1
|
Cifuentes SJ, Theran-Suarez NA, Rivera-Crespo C, Velez-Roman L, Thacker B, Glass C, Domenech M. Heparan Sulfate-Collagen Surface Multilayers Support Serum-Free Microcarrier Culture of Mesenchymal Stem Cells. ACS Biomater Sci Eng 2024; 10:5739-5751. [PMID: 39187752 DOI: 10.1021/acsbiomaterials.4c01008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The increasing cost of high-volume cultures and dependence on serum and growth factor supplementation limit the affordability of mesenchymal stromal cell (MSC) therapies. This has spurred interest in developing strategies that support adherent cell expansion while reducing raw material costs. Culture surfaces coated with sulfated glycosaminoglycans (GAGs), specifically heparan sulfate (HS), are an alternative to prolong growth factor retention in cell cultures. Unlike heparin, recombinant HS (rHS) offers strong binding affinity for multiple growth factors and extracellular matrix components, such as collagen I, without undesirable anticoagulant effects or xenobiotic health risks. The potential of rHS as a factor reservoir in MSC cultures remains underexplored. This study investigated the impact of rHS on the growth and anti-inflammatory properties of undifferentiated bone marrow MSCs in both planar and microcarrier-based cultures. It was hypothesized that rHS would enable MSC growth with minimal growth factor supplementation in a sulfation level-dependent manner. Cell culture surfaces were assembled via the layer-by-layer (LbL) method, combining alternating collagen I (COL) and rHS. These bilayers support cell adhesion and enable the incorporation of distinct sulfation levels on the culture surface. Examination of pro-mitogenic FGF and immunostimulatory IFN-γ release dynamics confirmed prolonged availability and sulfate level dependencies. Sulfated surfaces supported cell growth in low serum (2% FBS) and serum-free (SF) media at levels equivalent to standard culture conditions. Cell growth on rHS-coated surfaces in SF was comparable to that on heparin-coated surfaces and commercial surface-coated microcarriers in low serum. These growth benefits were observed in both planar and microcarrier (μCs) cultures. Additionally, rHS surfaces reduced β-galactosidase expression relative to uncoated surfaces, delaying cell senescence. Multivariate analysis of cytokines in conditioned media indicated that rHS-containing surfaces enhanced cytokine levels relative to uncoated surfaces during IFN-γ stimulation and correlated with decreased pro-inflammatory macrophage activity. Overall, utilizing highly sulfated rHS with COL reduces the need for exogenous growth factors and effectively supports MSC growth and anti-inflammatory potency on planar and microcarrier surfaces under minimal factor supplementation.
Collapse
Affiliation(s)
- Said J Cifuentes
- Bioengineering Graduate Program, University of Puerto Rico Mayaguez, Call Box 9000, Mayaguez, Puerto Rico 00681-9000, United States
- Bioengineering Department, Moffitt Cancer Center, Tampa, Florida 32611, United States
| | - Natalia A Theran-Suarez
- Chemical Engineering Department, University of Puerto Rico Mayaguez, 3550 General Atomics Ct, G02-102, Mayaguez, Puerto Rico 00681-9000, United States
| | - Carolina Rivera-Crespo
- Bioengineering Graduate Program, University of Puerto Rico Mayaguez, Call Box 9000, Mayaguez, Puerto Rico 00681-9000, United States
| | - Leonel Velez-Roman
- Bioengineering Graduate Program, University of Puerto Rico Mayaguez, Call Box 9000, Mayaguez, Puerto Rico 00681-9000, United States
| | - Bryan Thacker
- TEGA Therapeutics, Inc., 3550 General Atomics Ct, G02-102, San Diego, California 92121, United States
| | - Charles Glass
- TEGA Therapeutics, Inc., 3550 General Atomics Ct, G02-102, San Diego, California 92121, United States
| | - Maribella Domenech
- Bioengineering Graduate Program, University of Puerto Rico Mayaguez, Call Box 9000, Mayaguez, Puerto Rico 00681-9000, United States
- Chemical Engineering Department, University of Puerto Rico Mayaguez, 3550 General Atomics Ct, G02-102, Mayaguez, Puerto Rico 00681-9000, United States
| |
Collapse
|
2
|
Chen C, Zhan C, Huang X, Zhang S, Chen J. Three-dimensional printing of cell-laden bioink for blood vessel tissue engineering: influence of process parameters and components on cell viability. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:2411-2437. [PMID: 37725406 DOI: 10.1080/09205063.2023.2251781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/21/2023] [Indexed: 09/21/2023]
Abstract
Three-dimensional (3D) bioprinting is a potential therapeutic method for tissue engineering owing to its ability to prepare cell-laden tissue constructs. The properties of bioink are crucial to accurately control the printing structure. Meanwhile, the effect of process parameters on the precise structure is not nonsignificant. We investigated the correlation between process parameters of 3D bioprinting and the structural response of κ-carrageenan-based hydrogels to explore the controllable structure, printing resolution, and cell survival rate. Small-diameter (<6 mm) gel filaments with different structures were printed by varying the shear stress of the extrusion bioprinter to simulate the natural blood vessel structure. The cell viability of the scaffold was evaluated. The in vitro culture of human umbilical vein endothelium cells (HUVECs) on the κ-carrageenan (kc) and composite gels (carrageenan/carbon nanotube and carrageenan/sodium alginate) demonstrated that the cell attachment and proliferation on composite gels were better than those on pure kc. Our results revealed that the carrageenan-based composite bioinks offer better printability, sufficient mechanical stiffness, interconnectivity, and biocompatibility. This process can facilitate precise adjustment of the pore size, porosity, and pore distribution of the hydrogel structure by optimising the printing parameters as well as realise the precise preparation of the internal structure of the 3D hydrogel-based tissue engineering scaffold. Moreover, we obtained perfused tubular filament by 3D printing at optimal process parameters.
