here, we performed droplet-based single-cell RNA sequencing (scRNA-seq) of the human iPSCs after iPSC-EC differentiation. Droplet-based scRNA-seq enables analysis of thousands of cells in parallel, allowing comprehensive analysis of transcriptional heterogeneity. METHODS AND RESULTS Bona fide iPSC-EC cluster was identified by scRNA-seq, which expressed high levels of endothelial-specific genes. iPSC-ECs, sorted by CD144 antibody-conjugated magnetic sorting, exhibited standard endothelial morphology and function including tube formation, response to inflammatory signals, and production of NO. Nonendothelial cell populations resulting from the differentiation protocol were identified, which included immature cardiomyocytes, hepatic-like cells, and vascular smooth muscle cells. Furthermore, scRNA-seq analysis of purified iPSC-ECs revealed transcriptional heterogeneity with 4 major subpopulations, marked by robust enrichment of CLDN5, APLNR, GJA5, and ESM1 genes, respectively. CONCLUSIONS Massively parallel, droplet-based scRNA-seq allowed meticulous analysis of thousands of human iPSCs subjected to iPSC-EC differentiation. Results showed inefficiency of the differentiation technique, which can be improved with further studies based on identification of molecular signatures that inhibit expansion of nonendothelial cell types. Subtypes of bona fide human iPSC-ECs were also identified, allowing us to sort for iPSC-ECs with specific biological function and identity.
Collapse
Affiliation(s)
- David T. Paik
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiology
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Lei Tian
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiology
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Jaecheol Lee
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiology
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Nazish Sayed
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiology
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | | | - Siyeon Rhee
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - June-Wha Rhee
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiology
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Youngkyun Kim
- Stanford Cardiovascular Institute
- LG Chem, Ltd, Seoul, Republic of Korea
| | - Robert C. Wirka
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiology
| | - Jan W. Buikema
- Stanford Cardiovascular Institute
- Department of Cardiology, Utrecht Regenerative Medicine Center, Utrecht University, Utrecht, Netherlands
| | - Sean M. Wu
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiology
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Kristy Red-Horse
- Stanford Cardiovascular Institute
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Thomas Quertermous
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiology
| | - Joseph C. Wu
- Stanford Cardiovascular Institute
- Department of Medicine, Division of Cardiology
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| |
Collapse
|
32
|
A Novel Fragment Derived from Laminin-411 Facilitates Proliferation and Differentiation of Odontoblast-Like Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9465383. [PMID: 29854812 PMCID: PMC5966700 DOI: 10.1155/2018/9465383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/03/2018] [Indexed: 12/13/2022]
Abstract
The aim for the present study was to evaluate the in vitro effects of iMatrix-411 in odontoblast-like cells. To that end, iMatrix-411 was coated to both nontissue culture treated- (Non-PS) and tissue culture treated-polystyrene (TCPS) multiwells. MDPC-23 cells were seeded into noncoated (control) or coated wells. Optimal coating density and cell proliferation were assessed by cell counting kit-8 (CCK-8) at day two, day three, and day five. Osteo/odontogenic differentiation was evaluated by real-time RT-PCR and alkaline phosphatase (ALP) activity at days seven and eight, respectively. Calcific deposition of cells was visualized by alizarin red staining. Data were analyzed with post hoc Tukey HSD test (p < 0.05). Optimal coating density for iMatrix-411 was 8 μg/cm2. Exposure of MDPC-23 cells to iMatrix-411 in either non-PS or TCPS significantly enhanced proliferative activity. iMatrix-411 elevated ALP activity in both types of culture plates. iMatrix-411 significantly increased the mRNA level of OCN, BSP, OPN, ALP, and DMP-1. Meanwhile, it enhanced the expression of several integrin subunits: ITGA1, ITGA5, ITGAV, ITGB1, and ITGB5. Finally, iMatrix-411 also accelerated the mineralization at day eight in Non-PS. The results indicated iMatrix-411 stimulates proliferation and favours differentiation of odontoblast-like cells.
Collapse
|
33
|
Abstract
The basement membrane is a thin but dense, sheet-like specialized type of extracellular matrix that has remarkably diverse functions tailored to individual tissues and organs. Tightly controlled spatial and temporal changes in its composition and structure contribute to the diversity of basement membrane functions. These different basement membranes undergo dynamic transformations throughout animal life, most notably during development. Numerous developmental mechanisms are regulated or mediated by basement membranes, often by a combination of molecular and mechanical processes. A particularly important process involves cell transmigration through a basement membrane because of its link to cell invasion in disease. While developmental and disease processes share some similarities, what clearly distinguishes the two is dysregulation of cells and extracellular matrices in disease. With its relevance to many developmental and disease processes, the basement membrane is a vitally important area of research that may provide novel insights into biological mechanisms and development of innovative therapeutic approaches. Here we present a review of developmental and disease dynamics of basement membranes in Caenorhabditis elegans, Drosophila, and vertebrates.
