1
|
Subramanian B, Williams S, Karp S, Hennino MF, Jacas S, Lee M, Riella CV, Alper SL, Higgs HN, Pollak MR. Missense Mutant Gain-of-Function Causes Inverted Formin 2 (INF2)-Related Focal Segmental Glomerulosclerosis (FSGS). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.08.598088. [PMID: 38915495 PMCID: PMC11195136 DOI: 10.1101/2024.06.08.598088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Inverted formin-2 (INF2) gene mutations are among the most common causes of genetic focal segmental glomerulosclerosis (FSGS) with or without Charcot-Marie-Tooth (CMT) disease. Recent studies suggest that INF2, through its effects on actin and microtubule arrangement, can regulate processes including vesicle trafficking, cell adhesion, mitochondrial calcium uptake, mitochondrial fission, and T-cell polarization. Despite roles for INF2 in multiple cellular processes, neither the human pathogenic R218Q INF2 point mutation nor the INF2 knock-out allele is sufficient to cause disease in mice. This discrepancy challenges our efforts to explain the disease mechanism, as the link between INF2-related processes, podocyte structure, disease inheritance pattern, and their clinical presentation remains enigmatic. Here, we compared the kidney responses to puromycin aminonucleoside (PAN) induced injury between R218Q INF2 point mutant knock-in and INF2 knock-out mouse models and show that R218Q INF2 mice are susceptible to developing proteinuria and FSGS. This contrasts with INF2 knock-out mice, which show only a minimal kidney phenotype. Co-localization and co-immunoprecipitation analysis of wild-type and mutant INF2 coupled with measurements of cellular actin content revealed that the R218Q INF2 point mutation confers a gain-of-function effect by altering the actin cytoskeleton, facilitated in part by alterations in INF2 localization. Differential analysis of RNA expression in PAN-stressed heterozygous R218Q INF2 point-mutant and heterozygous INF2 knock-out mouse glomeruli showed that the adhesion and mitochondria-related pathways were significantly enriched in the disease condition. Mouse podocytes with R218Q INF2, and an INF2-mutant human patient's kidney organoid-derived podocytes with an S186P INF2 mutation, recapitulate the defective adhesion and mitochondria phenotypes. These results link INF2-regulated cellular processes to the onset and progression of glomerular disease. Thus, our data demonstrate that gain-of-function mechanisms drive INF2-related FSGS and explain the autosomal dominant inheritance pattern of this disease.
Collapse
|
2
|
Katagade V, Kandroo M, Ratnaparkhi A. Embryonic spatiotemporal expression pattern of Folded gastrulation suggests roles in multiple morphogenetic events and regulation by AbdA. G3 (BETHESDA, MD.) 2024; 14:jkae032. [PMID: 38366558 DOI: 10.1093/g3journal/jkae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/03/2023] [Accepted: 01/31/2024] [Indexed: 02/18/2024]
Abstract
In Drosophila, the signaling pathway activated by the ligand Folded gastrulation (Fog) is among the few known G protein-coupled receptor (GPCR) pathways to regulate cell shape change with a well-characterized role in gastrulation. However, an understanding of the spectrum of morphogenetic events regulated by Fog signaling is still lacking. Here, we present an analysis of the expression pattern and regulation of fog using a genome-engineered Fog::sfGFP line. We show that Fog expression is widespread and in tissues previously not associated with the signaling pathway including germ cells, trachea, and amnioserosa. In the central nervous system (CNS), we find that the ligand is expressed in multiple types of glia indicating a prominent role in the development of these cells. Consistent with this, we have identified 3 intronic enhancers whose expression in the CNS overlaps with Fog::sfGFP. Further, we show that enhancer-1, (fogintenh-1) located proximal to the coding exon is responsive to AbdA. Supporting this, we find that fog expression is downregulated in abdA mutants. Together, our study highlights the broad scope of Fog-GPCR signaling during embryogenesis and identifies Hox gene AbdA as a novel regulator of fog expression.
Collapse
Affiliation(s)
- Vrushali Katagade
- MACS-Agharkar Research Institute (Affiliated to Savitribai Phule Pune University), Developmental Biology Group, G.G. Agarkar Road, Pune 411 004, Maharashtra, India
| | - Manisha Kandroo
- MACS-Agharkar Research Institute (Affiliated to Savitribai Phule Pune University), Developmental Biology Group, G.G. Agarkar Road, Pune 411 004, Maharashtra, India
| | - Anuradha Ratnaparkhi
- MACS-Agharkar Research Institute (Affiliated to Savitribai Phule Pune University), Developmental Biology Group, G.G. Agarkar Road, Pune 411 004, Maharashtra, India
| |
Collapse
|
3
|
Ferreira SA, Tallia F, Heyraud A, Walker SA, Salzlechner C, Jones JR, Rankin SM. 3D printed hybrid scaffolds do not induce adverse inflammation in mice and direct human BM-MSC chondrogenesis in vitro. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100087. [PMID: 38312434 PMCID: PMC10835132 DOI: 10.1016/j.bbiosy.2024.100087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
Biomaterials that can improve the healing of articular cartilage lesions are needed. To address this unmet need, we developed novel 3D printed silica/poly(tetrahydrofuran)/poly(ε-caprolactone) (SiO2/PTHF/PCL-diCOOH) hybrid scaffolds. Our aim was to carry out essential studies to advance this medical device towards functional validation in pre-clinical trials. First, we show that the chemical composition, microarchitecture and mechanical properties of these scaffolds were not affected by sterilisation with gamma irradiation. To evaluate the systemic and local immunogenic reactivity of the sterilised 3D printed hybrid scaffolds, they were implanted subcutaneously into Balb/c mice. The scaffolds did not trigger a systemic inflammatory response over one week of implantation. The interaction between the host immune system and the implanted scaffold elicited a local physiological reaction with infiltration of mononuclear cells without any signs of a chronic inflammatory response. Then, we investigated how these 3D printed hybrid scaffolds direct chondrogenesis in vitro. Human bone marrow-derived mesenchymal stem/stromal cells (hBM-MSCs) seeded within the 3D printed hybrid scaffolds were cultured under normoxic or hypoxic conditions, with or without chondrogenic supplements. Chondrogenic differentiation assessed by both gene expression and protein production analyses showed that 3D printed hybrid scaffolds support hBM-MSC chondrogenesis. Articular cartilage-specific extracellular matrix deposition within these scaffolds was enhanced under hypoxic conditions (1.7 or 3.7 fold increase in the median of aggrecan production in basal or chondrogenic differentiation media). Our findings show that 3D printed SiO2/PTHF/PCL-diCOOH hybrid scaffolds have the potential to support the regeneration of cartilage tissue.
Collapse
Affiliation(s)
| | | | - Agathe Heyraud
- Department of Materials, Imperial College London, London, UK
| | - Simone A. Walker
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | - Julian R. Jones
- Department of Materials, Imperial College London, London, UK
| | - Sara M. Rankin
- National Heart & Lung Institute, Imperial College London, London, UK
| |
Collapse
|
4
|
Huang Y, Sun M, Lu Z, Zhong Q, Tan M, Wei Q, Zheng L. Role of integrin β1 and tenascin C mediate TGF-SMAD2/3 signaling in chondrogenic differentiation of BMSCs induced by type I collagen hydrogel. Regen Biomater 2024; 11:rbae017. [PMID: 38525326 PMCID: PMC10960929 DOI: 10.1093/rb/rbae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/24/2024] [Accepted: 02/09/2024] [Indexed: 03/26/2024] Open
Abstract
Cartilage defects may lead to severe degenerative joint diseases. Tissue engineering based on type I collagen hydrogel that has chondrogenic potential is ideal for cartilage repair. However, the underlying mechanisms of chondrogenic differentiation driven by type I collagen hydrogel have not been fully clarified. Herein, we explored potential collagen receptors and chondrogenic signaling pathways through bioinformatical analysis to investigate the mechanism of collagen-induced chondrogenesis. Results showed that the super enhancer-related genes induced by collagen hydrogel were significantly enriched in the TGF-β signaling pathway, and integrin-β1 (ITGB1), a receptor of collagen, was highly expressed in bone marrow mesenchymal stem cells (BMSCs). Further analysis showed genes such as COL2A1 and Tenascin C (TNC) that interacted with ITGB1 were significantly enriched in extracellular matrix (ECM) structural constituents in the chondrogenic induction group. Knockdown of ITGB1 led to the downregulation of cartilage-specific genes (SOX9, ACAN, COL2A1), SMAD2 and TNC, as well as the downregulation of phosphorylation of SMAD2/3. Knockdown of TNC also resulted in the decrease of cartilage markers, ITGB1 and the SMAD2/3 phosphorylation but overexpression of TNC showed the opposite trend. Finally, in vitro and in vivo experiments confirmed the involvement of ITGB1 and TNC in collagen-mediated chondrogenic differentiation and cartilage regeneration. In summary, we demonstrated that ITGB1 was a crucial receptor for chondrogenic differentiation of BMSCs induced by collagen hydrogel. It can activate TGF-SMAD2/3 signaling, followed by impacting TNC expression, which in turn promotes the interaction of ITGB1 and TGF-SMAD2/3 signaling to enhance chondrogenesis. These may provide concernful support for cartilage tissue engineering and biomaterials development.
Collapse
Affiliation(s)
- Yuanjun Huang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Miao Sun
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
| | - Zhenhui Lu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Life Science Institute, Guangxi Medical University, Nanning 530021, China
| | - Qiuling Zhong
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
| | - Manli Tan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Life Science Institute, Guangxi Medical University, Nanning 530021, China
| | - Qingjun Wei
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Life Science Institute, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
5
|
Hallström GF, Jones DL, Locke RC, Bonnevie ED, Kim SY, Laforest L, Garcia DC, Mauck RL. Microenvironmental mechanoactivation through Yap/Taz suppresses chondrogenic gene expression. Mol Biol Cell 2023; 34:ar73. [PMID: 37043309 PMCID: PMC10295477 DOI: 10.1091/mbc.e22-12-0543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/23/2023] [Accepted: 04/06/2023] [Indexed: 04/13/2023] Open
Abstract
Chondrocyte phenotype is preserved when cells are round and the actin cytoskeleton is cortical. Conversely, these cells rapidly dedifferentiate in vitro with increased mechanoactive Rho signaling, which increases cell size and causes large actin stress fiber to form. While the effects of Rho on chondrocyte phenotype are well established, the molecular mechanism is not yet fully elucidated. Yap, a transcriptional coregulator, is regulated by Rho in a mechanotransductive manner and can suppress chondrogenesis in vivo. Here, we sought to elucidate the relationship between mechanoactive Rho and Yap on chondrogenic gene expression. We first show that decreasing mechanoactive state through Rho inhibition results in a broad increase in chondrogenic gene expression. Next, we show that Yap and its coregulator Taz are negative regulators of chondrogenic gene expression, and removal of these factors promotes chondrogenesis even in environments that promote cell spreading. Finally, we establish that Yap/Taz is essential for translating Rho-mediated signals to negatively regulate chondrogenic gene expression, and that its removal negates the effects of increased Rho signaling. Together, these data indicate that Rho is a mechanoregulator of chondrogenic differentiation, and that its impact on chondrogenic expression is exerted principally through mechanically induced translocation and activity of Yap and Taz.
