51
|
Hoffecker IT, Arima Y, Iwata H. Tuning intercellular adhesion with membrane-anchored oligonucleotides. J R Soc Interface 2019; 16:20190299. [PMID: 31662069 DOI: 10.1098/rsif.2019.0299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adhesive interactions between cells play an integral role in development, differentiation and regeneration. Existing methods for controlling cell-cell cohesion and adhesion by manipulating protein expression are constrained by biological interdependencies, e.g. coupling of cadherins to actomyosin force-feedback mechanisms. We use oligonucleotides conjugated to PEGylated lipid anchors (ssDNAPEGDPPE) to introduce artificial cell-cell adhesion that is largely decoupled from the internal cytoskeleton. We describe cell-cell doublets with a mechanical model based on isotropic, elastic deformation of spheres to estimate the adhesion at the cell-cell interface. Physical manipulation of adhesion by modulating the PEG-lipid to ssDNAPEGDPPE ratio, and conversely treating with actin-depolymerizing cytochalasin D, resulted in decreases and increases in doublet contact area, respectively. Our data are relevant to the ongoing discussion over mechanisms of tissue surface tension and in agreement with models based on opposing cortical and cohesive forces. PEG-lipid modulation of doublet geometries resulted in a well-defined curve indicating continuity, enabling prescriptive calibration for controlling doublet geometry. Our study demonstrates tuning of basic doublet adhesion, laying the foundation for more complex multicellular adhesion control independent of protein expression.
Collapse
Affiliation(s)
- Ian T Hoffecker
- Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna väg 9, Solna 171 65, Sweden
| | - Yusuke Arima
- Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.,Institute for Materials Chemistry and Engineering, Kyushu University CE41, 744 Motoka, Nishi-Ku, Fukuoka 819-0395, Japan
| | - Hiroo Iwata
- Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.,The Compass to Healthy Life Research Complex Program, RIKEN, Kobe, Japan
| |
Collapse
|
52
|
Yazdani M, Shahdadfar A, Jackson CJ, Utheim TP. A Hyaluronan Hydrogel Scaffold for Culture of Human Oral Mucosal Epithelial Cells in Limbal Stem-Cell Therapy. Bioengineering (Basel) 2019; 6:E97. [PMID: 31652804 PMCID: PMC6955856 DOI: 10.3390/bioengineering6040097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 12/26/2022] Open
Abstract
Hyaluronan (HA), a major component of the extracellular matrix, plays a key role in cell proliferation, growth, survival, polarization and differentiation. We investigated the optimization of a HA hydrogel scaffold for culture of human oral mucosal epithelial cells (OMECs) for potential application in limbal stem cell therapy. The effect of the optimized scaffold on OMEC cell sheet morphology, cell metabolic activity and expression of genes associated with stemness, adherence and cell damage was studied. The results indicate that HA hydrogels crosslinked with polyethylene glycol diacrylate (PEGDA) failed to support OMEC attachment and growth. However, HA hydrogel scaffolds dried for three days and coated with 1 mg/mL collagen IV produced a full OMEC sheet. Cell morphology was comparable to control after three weeks culture, maintaining 76% metabolic activity. Of apoptosis-related genes, the pro-apoptotic markers CASP3 and BAX2 were upregulated and downregulated, respectively, compared to control whereas the anti-apoptotic marker BCL2 was downregulated. The expression level of stemness genes ΔNp63α and ABCG2 was significantly higher than control. Genes associated with improved scar-less wound healing (integrin-V) and protection of the ocular surface (cadherin-1) had ~3-fold increased expression. These data suggest that our optimized HA-hydrogel scaffold could enhance culture of OMEC cell sheets for use in ocular reconstruction.
Collapse
Affiliation(s)
- Mazyar Yazdani
- Department of Medical Biochemistry, Oslo University Hospital, Ullevål, 0450 Oslo, Norway.
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Ullevål, 0450 Oslo, Norway.
- The Norwegian Dry Eye Clinic, 0366 Oslo, Norway.
| | - Aboulghassem Shahdadfar
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Ullevål, 0450 Oslo, Norway.
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, 0318 Oslo, Norway.
| | - Catherine Joan Jackson
- Department of Medical Biochemistry, Oslo University Hospital, Ullevål, 0450 Oslo, Norway.
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, 0318 Oslo, Norway.
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, 0450 Oslo, Norway.
| | - Tor Paaske Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Ullevål, 0450 Oslo, Norway.
- The Norwegian Dry Eye Clinic, 0366 Oslo, Norway.
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, 0318 Oslo, Norway.
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, 0450 Oslo, Norway.
- Department of Ophthalmology, Stavanger University Hospital, 4011 Stavanger, Norway.
- Department of Ophthalmology, Sørlandet Hospital Arendal, 4604 Arendal, Norway.
| |
Collapse
|
53
|
San Juan BP, Garcia-Leon MJ, Rangel L, Goetz JG, Chaffer CL. The Complexities of Metastasis. Cancers (Basel) 2019; 11:E1575. [PMID: 31623163 PMCID: PMC6826702 DOI: 10.3390/cancers11101575] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/30/2022] Open
Abstract
Therapies that prevent metastatic dissemination and tumor growth in secondary organs are severely lacking. A better understanding of the mechanisms that drive metastasis will lead to improved therapies that increase patient survival. Within a tumor, cancer cells are equipped with different phenotypic and functional capacities that can impact their ability to complete the metastatic cascade. That phenotypic heterogeneity can be derived from a combination of factors, in which the genetic make-up, interaction with the environment, and ability of cells to adapt to evolving microenvironments and mechanical forces play a major role. In this review, we discuss the specific properties of those cancer cell subgroups and the mechanisms that confer or restrict their capacity to metastasize.
Collapse
Affiliation(s)
- Beatriz P San Juan
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst 2010, Australia.
- St Vincent's Clinical School, University of New South Wales Medicine, University of New South Wales, Darlinghurst 2010, Australia.
| | - Maria J Garcia-Leon
- INSERM UMR_S1109, Tumor Biomechanics, 67000 Strasbourg, France.
- Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France.
| | - Laura Rangel
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst 2010, Australia.
- St Vincent's Clinical School, University of New South Wales Medicine, University of New South Wales, Darlinghurst 2010, Australia.
| | - Jacky G Goetz
- INSERM UMR_S1109, Tumor Biomechanics, 67000 Strasbourg, France.
- Université de Strasbourg, 67000 Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France.
| | - Christine L Chaffer
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst 2010, Australia.
- St Vincent's Clinical School, University of New South Wales Medicine, University of New South Wales, Darlinghurst 2010, Australia.
| |
Collapse
|
54
|
Mesenchymal Stem Cells in the Adult Human Liver: Hype or Hope? Cells 2019; 8:cells8101127. [PMID: 31546729 PMCID: PMC6830330 DOI: 10.3390/cells8101127] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/18/2019] [Accepted: 09/21/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic liver diseases constitute a significant economic, social, and biomedical burden. Among commonly adopted approaches, only organ transplantation can radically help patients with end-stage liver pathologies. Cell therapy with hepatocytes as a treatment for chronic liver disease has demonstrated promising results. However, quality human hepatocytes are in short supply. Stem/progenitor cells capable of differentiating into functionally active hepatocytes provide an attractive alternative approach to cell therapy for liver diseases, as well as to liver-tissue engineering, drug screening, and basic research. The application of methods generally used to isolate mesenchymal stem cells (MSCs) and maintain them in culture to human liver tissue provides cells, designated here as liver MSCs. They have much in common with MSCs from other tissues, but differ in two aspects-expression of a range of hepatocyte-specific genes and, possibly, inherent commitment to hepatogenic differentiation. The aim of this review is to analyze data regarding liver MSCs, probably another type of liver stem/progenitor cells different from hepatic stellate cells or so-called hepatic progenitor cells. The review presents an analysis of the phenotypic characteristics of liver MSCs, their differentiation and therapeutic potential, methods for isolating these cells from human liver, and discusses issues of their origin and heterogeneity. Human liver MSCs are a fascinating object of fundamental research with a potential for important practical applications.
Collapse
|
55
|
Screening of perfused combinatorial 3D microenvironments for cell culture. Acta Biomater 2019; 96:222-236. [PMID: 31255663 DOI: 10.1016/j.actbio.2019.06.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 06/23/2019] [Accepted: 06/25/2019] [Indexed: 02/08/2023]
Abstract
Biomaterials combining biochemical and biophysical cues to establish close-to-extracellular matrix (ECM) models have been explored for cell expansion and differentiation purposes. Multivariate arrays are used as material-saving and rapid-to-analyze platforms, which enable selecting hit-spotted formulations targeting specific cellular responses. However, these systems often lack the ability to emulate dynamic mechanical aspects that occur in specific biological milieus and affect physiological phenomena including stem cells differentiation, tumor progression, or matrix modulation. We report a tailor-made strategy to address the combined effect of flow and biochemical composition of three-dimensional (3D) biomaterials on cellular response. We suggest a simple-to-implement device comprising (i) a perforated platform accommodating miniaturized 3D biomaterials and (ii) a bioreactor that enables the incorporation of the biomaterial-containing array into a disposable perfusion chamber. The system was upscaled to parallelizable setups, increasing the number of analyzed platforms per independent experiment. As a proof-of-concept, porous chitosan scaffolds with 1 mm diameter were functionalized with combinations of 5 ECM and cell-cell contact-mediating proteins, relevant for bone and dental regeneration, corresponding to 32 protein combinatorial formulations. Mesenchymal stem cells adhesion and production of an early osteogenic marker were assessed on-chip on static and under-flow dynamic perfusion conditions. Different hit-spotted biomaterial formulations were detected for the different flow regimes using direct image analysis. Cell-binding proteins still poorly explored as biomaterials components - amelogenin and E-cadherin - were here shown as relevant cell response modulators. Their combination with ECM cell-binding proteins - fibronectin, vitronectin, and type 1 collagen - rendered specific biomaterial combinations with high cell adhesion and ALP production under flow. The developed versatile system may be targeted at widespread tissue regeneration applications, and as a disease model/drug screening platform. STATEMENT OF SIGNIFICANCE: A perfusion system that enables cell culture in arrays of three-dimensional biomaterials under dynamic flow is reported. The effect of 31 cell-binding protein combinations in the adhesion and alkaline phosphatase (ALP) production of mesenchymal stem cells was assessed using a single bioreactor chamber. Flow perfusion was not only assessed as a classical enhancer/accelerator of cell growth and early osteogenic differentiation. We hypothesized that flow may affect cell-protein interactions, and that key components driving cell response may differ under static or dynamic regimes. Indeed, hit-spotted formulations that elicited highest cell attachment and ALP production on static cell culture differed from the ones detected for dynamic flow assays. The impacting role of poorly studied proteins as E-cadherin and amelogenin as biomaterial components was highlighted.
