1
|
Tian P, Koudis NM, Morais MRPT, Pickard A, Fresquet M, Adamson A, Derby B, Lennon R. Collagen IV assembly is influenced by fluid flow in kidney cell-derived matrices. Cells Dev 2024; 179:203923. [PMID: 38670459 DOI: 10.1016/j.cdev.2024.203923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/30/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
Kidney podocytes and endothelial cells assemble a complex and dynamic basement membrane that is essential for kidney filtration. Whilst many components of this specialised matrix are known, the influence of fluid flow on its assembly and organisation remains poorly understood. Using the coculture of podocytes and glomerular endothelial cells in a low-shear stress, high-flow bioreactor, we investigated the effect of laminar fluid flow on the composition and assembly of cell-derived matrix. With immunofluorescence and matrix image analysis we found flow-mediated remodelling of collagen IV. Using proteomic analysis of the cell-derived matrix we identified changes in both abundance and composition of matrix proteins under flow, including the collagen-modifying enzyme, prolyl 4-hydroxylase (P4HA1). To track collagen IV assembly, we used CRISPR-Cas9 to knock in the luminescent marker HiBiT to the endogenous COL4A2 gene in podocytes. With this system, we found that collagen IV was secreted and accumulated consistently under both static and flow conditions. However knockdown of P4HA1 in podocytes led to a reduction in the secretion of collagen IV and this was more pronounced under flow. Together, this work demonstrates the effect of fluid flow on the composition, modification, and organisation of kidney cell-derived matrix and provides an in vitro system for investigating flow-induced matrix alteration in the context of kidney development and disease.
Collapse
Affiliation(s)
- Pinyuan Tian
- Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, UK.
| | - Nikki-Maria Koudis
- Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Mychel R P T Morais
- Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, UK.
| | - Adam Pickard
- Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Maryline Fresquet
- Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, UK.
| | - Antony Adamson
- Genome Editing Unit Core Facility, Faculty of Biology, Medicine and Health, University of Manchester, UK.
| | - Brian Derby
- School of Materials, University of Manchester, UK.
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, UK; Royal Manchester Children's Hospital, Manchester, UK.
| |
Collapse
|
2
|
Lewis PA, Silajdžić E, Smith H, Bates N, Smith CA, Mancini FE, Knight D, Denning C, Brison DR, Kimber SJ. A secreted proteomic footprint for stem cell pluripotency. PLoS One 2024; 19:e0299365. [PMID: 38875182 PMCID: PMC11178176 DOI: 10.1371/journal.pone.0299365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 02/08/2024] [Indexed: 06/16/2024] Open
Abstract
With a view to developing a much-needed non-invasive method for monitoring the healthy pluripotent state of human stem cells in culture, we undertook proteomic analysis of the waste medium from cultured embryonic (Man-13) and induced (Rebl.PAT) human pluripotent stem cells (hPSCs). Cells were grown in E8 medium to maintain pluripotency, and then transferred to FGF2 and TGFβ deficient E6 media for 48 hours to replicate an early, undirected dissolution of pluripotency. We identified a distinct proteomic footprint associated with early loss of pluripotency in both hPSC lines, and a strong correlation with changes in the transcriptome. We demonstrate that multiplexing of four E8- against four E6- enriched secretome biomarkers provides a robust, diagnostic metric for the pluripotent state. These biomarkers were further confirmed by Western blotting which demonstrated consistent correlation with the pluripotent state across cell lines, and in response to a recovery assay.
Collapse
Affiliation(s)
- Philip A. Lewis
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Edina Silajdžić
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Helen Smith
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Nicola Bates
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Christopher A. Smith
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Fabrizio E. Mancini
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - David Knight
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Chris Denning
- Biodiscovery Institute, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Daniel R. Brison
- Royal Manchester Children’s Hospital, Manchester, United Kingdom
| | - Susan J. Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
3
|
Ahn Y, Jeong J, Choi KH, Lee DK, Lee M, Lee NY, Kim DY, Lee CK. Development of Reproducible and Scalable Culture Conditions for In Vitro Maintenance of Pig Embryonic Stem Cells Using the Sandoz Inbred Swiss Mouse Thioguanine-Resistant Ouabain-Resistant Cell Line as a Feeder Layer. Stem Cells Dev 2023; 32:747-757. [PMID: 37756363 DOI: 10.1089/scd.2023.0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023] Open
Abstract
Feeder cells play a crucial role in maintaining the pluripotency of embryonic stem cells (ESCs) by secreting various extrinsic regulators, such as extracellular matrix (ECM) proteins and growth factors. Although primary mouse embryonic fibroblasts (MEFs) are the most widely used feeder cell type for the culture of ESCs, they have inevitable disadvantages such as batch-to-batch variation and labor-intensive isolation processes. Here, we revealed that the Sandoz inbred Swiss Mouse (SIM) thioguanine-resistant ouabain-resistant (STO) cell line, an immortalized cell line established from mouse SIM embryonic fibroblasts, can be used as a feeder layer for in vitro culture of authentic pig ESCs instead of primary MEFs. First, the expression of genes encoding ECM proteins and growth factors was analyzed to compare their secretory functions as feeder cells. Quantitative real-time polymerase chain reaction (qPCR) showed that the gene expression of these pluripotency-associated factors was downregulated in STO cells compared to primary MEFs of similar density. Therefore, subsequent optimization of the culture conditions was attempted using higher STO cell densities. Notably, pig ESCs cultured on STO cell density of 3 × (187,500 cells/cm2) exhibited the most similar pluripotent state to pig ESCs cultured on primary MEF density of 1 × (62,500 cells/cm2), as determined by alkaline phosphatase staining, qPCR, and immunocytochemistry. In addition, pig ESCs cultured on STO cell density of 3 × formed complex teratoma containing multiple types of tissues derived from all three germ layers. Our culture conditions using optimal STO cell density can be applied to fields requiring reproducible and scalable production of pig ESCs, such as preclinical research and cellular agriculture.
Collapse
Affiliation(s)
- Yelim Ahn
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
| | - Jinsol Jeong
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
| | - Kwang-Hwan Choi
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
- Research and Development Center, Space F Corporation, Hwasung, South Korea
| | - Dong-Kyung Lee
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
- Research and Development Center, Space F Corporation, Hwasung, South Korea
| | - Mingyun Lee
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
| | - Na-Young Lee
- Department of Veterinary Pathology, College of Veterinary Medicine; Seoul National University, Seoul, South Korea
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine; Seoul National University, Seoul, South Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, South Korea
| |
Collapse
|
4
|
Yang X, Lu Y, Kuang Q, Wu Y, Tan X, Lan J, Qiang Z, Feng T. Human embryonic stem cells exert antitumor effects on prostate cancer cells in a co-culture microenvironment. Front Oncol 2023; 13:1164250. [PMID: 37313467 PMCID: PMC10258316 DOI: 10.3389/fonc.2023.1164250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/17/2023] [Indexed: 06/15/2023] Open
Abstract
Prostate cancer is currently the most common malignancy among men. Given the limitations of current conventional anticancer therapies, new high-risk treatments are urgently needed. Previous studies have shown that embryonic stem cells (ESCs) can reverse the tumorigenic phenotype of tumor cells. However, there are still challenges in using human ESCs (hESCs) directly in cancer treatment. To facilitate the practical application of hESCs, we established a co-culture system consisting of prostate cancer cell lines and hESCs and investigated the antitumor activity of the supernatant of the co-culture system (Co-Sp) in vitro and in vivo, as well as the underlying mechanisms involved. The Co-Sp decreased the viability of prostate cancer cells in a concentration-dependent manner, significantly inhibited colony formation, and induced cell cycle arrest at the G0/G1 phase of the cell cycle. In addition, Co-Sp promoted apoptosis of prostate cancer cells and inhibited cell migration and invasion. In vivo studies also revealed that Co-Sp inhibited tumor growth in the xenograft model. Mechanistic studies showed that Co-Sp reduced the expression of cyclin D1, cyclin E, CDK4, CDK2, MMP-9, MMP-1, and Bcl-2, and increased the expression of p21, cleaved caspase-9, cleaved caspase-3, cleaved PARP, and Bax in prostate cancer cells. Furthermore, the Co-Sp decreased the phosphorylation of PI3K, AKT, and mTOR in cells and tumor tissues. Taken together, our results indicated that the Co-Sp has potent antitumor activity and could directly inhibit tumor growth. Our findings provide a new and effective way for the application of hESCs in cancer therapy and contribute to a new strategy for clinical stem cell therapy.
