101
|
Chen L, Hu H, Qiu W, Shi K, Kassem M. Actin depolymerization enhances adipogenic differentiation in human stromal stem cells. Stem Cell Res 2018; 29:76-83. [DOI: 10.1016/j.scr.2018.03.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 10/17/2022] Open
|
102
|
Actin and myosin II modulate differentiation of pluripotent stem cells. PLoS One 2018; 13:e0195588. [PMID: 29664925 PMCID: PMC5903644 DOI: 10.1371/journal.pone.0195588] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 03/25/2018] [Indexed: 12/20/2022] Open
Abstract
Use of stem cell-based therapies in tissue engineering and regenerative medicine is hindered by efficient means of directed differentiation. For pluripotent stem cells, an initial critical differentiation event is specification to one of three germ lineages: endoderm, mesoderm, and ectoderm. Differentiation is known to be regulated by numerous extracellular and intracellular factors, but the role of the cytoskeleton during specification, or early differentiation, is still unknown. In these studies, we used agonists and antagonists to modulate actin polymerization and the actin-myosin molecular motor during spontaneous differentiation of embryonic stem cells in embryoid bodies. We found that inhibiting either actin polymerization or actin-myosin interactions led to a decrease in differentiation to the mesodermal lineage and an increase in differentiation to the endodermal lineage. Thus, targeting processes that regulate cytoskeletal tension may be effective in enhancing or inhibiting differentiation towards cells of the endodermal or mesodermal lineages, which include hepatocytes, islets, cardiomyocytes, endothelial cells, and osteocytes. Therefore, these fundamental findings demonstrate that modulation of the cytoskeleton may be useful in production for a range of cell-based therapies, including for liver, pancreatic, cardiac, vascular, and orthopedic applications.
Collapse
|
103
|
Zhuang J, Lin S, Dong L, Cheng K, Weng W. Magnetically actuated mechanical stimuli on Fe 3O 4/mineralized collagen coatings to enhance osteogenic differentiation of the MC3T3-E1 cells. Acta Biomater 2018; 71:49-60. [PMID: 29550443 DOI: 10.1016/j.actbio.2018.03.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 02/03/2018] [Accepted: 03/05/2018] [Indexed: 12/20/2022]
Abstract
Mechanical stimuli at the bone-implant interface are considered to activate the mechanotransduction pathway of the cell to improve the initial osseointegration establishment and to guarantee clinical success of the implant. However, control of the mechanical stimuli at the bone-implant interface still remains a challenge. In this study, we have designed a strategy of a magnetically responsive coating on which the mechanical stimuli is controlled because of coating deformation under static magnetic field (SMF). The iron oxide nanoparticle/mineralized collagen (IOP-MC) coatings were electrochemically codeposited on titanium substrates in different quantities of IOPs and distributions; the resulting coatings were verified to possess swelling behavior with flexibility same as that of hydrogel. The relative quantity of IOP to collagen and the IOP distribution in the coatings were demonstrated to play a critical role in mediating cell behavior. The cells present on the outer layer of the distributed IOP-MC (O-IOP-MC) coating with a mass ratio of 0.67 revealed the most distinct osteogenic differentiation activity being promoted, which could be attributed to the maximized mechanical stimuli with exposure to SMF. Furthermore, the enhanced osteogenic differentiation of the stimulated MC3T3-E1 cells originated from magnetically actuated mechanotransduction signaling pathway, embodying the upregulated expression of osteogenic-related and mechanotransduction-related genes. This work therefore provides a promising strategy for implementing mechanical stimuli to activate mechanotransduction on the bone-implant interface and thus to promote osseointegration. STATEMENT OF SIGNIFICANCE The magnetically actuated coating is designed to produce mechanical stimuli to cells for promoting osteogenic differentiation based on the coating deformation. Iron oxide nanoparticles (IOPs) were incorporated into the mineralized collagen coatings (MC) forming the composite coatings (IOP-MC) with spatially distributed IOPs, and the IOP-MC coatings with outer distributed IOPs (O-IOPs-MC) shows the maximized mechanical stimuli to cells with enhanced osteogenic differentiation under static magnetic field. The upregulated expression of the associated genes reveals that the enabled mechanotransduction signaling pathway is responsible for the promoted cellular osteogenic differentiation. This work therefore provides a promising strategy for implementing mechanical stimuli to activate mechanotransduction on the bone-implant interface to promote osseointegration.
Collapse
|
104
|
Schneider M, Angele P, Järvinen TA, Docheva D. Rescue plan for Achilles: Therapeutics steering the fate and functions of stem cells in tendon wound healing. Adv Drug Deliv Rev 2018; 129:352-375. [PMID: 29278683 DOI: 10.1016/j.addr.2017.12.016] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 12/01/2017] [Accepted: 12/22/2017] [Indexed: 02/07/2023]
Abstract
Due to the increasing age of our society and a rise in engagement of young people in extreme and/or competitive sports, both tendinopathies and tendon ruptures present a clinical and financial challenge. Tendon has limited natural healing capacity and often responds poorly to treatments, hence it requires prolonged rehabilitation in most cases. Till today, none of the therapeutic options has provided successful long-term solutions, meaning that repaired tendons do not recover their complete strength and functionality. Our understanding of tendon biology and healing increases only slowly and the development of new treatment options is insufficient. In this review, following discussion on tendon structure, healing and the clinical relevance of tendon injury, we aim to elucidate the role of stem cells in tendon healing and discuss new possibilities to enhance stem cell treatment of injured tendon. To date, studies mainly apply stem cells, often in combination with scaffolds or growth factors, to surgically created tendon defects. Deeper understanding of how stem cells and vasculature in the healing tendon react to growth factors, common drugs used to treat injured tendons and promising cellular boosters could help to develop new and more efficient ways to manage tendon injuries.
Collapse
|
105
|
Ji Y, Cao M, Liu J, Chen Y, Li X, Zhao J, Qu C. Rock signaling control PPARγ expression and actin polymerization during adipogenesis. Saudi J Biol Sci 2018; 24:1866-1870. [PMID: 29551937 PMCID: PMC5851925 DOI: 10.1016/j.sjbs.2017.11.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/09/2017] [Accepted: 11/09/2017] [Indexed: 11/25/2022] Open
Abstract
Aim: Adipogenesis is characterized by a strong interdependence between cell shape, cytoskeletal organization, and the onset of adipogenic gene expression. Here we investigated the role of the RhoA/ROCK pathway in adipogenesis. Result: High RhoA activity in the cell line C3H10T1/2 were generated (Named RhoA14V cells). Treatment of RhoA14V cells with Shield 1 following their differentiation into adipocytes resulted in the appearance of thick cortical actin filaments, and increased mRNA expression levels of RhoA, ROCK, p-MYPT1 and p-MLC, while PPARγ mRNA decreased. This resulted in decreased triglyceride synthesis and reduced expression of the adipogenic transcription factor PPARγ. These molecular changes were accompanied by reorganization of the actin cytoskeleton, during which ROCK signaling suppressed actin polymerization. Conclusion: ROCK signaling suppresses adipogenesis by controlling PPARγ expression and actin organization in adipocytes.
Collapse
Affiliation(s)
- Yuntao Ji
- School of Biology and Food Engineering, FuYang Normal University, Fuyang, Anhui 236041, China
| | - Meixia Cao
- School of Biology and Food Engineering, FuYang Normal University, Fuyang, Anhui 236041, China
| | - Jia Liu
- School of Biology and Food Engineering, FuYang Normal University, Fuyang, Anhui 236041, China
| | - Yanfei Chen
- School of Biology and Food Engineering, FuYang Normal University, Fuyang, Anhui 236041, China
| | - Xiaoli Li
- School of Biology and Food Engineering, FuYang Normal University, Fuyang, Anhui 236041, China
| | - Jing Zhao
- School of Biology and Food Engineering, FuYang Normal University, Fuyang, Anhui 236041, China
| | - Changqing Qu
- School of Biology and Food Engineering, FuYang Normal University, Fuyang, Anhui 236041, China
| |
Collapse
|
106
|
Duan W, Chen C, Haque M, Hayes D, Lopez MJ. Polymer-mineral scaffold augments in vivo equine multipotent stromal cell osteogenesis. Stem Cell Res Ther 2018. [PMID: 29523214 PMCID: PMC5845133 DOI: 10.1186/s13287-018-0790-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Use of bioscaffolds to direct osteogenic differentiation of adult multipotent stromal cells (MSCs) without exogenous proteins is a contemporary approach to bone regeneration. Identification of in vivo osteogenic contributions of exogenous MSCs on bioscaffolds after long-term implantation is vital to understanding cell persistence and effect duration. Methods This study was designed to quantify in vivo equine MSC osteogenesis on synthetic polymer scaffolds with distinct mineral combinations 9 weeks after implantation in a murine model. Cryopreserved, passage (P)1, equine bone marrow-derived MSCs (BMSC) and adipose tissue-derived MSCs (ASC) were culture expanded to P3 and immunophenotyped with flow cytometry. They were then loaded by spinner flask on to scaffolds composed of tricalcium phosphate (TCP)/hydroxyapatite (HA) (40:60; HT), polyethylene glycol (PEG)/poly-l-lactic acid (PLLA) (60:40; GA), or PEG/PLLA/TCP/HA (36:24:24:16; GT). Scaffolds with and without cells were maintained in static culture for up to 21 days or implanted subcutaneously in athymic mice that were radiographed every 3 weeks up to 9 weeks. In vitro cell viability and proliferation were determined. Explant composition (double-stranded (ds)DNA, collagen, sulfated glycosaminoglycan (sGAG), protein), equine and murine osteogenic target gene expression, microcomputed tomography (μCT) mineralization, and light microscopic structure were assessed. Results The ASC and BMSC number increased significantly in HT constructs between 7 and 21 days of culture, and BMSCs increased similarly in GT constructs. Radiographic opacity increased with time in GT-BMSC constructs. Extracellular matrix (ECM) components and dsDNA increased significantly in GT compared to HT constructs. Equine and murine osteogenic gene expression was highest in BMSC constructs with mineral-containing scaffolds. The HT constructs with either cell type had the highest mineral deposition based on μCT. Regardless of composition, scaffolds with cells had more ECM than those without, and osteoid was apparent in all BMSC constructs. Conclusions In this study, both exogenous and host MSCs appear to contribute to in vivo osteogenesis. Addition of mineral to polymer scaffolds enhances equine MSC osteogenesis over polymer alone, but pure mineral scaffold provides superior osteogenic support. These results emphasize the need for bioscaffolds that provide customized osteogenic direction of both exo- and endogenous MSCs for the best regenerative potential.