Collapse
|
56
|
Diao S, Yang H, Cao Y, Yang D, Fan Z. IGF2 enhanced the osteo-/dentinogenic and neurogenic differentiation potentials of stem cells from apical papilla. J Oral Rehabil 2019; 47 Suppl 1:55-65. [PMID: 31291686 DOI: 10.1111/joor.12859] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES In dental tissue engineering, niche is important for maintaining stem cell function and regenerating the dental tissues. However, there is limited knowledge for the growth factors in niche to maintain the function of stem cells. In this study, we investigated the effect of IGF2, a growth factor in stem cells from apical papilla (SCAPs) niche, on differentiation and proliferation potentials of SCAPs. MATERIALS AND METHODS Recombinant human IGF2 protein (rhIGF2) was used. Cell counting kit-8 assay, Carboxyfluorescein succinimidyl ester assay, alkaline phosphatase (ALP) activity, Alizarin Red staining, quantitative calcium analysis, immunofluorescence staining and real-time RT-PCR were performed to investigate the cell proliferation and differentiation potentials of SCAPs. And proteomic analysis was used to identify the differential secreted proteins. RESULTS By ALP activity assay, we found that 5 ng/mL rhIGF2 might be the optimal concentration for treatment. Then, Alizarin Red staining, quantitative calcium analysis and osteogenesis-related gene expression results showed that 5 ng/mL rhIGF2 could enhance the osteo-/dentinogenic differentiation potentials in SCAPs. Immunofluorescence staining and real-time RT-PCR results showed that neurogenic markers were significantly induced by 5 ng/mL rhIGF2 in SCAPs. Then, CCK-8 assay and CFSE assay results showed that 5 ng/mL rhIGF2 could enhance the cell proliferation in SCAPs. Furthermore, proteomic analysis showed that IGF2 could induce some secreted proteins which function related to the osteogenesis, neurogenesis and cell proliferation. CONCLUSIONS Our results identified that IGF2 might be the potential mediator in niche to promote SCAP function and dental tissue regeneration.
Collapse
Affiliation(s)
- Shu Diao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Department of Pediatric dentistry, Capital Medical University School of Stomatology, Beijing, China
| | - Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Yangyang Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Dongmei Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Department of Pediatric dentistry, Capital Medical University School of Stomatology, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
57
|
Godoy‐Parejo C, Deng C, Liu W, Chen G. Insulin Stimulates PI3K/AKT and Cell Adhesion to Promote the Survival of Individualized Human Embryonic Stem Cells. Stem Cells 2019; 37:1030-1041. [PMID: 31021484 PMCID: PMC6852186 DOI: 10.1002/stem.3026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 03/30/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022]
Abstract
Insulin is present in most maintenance media for human embryonic stem cells (hESCs), but little is known about its essential role in the cell survival of individualized cells during passage. In this article, we show that insulin suppresses caspase cleavage and apoptosis after dissociation. Insulin activates insulin-like growth factor (IGF) receptor and PI3K/AKT cascade to promote cell survival and its function is independent of rho-associated protein kinase regulation. During niche reformation after passaging, insulin activates integrin that is essential for cell survival. IGF receptor colocalizes with focal adhesion complex and stimulates protein phosphorylation involved in focal adhesion formation. Insulin promotes cell spreading on matrigel-coated surfaces and suppresses myosin light chain phosphorylation. Further study showed that insulin is also required for the cell survival on E-cadherin coated surface and in suspension, indicating its essential role in cell-cell adhesion. This work highlights insulin's complex roles in signal transduction and niche re-establishment in hESCs. Stem Cells 2019;37:1030-1041.
Collapse
Affiliation(s)
- Carlos Godoy‐Parejo
- Centre of Reproduction, Development, and Aging, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
| | - Chunhao Deng
- Centre of Reproduction, Development, and Aging, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
| | - Weiwei Liu
- Centre of Reproduction, Development, and Aging, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
- Bioimaging and Stem Cell Core Facility, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
| | - Guokai Chen
- Centre of Reproduction, Development, and Aging, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
- Bioimaging and Stem Cell Core Facility, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
- Institute of Translational Medicine, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
| |
Collapse
|
58
|
Greuel S, Hanci G, Böhme M, Miki T, Schubert F, Sittinger M, Mandenius CF, Zeilinger K, Freyer N. Effect of inoculum density on human-induced pluripotent stem cell expansion in 3D bioreactors. Cell Prolif 2019; 52:e12604. [PMID: 31069891 PMCID: PMC6668975 DOI: 10.1111/cpr.12604] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/15/2019] [Accepted: 02/20/2019] [Indexed: 12/18/2022] Open
Abstract
Objective For optimized expansion of human‐induced pluripotent stem cells (hiPSCs) with regards to clinical applications, we investigated the influence of the inoculum density on the expansion procedure in 3D hollow‐fibre bioreactors. Materials and Methods Analytical‐scale bioreactors with a cell compartment volume of 3 mL or a large‐scale bioreactor with a cell compartment volume of 17 mL were used and inoculated with either 10 × 106 or 50 × 106 hiPSCs. Cells were cultured in bioreactors over 15 days; daily measurements of biochemical parameters were performed. At the end of the experiment, the CellTiter‐Blue® Assay was used for culture activity evaluation and cell quantification. Also, cell compartment sections were removed for gene expression and immunohistochemistry analysis. Results The results revealed significantly higher values for cell metabolism, cell activity and cell yields when using the higher inoculation number, but also a more distinct differentiation. As large inoculation numbers require cost and time‐extensive pre‐expansion, low inoculation numbers may be used preferably for long‐term expansion of hiPSCs. Expansion of hiPSCs in the large‐scale bioreactor led to a successful production of 5.4 × 109 hiPSCs, thereby achieving sufficient cell amounts for clinical applications. Conclusions In conclusion, the results show a significant effect of the inoculum density on cell expansion, differentiation and production of hiPSCs, emphasizing the importance of the inoculum density for downstream applications of hiPSCs. Furthermore, the bioreactor technology was successfully applied for controlled and scalable production of hiPSCs for clinical use.
Collapse
Affiliation(s)
- Selina Greuel
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Güngör Hanci
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mike Böhme
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Toshio Miki
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - Michael Sittinger
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carl-Fredrik Mandenius
- Division of Biotechnology, Department of Physics, Chemistry and Biology (IFM), Linköping University, Linköping, Sweden
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nora Freyer
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
59
|
Nie Y, Wang W, Xu X, Zou J, Bhuvanesh T, Schulz B, Ma N, Lendlein A. Enhancement of human induced pluripotent stem cells adhesion through multilayer laminin coating. Clin Hemorheol Microcirc 2019; 70:531-542. [PMID: 30347612 DOI: 10.3233/ch-189318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bioengineered cell substrates are a highly promising tool to govern the differentiation of stem cells in vitro and to modulate the cellular behavior in vivo. While this technology works fine for adult stem cells, the cultivation of human induced pluripotent stem cells (hiPSCs) is challenging as these cells typically show poor attachment on the bioengineered substrates, which among other effects causes substantial cell death. Thus, very limited types of surfaces have been demonstrated suitable for hiPSC cultures. The multilayer coating approach that renders the surface with diverse chemical compositions, architectures, and functions can be used to improve the adhesion of hiPSCs on the bioengineered substrates. We hypothesized that a multilayer formation based on the attraction of molecules with opposite charges could functionalize the polystyrene (PS) substrates to improve the adhesion of hiPSCs. Polymeric substrates were stepwise coated, first with dopamine to form a polydopamine (PDA) layer, second with polylysine and last with Laminin-521. The multilayer formation resulted in the variation of hydrophilicity and chemical functionality of the surfaces. Hydrophilicity was detected using captive bubble method and the amount of primary and secondary amines on the surface was quantified by fluorescent staining. The PDA layer effectively immobilized the upper layers and thereby improved the attachment of hiPSCs. Cell adhesion was enhanced on the surfaces coated with multilayers, as compared to those without PDA and/or polylysine. Moreover, hiPSCs spread well over this multilayer laminin substrate. These cells maintained their proliferation capacity and differentiation potential. The multilayer coating strategy is a promising attempt for engineering polymer-based substrates for the cultivation of hiPSCs and of interest for expanding the application scope of hiPSCs.
Collapse
Affiliation(s)
- Yan Nie
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Jie Zou
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Thanga Bhuvanesh
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Burkhard Schulz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Teltow, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Teltow, Germany
| |
Collapse
|
60
|
Shuzui E, Kim MH, Kino-oka M. Anomalous cell migration triggers a switch to deviation from the undifferentiated state in colonies of human induced pluripotent stems on feeder layers. J Biosci Bioeng 2019; 127:246-255. [DOI: 10.1016/j.jbiosc.2018.07.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/02/2018] [Accepted: 07/24/2018] [Indexed: 01/07/2023]
|
61
|
FOSB⁻PCDHB13 Axis Disrupts the Microtubule Network in Non-Small Cell Lung Cancer. Cancers (Basel) 2019; 11:cancers11010107. [PMID: 30658436 PMCID: PMC6357195 DOI: 10.3390/cancers11010107] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 12/31/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is among the leading causes of human mortality. One reason for high rates of NSCLC mortality is that drug resistance is a major problem for both conventional chemotherapies and less-toxic targeted therapies. Thus, novel mechanistic insights into disease pathogenesis may benefit the development of urgently needed therapies. Here we show that FBJ murine osteosarcoma viral oncogene homolog B (FOSB) was induced by an antimicrobial peptide, tilapia piscidin-4 (TP4), through the dysregulation of mitochondrial Ca2+ homeostasis in NSCLC cells. Transcriptomic, chromatin immunoprecipitation quantitative PCR, and immunocytochemical studies reveal that protocadherin-β13 (PCDHB13) as a target of FOSB that was functionally associated with microtubule. Overexpression of either PCDHB13 or FOSB attenuated NSCLC growth and survival in vitro and in vivo. Importantly, downregulation of both FOSB and PCDHB13 was observed in NSCLC patients and was negatively correlated with pathological grade. These findings introduce the FOSB⁻PCDHB13 axis as a novel tumor suppressive pathway in NSCLC.
Collapse
|
62
|
Maintenance of an undifferentiated state of human-induced pluripotent stem cells through botulinum hemagglutinin-mediated regulation of cell behavior. J Biosci Bioeng 2019; 127:744-751. [PMID: 30660482 DOI: 10.1016/j.jbiosc.2018.11.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/14/2018] [Accepted: 11/29/2018] [Indexed: 01/01/2023]
Abstract
Applications of human induced pluripotent stem cell (hiPSC) culture are impaired by problems with long term maintenance of pluripotency. In this study, we report that exposure to botulinum hemagglutinin (HA), an E-cadherin function-blocking agent, suppressed deviation from an undifferentiated state in hiPSC colonies. Time-lapse imaging of live cells revealed that cells in central regions of colonies moved slowly and underwent a morphological change to a cobblestone-like shape via interaction between contacting cells, forming dense, multiple layers. Staining and migration analysis showed that actin stress fibers and paxillin spots were diminished in colony central regions, and this was associated with alteration of cellular morphology and migratory behavior. However, in culture with HA exposure, cells in the central and peripheral regions of hiPSC colonies were migratory and arranged in loose monolayers, resulting in relatively uniform dispersion of cells in colonies. We also found that a well-organized network of actin stress fibers was of significance in the central and peripheral regions of a colony, resulting in activation of paxillin and E-cadherin expression in hiPSCs. After routine application of HA for serial passages, hiPSCs remained pluripotent and capable of differentiating into all three germ layers. These observations indicate that relaxation of cell-cell junctions by HA induced rearrangements of the cytoskeleton and cell adhesion in hiPSC colonies by promoting migratory behaviors. These results suggest that this simple and readily reproducible culture strategy is a potentially useful tool for improving the robust and scalable maintenance of undifferentiated hiPSC cultures.