Collapse
Affiliation(s)
- Xinyue Yang
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yang Lu
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Qin Kuang
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yong Wu
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xin Tan
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jizhong Lan
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Zhe Qiang
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- Chongqing Academy of Chinese Materia Medica, Institute of Pharmacology Toxicology, Chongqing, China
| | - Tao Feng
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Cesare E, Urciuolo A, Stuart HT, Torchio E, Gesualdo A, Laterza C, Gagliano O, Martewicz S, Cui M, Manfredi A, Di Filippo L, Sabatelli P, Squarzoni S, Zorzan I, Betto RM, Martello G, Cacchiarelli D, Luni C, Elvassore N. 3D ECM-rich environment sustains the identity of naive human iPSCs. Cell Stem Cell 2022; 29:1703-1717.e7. [PMID: 36459970 DOI: 10.1016/j.stem.2022.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/07/2022] [Accepted: 11/09/2022] [Indexed: 12/03/2022]
Abstract
The establishment of in vitro naive human pluripotent stem cell cultures opened new perspectives for the study of early events in human development. The role of several transcription factors and signaling pathways have been characterized during maintenance of human naive pluripotency. However, little is known about the role exerted by the extracellular matrix (ECM) and its three-dimensional (3D) organization. Here, using an unbiased and integrated approach combining microfluidic cultures with transcriptional, proteomic, and secretome analyses, we found that naive, but not primed, hiPSC colonies are characterized by a self-organized ECM-rich microenvironment. Based on this, we developed a 3D culture system that supports robust long-term feeder-free self-renewal of naive hiPSCs and also allows direct and timely developmental morphogenesis simply by modulating the signaling environment. Our study opens new perspectives for future applications of naive hiPSCs to study critical stages of human development in 3D starting from a single cell.
Collapse
Affiliation(s)
- Elisa Cesare
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy; Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy
| | - Anna Urciuolo
- University College London Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; Institute of Pediatric Research IRP, Corso Stati Uniti, Padova 35127, Italy; Department of Molecular Medicine, University of Padova, Via G. Colombo 3, 35131 Padova, Italy
| | - Hannah T Stuart
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy; Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-BioCenter 1, 1030 Vienna, Austria
| | - Erika Torchio
- Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy
| | - Alessia Gesualdo
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy
| | - Cecilia Laterza
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy; Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy
| | - Onelia Gagliano
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy; Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy
| | - Sebastian Martewicz
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Meihua Cui
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Anna Manfredi
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy; Next Generation Diagnostic srl, Pozzuoli, Italy
| | - Lucio Di Filippo
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy; Next Generation Diagnostic srl, Pozzuoli, Italy
| | - Patrizia Sabatelli
- CNR - Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza" - Unit of Bologna, Bologna, Italy; IRCCS-Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Stefano Squarzoni
- CNR - Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza" - Unit of Bologna, Bologna, Italy; IRCCS-Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Irene Zorzan
- Epigenetics Programme, Babraham Institute, CB22 3AT Cambridge, UK
| | - Riccardo M Betto
- Department of Molecular Medicine, University of Padova, Via G. Colombo 3, 35131 Padova, Italy
| | - Graziano Martello
- Department of Biology, University of Padova, Via G. Colombo 3, Padova 35131, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy; Department of Translational Medicine, University of Naples "Federico II", Naples, Italy; School for Advanced Studies, Genomics and Experimental Medicine Program, University of Naples "Federico II", Naples, Italy
| | - Camilla Luni
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China; Department of Civil, Chemical, Environmental, and Materials Engineering (DICAM), University of Bologna, Via Terracini 28, Bologna 40131, Italy
| | - Nicola Elvassore
- Department of Industrial Engineering, University of Padova, 6/a Via Gradenigo, Padova 35131, Italy; Veneto Institute of Molecular Medicine, 2 Via Orus, Padova 35131, Italy; University College London Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.
| |
Collapse
|
6
|
Jurj A, Ionescu C, Berindan-Neagoe I, Braicu C. The extracellular matrix alteration, implication in modulation of drug resistance mechanism: friends or foes? J Exp Clin Cancer Res 2022; 41:276. [PMID: 36114508 PMCID: PMC9479349 DOI: 10.1186/s13046-022-02484-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/01/2022] [Indexed: 11/10/2022] Open
Abstract
The extracellular matrix (ECM) is an important component of the tumor microenvironment (TME), having several important roles related to the hallmarks of cancer. In cancer, multiple components of the ECM have been shown to be altered. Although most of these alterations are represented by the increased or decreased quantity of the ECM components, changes regarding the functional alteration of a particular ECM component or of the ECM as a whole have been described. These alterations can be induced by the cancer cells directly or by the TME cells, with cancer-associated fibroblasts being of particular interest in this regard. Because the ECM has this wide array of functions in the tumor, preclinical and clinical studies have assessed the possibility of targeting the ECM, with some of them showing encouraging results. In the present review, we will highlight the most relevant ECM components presenting a comprehensive description of their physical, cellular and molecular properties which can alter the therapy response of the tumor cells. Lastly, some evidences regarding important biological processes were discussed, offering a more detailed understanding of how to modulate altered signalling pathways and to counteract drug resistance mechanisms in tumor cells.
Collapse
Affiliation(s)
- Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania
| | - Calin Ionescu
- 7Th Surgical Department, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012, Cluj-Napoca, Romania
- Surgical Department, Municipal Hospital, 400139, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania.
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania.
- Research Center for Oncopathology and Translational Medicine (CCOMT), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, 540139, Targu Mures, Romania.
| |
Collapse
|
7
|
Hybrid Surface Nanostructures Using Chemical Vapor Deposition and Colloidal Self-Assembled Patterns for Human Mesenchymal Stem Cell Culture—A Preliminary Study. COATINGS 2022. [DOI: 10.3390/coatings12030311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Surface coatings are critical in biomaterials and biomedical devices. Chemical vapor deposition (CVD) is a well-known technology for the generation of thin films on a surface. However, the granular structures produced using CVD are rare. Recently, we used PPX-C, an excellent insulating material, for granular structure coating using CVD. Colloidal self-assembly is also a well-established method to generate granular structures named colloidal self-assembled patterns (cSAPs). In this study, we combined these two technologies to generate hierarchical granular structures and tested the biophysical effect of these hybrid surfaces on human bone marrow mesenchymal stem cells (hBMSCs). Two CVD-derived granular structures were made using water or glycerin droplets (i.e., CVD or GlyCVD surfaces). Water drops generate porous particles, while glycerin drops generate core–shell particles on the surface. These particles were dispersed randomly on the surface with sizes ranging from 1 to 20 μm. These CVD surfaces were hydrophobic (WCA ~ 80–110 degrees). On the other hand, a binary colloidal crystal (BCC), one type of cSAPs, composed of 5 μm Si and 400 nm carboxylated polystyrene (PSC) particles, had a close-packed structure and a hydrophilic surface (WCA ~ 45 degrees). The hybrid surfaces (i.e., CVD-BCC and GlyCVD-BCC) were smooth (Ra ~ 1.1–1.5 μm) and hydrophilic (WCA ~ 50 degrees), indicating a large surface coverage of BCC dominating the surface property. The hybrid surfaces were expected to be slightly negatively charged due to naturally charged CVD particles and negatively charged BCC particles. Cell adhesion was reduced on the hybrid surfaces, leading to an aggregated cell morphology, without reducing cell activity, compared to the flat control after 5 days. qPCR analysis showed that gene expression of type II collagen (COL2) was highly expressed on the GlyCVD-BCC without chemical induction after 3 and 14 days compared to the flat control. This proof-of-concept study demonstrates the potential of combining two technologies to make hybrid structures that can modulate stem cell attachment and differentiation.
Collapse
|
8
|
Pavarajarn W, Rungsiwiwut R, Numchaisrika P, Virutamasen P, Pruksananonda K. Human Caesarean scar-derived feeder cells: a novel feeder cell type for culturing human pluripotent stem cells without exogenous basic fibroblast growth factor supplementation. Reprod Fertil Dev 2021; 32:822-834. [PMID: 32527373 DOI: 10.1071/rd19128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 12/10/2019] [Indexed: 11/23/2022] Open
Abstract
In a feeder-dependent culture system of human pluripotent stem cells (hPSCs), coculture with mouse embryonic fibroblasts may limit the clinical use of hPSCs. The aim of this study was to determine the feasibility of using human Caesarean scar fibroblasts (HSFs) as feeder cells for the culture of hPSCs. HSFs were isolated and characterised and cocultured with hPSCs, and the pluripotency, differentiation ability and karyotypic stability of hPSCs were determined. Inactivated HSFs expressed genes (including inhibin subunit beta A (INHBA), bone morphogenetic protein 4 (BMP4), fibroblast growth factor 2 (FGF2), transforming growth factor-β1 (TGFB1), collagen alpha-1(I) (COL1A1) and fibronectin-1 (FN1) that have been implicated in the maintenance of hPSC pluripotency. When HSFs were used as feeder cells, the pluripotency and karyotypic stability of hPSC lines did not change after prolonged coculture. Interestingly, exogenous FGF2 could be omitted from the culture medium when HSFs were used as feeder cells for hESCs but not hiPSCs. hESCs cocultured with HSF feeder cells in medium without FGF2 supplementation maintained their pluripotency (as confirmed by the expression of pluripotency markers and genes), differentiated invitro into embryonic germ layers and maintained their normal karyotype. The present study demonstrates that HSFs are a novel feeder cell type for culturing hPSCs and that supplementation of exogenous FGF2 is not necessary for the Chula2.hES line.