Collapse
Affiliation(s)
- Grey F. Hallström
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Dakota L. Jones
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
| | - Ryan C. Locke
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Edward D. Bonnevie
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Sung Yeon Kim
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Lorielle Laforest
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
| | - Diana Cruz Garcia
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| |
Collapse
|
6
|
Cui L, Yang Z, Hong J, Zhu Z, Wang Z, Liu Z, Zheng W, Hao Y, He J, Ni P, Cheng G. Injectable and Degradable POSS-Polyphosphate-Polysaccharide Hybrid Hydrogel Scaffold for Cartilage Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:20625-20637. [PMID: 37078820 DOI: 10.1021/acsami.2c22947] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The limited self-repair capacity of articular cartilage has motivated the development of stem cell therapy based on artificial scaffolds that mimic the extracellular matrix (ECM) of cartilage tissue. In view of the specificity of articular cartilage, desirable tissue adhesiveness and stable mechanical properties under cyclic mechanical loads are critical for cartilage scaffolds. Herein, we developed an injectable and degradable organic-inorganic hybrid hydrogel as a cartilage scaffold based on polyhedral oligomeric silsesquioxane (POSS)-cored polyphosphate and polysaccharide. Specifically, acrylated 8-arm star-shaped POSS-poly(ethyl ethylene phosphate) (POSS-8PEEP-AC) was synthesized and cross-linked with thiolated hyaluronic acid (HA-SH) to form a degradable POSS-PEEP/HA hydrogel. Incorporation of POSS in the hydrogel increased the mechanical properties. The POSS-PEEP/HA hydrogel showed enzymatic biodegradability and favorable biocompatibility, supporting the growth and differentiation of human mesenchymal stem cells (hMSCs). The chondrogenic differentiation of encapsulated hMSCs was promoted by loading transforming growth factor-β3 (TGF-β3) in the hydrogel. In addition, the injectable POSS-PEEP/HA hydrogel was capable of adhering to rat cartilage tissue and resisting cyclic compression. Furthermore, in vivo results revealed that the transplanted hMSCs encapsulated in the POSS-PEEP/HA hydrogel scaffold significantly improved cartilage regeneration in rats, while the conjugation of TGF-β3 achieved a better therapeutic effect. The present work demonstrated the potential of the injectable, biodegradable, and mechanically enhanced POSS-PEEP/HA hybrid hydrogel as a scaffold biomaterial for cartilage regeneration.
Collapse
Affiliation(s)
- Leisha Cui
- School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Zun Yang
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Soochow University, Suzhou 215123, China
| | - Jing Hong
- School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Zhanchi Zhu
- School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Zhaojun Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Zhongqing Liu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Wenlong Zheng
- Suzhou Kowloon Hospital Shanghai Jiao Tong University School of Medicine, Suzhou 215021, Jiangsu, China
| | - Ying Hao
- School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| | - Jinlin He
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Soochow University, Suzhou 215123, China
| | - Peihong Ni
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Soochow University, Suzhou 215123, China
| | - Guosheng Cheng
- School of Nano-Tech and Nano Bionics, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, China
| |
Collapse
|
7
|
Tavasolian F, Inman RD. Biology and therapeutic potential of mesenchymal stem cell extracellular vesicles in axial spondyloarthritis. Commun Biol 2023; 6:413. [PMID: 37059822 PMCID: PMC10104809 DOI: 10.1038/s42003-023-04743-z] [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: 08/02/2022] [Accepted: 03/21/2023] [Indexed: 04/16/2023] Open
Abstract
Axial spondyloarthritis (AxSpA) is a chronic, inflammatory, autoimmune disease that predominantly affects the joints of the spine, causes chronic pain, and, in advanced stages, may result in spinal fusion. Recent developments in understanding the immunomodulatory and tissue-differentiating properties of mesenchymal stem cell (MSC) therapy have raised the possibility of applying such treatment to AxSpA. The therapeutic effectiveness of MSCs has been shown in numerous studies spanning a range of diseases. Several studies have been conducted examining acellular therapy based on MSC secretome. Extracellular vesicles (EVs) generated by MSCs have been proven to reproduce the impact of MSCs on target cells. These EVs are associated with immunological regulation, tissue remodeling, and cellular homeostasis. EVs' biological effects rely on their cargo, with microRNAs (miRNAs) integrated into EVs playing a particularly important role in gene expression regulation. In this article, we will discuss the impact of MSCs and EVs generated by MSCs on target cells and how these may be used as unique treatment strategies for AxSpA.
Collapse
Affiliation(s)
- Fataneh Tavasolian
- Spondylitis Program, Division of Rheumatology, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada
| | - Robert D Inman
- Spondylitis Program, Division of Rheumatology, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
- Departments of Medicine and Immunology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
Pham HL, Yang DH, Chae WR, Jung JH, Hoang TX, Lee NY, Kim JY. PDMS Micropatterns Coated with PDA and RGD Induce a Regulatory Macrophage-like Phenotype. MICROMACHINES 2023; 14:673. [PMID: 36985080 PMCID: PMC10052727 DOI: 10.3390/mi14030673] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 06/18/2023]
Abstract
Regulatory macrophages (Mreg) are a special cell type that present a potential therapeutic strategy for various inflammatory diseases. In vitro, Mreg generation mainly takes 7-10 days of treatment with chemicals, including cytokines. In the present study, we established a new approach for Mreg generation using a three-dimensional (3D) micropatterned polydimethylsiloxane (PDMS) surface coated with a natural biopolymer adhesive polydopamine (PDA) and the common cell adhesion peptide motif arginylglycylaspartic acid (RGD). The 3D PDMS surfaces were fabricated by photolithography and soft lithography techniques and were subsequently coated with an RGD+PDA mixture to form a surface that facilitates cell adhesion. Human monocytes (THP-1 cells) were cultured on different types of 2D or 3D micropatterns for four days, and the cell morphology, elongation, and Mreg marker expression were assessed using microscopic and flow cytometric analyses. The cells grown on the PDA+RGD-coated 3D micropatterns (20-µm width/20-µm space) exhibited the most elongated morphology and strongest expression levels of Mreg markers, such as CD163, CD206, CD209, CD274, MER-TK, TREM2, and DHRS9. The present study demonstrated that PDA+RGD-coated 3D PDMS micropatterns successfully induced Mreg-like cells from THP-1 cells within four days without the use of cytokines, suggesting a time- and cost-effective method to generate Mreg-like cells in vitro.
Collapse
Affiliation(s)
- Hoang Lan Pham
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Da Hyun Yang
- Department of BioNano Technology, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Woo Ri Chae
- Department of BioNano Technology, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Jong Hyeok Jung
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Thi Xoan Hoang
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Nae Yoon Lee
- Department of BioNano Technology, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Jae Young Kim
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| |
Collapse
|
9
|
Goh D, Yang Y, Lee EH, Hui JHP, Yang Z. Managing the Heterogeneity of Mesenchymal Stem Cells for Cartilage Regenerative Therapy: A Review. Bioengineering (Basel) 2023; 10:bioengineering10030355. [PMID: 36978745 PMCID: PMC10045936 DOI: 10.3390/bioengineering10030355] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/12/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Articular cartilage defects commonly result from trauma and are associated with significant morbidity. Since cartilage is an avascular, aneural, and alymphatic tissue with a poor intrinsic healing ability, the regeneration of functional hyaline cartilage remains a difficult clinical problem. Mesenchymal stem cells (MSCs) are multipotent cells with multilineage differentiation potential, including the ability to differentiate into chondrocytes. Due to their availability and ease of ex vivo expansion, clinicians are increasingly applying MSCs in the treatment of cartilage lesions. However, despite encouraging pre-clinical and clinical data, inconsistencies in MSC proliferative and chondrogenic potential depending on donor, tissue source, cell subset, culture conditions, and handling techniques remain a key barrier to widespread clinical application of MSC therapy in cartilage regeneration. In this review, we highlight the strategies to manage the heterogeneity of MSCs ex vivo for more effective cartilage repair, including reducing the MSC culture expansion period, and selecting MSCs with higher chondrogenic potential through specific genetic markers, surface markers, and biophysical attributes. The accomplishment of a less heterogeneous population of culture-expanded MSCs may improve the scalability, reproducibility, and standardisation of MSC therapy for clinical application in cartilage regeneration.
Collapse
Affiliation(s)
- Doreen Goh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Yanmeng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
- Correspondence: ; Tel.: +65-6516-5398
| |
Collapse
|
10
|
Nellinger S, Kluger PJ. How Mechanical and Physicochemical Material Characteristics Influence Adipose-Derived Stem Cell Fate. Int J Mol Sci 2023; 24:ijms24043551. [PMID: 36834966 PMCID: PMC9961531 DOI: 10.3390/ijms24043551] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/28/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are a subpopulation of mesenchymal stem cells. Compared to bone marrow-derived stem cells, they can be harvested with minimal invasiveness. ASCs can be easily expanded and were shown to be able to differentiate into several clinically relevant cell types. Therefore, this cell type represents a promising component in various tissue engineering and medical approaches (e.g., cell therapy). In vivo cells are surrounded by the extracellular matrix (ECM) that provides a wide range of tissue-specific physical and chemical cues, such as stiffness, topography, and chemical composition. Cells can sense the characteristics of their ECM and respond to them in a specific cellular behavior (e.g., proliferation or differentiation). Thus, in vitro biomaterial properties represent an important tool to control ASCs behavior. In this review, we give an overview of the current research in the mechanosensing of ASCs and current studies investigating the impact of material stiffens, topography, and chemical modification on ASC behavior. Additionally, we outline the use of natural ECM as a biomaterial and its interaction with ASCs regarding cellular behavior.
Collapse
Affiliation(s)
- Svenja Nellinger
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
| | - Petra Juliane Kluger
- School of Life Sciences, Reutlingen University, 72762 Reutlingen, Germany
- Correspondence: ; Tel.: +49-07121-271-2061
| |
Collapse
|
11
|
Qin S, Zhu J, Zhang G, Sui Q, Niu Y, Ye W, Ma G, Liu H. Research progress of functional motifs based on growth factors in cartilage tissue engineering: A review. Front Bioeng Biotechnol 2023; 11:1127949. [PMID: 36824354 PMCID: PMC9941568 DOI: 10.3389/fbioe.2023.1127949] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
Osteoarthritis is a chronic degenerative joint disease that exerts significant impacts on personal life quality, and cartilage tissue engineering is a practical treatment in clinical. Various growth factors are involved in cartilage regeneration and play important roles therein, which is the focus of current cartilage repair strategy. To compensate for the purification difficulty, high cost, poor metabolic stability, and circulating dilution of natural growth factors, the concept of functional motifs (also known as mimetic peptides) from original growth factor was introduced in recent studies. Here, we reviewed the selection mechanisms, biological functions, carrier scaffolds, and modification methods of growth factor-related functional motifs, and evaluated the repair performance in cartilage tissue engineering. Finally, the prospects of functional motifs in researches and clinical application were discussed.
Collapse
Affiliation(s)
- Shengao Qin
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Jiaman Zhu
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China,Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guangyong Zhang
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Qijia Sui
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Yimeng Niu
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China
| | - Weilong Ye
- School of Stomatology, Dalian Medical University, Dalian, China,Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China,*Correspondence: Weilong Ye, ; Guowu Ma, ; Huiying Liu,
| | - Guowu Ma
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China,*Correspondence: Weilong Ye, ; Guowu Ma, ; Huiying Liu,
| | - Huiying Liu
- School of Stomatology, Dalian Medical University, Dalian, China,Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian, China,*Correspondence: Weilong Ye, ; Guowu Ma, ; Huiying Liu,
| |
Collapse
|
12
|
Jalandhra GK, Molley TG, Hung TT, Roohani I, Kilian KA. In situ formation of osteochondral interfaces through "bone-ink" printing in tailored microgel suspensions. Acta Biomater 2023; 156:75-87. [PMID: 36055612 DOI: 10.1016/j.actbio.2022.08.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/21/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2023]
Abstract
Osteochondral tissue has a complex hierarchical structure spanning subchondral bone to articular cartilage. Biomaterials approaches to mimic and repair these interfaces have had limited success, largely due to challenges in fabricating composite hard-soft interfaces with living cells. Biofabrication approaches have emerged as attractive methods to form osteochondral analogues through additive assembly of hard and soft components. We have developed a unique printing platform that is able to integrate soft and hard materials concurrently through freeform printing of mineralized constructs within tunable microgel suspensions containing living cells. A library of microgels based on gelatin were prepared, where the stiffness of the microgels and a liquid "filler" phase can be tuned for bioprinting while simultaneously directing differentiation. Tuning microgel stiffness and filler content differentially directs chondrogenesis and osteogenesis within the same construct, demonstrating how this technique can be used to fabricate osteochondral interfaces in a single step. Printing of a rapidly setting calcium phosphate cement, so called "bone-ink" within a cell laden suspension bath further guides differentiation, where the cells adjacent to the nucleated hydroxyapatite phase undergo osteogenesis with cells in the surrounding medium undergoing chondrogenesis. In this way, bone analogues with hierarchical structure can be formed within cell-laden gradient soft matrices to yield multiphasic osteochondral constructs. This technique provides a versatile one-pot biofabrication approach without harsh post-processing which will aid efforts in bone disease modelling and tissue engineering. STATEMENT OF SIGNIFICANCE: This paper demonstrates the first example of a biofabrication approach to rapidly form osteochondral constructs in a single step under physiological conditions. Key to this advance is a tunable suspension of extracellular matrix microgels that are packed together with stem cells, providing a unique and modular scaffolding for guiding the simultaneous formation of bone and cartilage tissue. The physical properties of the suspension allow direct writing of a ceramic "bone-ink", resulting in an ordered structure of microscale hydrogels, living cells, and bone mimics in a single step. This platform reveals a simple approach to making complex skeletal tissue for disease modelling, with the possibility of repairing and replacing bone-cartilage interfaces in the clinic using a patient's own cells.