Collapse
|
63
|
Assessment of the Influence of Acetic Acid Residue on Type I Collagen during Isolation and Characterization. MATERIALS 2018; 11:ma11122518. [PMID: 30545004 PMCID: PMC6316942 DOI: 10.3390/ma11122518] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 12/03/2018] [Accepted: 12/10/2018] [Indexed: 01/13/2023]
Abstract
Various methods for isolation of type I collagen using acids, bases, enzymes, and their combinations have been applied. However, a lack of standardization exists among type I collagens isolated by various approaches. Consequently, in this study, we assessed the influence of acetic acid residue on type I collagen isolated by pepsin-acetic acid treatment, the fabrication of collagen-based porous scaffolds, and the seeded cells on collagen scaffolds. Unlike the isolated collagen dialyzed by deionized water (DDW), collagen dialyzed by 0.5 M acetic acid (DAC) exhibited structural and thermal denaturation. Both DDW- and DAC-based porous scaffolds at all collagen concentrations (0.5, 1 and 2% w/v) showed the high degree of porosity (>98%), and their pore morphologies were comparable at the same concentrations. However, the DDW- and DAC-based collagen scaffolds displayed significant differences in their physical properties (weight, thickness, and volume) and swelling behaviors. In particular, the weight losses induced by mechanical stimulation reflected the high degradation of DAC-collagen scaffolds. In cell culture experiments using adipose-derived stem cells (ADSCs), the characteristics of mesenchymal stem cell (MSC) did not change in both DDW- and DAC-collagen scaffolds for 10 days, although cells proliferated less in the DAC-collagen scaffolds. Our results suggest that the elimination of acetic acid residue from isolated collagen is recommended to produce collagen scaffolds that provide a stable environment for cells and cell therapy-related applications.
Collapse
|
64
|
Reprogramming mechanisms influence the maturation of hematopoietic progenitors from human pluripotent stem cells. Cell Death Dis 2018; 9:1090. [PMID: 30356076 PMCID: PMC6200746 DOI: 10.1038/s41419-018-1124-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/26/2018] [Accepted: 10/01/2018] [Indexed: 12/28/2022]
Abstract
Somatic cell nuclear transfer (SCNT) or the forced expression of transcription factors can be used to generate autologous pluripotent stem cells (PSCs). Although transcriptomic and epigenomic comparisons of isogenic human NT-embryonic stem cells (NT-ESCs) and induced PSCs (iPSCs) in the undifferentiated state have been reported, their functional similarities and differentiation potentials have not been fully elucidated. Our study showed that NT-ESCs and iPSCs derived from the same donors generally displayed similar in vitro commitment capacity toward three germ layer lineages as well as proliferative activity and clonogenic capacity. However, the maturation capacity of NT-ESC-derived hematopoietic progenitors was significantly greater than the corresponding capacity of isogenic iPSC-derived progenitors. Additionally, donor-dependent variations in hematopoietic specification and commitment capacity were observed. Transcriptome and methylome analyses in undifferentiated NT-ESCs and iPSCs revealed a set of genes that may influence variations in hematopoietic commitment and maturation between PSC lines derived using different reprogramming methods. Here, we suggest that genetically identical iPSCs and NT-ESCs could be functionally unequal due to differential transcription and methylation levels acquired during reprogramming. Our proof-of-concept study indicates that reprogramming mechanisms and genetic background could contribute to diverse functionalities between PSCs.
Collapse
|
65
|
Libby AR, Joy DA, So PL, Mandegar MA, Muncie JM, Mendoza-Camacho FN, Weaver VM, Conklin BR, McDevitt TC. Spatiotemporal mosaic self-patterning of pluripotent stem cells using CRISPR interference. eLife 2018; 7:36045. [PMID: 30298816 PMCID: PMC6177255 DOI: 10.7554/elife.36045] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 09/09/2018] [Indexed: 12/22/2022] Open
Abstract
Morphogenesis involves interactions of asymmetric cell populations to form complex multicellular patterns and structures comprised of distinct cell types. However, current methods to model morphogenic events lack control over cell-type co-emergence and offer little capability to selectively perturb specific cell subpopulations. Our in vitro system interrogates cell-cell interactions and multicellular organization within human induced pluripotent stem cell (hiPSC) colonies. We examined effects of induced mosaic knockdown of molecular regulators of cortical tension (ROCK1) and cell-cell adhesion (CDH1) with CRISPR interference. Mosaic knockdown of ROCK1 or CDH1 resulted in differential patterning within hiPSC colonies due to cellular self-organization, while retaining an epithelial pluripotent phenotype. Knockdown induction stimulates a transient wave of differential gene expression within the mixed populations that stabilized in coordination with observed self-organization. Mosaic patterning enables genetic interrogation of emergent multicellular properties, which can facilitate better understanding of the molecular pathways that regulate symmetry-breaking during morphogenesis. Embryos begin as a collection of similar cells, which progress in stages to form a huge variety of cell types in particular arrangements. These patterns of cells give rise to the different tissues and organs that make up the body. Although we often use ‘model’ organisms such as mice and frogs to study how embryos develop, our species has evolved unique ways to control organ development. Investigating these processes is difficult: we cannot experiment on human embryos, and our development is hard to recreate in test tubes. As a result, we do not fully understand how developing human cells specialize and organize. Libby et al. have now created a new system to study how different genes control cell organization. The system uses human pluripotent stem cells – cells that have the ability to specialize into any type of cell. Some of the stem cells are modified using a technique called inducible CRISPR interference, which makes it possible to reduce the activity of certain genes in these cells. Libby et al. used this technique to investigate how changes to the activity of two genes – called ROCK1 and CDH1 – affect how a mixed group of stem cells organized themselves. Cells that lacked ROCK1 formed bands near the edges of the group. Cells that lacked CDH1 segregated themselves from other cells, forming ‘islands’ inside the main group. The cells retained their ability to specialize into any type of cell after forming these patterns. However, specific groups of cells were more likely to become certain cell types. The method developed by Libby et al. can be used to study a range of complex tissue development and cell organization processes. Future work could create human tissue model systems for research into human disease or drug development.
Collapse
Affiliation(s)
- Ashley Rg Libby
- Gladstone Institute of Cardiovascular Disease, San Francisco, United States.,Developmental and Stem Cell Biology Graduate Program, University of California San Francisco, San Francisco, United States
| | - David A Joy
- Gladstone Institute of Cardiovascular Disease, San Francisco, United States.,Graduate Program in Bioengineering, University of California Berkeley, University of California San Francisco, San Francisco, United States
| | - Po-Lin So
- Gladstone Institute of Cardiovascular Disease, San Francisco, United States
| | | | - Jonathon M Muncie
- Graduate Program in Bioengineering, University of California Berkeley, University of California San Francisco, San Francisco, United States.,Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, United States
| | | | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, United States
| | - Bruce R Conklin
- Gladstone Institute of Cardiovascular Disease, San Francisco, United States.,Department of Medicine, Division of Genomic Medicine, University of California, San Francisco, United States
| | - Todd C McDevitt
- Gladstone Institute of Cardiovascular Disease, San Francisco, United States.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, United States
| |
Collapse
|
66
|
Gao X, Sprando RL, Yourick JJ. A Rapid and Highly Efficient Method for the Isolation, Purification, and Passaging of Human-Induced Pluripotent Stem Cells. Cell Reprogram 2018; 20:282-288. [DOI: 10.1089/cell.2018.0022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Xiugong Gao
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland
| | - Robert L. Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland
| | - Jeffrey J. Yourick
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland
| |
Collapse
|
67
|
Role of cell-secreted extracellular matrix formation in aggregate formation and stability of human induced pluripotent stem cells in suspension culture. J Biosci Bioeng 2018; 127:372-380. [PMID: 30249415 DOI: 10.1016/j.jbiosc.2018.08.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/16/2018] [Accepted: 08/23/2018] [Indexed: 11/22/2022]
Abstract
Clinical and industrial applications require large quantities of human induced pluripotent stem cells (hiPSCs); however, little is known regarding the mechanisms governing aggregate formation and stability in suspension culture. To address this, we determined differences in growth processes among hiPSC lines in suspension culture. Using an hiPSC aggregate suspension culture system, hiPSCs from different lines formed multicellular aggregates classified as large compact or small loose based on their size and morphology. Time-lapse observation of the growth processes of two different hiPSC lines revealed that the balance between cell division and the extent of subsequent cell death determined the final size and morphology of aggregates. Comparison of the cell survival and death of two hiPSC lines showed that the formation of small, loose aggregates was due to continued cell death during the exponential phase of growth, with apoptotic cells extruded from growing hiPSC aggregates by the concerted contraction of their neighbors. Western blot and immunofluorescent staining revealed that aggregate morphology and proliferative ability relied to a considerable extent upon secretion of the extracellular matrix (ECM). hiPSCs forming large compact and stable aggregates showed enhanced production of collagen type I in suspension culture at 120 h. Furthermore, these aggregates exhibited higher expression of E-cadherin and proliferation marker Ki-67 as compared with levels observed in small and loose aggregates at 120 h. These findings indicated that differences in both aggregate formation and stability in suspension culture among hiPSC lines were caused by differences in ECM secretion capacity.
Collapse
|
68
|
Zhang D, Lee H, Wang X, Rai A, Groot M, Jin Y. Exosome-Mediated Small RNA Delivery: A Novel Therapeutic Approach for Inflammatory Lung Responses. Mol Ther 2018; 26:2119-2130. [PMID: 30005869 PMCID: PMC6127502 DOI: 10.1016/j.ymthe.2018.06.007] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 06/04/2018] [Accepted: 06/07/2018] [Indexed: 02/08/2023] Open
Abstract
Exosomes (EXOs) are a type of extracellular nanovesicles released from living cells. Accumulating evidence suggests that EXOs are involved in the pathogenesis of human diseases, including lung conditions. In recent years, the potential of EXO-mediated drug delivery has gained increasing interest. In this report, we investigated whether inhaled EXOs serve as an efficient and practical delivery vehicle to activate or inhibit alveolar macrophages (AMs), subsequently modulating pulmonary immune responses. We first identified the recipient cells of the inhaled EXOs, which were labeled with PKH26. We found that only lung macrophages efficiently take up intratracheally instilled EXOs in vivo. Using modified calcium chloride-mediated transformation, we manipulated small RNA molecules in serum-derived EXOs, including siRNAs, microRNA (miRNA) mimics, and miRNA inhibitors. Via intratracheal instillation, we successfully delivered siRNA and miRNA mimics or inhibitors into lung macrophages using the serum-derived EXOs as vehicles. Furthermore, EXO siRNA or miRNA molecules are functional in modulating LPS-induced lung inflammation in vivo. Beneficially, serum-derived EXOs themselves do not trigger lung immune responses, adding more favorable features to serve as drug delivery agents. Collectively, we developed a novel protocol using serum-derived EXOs to deliver designated small RNA molecules into lung macrophages in vivo, potentially shedding light on future gene therapy of human lung diseases.
Collapse
Affiliation(s)
- Duo Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA
| | - Heedoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA
| | - Xiaoyun Wang
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Ashish Rai
- North Shore Medical Center, Salem Hospital, Boston, MA 01970, USA
| | - Michael Groot
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA.
| |
Collapse
|
69
|
Han SS, Shim HE, Park SJ, Kim BC, Lee DE, Chung HM, Moon SH, Kang SW. Safety and Optimization of Metabolic Labeling of Endothelial Progenitor Cells for Tracking. Sci Rep 2018; 8:13212. [PMID: 30181604 PMCID: PMC6123424 DOI: 10.1038/s41598-018-31594-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 08/20/2018] [Indexed: 12/13/2022] Open
Abstract
Metabolic labeling is one of the most powerful methods to label the live cell for in vitro and in vivo tracking. However, the cellular mechanisms by modified glycosylation due to metabolic agents are not fully understood. Therefore, metabolic labeling has not yet been widely used in EPC tracking and labeling. In this study, cell functional properties such as proliferation, migration and permeability and gene expression patterns of metabolic labeling agent-treated hUCB-EPCs were analyzed to demonstrate cellular effects of metabolic labeling agents. As the results, 10 μM Ac4ManNAz treatment had no effects on cellular function or gene regulations, however, higher concentration of Ac4ManNAz (>20 μM) led to the inhibition of functional properties (proliferation rate, viability and rate of endocytosis) and down-regulation of genes related to cell adhesion, PI3K/AKT, FGF and EGFR signaling pathways. Interestingly, the new blood vessel formation and angiogenic potential of hUCB-EPCs were not affected by Ac4ManNAz concentration. Based on our results, we suggest 10 μM as the optimal concentration of Ac4ManNAz for in vivo hUCB-EPC labeling and tracking. Additionally, we expect that our approach can be used for understanding the efficacy and safety of stem cell-based therapy in vivo.