Collapse
Affiliation(s)
- Wipawee Pavarajarn
- Graduate School, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4, Bangkok 10330, Thailand; and Human Embryonic Stem Cell Research Center, Reproductive Medicine Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4, Bangkok 10330, Thailand
| | - Ruttachuk Rungsiwiwut
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, 114 Sukhumvit 23, Bangkok 10110, Thailand
| | - Pranee Numchaisrika
- Human Embryonic Stem Cell Research Center, Reproductive Medicine Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4, Bangkok 10330, Thailand
| | - Pramuan Virutamasen
- Human Embryonic Stem Cell Research Center, Reproductive Medicine Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4, Bangkok 10330, Thailand
| | - Kamthorn Pruksananonda
- Human Embryonic Stem Cell Research Center, Reproductive Medicine Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4, Bangkok 10330, Thailand; and Corresponding author.
| |
Collapse
|
9
|
Sart S, Jeske R, Chen X, Ma T, Li Y. Engineering Stem Cell-Derived Extracellular Matrices: Decellularization, Characterization, and Biological Function. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:402-422. [DOI: 10.1089/ten.teb.2019.0349] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Sébastien Sart
- Hydrodynamics Laboratory, CNRS UMR7646, Ecole Polytechnique, Palaiseau, France
- Laboratory of Physical Microfluidics and Bioengineering, Department of Genome and Genetics, Institut Pasteur, Paris, France
| | - Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Xingchi Chen
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
10
|
Ke ZP, Xu YJ, Wang ZS, Sun J. RNA sequencing profiling reveals key mRNAs and long noncoding RNAs in atrial fibrillation. J Cell Biochem 2020; 121:3752-3763. [PMID: 31680326 DOI: 10.1002/jcb.29504] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/08/2019] [Indexed: 01/24/2023]
Abstract
Long noncoding RNAs (lncRNAs) are an emerging class of RNA species that could participate in some critical pathways and disease pathogenesis. However, the underlying molecular mechanism of lncRNAs in atrial fibrillation (AF) is still not fully understood. In the present study, we analyzed RNA-seq data of paired left and right atrial appendages from five patients with AF and other five patients without AF. Based on the gene expression profiles of 20 samples, we found that a majority of genes were aberrantly expressed in both left and right atrial appendages of patients with AF. Similarly, the dysregulated pathways in the left and right atrial appendages of patients with AF also bore a close resemblance. Moreover, we predicted regulatory lncRNAs that regulated the expression of adjacent protein-coding genes (PCGs) or interacted with proteins. We identified that NPPA and its antisense RNA NPPA-AS1 may participate in the pathogenesis of AF by regulating the muscle contraction. We also identified that RP11 - 99E15.2 and RP3 - 523K23.2 could interact with proteins ITGB3 and HSF2, respectively. RP11 - 99E15.2 and RP3 - 523K23.2 may participate in the pathogenesis of AF via regulating the extracellular matrix binding and the transcription of HSF2 target genes, respectively. The close association of the lncRNA-interacting proteins with AF further demonstrated that these two lncRNAs were also associated with AF. In conclusion, we have identified key regulatory lncRNAs implicated in AF, which not only improves our understanding of the lncRNA-related molecular mechanism underlying AF but also provides computationally predicted regulatory lncRNAs for AF researchers.
Collapse
Affiliation(s)
- Zun-Ping Ke
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Ying-Jia Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Zhang-Sheng Wang
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jian Sun
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Clinical Research Unit, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Tang SW, Tong WY, Pang SW, Voelcker NH, Lam YW. Deconstructing, Replicating, and Engineering Tissue Microenvironment for Stem Cell Differentiation. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:540-554. [PMID: 32242476 DOI: 10.1089/ten.teb.2020.0044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One of the most crucial components of regenerative medicine is the controlled differentiation of embryonic or adult stem cells into the desired cell lineage. Although most of the reported protocols of stem cell differentiation involve the use of soluble growth factors, it is increasingly evident that stem cells also undergo differentiation when cultured in the appropriate microenvironment. When cultured in decellularized tissues, for instance, stem cells can recapitulate the morphogenesis and functional specialization of differentiated cell types with speed and efficiency that often surpass the traditional growth factor-driven protocols. This suggests that the tissue microenvironment (TME) provides stem cells with a holistic "instructive niche" that harbors signals for cellular reprogramming. The translation of this into medical applications requires the decoding of these signals, but this has been hampered by the complexity of TME. This problem is often addressed by a reductionist approach, in which cells are exposed to substrates decorated with simple, empirically designed geometries, textures, and chemical compositions ("bottom-up" approach). Although these studies are invaluable in revealing the basic principles of mechanotransduction mechanisms, their physiological relevance is often uncertain. This review examines the recent progress of an alternative, "top-down" approach, in which the TME is treated as a holistic biological entity. This approach is made possible by recent advances in systems biology and fabrication technologies that enable the isolation, characterization, and reconstitution of TME. It is hoped that these new techniques will elucidate the nature of niche signals so that they can be extracted, replicated, and controlled. This review summarizes these emerging techniques and how the data they generated are changing our view on TME. Impact statement This review summarizes the current state of art of the understanding of instructive niche in the field of tissue microenvironment. Not only did we survey the use of different biochemical preparations as stimuli of stem cell differentiation and summarize the recent effort in dissecting the biochemical composition of these preparations, through the application of extracellular matrix (ECM) arrays and proteomics, but we also introduce the use of open-source, high-content immunohistochemistry projects in contributing to the understanding of tissue-specific composition of ECM. We believe this review would be highly useful for our peer researching in the same field. "Mr. Tulkinghorn is always the same… so oddly out of place and yet so perfectly at home." -Charles Dickens, Bleak House.
Collapse
Affiliation(s)
- Sze Wing Tang
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| | - Wing Yin Tong
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Stella W Pang
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong
| | - Nicolas H Voelcker
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Yun Wah Lam
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
12
|
Yemanyi F, Vranka J, Raghunathan V. Generating cell-derived matrices from human trabecular meshwork cell cultures for mechanistic studies. Methods Cell Biol 2020; 156:271-307. [PMID: 32222223 DOI: 10.1016/bs.mcb.2019.10.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ocular hypertension has been attributed to increased resistance to aqueous outflow often as a result of changes in trabecular meshwork (TM) extracellular matrix (ECM) using in vivo animal models (for example, by genetic manipulation) and ex vivo anterior segment perfusion organ cultures. These are, however, complex and difficult in dissecting molecular mechanisms and interactions. In vitro approaches to mimic the underlying substrate exist by manipulating either ECM topography, mechanics, or chemistry. These models best investigate the role of individual ECM protein(s) and/or substrate property, and thus do not recapitulate the multifactorial extracellular microenvironment; hence, mitigating its physiological relevance for mechanistic studies. Cell-derived matrices (CDMs), however, are capable of presenting a 3D-microenvironment rich in topography, chemistry, and whose mechanics can be tuned to better represent the network of native ECM constituents in vivo. Critically, the composition of CDMs may also be fine-tuned by addition of small molecules or relevant bioactive factors to mimic homeostasis or pathology. Here, we first provide a streamlined protocol for generating CDMs from TM cell cultures from normal or glaucomatous donor tissues. Second, we document how TM cells can be pharmacologically manipulated to obtain glucocorticoid-induced CDMs and how generated pristine CDMs can be manipulated with reagents like genipin. Finally, we summarize how CDMs may be used in mechanistic studies and discuss their probable application in future TM regenerative studies.
Collapse
Affiliation(s)
- Felix Yemanyi
- Department of Basic Sciences, University of Houston, Houston, TX, United States
| | - Janice Vranka
- Casey Eye Institute, Oregon Health and Science University, Portland, OR, United States
| | - VijayKrishna Raghunathan
- Department of Basic Sciences, University of Houston, Houston, TX, United States; The Ocular Surface Institute, College of Optometry, University of Houston, Houston, TX, United States; Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX, United States.
| |
Collapse
|
13
|
Nallanthighal S, Heiserman JP, Cheon DJ. The Role of the Extracellular Matrix in Cancer Stemness. Front Cell Dev Biol 2019; 7:86. [PMID: 31334229 PMCID: PMC6624409 DOI: 10.3389/fcell.2019.00086] [Citation(s) in RCA: 239] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 05/03/2019] [Indexed: 12/12/2022] Open
Abstract
As our understanding of cancer cell biology progresses, it has become clear that tumors are a heterogenous mixture of different cell populations, some of which contain so called "cancer stem cells" (CSCs). Hallmarks of CSCs include self-renewing capability, tumor-initiating capacity and chemoresistance. The extracellular matrix (ECM), a major structural component of the tumor microenvironment, is a highly dynamic structure and increasing evidence suggests that ECM proteins establish a physical and biochemical niche for CSCs. In cancer, abnormal ECM dynamics occur due to disrupted balance between ECM synthesis and secretion and altered expression of matrix-remodeling enzymes. Tumor-derived ECM is biochemically distinct in its composition and is stiffer compared to normal ECM. In this review, we will provide a brief overview of how different components of the ECM modulate CSC properties then discuss how physical, mechanical, and biochemical cues from the ECM drive cancer stemness. Given the fact that current CSC targeting therapies face many challenges, a better understanding of CSC-ECM interactions will be crucial to identify more effective therapeutic strategies to eliminate CSCs.