Collapse
Affiliation(s)
- Wei Duan
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA
| | - Cong Chen
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Masudul Haque
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA
| | - Daniel Hayes
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Mandi J Lopez
- Laboratory for Equine and Comparative Orthopedic Research, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
107
|
Morse A, Schindeler A, McDonald MM, Kneissel M, Kramer I, Little DG. Sclerostin Antibody Augments the Anabolic Bone Formation Response in a Mouse Model of Mechanical Tibial Loading. J Bone Miner Res 2018; 33:486-498. [PMID: 29090474 DOI: 10.1002/jbmr.3330] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/11/2017] [Accepted: 10/29/2017] [Indexed: 12/18/2022]
Abstract
Decreased activity or expression of sclerostin, an endogenous inhibitor of Wnt/β-catenin signaling, results in increased bone formation and mass. Antibodies targeting and neutralizing sclerostin (Scl-Ab) have been shown to increase bone mass and reduce fracture risk. Sclerostin is also important in modulating the response of bone to changes in its biomechanical environment. However, the effects of Scl-Ab on mechanotransduction are unclear, and it was speculated that the loading response may be altered for individuals receiving Scl-Ab therapy. To address this, we carried out a 2-week study of tibial cyclic compressive loading on C57Bl/6 mice treated with vehicle or 100 mg/kg/wk Scl-Ab. Increases in bone volume, density, and dynamic bone formation were found with loading, and the anabolic response was further increased by the combination of load and Scl-Ab. To investigate the underlying mechanism, gene profiling by RNA sequencing (RNAseq) was performed on tibias isolated from mice from all four experimental groups. Major alterations in Wnt/β-catenin gene expression were found with tibial loading, however not with Scl-Ab treatment alone. Notably, the combination of load and Scl-Ab elicited a synergistic response from a number of specific Wnt-related and mechanotransduction factors. An unexpected finding was significant upregulation of factors in the Rho GTPase signaling pathway with combination treatment. In summary, combination therapy had a more profound anabolic response than either Scl-Ab or loading treatment alone. The Wnt/β-catenin and Rho GTPase pathways were implicated within bone mechanotransduction and support the concept that bone mechanotransduction is likely to encompass a number of interconnected signaling pathways. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alyson Morse
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Michelle M McDonald
- Bone Biology Program, The Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | - David G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
108
|
Lin CW, Yang KC, Cheng NC, Tsai WB, Lou KL, Yu J. Evaluation of adhesion, proliferation, and differentiation of human adipose-derived stem cells on keratin. JOURNAL OF POLYMER RESEARCH 2018. [DOI: 10.1007/s10965-018-1446-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
109
|
Oliveira NM, Reis RL, Mano JF. Open Fluidics: A Cell Culture Flow System Developed Over Wettability Contrast-Based Chips. Adv Healthc Mater 2017; 6. [PMID: 29034587 DOI: 10.1002/adhm.201700638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 08/21/2017] [Indexed: 12/11/2022]
Abstract
Biological tissues are recurrently exposed to several dynamic mechanical forces that influence cell behavior. On this work, the focus is on the shear stress forces induced by fluid flow. The study of flow-induced effects on cells leads to important advances in cardiovascular, cancer, stem cell, and bone biology understanding. These studies are performed using cell culture flow (CCF) systems, mainly parallel plate flow chambers (PPFC), and microfluidic systems. Here, it is proposed an original CCF system based on the open fluidics concept. The system is developed using a planar superhydrophobic platform with hydrophilic paths. The paths work as channels to drive cell culture medium flows without using walls for liquid confinement. The liquid streams are controlled just based on the wettability contrast. To validate the concept, the effect of the shear stress stimulus in the osteogenic differentiation of C2C12 myoblast cells is studied. Combining bone morphogenic protein (specifically BMP-2) stimulation with this mechanical stimulus, a synergistic effect is found on osteoblast differentiation. This effect is confirmed by the enhancement of alkaline phosphatase activity, a well-known early marker of osteogenic differentiation. The suggested CCF system combines characteristics and advantages of both the PPFC and microfluidic systems.
Collapse
Affiliation(s)
- Nuno M. Oliveira
- 3B's Research Group - Biomaterials; Biodegradables and Biomimetics; University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; AvePark, Zona Industrial da Gandra; 4805-017 Barco GMR Portugal
- ICVS/3B's - PT Government Associate Laboratory; Braga/Guimarães 4710-057 Portugal
| | - Rui L. Reis
- 3B's Research Group - Biomaterials; Biodegradables and Biomimetics; University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; AvePark, Zona Industrial da Gandra; 4805-017 Barco GMR Portugal
- ICVS/3B's - PT Government Associate Laboratory; Braga/Guimarães 4710-057 Portugal
| | - João F. Mano
- 3B's Research Group - Biomaterials; Biodegradables and Biomimetics; University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; AvePark, Zona Industrial da Gandra; 4805-017 Barco GMR Portugal
- ICVS/3B's - PT Government Associate Laboratory; Braga/Guimarães 4710-057 Portugal
| |
Collapse
|
110
|
Carroll SF, Buckley CT, Kelly DJ. Cyclic Tensile Strain Can Play a Role in Directing both Intramembranous and Endochondral Ossification of Mesenchymal Stem Cells. Front Bioeng Biotechnol 2017; 5:73. [PMID: 29230389 PMCID: PMC5712005 DOI: 10.3389/fbioe.2017.00073] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/02/2017] [Indexed: 01/13/2023] Open
Abstract
Successfully regenerating damaged or diseased bone and other joint tissues will require a detailed understanding of how joint specific environmental cues regulate the fate of progenitor cells that are recruited or delivered to the site of injury. The goal of this study was to explore the role of cyclic tensile strain (CTS) in regulating the initiation of mesenchymal stem cell/multipotent stromal cell (MSC) differentiation, and specifically their progression along the endochondral pathway. To this end, we first explored the influence of CTS on the differentiation of MSCs in the absence of any specific growth factor, and secondly, we examined the influence of the long-term application of this mechanical stimulus on markers of endochondral ossification in MSCs maintained in chondrogenic culture conditions. A custom bioreactor was developed to apply uniaxial tensile deformation to bone marrow-derived MSCs encapsulated within physiological relevant 3D fibrin hydrogels. Mechanical loading, applied in the absence of soluble differentiation factors, was found to enhance the expression of both tenogenic (COL1A1) and osteogenic markers (BMP2, RUNX2, and ALPL), while suppressing markers of adipogenesis. No evidence of chondrogenesis was observed, suggesting that CTS can play a role in initiating direct intramembranous ossification. During long-term culture in the presence of a chondrogenic growth factor, CTS was shown to induce MSC re-organization and alignment, increase proteoglycan and collagen production, and to enhance the expression of markers associated with endochondral ossification (BMP2, RUNX2, ALPL, OPN, and COL10A1) in a strain magnitude-dependent manner. Taken together, these findings indicate that tensile loading may play a key role in promoting both intramembranous and endochondral ossification of MSCs in a context-dependent manner. In both cases, this loading-induced promotion of osteogenesis was correlated with an increase in the expression of the osteogenic growth factor BMP2. The results of this study demonstrate the potent role that extrinsic mechanical loading plays in guiding stem cell fate, which must be carefully considered when designing cell and tissue-engineering therapies if they are to realize their clinical potential.
Collapse
Affiliation(s)
- Simon F. Carroll
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Conor T. Buckley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
111
|
Sacco P, Furlani F, Cok M, Travan A, Borgogna M, Marsich E, Paoletti S, Donati I. Boric Acid Induced Transient Cross-Links in Lactose-Modified Chitosan (Chitlac). Biomacromolecules 2017; 18:4206-4213. [PMID: 29039653 DOI: 10.1021/acs.biomac.7b01237] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The present paper explores the effect of boric acid on Chitlac, a lactose-modified chitosan which had previously shown interesting biological and physical-chemical features. The herewith-reported experimental evidences demonstrated that boric acid binds to Chitlac, producing conformational and association effects on the chitosan derivative. The thermodynamics of boric acid binding to Chitlac was explored by means of 11B NMR, circular dichroism (CD), and UV-vis spectroscopy, while macromolecular effects were investigated by means of viscometry and dynamic light scattering (DLS). The experimental results revealed a chain-chain association when limited amounts of boric acid were added to Chitlac. However, upon exceeding a critical boric acid limit dependent on the polysaccharide concentration, the soluble aggregates disentangle. The rheological behavior of Chitlac upon treatment with boric acid was explored showing a dilatant behavior in conditions of steady flow. An uncommonly high dependence in the scaling law between the zero-shear viscosity and the concentration of Chitlac was found, i.e., η0 ∝ CCTL5.8, pointing to interesting potential implications of the present system in biomaterials development.
Collapse
Affiliation(s)
- Pasquale Sacco
- Department of Life Sciences, University of Trieste , Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Franco Furlani
- Department of Life Sciences, University of Trieste , Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Michela Cok
- Department of Life Sciences, University of Trieste , Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Andrea Travan
- Department of Life Sciences, University of Trieste , Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Massimiliano Borgogna
- Department of Life Sciences, University of Trieste , Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Eleonora Marsich
- Department of Medical, Surgical, and Health Sciences, University of Trieste , Piazza dell'Ospitale 1, I-34127 Trieste, Italy
| | - Sergio Paoletti
- Department of Life Sciences, University of Trieste , Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Ivan Donati
- Department of Life Sciences, University of Trieste , Via Licio Giorgieri 5, I-34127 Trieste, Italy
| |
Collapse
|
112
|
Gavazzo P, Petecchia L, Facci P, Vassalli M, Viti F. Controlled single-cell cyclic compression and transcription analysis: A pilot study. Biophys Chem 2017; 229:39-45. [DOI: 10.1016/j.bpc.2017.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/04/2017] [Accepted: 07/24/2017] [Indexed: 12/15/2022]
|
113
|
Kan C, Chen L, Hu Y, Lu H, Li Y, Kessler JA, Kan L. Microenvironmental factors that regulate mesenchymal stem cells: lessons learned from the study of heterotopic ossification. Histol Histopathol 2017; 32:977-985. [PMID: 28328009 PMCID: PMC5809774 DOI: 10.14670/hh-11-890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bone marrow contains a non-hematopoietic, clonogenic, multipotent population of stromal cells that are later called mesenchymal stem cells (MSC). Similar cells that share many common features with MSC are also found in other organs, which are thought to contribute both to normal tissue regeneration and to pathological processes such as heterotopic ossification (HO), the formation of ectopic bone in soft tissue. Understanding the microenvironmental factors that regulate MSC in vivo is essential both for understanding the biology of the stem cells and for effective translational applications of MSC. Unfortunately, this important aspect has been largely underappreciated. This review tries to raise the attention and highlight this critical issue by updating the relevant literature along with discussions of the key issues in the area.