Collapse
Affiliation(s)
- Sang-Soo Han
- Predictive Model Research Center, Korea Institute of Toxicology, Daejeon, Korea
| | - Hye-Eun Shim
- Predictive Model Research Center, Korea Institute of Toxicology, Daejeon, Korea
| | - Soon-Jung Park
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Byoung-Chul Kim
- The Genomics Institute, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Dong-Eun Lee
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeonbuk, Korea
| | - Hyung-Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Sung-Hwan Moon
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea.
| | - Sun-Woong Kang
- Predictive Model Research Center, Korea Institute of Toxicology, Daejeon, Korea.
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, Korea.
| |
Collapse
|
70
|
Topal T, Hong X, Xue X, Fan Z, Kanetkar N, Nguyen JT, Fu J, Deng CX, Krebsbach PH. Acoustic Tweezing Cytometry Induces Rapid Initiation of Human Embryonic Stem Cell Differentiation. Sci Rep 2018; 8:12977. [PMID: 30154528 PMCID: PMC6113316 DOI: 10.1038/s41598-018-30939-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/08/2018] [Indexed: 02/05/2023] Open
Abstract
Mechanical forces play critical roles in influencing human embryonic stem cell (hESC) fate. However, it remains largely uncharacterized how local mechanical forces influence hESC behavior in vitro. Here, we used an ultrasound (US) technique, acoustic tweezing cytometry (ATC), to apply targeted cyclic subcellular forces to hESCs via integrin-bound microbubbles (MBs). We found that ATC-mediated cyclic forces applied for 30 min to hESCs near the edge of a colony induced immediate global responses throughout the colony, suggesting the importance of cell-cell connection in the mechanoresponsiveness of hESCs to ATC-applied forces. ATC application generated increased contractile force, enhanced calcium activity, as well as decreased expression of pluripotency transcription factors Oct4 and Nanog, leading to rapid initiation of hESC differentiation and characteristic epithelial-mesenchymal transition (EMT) events that depend on focal adhesion kinase (FAK) activation and cytoskeleton (CSK) tension. These results reveal a unique, rapid mechanoresponsiveness and community behavior of hESCs to integrin-targeted cyclic forces.
Collapse
Affiliation(s)
- Tuğba Topal
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xiaowei Hong
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Zhenzhen Fan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Biomedical Engineering, Tianjin University, Tianjin, P.R. China
| | - Ninad Kanetkar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joe T Nguyen
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jianping Fu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Cheri X Deng
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA. .,Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Paul H Krebsbach
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA. .,Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109, USA. .,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA. .,Section of Periodontics, University of California, Los Angeles School of Dentistry, Los Angeles, CA, 90095, USA.
| |
Collapse
|
71
|
Liao S, Gan L, Qin W, Liu C, Mei Z. Inhibition of GSK3 and MEK induced cancer stem cell generation via the Wnt and MEK signaling pathways. Oncol Rep 2018; 40:2005-2013. [PMID: 30066938 PMCID: PMC6111576 DOI: 10.3892/or.2018.6600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 07/17/2018] [Indexed: 01/06/2023] Open
Abstract
Cancer stem cells (CSCs) are considered to be tumor-initiating cells, responsible for tumor invasive growth and dissemination to distant organ sites. Typically, radiation treatment and chemotherapy should target CSCs. However, current research investigating CSCs is impeded by the difficulty of isolating pure CSCs and maintaining them in vitro. In the present study, the synergistic inhibition of glycogen synthase kinase 3 and mitogen-activated protein kinase kinase using small molecules, CHIR99021 and PD184352, efficiently generated CSCs from immortalized human mammary epithelial cells (HMLEs) and resulted in the acquisition of mesenchymal traits and the expression of epithelial-mesenchymal transition markers. The cell proliferation, invasion and migration of HMLE cells were significantly promoted by CHIR99021 and PD184352 (P<0.05). Furthermore, the cell cycle was shifted from the G0/G1 phase to the G2/M phase, and the apoptotic rate was suppressed in HMLE cells following treatment with CHIR99021 and PD184352. Compared with control group, the stimulated cells exhibited an increased ability to form mammospheres and regenerate a tumor. In addition to these properties, the induced cells also exhibited notable chemotherapy resistance. In vivo, the treatment of cells with CHIR99021 and PD184352 promoted the growth of HMLE-engrafted tumor types. These results provide a practical strategy for the generation of CSCs using small molecules in vitro, which provides a cell resource that may be used for drug screening. Additionally, the present results additionally highlighted the synergistic functions of Wnt and mitogen-activated protein kinase kinase signaling pathways in tumorigenesis.
Collapse
Affiliation(s)
- Shengtao Liao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Li Gan
- Teaching and Research Section of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wanxiang Qin
- Department of Pain Management, Southwest Hospital, The First Affiliated Hospital of The Third Military Medical University, Chongqing 400038, P.R. China
| | - Chang Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Zhechuan Mei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
72
|
Comparison of growth kinetics between static and dynamic cultures of human induced pluripotent stem cells. J Biosci Bioeng 2018; 125:736-740. [DOI: 10.1016/j.jbiosc.2018.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 01/26/2023]
|
73
|
Huang D, Chen J, Yang C, Wang M. TPX2 silencing mediated by joint action of microvesicles and ultrasonic radiation inhibits the migration and invasion of SKOV3 cells. Mol Med Rep 2018; 17:7627-7635. [PMID: 29620263 PMCID: PMC5983958 DOI: 10.3892/mmr.2018.8810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 11/13/2017] [Indexed: 01/07/2023] Open
Abstract
Ovarian cancer, with its high morbidity, has one of the highest mortality rates among gynecological malignant tumors. Overexpression of targeting protein for Xklp2 (TPX2) has been identified in numerous malignant tumors. The present study sought to determine whether TPX2 silencing inhibited the growth and metastasis of ovarian cancer cells, and whether microvesicles‑ and ultrasonic radiation‑mediated small interfering (si)RNA‑TPX2 transfection may improve the therapeutic effect. The SKOV3 cell line, derived from papillary serous cytadenocarcinoma of the human ovary, was selected as a cell model. Cells were divided into five groups: Control, siRNA‑TPX2, siRNA‑TPX2 + microvesicle (M), siRNA‑TPX2 + ultrasonic irradiation (UI), and siRNA‑TPX2 + M + UI. Cell viability was evaluated under the aforementioned conditions via the Cell Counting kit 8 (CCK8) assay. Cell migration and invasion were detected using Transwell assays. The expression levels of associated genes, including epithelial cadherin (E‑cadherin), metalloproteinase inhibitor 2 (TIMP‑2), metastasis associated 1 (MTA1) and matrix metallopeptidase 2 (MMP2), were analyzed using reverse transcription‑quantitative polymerase chain reaction analysis and western blotting. MMP2 activity was determined using a gelatin zymography assay. The results suggested that TPX2 serves an important role in the development of SKOV3 cells; it is additionally able to inhibit cell migration and invasion by upregulating E‑cadherin and TIMP2, downregulating MMP2 and MTA1, and inhibiting the phosphorylation of p38 and c‑Jun N‑terminal kinase. The inhibitory effect of siRNA‑TPX2 on SKOV3 cellular metastasis in the presence of microvesicles and ultrasonic radiation was observed to be improved compared with the control. It is proposed that the combination of microvesicles and ultrasonic radiation with TPX2 silencing has the potential to be an effective gene therapy against ovarian cancer.
Collapse
Affiliation(s)
- Dong Huang
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Jianmin Chen
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Cuiyu Yang
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Minzhen Wang
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
74
|
Candiello J, Grandhi TSP, Goh SK, Vaidya V, Lemmon-Kishi M, Eliato KR, Ros R, Kumta PN, Rege K, Banerjee I. 3D heterogeneous islet organoid generation from human embryonic stem cells using a novel engineered hydrogel platform. Biomaterials 2018; 177:27-39. [PMID: 29883914 DOI: 10.1016/j.biomaterials.2018.05.031] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 05/19/2018] [Indexed: 01/05/2023]
Abstract
Organoids, which exhibit spontaneous organ specific organization, function, and multi-cellular complexity, are in essence the in vitro reproduction of specific in vivo organ systems. Recent work has demonstrated human pluripotent stem cells (hPSCs) as a viable regenerative cell source for tissue-specific organoid engineering. This is especially relevant for engineering islet organoids, due to the recent advances in generating functional beta-like cells from human pluripotent stem cells. In this study, we report specific engineering of regenerative islet organoids of precise size and cellular heterogeneity, using a novel hydrogel system, Amikagel. Amikagel facilitated controlled and spontaneous aggregation of human embryonic stem cell derived pancreatic progenitor cells (hESC-PP) into robust homogeneous spheroids. This platform further allowed fine control over the integration of multiple cell populations to produce heterogeneous spheroids, which is a necessity for complex organoid engineering. Amikagel induced hESC-PP spheroid formation enhanced pancreatic islet-specific Pdx-1 and NKX6.1 gene and protein expression, while also increasing the percentage of committed population. hESC-PP spheroids were further induced towards mature beta-like cells which demonstrated increased Beta-cell specific INS1 gene and C-peptide protein expression along with functional insulin production in response to in vitro glucose challenge. Further integration of hESC-PP with biologically relevant supporting endothelial cells resulted in multicellular organoids which demonstrated spontaneous maturation towards islet-specific INS1 gene and C-peptide protein expression along with a significantly developed extracellular matrix support system. These findings establish Amikagel -facilitated platform ideal for islet organoid engineering.
Collapse
Affiliation(s)
- Joseph Candiello
- Department of Bioengineering, University of Pittsburgh, PA, United States
| | | | - Saik Kia Goh
- Department of Bioengineering, University of Pittsburgh, PA, United States
| | - Vimal Vaidya
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA, United States
| | - Maya Lemmon-Kishi
- Department of Bioengineering, University of Pittsburgh, PA, United States
| | - Kiarash Rahmani Eliato
- Department of Physics, Center for Biological Physics, and Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Robert Ros
- Department of Physics, Center for Biological Physics, and Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Prashant N Kumta
- Department of Bioengineering, University of Pittsburgh, PA, United States; Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA, United States; Department of Mechanical Engineering and Material Science, University of Pittsburgh, PA, United States; Center for Complex Engineered Multifunctional Materials, University of Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, United States
| | - Kaushal Rege
- Chemical Engineering, Arizona State University, Tempe, AZ, United States
| | - Ipsita Banerjee
- Department of Bioengineering, University of Pittsburgh, PA, United States; Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, United States.
| |
Collapse
|
75
|
Curtis TM, Hannett JM, Harman RM, Puoplo NA, Van de Walle GR. The secretome of adipose-derived mesenchymal stem cells protects SH-SY5Y cells from arsenic-induced toxicity, independent of a neuron-like differentiation mechanism. Neurotoxicology 2018; 67:54-64. [PMID: 29660375 DOI: 10.1016/j.neuro.2018.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/15/2018] [Accepted: 04/11/2018] [Indexed: 01/01/2023]
Abstract
Arsenic exposure through contaminated food, water, and air causes irreversible neural damage and affects millions of people worldwide. Several studies have demonstrated that the secreted factors (secretome) from mesenchymal stromal/stem cells (MSCs) can promote neural recovery after several forms of injury including stroke and neurodegenerative diseases. The present study was conducted to determine if the secretome from adipose-derived MSCs (ADSCs) prevents arsenic damage to SH-SY5Y cells. To this end, human neuroblastoma cells (SH-SY5Y) were pre-treated with the secretome from ADSCs and then challenged with different concentrations of arsenic. After various doses and exposure times, the extent of neuronal injury was assessed using MTT reduction and LDH release assays as well as LIVE/DEAD staining. These data demonstrate that the ADSC secretome protects SH-SY5Y cells from arsenic-induced toxicity. Previous reports have shown that the secretome of MSCs can induce neuroblast differentiation and mature neurons are less susceptible to chemical-induced toxicity. In the current study, proliferation assays, neurite length assessment, and quantitative RT-PCR of differentiation markers indicated that the ADSC secretome does not induce SH-SY5Y differentiation into a mature neuron-like phenotype. In contrast, our results demonstrated that soluble factor(s) in the ADSC secretome enhance SH-SY5Y cell substrate-dependent adhesion. The present study is the first to illustrate that the secretome from ADSCs protects SH-SY5Y cells from arsenic-induced toxicity. Additionally, we showed that protection against arsenic toxicity is not dependent on SH-SY5Y cell differentiation into a mature neuron-like phenotype, but involves soluble factor(s) in the secretome that appear to enhance cell survival by an adhesion-dependent mechanism.