Collapse
Affiliation(s)
| | | | - Dong-Joo Cheon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| |
Collapse
|
14
|
Sharma S, Bollinger KE, Kodeboyina SK, Zhi W, Patton J, Bai S, Edwards B, Ulrich L, Bogorad D, Sharma A. Proteomic Alterations in Aqueous Humor From Patients With Primary Open Angle Glaucoma. Invest Ophthalmol Vis Sci 2019; 59:2635-2643. [PMID: 29847670 PMCID: PMC6733532 DOI: 10.1167/iovs.17-23434] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Purpose Primary open angle glaucoma (POAG) is the most prevalent form of glaucoma, accounting for approximately 90% of all cases. The aqueous humor (AH), a biological fluid in the anterior and posterior chambers of the eye, is involved in a multitude of functions including the maintenance of IOP and ocular homeostasis. This fluid is very close to the pathologic site and is also known to have a significant role in glaucoma pathogenesis. The purpose of this study was to identify proteomic alterations in AH from patients with POAG. Methods AH samples were extracted from 47 patients undergoing cataract surgery (controls: n = 32; POAG: n = 15). Proteomic analysis of the digested samples was accomplished by liquid-chromatography-mass spectrometry. The identified proteins were evaluated using a variety of statistical and bioinformatics methods. Results A total of 33 proteins were significantly altered in POAG subjects compared with the controls. The most abundant proteins in POAG subjects are IGKC (13.56-fold), ITIH4 (4.1-fold), APOC3 (3.36-fold), IDH3A (3.11-fold), LOC105369216 (2.98-fold). SERPINF2 (2.94-fold), NPC2 (2.88-fold), SUCLG2 (2.70-fold), KIAA0100 (2.29-fold), CNOT4 (2.23-fold), AQP4 (2.11-fold), COL18A1 (2.08-fold), NWD1 (2.07-fold), and TMEM120B (2.06-fold). A significant increasing trend in the odds ratios of having POAG was observed with increased levels of these proteins. Conclusion Proteins identified in this study are implicated in signaling, glycosylation, immune response, molecular transport, and lipid metabolism. The identified candidate proteins may be potential biomarkers associated with POAG development and may lead to more insight in understanding the mechanisms underlying the pathogenesis of this disease.
Collapse
Affiliation(s)
- Shruti Sharma
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States.,Department of Ophthalmology, Augusta University, Augusta, Georgia, United States.,James & Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Kathryn E Bollinger
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States.,James & Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Sai Karthik Kodeboyina
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Wenbo Zhi
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Jordan Patton
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| | - Shan Bai
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Blake Edwards
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| | - Lane Ulrich
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| | - David Bogorad
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| | - Ashok Sharma
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States.,Department of Population Health Sciences, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
15
|
Santoro R, Perrucci GL, Gowran A, Pompilio G. Unchain My Heart: Integrins at the Basis of iPSC Cardiomyocyte Differentiation. Stem Cells Int 2019; 2019:8203950. [PMID: 30906328 PMCID: PMC6393933 DOI: 10.1155/2019/8203950] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
The cellular response to the extracellular matrix (ECM) microenvironment mediated by integrin adhesion is of fundamental importance, in both developmental and pathological processes. In particular, mechanotransduction is of growing importance in groundbreaking cellular models such as induced pluripotent stem cells (iPSC), since this process may strongly influence cell fate and, thus, augment the precision of differentiation into specific cell types, e.g., cardiomyocytes. The decryption of the cellular machinery starting from ECM sensing to iPSC differentiation calls for new in vitro methods. Conveniently, engineered biomaterials activating controlled integrin-mediated responses through chemical, physical, and geometrical designs are key to resolving this issue and could foster clinical translation of optimized iPSC-based technology. This review introduces the main integrin-dependent mechanisms and signalling pathways involved in mechanotransduction. Special consideration is given to the integrin-iPSC linkage signalling chain in the cardiovascular field, focusing on biomaterial-based in vitro models to evaluate the relevance of this process in iPSC differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Rosaria Santoro
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Aoife Gowran
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
| | - Giulio Pompilio
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino IRCCS, via Carlo Parea 4, Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, via Festa del Perdono 7, Milan, Italy
| |
Collapse
|
16
|
Lee KJ, Comerford EJ, Simpson DM, Clegg PD, Canty-Laird EG. Identification and Characterization of Canine Ligament Progenitor Cells and Their Extracellular Matrix Niche. J Proteome Res 2019; 18:1328-1339. [DOI: 10.1021/acs.jproteome.8b00933] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Katie J Lee
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, United Kingdom
| | - Eithne J Comerford
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, United Kingdom
- School of Veterinary Science, Leahurst Campus, University of Liverpool, Chester High Road, Neston, CH64 7TE, United Kingdom
| | - Deborah M Simpson
- Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, United Kingdom
| | - Peter D Clegg
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, United Kingdom
- School of Veterinary Science, Leahurst Campus, University of Liverpool, Chester High Road, Neston, CH64 7TE, United Kingdom
- The MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Liverpool L7 8TX, United Kingdom
| | - Elizabeth G Canty-Laird
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, United Kingdom
- The MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Liverpool L7 8TX, United Kingdom
| |
Collapse
|
17
|
Cartilage oligomeric matrix protein is a novel notch ligand driving embryonic stem cell differentiation towards the smooth muscle lineage. J Mol Cell Cardiol 2018; 121:69-80. [PMID: 29981303 DOI: 10.1016/j.yjmcc.2018.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/06/2018] [Accepted: 07/03/2018] [Indexed: 12/18/2022]
Abstract
Cartilage oligomeric matrix protein (COMP), a protective component of vascular extracellular matrix (ECM), maintains the homeostasis of mature vascular smooth muscle cells (VSMCs). However, whether COMP modulates the differentiation of stem cells towards the smooth muscle lineage is still elusive. Firstly, purified mouse COMP directly induced mouse embryonic stem cell (ESC) differentiation into VSMCs both in vitro and in vivo, while the silencing of endogenous COMP markedly inhibited ESC-VSMC differentiation. RNA-Sequencing revealed that Notch signaling was significantly activated by COMP during ESC-VSMC differentiation, whereas the inhibition of Notch signaling attenuated COMP-directed ESC-VSMC differentiation. Furthermore, COMP deficiency inhibited Notch activation and VSMC differentiation in mice. Through silencing distinct Notch receptors, we identified that Notch1 mainly mediated COMP-initiated ESC-VSMC differentiation. Mechanistically, COMP N-terminus directly interacted with the EGF11-12 domain of Notch1 and activated Notch1 signaling, as evidenced by co-immunoprecipitation and mammalian two-hybrid assay. In conclusion, COMP served as a potential ligand of Notch1, thereby driving ESC-VSMC differentiation.
Collapse
|
18
|
Pedroni AC, Diniz IM, Abe GL, Moreira MS, Sipert CR, Marques MM. Photobiomodulation therapy and vitamin C on longevity of cell sheets of human dental pulp stem cells. J Cell Physiol 2018; 233:7026-7035. [DOI: 10.1002/jcp.26626] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 03/28/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Ana C.F. Pedroni
- Department of Restorative Dentistry, School of Dentistry University of Sao Paulo Sao Paulo Brazil
| | - Ivana M.A. Diniz
- Department of Restorative Dentistry, School of Dentistry University of Sao Paulo Sao Paulo Brazil
- Department of Restorative Dentistry, School of Dentistry Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Gabriela L. Abe
- Department of Restorative Dentistry, School of Dentistry University of Sao Paulo Sao Paulo Brazil
| | - Maria S. Moreira
- Post Graduation Program of the School of Dentistry Ibirapuera University Sao Paulo Brazil
| | - Carla R. Sipert
- Department of Restorative Dentistry, School of Dentistry University of Sao Paulo Sao Paulo Brazil
| | - Márcia M. Marques
- Department of Restorative Dentistry, School of Dentistry University of Sao Paulo Sao Paulo Brazil
| |
Collapse
|
19
|
Othman Z, Cillero Pastor B, van Rijt S, Habibovic P. Understanding interactions between biomaterials and biological systems using proteomics. Biomaterials 2018; 167:191-204. [PMID: 29571054 DOI: 10.1016/j.biomaterials.2018.03.020] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 03/02/2018] [Accepted: 03/12/2018] [Indexed: 12/22/2022]
Abstract
The role that biomaterials play in the clinical treatment of damaged organs and tissues is changing. While biomaterials used in permanent medical devices were required to passively take over the function of a damaged tissue in the long term, current biomaterials are expected to trigger and harness the self-regenerative potential of the body in situ and then to degrade, the foundation of regenerative medicine. To meet these different requirements, it is imperative to fully understand the interactions biomaterials have with biological systems, in space and in time. This knowledge will lead to a better understanding of the regenerative capabilities of biomaterials aiding their design with improved functionalities (e.g. biocompatibility, bioactivity). Proteins play a pivotal role in the interaction between biomaterials and cells or tissues. Protein adsorption on the material surface is the very first event of this interaction, which is determinant for the subsequent processes of cell growth, differentiation, and extracellular matrix formation. Against this background, the aim of the current review is to provide insight in the current knowledge of the role of proteins in cell-biomaterial and tissue-biomaterial interactions. In particular, the focus is on proteomics studies, mainly using mass spectrometry, and the knowledge they have generated on protein adsorption of biomaterials, protein production by cells cultured on materials, safety and efficacy of new materials based on nanoparticles and the analysis of extracellular matrices and extracellular matrix-derived products. In the outlook, the potential and limitations of this approach are discussed and mass spectrometry imaging is presented as a powerful technique that complements existing mass spectrometry techniques by providing spatial molecular information about the material-biological system interactions.