Collapse
Affiliation(s)
- Chongshuai Chen
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Congcong Zhan
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xia Huang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Shanfeng Zhang
- Experimental Center for Basic Medicine, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Junying Chen
- School of Chemical Engineering, Zhengzhou University, Zhengzhou, Henan, P.R. China
| |
Collapse
|
3
|
Nile M, Folwaczny M, Wichelhaus A, Baumert U, Janjic Rankovic M. Fluid flow shear stress and tissue remodeling-an orthodontic perspective: evidence synthesis and differential gene expression network analysis. Front Bioeng Biotechnol 2023; 11:1256825. [PMID: 37795174 PMCID: PMC10545883 DOI: 10.3389/fbioe.2023.1256825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/28/2023] [Indexed: 10/06/2023] Open
Abstract
Introduction: This study aimed to identify and analyze in vitro studies investigating the biological effect of fluid-flow shear stress (FSS) on cells found in the periodontal ligament and bone tissue. Method: We followed the PRISMA guideline for systematic reviews. A PubMed search strategy was developed, studies were selected according to predefined eligibility criteria, and the risk of bias was assessed. Relevant data related to cell source, applied FSS, and locus-specific expression were extracted. Based on this evidence synthesis and, as an original part of this work, analysis of differential gene expression using over-representation and network-analysis was performed. Five relevant publicly available gene expression datasets were analyzed using gene set enrichment analysis (GSEA). Result: A total of 6,974 articles were identified. Titles and abstracts were screened, and 218 articles were selected for full-text assessment. Finally, 120 articles were included in this study. Sample size determination and statistical analysis related to methodological quality and the ethical statement item in reporting quality were most frequently identified as high risk of bias. The analyzed studies mostly used custom-made fluid-flow apparatuses (61.7%). FSS was most frequently applied for 0.5 h, 1 h, or 2 h, whereas FSS magnitudes ranged from 6 to 20 dyn/cm2 depending on cell type and flow profile. Fluid-flow frequencies of 1 Hz in human cells and 1 and 5 Hz in mouse cells were mostly applied. FSS upregulated genes/metabolites responsible for tissue formation (AKT1, alkaline phosphatase, BGLAP, BMP2, Ca2+, COL1A1, CTNNB1, GJA1, MAPK1/MAPK3, PDPN, RUNX2, SPP1, TNFRSF11B, VEGFA, WNT3A) and inflammation (nitric oxide, PGE-2, PGI-2, PTGS1, PTGS2). Protein-protein interaction networks were constructed and analyzed using over-representation analysis and GSEA to identify shared signaling pathways. Conclusion: To our knowledge, this is the first review giving a comprehensive overview and discussion of methodological technical details regarding fluid flow application in 2D cell culture in vitro experimental conditions. Therefore, it is not only providing valuable information about cellular molecular events and their quantitative and qualitative analysis, but also confirming the reproducibility of previously published results.
Collapse
Affiliation(s)
- Mustafa Nile
- Department of Orthodontics and Dentofacial Orthopedics, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Folwaczny
- Department of Conservative Dentistry and Periodontology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Andrea Wichelhaus
- Department of Orthodontics and Dentofacial Orthopedics, LMU University Hospital, LMU Munich, Munich, Germany
| | - Uwe Baumert
- Department of Orthodontics and Dentofacial Orthopedics, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mila Janjic Rankovic
- Department of Orthodontics and Dentofacial Orthopedics, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
4
|
Yamada S, Yassin MA, Torelli F, Hansmann J, Green JBA, Schwarz T, Mustafa K. Unique osteogenic profile of bone marrow stem cells stimulated in perfusion bioreactor is Rho-ROCK-mediated contractility dependent. Bioeng Transl Med 2023; 8:e10509. [PMID: 37206242 PMCID: PMC10189446 DOI: 10.1002/btm2.10509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/19/2023] Open
Abstract
The fate determination of bone marrow mesenchymal stem/stromal cells (BMSC) is tightly regulated by mechanical cues, including fluid shear stress. Knowledge of mechanobiology in 2D culture has allowed researchers in bone tissue engineering to develop 3D dynamic culture systems with the potential for clinical translation in which the fate and growth of BMSC are mechanically controlled. However, due to the complexity of 3D dynamic cell culture compared to the 2D counterpart, the mechanisms of cell regulation in the dynamic environment remain relatively undescribed. In the present study, we analyzed the cytoskeletal modulation and osteogenic profiles of BMSC under fluid stimuli in a 3D culture condition using a perfusion bioreactor. BMSC subjected to fluid shear stress (mean 1.56 mPa) showed increased actomyosin contractility, accompanied by the upregulation of mechanoreceptors, focal adhesions, and Rho GTPase-mediated signaling molecules. Osteogenic gene expression profiling revealed that fluid shear stress promoted the expression of osteogenic markers differently from chemically induced osteogenesis. Osteogenic marker mRNA expression, type 1 collagen formation, ALP activity, and mineralization were promoted in the dynamic condition, even in the absence of chemical supplementation. The inhibition of cell contractility under flow by Rhosin chloride, Y27632, MLCK inhibitor peptide-18, or Blebbistatin revealed that actomyosin contractility was required for maintaining the proliferative status and mechanically induced osteogenic differentiation in the dynamic culture. The study highlights the cytoskeletal response and unique osteogenic profile of BMSC in this type of dynamic cell culture, stepping toward the clinical translation of mechanically stimulated BMCS for bone regeneration.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| | - Mohammed A. Yassin
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| | - Francesco Torelli
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| | - Jan Hansmann
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISCWürzburgGermany
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital WürzburgWürzburgGermany
- Department of Electrical EngineeringUniversity of Applied Sciences Würzburg‐SchweinfurtSchweinfurtGermany
| | - Jeremy B. A. Green
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonUK
| | - Thomas Schwarz
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISCWürzburgGermany
| | - Kamal Mustafa
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| |
Collapse
|
5
|
Ghasemzadeh-Hasankolaei M, Miranda JM, Correia CR, Mano JF. Viscous Microcapsules as Microbioreactors to Study Mesenchymal Stem/Stromal Cells Osteolineage Commitment. SMALL METHODS 2023:e2201503. [PMID: 37029584 DOI: 10.1002/smtd.202201503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/07/2023] [Indexed: 06/19/2023]
Abstract
It is essential to design a multifunctional well-controlled platform to transfer mechanical cues to the cells in different magnitudes. This study introduces a platform, a miniaturized bioreactor, which enables to study the effect of shear stress in microsized compartmentalized structures. In this system, the well-established cell encapsulation system of liquefied capsules (LCs) is used as microbioreactors in which the encapsulated cells are exposed to variable core viscosities to experience different mechanical forces under a 3D dynamic culture. The LC technology is joined with electrospraying to produce such microbioreactors at high rates, thus allowing the application of microcapsules for high-throughput screening. Using this platform for osteogenic differentiation as an example, shows that microbioreactors with higher core viscosity which produce higher shear stress lead to significantly higher osteogenic characteristics. Moreover, in this system the forces experienced by cells in each LC are simulated by computational modeling. The maximum wall shear stress applied to the cells inside the bioreactor with low, and high core viscosity environment is estimated to be 297 and 1367 mPa, respectively, for the experimental setup employed. This work outlines the potential of LC microbioreactors as a reliable in vitro customizable platform with a wide range of applications.
Collapse
Affiliation(s)
- Maryam Ghasemzadeh-Hasankolaei
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - João M Miranda
- CEFT-Tranport Phenomena Research Center, Department of Chemical Engineering, Faculdade de Engenharia da Universidade do Porto (FEUP), Rua Dr. Roberto Frias, Porto, 4200-465, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, Porto, 4200-465, Portugal
| | - Clara R Correia
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - João F Mano
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
6
|
Han J, Park S, Kim JE, Park B, Hong Y, Lim JW, Jeong S, Son H, Kim HB, Seonwoo H, Jang KJ, Chung JH. Development of a Scaffold-on-a-Chip Platform to Evaluate Cell Infiltration and Osteogenesis on the 3D-Printed Scaffold for Bone Regeneration. ACS Biomater Sci Eng 2023; 9:968-977. [PMID: 36701173 DOI: 10.1021/acsbiomaterials.2c01367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Developing a scaffold for efficient and functional bone regeneration remains challenging. To accomplish this goal, a "scaffold-on-a-chip" device was developed as a platform to aid with the evaluation process. The device mimics a microenvironment experienced by a transplanted bone scaffold. The device contains a circular space at the center for scaffold insert and microfluidic channel that encloses the space. Such a design allows for monitoring of cell behavior at the blood-scaffold interphase. MC3T3-E1 cells were cultured with three different types of scaffold inserts to test its capability as an evaluation platform. Cellular behaviors, including migration, morphology, and osteogenesis with each scaffold, were analyzed through fluorescence images of live/dead assay and immunocytochemistry. Cellular behaviors, such as migration, morphology, and osteogenesis, were evaluated. The results revealed that our platform could effectively evaluate the osteoconductivity and osteoinductivity of scaffolds with various properties. In conclusion, our proposed platform is expected to replace current in vivo animal models as a highly relevant in vitro platform and can contribute to the fundamental study of bone regeneration.