Collapse
Affiliation(s)
- Theresa M Curtis
- Department of Biological Sciences, State University of New York at Cortland, Cortland, NY, United States.
| | - Joseph M Hannett
- Department of Biological Sciences, State University of New York at Cortland, Cortland, NY, United States
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Nicholas A Puoplo
- Department of Biological Sciences, State University of New York at Cortland, Cortland, NY, United States
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
76
|
Lin R, Wang Y, Ji K, Liu Z, Xiao S, Zhou D, Chen Q, Shi B. Bioinformatics analysis to screen key genes implicated in the differentiation of induced pluripotent stem cells to hepatocytes. Mol Med Rep 2018; 17:4351-4359. [PMID: 29328449 PMCID: PMC5802208 DOI: 10.3892/mmr.2018.8385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 01/27/2017] [Indexed: 12/11/2022] Open
Abstract
Due to the lack of potential organs, hepatocellular transplantation has been considered for treating end-stage liver disease. Induced pluripotent stem cells (iPSCs) are reverted from somatic cells and are able to differentiate into hepatocytes. The present study aimed to investigate the mechanisms underlying iPSC differentiation to hepatocytes. GSE66076 was downloaded from the Gene Expression Omnibus; this database includes data from 3 undifferentiated (T0), 3 definitive endoderm (T5), and 3 early hepatocyte (T24) samples across hepatic‑directed differentiation of iPSCs. Differentially expressed genes (DEGs) between T0 and T5 or T24 samples were identified using the linear models for microarray data package in Bioconductor, and enrichment analyses were performed. Using the weighted correlation network analysis package in R, clusters were identified for the merged DEGs. Cytoscape was used to construct protein‑protein interaction (PPI) networks for DEGs identified to belong to significant clusters. Using the ReactomeFI plugin in Cytoscape, functional interaction (FI) networks were constructed for the common genes. A total of 433 and 1,342 DEGs were identified in the T5 and T24 samples respectively, compared with the T0 samples. Blue and turquoise clusters were identified as significant gene clusters. In the PPI network for DEGs in the blue cluster, the key node fibroblast growth factor 2 (FGF2) could interact with bone morphogenetic protein 2 (BMP2). Cyclin‑dependent kinase 1 (CDK1) was demonstrated to have the highest degree (degree=71) in the PPI network for DEGs in the turquoise cluster. Enrichment analysis for the common genes, including hepatocyte nuclear factor 4α (HNF4A) and epidermal growth factor (EGF), in the FI network indicated that EGF and FGF2 were enriched in the Ras and Rap1 signaling pathways. The present results suggest that FGF2, BMP2, CDK1, HNF4A and EGF may participate in the differentiation of iPSCs into hepatocytes.
Collapse
Affiliation(s)
- Rui Lin
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University Medical School, Shanghai 200065, P.R. China
| | - Yufeng Wang
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University Medical School, Shanghai 200065, P.R. China
| | - Kun Ji
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University Medical School, Shanghai 200065, P.R. China
| | - Zhongyan Liu
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University Medical School, Shanghai 200065, P.R. China
| | - Shuai Xiao
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University Medical School, Shanghai 200065, P.R. China
| | - Dehua Zhou
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University Medical School, Shanghai 200065, P.R. China
| | - Quanning Chen
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University Medical School, Shanghai 200065, P.R. China
| | - Baomin Shi
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University Medical School, Shanghai 200065, P.R. China
| |
Collapse
|
77
|
Abdal Dayem A, Lee S, Y. Choi H, Cho SG. The Impact of Adhesion Molecules on the In Vitro Culture and Differentiation of Stem Cells. Biotechnol J 2018; 13:1700575. [DOI: 10.1002/biot.201700575] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology; Incurable Disease Animal Model and Stem Cell Institute (IDASI); Konkuk University; 120 Neungdong-ro Gwangjin-gu 05029 Seoul Republic of Korea
| | - Soobin Lee
- Department of Stem Cell and Regenerative Biotechnology; Incurable Disease Animal Model and Stem Cell Institute (IDASI); Konkuk University; 120 Neungdong-ro Gwangjin-gu 05029 Seoul Republic of Korea
| | - Hye Y. Choi
- Department of Stem Cell and Regenerative Biotechnology; Incurable Disease Animal Model and Stem Cell Institute (IDASI); Konkuk University; 120 Neungdong-ro Gwangjin-gu 05029 Seoul Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology; Incurable Disease Animal Model and Stem Cell Institute (IDASI); Konkuk University; 120 Neungdong-ro Gwangjin-gu 05029 Seoul Republic of Korea
| |
Collapse
|
78
|
Nath SC, Tokura T, Kim M, Kino‐oka M. Botulinum hemagglutinin‐mediated in situ break‐up of human induced pluripotent stem cell aggregates for high‐density suspension culture. Biotechnol Bioeng 2018; 115:910-920. [DOI: 10.1002/bit.26526] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/04/2017] [Accepted: 12/22/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Suman C. Nath
- Department of BiotechnologyGraduate School of EngineeringOsaka UniversitySuitaOsakaJapan
| | - Tomohiro Tokura
- Department of BiotechnologyGraduate School of EngineeringOsaka UniversitySuitaOsakaJapan
- Fujimori Kogyo Co. Ltd.ShinjukuTokyoJapan
| | - Mee‐Hae Kim
- Department of BiotechnologyGraduate School of EngineeringOsaka UniversitySuitaOsakaJapan
| | - Masahiro Kino‐oka
- Department of BiotechnologyGraduate School of EngineeringOsaka UniversitySuitaOsakaJapan
| |
Collapse
|
79
|
Long-Term Maintenance of Human Pluripotent Stem Cells on cRGDfK-Presenting Synthetic Surfaces. Sci Rep 2018; 8:701. [PMID: 29335618 PMCID: PMC5768753 DOI: 10.1038/s41598-018-19209-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022] Open
Abstract
Synthetic human pluripotent stem cell (hPSC) culture surfaces with defined physical and chemical properties will facilitate improved research and therapeutic applications of hPSCs. In this study, synthetic surfaces for hPSC culture in E8 medium were produced for screening by modifying two polymer brush coatings [poly(acrylamide-co-acrylic acid) (PAAA) and poly(acrylamide-co-propargyl acrylamide) (PAPA)] to present single peptides. Adhesion of hPSC colonies was more consistently observed on surfaces modified with cRGDfK compared to surfaces modified with other peptide sequences tested. PAPA-coated polystyrene flasks with coupled cRGDfK (cRGDfK-PAPA) were then used for long-term studies of three hPSC lines (H9, hiPS-NHF1.3, Genea-02). Cell lines maintained for ten passages on cRGDfK-PAPA were assessed for colony morphology, proliferation rate, maintenance of OCT4 expression, cell viability at harvest, teratoma formation potential, and global gene expression as assessed by the PluriTest™ assay. cRGDfK-PAPA and control cultures maintained on Geltrex™ produced comparable results in most assays. No karyotypic abnormalities were detected in cultures maintained on cRGDfK-PAPA, while abnormalities were detected in cultures maintained on Geltrex™, StemAdhere™ or Synthemax™. This is the first report of long term maintenance of hPSC cultures on the scalable, stable, and cost-effective cRGDfK-PAPA coating.
Collapse
|
80
|
Green DW, Watson GS, Watson JA, Lee JM, Jung HS. Use of Tethered Hydrogel Microcoatings for Mesenchymal Stem Cell Equilibrium, Differentiation, and Self-Organization into Microtissues. ACTA ACUST UNITED AC 2017; 1:e1700116. [PMID: 32646160 DOI: 10.1002/adbi.201700116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/08/2017] [Indexed: 01/29/2023]
Abstract
Therapeutic adult mesenchymal stem cells (MSCs) lose multipotency and multilineage specialization in culture and after transplantation due to the absence of complex biological architecture. Here, it is shown that a transient ultrathin covering of permeable biomaterial can be differentially formulated to either preserve multipotency or induce multidifferentiation. Accordingly, populations of single, spherical MSCs in suspended media with high selectivity and specificity can be coated. Assembly of single, double, and triple hydrogel layers at MSC membranes is initiated by first attaching MSC-specific immunoglobulins onto CD90 or Stro-1 receptors and UEA-1 and soybean lectins. A secondary biotinylated immunoglobulin is targeted for avidin binding, which becomes an attractor for biotinylated alginate or hyaluronate, which are subsequently stiffened and gelled, in situ around the entire cell surface. Alginate microcoatings permeated with mobile BMP-2-induced osteospecialized tissue, vascular endothelial growth factor (VEGF) induced microcapillary formation, while microcoatings, with selected basement membrane proteins, preserve the multipotent phenotype of MSCs, for continuing rounds of culture and directed specialization. Furthermore, forced packing of microcoated MSC populations creates prototypical tissue compartments: the coating partially simulating the extracellular matrix structures. Remarkably, microcoated MSC clusters show a tremendous simulation of a common embryological tissue transformation into the epithelium. Thus, confinement of free morphology exerts another control on tissue specialization and formation.