Collapse
Affiliation(s)
- Ziryan Othman
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Berta Cillero Pastor
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Sabine van Rijt
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Pamela Habibovic
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
20
|
Soteriou D, Fuchs Y. A matter of life and death: stem cell survival in tissue regeneration and tumour formation. Nat Rev Cancer 2018; 18:187-201. [PMID: 29348578 DOI: 10.1038/nrc.2017.122] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In recent years, great strides have been made in our understanding of how stem cells (SCs) govern tissue homeostasis and regeneration. The inherent longevity of SCs raises the possibility that the unique protective mechanisms in these cells might also be involved in tumorigenesis. In this Opinion article, we discuss how SCs are protected throughout their lifespan, focusing on quiescent behaviour, DNA damage response and programmed cell death. We briefly examine the roles of adult SCs and progenitors in tissue repair and tumorigenesis and explore how signals released from dying or dormant cells influence the function of healthy or aberrant SCs. Important insight into the mechanisms that regulate SC death and survival, as well as the 'legacy' imparted by departing cells, may unlock novel avenues for regenerative medicine and cancer therapy.
Collapse
Affiliation(s)
- Despina Soteriou
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology; the Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology; and the Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology; the Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology; and the Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200, Israel
| |
Collapse
|
21
|
Mouse embryonic fibroblast (MEF)/BMP4-conditioned medium enhanced multipotency of human dental pulp cells. J Mol Histol 2017; 49:17-26. [DOI: 10.1007/s10735-017-9743-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/23/2017] [Indexed: 12/13/2022]
|
22
|
Vitillo L, Baxter M, Iskender B, Whiting P, Kimber SJ. Integrin-Associated Focal Adhesion Kinase Protects Human Embryonic Stem Cells from Apoptosis, Detachment, and Differentiation. Stem Cell Reports 2017; 7:167-76. [PMID: 27509133 PMCID: PMC4983079 DOI: 10.1016/j.stemcr.2016.07.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 07/08/2016] [Accepted: 07/08/2016] [Indexed: 11/25/2022] Open
Abstract
Human embryonic stem cells (hESCs) can be maintained in a fully defined niche on extracellular matrix substrates, to which they attach through integrin receptors. However, the underlying integrin signaling mechanisms, and their contribution to hESC behavior, are largely unknown. Here, we show that focal adhesion kinase (FAK) transduces integrin activation and supports hESC survival, substrate adhesion, and maintenance of the undifferentiated state. After inhibiting FAK kinase activity we show that hESCs undergo cell detachment-dependent apoptosis or differentiation. We also report deactivation of FAK downstream targets, AKT and MDM2, and upregulation of p53, all key players in hESC regulatory networks. Loss of integrin activity or FAK also induces cell aggregation, revealing a role in the cell-cell interactions of hESCs. This study provides insight into the integrin signaling cascade activated in hESCs and reveals in FAK a key player in the maintenance of hESC survival and undifferentiated state.
Collapse
Affiliation(s)
- Loriana Vitillo
- North West Embryonic Stem Cell Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Melissa Baxter
- North West Embryonic Stem Cell Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Banu Iskender
- North West Embryonic Stem Cell Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Paul Whiting
- Pfizer Neusentis, The Portway Building, Granta Park, Cambridge CB21 6GS, UK
| | - Susan J Kimber
- North West Embryonic Stem Cell Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
23
|
Kaukonen R, Jacquemet G, Hamidi H, Ivaska J. Cell-derived matrices for studying cell proliferation and directional migration in a complex 3D microenvironment. Nat Protoc 2017; 12:2376-2390. [PMID: 29048422 DOI: 10.1038/nprot.2017.107] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
2D surfaces offer simple analysis of cells in culture, yet these often yield different cell morphologies and responses from those observed in vivo. Considerable effort has therefore been expended on the generation of more tissue-like environments for the study of cell behavior in vitro. Purified matrix proteins provide a 3D scaffold that better mimics the in vivo situation; however, these are far removed from the complex tissue composition seen in vivo. Cell-derived matrices (CDMs) offer a more physiologically relevant alternative for studying in vivo-like cell behavior in vitro. In the protocol described here, fibroblasts cultured on gelatin-coated surfaces are maintained in the presence of ascorbic acid to strengthen matrix deposition over 1-3 weeks. The resulting fibrillar CDMs are denuded of cells, and other cells are subsequently cultured on them, after which their behavior is monitored. We also demonstrate how to use CDMs as an in vivo-relevant reductionist model for studying tumor-stroma-induced changes in carcinoma cell proliferation and migration.
Collapse
Affiliation(s)
- Riina Kaukonen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Guillaume Jacquemet
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,Department of Biochemistry, University of Turku, Turku, Finland
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Human pluripotent stem cells (hPSCs) are anchorage-dependent cells that can be cultured on a variety of matrices and express integrins and the machinery for integrin signaling. Until recently, there has been limited understanding of exactly how integrin signaling regulates pluripotent stem cell (PSC) behavior. This review summarizes our knowledge of how integrins and focal adhesion kinase (FAK) regulate different aspects of hPSC biology. RECENT FINDINGS The latest research suggests that mouse and human embryonic stem cells utilize similar integrin signaling players but with different biological outcomes, reflecting the known developmental difference in their pluripotent status. Notably, attachment cues via FAK signaling are crucial for hPSCs survival and pluripotency maintenance. FAK may be found cortically but also in the nucleus of hPSCs intersecting core pluripotency networks. SUMMARY Integrins and FAK have been consigned to the conventional role of cell adhesion receptor systems in PSCs. This review highlights data indicating that they are firmly integrated in pluripotency circuits, with implications for both research PSC culture and scale up and use in clinical applications.
Collapse
Affiliation(s)
- Loriana Vitillo
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, University of Manchester, Michael Smith Building, Oxford Rd, Manchester, M13 9PT UK
| | - Susan J. Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, University of Manchester, Michael Smith Building, Oxford Rd, Manchester, M13 9PT UK
| |
Collapse
|
25
|
Taleahmad S, Mirzaei M, Samadian A, Hassani SN, Haynes PA, Salekdeh GH, Baharvand H. Low Focal Adhesion Signaling Promotes Ground State Pluripotency of Mouse Embryonic Stem Cells. J Proteome Res 2017; 16:3585-3595. [DOI: 10.1021/acs.jproteome.7b00322] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
| | | | | | | | | | - Ghasem Hosseini Salekdeh
- Department
of Systems Biology, Agricultural Biotechnology, Research Institute of Iran, Karaj, Iran
| | - Hossein Baharvand
- Department
of Developmental Biology, University of Science and Culture, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| |
Collapse
|
26
|
Baek JA, Seol HW, Jung J, Kim HS, Oh SK, Choi YM. Clean-Up Human Embryonic Stem Cell Lines Using Humanized Culture Condition. Tissue Eng Regen Med 2017; 14:453-464. [PMID: 30603501 DOI: 10.1007/s13770-017-0053-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 03/19/2017] [Accepted: 03/21/2017] [Indexed: 11/28/2022] Open
Abstract
Human embryonic stem cell (hESC) culture system has been changing culture conditions from conventional to xeno-free for therapeutic cell applications, and N-glycolylneuraminic acid (Neu5Gc) could be a useful indicator of xenogeneic contaminations in hESCs because human cells can no longer produce it genetically. We set up the humanized culture condition using commercially available humanized materials and two different adaptation methods: sequential or direct. SNUhES4 and H1 hESC lines, previously established in conventional culture conditions, were maintained using the humanized culture condition and were examined for the presence of Neu5Gc. The hESCs showed the same morphology and character as those of the conventional culture condition. Moreover, they were negative for Neu5Gc within two passages without loss of pluripotency. This study suggested that this method can effectively cleanse previously established hESC lines, bringing them one step closer to being clinical-grade hESCs.