Collapse
Affiliation(s)
- Chen Kan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lijun Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yangyang Hu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Haimei Lu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yuyun Li
- Department of Medical Laboratory Science, Bengbu Medical College, Bengbu, China
| | - John A Kessler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lixin Kan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Department of Medical Laboratory Science, Bengbu Medical College, Bengbu, China
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
114
|
McMichael BK, Jeong YH, Auerbach JA, Han CM, Sedlar R, Shettigar V, Bähler M, Agarwal S, Kim DG, Lee BS. The RhoGAP Myo9b Promotes Bone Growth by Mediating Osteoblastic Responsiveness to IGF-1. J Bone Miner Res 2017; 32:2103-2115. [PMID: 28585695 DOI: 10.1002/jbmr.3192] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 05/26/2017] [Accepted: 06/05/2017] [Indexed: 12/31/2022]
Abstract
The Ras homolog A (RhoA) subfamily of Rho guanosine triphosphatases (GTPases) regulates actin-based cellular functions in bone such as differentiation, migration, and mechanotransduction. Polymorphisms or genetic ablation of RHOA and some of its regulatory guanine exchange factors (GEFs) have been linked to poor bone health in humans and mice, but the effects of RhoA-specific GTPase-activating proteins (GAPs) on bone quality have not yet been identified. Therefore, we examined the consequences of RhoGAP Myo9b gene knockout on bone growth, phenotype, and cellular activity. Male and female mice lacking both alleles demonstrated growth retardation and decreased bone formation rates during early puberty. These mice had smaller, weaker bones by 4 weeks of age, but only female KOs had altered cellular numbers, with fewer osteoblasts and more osteoclasts. By 12 weeks of age, bone quality in KOs worsened. In contrast, 4-week-old heterozygotes demonstrated bone defects that resolved by 12 weeks of age. Throughout, Myo9b ablation affected females more than males. Osteoclast activity appeared unaffected. In primary osteogenic cells, Myo9b was distributed in stress fibers and focal adhesions, and its absence resulted in poor spreading and eventual detachment from culture dishes. Similarly, MC3T3-E1 preosteoblasts with transiently suppressed Myo9b levels spread poorly and contained decreased numbers of focal adhesions. These cells also demonstrated reduced ability to undergo IGF-1-induced spreading or chemotaxis toward IGF-1, though responses to PDGF and BMP-2 were unaffected. IGF-1 receptor (IGF1R) activation was normal in cells with diminished Myo9b levels, but the activated receptor was redistributed from stress fibers and focal adhesions into nuclei, potentially affecting receptor accessibility and gene expression. These results demonstrate that Myo9b regulates a subset of RhoA-activated processes necessary for IGF-1 responsiveness in osteogenic cells, and is critical for normal bone formation in growing mice. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Yong-Hoon Jeong
- College of Dentistry, The Ohio State University, Columbus, OH, USA
| | | | - Cheol-Min Han
- College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Ryan Sedlar
- College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Vikram Shettigar
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Martin Bähler
- Institut für Molekulare Zellbiologie, Universität Münster, Münster, Germany
| | - Sudha Agarwal
- College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Do-Gyoon Kim
- College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Beth S Lee
- College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
115
|
Islam A, Mbimba T, Younesi M, Akkus O. Effects of substrate stiffness on the tenoinduction of human mesenchymal stem cells. Acta Biomater 2017; 58:244-253. [PMID: 28602855 DOI: 10.1016/j.actbio.2017.05.058] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/02/2017] [Accepted: 05/31/2017] [Indexed: 12/29/2022]
Abstract
Extracellular matrix modulus plays an important role in regulating cell morphology, proliferation and differentiation during regular and diseased states. Although the effects of substrate topography and modulus on MSC differentiation are well known with respect to osteogenesis and adipogenesis, there has been relatively little investigation on the effects of this phenomenon on tenogenesis. Furthermore, relative roles of topographical factors (matrix alignment vs. matrix modulus) in inducing tenogenic differentiation is not well understood. In this study we investigated the effects of modulus and topographical alignment of type I collagen substrate on tendon differentiation. Type I collagen sheet substrates with random topographical alignment were fabricated with their moduli tuned in the range of 0.1, 1, 10 and 100MPa by using electrocompaction and controlled crosslinking. In one of the groups, topographical alignment was introduced at 10MPa stiffness, by controlled unidirectional stretching of the sheet. RT-PCR, immunohistochemistry and immunofluorescence results showed that mimicking the tendon topography, i.e. increasing the substrate modulus as well as alignment increased the tenogenic differentiation. Higher substrate modulus increased the expression of COLI, COLIII, COMP and TSP-4 about 2-3-fold and increased the production of COLI, COLIII and TSP-4 about 2-4-fold. Substrate alignment up regulated COLIII and COMP expression by 2-fold. Therefore, the tenoinductive collagen material model developed in this study can be used in the research and development of tissue engineering tendon repair constructs in future. STATEMENT OF SIGNIFICANCE Although the effects of substrate topography and modulus on MSC differentiation are well known with respect to osteogenesis and adipogenesis, there has been relatively little investigation on the effects of this phenomenon on tenogenesis. Furthermore, a relative role of topographical factors (matrix alignment vs. matrix modulus) in inducing tenogenic differentiation is not well understood. We investigated the effects of modulus and topographical alignment of type I collagen substrate on tendon differentiation. This study showed mimicking the tendon topography, i.e. increasing the substrate modulus as well as alignment increased the tenogenic differentiation. Therefore, the tenoinductive collagen material model developed in this study can be used in the research and development of tissue engineering tendon repair constructs in future.
Collapse
|
116
|
Barata D, Dias P, Wieringa P, van Blitterswijk C, Habibovic P. Cell-instructive high-resolution micropatterned polylactic acid surfaces. Biofabrication 2017; 9:035004. [PMID: 28671108 DOI: 10.1088/1758-5090/aa7d24] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Micro and nanoscale topographical structuring of biomaterial surfaces has been a valuable tool for influencing cell behavior, including cell attachment, proliferation and differentiation. However, most fabrication techniques for surface patterning of implantable biomaterials suffer from a limited resolution, not allowing controlled generation of sub-cellular three-dimensional features. Here, a direct laser lithography technique based on two-photon absorption was used to construct several patterns varying in size between 500 nm and 15 μm. Through replication via an intermediate mold, the patterns were transferred into polylactic acid (PLA), a widely used biomedical polymer, while retaining the original geometry. An osteoblast-like cell line, MG-63 was used for characterizing the morphological response to the topographical patterns. The results indicated that semi-continuous (dashed) lines, with a height of 1 μm were able to induce cell elongation in the direction of the lines. However, when dashes with a height of 0.5 μm were combined with perpendicularly crossing continuous lines (rails) with a height of 8 μm, the contact guidance effect of the dashes was lost and elongation of the cells was observed in the direction of the larger features. A second pattern, consisting of different arrays of pillars showed that, depending on the pillar height, the cells were either able to spread over the pattern or were confined between the pattern features. These differences in the ability of cells to spread further resulted in the formation of tension forces through stress fibers and displacement of vimentin. The method for high-resolution micropatterning of PLA as presented here can also be applied to other biomedical polymers, making it useful both for fundamental studies and for designing new biomaterials with improved functionality.
Collapse
Affiliation(s)
- David Barata
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Overijssel, Netherlands. Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Limburg, Netherlands
| | | | | | | | | |
Collapse
|
117
|
Mata A, Azevedo HS, Botto L, Gavara N, Su L. New Bioengineering Breakthroughs and Enabling Tools in Regenerative Medicine. CURRENT STEM CELL REPORTS 2017; 3:83-97. [PMID: 28596936 PMCID: PMC5445180 DOI: 10.1007/s40778-017-0081-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW In this review, we provide a general overview of recent bioengineering breakthroughs and enabling tools that are transforming the field of regenerative medicine (RM). We focus on five key areas that are evolving and increasingly interacting including mechanobiology, biomaterials and scaffolds, intracellular delivery strategies, imaging techniques, and computational and mathematical modeling. RECENT FINDINGS Mechanobiology plays an increasingly important role in tissue regeneration and design of therapies. This knowledge is aiding the design of more precise and effective biomaterials and scaffolds. Likewise, this enhanced precision is enabling ways to communicate with and stimulate cells down to their genome. Novel imaging technologies are permitting visualization and monitoring of all these events with increasing resolution from the research stages up to the clinic. Finally, algorithmic mining of data and soft matter physics and engineering are creating growing opportunities to predict biological scenarios, device performance, and therapeutic outcomes. SUMMARY We have found that the development of these areas is not only leading to revolutionary technological advances but also enabling a conceptual leap focused on targeting regenerative strategies in a holistic manner. This approach is bringing us ever more closer to the reality of personalized and precise RM.
Collapse
Affiliation(s)
- Alvaro Mata
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary University of London, London, E1 4NS UK
| | - Helena S. Azevedo
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary University of London, London, E1 4NS UK
| | - Lorenzo Botto
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary University of London, London, E1 4NS UK
| | - Nuria Gavara
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary University of London, London, E1 4NS UK
| | - Lei Su
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary University of London, London, E1 4NS UK
| |
Collapse
|
118
|
Camarero-Espinosa S, Cooper-White J. Tailoring biomaterial scaffolds for osteochondral repair. Int J Pharm 2017; 523:476-489. [DOI: 10.1016/j.ijpharm.2016.10.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/11/2016] [Accepted: 10/17/2016] [Indexed: 12/11/2022]
|
119
|
Malfroy Camine V, Terrier A, Pioletti DP. Micromotion-induced peri-prosthetic fluid flow around a cementless femoral stem. Comput Methods Biomech Biomed Engin 2017; 20:730-736. [PMID: 28271719 DOI: 10.1080/10255842.2017.1296954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Micromotion-induced interstitial fluid flow at the bone-implant interface has been proposed to play an important role in aseptic loosening of cementless implants. High fluid velocities are thought to promote aseptic loosening through activation of osteoclasts, shear stress induced control of mesenchymal stem cells differentiation, or transport of molecules. In this study, our objectives were to characterize and quantify micromotion-induced fluid flow around a cementless femoral stem using finite element modeling. With a 2D model of the bone-implant interface and full-factorial design, we first evaluated the relative influence of material properties, and bone-implant micromotion and gap on fluid velocity. Transverse sections around a femoral stem were built from computed tomography images, while boundary conditions were obtained from experimental measurements on the same femur. In a second step, a 3D model was built from the same data-set to estimate the shear stress experienced by cells hosted in the peri-implant tissues. The full-factorial design analysis showed that local micromotion had the most influence on peak fluid velocity at the interface. Remarkable variations in fluid velocity were observed in the macrostructures at the surface of the implant in the 2D transverse sections of the stem. The 3D model predicted peak fluid velocities extending up to 2.2 mm/s in the granulation tissue and to 3.9 mm/s in the trabecular bone. Peak shear stresses on the cells hosted in these tissues ranged from 0.1 to 12.5 Pa. These results offer insight into mechanical stimuli encountered at the bone-implant interface.