Collapse
Affiliation(s)
- Jinsub Han
- Department of Biosystems Engineering, Seoul National University, Seoul 08826, Korea.,Convergence Major in Global Smart Farm, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Sangbae Park
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Jae Eun Kim
- Department of Biosystems Engineering, Seoul National University, Seoul 08826, Korea
| | - Byeongjoo Park
- Department of Biosystems Engineering, Seoul National University, Seoul 08826, Korea
| | - Yeonggeol Hong
- Department of Bio-Systems Engineering, Institute of Smart Farm, Gyeongsang National University, Jinju 52828, Korea
| | - Jae Woon Lim
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Seung Jeong
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Hyunmok Son
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Hong Bae Kim
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Hoon Seonwoo
- Department of Convergent Biosystems Engineering, College of Life Sciences and Natural Resources, Sunchon National University, Suncheon 57922, Korea.,Interdisciplinary Program in IT-Bio Convergence System, Sunchon National University, Suncheon 57922, Korea
| | - Kyoung-Je Jang
- Department of Bio-Systems Engineering, Institute of Smart Farm, Gyeongsang National University, Jinju 52828, Korea.,Institute of Agriculture & Life Science, Gyeongsang National University, Jinju 52828, Korea
| | - Jong Hoon Chung
- Department of Biosystems Engineering, Seoul National University, Seoul 08826, Korea.,Convergence Major in Global Smart Farm, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.,Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
7
|
Song W, Bo X, Ma X, Hou K, Li D, Geng W, Zeng J. Craniomaxillofacial derived bone marrow mesenchymal stem/stromal cells (BMSCs) for craniomaxillofacial bone tissue engineering: A literature review. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2022; 123:e650-e659. [PMID: 35691558 DOI: 10.1016/j.jormas.2022.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/06/2022] [Accepted: 06/07/2022] [Indexed: 11/20/2022]
|
8
|
Zhao Y, Richardson K, Yang R, Bousraou Z, Lee YK, Fasciano S, Wang S. Notch signaling and fluid shear stress in regulating osteogenic differentiation. Front Bioeng Biotechnol 2022; 10:1007430. [PMID: 36277376 PMCID: PMC9581166 DOI: 10.3389/fbioe.2022.1007430] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Osteoporosis is a common bone and metabolic disease that is characterized by bone density loss and microstructural degeneration. Human bone marrow-derived mesenchymal stem cells (hMSCs) are multipotent progenitor cells with the potential to differentiate into various cell types, including osteoblasts, chondrocytes, and adipocytes, which have been utilized extensively in the field of bone tissue engineering and cell-based therapy. Although fluid shear stress plays an important role in bone osteogenic differentiation, the cellular and molecular mechanisms underlying this effect remain poorly understood. Here, a locked nucleic acid (LNA)/DNA nanobiosensor was exploited to monitor mRNA gene expression of hMSCs that were exposed to physiologically relevant fluid shear stress to examine the regulatory role of Notch signaling during osteogenic differentiation. First, the effects of fluid shear stress on cell viability, proliferation, morphology, and osteogenic differentiation were investigated and compared. Our results showed shear stress modulates hMSCs morphology and osteogenic differentiation depending on the applied shear and duration. By incorporating this LNA/DNA nanobiosensor and alkaline phosphatase (ALP) staining, we further investigated the role of Notch signaling in regulating osteogenic differentiation. Pharmacological treatment is applied to disrupt Notch signaling to investigate the mechanisms that govern shear stress induced osteogenic differentiation. Our experimental results provide convincing evidence supporting that physiologically relevant shear stress regulates osteogenic differentiation through Notch signaling. Inhibition of Notch signaling mediates the effects of shear stress on osteogenic differentiation, with reduced ALP enzyme activity and decreased Dll4 mRNA expression. In conclusion, our results will add new information concerning osteogenic differentiation of hMSCs under shear stress and the regulatory role of Notch signaling. Further studies may elucidate the mechanisms underlying the mechanosensitive role of Notch signaling in stem cell differentiation.
Collapse
Affiliation(s)
- Yuwen Zhao
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, United States
- Department of Bioengineering, Lehigh University, Bethlehem, PA, United States
| | - Kiarra Richardson
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, United States
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Rui Yang
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, United States
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States
| | - Zoe Bousraou
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, United States
| | - Yoo Kyoung Lee
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, United States
| | - Samantha Fasciano
- Department of Cellular and Molecular Biology, University of New Haven, West Haven, CT, United States
| | - Shue Wang
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, United States
- *Correspondence: Shue Wang,
| |
Collapse
|
9
|
Yamada S, Yassin MA, Schwarz T, Mustafa K, Hansmann J. Optimization and Validation of a Custom-Designed Perfusion Bioreactor for Bone Tissue Engineering: Flow Assessment and Optimal Culture Environmental Conditions. Front Bioeng Biotechnol 2022; 10:811942. [PMID: 35402393 PMCID: PMC8990132 DOI: 10.3389/fbioe.2022.811942] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/07/2022] [Indexed: 11/29/2022] Open
Abstract
Various perfusion bioreactor systems have been designed to improve cell culture with three-dimensional porous scaffolds, and there is some evidence that fluid force improves the osteogenic commitment of the progenitors. However, because of the unique design concept and operational configuration of each study, the experimental setups of perfusion bioreactor systems are not always compatible with other systems. To reconcile results from different systems, the thorough optimization and validation of experimental configuration are required in each system. In this study, optimal experimental conditions for a perfusion bioreactor were explored in three steps. First, an in silico modeling was performed using a scaffold geometry obtained by microCT and an expedient geometry parameterized with porosity and permeability to assess the accuracy of calculated fluid shear stress and computational time. Then, environmental factors for cell culture were optimized, including the volume of the medium, bubble suppression, and medium evaporation. Further, by combining the findings, it was possible to determine the optimal flow rate at which cell growth was supported while osteogenic differentiation was triggered. Here, we demonstrated that fluid shear stress up to 15 mPa was sufficient to induce osteogenesis, but cell growth was severely impacted by the volume of perfused medium, the presence of air bubbles, and medium evaporation, all of which are common concerns in perfusion bioreactor systems. This study emphasizes the necessity of optimization of experimental variables, which may often be underreported or overlooked, and indicates steps which can be taken to address issues common to perfusion bioreactors for bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Centre of Translational Oral Research, Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
- *Correspondence: Shuntaro Yamada, ; Jan Hansmann,