Collapse
Affiliation(s)
- Gagan K Jalandhra
- School of Materials Science and Engineering, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052
| | - Thomas G Molley
- School of Materials Science and Engineering, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052
| | - Tzong-Tyng Hung
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney NSW 2052
| | - Iman Roohani
- School of Chemistry, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052
| | - Kristopher A Kilian
- School of Materials Science and Engineering, University of New South Wales, Sydney NSW 2052; School of Chemistry, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052.
| |
Collapse
|
13
|
Tassinari R, Olivi E, Cavallini C, Taglioli V, Zannini C, Marcuzzi M, Fedchenko O, Ventura C. Mechanobiology: A landscape for reinterpreting stem cell heterogeneity and regenerative potential in diseased tissues. iScience 2022; 26:105875. [PMID: 36647385 PMCID: PMC9839966 DOI: 10.1016/j.isci.2022.105875] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mechanical forces play a fundamental role in cellular dynamics from the molecular level to the establishment of complex heterogeneity in somatic and stem cells. Here, we highlight the role of cytoskeletal mechanics and extracellular matrix in generating mechanical forces merging into oscillatory synchronized patterns. We discuss how cellular mechanosensing/-transduction can be modulated by mechanical forces to control tissue metabolism and set the basis for nonpharmacologic tissue rescue. Control of bone anabolic activity and repair, as well as obesity prevention, through a fine-tuning of the stem cell morphodynamics are highlighted. We also discuss the use of mechanical forces in the treatment of cardiovascular diseases and heart failure through the fine modulation of stem cell metabolic activity and regenerative potential. We finally focus on the new landscape of delivering specific mechanical stimuli to reprogram tissue-resident stem cells and enhance our self-healing potential, without the need for stem cell or tissue transplantation.
Collapse
Affiliation(s)
| | - Elena Olivi
- ELDOR LAB, via Corticella 183, 40129 Bologna, Italy
| | | | | | | | - Martina Marcuzzi
- NIBB, National Institute of Biostructures and Biosystems, National Laboratory of Molecular Biology and Stem Cell Engineering, via Corticella 183, 40129 Bologna, Italy
| | - Oleksandra Fedchenko
- NIBB, National Institute of Biostructures and Biosystems, National Laboratory of Molecular Biology and Stem Cell Engineering, via Corticella 183, 40129 Bologna, Italy
| | - Carlo Ventura
- ELDOR LAB, via Corticella 183, 40129 Bologna, Italy,NIBB, National Institute of Biostructures and Biosystems, National Laboratory of Molecular Biology and Stem Cell Engineering, via Corticella 183, 40129 Bologna, Italy,Corresponding author
| |
Collapse
|
14
|
Liu Q, Dai W, Gao Y, Dong L, Jia H, Li S, Guo L, Fan Y, Zhang X. The synergistic regulation of chondrogenesis by collagen-based hydrogels and cell co-culture. Acta Biomater 2022; 154:194-211. [PMID: 36309191 DOI: 10.1016/j.actbio.2022.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/19/2022] [Accepted: 10/07/2022] [Indexed: 01/24/2023]
Abstract
The suitable seeding cells and scaffolds are very important for tissue engineering to create functional cartilage. Although the physicochemical properties of scaffold and co-culture system of mesenchymal stem cells (MSCs) and chondrocytes could affect functional properties of engineered cartilage tissues respectively, the combined effects of them on chondrogenesis is currently unknown. Herein, methacrylated collagen (CMA30 and CMA80) hydrogels with different degradation rate and stiffness were prepared. The MSCs and chondrocytes were co-cultured or monocultured in collagen, CMA30 and CMA80 hydrogels in vitro or in vivo. The results demonstrated that cell spreading and proliferation was regulated by degradation rate and stiffness of hydrogels. Compared to single MSCs culture, co-culture cells in all collagen-based hydrogels significantly improved chondrogenesis. CMA30 hydrogel with moderate degradation rate and low storage modulus was the most effective for co-culture system to promote chondrogenesis compared to Col and CMA80 hydrogel in vitro culture, while there was no obvious difference between CMA30 and CMA80 hydrogel in vivo. Furthermore, the intercellular substance exchange was very important for co-culture system to maintain the positive effect on chondrogenesis. Overall, the current study highlights the synergistic effects of the physicochemical properties of collagen-based hydrogel and co-culture system on cartilage formation. STATEMENT OF SIGNIFICANCE: Scaffolds and cells play a key role in cartilage tissue engineering. The combined effects of physicochemical properties of collagen hydrogels and co-culture system (MSCs and chondrocytes) on chondrogenesis is unknown. In contrast to the studies that investigated the effect of single factor (scaffolds or cells) on cartilage formation, this manuscript explored the synergistic regulation of both scaffold properties and biological factors on chondrogenesis, and provided a promising strategy for cartilage tissue engineering.
Collapse
Affiliation(s)
- Qingli Liu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Wenling Dai
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Yongli Gao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Longpeng Dong
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Hengxing Jia
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Shikui Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Likun Guo
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China.
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| |
Collapse
|
15
|
Molley TG, Hung TT, Kilian KA. Cell-Laden Gradient Microgel Suspensions for Spatial Control of Differentiation During Biofabrication. Adv Healthc Mater 2022; 11:e2201122. [PMID: 35866537 PMCID: PMC9780160 DOI: 10.1002/adhm.202201122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/21/2022] [Indexed: 01/28/2023]
Abstract
During tissue development, stem and progenitor cells form functional tissue with high cellular diversity and intricate micro- and macro-architecture. Current approaches have attempted to replicate this process with materials cues or through spontaneous cell self-organization. However, cell-directed and materials-directed organization are required simultaneously to achieve biomimetic structure and function. Here, it is shown how integrating live adipose derived stem cells with gradient microgel suspensions steers divergent differentiation outcomes. Microgel matrices composed of small particles are found to promote adipogenic differentiation, while larger particles fostered increased cell spreading and osteogenic differentiation. Tuning the matrix formulation demonstrates that early cell adhesion and spreading dictate differentiation outcome. Combining small and large microgels into gradients spatially directs proliferation and differentiation over time. After 21 days of culture, osteogenic conditions foster significant mineralization within the individual microgels, thereby providing cell-directed changes in composition and mechanics within the gradient porous scaffold. Freeform printing of high-density cell suspensions is performed across these gradients to demonstrate the potential for hierarchical tissue biofabrication. Interstitial porosity influences cell expansion from the print and microgel size guides spatial differentiation, thereby providing scope to fabricate tissue gradients at multiple scales through integrated and printed cell populations.
Collapse
Affiliation(s)
- Thomas G Molley
- School of Materials Science and Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Tzong-Tyng Hung
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Kristopher A Kilian
- School of Materials Science and Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
16
|
Chen T, Cao F, Peng W, Wei R, Xu Q, Feng B, Wang J, Weng J, Wang M, Zhang X. Optimal regeneration and repair of critical size articular cartilage driven by endogenous CLECSF1. Biomed Signal Process Control 2022. [DOI: 10.1016/j.bspc.2022.103898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
17
|
Liu S, Kanchanawong P. Emerging interplay of cytoskeletal architecture, cytomechanics and pluripotency. J Cell Sci 2022; 135:275761. [PMID: 35726598 DOI: 10.1242/jcs.259379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pluripotent stem cells (PSCs) are capable of differentiating into all three germ layers and trophoblasts, whereas tissue-specific adult stem cells have a more limited lineage potency. Although the importance of the cytoskeletal architecture and cytomechanical properties in adult stem cell differentiation have been widely appreciated, how they contribute to mechanotransduction in PSCs is less well understood. Here, we discuss recent insights into the interplay of cellular architecture, cell mechanics and the pluripotent states of PSCs. Notably, the distinctive cytomechanical and morphodynamic profiles of PSCs are accompanied by a number of unique molecular mechanisms. The extent to which such mechanobiological signatures are intertwined with pluripotency regulation remains an open question that may have important implications in developmental morphogenesis and regenerative medicine.
Collapse
Affiliation(s)
- Shiying Liu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore.,Department of Biomedical Engineering, National University of Singapore, Singapore 117411, Republic of Singapore
| |
Collapse
|
18
|
Decellularised Cartilage ECM Culture Coatings Drive Rapid and Robust Chondrogenic Differentiation of Human Periosteal Cells. Bioengineering (Basel) 2022; 9:bioengineering9050203. [PMID: 35621481 PMCID: PMC9137502 DOI: 10.3390/bioengineering9050203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
The control of cell behaviour in an effort to create highly homogeneous cultures is becoming an area of intense research, both to elucidate fundamental biology and for regenerative applications. The extracellular matrix (ECM) controls many cellular processes in vivo, and as such is a rich source of cues that may be translated in vitro. Herein, we describe the creation of cell culture coatings from porcine decellularised hyaline cartilage through enzymatic digestion. Surprisingly, heat-mediated sterilisation created a coating with the capacity to rapidly and robustly induce chondrogenic differentiation of human periosteal cells. This differentiation was validated through the alteration of cell phenotype from a fibroblastic to a cuboidal/cobblestone chondrocyte-like appearance. Moreover, chondrogenic gene expression further supported this observation, where cells cultured on heat sterilised ECM-coated plastic displayed higher expression of COL2A1, ACAN and PRG4 (p < 0.05) compared to non-coated plastic cultures. Interestingly, COL2A1 and ACAN expression in this context were sensitive to initial cell density; however, SOX9 expression appeared to be mainly driven by the coating independent of seeding density. The creation of a highly chondrogenic coating may provide a cost-effective solution for the differentiation and/or expansion of human chondrocytes aimed towards cartilage repair strategies.
Collapse
|
19
|
Che H, Selig M, Rolauffs B. Micro-patterned cell populations as advanced pharmaceutical drugs with precise functional control. Adv Drug Deliv Rev 2022; 184:114169. [PMID: 35217114 DOI: 10.1016/j.addr.2022.114169] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/29/2022]
Abstract
Human cells are both advanced pharmaceutical drugs and 'drug deliverers'. However, functional control prior to or after cell implantation remains challenging. Micro-patterning cells through geometrically defined adhesion sites allows controlling morphogenesis, polarity, cellular mechanics, proliferation, migration, differentiation, stemness, cell-cell interactions, collective cell behavior, and likely immuno-modulatory properties. Consequently, generating micro-patterned therapeutic cells is a promising idea that has not yet been realized and few if any steps have been undertaken in this direction. This review highlights potential therapeutic applications, summarizes comprehensively the many cell functions that have been successfully controlled through micro-patterning, details the established micro-pattern designs, introduces the available fabrication technologies to the non-specialized reader, and suggests a quality evaluation score. Such a broad review is not yet available but would facilitate the manufacturing of therapeutically patterned cell populations using micro-patterned cell-instructive biomaterials for improved functional control as drug delivery systems in the context of cells as pharmaceutical products.
