1
|
Abbasloo F, Vahidi B, Khani MM, Sigaroodi F, Sarbandi RR. Osteogenic differentiation of mesenchymal stem cell on poly sorbitol sebacate scaffold under shear stress in a bioreactor. Tissue Cell 2025; 93:102715. [PMID: 39787937 DOI: 10.1016/j.tice.2024.102715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 12/28/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025]
Abstract
Mechanical loading plays a pivotal role in regulating bone anabolic processes. Understanding the optimal mechanical loading parameters for cellular responses is critical for advancing strategies in orthopedic bioreactor-based bone tissue engineering. This study developed a poly (sorbitol sebacate) (PSS) filmscaffold with a sorbitol-to-sebacic acid molar ratio of 1:4. The scaffold underwent extensive characterization, including physical and mechanical property evaluations, biocompatibility assessments, and cell adhesion analysis. The Young's modulus of the PSS polymer was determined to be 6.81 ± 0.44 MPa under dry conditions, 6.37 ± 1.09 MPa in a wet state, and 6.38 ± 0.71 MPa after ethanol washing (70 %). The average contact angle of the PSS film was measured at 88.806 ± 1.644°, indicating moderate hydrophilicity. To investigate the osteogenic potential, a fluid flow inducing a shear stress of 1 Pa at a frequency of 1 Hz was applied to mesenchymal stem cells (MSCs) cultured on the PSS scaffold. Cells were exposed to dynamic fluid flow for one hour daily on days 4, 5, 6, and 7 of culture, followed by a static culture period of 14 days. The expression of osteogenic differentiation markers, including osteopontin (OPN), osteocalcin (OCN), type I collagen, and calcium deposition, was significantly elevated under dynamic conditions compared to static culture. This study highlights the importance of mechanical stimulation in enhancing MSC osteogenesis and underscores the osteoconductive properties of the PSS scaffold. These findings provide valuable insights into scaffold design and mechanical loading strategies for laboratory-based bone tissue engineering applications.
Collapse
Affiliation(s)
- Fatemeh Abbasloo
- Department of Medical Technology and Tissue engineering, Faculty of Life Science Engineering, School of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran
| | - Bahman Vahidi
- Department of Medical Technology and Tissue engineering, Faculty of Life Science Engineering, School of Interdisciplinary Science and Technology, University of Tehran, Tehran, Iran.
| | - Mohammad-Mehdi Khani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Faraz Sigaroodi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Ramezani Sarbandi
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
2
|
Zhao H, Tu X. The potential key genes within focal adhesion that regulate mesenchymal stem cells osteogenesis or adipogenesis in microgravity related disuse osteoporosis: an integrated analysis. Front Endocrinol (Lausanne) 2025; 16:1469400. [PMID: 40130165 PMCID: PMC11930814 DOI: 10.3389/fendo.2025.1469400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/14/2025] [Indexed: 03/26/2025] Open
Abstract
This study aimed to identify key genes related to focal adhesions (FA) and cells involved in osteoblast (OS) and adipocyte (AD) differentiation in osteoporosis. A mouse model of disuse osteoporosis was made by hindlimbs unloading (HLU)/Tail - suspension. Micro - CT and histological analysis were done, and differentially expressed genes (DEGs) from GSE100930 were analyzed. Soft clustering on GSE80614 OS/AD samples found FA - related candidate genes. protein-protein interaction (PPI) network and cytoHubba's Degree algorithm identified key FA - genes, validated by quantitative polymerase chain reaction (qPCR). Key OS/AD - associated cells were identified by single - cell analysis. The mouse model showed decreased bone density, microstructure damage, increased marrow adiposity, and altered gene expression. Key FA - related genes for osteogenesis (ITGB3, LAMC1, COL6A3, ITGA8, PDGFRB) and adipogenesis (ITGB3, ITGA4, LAMB1, ITGA8, LAMA4) were found and validated. Key cells (chondrocyte, adipocyte, and osteoblast progenitors) are involved in specific pathways, with osteoblast progenitors having stronger interactions. Pseudotime analysis implies differentiation from chondrocyte progenitors to adipocyte, then osteoblast progenitors. This study provides new insights for disuse osteoporosis research.