Collapse
Affiliation(s)
- Valérie Malfroy Camine
- a Laboratory of Biomechanical Orthopedics , Ecole Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| | - Alexandre Terrier
- a Laboratory of Biomechanical Orthopedics , Ecole Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| | - Dominique P Pioletti
- a Laboratory of Biomechanical Orthopedics , Ecole Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| |
Collapse
|
120
|
Stavenschi E, Labour MN, Hoey DA. Oscillatory fluid flow induces the osteogenic lineage commitment of mesenchymal stem cells: The effect of shear stress magnitude, frequency, and duration. J Biomech 2017; 55:99-106. [PMID: 28256244 DOI: 10.1016/j.jbiomech.2017.02.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/28/2016] [Accepted: 02/11/2017] [Indexed: 01/12/2023]
Abstract
A potent regulator of bone anabolism is physical loading. However, it is currently unclear whether physical stimuli such as fluid shear within the marrow cavity is sufficient to directly drive the osteogenic lineage commitment of resident mesenchymal stem cells (MSC). Therefore, the objective of the study is to employ a systematic analysis of oscillatory fluid flow (OFF) parameters predicted to occur in vivo on early MSC osteogenic responses and late stage lineage commitment. MSCs were exposed to OFF of 1Pa, 2Pa and 5Pa magnitudes at frequencies of 0.5Hz, 1Hz and 2Hz for 1h, 2h and 4h of stimulation. Our findings demonstrate that OFF elicits a positive osteogenic response in MSCs in a shear stress magnitude, frequency, and duration dependent manner that is gene specific. Based on the mRNA expression of osteogenic markers Cox2, Runx2 and Opn after short-term fluid flow stimulation, we identified that a regime of 2Pa shear magnitude and 2Hz frequency induces the most robust and reliable upregulation in osteogenic gene expression. Furthermore, long-term mechanical stimulation utilising this regime, elicits a significant increase in collagen and mineral deposition when compared to static control demonstrating that mechanical stimuli predicted within the marrow is sufficient to directly drive osteogenesis.
Collapse
Affiliation(s)
- Elena Stavenschi
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Dept. of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Marie-Noelle Labour
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Dept. of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - David A Hoey
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Dept. of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland; Dept. of Mechanical, Aeronautical and Biomedical Engineering, University of Limerick, Ireland; Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, Dublin 2, Ireland.
| |
Collapse
|
121
|
Abstract
The primary cilium is a mechanosensor in a variety of mammalian cell types, initiating and directing intracellular signalling cascades in response to external stimuli. When primary cilia formation is disrupted, cells have diminished mechanosensitivity and an abrogated response to mechanical stimulation. Due to this important role, we hypothesised that increasing primary cilia length would enhance the downstream response and therefore, mechanosensitivity. To test this hypothesis, we increased osteocyte primary cilia length with fenoldopam and lithium and found that cells with longer primary cilia were more mechanosensitive. Furthermore, fenoldopam treatment potentiated adenylyl cyclase activity and was able to recover primary cilia form and sensitivity in cells with impaired cilia. This work demonstrates that modulating the structure of the primary cilium directly impacts cellular mechanosensitivity. Our results implicate cilium length as a potential therapeutic target for combating numerous conditions characterised by impaired cilia function.
Collapse
|
122
|
Zhuoli Z, Ling Z, Ruiyang M, Xueqi G. [Influences of vibration on rapid osteogenic response of osteoblasts]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2017; 35:68-72. [PMID: 28326730 DOI: 10.7518/hxkq.2017.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study investigated the rapid response of osteoblasts, which were derived from low-magnitude high-frequency vibration (LMHFV). Refractory period-derived memory response was also observed. METHODS MC3T3-E1 cells were incubated and received LMHFV stimulation (0.49 g, 40 Hz) for 30 min. After application of LMHFV, mRNA levels of earlier osteogenic differentiation markers Runt-related transcription factor 2 (Runx2), collagen typeⅠ(Col-Ⅰ), and alkaline phosphatase (ALP) were immediately detected by real-time fluorescence quantitative polymerase chain reaction in the absence or presence of antioxidant. Simultaneously, concentrations of mitochondrial reactive oxygen species (ROS) and average mitochondrial length were also measured. RESULTS Osteoblasts in the vibration group showed decreased gene expressions of Runx2, Col-Ⅰ, and ALP (P<0.01) and increased levels of mitochondrial ROS (P<0.01) and shortened mitochondria (P<0.01), whereas antioxidant treatment resulted in recovery from changes in the above indicators (P<0.01). CONCLUSIONS LMHFV can downregulate mRNA levels of early osteogenic differentiation markers, promote ROS generation, and mitochondrial fission.
.
Collapse
Affiliation(s)
- Zhu Zhuoli
- State Key Laboratory of Oral Diseases, Dept. of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhang Ling
- State Key Laboratory of Oral Diseases, Dept. of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ma Ruiyang
- State Key Laboratory of Oral Diseases, Dept. of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Gan Xueqi
- State Key Laboratory of Oral Diseases, Dept. of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
123
|
Ye Y, Jing X, Li N, Wu Y, Li B, Xu T. Icariin promotes proliferation and osteogenic differentiation of rat adipose-derived stem cells by activating the RhoA-TAZ signaling pathway. Biomed Pharmacother 2017; 88:384-394. [PMID: 28122303 DOI: 10.1016/j.biopha.2017.01.075] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/02/2017] [Accepted: 01/12/2017] [Indexed: 12/18/2022] Open
Abstract
Icariin, the main active flavonoid glucoside isolated from Herba epimedii, has been demonstrated to be a potential alternative therapy to prevent postmenopausal osteoporosis. Icariin has also been shown to regulate the proliferation and osteogenic differentiation of rat bone marrow stromal cells (rBMSCs). However, the detailed molecular mechanism of icariin has remained largely unknown. Besides, no investigation has focused on the relevance of icariin in the regulation of rat adipose-derived stem cells (rASCs) proliferation and osteogenic differentiation. In the present study, we detected that icariin promotes proliferation and osteogenic differentiation of rASCs in a concentration range from 10-8M to 10-6M, with 10-7M to be the optimal concentration. We found that 10-7M icariin significantly increased active RhoA protein expression and ROCK substrate molecule p-MYPT1 expression in rASCs. C3 (the RhoA inhibitor) treatment abrogated the increased proliferation and osteogenic differentiation of rASCs induced by icariin. Interestingly, we also found that C3 abrogated the activation of TAZ induced by icariin. Depletion of TAZ by siRNA transfection significantly blocked icariin promoted proliferation and osteogenic differentiation of rASCs. However, icariin induced active RhoA protein expression was not affected by TAZ specific siRNA transfection, suggesting that RhoA lies upstream of TAZ. Taken together, our data indicate that icariin promotes proliferation and osteogenic differentiation of rASCs by activating the RhoA-TAZ signaling pathway.
Collapse
Affiliation(s)
- Yaping Ye
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xingzhi Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Na Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yingxing Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bingbing Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
124
|
Takeshita N, Hasegawa M, Sasaki K, Seki D, Seiryu M, Miyashita S, Takano I, Oyanagi T, Miyajima Y, Takano-Yamamoto T. In vivo expression and regulation of genes associated with vascularization during early response of sutures to tensile force. J Bone Miner Metab 2017; 35:40-51. [PMID: 26825658 DOI: 10.1007/s00774-016-0737-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 01/04/2016] [Indexed: 12/14/2022]
Abstract
Sutures are fibrous tissues that connect bones in craniofacial skeletal complexes. Cranio- and dentofacial skeletal deformities in infant and adolescent patients can be treated by applying tensile force to sutures to induce sutural bone formation. The early gene expression induced by mechanical stress is essential for bone formation in long bones; however, early gene expression during sutural bone formation induced by tensile force is poorly characterized. In vivo studies are essential to evaluate molecular responses to mechanical stresses in heterogeneous cell populations, such as sutures. In this paper we examined in vivo early gene expression and the underlying regulatory mechanism for this expression in tensile-force-applied cranial sutures, focusing on genes involved in vascularization. Tensile force upregulated expression of vascular factors, such as vascular endothelial growth factor (Vegf) and endothelial cell markers, in sutures within 3 h. The expression of connective tissue growth factor (Ctgf) and Rho-associated coiled-coil containing protein kinase 2 (Rock2) was also upregulated by tensile force. A CTGF-neutralizing antibody and the ROCK inhibitor, Y-27632, abolished tensile-force-induced Vegf expression. Moreover, tensile force activated extracellular signal-related kinase 1/2 (ERK1/2) signaling in sagittal sutures, and the ERK1/2 inhibitor, U0126, partially inhibited tensile-force-induced Ctgf expression. These results indicate that tensile force induces in vivo gene expression associated with vascularization early in tensile-force-induced sutural bone formation. Moreover, the early induction of Vegf gene expression is regulated by CTGF and ROCK2.
Collapse
Affiliation(s)
- Nobuo Takeshita
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Masakazu Hasegawa
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Kiyo Sasaki
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Daisuke Seki
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Masahiro Seiryu
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Shunro Miyashita
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Ikuko Takano
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Toshihito Oyanagi
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yuki Miyajima
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Teruko Takano-Yamamoto
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
125
|
Chen D, Sarkar S, Candia J, Florczyk SJ, Bodhak S, Driscoll MK, Simon CG, Dunkers JP, Losert W. Machine learning based methodology to identify cell shape phenotypes associated with microenvironmental cues. Biomaterials 2016; 104:104-18. [PMID: 27449947 PMCID: PMC11305428 DOI: 10.1016/j.biomaterials.2016.06.040] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/17/2016] [Accepted: 06/19/2016] [Indexed: 01/02/2023]
Abstract
Cell morphology has been identified as a potential indicator of stem cell response to biomaterials. However, determination of cell shape phenotype in biomaterials is complicated by heterogeneous cell populations, microenvironment heterogeneity, and multi-parametric definitions of cell morphology. To associate cell morphology with cell-material interactions, we developed a shape phenotyping framework based on support vector machines. A feature selection procedure was implemented to select the most significant combination of cell shape metrics to build classifiers with both accuracy and stability to identify and predict microenvironment-driven morphological differences in heterogeneous cell populations. The analysis was conducted at a multi-cell level, where a "supercell" method used average shape measurements of small groups of single cells to account for heterogeneous populations and microenvironment. A subsampling validation algorithm revealed the range of supercell sizes and sample sizes needed for classifier stability and generalization capability. As an example, the responses of human bone marrow stromal cells (hBMSCs) to fibrous vs flat microenvironments were compared on day 1. Our analysis showed that 57 cells (grouped into supercells of size 4) are the minimum needed for phenotyping. The analysis identified that a combination of minor axis length, solidity, and mean negative curvature were the strongest early shape-based indicator of hBMSCs response to fibrous microenvironment.