| | - Mohammed A. Yassin
- Centre of Translational Oral Research, Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Thomas Schwarz
- Translational Centre Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany
| | - Kamal Mustafa
- Centre of Translational Oral Research, Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Jan Hansmann
- Translational Centre Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
- Department Electrical Engineering, University of Applied Sciences Würzburg-Schweinfurt, Würzburg, Germany
- *Correspondence: Shuntaro Yamada, ; Jan Hansmann,
| |
Collapse
|
10
|
Gould NR, Leser JM, Torre OM, Khairallah RJ, Ward CW, Stains JP. In vitro Fluid Shear Stress Induced Sclerostin Degradation and CaMKII Activation in Osteocytes. Bio Protoc 2021; 11:e4251. [PMID: 35005095 PMCID: PMC8678913 DOI: 10.21769/bioprotoc.4251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/18/2021] [Accepted: 09/27/2021] [Indexed: 11/19/2023] Open
Abstract
Bone is a dynamic tissue that adapts to changes in its mechanical environment. Mechanical stimuli pressurize interstitial fluid in the lacunar-canalicular system within the bone matrix, causing fluid shear stress (FSS) across bone embedded, mechano-sensitive osteocytes. Therefore, modeling this mechanical stimulus in vitro is vital for identifying mechano-transduction cascades that contribute to the regulation of mechano-responsive proteins, such as the Wnt/β-catenin antagonist, sclerostin, which is reduced in response to FSS. Recently, we reported the rapid post-translational degradation of sclerostin protein in bone cells following FSS. Given the fundamental nature of sclerostin to bone physiology and the nuances of studying its rapid post-translational control, here, we detail our FSS protocol, and adaptations that can be made, to stimulate Ocy454 osteocyte-like cells to study sclerostin protein in vitro. While this protocol is optimized for detecting sclerostin degradation by western blot, this protocol can be adapted to examine transcriptional changes with RT-qPCR, cellular dynamics with live cell imaging, or secreted factors in the FSS buffer. This protocol utilizes 3D-printed FSS tips that are compatible with commercially available 96-well plates, allowing for high experimental accessibility, versatility, and throughput. However, this protocol can be adapted for any FSS chamber. It can also be combined with pharmacological inhibitors or genetic manipulations to interrogate the role of specific cellular components. In all, this experimental set-up and protocol is highly adaptable to allow for many experimental outcomes to examine many aspects of cell mechano-transduction.
Collapse
Affiliation(s)
- Nicole R. Gould
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, USA
| | - Jenna M. Leser
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, USA
| | - Olivia M. Torre
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, USA
| | | | - Christopher W. Ward
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, USA
| | - Joseph P. Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
11
|
Qin X, Zhang K, Qiu J, Wang N, Qu K, Cui Y, Huang J, Luo L, Zhong Y, Tian T, Wu W, Wang Y, Wang G. Uptake of oxidative stress-mediated extracellular vesicles by vascular endothelial cells under low magnitude shear stress. Bioact Mater 2021; 9:397-410. [PMID: 34820579 PMCID: PMC8586717 DOI: 10.1016/j.bioactmat.2021.10.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/11/2021] [Accepted: 10/20/2021] [Indexed: 11/17/2022] Open
Abstract
Extracellular vesicles (EVs) are increasingly used as delivery vehicles for drugs and bioactive molecules, which usually require intravascular administration. The endothelial cells covering the inner surface of blood vessels are susceptible to the shear stress of blood flow. Few studies demonstrate the interplay of red blood cell-derived EVs (RBCEVs) and endothelial cells. Thus, the phagocytosis of EVs by vascular endothelial cells during blood flow needs to be elucidated. In this study, red blood cell-derived extracellular vesicles (RBCEVs) were constructed to investigate endothelial cell phagocytosis in vitro and animal models. Results showed that low magnitude shear stress including low shear stress (LSS) and oscillatory shear stress (OSS) could promote the uptake of RBCEVs by endothelial cells in vitro. In addition, in zebrafish and mouse models, RBCEVs tend to be internalized by endothelial cells under LSS or OSS. Moreover, RBCEVs are easily engulfed by endothelial cells in atherosclerotic plaques exposed to LSS or OSS. In terms of mechanism, oxidative stress induced by LSS is part of the reason for the increased uptake of endothelial cells. Overall, this study shows that vascular endothelial cells can easily engulf EVs in areas of low magnitude shear stress, which will provide a theoretical basis for the development and utilization of EVs-based nano-drug delivery systems in vivo. We recently reported that endothelial cells were amateur phagocytic cells for RBCEVs engulfment. Low magnitude shear stress (LSS and OSS) can increase the uptake of RBCEVs by endothelial cells in vitro and in vivo. ROS induced by low magnitude shear stress acts as an accelerator to enhance endothelial cells uptake of RBCEVs.
Collapse
Affiliation(s)
- Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Nan Wang
- The Nanoscience Centre, University of Cambridge, Cambridge, CB3 0FF, UK
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yuliang Cui
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Junli Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Li Luo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Tian Tian
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| |
Collapse
|
12
|
Manokawinchoke J, Pavasant P, Limjeerajarus CN, Limjeerajarus N, Osathanon T, Egusa H. Mechanical loading and the control of stem cell behavior. Arch Oral Biol 2021; 125:105092. [PMID: 33652301 DOI: 10.1016/j.archoralbio.2021.105092] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/08/2021] [Accepted: 02/21/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Mechanical stimulation regulates many cell responses. The present study describes the effects of different in vitro mechanical stimulation approaches on stem cell behavior. DESIGN The narrative review approach was performed. The articles published in English language that addressed the effects of mechanical force on stem cells were searched on Pubmed and Scopus database. The effects of extrinsic mechanical force on stem cell response was reviewed and discussed. RESULTS Cells sense mechanical stimuli by the function of mechanoreceptors and further transduce force stimulation into intracellular signaling. Cell responses to mechanical stimuli depend on several factors including type, magnitude, and duration. Further, similar mechanical stimuli exhibit distinct cell responses based on numerous factors including cell type and differentiation stage. Various mechanical applications modulate stemness maintenance and cell differentiation toward specific lineages. CONCLUSIONS Mechanical force application modulates stemness maintenance and differentiation. Modification of force regimens could be utilized to precisely control appropriate stem cell behavior toward specific applications.
Collapse
Affiliation(s)
- Jeeranan Manokawinchoke
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan.
| | - Prasit Pavasant
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Chalida Nakalekha Limjeerajarus
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Nuttapol Limjeerajarus
- Research Center for Advanced Energy Technology, Faculty of Engineering, Thai-Nichi Institute of Technology, Bangkok, 10250, Thailand.