Collapse
Affiliation(s)
- Hui Che
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215006, China
| | - Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany.
| |
Collapse
|
20
|
Barlian A, Saputri DHA, Hernando A, Khoirinaya C, Prajatelistia E, Tanoto H. Spidroin striped micropattern promotes chondrogenic differentiation of human Wharton's jelly mesenchymal stem cells. Sci Rep 2022; 12:4837. [PMID: 35319008 PMCID: PMC8941093 DOI: 10.1038/s41598-022-08982-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 03/14/2022] [Indexed: 11/29/2022] Open
Abstract
Cartilage tissue engineering, particularly micropattern, can influence the biophysical properties of mesenchymal stem cells (MSCs) leading to chondrogenesis. In this research, human Wharton’s jelly MSCs (hWJ-MSCs) were grown on a striped micropattern containing spider silk protein (spidroin) from Argiope appensa. This research aims to direct hWJ-MSCs chondrogenesis using micropattern made of spidroin bioink as opposed to fibronectin that often used as the gold standard. Cells were cultured on striped micropattern of 500 µm and 1000 µm width sizes without chondrogenic differentiation medium for 21 days. The immunocytochemistry result showed that spidroin contains RGD sequences and facilitates cell adhesion via integrin β1. Chondrogenesis was observed through the expression of glycosaminoglycan, type II collagen, and SOX9. The result on glycosaminoglycan content proved that 1000 µm was the optimal width to support chondrogenesis. Spidroin micropattern induced significantly higher expression of SOX9 mRNA on day-21 and SOX9 protein was located inside the nucleus starting from day-7. COL2A1 mRNA of spidroin micropattern groups was downregulated on day-21 and collagen type II protein was detected starting from day-14. These results showed that spidroin micropattern enhances chondrogenic markers while maintains long-term upregulation of SOX9, and therefore has the potential as a new method for cartilage tissue engineering.
Collapse
Affiliation(s)
- Anggraini Barlian
- School of Life Sciences and Technology, Bandung Institute of Technology, Bandung, West Java, 40132, Indonesia. .,Research Center for Nanosciences and Nanotechnology, Bandung Institute of Technology, Bandung, West Java, 40132, Indonesia.
| | - Dinda Hani'ah Arum Saputri
- School of Life Sciences and Technology, Bandung Institute of Technology, Bandung, West Java, 40132, Indonesia
| | - Adriel Hernando
- School of Life Sciences and Technology, Bandung Institute of Technology, Bandung, West Java, 40132, Indonesia
| | - Candrani Khoirinaya
- School of Life Sciences and Technology, Bandung Institute of Technology, Bandung, West Java, 40132, Indonesia
| | - Ekavianty Prajatelistia
- Faculty of Mechanical and Aerospace Engineering, Bandung Institute of Technology, Bandung, West Java, 40132, Indonesia
| | - Hutomo Tanoto
- Faculty of Mechanical and Aerospace Engineering, Bandung Institute of Technology, Bandung, West Java, 40132, Indonesia
| |
Collapse
|
21
|
Tuvshindorj U, Trouillet V, Vasilevich A, Koch B, Vermeulen S, Carlier A, Alexander MR, Giselbrecht S, Truckenmüller R, de Boer J. The Galapagos Chip Platform for High-Throughput Screening of Cell Adhesive Chemical Micropatterns. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105704. [PMID: 34985808 DOI: 10.1002/smll.202105704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/04/2021] [Indexed: 06/14/2023]
Abstract
In vivo cells reside in a complex extracellular matrix (ECM) that presents spatially distributed biochemical and -physical cues at the nano- to micrometer scales. Chemical micropatterning is successfully used to generate adhesive islands to control where and how cells attach and restore cues of the ECM in vitro. Although chemical micropatterning has become a powerful tool to study cell-material interactions, only a fraction of the possible micropattern designs was covered so far, leaving many other possible designs still unexplored. Here, a high-throughput screening platform called "Galapagos chip" is developed. It contains a library of 2176 distinct subcellular chemical patterns created using mathematical algorithms and a straightforward UV-induced two-step surface modification. This approach enables the immobilization of ligands in geometrically defined regions onto cell culture substrates. To validate the system, binary RGD/polyethylene glycol patterns are prepared on which human mesenchymal stem cells are cultured, and the authors observe how different patterns affect cell and organelle morphology. As proof of concept, the cells are stained for the mechanosensitive YAP protein, and, using a machine-learning algorithm, it is demonstrated that cell shape and YAP nuclear translocation correlate. It is concluded that the Galapagos chip is a versatile platform to screen geometrical aspects of cell-ECM interaction.
Collapse
Affiliation(s)
- Urandelger Tuvshindorj
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
- Department of Biomedical Engineering and Institute, for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Vanessa Trouillet
- Institute for Applied Materials and Karlsruhe Nano Micro Facility, Karlsruhe Institute of Technology, 76344, Eggenstein-Leopoldshafen, Germany
| | - Aliaksei Vasilevich
- Department of Biomedical Engineering and Institute, for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Britta Koch
- Advanced Materials and Healthcare Technologies Division, The School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Steven Vermeulen
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Morgan R Alexander
- Advanced Materials and Healthcare Technologies Division, The School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering and Institute, for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| |
Collapse
|
22
|
Aprile P, Whelan IT, Sathy BN, Carroll SF, Kelly DJ. Soft Hydrogel Environments that Facilitate Cell Spreading and Aggregation Preferentially Support Chondrogenesis of Adult Stem Cells. Macromol Biosci 2022; 22:e2100365. [PMID: 35171524 DOI: 10.1002/mabi.202100365] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/14/2022] [Indexed: 11/10/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) represent a promising cell type for treating damaged and diseased synovial joints. The therapeutic potential of MSCs will be facilitated by the engineering of biomaterial environments capable of directing their fate. Here we explored the interplay between matrix elasticity and cell morphology in regulating the chondrogenic differentiation of MSCs when seeded onto or encapsulated within hydrogels made of interpenetrating networks (IPN) of alginate and collagen type I. This IPN system enabled the independent control of substrate stiffness (in 2D and in 3D) and cell morphology (3D only). In a 2D culture environment, the expression of chondrogenic markers SOX9, ACAN and COL2 increased on a soft substrate, which correlated with increased SMAD2/3 nuclear localization, enhanced MSCs condensation and the formation of larger cellular aggregates. The encapsulation of spread MSCs within a soft IPN dramatically increased the expression of cartilage-specific genes, which was linked to higher levels of cellular condensation and nuclear SMAD2/3 localization. Surprisingly, cells forced to adopt a more rounded morphology within the same soft IPNs expressed higher levels of the osteogenic markers RUNX2 and COL1. The insight provided by this study suggests that a mechanobiology informed approach to biomaterial development will be integral to the development of successful cartilage tissue engineering strategies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Paola Aprile
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Ian T Whelan
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,CÚRAM Center for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Binulal N Sathy
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Centre for Nanoscience and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Simon F Carroll
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,CÚRAM Center for Research in Medical Devices, National University of Ireland, Galway, Ireland.,The Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Trinity College Dublin, Ireland
| |
Collapse
|
23
|
Zhao Y, Sun Q, Huo B. Focal adhesion regulates osteogenic differentiation of mesenchymal stem cells and osteoblasts. BIOMATERIALS TRANSLATIONAL 2021; 2:312-322. [PMID: 35837413 PMCID: PMC9255797 DOI: 10.12336/biomatertransl.2021.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/29/2021] [Accepted: 12/10/2021] [Indexed: 11/18/2022]
Abstract
Focal adhesions are large macromolecular assemblies through which cells are connected with the extracellular matrix so that extracellular signals can be transmitted inside cells. Some studies have focused on the effect of cell shape on the differentiation of stem cells, but little attention has been paid to focal adhesion. In the present study, mesenchymal stem cells (MSCs) and osteoblast-like MC3T3-E1 cells were seeded onto micropatterned substrates on which circular adhesive islands with different spacing and area were created for focal adhesion. Results showed that the patterns of focal adhesion changed cell morphology but did not affect cell survival. For MSCs cultured for 3 days, patterns with small circles and large spacing promoted osteogenesis. For MSCs cultured for 7 days, patterns with large circles and spacing enhanced osteogenesis. For MC3T3-E1 cells, the patterns of focal adhesion had no effect on cell differentiation after 3 days of culture, but patterns with small circles and spacing improved osteogenic differentiation after 7 days. Moreover, the assembly of F-actin, phosphorylation of myosin, and nuclear translocation of yes-associated proteins (YAP) were consistent with the expression of differentiation markers, indicating that the pattern of focal adhesion may affect the osteogenesis of MSCs and osteoblasts through changes in cytoskeletal tension and nuclear localisation of YAP.
Collapse
Affiliation(s)
| | | | - Bo Huo
- Corresponding author: Bo Huo,
| |
Collapse
|
24
|
Brito Barrera YA, Husteden C, Alherz J, Fuhrmann B, Wölk C, Groth T. Extracellular matrix-inspired surface coatings functionalized with dexamethasone-loaded liposomes to induce osteo- and chondrogenic differentiation of multipotent stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112516. [PMID: 34857295 DOI: 10.1016/j.msec.2021.112516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 10/20/2022]
Abstract
Biomimetic surface coatings can be combined with conventional implants to mimic the extracellular matrix (ECM) of the surrounding tissue to make them more biocompatible. Layer-by-layer technique (LbL) can be used for making surface coatings by alternating adsorption of polyanions and polycations from aqueous solutions without need of chemical reactions. Here, polyelectrolyte multilayer (PEM) systems is made of hyaluronic acid (HA) as polyanion and Collagen I (Col) as polycation to mimic the ECM of connective tissue. The PEM are combined with dexamethasone (Dex)-loaded liposomes to achieve a local delivery and protection of this drug for stimulation of osteo- and chondrogenic differentiation of multipotent stem cells. The liposomes possess a positive surface charge that is required for immobilization on the PEM. The surface properties of PEM system show a positive zeta potential after liposome adsorption and a decrease in wettability, both promoting cell adhesion and spreading of C3H10T1/2 multipotent embryonic mouse fibroblasts. Differentiation of C3H10T1/2 was more prominent on the PEM system with embedded Dex-loaded liposomes compared to the basal PEM system and the use of free Dex-loaded liposomes in the supernatant. This was evident by immunohistochemical staining and an upregulation of the expression of genes, which play a key role in osteogenesis (RunX2, ALP, Osteocalcin (OCN)) and chondrogenesis (Sox9, aggrecan (ACAN), collagen type II), determined by quantitative Real-time polymerase chain reaction (qRT-PCR) after 21 days. These findings indicate that the designed liposome-loaded PEM system have high potential for use as drug delivery systems for implant coatings that can induce bone and cartilage differentiation needed for example in osteochondral implants.
Collapse
Affiliation(s)
- Yazmin A Brito Barrera
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Heinrich-Damerow-Strasse 4, 06120 Halle (Saale), Germany
| | - Catharina Husteden
- Medicinal Chemistry Department, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Jumanah Alherz
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Heinrich-Damerow-Strasse 4, 06120 Halle (Saale), Germany
| | - Bodo Fuhrmann
- Interdisciplinary Center of Materials Science, Martin Luther University Halle-Wittenberg, D-06099 Halle (Saale), Germany
| | - Christian Wölk
- Pharmaceutical Technology, Institute of Pharmacy, Faculty of Medicine, Leipzig University, 04317 Leipzig, Germany
| | - Thomas Groth
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Heinrich-Damerow-Strasse 4, 06120 Halle (Saale), Germany; Interdisciplinary Center of Materials Science, Martin Luther University Halle-Wittenberg, D-06099 Halle (Saale), Germany; Laboratory of Biomedical Nanotechnologies, Institute of Bionic Technologies and Engineering, I.M. Sechenov First Moscow State University, 119991, Trubetskaya street 8, Moscow, Russian Federation.
| |
Collapse
|
25
|
Vaidyanathan K, Wang C, Krajnik A, Yu Y, Choi M, Lin B, Jang J, Heo SJ, Kolega J, Lee K, Bae Y. A machine learning pipeline revealing heterogeneous responses to drug perturbations on vascular smooth muscle cell spheroid morphology and formation. Sci Rep 2021; 11:23285. [PMID: 34857846 PMCID: PMC8640073 DOI: 10.1038/s41598-021-02683-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
Machine learning approaches have shown great promise in biology and medicine discovering hidden information to further understand complex biological and pathological processes. In this study, we developed a deep learning-based machine learning algorithm to meaningfully process image data and facilitate studies in vascular biology and pathology. Vascular injury and atherosclerosis are characterized by neointima formation caused by the aberrant accumulation and proliferation of vascular smooth muscle cells (VSMCs) within the vessel wall. Understanding how to control VSMC behaviors would promote the development of therapeutic targets to treat vascular diseases. However, the response to drug treatments among VSMCs with the same diseased vascular condition is often heterogeneous. Here, to identify the heterogeneous responses of drug treatments, we created an in vitro experimental model system using VSMC spheroids and developed a machine learning-based computational method called HETEROID (heterogeneous spheroid). First, we established a VSMC spheroid model that mimics neointima-like formation and the structure of arteries. Then, to identify the morphological subpopulations of drug-treated VSMC spheroids, we used a machine learning framework that combines deep learning-based spheroid segmentation and morphological clustering analysis. Our machine learning approach successfully showed that FAK, Rac, Rho, and Cdc42 inhibitors differentially affect spheroid morphology, suggesting that multiple drug responses of VSMC spheroid formation exist. Overall, our HETEROID pipeline enables detailed quantitative drug characterization of morphological changes in neointima formation, that occurs in vivo, by single-spheroid analysis.