Collapse
Affiliation(s)
- Desu Chen
- Biophysics Program, University of Maryland, College Park, MD, United States
| | - Sumona Sarkar
- Biosystems & Biomaterials Division, National Institute of Standards & Technology, Gaithersburg, MD, United States
| | - Julián Candia
- Department of Physics, University of Maryland, College Park, MD, United States; School of Medicine, University of Maryland, Baltimore, MD, United States; Center for Human Immunology, National Institutes of Health, Bethesda, MD, United States
| | - Stephen J Florczyk
- Biosystems & Biomaterials Division, National Institute of Standards & Technology, Gaithersburg, MD, United States
| | - Subhadip Bodhak
- Biosystems & Biomaterials Division, National Institute of Standards & Technology, Gaithersburg, MD, United States
| | - Meghan K Driscoll
- Department of Physics, University of Maryland, College Park, MD, United States
| | - Carl G Simon
- Biosystems & Biomaterials Division, National Institute of Standards & Technology, Gaithersburg, MD, United States
| | - Joy P Dunkers
- Biosystems & Biomaterials Division, National Institute of Standards & Technology, Gaithersburg, MD, United States
| | - Wolfgang Losert
- Department of Physics, University of Maryland, College Park, MD, United States.
| |
Collapse
|
126
|
The molecular basis of bone mechanotransduction. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2016; 16:221-36. [PMID: 27609037 PMCID: PMC5114345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The skeleton has the ability to perfectly adapt to external forces of the operating environment, by altering its morphology and metabolism in order to meet different needs. This unique adaptive capacity of the skeleton creates an interesting range of biological questions concerning the perception of mechanical or other kinds of signals, the type of receptor, and the molecular pathways involved in this adaptation. Studies of the characteristics of the cellular engineering provide a host of new information that confers to osteocytes the role of the protagonist in the perception and regulation of mechanical effects on the skeleton. The identity of mechanoreceptors is manifold and concerns ion channels, integrins, cell membrane, the cytoskeleton, and other systems. A similar multiplicity characterizes the intracellular signaling. This review describes recent data concerning the outward force reception systems and intracellular transduction pathways of information transfer leading to the continuous adaptation of bone tissue. Increased appreciation of the importance of the mechanical environment in regulating and determining the effectiveness of structural adjustment of the skeleton defines new horizons for the discovery of novel therapeutic approaches to diseases associated with bone loss.
Collapse
|
127
|
Yang K, Wu Y, Cheng P, Zhang J, Yang C, Pi B, Ye Y, You H, Chen A, Xu T, Guo F, Qi J. YAP and ERK mediated mechanical strain-induced cell cycle progression through RhoA and cytoskeletal dynamics in rat growth plate chondrocytes. J Orthop Res 2016; 34:1121-9. [PMID: 26694636 DOI: 10.1002/jor.23138] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 12/14/2015] [Indexed: 02/04/2023]
Abstract
Yes-associated protein (YAP) and extracellular signal-regulated kinase (ERK) have been considered as key regulators in tissue homeostasis, organ development, and tumor formation. However, the roles of YAP and ERK in the mediating strain mechanosensing in the growth plate cartilage have not been determined. In this study, chondrocytes obtained from the growth plate cartilage of 2-week-old Sprague-Dawley rats were subjected to the mechanical strain with different magnitudes and durations at a frequency of 0.5 Hz. We found that YAP and ERK activation in response to mechanical strain was time and magnitude dependent. Pretreatment with a RhoA inhibitor (C3 toxin) or a microfilament cytoskeleton disrupting reagent (cytochalasin D) could suppress their activation. In addition, activated YAP and ERK were able to induce cell cycle progression by up-regulating the expression of cell cycle-related genes. These results shed new light on the function of YAP and ERK in mechanical strain-promoted growth plate development. Our results also provided evidence that RhoA and cytoskeletal dynamics are required for this mechanotransduction. © 2015 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1121-1129, 2016.
Collapse
Affiliation(s)
- Kaixiang Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, P. R. China.,Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, Jiangsu Province, P. R. China
| | - Yingxing Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, P. R. China
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, P. R. China
| | - Jinming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, P. R. China
| | - Chengyuan Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, Jiangsu Province, P. R. China
| | - Bin Pi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, Jiangsu Province, P. R. China
| | - Yaping Ye
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, P. R. China
| | - Hongbo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, P. R. China
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, P. R. China
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, P. R. China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, P. R. China
| | - Jun Qi
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, P. R. China
| |
Collapse
|
128
|
Bo Z, Bin G, Jing W, Cuifang W, Liping A, Jinglin M, Jin J, Xiaoyi T, Cong C, Ning D, Yayi X. Fluid shear stress promotes osteoblast proliferation via the Gαq-ERK5 signaling pathway. Connect Tissue Res 2016; 57:299-306. [PMID: 27115838 DOI: 10.1080/03008207.2016.1181063] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Fluid shear stress (FSS) is a ubiquitous mechanical stimulus that potently promotes osteoblast proliferation. Previously, we reported that extracellular signal-regulated kinase 5 (ERK5) is essential for FSS-induced osteoblast proliferation. However, the precise mechanism by which FSS promotes osteoblast proliferation via ERK5 activation is poorly understood. The aim of this study was to determine the critical role of Gαq in FSS-induced ERK5 phosphorylation and osteoblast proliferation, as well as the downstream targets of the Gαq-ERK5 pathway. MC3T3-E1 cells were transfected with 50 nM Gαq siRNA, treated with 5 mM XMD8-92 (a highly selective inhibitor of ERK5 activity), and/or exposed to FSS (12 dyn/cm(2)). Cell proliferation was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The protein expression levels of Gαq, P-ERK5, ERK5, Cyclin B1, and CDK1 were analyzed by Western blot. Physiological FSS exposure for 60 min remarkably promoted MC3T3-E1 cell proliferation, however, this effect was suppressed by siRNA-mediated Gαq knockdown or inhibition of ERK5 activity by XMD8-92 treatment, suggesting that Gαq and ERK5 might modulate FSS-increased osteoblast proliferation. Furthermore, ERK5 phosphorylation was dramatically inhibited by Gαq siRNA. In addition, our study further revealed that FSS treatment of MC3T3-E1 cells for 60 min markedly upregulated the protein expression levels of Cyclin B1 and CDK1, and this increased expression was predominantly blocked by Gαq siRNA or XMD8-92 treatment. We propose that FSS acts on the Gαq-ERK5 signaling pathway to upregulate Cyclin B1 and CDK1 expression, thereby resulting in MC3T3-E1 cell proliferation. Thus, the Gαq-ERK5 signaling pathway may provide useful information regarding the treatment of bone metabolic disease.
Collapse
Affiliation(s)
- Zhang Bo
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Geng Bin
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Wang Jing
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Wang Cuifang
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - An Liping
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Ma Jinglin
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Jiang Jin
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Tan Xiaoyi
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Chen Cong
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Ding Ning
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Xia Yayi
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| |
Collapse
|
129
|
Abstract
Mechanical loading is a potent anabolic regulator of bone mass, and the first line of defense for bone loss is weight-bearing exercise. Likewise, protected weight bearing is the first prescribed physical therapy following orthopedic reconstructive surgery. In both cases, enhancement of new bone formation is the goal. Our understanding of the physical cues, mechanisms of force sensation, and the subsequent cellular response will help identify novel physical and therapeutic treatments for age- and disuse-related bone loss, delayed- and nonunion fractures, and significant bony defects. This review highlights important new insights into the principles and mechanisms governing mechanical adaptation of the skeleton during homeostasis and repair and ends with a summary of clinical implications stemming from our current understanding of how bone adapts to biophysical force.
Collapse
|
130
|
Hoon JL, Tan MH, Koh CG. The Regulation of Cellular Responses to Mechanical Cues by Rho GTPases. Cells 2016; 5:cells5020017. [PMID: 27058559 PMCID: PMC4931666 DOI: 10.3390/cells5020017] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/21/2022] Open
Abstract
The Rho GTPases regulate many cellular signaling cascades that modulate cell motility, migration, morphology and cell division. A large body of work has now delineated the biochemical cues and pathways, which stimulate the GTPases and their downstream effectors. However, cells also respond exquisitely to biophysical and mechanical cues such as stiffness and topography of the extracellular matrix that profoundly influence cell migration, proliferation and differentiation. As these cellular responses are mediated by the actin cytoskeleton, an involvement of Rho GTPases in the transduction of such cues is not unexpected. In this review, we discuss an emerging role of Rho GTPase proteins in the regulation of the responses elicited by biophysical and mechanical stimuli.
Collapse
Affiliation(s)
- Jing Ling Hoon
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
| | - Mei Hua Tan
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
| | - Cheng-Gee Koh
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
- Mechanobiology Institute, Singapore 117411, Singapore.
| |
Collapse
|
131
|
Ghaemi RV, Vahidi B, Sabour MH, Haghighipour N, Alihemmati Z. Fluid-Structure Interactions Analysis of Shear-Induced Modulation of a Mesenchymal Stem Cell: An Image-Based Study. Artif Organs 2016; 40:278-287. [PMID: 26333040 DOI: 10.1111/aor.12547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although effects of biochemical modulation of stem cells have been widely investigated, only recent advances have been made in the identification of mechanical conditioning on cell signaling pathways. Experimental investigations quantifying the micromechanical environment of mesenchymal stem cells (MSCs) are challenging while computational approaches can predict their behavior due to in vitro stimulations. This study introduces a 3D cell-specific finite element model simulating large deformations of MSCs. Here emphasizing cell mechanical modulation which represents the most challenging multiphysics phenomena in sub-cellular level, we focused on an approach attempting to elicit unique responses of a cell under fluid flow. Fluorescent staining of MSCs was performed in order to visualize the MSC morphology and develop a geometrically accurate model of it based on a confocal 3D image. We developed a 3D model of a cell fixed in a microchannel under fluid flow and then solved the numerical model by fluid-structure interactions method. By imposing flow characteristics representative of vigorous in vitro conditions, the model predicts that the employed external flow induces significant localized effective stress in the nucleo-cytoplasmic interface and average cell deformation of about 40%. Moreover, it can be concluded that a lower strain level is made in the cell by the oscillatory flow as compared with steady flow, while same ranges of effective stress are recorded inside the cell in both conditions. The deeper understanding provided by this study is beneficial for better design of single cell in vitro studies.