Collapse
Affiliation(s)
- Jin Ah Baek
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea
| | - Hye Won Seol
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea
| | - Juwon Jung
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea
| | - Hee Sun Kim
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea.,2Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Sun Kyung Oh
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea
| | - Young Min Choi
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea.,2Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| |
Collapse
|
27
|
Protein Kinases in Pluripotency—Beyond the Usual Suspects. J Mol Biol 2017; 429:1504-1520. [DOI: 10.1016/j.jmb.2017.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/14/2022]
|
28
|
|
29
|
Cierna Z, Mego M, Jurisica I, Machalekova K, Chovanec M, Miskovska V, Svetlovska D, Kalavska K, Rejlekova K, Kajo K, Mardiak J, Babal P. Fibrillin-1 (FBN-1) a new marker of germ cell neoplasia in situ. BMC Cancer 2016; 16:597. [PMID: 27487789 PMCID: PMC4973050 DOI: 10.1186/s12885-016-2644-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 07/28/2016] [Indexed: 01/09/2023] Open
Abstract
Background Germ cell neoplasia in situ (GCNIS), is preinvasive stage of testicular germ cell tumours (TGCTs). Fibrillins, which are integral components of microfibrils are suggested to be involved in cancer pathogenesis and maintenance of embryonic stem cells pluripotency. The aim of this study was to examine fibrillin-1 (FBN-1) expression in TGCTs patients. Methods Surgical specimens from 203 patients with TGCTs were included into the translational study. FBN-1 expression was evaluated in the tumour tissue, in GCNIS and in adjacent non-neoplastic testicular tissue in all available cases. Tissue samples were processed by the tissue microarray method. FBN-1 was detected by immunohistochemistry using goat polyclonal antibody and the expression was evaluated by the multiplicative quickscore (QS). Results The highest FBN-1 positivity was detected in GCNIS (mean QS = 11.30), with overexpression of FBN-1 (QS >9) in the majority (77.1 %) of cases. Expression of FBN-1 in all subtypes of TGCTs was significantly lower in comparison to expression in GCNIS (all p <0.001). Seminoma had significantly higher expression compared to EC, ChC and TER (all p <0.05), but not to YST (p = 0.84). In non-neoplastic testicular tissue the FBN-1 positivity was very low (mean QS = 0.02). Sensitivity, specificity, positive and negative predictive value of FBN-1 expression for diagnosis of GCNIS were 97.1, 98.8, 98.6 and 97.7 %. Conclusions FBN-1 is overexpressed in TGCTs and especially in GCNIS when compared to non-neoplastic testicular tissue in patients with germ cell tumors and could be involved in germ cell neoplasia in situ development.
Collapse
Affiliation(s)
- Z Cierna
- Department of Pathology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - M Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia. .,Translational Research Unit, Faculty of Medicine, Comenius University, Bratislava, Slovakia. .,National Cancer Institute, Bratislava, Slovakia. .,2nd Department of Oncology, Faculty of Medicine, National Cancer Institute, Comenius University, Klenova 1, 833 10, Bratislava, Slovak Republic.
| | - I Jurisica
- Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Canada
| | - K Machalekova
- St. Elisabeth Cancer Institute, Bratislava, Slovakia
| | - M Chovanec
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,National Cancer Institute, Bratislava, Slovakia
| | - V Miskovska
- St. Elisabeth Cancer Institute, Bratislava, Slovakia.,1st Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - D Svetlovska
- Translational Research Unit, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,National Cancer Institute, Bratislava, Slovakia
| | - K Kalavska
- Translational Research Unit, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,National Cancer Institute, Bratislava, Slovakia
| | - K Rejlekova
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,National Cancer Institute, Bratislava, Slovakia
| | - K Kajo
- St. Elisabeth Cancer Institute, Bratislava, Slovakia
| | - J Mardiak
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,National Cancer Institute, Bratislava, Slovakia
| | - P Babal
- Department of Pathology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
30
|
Zhang L, Xu Y, Xu J, Wei Y, Xu X. Protein kinase A inhibitor, H89, significantly enhances survival rate of dissociated human embryonic stem cells following cryopreservation. Cell Prolif 2016; 49:589-98. [PMID: 27484641 DOI: 10.1111/cpr.12278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/01/2016] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES Human embryonic stem cells (hESCs) have huge potential for establishment of disease models and for treating degenerative diseases. However, the extremely low survival level of dissociated hESCs following cryopreservation is been a tremendous problem to allow for their rapid expansion, genetic manipulation and future medical applications. In this study, we have aimed to develop an efficient strategy to improve survival of dissociated hESCs after cryopreservation. MATERIALS AND METHODS Human embryonic stem cells (H9 line), dissociated into single cells, were cryopreserved using the slow-freezing method. Viable cells and their colony numbers in culture after cryopreservation were evaluated when treated with protein kinase A inhibitor H89. Western blotting was carried out to investigate mechanisms of low survival levels of dissociated hESCs following cryopreservation. Immunofluorescence, reverse transcription-polymerase chain reaction (RT-PCR), in vitro and in vivo differentiation were performed to testify to pluripotency and differentiation ability of hte cryopreserved cells treated with H89. RESULTS H89 significantly improved survival level of dissociated hESCs after cryopreservation through ROCK inhibition. H89-treated cells still maintained their pluripotency and differentiation capacity. CONCLUSIONS This new approach for cryopreservation of single hESCs, using H89, can promote potential use of hESCs in regenerative medicine in the future.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanqing Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jiandong Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuping Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xia Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
31
|
Reprogramming bladder cancer cells for studying cancer initiation and progression. Tumour Biol 2016; 37:13237-13245. [DOI: 10.1007/s13277-016-5226-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 07/14/2016] [Indexed: 10/21/2022] Open
|
32
|
Langhans MT, Yu S, Tuan RS. Stem Cells in Skeletal Tissue Engineering: Technologies and Models. Curr Stem Cell Res Ther 2016; 11:453-474. [PMID: 26423296 DOI: 10.2174/1574888x10666151001115248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/01/2015] [Accepted: 04/01/2015] [Indexed: 12/14/2022]
Abstract
This review surveys the use of pluripotent and multipotent stem cells in skeletal tissue engineering. Specific emphasis is focused on evaluating the function and activities of these cells in the context of development in vivo, and how technologies and methods of stem cell-based tissue engineering for stem cells must draw inspiration from developmental biology. Information on the embryonic origin and in vivo differentiation of skeletal tissues is first reviewed, to shed light on the persistence and activities of adult stem cells that remain in skeletal tissues after embryogenesis. Next, the development and differentiation of pluripotent stem cells is discussed, and some of their advantages and disadvantages in the context of tissue engineering are presented. The final section highlights current use of multipotent adult mesenchymal stem cells, reviewing their origin, differentiation capacity, and potential applications to tissue engineering.
Collapse
Affiliation(s)
| | | | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA 15219, USA.
| |
Collapse
|
33
|
Caiazzo M, Okawa Y, Ranga A, Piersigilli A, Tabata Y, Lutolf MP. Defined three-dimensional microenvironments boost induction of pluripotency. NATURE MATERIALS 2016; 15:344-52. [PMID: 26752655 DOI: 10.1038/nmat4536] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 12/08/2015] [Indexed: 05/25/2023]
Abstract
Since the discovery of induced pluripotent stem cells (iPSCs), numerous approaches have been explored to improve the original protocol, which is based on a two-dimensional (2D) cell-culture system. Surprisingly, nothing is known about the effect of a more biologically faithful 3D environment on somatic-cell reprogramming. Here, we report a systematic analysis of how reprogramming of somatic cells occurs within engineered 3D extracellular matrices. By modulating microenvironmental stiffness, degradability and biochemical composition, we have identified a previously unknown role for biophysical effectors in the promotion of iPSC generation. We find that the physical cell confinement imposed by the 3D microenvironment boosts reprogramming through an accelerated mesenchymal-to-epithelial transition and increased epigenetic remodelling. We conclude that 3D microenvironmental signals act synergistically with reprogramming transcription factors to increase somatic plasticity.
Collapse
Affiliation(s)
- Massimiliano Caiazzo
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Yuya Okawa
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Adrian Ranga
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | | | - Yoji Tabata
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science (SB), EPFL, 1015 Lausanne, Switzerland
| |
Collapse
|
34
|
Human Umbilical Cord Blood-Derived Serum for Culturing the Supportive Feeder Cells of Human Pluripotent Stem Cell Lines. Stem Cells Int 2015; 2016:4626048. [PMID: 26839561 PMCID: PMC4709772 DOI: 10.1155/2016/4626048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/02/2015] [Accepted: 09/13/2015] [Indexed: 12/24/2022] Open
Abstract
Although human pluripotent stem cells (hPSCs) can proliferate robustly on the feeder-free culture system, genetic instability of hPSCs has been reported in such environment. Alternatively, feeder cells enable hPSCs to maintain their pluripotency. The feeder cells are usually grown in a culture medium containing fetal bovine serum (FBS) prior to coculture with hPSCs. The use of FBS might limit the clinical application of hPSCs. Recently, human cord blood-derived serum (hUCS) showed a positive effect on culture of mesenchymal stem cells. It is interesting to test whether hUCS can be used for culture of feeder cells of hPSCs. This study was aimed to replace FBS with hUCS for culturing the human foreskin fibroblasts (HFFs) prior to feeder cell preparation. The results showed that HFFs cultured in hUCS-containing medium (HFF-hUCS) displayed fibroblastic features, high proliferation rates, short population doubling times, and normal karyotypes after prolonged culture. Inactivated HFF-hUCS expressed important genes, including Activin A, FGF2, and TGFβ1, which have been implicated in the maintenance of hPSC pluripotency. Moreover, hPSC lines maintained pluripotency, differentiation capacities, and karyotypic stability after being cocultured for extended period with inactivated HFF-hUCS. Therefore, the results demonstrated the benefit of hUCS for hPSCs culture system.