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan.
| |
Collapse
|
13
|
Yamada S, Yassin MA, Schwarz T, Hansmann J, Mustafa K. Induction of osteogenic differentiation of bone marrow stromal cells on 3D polyester-based scaffolds solely by subphysiological fluidic stimulation in a laminar flow bioreactor. J Tissue Eng 2021; 12:20417314211019375. [PMID: 34262684 PMCID: PMC8243246 DOI: 10.1177/20417314211019375] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/04/2021] [Indexed: 01/09/2023] Open
Abstract
The fatal determination of bone marrow mesenchymal stem/stromal cells (BMSC) is closely associated with mechano-environmental factors in addition to biochemical clues. The aim of this study was to induce osteogenesis in the absence of chemical stimuli using a custom-designed laminar flow bioreactor. BMSC were seeded onto synthetic microporous scaffolds and subjected to the subphysiological level of fluid flow for up to 21 days. During the perfusion, cell proliferation was significantly inhibited. There were also morphological changes, with F-actin polymerisation and upregulation of ROCK1. Notably, in BMSC subjected to flow, mRNA expression of osteogenic markers was significantly upregulated and RUNX2 was localised in the nuclei. Further, under perfusion, there was greater deposition of collagen type 1 and calcium onto the scaffolds. The results confirm that an appropriate level of fluid stimuli preconditions BMSC towards the osteoblastic lineage on 3D scaffolds in the absence of chemical stimulation, which highlights the utility of flow bioreactors in bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| | - Mohammed Ahmed Yassin
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| | - Thomas Schwarz
- Fraunhofer Institute for Silicate
Research ISC, Translational Center Regenerative Therapies, Wurzburg, Bayern,
Germany
| | - Jan Hansmann
- Fraunhofer Institute for Silicate
Research ISC, Translational Center Regenerative Therapies, Wurzburg, Bayern,
Germany
- Chair of Tissue Engineering and
Regenerative Medicine, University Hospital Würzburg, Germany
- Department Electrical Engineering,
University of Applied Sciences Würzburg-Schweinfurt, Germany
| | - Kamal Mustafa
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| |
Collapse
|
14
|
Matichescu A, Ardelean LC, Rusu LC, Craciun D, Bratu EA, Babucea M, Leretter M. Advanced Biomaterials and Techniques for Oral Tissue Engineering and Regeneration-A Review. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E5303. [PMID: 33238625 PMCID: PMC7700200 DOI: 10.3390/ma13225303] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022]
Abstract
The reconstruction or repair of oral and maxillofacial functionalities and aesthetics is a priority for patients affected by tooth loss, congenital defects, trauma deformities, or various dental diseases. Therefore, in dental medicine, tissue reconstruction represents a major interest in oral and maxillofacial surgery, periodontics, orthodontics, endodontics, and even daily clinical practice. The current clinical approaches involve a vast array of techniques ranging from the traditional use of tissue grafts to the most innovative regenerative procedures, such as tissue engineering. In recent decades, a wide range of both artificial and natural biomaterials and scaffolds, genes, stem cells isolated from the mouth area (dental follicle, deciduous teeth, periodontal ligament, dental pulp, salivary glands, and adipose tissue), and various growth factors have been tested in tissue engineering approaches in dentistry, with many being proven successful. However, to fully eliminate the problems of traditional bone and tissue reconstruction in dentistry, continuous research is needed. Based on a recent literature review, this paper creates a picture of current innovative strategies applying dental stem cells for tissue regeneration in different dental fields and maxillofacial surgery, and offers detailed information regarding the available scientific data and practical applications.
Collapse
Affiliation(s)
- Anamaria Matichescu
- Department of Preventive Dentistry, Community and Oral Health, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Lavinia Cosmina Ardelean
- Department of Technology of Materials and Devices in Dental Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Laura-Cristina Rusu
- Department of Oral Pathology, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (L.-C.R.); (D.C.); (M.B.)
| | - Dragos Craciun
- Department of Oral Pathology, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (L.-C.R.); (D.C.); (M.B.)
| | - Emanuel Adrian Bratu
- Department of Implant Supported Restorations, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Marius Babucea
- Department of Oral Pathology, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (L.-C.R.); (D.C.); (M.B.)
| | - Marius Leretter
- Department of Prosthodontics, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania;
| |
Collapse
|
15
|
Pourlak T, Pourlak T, Ghodrati M, Mortazavi A, Dolati S, Yousefi M. Usage of stem cells in oral and maxillofacial region. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2020; 122:441-452. [PMID: 33099018 DOI: 10.1016/j.jormas.2020.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/07/2020] [Accepted: 10/06/2020] [Indexed: 11/30/2022]
Abstract
Malformations of the maxillofacial region has disturbing psychosocial effects and causes enormous socioeconomic concerns. The management of maxillofacial defects caused by congenital anomalies, trauma, osteoporotic fractures, periodontitis, or cancer treatment is challenging for oral and maxillofacial surgeons. Numerous approaches have been recommended for the managing of these deficiencies. The traditional treatment for maxillofacial defects or their repair is an intricate process by autologous bone grafts from the scapula, ribs, fibula, or iliac crest origins. Regenerative medicine is well thought-out as a perfect substitute approach for autologous bone grafts to renovate bone deficiencies. The use of stem cells has improved results and offered a technique to reconstruct craniofacial bone defects. The field of tissue engineering for the regeneration of maxillofacial needs integration of biochemical and biomaterial engineering aspects with cell transplantation to generate better-quality biomimetic scaffolds, prevascularize three-dimensional (3D) tissue structures, and engineer the composite interface of diverse facial tissues. In this review, we have discussed the application of different adult stem cells to repair oral and maxillofacial defects in animal models and clinical trials.
Collapse
Affiliation(s)
- T Pourlak
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - T Pourlak
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - M Ghodrati
- Department of Endodontics, Dental and Periodental Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - A Mortazavi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - S Dolati
- Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - M Yousefi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
da Silva Madaleno C, Jatzlau J, Knaus P. BMP signalling in a mechanical context - Implications for bone biology. Bone 2020; 137:115416. [PMID: 32422297 DOI: 10.1016/j.bone.2020.115416] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 01/12/2023]
Abstract
Bone Morphogenetic Proteins (BMPs) are extracellular multifunctional signalling cytokines and members of the TGFβ super family. These pleiotropic growth factors crucially promote bone formation, remodeling and healing after injury. Additionally, bone homeostasis is systematically regulated by mechanical inputs from the environment, which are incorporated into the bone cells' biochemical response. These inputs range from compression and tension induced by the movement of neighboring muscle, to fluid shear stress induced by interstitial fluid flow in the canaliculi and in the vascular system. Although BMPs are widely applied in a clinic context to promote fracture healing, it is still elusive how mechanical inputs modulate this signalling pathway, hindering an efficient and side-effect free application of these ligands in bone healing. This review aims to summarize the current understanding in how mechanical cues (tension, compression, shear force and hydrostatic pressure) and substrate stiffness modulate BMP signalling. We highlight the time-dependent effects in modulating immediate early up to long-term effects of mechano-BMP crosstalk during bone formation and remodeling, considering the interplay with other already established mechanosensitive pathways, such as MRTF/SRF and Hippo signalling.