Collapse
|
34
|
Shear stress: An essential driver of endothelial progenitor cells. J Mol Cell Cardiol 2018; 118:46-69. [PMID: 29549046 DOI: 10.1016/j.yjmcc.2018.03.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023]
Abstract
The blood flow through vessels produces a tangential, or shear, stress sensed by their innermost layer (i.e., endothelium) and representing a major hemodynamic force. In humans, endothelial repair and blood vessel formation are mainly performed by circulating endothelial progenitor cells (EPCs) characterized by a considerable expression of vascular endothelial growth factor receptor 2 (VEGFR2), CD34, and CD133, pronounced tube formation activity in vitro, and strong reendothelialization or neovascularization capacity in vivo. EPCs have been proposed as a promising agent to induce reendothelialization of injured arteries, neovascularization of ischemic tissues, and endothelialization or vascularization of bioartificial constructs. A number of preconditioning approaches have been suggested to improve the regenerative potential of EPCs, including the use of biophysical stimuli such as shear stress. However, in spite of well-defined influence of shear stress on mature endothelial cells (ECs), articles summarizing how it affects EPCs are lacking. Here we discuss the impact of shear stress on homing, paracrine effects, and differentiation of EPCs. Unidirectional laminar shear stress significantly promotes homing of circulating EPCs to endothelial injury sites, induces anti-thrombotic and anti-atherosclerotic phenotype of EPCs, increases their capability to form capillary-like tubes in vitro, and enhances differentiation of EPCs into mature ECs in a dose-dependent manner. These effects are mediated by VEGFR2, Tie2, Notch, and β1/3 integrin signaling and can be abrogated by means of complementary siRNA/shRNA or selective pharmacological inhibitors of the respective proteins. Although the testing of sheared EPCs for vascular tissue engineering or regenerative medicine applications is still an unaccomplished task, favorable effects of unidirectional laminar shear stress on EPCs suggest its usefulness for their preconditioning.
Collapse
|
35
|
Watanabe D, Koyanagi‐Aoi M, Taniguchi‐Ikeda M, Yoshida Y, Azuma T, Aoi T. The Generation of Human γδT Cell-Derived Induced Pluripotent Stem Cells from Whole Peripheral Blood Mononuclear Cell Culture. Stem Cells Transl Med 2018; 7:34-44. [PMID: 29164800 PMCID: PMC5746152 DOI: 10.1002/sctm.17-0021] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 10/20/2017] [Indexed: 12/29/2022] Open
Abstract
γδT cells constitute a small proportion of lymphocytes in peripheral blood. Unlike αβT cells, the anti-tumor activities are exerted through several different pathways in a MHC-unrestricted manner. Thus, immunotherapy using γδT cells is considered to be effective for various types of cancer. Occasionally, however, ex vivo expanded cells are not as effective as expected due to cell exhaustion. To overcome the issue of T-cell exhaustion, researchers have generated induced pluripotent stem cells (iPSCs) that harbor the same T-cell receptor (TCR) genes as their original T-cells, which provide nearly limitless sources for antigen-specific cytotoxic T lymphocytes (CTLs). However, these technologies have focused on αβT cells and require a population of antigen-specific CTLs, which are purified by cell sorting with HLA-peptide multimer, as the origin of iPS cells. In the present study, we aimed to develop an efficient and convenient system for generating iPSCs that harbor rearrangements of the TCRG and TCRD gene regions (γδT-iPSCs) without cell-sorting. We stimulated human whole peripheral blood mononuclear cell (PBMC) culture using Interleukin-2 and Zoledronate to activate γδT cells. Gene transfer into those cells with the Sendai virus vector resulted in γδT cell-dominant expression of exogenous genes. The introduction of reprogramming factors into the stimulated PBMC culture allowed us to establish iPSC lines. Around 70% of the established lines carried rearrangements at the TCRG and TCRD gene locus. The γδT-iPSCs could differentiate into hematopoietic progenitors. Our technology will pave the way for new avenues toward novel immunotherapy that can be applied for various types of cancer. Stem Cells Translational Medicine 2018;7:34-44.