Collapse
Affiliation(s)
- Roza Vaez Ghaemi
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
- Department of Chemical and Biological Engineering, Faculty of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Bahman Vahidi
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mohammad Hossein Sabour
- Department of Aerospace Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | | | - Zakieh Alihemmati
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| |
Collapse
|
132
|
Sonam S, Sathe SR, Yim EKF, Sheetz MP, Lim CT. Cell contractility arising from topography and shear flow determines human mesenchymal stem cell fate. Sci Rep 2016; 6:20415. [PMID: 26879739 PMCID: PMC4754642 DOI: 10.1038/srep20415] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/06/2016] [Indexed: 01/13/2023] Open
Abstract
Extracellular matrix (ECM) of the human Mesenchymal Stem Cells (MSCs) influences intracellular tension and is known to regulate stem cell fate. However, little is known about the physiological conditions in the bone marrow, where external forces such as fluid shear stress, apart from the physical characteristics of the ECM, influence stem cell response. Here, we hypothesize that substrate topography and fluid shear stress alter the cellular contractile forces, influence the genetic expression of the stem cells and hence alter their lineage. When fluid shear stress was applied, human MSCs with higher contractility (seeded on 1 μm wells) underwent osteogenesis, whereas those with lower contractility (seeded on 2 μm gratings) remained multipotent. Compared to human MSCs seeded on gratings, those seeded on wells exhibited altered alignment and an increase in the area and number of focal adhesions. When actomyosin contractility was inhibited, human MSCs did not exhibit differentiation, regardless of the topographical feature they were being cultured on. We conclude that the stresses generated by the applied fluid flow impinge on cell contractility to drive the stem cell differentiation via the contractility of the stem cells.
Collapse
Affiliation(s)
- Surabhi Sonam
- Mechanobiology Institute, National University of Singapore, 117411, Singapore.,Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Sharvari R Sathe
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Evelyn K F Yim
- Mechanobiology Institute, National University of Singapore, 117411, Singapore.,Department of Biomedical Engineering, National University of Singapore, 117583, Singapore.,Department of Surgery, National University of Singapore, 119228, Singapore
| | - Michael P Sheetz
- Mechanobiology Institute, National University of Singapore, 117411, Singapore.,Department of Biological Sciences, Columbia University, New York, 10027, USA
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, 117411, Singapore.,Department of Biomedical Engineering, National University of Singapore, 117583, Singapore.,Department of Mechanical Engineering, National University of Singapore, 117575, Singapore
| |
Collapse
|
133
|
Brown GN, Sattler RL, Guo XE. Experimental studies of bone mechanoadaptation: bridging in vitro and in vivo studies with multiscale systems. Interface Focus 2016; 6:20150071. [PMID: 26855756 DOI: 10.1098/rsfs.2015.0071] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Despite advancements in technology and science over the last century, the mechanisms underlying Wolff's law-bone structure adaptation in response to physical stimuli-remain poorly understood, limiting the ability to effectively treat and prevent skeletal diseases. A challenge to overcome in the study of the underlying mechanisms of this principle is the multiscale nature of mechanoadaptation. While there exist in silico systems that are capable of studying across these scales, experimental studies are typically limited to interpretation at a single dimension or time point. For instance, studies of single-cell responses to defined physical stimuli offer only a limited prediction of the whole bone response, while overlapping pathways or compensatory mechanisms complicate the ability to isolate critical targets in a whole animal model. Thus, there exists a need to develop experimental systems capable of bridging traditional experimental approaches and informing existing multiscale theoretical models. The purpose of this article is to review the process of mechanoadaptation and inherent challenges in studying its underlying mechanisms, discuss the limitations of traditional experimental systems in capturing the many facets of this process and highlight three multiscale experimental systems which bridge traditional approaches and cover relatively understudied time and length scales in bone adaptation.
Collapse
Affiliation(s)
- Genevieve N Brown
- Bone Bioengineering Laboratory, Department of Biomedical Engineering , Columbia University , New York, NY 10027 , USA
| | - Rachel L Sattler
- Bone Bioengineering Laboratory, Department of Biomedical Engineering , Columbia University , New York, NY 10027 , USA
| | - X Edward Guo
- Bone Bioengineering Laboratory, Department of Biomedical Engineering , Columbia University , New York, NY 10027 , USA
| |
Collapse
|
134
|
Singh RK, Awasthi S, Dhayalan A, Ferreira JMF, Kannan S. Deposition, structure, physical and invitro characteristics of Ag-doped β-Ca3(PO4)2/chitosan hybrid composite coatings on Titanium metal. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 62:692-701. [PMID: 26952474 DOI: 10.1016/j.msec.2016.02.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 01/28/2016] [Accepted: 02/03/2016] [Indexed: 01/06/2023]
Abstract
Pure and five silver-doped (0-5Ag) β-tricalcium phosphate [β-TCP, β-Ca3(PO4)2]/chitosan composite coatings were deposited on Titanium (Ti) substrates and their properties that are relevant for applications in hard tissue replacements were assessed. Silver, β-TCP and chitosan were combined to profit from their salient and complementary antibacterial and biocompatible features.The β-Ca3(PO4)2 powders were synthesized by co-precipitation. The characterization results confirmed the Ag(+) occupancy at the crystal lattice of β-Ca3(PO4)2. The Ag-dopedβ-Ca3(PO4)2/chitosan composite coatings deposited by electrophoresis showed good antibacterial activity and exhibited negative cytotoxic effects towards the human osteosarcoma cell line MG-63. The morphology of the coatings was observed by SEM and their efficiency against corrosion of metallic substrates was determined through potentiodynamic polarization tests.
Collapse
Affiliation(s)
- Ram Kishore Singh
- Centre for Nanoscience and Technology, Pondicherry University, Puducherry605 014, India
| | - Sharad Awasthi
- Department of Biotechnology, Pondicherry University, Puducherry605 014, India
| | - Arunkumar Dhayalan
- Department of Biotechnology, Pondicherry University, Puducherry605 014, India
| | - J M F Ferreira
- Department of Materials and Ceramics Engineering, University of Aveiro, CICECO, Aveiro3810 193, Portugal
| | - S Kannan
- Centre for Nanoscience and Technology, Pondicherry University, Puducherry605 014, India.
| |
Collapse
|
135
|
Chua ILS, Kim HW, Lee JH. Signaling of extracellular matrices for tissue regeneration and therapeutics. Tissue Eng Regen Med 2016; 13:1-12. [PMID: 30603379 DOI: 10.1007/s13770-016-9075-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/18/2015] [Accepted: 11/02/2015] [Indexed: 12/17/2022] Open
Abstract
Cells receive important regulatory signals from their extracellular matrix (ECM) and the physical property of the ECM regulates important cellular behaviors like cell proliferation, migration and differentiation. A large part of tissue formation and regeneration depends on cellular interaction with its ECM. A comprehensive understanding of the mechanistic biochemical pathway of the ECM components is necessary for the design of a biomaterial scaffold for tissue engineering. Depending on the type of tissue, the ECM requirement might be different and this would influence its downstream intracellular cell signaling. Here, we reviewed the ECM and its signaling pathway by discussing: 1) classification of the ECM into hard, elastic and soft tissue based on its physical properties, 2) proliferation and differentiation control of the ECM, 3) roles of membrane receptor and its intracellular regulation factor, 4) ECM remodeling via inside-out signaling. By providing a comprehensive overview of the ECM's role in mechanotransduction and the self-regulatory effect of cells back on the ECM, we hope to provide a better insight of the physical and biochemical cues from the ECM. A sound understanding on the in vivo ECM has implication on the choice of materials and surface coating of biomimetic scaffolds used for tissue regeneration and therapeutics in a cell-free scaffold.
Collapse
Affiliation(s)
- Ing Loon Sean Chua
- 1Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore City, Singapore
| | - Hae-Won Kim
- 2Department of Nanobiomedical Sciences and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea.,3Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Korea.,4Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Korea
| | - Jae Ho Lee
- 1Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore City, Singapore.,2Department of Nanobiomedical Sciences and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea.,3Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Korea
| |
Collapse
|
136
|
Reactive oxygen species regulatory mechanisms associated with rapid response of MC3T3-E1 cells for vibration stress. Biochem Biophys Res Commun 2016; 470:510-515. [DOI: 10.1016/j.bbrc.2016.01.120] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 01/20/2016] [Indexed: 01/15/2023]
|
137
|
Puts R, Ruschke K, Ambrosi TH, Kadow-Romacker A, Knaus P, Jenderka KV, Raum K. A Focused Low-Intensity Pulsed Ultrasound (FLIPUS) System for Cell Stimulation: Physical and Biological Proof of Principle. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2016; 63:91-100. [PMID: 26552085 DOI: 10.1109/tuffc.2015.2498042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Quantitative ultrasound (QUS) is a promising technique for bone tissue evaluation. Highly focused transducers used for QUS also have the capability to be applied for tissue-regenerative purposes and can provide spatially limited deposition of acoustic energy. We describe a focused low-intensity pulsed ultrasound (FLIPUS) system, which has been developed for the stimulation of cell monolayers in the defocused far field of the transducer through the bottom of the well plate. Tissue culture well plates, carrying the cells, were incubated in a special chamber, immersed in a temperature-controlled water tank. A stimulation frequency of 3.6 MHz provided an optimal sound transmission through the polystyrene well plate. The ultrasound was pulsed for 20 min daily at 100-Hz repetition frequency with 27.8% duty cycle. The calibrated output intensity corresponded to I(SATA) = 44.5 ± 7.1 mW/cm2, which is comparable to the most frequently reported nominal output levels in LIPUS studies. No temperature change by the ultrasound exposure was observed in the well plate. The system was used to stimulate rat mesenchymal stem cells (rMSCs). The applied intensity had no apoptotic effect and enhanced the expression of osteogenic markers, i.e., osteopontin (OPN), collagen 1 (Col-1), the osteoblast-specific transcription factor-Runx-2 and E11 protein, an early osteocyte marker, in stimulated cells on day 5. The proposed FLIPUS setup opens new perspectives for the evaluation of the mechanistic effects of LIPUS.
Collapse
|
138
|
Seo JH, Hirata M, Kakinoki S, Yamaoka T, Yui N. Dynamic polyrotaxane-coated surface for effective differentiation of mouse induced pluripotent stem cells into cardiomyocytes. RSC Adv 2016. [DOI: 10.1039/c6ra03967g] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Increasing molecular mobility of hydrated polyrotaxane (PRX)-coated surfaces was effective to promote the differentiation of mouse induced pluripotent stem cells (iPS cells) into cardiomyocytes.