Collapse
Affiliation(s)
- Kalyanaraman Vaidyanathan
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA
| | - Chuangqi Wang
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Amanda Krajnik
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA
| | - Yudong Yu
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Moses Choi
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Bolun Lin
- Department of Computer Science, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Junbong Jang
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Su-Jin Heo
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John Kolega
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA
| | - Kwonmoo Lee
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA.
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA.
| |
Collapse
|
26
|
Walters B, Turner PA, Rolauffs B, Hart ML, Stegemann JP. Controlled Growth Factor Delivery and Cyclic Stretch Induces a Smooth Muscle Cell-like Phenotype in Adipose-Derived Stem Cells. Cells 2021; 10:cells10113123. [PMID: 34831345 PMCID: PMC8624888 DOI: 10.3390/cells10113123] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are an abundant and easily accessible multipotent stem cell source with potential application in smooth muscle regeneration strategies. In 3D collagen hydrogels, we investigated whether sustained release of growth factors (GF) PDGF-AB and TGF-β1 from GF-loaded microspheres could induce a smooth muscle cell (SMC) phenotype in ASCs, and if the addition of uniaxial cyclic stretch could enhance the differentiation level. This study demonstrated that the combination of cyclic stretch and GF release over time from loaded microspheres potentiated the differentiation of ASCs, as quantified by protein expression of early to late SMC differentiation markers (SMA, TGLN and smooth muscle MHC). The delivery of GFs via microspheres produced large ASCs with a spindle-shaped, elongated SMC-like morphology. Cyclic strain produced the largest, longest, and most spindle-shaped cells regardless of the presence or absence of growth factors or the growth factor delivery method. Protein expression and cell morphology data confirmed that the sustained release of GFs from GF-loaded microspheres can be used to promote the differentiation of ASCs into SMCs and that the addition of uniaxial cyclic stretch significantly enhances the differentiation level, as quantified by intermediate and late SMC markers and a SMC-like elongated cell morphology.
Collapse
Affiliation(s)
- Brandan Walters
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Paul A. Turner
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Bernd Rolauffs
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
| | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| |
Collapse
|
27
|
Review on material parameters to enhance bone cell function in vitro and in vivo. Biochem Soc Trans 2021; 48:2039-2050. [PMID: 32940685 DOI: 10.1042/bst20200210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
Bone plays critical roles in support, protection, movement, and metabolism. Although bone has an innate capacity for regeneration, this capacity is limited, and many bone injuries and diseases require intervention. Biomaterials are a critical component of many treatments to restore bone function and include non-resorbable implants to augment bone and resorbable materials to guide regeneration. Biomaterials can vary considerably in their biocompatibility and bioactivity, which are functions of specific material parameters. The success of biomaterials in bone augmentation and regeneration is based on their effects on the function of bone cells. Such functions include adhesion, migration, inflammation, proliferation, communication, differentiation, resorption, and vascularization. This review will focus on how different material parameters can enhance bone cell function both in vitro and in vivo.
Collapse
|
28
|
Shen H, He Y, Wang N, Fritch MR, Li X, Lin H, Tuan RS. Enhancing the potential of aged human articular chondrocytes for high-quality cartilage regeneration. FASEB J 2021; 35:e21410. [PMID: 33617078 DOI: 10.1096/fj.202002386r] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/03/2021] [Accepted: 01/19/2021] [Indexed: 11/11/2022]
Abstract
Autologous chondrocyte implantation (ACI) is a regenerative procedure used to treat focal articular cartilage defects in knee joints. However, age has been considered as a limiting factor and ACI is not recommended for patients older than 40-50 years of age. One reason for this may be due to the reduced capacity of aged chondrocytes in generating new cartilage. Currently, the underlying mechanism contributing to aging-associated functional decline in chondrocytes is not clear and no proven approach exists to reverse chondrocyte aging. Given that chondrocytes in healthy hyaline cartilage typically display a spherical shape, believed to be essential for chondrocyte phenotype stability, we hypothesize that maintaining aged chondrocytes in a suspension culture that forces the cells to adopt a round morphology may help to "rejuvenate" them to a younger state, thus, leading to enhanced cartilage regeneration. Chondrocytes isolated from aged donors displayed reduced proliferation potential and impaired capacity in generating hyaline cartilage, compared to cells isolated from young donors, indicated by increased hypertrophy and cellular senescence. To test our hypothesis, the "old" chondrocytes were seeded as a suspension onto an agarose-based substratum, where they maintained a round morphology. After the 3-day suspension culture, aged chondrocytes displayed enhanced replicative capacity, compared to those grown adherent to tissue culture plastic. Moreover, chondrocytes subjected to suspension culture formed new cartilage in vitro with higher quality and quantity, with enhanced cartilage matrix deposition, concomitant with lower levels of hypertrophy and cellular senescence markers. Mechanistic analysis suggested the involvement of the RhoA and ERK1/2 signaling pathways in the "rejuvenation" process. In summary, our study presents a robust and straightforward method to enhance the function of aged human chondrocytes, which can be conveniently used to generate a large number of high-quality chondrocytes for ACI application in the elderly.
Collapse
Affiliation(s)
- He Shen
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yuchen He
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ning Wang
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Madalyn R Fritch
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xinyu Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rocky S Tuan
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
29
|
Bi J, Li Q, Yang Z, Cai L, Lv T, Yang X, Yan L, Liu X, Wang Q, Fu X, Xiao R. CXCL2 Impairs Functions of Bone Marrow Mesenchymal Stem Cells and Can Serve as a Serum Marker in High-Fat Diet-Fed Rats. Front Cell Dev Biol 2021; 9:687942. [PMID: 34327200 PMCID: PMC8315099 DOI: 10.3389/fcell.2021.687942] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/17/2021] [Indexed: 12/27/2022] Open
Abstract
In modern society excessive consumption of a high-fat diet (HFD) is a significant risk factor for many diseases such as diabetes, osteoarthritis and certain cancers. Resolving cellular and molecular mechanisms underlying HFD-associated disorders is of great importance to human health. Mesenchymal stem cells (MSCs) are key players in tissue homeostasis and adversely affected by prolonged HFD feeding. Low-grade systemic inflammation induced by HFD is characterized by increased levels of pro-inflammatory cytokines and alters homeostasis in many organs. However, whether, which and how HFD associated inflammatory cytokines impair MSCs remain unclear. Here we demonstrated that HFD induced serum cytokines disturbances, especially a continuous elevation of serum CXCL2 level in rats. Coincidentally, the differentially expressed genes (DEGs) of bone marrow MSCs (BMSCs) which functions were impaired in HFD rats were enriched in cytokine signaling. Further mechanism analysis revealed that CXCL2 treatment in vitro suppresses the adipogenic potential of BMSCs via Rac1 activation, and promoted BMSC migration and senescence by inducing over-production of ELMO1 and reactive oxygen species (ROS) respectively. Moreover, we found that although glycolipid metabolism indicators can be corrected, the CXCL2 elevation and BMSC dysfunctions cannot be fully rescued by diet correction and anti-inflammatory aspirin treatment, indicating the long-lasting deleterious effects of HFD on serum CXCL2 levels and BMSC functions. Altogether, our findings identify CXCL2 as an important regulator in BMSCs functions and may serve as a serum marker to indicate the BMSC dysfunctions induced by HFD. In addition, our findings underscore the intricate link among high-fat intake, chronic inflammation and BMSC dysfunction which may facilitate development of protective strategies for HFD associated diseases.
Collapse
Affiliation(s)
- Jianhai Bi
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiuchen Li
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhigang Yang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Cai
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Lv
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xun Yang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Yan
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xia Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Wang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Integrin-Linked Kinase (ILK) Regulates Urinary Stem Cells Differentiation into Smooth Muscle via NF- κB Signal Pathway. Stem Cells Int 2021; 2021:6633111. [PMID: 33854551 PMCID: PMC8019365 DOI: 10.1155/2021/6633111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/28/2021] [Accepted: 03/09/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives Urinary stem cells (USCs) have the capacity for unlimited growth and are promising tools for the investigations of cell differentiation and urinary regeneration. However, the limited life span significantly restricts their usefulness. This study is aimed at exploring the effect of integrin-linked kinase (ILK) on the smooth muscle cells (SMCs) differentiation of the dog USCs and investigating its molecular mechanism. Methods An immortalized USCs cell line with the molecular markers and biological functions was prepared. After successfully inducing the differentiation of USCs into SMCs, the expression level of the unique key factor and its mechanisms in this process was determined through real-time polymerase chain reaction, Western blot, or Immunofluorescence staining. Results We found that high cell density promoted USCs differentiation SMCs, and ILK was necessary for USCs differentiation into SMCs. Knocking down ILK decreased the expression of SMCs specific-marker, while using a selective ILK agonist increased the expression of SMCs specific-marker. Furthermore, ILK regulated SMCs differentiation in part through the activation of NF-κB pathway in USCs. A NF-κB activity assay showed overexpression of ILK could significantly upregulate NF-κB p50 expression, and NF-κB p50 acts as downstream signal molecular of ILK. Conclusion High cell density induces the differentiation of USCs into SMCs, and ILK is a key regulator of myogenesis. Furthermore, NF-κB signaling pathway might play a crucial role in this process.
Collapse
|
31
|
Wang S, Hashemi S, Stratton S, Arinzeh TL. The Effect of Physical Cues of Biomaterial Scaffolds on Stem Cell Behavior. Adv Healthc Mater 2021; 10:e2001244. [PMID: 33274860 DOI: 10.1002/adhm.202001244] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/09/2020] [Indexed: 02/06/2023]
Abstract
Stem cells have been sought as a promising cell source in the tissue engineering field due to their proliferative capacity as well as differentiation potential. Biomaterials have been utilized to facilitate the delivery of stem cells in order to improve their engraftment and long-term viability upon implantation. Biomaterials also have been developed as scaffolds to promote stem cell induced tissue regeneration. This review focuses on the latter where the biomaterial scaffold is designed to provide physical cues to stem cells in order to promote their behavior for tissue formation. Recent work that explores the effect of scaffold physical properties, topography, mechanical properties and electrical properties, is discussed. Although still being elucidated, the biological mechanisms, including cell shape, focal adhesion distribution, and nuclear shape, are presented. This review also discusses emerging areas and challenges in clinical translation.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | - Sharareh Hashemi
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | - Scott Stratton
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | | |
Collapse
|
32
|
Transcriptional activators YAP/TAZ and AXL orchestrate dedifferentiation, cell fate, and metastasis in human osteosarcoma. Cancer Gene Ther 2021; 28:1325-1338. [PMID: 33408328 PMCID: PMC8636268 DOI: 10.1038/s41417-020-00281-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/19/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022]
Abstract
Osteosarcoma (OS) is a molecularly heterogeneous, aggressive, poorly differentiated pediatric bone cancer that frequently spreads to the lung. Relatively little is known about phenotypic and epigenetic changes that promote lung metastases. To identify key drivers of metastasis, we studied human CCH-OS-D OS cells within a previously described rat acellular lung (ACL) model that preserves the native lung architecture, extracellular matrix, and capillary network. This system identified a subset of cells—termed derived circulating tumor cells (dCTCs)—that can migrate, intravasate, and spread within a bioreactor-perfused capillary network. Remarkably, dCTCs highly expressed epithelial-to-mesenchymal transition (EMT)-associated transcription factors (EMT-TFs), such as ZEB1, TWIST, and SOX9, which suggests that they undergo cellular reprogramming toward a less differentiated state by coopting the same epigenetic machinery used by carcinomas. Since YAP/TAZ and AXL tightly regulate the fate and plasticity of normal mesenchymal cells in response to microenvironmental cues, we explored whether these proteins contributed to OS metastatic potential using an isogenic pair of human OS cell lines that differ in AXL expression. We show that AXL inhibition significantly reduced the number of MG63.2 pulmonary metastases in murine models. Collectively, we present a laboratory-based method to detect and characterize a pure population of dCTCs, which provides a unique opportunity to study how OS cell fate and differentiation contributes to metastatic potential. Though the important step of clinical validation remains, our identification of AXL, ZEB1, and TWIST upregulation raises the tantalizing prospect that EMT-TF-directed therapies might expand the arsenal of therapies used to combat advanced-stage OS.