Collapse
Affiliation(s)
- Carolina da Silva Madaleno
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Berlin Brandenburg School of Regenerative Therapies (BSRT), Charité Universitätsmedizin, Berlin, Germany
| | - Jerome Jatzlau
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Berlin Brandenburg School of Regenerative Therapies (BSRT), Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
17
|
Zhang P, Zhang C, Li J, Han J, Liu X, Yang H. The physical microenvironment of hematopoietic stem cells and its emerging roles in engineering applications. Stem Cell Res Ther 2019; 10:327. [PMID: 31744536 PMCID: PMC6862744 DOI: 10.1186/s13287-019-1422-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/22/2019] [Accepted: 09/23/2019] [Indexed: 12/18/2022] Open
Abstract
Stem cells are considered the fundamental underpinnings of tissue biology. The stem cell microenvironment provides factors and elements that play significant roles in controlling the cell fate direction. The bone marrow is an important environment for functional hematopoietic stem cells in adults. Remarkable progress has been achieved in the area of hematopoietic stem cell fate modulation based on the recognition of biochemical factors provided by bone marrow niches. In this review, we focus on emerging evidence that hematopoietic stem cell fate is altered in response to a variety of microenvironmental physical cues, such as geometric properties, matrix stiffness, and mechanical forces. Based on knowledge of these biophysical cues, recent developments in harnessing hematopoietic stem cell niches ex vivo are also discussed. A comprehensive understanding of cell microenvironments helps provide mechanistic insights into pathophysiological mechanisms and underlies biomaterial-based hematopoietic stem cell engineering.
Collapse
Affiliation(s)
- Pan Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Chen Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Jing Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Jiyang Han
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Xiru Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China.
- Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China.
| |
Collapse
|
18
|
Ferrell N, Sandoval RM, Molitoris BA, Brakeman P, Roy S, Fissell WH. Application of physiological shear stress to renal tubular epithelial cells. Methods Cell Biol 2019; 153:43-67. [PMID: 31395384 DOI: 10.1016/bs.mcb.2019.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Renal tubular epithelial cells are consistently exposed to flow of glomerular filtrate that creates fluid shear stress at the apical cell surface. This biophysical stimulus regulates several critical renal epithelial cell functions, including transport, protein uptake, and barrier function. Defining the in vivo mechanical conditions in the kidney tubule is important for accurately recapitulating these conditions in vitro. Here we provide a summary of the fluid flow conditions in the kidney and how this translates into different levels of fluid shear stress down the length of the nephron. A detailed method is provided for measuring fluid flow in the proximal tubule by intravital microscopy. Devices to mimic in vivo fluid shear stress for in vitro studies are discussed, and we present two methods for culture and analysis of renal tubule epithelial cells exposed physiological levels of fluid shear stress. The first is a microfluidic device that permits application of controlled shear stress to cells cultured on porous membranes. The second is culture of renal tubule cells on an orbital shaker. Each method has advantages and disadvantages that should be considered in the context of the specific experimental objectives.
Collapse
Affiliation(s)
- Nicholas Ferrell
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States.
| | - Ruben M Sandoval
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Bruce A Molitoris
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Paul Brakeman
- Department of Pediatrics, University of California, San Francisco, CA, United States
| | - Shuvo Roy
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, United States
| | - William H Fissell
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
19
|
Choi HY, Yang GM, Dayem AA, Saha SK, Kim K, Yoo Y, Hong K, Kim JH, Yee C, Lee KM, Cho SG. Hydrodynamic shear stress promotes epithelial-mesenchymal transition by downregulating ERK and GSK3β activities. Breast Cancer Res 2019; 21:6. [PMID: 30651129 PMCID: PMC6335853 DOI: 10.1186/s13058-018-1071-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/26/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) occurs in the tumor microenvironment and presents an important mechanism of tumor cell intravasation, stemness acquisition, and metastasis. During metastasis, tumor cells enter the circulation to gain access to distant tissues, but how this fluid microenvironment influences cancer cell biology is poorly understood. METHODS AND RESULTS Here, we present both in vivo and in vitro evidence that EMT-like transition also occurs in circulating tumor cells (CTCs) as a result of hydrodynamic shear stress (+SS), which promotes conversion of CD24middle/CD44high/CD133middle/CXCR4low/ALDH1low primary patient epithelial tumor cells into specific high sphere-forming CD24low/CD44low/CD133high/CXCR4high/ALDH1high cancer stem-like cells (CSLCs) or tumor-initiating cells (TICs) with elevated tumor progression and metastasis capacity in vitro and in vivo. We demonstrate that conversion of CSLCs/TICs from epithelial tumor cells via +SS is dependent on reactive oxygen species (ROS)/nitric oxide (NO) generation, and suppression of extracellular signal-related kinase (ERK)/glycogen synthase kinase (GSK)3β, a mechanism similar to that operating in embryonic stem cells to prevent their differentiation while promoting self-renewal. CONCLUSION Fluid shear stress experienced during systemic circulation of human breast tumor cells can lead to specific acquisition of mesenchymal stem cell (MSC)-like potential that promotes EMT, mesenchymal-epithelial transition, and metastasis to distant organs. Our data revealed that biomechanical forces appeared to be important microenvironmental factors that not only drive hematopoietic development but also lead to acquisition of CSLCs/TIC potential in cancer metastasis. Our data highlight that +SS is a critical factor that promotes the conversion of CTCs into distinct TICs in blood circulation by endowing plasticity to these cells and by maintaining their self-renewal signaling pathways.
Collapse
Affiliation(s)
- Hye Yeon Choi
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Gwang-Mo Yang
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Subbroto Kumar Saha
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Kyeongseok Kim
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Youngbum Yoo
- Department of Surgery, Konkuk University School of Medicine, Seoul, 05030, Republic of Korea
| | - Kwonho Hong
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Jin-Hoi Kim
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Cassian Yee
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Kyung-Mi Lee
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 26-1 Anam-dong, Sungbuk-gu, Seoul, 02841, Republic of Korea.
| | - Ssang-Goo Cho
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| |
Collapse
|
20
|
Shaito A, Saliba J, Husari A, El-Harakeh M, Chhouri H, Hashem Y, Shihadeh A, El-Sabban M. Electronic Cigarette Smoke Impairs Normal Mesenchymal Stem Cell Differentiation. Sci Rep 2017; 7:14281. [PMID: 29079789 PMCID: PMC5660168 DOI: 10.1038/s41598-017-14634-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022] Open
Abstract
Electronic cigarettes (e-cigarettes) are promoted as low-risk alternatives to combustible cigarettes. However, the effects of chronic inhalation of potential toxicants emitted by ecigarettes remain largely unexamined. It is conceivable that smoking-induced chronic diseases result in cellular injury, in the absence of effective repair by stem cells. This study evaluates the effect of cigarette and e-cigarette aerosol extracts on the survival and differentiation of bone marrow-derived mesenchymal stem cells (MSCs). MSC growth and osteogenic differentiation were examined after exposure to smoke extracts. Data revealed detrimental effects of both cigarette and e-cigarette extracts on MSC morphology and growth. Levels and activity of alkaline phosphatase, an osteogenic marker, decreased and induction of osteoblastic differentiation was impaired. Both smoke extracts prevented osteogenic differentiation from progressing, evident by decreased expression of terminal osteogenic markers and mineralization. Elevated levels of reactive oxygen species (ROS) were detected in cells exposed to smoke extracts. Moreover, decreased differentiation potential was concomitant with severe down-regulation of Connexin 43 expression, leading to the loss of gap junction-mediated communication, which together with elevated ROS levels, could explain decreased proliferation and loss of differentiation potential. Hence, e-cigarettes present similar risk as combustible cigarettes with respect to tissue repair impairment.