Collapse
Affiliation(s)
- David W Green
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea.,Oral Biosciences, Faculty of Dentistry, The University of Hong Kong, Sai Ying Pun, Hong Kong, SAR
| | - Gregory S Watson
- School of Science and Engineering, University of the Sunshine Coast, Hervey Bay, QLD, 4655, Australia
| | - Jolanta A Watson
- School of Science and Engineering, University of the Sunshine Coast, Hervey Bay, QLD, 4655, Australia
| | - Jong-Min Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Han-Sung Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea.,Oral Biosciences, Faculty of Dentistry, The University of Hong Kong, Sai Ying Pun, Hong Kong, SAR
| |
Collapse
|
81
|
Maurizi G, Petäistö T, Maurizi A, Della Guardia L. Key-genes regulating the liposecretion process of mature adipocytes. J Cell Physiol 2017; 233:3784-3793. [PMID: 28926092 DOI: 10.1002/jcp.26188] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/14/2017] [Indexed: 12/13/2022]
Abstract
White mature adipocytes (MAs) are plastic cells able to reversibly transdifferentiate toward fibroblast-like cells maintaining stem cell gene signatures. The main morphologic aspect of this transdifferentiation process, called liposecretion, is the secretion of large lipid droplets and the development of organelles necessary for exocrine secretion. There is a considerable interest in the adipocyte plastic properties involving liposecretion process, but the molecular details are incompletely explored. This review analyzes the gene expression of MAs isolated from human subcutaneous fat tissue with respect to bone marrow (BM)-derived mesenchymal stem cells (MSC) focusing on gene regulatory pathways involved into cellular morphology changes, cellular proliferation and transports of molecules through the membrane, suggesting potential ways to guide liposecretion. In particular, Wnt, MAPK/ERK, and AKT pathways were accurately described, studying up- and down-stream molecules involved. Moreover, adipogenic extra- and intra-cellular interactions were analyzed studying the role of CDH2, CDH11, ITGA5, E-Syt1, PAI-1, IGF1, and INHBB genes. Additionally, PLIN1 and PLIN2 could be key-genes of liposecretion process regulating molecules transport through the membrane. All together data demonstrated that liposecretion is regulated through a complex molecular networks that are able to respond to microenvironment signals, cytokines, and growth factors. Autocrine as well as external signaling molecules might activate liposecretion affecting adipocytes physiology.
Collapse
Affiliation(s)
| | - Tiina Petäistö
- Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Angela Maurizi
- Chirurgia Generale, ASUR Regione Marche, Ospedale "Carlo Urbani", Jesi, Italy
| | - Lucio Della Guardia
- Dipartimento di Sanità Pubblica, Medicina Sperimentale e Forense, Unità di Scienza dell'Alimentazione, Università degli stui di Pavia, Pavia, Italy
| |
Collapse
|
82
|
Kim MH, Matsubara Y, Fujinaga Y, Kino-Oka M. A Simple and Robust Method for Culturing Human-Induced Pluripotent Stem Cells in an Undifferentiated State Using Botulinum Hemagglutinin. Biotechnol J 2017; 13. [PMID: 29027750 DOI: 10.1002/biot.201700384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/24/2017] [Indexed: 01/18/2023]
Abstract
Clinical and industrial applications of human-induced pluripotent stem cells (hiPSCs) is hindered by the lack of robust culture strategies capable of sustaining a culture in an undifferentiated state. Here, a simple and robust hiPSC-culture-propagation strategy incorporating botulinum hemagglutinin (HA)-mediated selective removal of cells deviating from an undifferentiated state is developed. After HA treatment, cell-cell adhesion is disrupted, and deviated cells detached from the central region of the colony to subsequently form tight monolayer colonies following prolonged incubation. The authors find that the temporal and dose-dependent activity of HA regulated deviated-cell removal and recoverability after disruption of cell-cell adhesion in hiPSC colonies. The effects of HA are confirmed under all culture conditions examined, regardless of hiPSC line and feeder-dependent or -free culture conditions. After routine application of our HA-treatment paradigm for serial passages, hiPSCs maintains expression of pluripotent markers and readily forms embryoid bodies expressing markers for all three germ-cell layers. This method enables highly efficient culturing of hiPSCs and use of entire undifferentiated portions without having to pick deviated cells manually. This simple and readily reproducible culture strategy is a potentially useful tool for improving the robust and scalable maintenance of undifferentiated hiPSC cultures.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshifumi Matsubara
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yukako Fujinaga
- Laboratory for Infection Cell Biology, International Research Centre for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Bacteriology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara, Kanazawa, Ishikawa, 920-8640, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
83
|
Nemati S, Rezabakhsh A, Khoshfetrat AB, Nourazarian A, Biray Avci Ç, Goker Bagca B, Alizadeh Sardroud H, Khaksar M, Ahmadi M, Delkhosh A, Sokullu E, Rahbarghazi R. Alginate-gelatin encapsulation of human endothelial cells promoted angiogenesis in in vivo and in vitro milieu. Biotechnol Bioeng 2017; 114:2920-2930. [DOI: 10.1002/bit.26395] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/31/2017] [Accepted: 08/01/2017] [Indexed: 01/04/2023]
Affiliation(s)
- Sorour Nemati
- Chemical Engineering Faculty; Sahand University of Technology; Tabriz Iran
- Stem Cell Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Aysa Rezabakhsh
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Tabriz University of Medical Sciences; Tabriz Iran
| | | | - Alireza Nourazarian
- Department of Biochemistry and Clinical Laboratories; Faculty of Medicine; Tabriz University of Medical Sciences; Tabriz Iran
| | - Çığır Biray Avci
- Department of Medical Biology; Faculty of Medicine; Ege University; Izmir Turkey
| | - Bakiye Goker Bagca
- Department of Medical Biology; Faculty of Medicine; Ege University; Izmir Turkey
| | | | - Majid Khaksar
- Stem Cell Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Mahdi Ahmadi
- Department of Physiology; Faculty of Medicine; Tabriz University of Medical Sciences; Tabriz Iran
| | - Aref Delkhosh
- Stem Cell Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Emel Sokullu
- Izmir Katip Celebi University; Bioengineering Department; Izmir Turkey
- Harvard Medical School; Division of Biomedical Engineering at Brigham and Women's Hospital, Harvard-MIT Health Sciences and Technology; Cambridge MA
| | - Reza Rahbarghazi
- Stem Cell Research Center; Tabriz University of Medical Sciences; Tabriz Iran
- Department of Applied Cell Sciences; Faculty of Advanced Medical Sciences; Tabriz University of Medical Sciences; Tabriz Iran
| |
Collapse
|
84
|
Sart S, Bejoy J, Li Y. Characterization of 3D pluripotent stem cell aggregates and the impact of their properties on bioprocessing. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.05.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
85
|
Närvä E, Stubb A, Guzmán C, Blomqvist M, Balboa D, Lerche M, Saari M, Otonkoski T, Ivaska J. A Strong Contractile Actin Fence and Large Adhesions Direct Human Pluripotent Colony Morphology and Adhesion. Stem Cell Reports 2017. [PMID: 28625538 PMCID: PMC5511101 DOI: 10.1016/j.stemcr.2017.05.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cell-type-specific functions and identity are tightly regulated by interactions between the cell cytoskeleton and the extracellular matrix (ECM). Human pluripotent stem cells (hPSCs) have ultimate differentiation capacity and exceptionally low-strength ECM contact, yet the organization and function of adhesion sites and associated actin cytoskeleton remain poorly defined. We imaged hPSCs at the cell-ECM interface with total internal reflection fluorescence microscopy and discovered that adhesions at the colony edge were exceptionally large and connected by thick ventral stress fibers. The actin fence encircling the colony was found to exert extensive Rho-ROCK-myosin-dependent mechanical stress to enforce colony morphology, compaction, and pluripotency and to define mitotic spindle orientation. Remarkably, differentiation altered adhesion organization and signaling characterized by a switch from ventral to dorsal stress fibers, reduced mechanical stress, and increased integrin activity and cell-ECM adhesion strength. Thus, pluripotency appears to be linked to unique colony organization and adhesion structure. Human pluripotent colonies have exceptional actin structure and focal adhesions Contraction-dependent tight colony compaction enforces pluripotency Colony morphology is maintained by edge-oriented cell divisions Differentiation alters actin orientation, integrin activity, and adhesion strength
Collapse
Affiliation(s)
- Elisa Närvä
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Aki Stubb
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Camilo Guzmán
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Matias Blomqvist
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Diego Balboa
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Martina Lerche
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Markku Saari
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Timo Otonkoski
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland; Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki 00290, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku 20520, Finland; Department of Biochemistry and Food Chemistry, University of Turku, Turku 20520, Finland.
| |
Collapse
|
86
|
Nath SC, Horie M, Nagamori E, Kino-Oka M. Size- and time-dependent growth properties of human induced pluripotent stem cells in the culture of single aggregate. J Biosci Bioeng 2017; 124:469-475. [PMID: 28601606 DOI: 10.1016/j.jbiosc.2017.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/05/2017] [Accepted: 05/11/2017] [Indexed: 12/21/2022]
Abstract
Aggregate culture of human induced pluripotent stem cells (hiPSCs) is a promising method to obtain high number of cells for cell therapy applications. This study quantitatively evaluated the effects of initial cell number and culture time on the growth of hiPSCs in the culture of single aggregate. Small size aggregates ((1.1 ± 0.4) × 101-(2.8 ± 0.5) × 101 cells/aggregate) showed a lower growth rate in comparison to medium size aggregates ((8.8 ± 0.8) × 101-(6.8 ± 1.1) × 102 cells/aggregate) during early-stage of culture (24-72 h). However, when small size aggregates were cultured in conditioned medium, their growth rate increased significantly. On the other hand, large size aggregates ((1.1 ± 0.2) × 103-(3.5 ± 1.1) × 103 cells/aggregate) showed a lower growth rate and lower expression level of proliferation marker (ki-67) in the center region of aggregate in comparison to medium size aggregate during early-stage of culture. Medium size aggregates showed the highest growth rate during early-stage of culture. Furthermore, hiPSCs proliferation was dependent on culture time because the growth rate decreased significantly during late-stage of culture (72-120 h) at which point collagen type I accumulated on the periphery of aggregate, suggesting blockage of diffusive transport of nutrients, oxygen and metabolites into and out of the aggregates. Consideration of initial cell number and culture time are important to maintain balance between autocrine factors secretion and extracellular matrix accumulation on the aggregate periphery to achieve optimal growth of hiPSCs in the culture of single aggregate.
Collapse
Affiliation(s)
- Suman C Nath
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Masanobu Horie
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Eiji Nagamori
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
87
|
Bu J, Kang YT, Lee YS, Kim J, Cho YH, Moon BI. Lab on a fabric: Mass producible and low-cost fabric filters for the high-throughput viable isolation of circulating tumor cells. Biosens Bioelectron 2017; 91:747-755. [DOI: 10.1016/j.bios.2017.01.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/02/2017] [Accepted: 01/19/2017] [Indexed: 01/01/2023]
|
88
|
Cadherins Associate with Distinct Stem Cell-Related Transcription Factors to Coordinate the Maintenance of Stemness in Triple-Negative Breast Cancer. Stem Cells Int 2017; 2017:5091541. [PMID: 28392805 PMCID: PMC5368378 DOI: 10.1155/2017/5091541] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/05/2017] [Accepted: 01/17/2017] [Indexed: 12/27/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer with poor prognosis and is enriched in cancer stem cells (CSCs). However, it is not completely understood how the CSCs were maintained in TNBC. In this study, by analyzing The Cancer Genome Atlas (TCGA) provisional datasets and several small-size breast datasets, we found that cadherins (CDHs) 2, 4, 6, and 17 were frequently amplified/overexpressed in 47% of TNBC while E-cadherin (CDH1) was downregulated/mutated at 10%. The alterations of CDH2/4/6/17 were strongly associated with the elevated levels of several stem cell-related transcription factors (SC-TFs) including FOXM1, MCM2, WWTR1, SNAI1, and SOX9. CDH2/4/6/17-enriched genes including FOXM1 and MCM2 were also clustered and regulated by NFY (nuclear transcription factor Y) and/or EVI1/MECOM. Meanwhile, these SC-TFs including NFYA were upregulated in TNBC cells, but they were downregulated in luminal type of cells. Furthermore, small compounds might be predicted via the Connectivity Map analysis to target TNBC with the alterations of CDH2/4/6/17 and SC-TFs. Together with the important role of these SC-TFs in the stem cell regulation, our data provide novel insights into the maintenance of CSCs in TNBC and the discovery of these SC-TFs associated with the alterations of CDH2/4/6/17 has an implication in targeted therapy of TNBC.