Collapse
|
33
|
Thompson CL, Fu S, Knight MM, Thorpe SD. Mechanical Stimulation: A Crucial Element of Organ-on-Chip Models. Front Bioeng Biotechnol 2020; 8:602646. [PMID: 33363131 PMCID: PMC7758201 DOI: 10.3389/fbioe.2020.602646] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Organ-on-chip (OOC) systems recapitulate key biological processes and responses in vitro exhibited by cells, tissues, and organs in vivo. Accordingly, these models of both health and disease hold great promise for improving fundamental research, drug development, personalized medicine, and testing of pharmaceuticals, food substances, pollutants etc. Cells within the body are exposed to biomechanical stimuli, the nature of which is tissue specific and may change with disease or injury. These biomechanical stimuli regulate cell behavior and can amplify, annul, or even reverse the response to a given biochemical cue or drug candidate. As such, the application of an appropriate physiological or pathological biomechanical environment is essential for the successful recapitulation of in vivo behavior in OOC models. Here we review the current range of commercially available OOC platforms which incorporate active biomechanical stimulation. We highlight recent findings demonstrating the importance of including mechanical stimuli in models used for drug development and outline emerging factors which regulate the cellular response to the biomechanical environment. We explore the incorporation of mechanical stimuli in different organ models and identify areas where further research and development is required. Challenges associated with the integration of mechanics alongside other OOC requirements including scaling to increase throughput and diagnostic imaging are discussed. In summary, compelling evidence demonstrates that the incorporation of biomechanical stimuli in these OOC or microphysiological systems is key to fully replicating in vivo physiology in health and disease.
Collapse
Affiliation(s)
- Clare L Thompson
- Centre for Predictive in vitro Models, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Su Fu
- Centre for Predictive in vitro Models, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Martin M Knight
- Centre for Predictive in vitro Models, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Stephen D Thorpe
- UCD School of Medicine, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
34
|
Ejeian F, Razmjou A, Nasr-Esfahani MH, Mohammad M, Karamali F, Ebrahimi Warkiani M, Asadnia M, Chen V. ZIF-8 Modified Polypropylene Membrane: A Biomimetic Cell Culture Platform with a View to the Improvement of Guided Bone Regeneration. Int J Nanomedicine 2020; 15:10029-10043. [PMID: 33335393 PMCID: PMC7737945 DOI: 10.2147/ijn.s269169] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Despite the significant advances in modeling of biomechanical aspects of cell microenvironment, it remains a major challenge to precisely mimic the physiological condition of the particular cell niche. Here, the metal-organic frameworks (MOFs) have been introduced as a feasible platform for multifactorial control of cell-substrate interaction, given the wide range of physical and mechanical properties of MOF materials and their structural flexibility. RESULTS In situ crystallization of zeolitic imidazolate framework-8 (ZIF-8) on the polydopamine (PDA)-modified membrane significantly raised surface energy, wettability, roughness, and stiffness of the substrate. This modulation led to an almost twofold increment in the primary attachment of dental pulp stem cells (DPSCs) compare to conventional plastic culture dishes. The findings indicate that polypropylene (PP) membrane modified by PDA/ZIF-8 coating effectively supports the growth and proliferation of DPSCs at a substantial rate. Further analysis also displayed the exaggerated multilineage differentiation of DPSCs with amplified level of autocrine cell fate determination signals, like BSP1, BMP2, PPARG, FABP4, ACAN, and COL2A. Notably, osteogenic markers were dramatically overexpressed (more than 100-folds rather than tissue culture plate) in response to biomechanical characteristics of the ZIF-8 layer. CONCLUSION Hence, surface modification of cell culture platforms with MOF nanostructures proposed as a powerful nanomedical approach for selectively guiding stem cells for tissue regeneration. In particular, PP/PDA/ZIF-8 membrane presented ideal characteristics for using as a barrier membrane for guided bone regeneration (GBR) in periodontal tissue engineering.
Collapse
Affiliation(s)
- Fatemeh Ejeian
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan73441-81746, Iran
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Amir Razmjou
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan73441-81746, Iran
- UNESCO Center for Membrane Technology, School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Munirah Mohammad
- UNESCO Center for Membrane Technology, School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Fereshteh Karamali
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Mohsen Asadnia
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Vicki Chen
- School of Chemical Engineering, University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
35
|
Fearing BV, Speer JE, Jing L, Kalathil A, P. Kelly M, M. Buchowski J, P. Zebala L, Luhmann S, C. Gupta M, A. Setton L. Verteporfin treatment controls morphology, phenotype, and global gene expression for cells of the human nucleus pulposus. JOR Spine 2020; 3:e1111. [PMID: 33392449 PMCID: PMC7770208 DOI: 10.1002/jsp2.1111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Cells of the nucleus pulposus (NP) are essential contributors to extracellular matrix synthesis and function of the intervertebral disc. With age and degeneration, the NP becomes stiffer and more dehydrated, which is associated with a loss of phenotype and biosynthetic function for its resident NP cells. Also, with aging, the NP cell undergoes substantial morphological changes from a rounded shape with pronounced vacuoles in the neonate and juvenile, to one that is more flattened and spread with a loss of vacuoles. Here, we make use of the clinically relevant pharmacological treatment verteporfin (VP), previously identified as a disruptor of yes-associated protein-TEA domain family member-binding domain (TEAD) signaling, to promote morphological changes in adult human NP cells in order to study variations in gene expression related to differences in cell shape. Treatment of adult, degenerative human NP cells with VP caused a shift in morphology from a spread, fibroblastic-like shape to a rounded, clustered morphology with decreased transcriptional activity of TEAD and serum-response factor. These changes were accompanied by an increased expression of vacuoles, NP-specific gene markers, and biosynthetic activity. The contemporaneous observation of VP-induced changes in cell shape and prominent, time-dependent changes within the transcriptome of NP cells occurred over all timepoints in culture. Enriched gene sets with the transition to VP-induced cell rounding suggest a major role for cell adhesion, cytoskeletal remodeling, vacuolar lumen, and MAPK activity in the NP phenotypic and functional response to changes in cell shape.
Collapse
Affiliation(s)
- Bailey V. Fearing
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
- Department of Orthopaedic SurgeryAtrium Health Musculoskeletal InstituteCharlotteNorth CarolinaUSA
| | - Julie E. Speer
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Liufang Jing
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Aravind Kalathil
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Michael P. Kelly
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Jacob M. Buchowski
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Lukas P. Zebala
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Scott Luhmann
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Munish C. Gupta
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Lori A. Setton
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| |
Collapse
|
36
|
Wu KC, Weng HK, Hsu YS, Huang PJ, Wang YK. Aqueous extract of Arctium lappa L. root (burdock) enhances chondrogenesis in human bone marrow-derived mesenchymal stem cells. BMC Complement Med Ther 2020; 20:364. [PMID: 33228629 PMCID: PMC7686739 DOI: 10.1186/s12906-020-03158-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 11/13/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Arctium lappa L. root (burdock root) has long been recommended for the treatment of different diseases in traditional Chinese medicine. Burdock root possesses anti-oxidative, anti-inflammatory, anti-cancer, and anti-microbial activities. The aim of the study was to elucidate whether aqueous extract of burdock root regulates mesenchymal stem cell proliferation and differentiation. METHODS Human bone marrow-derived mesenchymal stem cells in 2D high density culture and in 3D micromass pellets were treated with chondrogenic induction medium and chondral basal medium in the absence or presence of aqueous extract of burdock root. The chondrogenic differentiation was accessed by staining glucosaminoglycans, immunostaining SOX9 and type II collagen and immuonblotting of SOX9, aggrecan and type II collagen. RESULTS Treatment of aqueous extract of burdock root increased the cell proliferation of hMSCs. It did not have significant effect on osteogenic and adipogenic differentiation, but significantly enhanced chondrogenic induction medium-induced chondrogenesis. The increment was dose dependent, as examined by staining glucosaminoglycans, SOX9, and type II collagen and immunobloting of SOX9, aggrecan and type II collagen in 2D and 3D cultures. In the presence of supplemental materials, burdock root aqueous extract showed equivalent chondrogenic induction capability to that of TGF-β. CONCLUSIONS The results demonstrate that aqueous extract of Arctium lappa L. root promotes chondrogenic medium-induced chondrogenic differentiation. The aqueous extract of burdock root can even be used alone to stimulate chondrogenic differentiation. The study suggests that the aqueous extract of burdock root can be used as an alternative strategy for treatment purposes.
Collapse
Affiliation(s)
- King-Chuen Wu
- Department of Nursing, Chang Gung University of Science and Technology, Chia-Yi County, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Chiayi County, Taiwan
| | - Hung-Kai Weng
- Department of Orthopaedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Yun-Shang Hsu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Pin-Jia Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Yang-Kao Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan. .,Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan City, Taiwan.
| |
Collapse
|
37
|
Sun T, Shi Q, Liang Q, Yao Y, Wang H, Sun J, Huang Q, Fukuda T. Fabrication of vascular smooth muscle-like tissues based on self-organization of circumferentially aligned cells in microengineered hydrogels. LAB ON A CHIP 2020; 20:3120-3131. [PMID: 32756693 DOI: 10.1039/d0lc00544d] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Circumferential alignment of vascular smooth muscle cells (vSMCs) is critical to form an in vivo-like vascular smooth muscle layer in vitro. Although many techniques to elicit such an alignment on 3D substrates have been demonstrated, it remains a challenge to recapitulate the circumferential cellular alignment of vascular smooth muscle tissues in 3D hydrogels. Here, we propose a spring-like gelatin methacrylate (GelMA) structure formed by semi-automated reeling of a core-shell microfiber at the micro-scale. The resulting structures facilitate circumferential alignment and self-organization of encapsulated human mesenchymal stem cells (MSCs) into multilayer spring-like cellular structures. Based on the permeable tubular lumens of these structures, a perfusion culture micro-system is developed to further facilitate the vSMC differentiation of MSCs under the effect of TGF-β1. We also evaluated the MSC contraction-induced shrinkage of the resulting cellular structures. These results demonstrate the successful in vitro regeneration of vascular smooth muscle (vSM)-like tissues in 3D environments. Compared with the substrate surface, the porous structure in hydrogels is more similar to cell microenvironments in vivo. Thus, this approach may be used to develop an in vitro model for the study of vascular tissue regeneration and the mechanism of vascular remolding during hypertension.