Collapse
Affiliation(s)
- A Shaito
- Department of Biological and Chemical Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut, Lebanon
| | - J Saliba
- Department of Biology, Faculty of Science, Lebanese University, Beirut, Lebanon
| | - A Husari
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - M El-Harakeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - H Chhouri
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Y Hashem
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - A Shihadeh
- Department of Mechanical Engineering, Faculty of Engineering, American University of Beirut, Beirut, Lebanon
| | - M El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
21
|
Sun YX, Zhang JF, Xu J, Xu LL, Wu TY, Wang B, Pan XH, Li G. MicroRNA-144-3p inhibits bone formation in distraction osteogenesis through targeting Connexin 43. Oncotarget 2017; 8:89913-89922. [PMID: 29163798 PMCID: PMC5685719 DOI: 10.18632/oncotarget.20984] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/17/2017] [Indexed: 11/25/2022] Open
Abstract
Distraction osteogenesis (DO), one of effective therapies for bone regeneration, has been received more attention in recent years. However, the underlying mechanism remains elusive. Recently, microRNAs (miRNAs) have been reported to play important roles in regulating osteogenesis and bone formation. We therefore provided the hypothesis that miRNAs could involve in the DO-mediated bone regeneration. After successfully established the DO model of rats, a miRNA microarray was performed to find the differently expressed miRNAs in DO and control groups in this study. As one of the most downregulated miRNAs, miR-144-3p was found to be decreased during osteogenic differentiation in mesenchymal stem cells of rats (rBMSCs) and DO model. And miR-144-3p overexpression suppressed the osteogenesis while its inhibitor promoted osteogenesis. Furthermore, Connexin-43, an essential regulator for osteogenesis, was validated to be a novel target for miR-144-3p. Finally, miR-144-3p inhibitor modified MSCs promoted mineralization of distracted bone in rat DO model. In conclusion, miR-144-3p was found to regulate osteogenesis and inhibition of miR-144-3p resulted in acceleration of mineralization of DO, which not only give clues to understanding the mechanism of DO but also provide a potential therapeutic target in clinical practice.
Collapse
Affiliation(s)
- Yu-Xin Sun
- Department of Orthopaedics and Traumatology, Bao-An District People's Hospital, Shenzhen, PR China.,Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Jin-Fang Zhang
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Jia Xu
- Department of Orthopaedics Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, PR China
| | - Liang-Liang Xu
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Tian-Yi Wu
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Bin Wang
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Xiao-Hua Pan
- Department of Orthopaedics and Traumatology, Bao-An District People's Hospital, Shenzhen, PR China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Bao-An District People's Hospital, Shenzhen, PR China.,Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences and Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China.,Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| |
Collapse
|
22
|
|
23
|
Huselstein C, Rahouadj R, de Isla N, Bensoussan D, Stoltz JF, Li YP. Mechanobiology of mesenchymal stem cells: Which interest for cell-based treatment? Biomed Mater Eng 2017; 28:S47-S56. [PMID: 28372277 DOI: 10.3233/bme-171623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Thanks to their immune properties, the mesenchymal stem cells (MSC) are a promising source for cell therapy. Current clinical trials show that MSC administrated to patients can treat different diseases (graft-versus-host disease (GVHD), liver cirrhosis, systemic lupus, erythematosus, rheumatoid arthritis, type I diabetes…). In this case, the most common mode of cell administration is the intravenous injection, and the hemodynamic environment of cells induced by blood circulation could interfere on their behavior during the migration and homing towards the injured site. After a brief review of the mechanobiology concept, this paper will help in understanding how the mechanical environment could interact with MSC behavior once they are injected to patient in cell-based treatment.
Collapse
Affiliation(s)
- Céline Huselstein
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54500 Vandœuvre-lès-Nancy, France.,Université de Lorraine, 54000 Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, F-54505 Vandœuvre-lès-Nancy, France
| | - R Rahouadj
- Université de Lorraine, 54000 Nancy, France.,UMR 7563 CNRS-Université de Lorraine, LEMTA, Vandœuvre-lès-Nancy, France
| | - N de Isla
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54500 Vandœuvre-lès-Nancy, France.,Université de Lorraine, 54000 Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, F-54505 Vandœuvre-lès-Nancy, France
| | - D Bensoussan
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54500 Vandœuvre-lès-Nancy, France.,Université de Lorraine, 54000 Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, F-54505 Vandœuvre-lès-Nancy, France.,CHU de Nancy, Unité de Thérapie Cellulaire, banque de Tissus, 54500 Vandœuvre-lès-Nancy, France
| | - J F Stoltz
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54500 Vandœuvre-lès-Nancy, France.,Université de Lorraine, 54000 Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, F-54505 Vandœuvre-lès-Nancy, France
| | - Y P Li
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| |
Collapse
|
24
|
Chalisserry EP, Nam SY, Park SH, Anil S. Therapeutic potential of dental stem cells. J Tissue Eng 2017; 8:2041731417702531. [PMID: 28616151 PMCID: PMC5461911 DOI: 10.1177/2041731417702531] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/12/2017] [Indexed: 12/13/2022] Open
Abstract
Stem cell biology has become an important field in regenerative medicine and tissue engineering therapy since the discovery and characterization of mesenchymal stem cells. Stem cell populations have also been isolated from human dental tissues, including dental pulp stem cells, stem cells from human exfoliated deciduous teeth, stem cells from apical papilla, dental follicle progenitor cells, and periodontal ligament stem cells. Dental stem cells are relatively easily obtainable and exhibit high plasticity and multipotential capabilities. The dental stem cells represent a gold standard for neural-crest-derived bone reconstruction in humans and can be used for the repair of body defects in low-risk autologous therapeutic strategies. The bioengineering technologies developed for tooth regeneration will make substantial contributions to understand the developmental process and will encourage future organ replacement by regenerative therapies in a wide variety of organs such as the liver, kidney, and heart. The concept of developing tooth banking and preservation of dental stem cells is promising. Further research in the area has the potential to herald a new dawn in effective treatment of notoriously difficult diseases which could prove highly beneficial to mankind in the long run.