Collapse
|
35
|
Leha A, Moens N, Meleckyte R, Culley OJ, Gervasio MK, Kerz M, Reimer A, Cain SA, Streeter I, Folarin A, Stegle O, Kielty CM, Durbin R, Watt FM, Danovi D. A high-content platform to characterise human induced pluripotent stem cell lines. Methods 2015; 96:85-96. [PMID: 26608109 PMCID: PMC4773406 DOI: 10.1016/j.ymeth.2015.11.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/11/2015] [Accepted: 11/17/2015] [Indexed: 01/16/2023] Open
Abstract
iPSCs show inter/intra-line/donor-variability hampering characterisation. HipSci generates, banks and provides iPSCs from hundreds of individual donors. iPSCs respond to different human plasma fibronectin concentrations on 96-well assays. Phenotypic features: cell number, proliferation, morphology and intercellular adhesion. The methodologies described can be tailored for disease-modelling and other cell types.
Induced pluripotent stem cells (iPSCs) provide invaluable opportunities for future cell therapies as well as for studying human development, modelling diseases and discovering therapeutics. In order to realise the potential of iPSCs, it is crucial to comprehensively characterise cells generated from large cohorts of healthy and diseased individuals. The human iPSC initiative (HipSci) is assessing a large panel of cell lines to define cell phenotypes, dissect inter- and intra-line and donor variability and identify its key determinant components. Here we report the establishment of a high-content platform for phenotypic analysis of human iPSC lines. In the described assay, cells are dissociated and seeded as single cells onto 96-well plates coated with fibronectin at three different concentrations. This method allows assessment of cell number, proliferation, morphology and intercellular adhesion. Altogether, our strategy delivers robust quantification of phenotypic diversity within complex cell populations facilitating future identification of the genetic, biological and technical determinants of variance. Approaches such as the one described can be used to benchmark iPSCs from multiple donors and create novel platforms that can readily be tailored for disease modelling and drug discovery.
Collapse
Affiliation(s)
- Andreas Leha
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Nathalie Moens
- HipSci Cell Phenotyping, Centre for Stem Cells and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Ruta Meleckyte
- HipSci Cell Phenotyping, Centre for Stem Cells and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Oliver J Culley
- HipSci Cell Phenotyping, Centre for Stem Cells and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Mia K Gervasio
- HipSci Cell Phenotyping, Centre for Stem Cells and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Maximilian Kerz
- HipSci Cell Phenotyping, Centre for Stem Cells and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK; NIHR Biomedical Research Centre for Mental Health Informatics Core, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Andreas Reimer
- HipSci Cell Phenotyping, Centre for Stem Cells and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Stuart A Cain
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Ian Streeter
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Amos Folarin
- NIHR Biomedical Research Centre for Mental Health Informatics Core, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Cay M Kielty
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | | | - Richard Durbin
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Fiona M Watt
- HipSci Cell Phenotyping, Centre for Stem Cells and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Davide Danovi
- HipSci Cell Phenotyping, Centre for Stem Cells and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|
36
|
Applying Proteomics to Investigate Extracellular Matrix in Health and Disease. CURRENT TOPICS IN MEMBRANES 2015; 76:171-96. [PMID: 26610914 DOI: 10.1016/bs.ctm.2015.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The molecular composition of basement membranes (BMs) has traditionally been investigated by candidate-based approaches leading to the identification of key structural components as described in previous chapters. Laminins, collagen IV, nidogens, perlecan, and type XV/XVIII collagen are integral to BMs with isoforms showing tissue specificity. More recently the application of mass spectrometry (MS)-based proteomics has led to the discovery of many more structural and regulatory components of BMs and more broadly, extracellular matrix (ECM). These investigations have revealed tissue-specific signatures of between 100 and 150 ECM components, demonstrating the complexity of the extracellular niche. In addition to providing a structural scaffold for cells, ECM is a dynamic extracellular environment capable of regulating the physical properties of tissues. Global investigations of ECM with proteomics in turn enable systems level analyses and when applied to health and disease states these investigations provide insights into pathways regulating matrix dysregulation. This chapter focuses on the methods used to extract ECM and on the analysis of its composition using MS-based proteomics, and it provides examples of how these approaches have been used to investigate health and disease states.
Collapse
|
37
|
Pettinato G, Wen X, Zhang N. Engineering Strategies for the Formation of Embryoid Bodies from Human Pluripotent Stem Cells. Stem Cells Dev 2015; 24:1595-609. [PMID: 25900308 DOI: 10.1089/scd.2014.0427] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are powerful tools for regenerative therapy and studying human developmental biology, attributing to their ability to differentiate into many functional cell types in the body. The main challenge in realizing hPSC potential is to guide their differentiation in a well-controlled manner. One way to control the cell differentiation process is to recapitulate during in vitro culture the key events in embryogenesis to obtain the three developmental germ layers from which all cell types arise. To achieve this goal, many techniques have been tested to obtain a cellular cluster, an embryoid body (EB), from both mouse and hPSCs. Generation of EBs that are homogeneous in size and shape would allow directed hPSC differentiation into desired cell types in a more synchronous manner and define the roles of cell-cell interaction and spatial organization in lineage specification in a setting similar to in vivo embryonic development. However, previous success in uniform EB formation from mouse PSCs cannot be extrapolated to hPSCs possibly due to the destabilization of adherens junctions on cell surfaces during the dissociation into single cells, making hPSCs extremely vulnerable to cell death. Recently, new advances have emerged to form uniform human embryoid bodies (hEBs) from dissociated single cells of hPSCs. In this review, the existing methods for hEB production from hPSCs and the results on the downstream differentiation of the hEBs are described with emphases on the efficiency, homogeneity, scalability, and reproducibility of the hEB formation process and the yield in terminal differentiation. New trends in hEB production and directed differentiation are discussed.
Collapse
Affiliation(s)
- Giuseppe Pettinato
- 1 Department of Biomedical Engineering, Virginia Commonwealth University , Richmond, Virginia.,2 Department of Chemical and Life Science Engineering, Virginia Commonwealth University , Richmond, Virginia
| | - Xuejun Wen
- 2 Department of Chemical and Life Science Engineering, Virginia Commonwealth University , Richmond, Virginia.,3 Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Ning Zhang
- 1 Department of Biomedical Engineering, Virginia Commonwealth University , Richmond, Virginia
| |
Collapse
|
38
|
Randles MJ, Woolf AS, Huang JL, Byron A, Humphries JD, Price KL, Kolatsi-Joannou M, Collinson S, Denny T, Knight D, Mironov A, Starborg T, Korstanje R, Humphries MJ, Long DA, Lennon R. Genetic Background is a Key Determinant of Glomerular Extracellular Matrix Composition and Organization. J Am Soc Nephrol 2015; 26:3021-34. [PMID: 25896609 DOI: 10.1681/asn.2014040419] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 02/16/2015] [Indexed: 12/27/2022] Open
Abstract
Glomerular disease often features altered histologic patterns of extracellular matrix (ECM). Despite this, the potential complexities of the glomerular ECM in both health and disease are poorly understood. To explore whether genetic background and sex determine glomerular ECM composition, we investigated two mouse strains, FVB and B6, using RNA microarrays of isolated glomeruli combined with proteomic glomerular ECM analyses. These studies, undertaken in healthy young adult animals, revealed unique strain- and sex-dependent glomerular ECM signatures, which correlated with variations in levels of albuminuria and known predisposition to progressive nephropathy. Among the variation, we observed changes in netrin 4, fibroblast growth factor 2, tenascin C, collagen 1, meprin 1-α, and meprin 1-β. Differences in protein abundance were validated by quantitative immunohistochemistry and Western blot analysis, and the collective differences were not explained by mutations in known ECM or glomerular disease genes. Within the distinct signatures, we discovered a core set of structural ECM proteins that form multiple protein-protein interactions and are conserved from mouse to man. Furthermore, we found striking ultrastructural changes in glomerular basement membranes in FVB mice. Pathway analysis of merged transcriptomic and proteomic datasets identified potential ECM regulatory pathways involving inhibition of matrix metalloproteases, liver X receptor/retinoid X receptor, nuclear factor erythroid 2-related factor 2, notch, and cyclin-dependent kinase 5. These pathways may therefore alter ECM and confer susceptibility to disease.