Collapse
Affiliation(s)
- Tao Sun
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, 100081, People's Republic of China.
| | - Qing Shi
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, 100081, People's Republic of China. and Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, 100081, People's Republic of China
| | - Qian Liang
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, 100081, People's Republic of China
| | - Yibing Yao
- Department of Geriatric Oncology, Fourth Medical Center of PLA General Hospital, 100048, People's Republic of China
| | - Huaping Wang
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, 100081, People's Republic of China
| | - Junzhong Sun
- Department of Geriatric Oncology, Fourth Medical Center of PLA General Hospital, 100048, People's Republic of China
| | - Qiang Huang
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, 100081, People's Republic of China
| | - Toshio Fukuda
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, 100081, People's Republic of China.
| |
Collapse
|
38
|
Domingues C, Geraldo AM, Anjo SI, Matos A, Almeida C, Caramelo I, Lopes-da-Silva JA, Paiva A, Carvalho J, Pires das Neves R, Manadas B, Grãos M. Cofilin-1 Is a Mechanosensitive Regulator of Transcription. Front Cell Dev Biol 2020; 8:678. [PMID: 32903827 PMCID: PMC7438942 DOI: 10.3389/fcell.2020.00678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanical properties of the extracellular environment are interrogated by cells and integrated through mechanotransduction. Many cellular processes depend on actomyosin-dependent contractility, which is influenced by the microenvironment’s stiffness. Here, we explored the influence of substrate stiffness on the proteome of proliferating undifferentiated human umbilical cord-matrix mesenchymal stem/stromal cells. The relative abundance of several proteins changed significantly by expanding cells on soft (∼3 kPa) or stiff substrates (GPa). Many such proteins are associated with the regulation of the actin cytoskeleton, a major player of mechanotransduction and cell physiology in response to mechanical cues. Specifically, Cofilin-1 levels were elevated in cells cultured on soft comparing with stiff substrates. Furthermore, Cofilin-1 was de-phosphorylated (active) and present in the nuclei of cells kept on soft substrates, in contrast with phosphorylated (inactive) and widespread distribution in cells on stiff. Soft substrates promoted Cofilin-1-dependent increased RNA transcription and faster RNA polymerase II-mediated transcription elongation. Cofilin-1 is part of a novel mechanism linking mechanotransduction and transcription.
Collapse
Affiliation(s)
- Catarina Domingues
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - A Margarida Geraldo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Sandra Isabel Anjo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - André Matos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Polytechnic Institute of Coimbra, Coimbra College of Agriculture, Coimbra, Portugal
| | - Cláudio Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Polytechnic Institute of Coimbra, Coimbra College of Agriculture, Coimbra, Portugal
| | - Inês Caramelo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | | | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Coimbra, Portugal
| | - João Carvalho
- Centro de Física da Universidade de Coimbra (CFisUC), Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Ricardo Pires das Neves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - Bruno Manadas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Mário Grãos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal.,Biocant, Technology Transfer Association, Cantanhede, Portugal
| |
Collapse
|
39
|
Selig M, Lauer JC, Hart ML, Rolauffs B. Mechanotransduction and Stiffness-Sensing: Mechanisms and Opportunities to Control Multiple Molecular Aspects of Cell Phenotype as a Design Cornerstone of Cell-Instructive Biomaterials for Articular Cartilage Repair. Int J Mol Sci 2020; 21:E5399. [PMID: 32751354 PMCID: PMC7432012 DOI: 10.3390/ijms21155399] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/23/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Since material stiffness controls many cell functions, we reviewed the currently available knowledge on stiffness sensing and elucidated what is known in the context of clinical and experimental articular cartilage (AC) repair. Remarkably, no stiffness information on the various biomaterials for clinical AC repair was accessible. Using mRNA expression profiles and morphology as surrogate markers of stiffness-related effects, we deduced that the various clinically available biomaterials control chondrocyte (CH) phenotype well, but not to equal extents, and only in non-degenerative settings. Ample evidence demonstrates that multiple molecular aspects of CH and mesenchymal stromal cell (MSC) phenotype are susceptible to material stiffness, because proliferation, migration, lineage determination, shape, cytoskeletal properties, expression profiles, cell surface receptor composition, integrin subunit expression, and nuclear shape and composition of CHs and/or MSCs are stiffness-regulated. Moreover, material stiffness modulates MSC immuno-modulatory and angiogenic properties, transforming growth factor beta 1 (TGF-β1)-induced lineage determination, and CH re-differentiation/de-differentiation, collagen type II fragment production, and TGF-β1- and interleukin 1 beta (IL-1β)-induced changes in cell stiffness and traction force. We then integrated the available molecular signaling data into a stiffness-regulated CH phenotype model. Overall, we recommend using material stiffness for controlling cell phenotype, as this would be a promising design cornerstone for novel future-oriented, cell-instructive biomaterials for clinical high-quality AC repair tissue.
Collapse
Affiliation(s)
- Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Jasmin C. Lauer
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Melanie L. Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| |
Collapse
|
40
|
Jiang D, Scharffetter-Kochanek K. Mesenchymal Stem Cells Adaptively Respond to Environmental Cues Thereby Improving Granulation Tissue Formation and Wound Healing. Front Cell Dev Biol 2020; 8:697. [PMID: 32850818 PMCID: PMC7403200 DOI: 10.3389/fcell.2020.00697] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
Granulation tissue formation constitutes a key step during wound healing of the skin and other organs. Granulation tissue concomitantly initiates regenerative M2 macrophages polarization, fibroblast proliferation, myofibroblast differentiation with subsequent contraction of the wound, new vessel formation, and matrix deposition. Impaired granulation tissue formation either leads to delayed wound healing or excessive scar formation, conditions with high morbidity and mortality. Accumulating evidence has demonstrated that mesenchymal stem cell (MSC)-based therapy is a promising strategy to ameliorate defects in granulation tissue formation and to successfully treat non-healing chronic wounds. In this review we give an updated overview of how therapeutically administered MSCs ensure a balanced granulation tissue formation, and furthermore discuss the cellular and molecular mechanisms underlying the adaptive responses of MSCs to cue in their direct neighborhood. Improved understanding of the interplay between the exogenous MSCs and their niche in granulation tissue will foster the development of MSC-based therapies tailored for difficult-to-treat non-healing wounds.
Collapse
Affiliation(s)
- Dongsheng Jiang
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
| | | |
Collapse
|
41
|
Frassica MT, Grunlan MA. Perspectives on Synthetic Materials to Guide Tissue Regeneration for Osteochondral Defect Repair. ACS Biomater Sci Eng 2020; 6:4324-4336. [PMID: 33455185 DOI: 10.1021/acsbiomaterials.0c00753] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regenerative engineering holds the potential to treat clinically pervasive osteochondral defects (OCDs). In a synthetic materials-guided approach, the scaffold's chemical and physical properties alone instruct cellular behavior in order to effect regeneration, referred to herein as "instructive" properties. While this alleviates the costs and off-target risks associated with exogenous growth factors, the scaffold must be potently instructive to achieve tissue growth. Moreover, toward achieving functionality, such a scaffold should also recapitulate the spatial complexity of the osteochondral tissues. Thus, in addition to the regeneration of the articular cartilage and underlying cancellous bone, the complex osteochondral interface, composed of calcified cartilage and subchondral bone, should also be restored. In this Perspective, we highlight recent synthetic-based, instructive osteochondral scaffolds that have leveraged new material chemistries as well as innovative fabrication strategies. In particular, scaffolds with spatially complex chemical and morphological features have been prepared with electrospinning, solvent-casting-particulate-leaching, freeze-drying, and additive manufacturing. While few synthetic scaffolds have advanced to clinical studies to treat OCDs, these recent efforts point to the promising use of the chemical and physical properties of synthetic materials for regeneration of osteochondral tissues.
Collapse
Affiliation(s)
- Michael T Frassica
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-2120, United States
| | - Melissa A Grunlan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-2120, United States.,Department of Materials Science & Engineering, Texas A&M University, College Station, Texas 77843-3003, United States.,Department of Chemistry, Texas A&M University, College Station, Texas 77843-3255, United States
| |
Collapse
|
42
|
Wang Y, Xiao Y, Long S, Fan Y, Zhang X. Role of N-Cadherin in a Niche-Mimicking Microenvironment for Chondrogenesis of Mesenchymal Stem Cells In Vitro. ACS Biomater Sci Eng 2020; 6:3491-3501. [PMID: 33463167 DOI: 10.1021/acsbiomaterials.0c00149] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
During the development of natural cartilage, mesenchymal condensation is the starting event of chondrogenesis, and mesenchymal stem cells (MSCs) experienced a microenvironment transition from primarily cell-cell interactions to a later stage, where cell-extracellular matrix (ECM) interactions dominate. Although micromass pellet culture has been developed to mimic mesenchymal condensation in vitro, the molecular mechanism remains elusive, and the transition from cell-cell to cell-ECM interactions has been poorly recapitulated. In this study, we first constructed MSC microspheres (MMs) and investigated their chondrogenic differentiation with functional blocking of N-cadherin. The results showed that early cartilage differentiation and cartilage-specific matrix deposition of MSCs in the group with the N-cadherin antibody were significantly postponed. Next, poly(l-lysine) treatment was transiently applied to promote the expression of N-cadherin gene, CDH2, and the treatment-promoted MSC chondrogenesis. Upon one-day culture in MMs with established cell-cell adhesions, collagen hydrogel-encapsulated MMs (CMMs) were constructed to simulate the cell-ECM interactions, and the collagen microenvironment compensated the inhibitory effects from N-cadherin blocking. Surprisingly, chondrogenic-differentiated cell migration, which has important implications in cartilage repair and integration, was found in the CMMs without N-cadherin blocking. In conclusion, our study demonstrated that N-cadherin plays the critical role in early mesenchymal condensation, and the collagen hydrogel provides a supportive microenvironment for late chondrogenic differentiation. Therefore, sequential presentations of cell-cell adhesion and cell-ECM interaction in an engineered microenvironment seem to be a promising strategy to facilitate MSC chondrogenic differentiation.
Collapse
Affiliation(s)
- Yonghui Wang
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning 530021, China
| | - Yun Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Shihe Long
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning 530021, China.,National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
43
|
Begum R, Perriman AW, Su B, Scarpa F, Kafienah W. Chondroinduction of Mesenchymal Stem Cells on Cellulose-Silk Composite Nanofibrous Substrates: The Role of Substrate Elasticity. Front Bioeng Biotechnol 2020; 8:197. [PMID: 32266231 PMCID: PMC7096586 DOI: 10.3389/fbioe.2020.00197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 02/28/2020] [Indexed: 01/09/2023] Open
Abstract
Smart biomaterials with an inherent capacity to elicit specific behaviors in lieu of biological prompts would be advantageous for regenerative medicine applications. In this work, we employ an electrospinning technique to model the in vivo nanofibrous extracellular matrix (ECM) of cartilage using a chondroinductive cellulose and silk polymer blend (75:25 ratio). This natural polymer composite is directly electrospun for the first time, into nanofibers without post-spun treatment, using a trifluoroacetic acid and acetic acid cosolvent system. Biocompatibility of the composite nanofibres with human mesenchymal stem cells (hMSCs) is demonstrated and its inherent capacity to direct chondrogenic stem cell differentiation, in the absence of stimulating growth factors, is confirmed. This chondrogenic stimulation could be countered biochemically using fibroblast growth factor-2, a growth factor used to enhance the proliferation of hMSCs. Furthermore, the potential mechanisms driving this chondroinduction at the cell-biomaterial interface is investigated. Composite substrates are fabricated as two-dimensional film surfaces and cultured with hMSCs in the presence of chemicals that interfere with their biochemical and mechanical signaling pathways. Preventing substrate surface elasticity transmission resulted in a significant downregulation of chondrogenic gene expression. Interference with the classical chondrogenic Smad2/3 phosphorylation pathway did not impact chondrogenesis. The results highlight the importance of substrate mechanical elasticity on hMSCs chondroinduction and its independence to known chondrogenic biochemical pathways. The newly fabricated scaffolds provide the foundation for designing a robust, self-inductive, and cost-effective biomimetic biomaterial for cartilage tissue engineering.