Collapse
Affiliation(s)
- Elna Paul Chalisserry
- Interdisciplinary Program of Marine-Bio, Electrical & Mechanical Engineering, Pukyong National University, Busan, Korea
- Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, Korea
| | - Seung Yun Nam
- Interdisciplinary Program of Marine-Bio, Electrical & Mechanical Engineering, Pukyong National University, Busan, Korea
- Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, Korea
- Department of Biomedical Engineering, Pukyong National University, Busan, South Korea
| | - Sang Hyug Park
- Interdisciplinary Program of Marine-Bio, Electrical & Mechanical Engineering, Pukyong National University, Busan, Korea
- Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, Korea
- Department of Biomedical Engineering, Pukyong National University, Busan, South Korea
| | - Sukumaran Anil
- Division of Periodontics, Department of Preventive Dental Sciences, College of Dentistry Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
25
|
Wittkowske C, Reilly GC, Lacroix D, Perrault CM. In Vitro Bone Cell Models: Impact of Fluid Shear Stress on Bone Formation. Front Bioeng Biotechnol 2016; 4:87. [PMID: 27896266 PMCID: PMC5108781 DOI: 10.3389/fbioe.2016.00087] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 10/25/2016] [Indexed: 01/06/2023] Open
Abstract
This review describes the role of bone cells and their surrounding matrix in maintaining bone strength through the process of bone remodeling. Subsequently, this work focusses on how bone formation is guided by mechanical forces and fluid shear stress in particular. It has been demonstrated that mechanical stimulation is an important regulator of bone metabolism. Shear stress generated by interstitial fluid flow in the lacunar-canalicular network influences maintenance and healing of bone tissue. Fluid flow is primarily caused by compressive loading of bone as a result of physical activity. Changes in loading, e.g., due to extended periods of bed rest or microgravity in space are associated with altered bone remodeling and formation in vivo. In vitro, it has been reported that bone cells respond to fluid shear stress by releasing osteogenic signaling factors, such as nitric oxide, and prostaglandins. This work focusses on the application of in vitro models to study the effects of fluid flow on bone cell signaling, collagen deposition, and matrix mineralization. Particular attention is given to in vitro set-ups, which allow long-term cell culture and the application of low fluid shear stress. In addition, this review explores what mechanisms influence the orientation of collagen fibers, which determine the anisotropic properties of bone. A better understanding of these mechanisms could facilitate the design of improved tissue-engineered bone implants or more effective bone disease models.
Collapse
Affiliation(s)
- Claudia Wittkowske
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK; INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Gwendolen C Reilly
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK; Department of Material Science, University of Sheffield, Sheffield, UK
| | - Damien Lacroix
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK; INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Cecile M Perrault
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK; INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| |
Collapse
|
26
|
Autologous bone marrow mesenchymal stem cells associated with tantalum rod implantation and vascularized iliac grafting for the treatment of end-stage osteonecrosis of the femoral head. BIOMED RESEARCH INTERNATIONAL 2015; 2015:240506. [PMID: 25802840 PMCID: PMC4352743 DOI: 10.1155/2015/240506] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 10/30/2014] [Accepted: 11/02/2014] [Indexed: 01/24/2023]
Abstract
Tantalum rod implantation with vascularized iliac grafting has been reported to be an effective method for the treatment of young patients with osteonecrosis of the femoral head (ONFH) to avert the need for total hip arthroplasty (THA). However, there have been unsatisfactory success rates for end-stage ONFH. The authors describe a modified technique using bone marrow mesenchymal stem cells (BMMSCs) associated with porous tantalum rod implantation combined with vascularized iliac grafting for the treatment of end-stage ONFH. A total of 24 patients (31 hips) with end-stage ONFH were treated with surgery; ARCO IIIc stage was observed in 19 hips and ARCO IV stage was observed in 12 hips. All patients were followed for a mean time of 64.35 ± 13.03 months (range 26–78). Operations on only five hips were converted to THA. The joint-preserving success rate of the entire group was 89.47% for ARCO stage IIIc and 75% for ARCO stage IV. The mean Harris hip score of the 31 hips improved significantly from 38.74 ± 5.88 points (range 22–50) to 77.23 ± 14.75 points (range 33–95). This intervention was safe and effective in delaying or avoiding total hip replacement for end-stage ONFH.
Collapse
|
27
|
Liu J, Yu F, Sun Y, Jiang B, Zhang W, Yang J, Xu GT, Liang A, Liu S. Concise Reviews: Characteristics and Potential Applications of Human Dental Tissue-Derived Mesenchymal Stem Cells. Stem Cells 2015; 33:627-38. [PMID: 25447379 DOI: 10.1002/stem.1909] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 10/21/2014] [Accepted: 11/07/2014] [Indexed: 12/16/2022]
Affiliation(s)
- Junjun Liu
- Department of Ophthalmology; Shanghai Tenth People's Hospital
| | - Fang Yu
- Laboratory of Oral Biomedical Science and Translational Medicine, School of Stomatology; Tongji University; Shanghai People's Republic of China
| | - Yao Sun
- Laboratory of Oral Biomedical Science and Translational Medicine, School of Stomatology; Tongji University; Shanghai People's Republic of China
| | - Beizhan Jiang
- Laboratory of Oral Biomedical Science and Translational Medicine, School of Stomatology; Tongji University; Shanghai People's Republic of China
| | - Wenjun Zhang
- Translational Center for Stem Cell Research, Tongji Hospital; Tongji University School of Medicine; Shanghai People's Republic of China
| | - Jianhua Yang
- Department of Ophthalmology; Shanghai Tenth People's Hospital
| | - Guo-Tong Xu
- Department of Ophthalmology; Shanghai Tenth People's Hospital
| | - Aibin Liang
- Translational Center for Stem Cell Research, Tongji Hospital; Tongji University School of Medicine; Shanghai People's Republic of China
| | - Shangfeng Liu
- Department of Ophthalmology; Shanghai Tenth People's Hospital
| |
Collapse
|
28
|
Godby RC, Munjal C, Opoka AM, Smith JM, Yutzey KE, Narmoneva DA, Hinton RB. Cross Talk between NOTCH Signaling and Biomechanics in Human Aortic Valve Disease Pathogenesis. J Cardiovasc Dev Dis 2014; 1:237-256. [PMID: 29552567 PMCID: PMC5856658 DOI: 10.3390/jcdd1030237] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aortic valve disease is a burgeoning public health problem associated with significant mortality. Loss of function mutations in NOTCH1 cause bicuspid aortic valve (BAV) and calcific aortic valve disease. Because calcific nodules manifest on the fibrosa side of the cusp in low fluidic oscillatory shear stress (OSS), elucidating pathogenesis requires approaches that consider both molecular and mechanical factors. Therefore, we examined the relationship between NOTCH loss of function (LOF) and biomechanical indices in healthy and diseased human aortic valve interstitial cells (AVICs). An orbital shaker system was used to apply cyclic OSS, which mimics the cardiac cycle and hemodynamics experienced by AVICs in vivo. NOTCH LOF blocked OSS-induced cell alignment in human umbilical vein endothelial cells (HUVECs), whereas AVICs did not align when subjected to OSS under any conditions. In healthy AVICs, OSS resulted in decreased elastin (ELN) and α-SMA (ACTA2). NOTCH LOF was associated with similar changes, but in diseased AVICs, NOTCH LOF combined with OSS was associated with increased α-SMA expression. Interestingly, AVICs showed relatively higher expression of NOTCH2 compared to NOTCH1. Biomechanical interactions between endothelial and interstitial cells involve complex NOTCH signaling that contributes to matrix homeostasis in health and disorganization in disease.
Collapse
Affiliation(s)
- Richard C. Godby
- Division of Cardiology, the Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Charu Munjal
- Division of Cardiology, the Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Amy M. Opoka
- Division of Cardiology, the Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - J. Michael Smith
- TriHealth Heart Institute, Cardio-Thoracic Surgery, Good Samaritan Hospital, Cincinnati, OH 45242, USA
| | - Katherine E. Yutzey
- Molecular Cardiovascular Biology, the Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Daria A. Narmoneva
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Robert B. Hinton
- Division of Cardiology, the Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-513-636-0389; Fax: +1-513-636-5958
| |
Collapse
|