Collapse
Affiliation(s)
- Ji-Hun Seo
- Institute of Biomaterials and Bioengineering
- Tokyo Medical and Dental University
- Tokyo 101-0062
- Japan
- Department of Materials Science and Engineering
| | - Mitsuhi Hirata
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center Research Institute
- Osaka 565-8565
- Japan
- JST-CREST
| | - Sachiro Kakinoki
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center Research Institute
- Osaka 565-8565
- Japan
- JST-CREST
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center Research Institute
- Osaka 565-8565
- Japan
- JST-CREST
| | - Nobuhiko Yui
- Institute of Biomaterials and Bioengineering
- Tokyo Medical and Dental University
- Tokyo 101-0062
- Japan
- JST-CREST
| |
Collapse
|
139
|
Staines KA, Prideaux M, Allen S, Buttle DJ, Pitsillides AA, Farquharson C. E11/Podoplanin Protein Stabilization Through Inhibition of the Proteasome Promotes Osteocyte Differentiation in Murine in Vitro Models. J Cell Physiol 2015; 231:1392-404. [PMID: 26639105 PMCID: PMC4832367 DOI: 10.1002/jcp.25282] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/04/2015] [Indexed: 11/08/2022]
Abstract
The transmembrane glycoprotein E11 is considered critical in early osteoblast-osteocyte transitions (osteocytogenesis), however its function and regulatory mechanisms are still unknown. Using the late osteoblast MLO-A5 cell line we reveal increased E11 protein/mRNA expression (P < 0.001) concomitant with extensive osteocyte dendrite formation and matrix mineralization (P < 0.001). Transfection with E11 significantly increased mRNA levels (P < 0.001), but immunoblotting failed to detect any correlative increases in E11 protein levels, suggestive of post-translational degradation. We found that exogenous treatment of MLO-A5 and osteocytic IDG-SW3 cells with 10 μM ALLN (calpain and proteasome inhibitor) stabilized E11 protein levels and induced a profound increase in osteocytic dendrite formation (P < 0.001). Treatment with other calpain inhibitors failed to promote similar osteocytogenic changes, suggesting that these effects of ALLN rely upon its proteasome inhibitor actions. Accordingly we found that proteasome-selective inhibitors (MG132/lactacystin/ Bortezomib/Withaferin-A) produced similar dose-dependent increases in E11 protein levels in MLO-A5 and primary osteoblast cells. This proteasomal targeting was confirmed by immunoprecipitation of ubiquitinylated proteins, which included E11, and by increased levels of ubiquitinylated E11 protein upon addition of the proteasome inhibitors MG132/Bortezomib. Activation of RhoA, the small GTPase, was found to be increased concomitant with the peak in E11 levels and its downstream signaling was also observed to promote MLO-A5 cell dendrite formation. Our data indicate that a mechanism reliant upon blockade of proteasome-mediated E11 destabilization contributes to osteocytogenesis and that this may involve downstream targeting of RhoA. This work adds to our mechanistic understanding of the factors regulating bone homeostasis, which may lead to future therapeutic approaches.
Collapse
Affiliation(s)
- Katherine A Staines
- Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| | - Matt Prideaux
- The University of Adelaide, North Terrace, Adelaide, Australia
| | - Steve Allen
- Royal Veterinary College, Royal College Street, London, United Kingdom
| | - David J Buttle
- Department of Infection and Immunity, The University of Sheffield Medical School, Beech Hill Road, Sheffield, United Kingdom
| | | | - Colin Farquharson
- Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| |
Collapse
|
140
|
Sart S, Agathos SN, Li Y, Ma T. Regulation of mesenchymal stem cell 3D microenvironment: From macro to microfluidic bioreactors. Biotechnol J 2015; 11:43-57. [PMID: 26696441 DOI: 10.1002/biot.201500191] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 11/02/2015] [Accepted: 11/30/2015] [Indexed: 12/12/2022]
Abstract
Human mesenchymal stem cells (hMSCs) have emerged as an important cell type in cell therapy and tissue engineering. In these applications, maintaining the therapeutic properties of hMSCs requires tight control of the culture environments and the structural cell organizations. Bioreactor systems are essential tools to achieve these goals in the clinical-scale expansion and tissue engineering applications. This review summarizes how different bioreactors provide cues to regulate the structure and the chemico-mechanical microenvironment of hMSCs with a focus on 3D organization. In addition to conventional bioreactors, recent advances in microfluidic bioreactors as a novel approach to better control the hMSC microenvironment are also discussed. These advancements highlight the key role of bioreactor systems in preserving hMSC's functional properties by providing dynamic and temporal regulation of in vitro cellular microenvironment.
Collapse
Affiliation(s)
- Sébastien Sart
- Hydrodynamics Laboratory, CNRS UMR7646, Ecole Polytechnique, Palaiseau, France
| | - Spiros N Agathos
- Laboratory of Bioengineering, Catholic University of Louvain, Louvain-la-Neuve, Belgium
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA.
| |
Collapse
|
141
|
Heo SJ, Thorpe SD, Driscoll TP, Duncan RL, Lee DA, Mauck RL. Biophysical Regulation of Chromatin Architecture Instills a Mechanical Memory in Mesenchymal Stem Cells. Sci Rep 2015; 5:16895. [PMID: 26592929 PMCID: PMC4655352 DOI: 10.1038/srep16895] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 10/20/2015] [Indexed: 12/23/2022] Open
Abstract
Mechanical cues direct the lineage commitment of mesenchymal stem cells (MSCs). In this study, we identified the operative molecular mechanisms through which dynamic tensile loading (DL) regulates changes in chromatin organization and nuclear mechanics in MSCs. Our data show that, in the absence of exogenous differentiation factors, short term DL elicits a rapid increase in chromatin condensation, mediated by acto-myosin based cellular contractility and the activity of the histone-lysine N-methyltransferase EZH2. The resulting change in chromatin condensation stiffened the MSC nucleus, making it less deformable when stretch was applied to the cell. We also identified stretch induced ATP release and purinergic calcium signaling as a central mediator of this chromatin condensation process. Further, we showed that DL, through differential stabilization of the condensed chromatin state, established a ‘mechanical memory’ in these cells. That is, increasing strain levels and number of loading events led to a greater degree of chromatin condensation that persisted for longer periods of time after the cessation of loading. These data indicate that, with mechanical perturbation, MSCs develop a mechanical memory encoded in structural changes in the nucleus which may sensitize them to future mechanical loading events and define the trajectory and persistence of their lineage specification.
Collapse
Affiliation(s)
- Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen D Thorpe
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Tristan P Driscoll
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.,Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, PA, USA
| | - Randall L Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - David A Lee
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.,Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, PA, USA
| |
Collapse
|
142
|
Shao J, Zhang W, Yang T. Using mesenchymal stem cells as a therapy for bone regeneration and repairing. Biol Res 2015; 48:62. [PMID: 26530042 PMCID: PMC4630918 DOI: 10.1186/s40659-015-0053-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023] Open
Abstract
Bone is a unique tissue which could regenerate completely after injury rather than heal itself with a scar. Compared with other tissues the difference is that, during bone repairing and regeneration, after the inflammatory phase the mesenchymal stem cells (MSCs) are recruited to the injury site and differentiate into either chondroblasts or osteoblasts precursors, leading to bone repairing and regeneration. Besides these two precursors, the MSCs can also differentiate into adipocyte precursors, skeletal muscle precursors and some other mesodermal cells. With this multilineage potentiality, the MSCs are probably used to cure bone injury and other woundings in the near future. Here we will introduce the recent developments in understanding the mechanism of MSCs action in bone regeneration and repairing.
Collapse
Affiliation(s)
- Jin Shao
- Department of Orthopaedics, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai, 200135, China.
| | - Weiwei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Tieyi Yang
- Department of Orthopaedics, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai, 200135, China.
| |
Collapse
|
143
|
Miller GJ, Gerstenfeld LC, Morgan EF. Mechanical microenvironments and protein expression associated with formation of different skeletal tissues during bone healing. Biomech Model Mechanobiol 2015; 14:1239-53. [PMID: 25822264 PMCID: PMC5608650 DOI: 10.1007/s10237-015-0670-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 03/23/2015] [Indexed: 11/29/2022]
Abstract
Uncovering the mechanisms of the sensitivity of bone healing to mechanical factors is critical for understanding the basic biology and mechanobiology of the skeleton, as well as for enhancing clinical treatment of bone injuries. This study refined an experimental method of measuring the strain microenvironment at the site of a bone injury during bone healing. This method used a rat model in which a well-controlled bending motion was applied to an osteotomy to induce the formation of pseudarthrosis that is composed of a range of skeletal tissues, including woven bone, cartilage, fibrocartilage, fibrous tissue, and clot tissue. The goal of this study was to identify both the features of the strain microenvironment associated with formation of these different tissues and the expression of proteins frequently implicated in sensing and transducing mechanical cues. By pairing the strain measurements with histological analyses that identified the regions in which each tissue type formed, we found that formation of the different tissue types occurs in distinct strain microenvironments and that the type of tissue formed is correlated most strongly to the local magnitudes of extensional and shear strains. Weaker correlations were found for dilatation. Immunohistochemical analyses of focal adhesion kinase and rho family proteins RhoA and CDC42 revealed differences within the cartilaginous tissues in the calluses from the pseudarthrosis model as compared to fracture calluses undergoing normal endochondral bone repair. These findings suggest the involvement of these proteins in the way by which mechanical stimuli modulate the process of cartilage formation during bone healing.
Collapse
Affiliation(s)
- Gregory J Miller
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Louis C Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Elise F Morgan
- Department of Mechanical Engineering, Boston University, Boston, MA, USA.
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
144
|
Kim H, Kim I, Choi HJ, Kim SY, Yang EG. Neuron-like differentiation of mesenchymal stem cells on silicon nanowires. NANOSCALE 2015; 7:17131-17138. [PMID: 26422757 DOI: 10.1039/c5nr05787f] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The behavior of mammalian cells on vertical nanowire (NW) arrays, including cell spreading and the dynamic distribution of focal adhesions and cytoskeletal proteins, has been intensively studied to extend the implications for cellular manipulations in vitro. Prompted by the result that cells on silicon (Si) NWs showed morphological changes and reduced migration rates, we have explored the transition of mesenchymal stem cells into a neuronal lineage by using SiNWs with varying lengths. When human mesenchymal stem cells (hMSCs) were cultured on the longest SiNWs for 3 days, most of the cells exhibited elongated shapes with neurite-like extensions and dot-like focal adhesions that were prominently observed along with actin filaments. Under these circumstances, the cell motility analyzed by live cell imaging was found to decrease due to the presence of SiNWs. In addition, the slowed growth rate, as well as the reduced population of S phase cells, suggested that the cell cycle was likely arrested in response to the differentiation process. Furthermore, we measured the mRNA levels of several lineage-specific markers to confirm that the SiNWs actually induced neuron-like differentiation of the hMSCs while hampering their osteogenic differentiation. Taken together, our results implied that SiNWs were capable of inducing active reorganization of cellular behaviors, collectively guiding the fate of hMSCs into the neural lineage even in the absence of any inducing reagent.