Collapse
Affiliation(s)
- Runa Begum
- Faculty of Biomedical Sciences, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Adam W Perriman
- Faculty of Biomedical Sciences, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Bo Su
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Fabrizio Scarpa
- Bristol Composites Institute (ACCIS), University of Bristol, Bristol, United Kingdom
| | - Wael Kafienah
- Faculty of Biomedical Sciences, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
44
|
Lee H, Ahn J, Jung C, Jeung Y, Cho H, Son MJ, Chung K. Optimization of 3D hydrogel microenvironment for enhanced hepatic functionality of primary human hepatocytes. Biotechnol Bioeng 2020; 117:1864-1876. [DOI: 10.1002/bit.27328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/03/2020] [Accepted: 03/07/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Ho‐Joon Lee
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
| | - Jiwon Ahn
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
| | - Cho‐Rock Jung
- Gene Therapy UnitKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
- Department of Functional GenomicsKorea University of Science and Technology (UST) Daejeon Republic of Korea
| | - Yun‐Ji Jeung
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
| | - Hyun‐Soo Cho
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
| | - Myung Jin Son
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
- Department of Functional GenomicsKorea University of Science and Technology (UST) Daejeon Republic of Korea
| | - Kyung‐Sook Chung
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
- Department of Functional GenomicsKorea University of Science and Technology (UST) Daejeon Republic of Korea
- Biomedical Translational Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB) Daejeon Republic of Korea
| |
Collapse
|
45
|
Abstract
Tendons connect muscles to bones to transfer the forces necessary for movement. Cell-cell junction proteins, cadherins and connexins, may play a role in tendon development and injury. In this review, we begin by highlighting current understanding of how cell-cell junctions may regulate embryonic tendon development and differentiation. We then examine cell-cell junctions in postnatal tendon, before summarizing the role of cadherins and connexins in adult tendons. More information exists regarding the role of cell-cell junctions in the formation and homeostasis of other musculoskeletal tissues, namely cartilage and bone. Therefore, to inform future tendon studies, we include a brief survey of cadherins and connexins in chondrogenesis and osteogenesis, and summarize how cell-cell junctions are involved in some musculoskeletal tissue pathologies. An enhanced understanding of how cell-cell junctions participate in tendon development, maintenance, and disease will benefit future regenerative strategies.
Collapse
Affiliation(s)
| | - Jett B Murray
- Biological Engineering, University of Idaho, Moscow, ID
| | | |
Collapse
|
46
|
McColloch A, Rabiei M, Rabbani P, Bowling A, Cho M. Correlation between Nuclear Morphology and Adipogenic Differentiation: Application of a Combined Experimental and Computational Modeling Approach. Sci Rep 2019; 9:16381. [PMID: 31705037 PMCID: PMC6842088 DOI: 10.1038/s41598-019-52926-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 10/22/2019] [Indexed: 12/18/2022] Open
Abstract
Stem cells undergo drastic morphological alterations during differentiation. While extensive studies have been performed to examine the cytoskeletal remodeling, there is a growing interest to determine the morphological, structural and functional changes of the nucleus. The current study is therefore aimed at quantifying the extent of remodeling of the nuclear morphology of human mesenchymal stem cells during biochemically-induced adipogenic differentiation. Results show the size of nuclei decreased exponentially over time as the lipid accumulation is up-regulated. Increases in the lipid accumulation appear to lag the nuclear reorganization, suggesting the nuclear deformation is a prerequisite to adipocyte maturation. Furthermore, the lamin A/C expression was increased and redistributed to the nuclear periphery along with a subsequent increase in the nuclear aspect ratio. To further assess the role of the nucleus, a nuclear morphology with a high aspect ratio was achieved using microcontact-printed substrate. The cells with an elongated nuclear shape did not efficiently undergo adipogenesis, suggesting the cellular and nuclear processes associated with stem cell differentiation at the early stage of adipogenesis cause a change in the nuclear morphology and cannot be abrogated by the morphological cues. In addition, a novel computational biomechanical model was generated to simulate the nuclear shape change during differentiation and predict the forces acting upon the nucleus. This effort led to the development of computational scaling approach to simulate the experimentally observed adipogenic differentiation processes over 15 days in less than 1.5 hours.
Collapse
Affiliation(s)
- Andrew McColloch
- University of Texas at Arlington, Department of Biomedical Engineering, Arlington, 76010, USA
| | - Manoochehr Rabiei
- University of Texas at Arlington, Department of Mechanical and Aerospace Engineering, Arlington, TX, 76010, USA
| | - Parisa Rabbani
- University of Texas at Arlington, Department of Biomedical Engineering, Arlington, 76010, USA
| | - Alan Bowling
- University of Texas at Arlington, Department of Mechanical and Aerospace Engineering, Arlington, TX, 76010, USA
| | - Michael Cho
- University of Texas at Arlington, Department of Biomedical Engineering, Arlington, 76010, USA.
| |
Collapse
|
47
|
Zhu Y, Liu X, Wu J, Wong TM, Feng X, Yang C, Wu S, Zheng Y, Liu X, Cheung KMC, Yeung KWK. Micro- and Nanohemispherical 3D Imprints Modulate the Osteogenic Differentiation and Mineralization Tendency of Bone Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:35513-35524. [PMID: 31507175 DOI: 10.1021/acsami.9b05521] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Surface topography has been reported to play a key role in modulating cell behaviors, yet the mechanism through which it modulates these behaviors is not fully understood, especially in the case of three-dimensional (3D) topographies. In this study, a series of novel hemispherical 3D imprints ranging from the nanoscale to the microscale were prepared on titanium (Ti) surfaces using a customized interfacial lithography method. Mouse embryo osteoblast precursor cells (MC3T3-E1) were selected to investigate the solitary effect of specific hemispherical 3D imprints on cellular behaviors. The results indicated that varied hemispherical 3D imprints can affect the formation of filopodia and the arrangement of the cytoskeleton in different ways. Specifically, they can alter the spreading morphologies of cells and lead to deformation of the nucleus, which eventually affects cell proliferation and osteogenic differentiation. Cells cultured on different hemispherical 3D imprints exhibited promoted proliferation and osteogenic differentiation to different degrees; for example, cells cultured on 90 and 500 nm hemispherical imprints formed abundant filopodia and exhibited the highest alkaline phosphatase activity and osteogenic gene expression, respectively. Four-week tibia implantation also confirmed that 90 nm hemispherical imprints improved the osteogenic ability in vivo compared with an unpatterned Ti substrate. In addition to promoted proliferation, colonization of more cells on the surface of implants and induction of rapid osteogenic differentiation can occur. Our work provides a rational way to balance cell proliferation and differentiation, which can accelerate bone integration of an implant and host tissue.
Collapse
Affiliation(s)
- Yizhou Zhu
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine , The University of Hong Kong , Pokfulam, Hong Kong 999077 , China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology , The University of Hong Kong-Shenzhen Hospital , Shenzhen 518053 , China
| | - Xiangmei Liu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering , Hubei University , Wuhan 430062 , China
| | - Jun Wu
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology , The University of Hong Kong-Shenzhen Hospital , Shenzhen 518053 , China
| | - Tak Man Wong
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine , The University of Hong Kong , Pokfulam, Hong Kong 999077 , China
| | - Xiaobo Feng
- Department of Orthopaedics, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China
| | - Shuilin Wu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering , Hubei University , Wuhan 430062 , China
- School of Materials Science & Engineering , Tianjin University , Tianjin 300350 , China
| | - Yufeng Zheng
- State Key Laboratory for Turbulence and Complex System and Department of Materials Science and Engineering, College of Engineering , Peking University , Beijing 100871 , China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics , Chinese Academy of Sciences , Shanghai 200050 , China
| | - Kenneth M C Cheung
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine , The University of Hong Kong , Pokfulam, Hong Kong 999077 , China
| | - Kelvin W K Yeung
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine , The University of Hong Kong , Pokfulam, Hong Kong 999077 , China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology , The University of Hong Kong-Shenzhen Hospital , Shenzhen 518053 , China
- China Orthopedic Regenerative Medicine Group (CORMed) , Hangzhou 310058 , China
| |
Collapse
|
48
|
Viscoelasticity in natural tissues and engineered scaffolds for tissue reconstruction. Acta Biomater 2019; 97:74-92. [PMID: 31400521 DOI: 10.1016/j.actbio.2019.08.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/23/2019] [Accepted: 08/06/2019] [Indexed: 02/05/2023]
Abstract
Viscoelasticity of living tissues plays a critical role in tissue homeostasis and regeneration, and its implication in disease development and progression is being recognized recently. In this review, we first explored the state of knowledge regarding the potential application of tissue viscoelasticity in disease diagnosis. In order to better characterize viscoelasticity with local resolution and non-invasiveness, emerging characterization methods have been developed with the potential to be supplemented to existing facilities. To understand cellular responses to matrix viscoelastic behaviors in vitro, hydrogels made of natural polymers have been developed and the relationships between their molecular structure and viscoelastic behaviors, are elucidated. Moreover, how cells perceive the viscoelastic microenvironment and cellular responses including cell attachment, spreading, proliferation, differentiation and matrix production, have been discussed. Finally, some future perspective on an integrated mechanobiological comprehension of the viscoelastic behaviors involved in tissue homeostasis, cellular responses and biomaterial design are highlighted. STATEMENT OF SIGNIFICANCE: Tissue- or organ-scale viscoelastic behavior is critical for homeostasis, and the molecular basis and cellular responses of viscoelastic materials at micro- or nano-scale are being recognized recently. We summarized the potential applications of viscoelasticity in disease diagnosis enabled by emerging non-invasive characterization technologies, and discussed the underlying mechanism of viscoelasticity of hydrogels and current understandings of cell regulatory functions of them. With a growing understanding of the molecular basis of hydrogel viscoelasticity and recognition of its regulatory functions on cell behaviors, it is important to bring the clinical insights on how these characterization technologies and engineered materials may contribute to disease diagnosis and treatment. This review explains the basics in characterizing viscoelasticity with our hope to bridge the gap between basic research and clinical applications.
Collapse
|
49
|
Gegg C, Yang F. The Effects of ROCK Inhibition on Mesenchymal Stem Cell Chondrogenesis Are Culture Model Dependent. Tissue Eng Part A 2019; 26:130-139. [PMID: 31411113 DOI: 10.1089/ten.tea.2019.0068] [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] [Indexed: 12/13/2022] Open
Abstract
Rho-associated protein kinase (ROCK) signaling correlates with cell shape, with decreased cell spreading accompanied by decreased ROCK activity. However, how cell shape and ROCK activity impact the chondrogenesis of mesenchymal stem cells (MSCs) remains inconclusive. Here we examine the effects of ROCK inhibition on human MSC chondrogenesis in four different culture models, including three-dimensional (3D) microribbon (μRB) scaffolds, two-dimensional hydrogel (2D-HG) substrates, 3D hydrogels (3D-HGs), and pellet. For each culture model involving biomaterials, four polymers were compared, including gelatin, chondroitin sulfate, hyaluronic acid, and polyethylene glycol. ROCK inhibition decreased MSC chondrogenesis in μRB model, enhanced chondrogenesis in pellet, and had minimal effect in 2D-HG or 3D-HG models. Furthermore, we demonstrate that MSC chondrogenesis cannot be predicted using ROCK signaling alone. While varying biomaterial compositions can impact the amount or phenotype of resulting cartilage, varying biomaterials did not change the chondrogenic response to ROCK inhibition within each culture model. Regardless of culture model or ROCK expression, increased cartilage formation was always accompanied by enhanced N-cadherin expression and production, suggesting that N-cadherin is a robust marker to select culture conditions that promote chondrogenesis. Together, the results from this study may be used to enhance MSC-based cartilage regeneration in different culture models. Impact Statement Here we assessed the effects of Rho-associated protein kinase (ROCK) inhibition on mesenchymal stem cell (MSC) chondrogenesis in different culture models, including three-dimensional (3D) microribbon scaffolds, two-dimensional hydrogel substrates, 3D hydrogels, and pellet culture. Our results demonstrate that effects of ROCK inhibition on MSC chondrogenesis differ substantially depending on culture models. Furthermore, MSC chondrogenesis cannot be predicted using ROCK signaling alone. The results from this study fill in a gap of knowledge in the correlation between ROCK signaling and MSC chondrogenesis, which may be used to enhance MSC-based cartilage regeneration in different culture models.
Collapse
Affiliation(s)
- Courtney Gegg
- Department of Bioengineering, Stanford University Schools of Engineering and Medicine, Stanford, California
| | - Fan Yang
- Department of Bioengineering and Orthopedic Surgery, Stanford University Schools of Engineering and Medicine, Stanford, California
| |
Collapse
|
50
|
Li YY, Lam KL, Chen AD, Zhang W, Chan BP. Collagen microencapsulation recapitulates mesenchymal condensation and potentiates chondrogenesis of human mesenchymal stem cells – A matrix-driven in vitro model of early skeletogenesis. Biomaterials 2019; 213:119210. [DOI: 10.1016/j.biomaterials.2019.05.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/28/2019] [Accepted: 05/10/2019] [Indexed: 01/01/2023]
|