Collapse
|
89
|
Botulinum hemagglutinin-mediated selective removal of cells deviating from the undifferentiated state in hiPSC colonies. Sci Rep 2017; 7:93. [PMID: 28273902 PMCID: PMC5428320 DOI: 10.1038/s41598-017-00083-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/31/2017] [Indexed: 01/05/2023] Open
Abstract
The undifferentiated state of human induced pluripotent stem cells (hiPSCs) depends on their cell–cell and cell–substrate adhesions. In this study, we report that exposure to botulinum hemagglutinin (HA), an E-cadherin function-blocking agent, selectively removed cells that deviated from the undifferentiated state in hiPSC colonies. After HA treatment, cell–cell adhesion was disrupted, deviated cells detached from colony centers, and dividing cells filled these spaces. Because E-cadherin-mediated adhesion was disrupted in undifferentiated cells, stress-fiber formation and focal adhesions were diminished; however, these were subsequently restored, and the cells retained expression of undifferentiated stem cell markers and their differentiation potential. In contrast, actin structures and focal adhesions were lost from deviated cells, and they subsequently died. In undifferentiated and deviated cells, the cadherin/integrin-regulator Rap1 was localized at cell–cell adhesions and in the cytoplasm, respectively. Concurrent HA and Rap1-inhibitor treatment accelerated the deviated-cell detachment and delayed the recovery of hiPSC morphology, but this effect was significantly attenuated by co-treatment with Rap1 activator. Thus, Rap1 regulated E-cadherin–integrin interplay in hiPSC colonies exhibiting deviation, while HA-mediated selective removal of these deviated cells helped maintain the undifferentiated state in the remaining hiPSCs.
Collapse
|
90
|
Neganova I, Chichagova V, Armstrong L, Lako M. A critical role for p38MAPK signalling pathway during reprogramming of human fibroblasts to iPSCs. Sci Rep 2017; 7:41693. [PMID: 28155868 PMCID: PMC5290526 DOI: 10.1038/srep41693] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/09/2016] [Indexed: 01/17/2023] Open
Abstract
Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) holds enormous promise for regenerative medicine. Reprogramming is a stepwise process with well-defined stages of initiation, maturation and stabilisation which are critically dependent on interactions between key pluripotency transcription factors, epigenetic regulators and signalling pathways. In this manuscript we have investigated the role of p38 MAPK signalling pathway and have shown a subpopulation- and phase-specific pattern of activation occurring during the initiation and maturation stage of reprogramming in partially and fully reprogrammed cells respectively. Downregulation of p38 MAPK activity via RNA interference or small molecule inhibitor led to cell accumulation in G1 phase of the cell cycle and reduced expression of cell cycle regulators during the initiation stage of reprogramming. This was associated with a significant downregulation of key pluripotency marker expression, disruption of mesenchymal to epithelial transition (MET), increased expression of differentiation markers and presence of partially reprogrammed cells which retained a typical gene expression profile of mesendodermal cells and were unable to progress to fully reprogrammed phenotype. Together our data indicate an important role for p38 MAPK activity in proliferation, MET progression and establishment of pluripotent phenotype, which are necessary steps for the development of human iPSCs.
Collapse
Affiliation(s)
- Irina Neganova
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, NE1 3BZ, UK
| | - Valeria Chichagova
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, NE1 3BZ, UK
| | - Lyle Armstrong
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, NE1 3BZ, UK
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, NE1 3BZ, UK
| |
Collapse
|
91
|
Varun D, Srinivasan GR, Tsai YH, Kim HJ, Cutts J, Petty F, Merkley R, Stephanopoulos N, Dolezalova D, Marsala M, Brafman DA. A robust vitronectin-derived peptide for the scalable long-term expansion and neuronal differentiation of human pluripotent stem cell (hPSC)-derived neural progenitor cells (hNPCs). Acta Biomater 2017; 48:120-130. [PMID: 27989923 DOI: 10.1016/j.actbio.2016.10.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/03/2016] [Accepted: 10/26/2016] [Indexed: 12/22/2022]
Abstract
Despite therapeutic advances, neurodegenerative diseases and disorders remain some of the leading causes of mortality and morbidity in the United States. Therefore, cell-based therapies to replace lost or damaged neurons and supporting cells of the central nervous system (CNS) are of great therapeutic interest. To that end, human pluripotent stem cell (hPSC) derived neural progenitor cells (hNPCs) and their neuronal derivatives could provide the cellular 'raw material' needed for regenerative medicine therapies for a variety of CNS disorders. In addition, hNPCs derived from patient-specific hPSCs could be used to elucidate the underlying mechanisms of neurodegenerative diseases and identify potential drug candidates. However, the scientific and clinical application of hNPCs requires the development of robust, defined, and scalable substrates for their long-term expansion and neuronal differentiation. In this study, we rationally designed a vitronectin-derived peptide (VDP) that served as an adhesive growth substrate for the long-term expansion of several hNPC lines. Moreover, VDP-coated surfaces allowed for the directed neuronal differentiation of hNPC at levels similar to cells differentiated on traditional extracellular matrix protein-based substrates. Overall, the ability of VDP to support the long-term expansion and directed neuronal differentiation of hNPCs will significantly advance the future translational application of these cells in treating injuries, disorders, and diseases of the CNS.
Collapse
|
92
|
Leong MF, Lu HF, Lim TC, Du C, Ma NK, Wan AC. Electrospun polystyrene scaffolds as a synthetic substrate for xeno-free expansion and differentiation of human induced pluripotent stem cells. Acta Biomater 2016; 46:266-277. [PMID: 27667015 DOI: 10.1016/j.actbio.2016.09.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 09/16/2016] [Accepted: 09/22/2016] [Indexed: 01/01/2023]
Abstract
The use of human induced pluripotent stem cells (hiPSCs) for clinical tissue engineering applications requires expansion and differentiation of the cells using defined, xeno-free substrates. The screening and selection of suitable synthetic substrates however, is tedious, as their performance relies on the inherent material properties. In the present work, we demonstrate an alternative concept for xeno-free expansion and differentiation of hiPSCs using synthetic substrates, which hinges on the structure-function relationship between electrospun polystyrene scaffolds (ESPS) and pluripotent stem cell growth. ESPS of differential porosity was obtained by fusing the fibers at different temperatures. The more porous, loosely fused scaffolds were found to efficiently trap the cells, leading to a large number of three-dimensional (3D) aggregates which were shown to be pluripotent colonies. Immunostaining, PCR analyses, in vitro differentiation and in vivo teratoma formation studies demonstrated that these hiPSC aggregates could be cultured for up to 10 consecutive passages (P10) with maintenance of pluripotency. Flow cytometry showed that more than 80% of the cell population stained positive for the pluripotent marker OCT4 at P1, P5 and P10. P10 cells could be differentiated to neuronal-like cells and cultured within the ESPS for up to 18months. Our results suggest the usefulness of a generic class of synthetic substrates, exemplified by ESPS, for 'trapped aggregate culture' of hiPSCs. STATEMENT OF SIGNIFICANCE To realize the potential of human induced pluripotent stem cells (hiPSCs) in clinical medicine, robust, xeno-free substrates for expansion and differentiation of iPSCs are required. In the existing literature, synthetic materials have been reported that meet the requirement for non-xenogeneic substrates. However, the self-renewal and differentiation characteristics of hiPSCs are affected differently by the biocompatibility and physico-chemical properties of individual substrates. Although some rules based on chemical structure and substrate rigidity have been developed, most of these efforts are still empirical, and most synthetic substrates must still be rigorously screened for suitability. In this paper, we demonstrate an alternative concept for xeno-free expansion and differentiation of hiPSCs using synthetic substrates, which hinges on the structure-function relationship between electrospun polystyrene scaffolds (ESPS) and pluripotent stem cell growth. ESPS of differential porosity was obtained by fusing the fibers at different temperatures. The more porous, loosely fused scaffold was found to efficiently trap the cells, leading to a large number of three-dimensional (3D) aggregates. In the form of these trapped aggregates, we showed that hiPSCs could be cultured for up to 10 consecutive passages (P10) with maintenance of pluripotency, following which they could be differentiated to a chosen lineage. We believe that this novel, generic class of synthetic substrates that employs 'trapped aggregate culture' for expansion and differentiation of hiPSCs is an important conceptual advance, and would be of high interest to the readership of Acta Biomaterialia.
Collapse
|
93
|
Sulaiman A, Li L, Wang L. E-cadherin adhesion-mediated Wnt activation for mesoderm specification in human embryonic stem cells needs a soft mattress. Stem Cell Investig 2016; 3:77. [PMID: 28066779 DOI: 10.21037/sci.2016.10.12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 10/28/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Andrew Sulaiman
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Li Li
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada;; Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada;; Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada
| |
Collapse
|
94
|
Kim H, Kang DH, Koo KH, Lee S, Kim SM, Kim J, Yoon MH, Kim SY, Yang EG. Vertical nanocolumn-assisted pluripotent stem cell colony formation with minimal cell-penetration. NANOSCALE 2016; 8:18087-18097. [PMID: 27714141 DOI: 10.1039/c6nr06203b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The biological applications of vertical nanostructures mostly rely on their intracellular accessibility through the cellular membrane by promoting cell-to-nanostructure interactions. Herein, we report a seemingly counter-intuitive approach for the spontaneous formation of mouse induced pluripotent stem cell (iPSC)-derived three-dimensional spherical colonies with unlimited self-renewal and differentiation potential. The comprehensive analyses of iPSCs cultured on vertical silicon nanocolumn arrays (vSNAs) with various nanocolumn geometries show reduced cell-to-substrate adhesion and enhanced cell-to-cell interactions under optimized vSNA conditions, successfully accommodating the spontaneous production of iPSC-derived spherical colonies. Remarkably, these colonies which were only minimally penetrated by and thereby easily harvested from wafer-sized vSNAs display a substantial increase in pluripotency marker expression and successfully differentiate into three germ layers. Our vSNAs capable of large-scale fabrication, efficient for spherical colony formation, and reusable for multiple iPSC culture could serve as a broad-impact culture platform for stem cell research.
Collapse
Affiliation(s)
- Hyunju Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea.
| | - Dong Hee Kang
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, 261 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Republic of Korea.
| | - Kyung Hee Koo
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea.
| | - Seyeong Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, 261 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Republic of Korea.
| | - Seong-Min Kim
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, 261 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Republic of Korea.
| | - Janghwan Kim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, 34141, Republic of Korea and Department of Functional Genomics, Korea University of Science and Technology (UST), KRIBB campus, Daejeon, 34141, Republic of Korea
| | - Myung-Han Yoon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, 261 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Republic of Korea.
| | - So Yeon Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea. and Department of Biomedical Engineering, Korea University of Science and Technology (UST), KIST campus, Seoul 02792, Republic of Korea
| | - Eun Gyeong Yang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea. and Department of Biological Chemistry, Korea University of Science and Technology (UST), KIST campus, Seoul 02792, Republic of Korea
| |
Collapse
|
95
|
Hadjiantoniou SV, Sean D, Ignacio M, Godin M, Slater GW, Pelling AE. Physical confinement signals regulate the organization of stem cells in three dimensions. J R Soc Interface 2016; 13:20160613. [PMID: 27798278 PMCID: PMC5095220 DOI: 10.1098/rsif.2016.0613] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/05/2016] [Indexed: 01/25/2023] Open
Abstract
During embryogenesis, the spherical inner cell mass (ICM) proliferates in the confined environment of a blastocyst. Embryonic stem cells (ESCs) are derived from the ICM, and mimicking embryogenesis in vitro, mouse ESCs (mESCs) are often cultured in hanging droplets. This promotes the formation of a spheroid as the cells sediment and aggregate owing to increased physical confinement and cell-cell interactions. In contrast, mESCs form two-dimensional monolayers on flat substrates and it remains unclear if the difference in organization is owing to a lack of physical confinement or increased cell-substrate versus cell-cell interactions. Employing microfabricated substrates, we demonstrate that a single geometric degree of physical confinement on a surface can also initiate spherogenesis. Experiment and computation reveal that a balance between cell-cell and cell-substrate interactions finely controls the morphology and organization of mESC aggregates. Physical confinement is thus an important regulatory cue in the three-dimensional organization and morphogenesis of developing cells.