Collapse
Affiliation(s)
- Michael J Randles
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom; Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - Adrian S Woolf
- Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - Jennifer L Huang
- Developmental Biology and Cancer Program, Institute of Child Health, University College London, London, United Kingdom
| | - Adam Byron
- Edinburgh Cancer Research United Kingdom Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom; and
| | - Jonathan D Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Karen L Price
- Developmental Biology and Cancer Program, Institute of Child Health, University College London, London, United Kingdom
| | - Maria Kolatsi-Joannou
- Developmental Biology and Cancer Program, Institute of Child Health, University College London, London, United Kingdom
| | - Sophie Collinson
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Thomas Denny
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom; Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - David Knight
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Aleksandr Mironov
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Toby Starborg
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - David A Long
- Developmental Biology and Cancer Program, Institute of Child Health, University College London, London, United Kingdom
| | - Rachel Lennon
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom; Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom;
| |
Collapse
|
39
|
Holley RJ, Tai G, Williamson AJK, Taylor S, Cain SA, Richardson SM, Merry CLR, Whetton AD, Kielty CM, Canfield AE. Comparative quantification of the surfaceome of human multipotent mesenchymal progenitor cells. Stem Cell Reports 2015; 4:473-88. [PMID: 25684225 PMCID: PMC4375938 DOI: 10.1016/j.stemcr.2015.01.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 01/12/2015] [Accepted: 01/12/2015] [Indexed: 12/23/2022] Open
Abstract
Mesenchymal progenitor cells have great therapeutic potential, yet incomplete characterization of their cell-surface interface limits their clinical exploitation. We have employed subcellular fractionation with quantitative discovery proteomics to define the cell-surface interface proteome of human bone marrow mesenchymal stromal/stem cells (MSCs) and human umbilical cord perivascular cells (HUCPVCs). We compared cell-surface-enriched fractions from MSCs and HUCPVCs (three donors each) with adult mesenchymal fibroblasts using eight-channel isobaric-tagging mass spectrometry, yielding relative quantification on >6,000 proteins with high confidence. This approach identified 186 upregulated mesenchymal progenitor biomarkers. Validation of 10 of these markers, including ROR2, EPHA2, and PLXNA2, confirmed upregulated expression in mesenchymal progenitor populations and distinct roles in progenitor cell proliferation, migration, and differentiation. Our approach has delivered a cell-surface proteome repository that now enables improved selection and characterization of human mesenchymal progenitor populations.
Collapse
Affiliation(s)
- Rebecca J Holley
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Guangping Tai
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Andrew J K Williamson
- Faculty of Medical and Human Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Samuel Taylor
- Faculty of Medical and Human Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Stuart A Cain
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Stephen M Richardson
- Faculty of Medical and Human Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Catherine L R Merry
- Faculty of Engineering and Physical Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Anthony D Whetton
- Faculty of Medical and Human Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Cay M Kielty
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK.
| | - Ann E Canfield
- Faculty of Medical and Human Sciences, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
40
|
Revealing cytokine-induced changes in the extracellular matrix with secondary ion mass spectrometry. Acta Biomater 2015; 14:70-83. [PMID: 25523877 DOI: 10.1016/j.actbio.2014.12.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/22/2014] [Accepted: 12/08/2014] [Indexed: 11/23/2022]
Abstract
Cell-secreted matrices (CSMs), where extracellular matrix (ECM) deposited by monolayer cell cultures is decellularized, have been increasingly used to produce surfaces that may be reseeded with cells. Such surfaces are useful to help us understand cell-ECM interactions in a microenvironment closer to the in vivo situation than synthetic substrates with adsorbed proteins. We describe the production of CSMs from mouse primary osteoblasts (mPObs) exposed to cytokine challenge during matrix secretion, mimicking in vivo inflammatory environments. Time-of-flight secondary ion mass spectrometry data revealed that CSMs with cytokine challenge at day 7 or 12 of culture can be chemically distinguished from one another and from untreated CSM using multivariate analysis. Comparison of the differences with reference spectra from adsorbed protein mixtures points towards cytokine challenge resulting in a decrease in collagen content. This is supported by immunocytochemical and histological staining, demonstrating a 44% loss of collagen mass and a 32% loss in collagen I coverage. CSM surfaces demonstrate greater cell adhesion than adsorbed ECM proteins. When mPObs were reseeded onto cytokine-challenged CSMs they exhibited reduced adhesion and elongated morphology compared to untreated CSMs. Such changes may direct subsequent cell fate and function, and provide insights into pathological responses at sites of inflammation.
Collapse
|
41
|
Dreixler JC, Poston JN, Balyasnikova I, Shaikh AR, Tupper KY, Conway S, Boddapati V, Marcet MM, Lesniak MS, Roth S. Delayed administration of bone marrow mesenchymal stem cell conditioned medium significantly improves outcome after retinal ischemia in rats. Invest Ophthalmol Vis Sci 2014; 55:3785-96. [PMID: 24699381 DOI: 10.1167/iovs.13-11683] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Delayed treatment after ischemia is often unsatisfactory. We hypothesized that injection of bone marrow stem cell (BMSC) conditioned medium after ischemia could rescue ischemic retina, and in this study we characterized the functional and histological outcomes and mechanisms of this neuroprotection. METHODS Retinal ischemia was produced in adult Wistar rats by increasing intraocular pressure for 55 minutes. Conditioned medium (CM) from rat BMSCs or unconditioned medium (uCM) was injected into the vitreous 24 hours after the end of ischemia. Recovery was assessed 7 days after ischemia using electroretinography, at which time we euthanized the animals and then prepared 4-μm-thick paraffin-embedded retinal sections. TUNEL and Western blot were used to identify apoptotic cells and apoptosis-related gene expression 24 hours after injections; that is, 48 hours after ischemia. Protein content in CM versus uCM was studied using tandem mass spectrometry, and bioinformatics methods were used to model protein interactions. RESULTS Intravitreal injection of CM 24 hours after ischemia significantly improved retinal function and attenuated cell loss in the retinal ganglion cell layer. CM attenuated postischemic apoptosis and apoptosis-related gene expression. By spectral counting, 19 proteins that met stringent identification criteria were increased in the CM compared to uCM; the majority were extracellular matrix proteins that mapped into an interactional network together with other proteins involved in cell growth and adhesion. CONCLUSIONS By restoring retinal function, attenuating apoptosis, and preventing retinal cell loss after ischemia, CM is a robust means of delayed postischemic intervention. We identified some potential candidate proteins for this effect.
Collapse
Affiliation(s)
- John C Dreixler
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, United States
| | - Jacqueline N Poston
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Irina Balyasnikova
- Department of Surgery (Neurosurgery), The University of Chicago, Chicago, Illinois, United States
| | - Afzhal R Shaikh
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, United States
| | - Kelsey Y Tupper
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, United States
| | - Sineadh Conway
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, United States
| | - Venkat Boddapati
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, United States
| | - Marcus M Marcet
- Department of Surgery (Ophthalmology and Visual Science), The University of Chicago, Chicago, Illinois, United States
| | - Maciej S Lesniak
- Department of Surgery (Neurosurgery), The University of Chicago, Chicago, Illinois, United States
| | - Steven Roth
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
42
|
Byron A, Randles MJ, Humphries JD, Mironov A, Hamidi H, Harris S, Mathieson PW, Saleem MA, Satchell SC, Zent R, Humphries MJ, Lennon R. Glomerular cell cross-talk influences composition and assembly of extracellular matrix. J Am Soc Nephrol 2014; 25:953-66. [PMID: 24436469 DOI: 10.1681/asn.2013070795] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The glomerular basement membrane (GBM) is a specialized extracellular matrix (ECM) compartment within the glomerulus that contains tissue-restricted isoforms of collagen IV and laminin. It is integral to the capillary wall and therefore, functionally linked to glomerular filtration. Although the composition of the GBM has been investigated with global and candidate-based approaches, the relative contributions of glomerular cell types to the production of ECM are not well understood. To characterize specific cellular contributions to the GBM, we used mass spectrometry-based proteomics to analyze ECM isolated from podocytes and glomerular endothelial cells in vitro. These analyses identified cell type-specific differences in ECM composition, indicating distinct contributions to glomerular ECM assembly. Coculture of podocytes and endothelial cells resulted in an altered composition and organization of ECM compared with monoculture ECMs, and electron microscopy revealed basement membrane-like ECM deposition between cocultured cells, suggesting the involvement of cell-cell cross-talk in the production of glomerular ECM. Notably, compared with monoculture ECM proteomes, the coculture ECM proteome better resembled a tissue-derived glomerular ECM dataset, indicating its relevance to GBM in vivo. Protein network analyses revealed a common core of 35 highly connected structural ECM proteins that may be important for glomerular ECM assembly. Overall, these findings show the complexity of the glomerular ECM and suggest that both ECM composition and organization are context-dependent.
Collapse
Affiliation(s)
- Adam Byron
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences and
| | - Michael J Randles
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences and Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Aleksandr Mironov
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences and
| | - Hellyeh Hamidi
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences and
| | - Shelley Harris
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - Peter W Mathieson
- Academic Renal Unit, Faculty of Medicine and Dentistry, University of Bristol, Bristol, United Kingdom
| | - Moin A Saleem
- Academic Renal Unit, Faculty of Medicine and Dentistry, University of Bristol, Bristol, United Kingdom
| | - Simon C Satchell
- Academic Renal Unit, Faculty of Medicine and Dentistry, University of Bristol, Bristol, United Kingdom
| | - Roy Zent
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Medicine, Veterans Affairs Hospital, Nashville, Tennessee
| | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences and
| | - Rachel Lennon
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences and Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom;
| |
Collapse
|
43
|
Establishment and characterization of human engineered cells stably expressing large extracellular matrix proteins. Arch Pharm Res 2013; 37:149-56. [DOI: 10.1007/s12272-013-0294-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 11/10/2013] [Indexed: 10/26/2022]
|