Collapse
Affiliation(s)
- Daisuke Watanabe
- Division of Advanced Medical Science, Graduate School of Science, Technology and InnovationKobe UniversityKobeJapan
- Department of iPS cell Applications, Kobe UniversityKobeJapan
- Division of Gastroenterology, Department of Internal Medicine, Kobe UniversityKobeJapan
| | - Michiyo Koyanagi‐Aoi
- Division of Advanced Medical Science, Graduate School of Science, Technology and InnovationKobe UniversityKobeJapan
- Department of iPS cell Applications, Kobe UniversityKobeJapan
- Center for Human Resource Development for Regenerative Medicine, Kobe University HospitalKobeJapan
| | | | - Yukiko Yoshida
- Division of Advanced Medical Science, Graduate School of Science, Technology and InnovationKobe UniversityKobeJapan
- Department of iPS cell Applications, Kobe UniversityKobeJapan
- Division of Gastroenterology, Department of Internal Medicine, Kobe UniversityKobeJapan
| | - Takeshi Azuma
- Division of Gastroenterology, Department of Internal Medicine, Kobe UniversityKobeJapan
| | - Takashi Aoi
- Division of Advanced Medical Science, Graduate School of Science, Technology and InnovationKobe UniversityKobeJapan
- Department of iPS cell Applications, Kobe UniversityKobeJapan
- Center for Human Resource Development for Regenerative Medicine, Kobe University HospitalKobeJapan
| |
Collapse
|
36
|
Hamano S, Tomokiyo A, Hasegawa D, Yoshida S, Sugii H, Mitarai H, Fujino S, Wada N, Maeda H. Extracellular Matrix from Periodontal Ligament Cells Could Induce the Differentiation of Induced Pluripotent Stem Cells to Periodontal Ligament Stem Cell-Like Cells. Stem Cells Dev 2017; 27:100-111. [PMID: 29160151 DOI: 10.1089/scd.2017.0077] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The periodontal ligament (PDL) plays an important role in anchoring teeth in the bone socket. Damage to the PDL, such as after severe inflammation, can be treated with a therapeutic strategy that uses stem cells derived from PDL tissue (PDLSCs), a strategy that has received intense scrutiny over the past decade. However, there is an insufficient number of PDLSCs within the PDL for treating such damage. Therefore, we sought to induce the differentiation of induced pluripotent stem (iPS) cells into PDLSCs as an initial step toward PDL therapy. To this end, we first induced iPS cells into neural crest (NC)-like cells. We then captured the p75 neurotrophic receptor-positive cells (iPS-NC cells) and cultured them on an extracellular matrix (ECM) produced by human PDL cells (iPS-NC-PDL cells). These iPS-NC-PDL cells showed reduced expression of embryonic stem cell and NC cell markers as compared with iPS and iPS-NC cells, and enrichment of mesenchymal stem cell markers. The cells also had a higher proliferative capacity, multipotency, and elevated expression of PDL-related markers than iPS-NC cells cultured on fibronectin and laminin (iPS-NC-FL cells) or ECM produced by human skin fibroblast cells (iPS-NC-SF cells). Overall, we present a culture method to produce high number of PDLSC-like cells from iPS cells as a first step toward a strategy for PDL regeneration.
Collapse
Affiliation(s)
- Sayuri Hamano
- 1 Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University , Fukuoka, Japan .,2 OBT Research Center, Faculty of Dental Science, Kyushu University , Fukuoka, Japan
| | - Atsushi Tomokiyo
- 3 Department of Endodontology, Kyushu University Hospital , Fukuoka, Japan
| | - Daigaku Hasegawa
- 3 Department of Endodontology, Kyushu University Hospital , Fukuoka, Japan
| | - Shinichiro Yoshida
- 3 Department of Endodontology, Kyushu University Hospital , Fukuoka, Japan
| | - Hideki Sugii
- 3 Department of Endodontology, Kyushu University Hospital , Fukuoka, Japan
| | - Hiromi Mitarai
- 3 Department of Endodontology, Kyushu University Hospital , Fukuoka, Japan
| | - Shoko Fujino
- 1 Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University , Fukuoka, Japan
| | - Naohisa Wada
- 4 Division of General Dentistry, Kyushu University Hospital , Fukuoka, Japan
| | - Hidefumi Maeda
- 1 Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University , Fukuoka, Japan .,3 Department of Endodontology, Kyushu University Hospital , Fukuoka, Japan
| |
Collapse
|