Collapse
Affiliation(s)
- Sebastian V Hadjiantoniou
- Department of Biology, University of Ottawa, Gendron Hall, 30 Marie Curie, Ottawa, Ontario, Canada K1N 6N5
| | - David Sean
- Department of Physics, University of Ottawa, MacDonald Hall, 150 Louis Pasteur, Ottawa, Ontario, Canada K1N 6N5
| | - Maxime Ignacio
- Department of Physics, University of Ottawa, MacDonald Hall, 150 Louis Pasteur, Ottawa, Ontario, Canada K1N 6N5
| | - Michel Godin
- Department of Physics, University of Ottawa, MacDonald Hall, 150 Louis Pasteur, Ottawa, Ontario, Canada K1N 6N5
- Department of Mechanical Engineering, University of Ottawa, Site Building, 800 King Edward Avenue, Ottawa, Ontario, Canada K1N 6N5
- Ottawa-Carleton Institute for Biomedical Engineering, Ottawa, Ontario, Canada K1N 6N5
| | - Gary W Slater
- Department of Physics, University of Ottawa, MacDonald Hall, 150 Louis Pasteur, Ottawa, Ontario, Canada K1N 6N5
| | - Andrew E Pelling
- Department of Biology, University of Ottawa, Gendron Hall, 30 Marie Curie, Ottawa, Ontario, Canada K1N 6N5
- Department of Physics, University of Ottawa, MacDonald Hall, 150 Louis Pasteur, Ottawa, Ontario, Canada K1N 6N5
- Institute for Science, University of Ottawa, Society and Policy, Desmarais Building, 55 Laurier Avenue East, Ottawa, Ontario, Canada K1N 6N5
| |
Collapse
|
96
|
Zhao XM, Cui LS, Hao HS, Wang HY, Zhao SJ, Du WH, Wang D, Liu Y, Zhu HB. Transcriptome analyses of inner cell mass and trophectoderm cells isolated by magnetic-activated cell sorting from bovine blastocysts using single cell RNA-seq. Reprod Domest Anim 2016; 51:726-35. [PMID: 27440443 DOI: 10.1111/rda.12737] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/21/2016] [Indexed: 12/31/2022]
Abstract
Research on bovine embryonic stem cells (bESCs) has been hampered because bESCs are cultured in conditions that are based on information obtained from culturing mouse and human inner cell mass (ICM) cells. The aim of this study was to compare gene expression in ICM and trophectoderm (TE) cell lineages of bovine embryos and to discuss the findings relative to information available for mice and humans. We separated a high-purity (>90%) ICM and TE from bovine blastocysts by magnetic-activated cell sorting and analysed their transcriptomes by single cell RNA-seq. Differentially expressed genes (DEGs) were assessed using Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) databases. Finally, qRT-PCR was performed to validate the RNA-seq results. From 207 DEGs identified (adjusted p ≤ .05; fold change ≥2), 159 and 48 had greater expression in the ICM and TE cells respectively. We validated 27 genes using qRT-PCR and found their expression patterns were mostly similar to those of RNA-seq, including 12 novel ICM-dominant (HNF4A, CCL24, FGFR4, IFITM3, PTCHD2, GJB5, FN1, KLK7, PRDM14, GRP, FGF19 and GCM1) and two novel TE-dominant (SLC10A1 and WNT4) genes. Bioinformatics analysis showed that these DEGs are involved in many important pathways, such as MAPK and cancer cell pathways, and these pathways have been shown to play essential roles in mouse and human ESCs in the self-renewal and pluripotent maintenance. As a conclusion, there were sufficient differences to allow us to conclude that the control of pluripotency in bovine ICM cells is species-specific.
Collapse
Affiliation(s)
- X-M Zhao
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - L-S Cui
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - H-S Hao
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - H-Y Wang
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - S-J Zhao
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - W-H Du
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - D Wang
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Y Liu
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - H-B Zhu
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China.
| |
Collapse
|
97
|
Zhang Y, Mao H, Qian M, Hu F, Cao L, Xu K, Shuai Q, Gao C, Lang R, Akaike T, Yang J. Surface modification with E-cadherin fusion protein for mesenchymal stem cell culture. J Mater Chem B 2016; 4:4267-4277. [PMID: 32263408 DOI: 10.1039/c6tb00765a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
To effectively expand human mesenchymal stem cells (hMSCs) in vitro without affecting their innate biological properties, a fusion protein (hE-cad-Fc) consisting of a human E-cadherin extracellular domain and an immunoglobulin G Fc region was fabricated and used as a biomimetic matrix for MSC culture surface modification. The results showed that cells cultured on hE-cad-Fc-modified polystyrene surfaces exhibited improved proliferation and paracrine functions compared with cells cultured on unmodified and collagen-modified polystyrene surfaces. Meanwhile, surfaces modified with hE-cad-Fc effectively inhibited cell apoptosis even under the serum deprivation conditions. Additionally, the hE-cad-Fc not only up-regulated the expression of β-catenin in MSCs and stimulated the cellular membrane complex of E-cadherin/β-catenin, but also effectively activated the intracellular signals such as EGFR, AKT and ERK phosphorylation. Therefore, hE-cad-Fc appeared to be a promising candidate for biological surface modification and stem cell culture.
Collapse
Affiliation(s)
- Yan Zhang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Row S, Liu Y, Alimperti S, Agarwal SK, Andreadis ST. Cadherin-11 is a novel regulator of extracellular matrix synthesis and tissue mechanics. J Cell Sci 2016; 129:2950-61. [PMID: 27311482 DOI: 10.1242/jcs.183772] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 06/10/2016] [Indexed: 01/20/2023] Open
Abstract
We discovered that Cadherin-11 (CDH11) regulates collagen and elastin synthesis, both affecting the mechanical properties and contractile function of animal tissues. Using a Cdh11-null mouse model, we observed a significant reduction in the mechanical properties [Youngs' modulus and ultimate tensile strength (UTS)] of Cdh11(-/-) as compared to wild-type (WT) mouse tissues, such as the aorta, bladder and skin. The deterioration of mechanical properties (Youngs' modulus and UTS) was accompanied by reduced collagen and elastin content in Cdh11(-/-) mouse tissues as well as in cells in culture. Similarly, knocking down CDH11 abolished collagen and elastin synthesis in human cells, and consequently reduced their ability to generate force. Conversely, engagement of CDH11 through homophilic interactions, led to swift activation of the TGF-β and ROCK pathways as evidenced by phosphorylation of downstream effectors. Subsequently, activation of the key transcription factors, MRTF-A (also known as MKL1) and MYOCD led to significant upregulation of collagen and elastin genes. Taken together, our results demonstrate a novel role of adherens junctions in regulating extracellular matrix (ECM) synthesis with implications for many important biological processes, including maintenance of tissue integrity, wound healing and tissue regeneration.
Collapse
Affiliation(s)
- Sindhu Row
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, NY 14260, USA
| | - Yayu Liu
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, NY 14260, USA
| | - Stella Alimperti
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, NY 14260, USA
| | - Sandeep K Agarwal
- Section of Allergy, Immunology, and Rheumatology Biology, Inflammation Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, NY 14260, USA Department of Biomedical Engineering, University at Buffalo, State University of New York, Amherst, NY 14260, USA Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA
| |
Collapse
|
99
|
Horiguchi I, Sakai Y. Serum replacement with albumin-associated lipids prevents excess aggregation and enhances growth of induced pluripotent stem cells in suspension culture. Biotechnol Prog 2016; 32:1009-16. [PMID: 27193385 DOI: 10.1002/btpr.2301] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 05/10/2016] [Indexed: 01/08/2023]
Abstract
Suspension culture systems are currently under investigation for the mass production of pluripotent stem (PS) cells for tissue engineering; however, the control of cell aggregation in suspension culture remains challenging. Existing methods to control aggregation such as microwell culture are difficult to scale up. To address this issue, in this study a novel method that incorporates the addition of KnockOut Serum Replacement (KSR) to the PS cell culture medium was described. The method regulated cellular aggregation and significantly improved cell growth (a 2- to 10-fold increase) without any influence on pluripotency. In addition, albumin-associated lipids as the major working ingredient of KSR responsible for this inhibition of aggregation were identified. This is one of the simplest methods described to date to control aggregation and requires only chemically synthesizable reagents. Thus, this method has the potential to simplify the mass production process of PS cells and thus lower their cost. © 2016 American Institute of Chemical Engineers Biotechnol. Prog., 32:1009-1016, 2016.
Collapse
Affiliation(s)
- Ikki Horiguchi
- Dept. of Chemical System Engineering, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Dept. of Chemical System Engineering, The University of Tokyo, Tokyo, Japan.,Center for International Research on Integrative Biomedical Systems, Inst. of Industrial Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
100
|
Pomeroy JE, Hough SR, Davidson KC, Quaas AM, Rees JA, Pera MF. Stem Cell Surface Marker Expression Defines Late Stages of Reprogramming to Pluripotency in Human Fibroblasts. Stem Cells Transl Med 2016; 5:870-82. [PMID: 27160704 DOI: 10.5966/sctm.2015-0250] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/23/2016] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Our current understanding of the induction of pluripotency by defined factors indicates that this process occurs in discrete stages characterized by specific alterations in the cellular transcriptome and epigenome. However, the final phase of the reprogramming process is incompletely understood. We sought to generate tools to characterize the transition to a fully reprogramed state. We used combinations of stem cell surface markers to isolate colonies emerging after transfection of human fibroblasts with reprogramming factors and then analyzed their expression of genes associated with pluripotency and early germ lineage specification. We found that expression of a subset of these genes, including the cell-cell adhesion molecule CDH3, characterized a late stage in the reprogramming process. Combined live-cell staining with the antibody GCTM-2 and anti-CDH3 during reprogramming identified colonies of cells that showed gene expression patterns very similar to those of embryonic stem cell or established induced pluripotent stem cell lines, and gave rise to stable induced pluripotent stem cell lines at high frequency. Our findings will facilitate studies of the final stages of reprogramming of human cells to pluripotency and will provide a simple means for prospective identification of fully reprogrammed cells. SIGNIFICANCE Reprogramming of differentiated cells back to an embryonic pluripotent state has wide ranging applications in understanding and treating human disease. However, how cells traverse the barriers on the journey to pluripotency still is not fully understood. This report describes tools to study the late stages of cellular reprogramming. The findings enable a more precise approach to dissecting the final phases of conversion to pluripotency, a process that is particularly poorly defined. The results of this study also provide a simple new method for the selection of fully reprogrammed cells, which could enhance the efficiency of derivation of cell lines for research and therapy.
Collapse
Affiliation(s)
- Jordan E Pomeroy
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Shelley R Hough
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, California, USA Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Kathryn C Davidson
- University of Melbourne and Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia Australian Institute of Regenerative Medicine, Monash University, Clayton, Victoria, Australia
| | - Alex M Quaas
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jordan A Rees
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Martin F Pera
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, California, USA Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia The Florey Neuroscience and Mental Health Institute, Parkville, Victoria, Australia
| |
Collapse
|