Collapse
Affiliation(s)
- Hyunju Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 136-791, South Korea.
| | | | | | | | | |
Collapse
|
145
|
Rho/Rock signal transduction pathway is required for MSC tenogenic differentiation. Bone Res 2015; 3:15015. [PMID: 26509098 PMCID: PMC4605238 DOI: 10.1038/boneres.2015.15] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/20/2015] [Accepted: 04/20/2015] [Indexed: 01/20/2023] Open
Abstract
Mesenchymal stem cell (MSC)-based treatments have shown promise for improving tendon healing and repair. MSCs have the potential to differentiate into multiple lineages in response to select chemical and physical stimuli, including into tenocytes. Cell elongation and cytoskeletal tension have been shown to be instrumental to the process of MSC differentiation. Previous studies have shown that inhibition of stress fiber formation leads MSCs to default toward an adipogenic lineage, which suggests that stress fibers are required for MSCs to sense the environmental factors that can induce differentiation into tenocytes. As the Rho/ROCK signal transduction pathway plays a critical role in both stress fiber formation and in cell sensation, we examined whether the activation of this pathway was required when inducing MSC tendon differentiation using rope-like silk scaffolds. To accomplish this, we employed a loss-of-function approach by knocking out ROCK, actin and myosin (two other components of the pathway) using the specific inhibitors Y-27632, Latrunculin A and blebbistatin, respectively. We demonstrated that independently disrupting the cytoskeleton and the Rho/ROCK pathway abolished the expression of tendon differentiation markers and led to a loss of spindle morphology. Together, these studies suggest that the tension that is generated by MSC elongation is essential for MSC teno-differentiation and that the Rho/ROCK pathway is a critical mediator of tendon differentiation on rope-like silk scaffolds.
Collapse
|
146
|
Koide H, Holmbeck K, Lui JC, Guo XC, Driggers P, Chu T, Tatsuno I, Quaglieri C, Kino T, Baron J, Young MF, Robey PG, Segars JH. Mice Deficient in AKAP13 (BRX) Are Osteoporotic and Have Impaired Osteogenesis. J Bone Miner Res 2015; 30:1887-95. [PMID: 25892096 PMCID: PMC4590282 DOI: 10.1002/jbmr.2534] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 04/03/2015] [Accepted: 04/15/2015] [Indexed: 01/23/2023]
Abstract
Mechanical stimulation is crucial to bone growth and triggers osteogenic differentiation through a process involving Rho and protein kinase A. We previously cloned a gene (AKAP13, aka BRX) encoding a protein kinase A-anchoring protein in the N-terminus, a guanine nucleotide-exchange factor for RhoA in the mid-section, coupled to a carboxyl region that binds to estrogen and glucocorticoid nuclear receptors. Because of the critical role of Rho, estrogen, and glucocorticoids in bone remodeling, we examined the multifunctional role of Akap13. Akap13 was expressed in bone, and mice haploinsufficient for Akap13 (Akap13(+/-)) displayed reduced bone mineral density, reduced bone volume/total volume, and trabecular number, and increased trabecular spacing; resembling the changes observed in osteoporotic bone. Consistent with the osteoporotic phenotype, Colony forming unit-fibroblast numbers were diminished in Akap13(+/-) mice, as were osteoblast numbers and extracellular matrix production when compared to control littermates. Transcripts of Runx2, an essential transcription factor for the osteogenic lineage, and alkaline phosphatase (Alp), an indicator of osteogenic commitment, were both reduced in femora of Akap13(+/-) mice. Knockdown of Akap13 reduced levels of Runx2 and Alp transcripts in immortalized bone marrow stem cells. These findings suggest that Akap13 haploinsufficient mice have a deficiency in early osteogenesis with a corresponding reduction in osteoblast number, but no impairment of mature osteoblast activity.
Collapse
Affiliation(s)
- Hisashi Koide
- Unit of Reproductive Endocrinology, Program in Reproductive and Adult Endocrinology (PRAE), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kenn Holmbeck
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Julian C Lui
- Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Xiaoxiao C Guo
- Unit of Reproductive Endocrinology, Program in Reproductive and Adult Endocrinology (PRAE), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Paul Driggers
- Unit of Reproductive Endocrinology, Program in Reproductive and Adult Endocrinology (PRAE), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Tiffany Chu
- Unit of Reproductive Endocrinology, Program in Reproductive and Adult Endocrinology (PRAE), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Ichiro Tatsuno
- Center for Diabetes, Metabolism and Endocrinology, Toho University Sakura Medical Center, Chiba, Japan
| | - Caroline Quaglieri
- Unit of Reproductive Endocrinology, Program in Reproductive and Adult Endocrinology (PRAE), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Tomoshige Kino
- Unit of Reproductive Endocrinology, Program in Reproductive and Adult Endocrinology (PRAE), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jeffrey Baron
- Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Marian F Young
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health (NIH), Bethesda, MD, USA
| | - James H Segars
- Unit of Reproductive Endocrinology, Program in Reproductive and Adult Endocrinology (PRAE), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
147
|
Tai IC, Wang YH, Chen CH, Chuang SC, Chang JK, Ho ML. Simvastatin enhances Rho/actin/cell rigidity pathway contributing to mesenchymal stem cells' osteogenic differentiation. Int J Nanomedicine 2015; 10:5881-94. [PMID: 26451103 PMCID: PMC4590348 DOI: 10.2147/ijn.s84273] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Recent studies have indicated that statins induce osteogenic differentiation both in vitro and in vivo. The molecular mechanism of statin-stimulated osteogenesis is unknown. Activation of RhoA signaling increases cytoskeletal tension, which plays a crucial role in the osteogenic differentiation of mesenchymal stem cells. We thus hypothesized that RhoA signaling is involved in simvastatin-induced osteogenesis in bone marrow mesenchymal stem cells. We found that although treatment with simvastatin shifts localization of RhoA protein from the membrane to the cytosol, the treatment still activates RhoA dose-dependently because it reduces the association with RhoGDIα. Simvastatin also increased the expression of osteogenic proteins, density of actin filament, the number of focal adhesions, and cellular tension. Furthermore, disrupting actin cytoskeleton or decreasing cell rigidity by using chemical agents reduced simvastatin-induced osteogenic differentiation. In vivo study also confirms that density of actin filament is increased in simvastatin-induced ectopic bone formation. Our study is the first to demonstrate that maintaining intact actin cytoskeletons and enhancing cell rigidity are crucial in simvastatin-induced osteogenesis. The results suggested that simvastatin, which is an osteoinductive factor and acts by increasing actin filament organization and cell rigidity combined with osteoconductive biomaterials, may benefit stem-cell-based bone regeneration.
Collapse
Affiliation(s)
- I-Chun Tai
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yao-Hsien Wang
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shu-Chun Chuang
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Je-Ken Chang
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan ; Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Marine Biotechnology and Resources, National Sun Yat-sen UniVersity, Kaohsiung, Taiwan
| |
Collapse
|
148
|
Li J, Hu C, Han L, Liu L, Jing W, Tang W, Tian W, Long J. MiR-154-5p regulates osteogenic differentiation of adipose-derived mesenchymal stem cells under tensile stress through the Wnt/PCP pathway by targeting Wnt11. Bone 2015; 78:130-141. [PMID: 25959411 DOI: 10.1016/j.bone.2015.05.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/13/2015] [Accepted: 05/02/2015] [Indexed: 01/10/2023]
Abstract
Mechanical stress is a well-acknowledged positive regulatory factor for osteogenic differentiation of adipose- derived mesenchymal stem cells (ADSCs). However, the molecular mechanisms associated with micro-RNAs (miRNAs) whereby ADSCs respond to mechanical stimuli remain elusive. We investigated the mechanism of mechanotransduction from the miRNA perspective in the osteogenic differentiation of ADSCs under tensile stress. Microarray analysis showed that miR-154-5p was remarkably downregulated when ADSCs were subjected to mechanical tension. Bioinformatics analysis with luciferase reporter assays demonstrated that Wnt11 3'UTR was a new direct target of miR-154-5p. Under tensile stress, lentivirus-mediated gain- or loss-of-function studies revealed that forced expression of miR-154-5p inhibited osteogenic differentiation of ADSCs, whereas inhibition of endogenous miR-154-5p with its antisense oligonucleotide (ASO-154-5p) obviously promoted osteogenic differentiation. Furthermore, miR-154-5p overexpression decreased activity of the non-canonical Wnt/PCP (RhoA-ROCK) pathway, as indicated by lower expression of Wnt11, active RhoA and ROCKII in miR-154-5p-treated ADSCs. By contrast, miR-154-5p inhibition activated the Wnt/PCP signals. Taken together, these results demonstrate that, under tensile stress, miR-154-5p negatively regulates ADSCs osteogenic differentiation through the Wnt/PCP pathway by directly targeting Wnt11. This novel regulatory pathway provides new insights into the molecular mechanism of mechanotransduction in osteogenic differentiation of ADSCs.
Collapse
Affiliation(s)
- Jianwei Li
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China
| | - Chen Hu
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China
| | - Lu Han
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China
| | - Lei Liu
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China
| | - Wei Jing
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China
| | - Wei Tang
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China
| | - Weidong Tian
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China
| | - Jie Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China; Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
149
|
Aisha M, Nor-Ashikin M, Sharaniza A, Nawawi H, Froemming G. Orbital fluid shear stress promotes osteoblast metabolism, proliferation and alkaline phosphates activity in vitro. Exp Cell Res 2015; 337:87-93. [DOI: 10.1016/j.yexcr.2015.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 07/05/2015] [Accepted: 07/06/2015] [Indexed: 01/17/2023]
|
150
|
Pearson NC, Oliver JM, Shipley RJ, Waters SL. A multiphase model for chemically- and mechanically- induced cell differentiation in a hollow fibre membrane bioreactor: minimising growth factor consumption. Biomech Model Mechanobiol 2015; 15:683-700. [PMID: 26276678 DOI: 10.1007/s10237-015-0717-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/04/2015] [Indexed: 11/30/2022]
Abstract
We present a simplified two-dimensional model of fluid flow, solute transport, and cell distribution in a hollow fibre membrane bioreactor. We consider two cell populations, one undifferentiated and one differentiated, with differentiation stimulated either by growth factor alone, or by both growth factor and fluid shear stress. Two experimental configurations are considered, a 3-layer model in which the cells are seeded in a scaffold throughout the extracapillary space (ECS), and a 4-layer model in which the cell-scaffold construct occupies a layer surrounding the outside of the hollow fibre, only partially filling the ECS. Above this is a region of free-flowing fluid, referred to as the upper fluid layer. Following previous models by the authors (Pearson et al. in Math Med Biol, 2013, Biomech Model Mechanbiol 1-16, 2014a, we employ porous mixture theory to model the dynamics of, and interactions between, the cells, scaffold, and fluid in the cell-scaffold construct. We use this model to determine operating conditions (experiment end time, growth factor inlet concentration, and inlet fluid fluxes) which result in a required percentage of differentiated cells, as well as maximising the differentiated cell yield and minimising the consumption of expensive growth factor.
Collapse
Affiliation(s)
- Natalie C Pearson
- OCIAM, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - James M Oliver
- OCIAM, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - Rebecca J Shipley
- Biomechanical Engineering Group, UCL Mechanical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Sarah L Waters
- OCIAM, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK.
| |
Collapse
|