Collapse
Affiliation(s)
| | - Xiaolin Tu
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Liu M, Wu A, Liu J, Huang HW, Li Y, Shi Q, Huang Q, Wang H. Arched microfluidic channel for the promotion of axonal growth performance. iScience 2024; 27:110885. [PMID: 39319262 PMCID: PMC11419798 DOI: 10.1016/j.isci.2024.110885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/02/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024] Open
Abstract
Uniformly distributed fluid shear stress can promote axonal growth, aiding in the efficient construction of functional neural interfaces. However, challenges remain in the construction of the micro-scale environment with a uniform fluidic stress distribution. In this study, we designed and fabricated a microfluidic chip with arched-section microfluidic channels (AMCs) to increase primary cortical neuron growth rate and terminal number by constructing a uniform-stress-distributed environment. Inspired by the three-dimensional (3D) microenvironment where cerebrospinal-fluid-contacting neurons are located, the surface curvature of the traditional rectangular-section microfluidic channel (RMC) was adjusted to construct structures with 3D curved surfaces. Compared with those on the RMC chips, the average growth rate of the axons on the AMC chips increased by 8.9% within 19 days, and the average number of terminals increased by 14.9%. This platform provides a structure that can effectively promote neuron growth and has potential in constructing more complex functional neural interfaces.
Collapse
Affiliation(s)
- Menghua Liu
- Intelligent Robotics Institute, School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Anping Wu
- Intelligent Robotics Institute, School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Jiaxin Liu
- Intelligent Robotics Institute, School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Hen-Wei Huang
- Laboratory for Translational Engineering, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yang Li
- Peking University First Hospital, Xicheng District, Beijing 100034, China
| | - Qing Shi
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China
| | - Qiang Huang
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China
| | - Huaping Wang
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, China
| |
Collapse
|
4
|
Yang L, Chen H, Yang C, Hu Z, Jiang Z, Meng S, Liu R, Huang L, Yang K. Research progress on the regulatory mechanism of integrin-mediated mechanical stress in cells involved in bone metabolism. J Cell Mol Med 2024; 28:e18183. [PMID: 38506078 PMCID: PMC10951882 DOI: 10.1111/jcmm.18183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/14/2024] [Accepted: 02/04/2024] [Indexed: 03/21/2024] Open
Abstract
Mechanical stress is an internal force between various parts of an object that resists external factors and effects that cause an object to deform, and mechanical stress is essential for various tissues that are constantly subjected to mechanical loads to function normally. Integrins are a class of transmembrane heterodimeric glycoprotein receptors that are important target proteins for the action of mechanical stress stimuli on cells and can convert extracellular physical and mechanical signals into intracellular bioelectrical signals, thereby regulating osteogenesis and osteolysis. Integrins play a bidirectional regulatory role in bone metabolism. In this paper, relevant literature published in recent years is reviewed and summarized. The characteristics of integrins and mechanical stress are introduced, as well as the mechanisms underlying responses of integrin to mechanical stress stimulation. The paper focuses on integrin-mediated mechanical stress in different cells involved in bone metabolism and its associated signalling mechanisms. The purpose of this review is to provide a theoretical basis for the application of integrin-mediated mechanical stress to the field of bone tissue repair and regeneration.
Collapse
Affiliation(s)
- Li Yang
- Department of Periodontology, Hospital of StomatologyZunyi Medical UniversityZunyiChina
| | - Hong Chen
- Department of Periodontology, Hospital of StomatologyZunyi Medical UniversityZunyiChina
| | - Chanchan Yang
- Department of Periodontology, Hospital of StomatologyZunyi Medical UniversityZunyiChina
| | - Zhengqi Hu
- Department of Periodontology, Hospital of StomatologyZunyi Medical UniversityZunyiChina
| | - Zhiliang Jiang
- Department of Periodontology, Hospital of StomatologyZunyi Medical UniversityZunyiChina
| | - Shengzi Meng
- Department of Periodontology, Hospital of StomatologyZunyi Medical UniversityZunyiChina
| | | | - Lan Huang
- Department of Periodontology, Hospital of StomatologyZunyi Medical UniversityZunyiChina
| | | |
Collapse
|
5
|
Zhou Y, Guo P, Jin Z, Chai M, Zhang S, Wang X, Tan WS, Zhou Y. Fluid shear force and hydrostatic pressure jointly promote osteogenic differentiation of BMSCs by activating YAP1 and NFAT2. Biotechnol J 2024; 19:e2300714. [PMID: 38622793 DOI: 10.1002/biot.202300714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 04/17/2024]
Abstract
Natural bone tissue features a complex mechanical environment, with cells responding to diverse mechanical stimuli, including fluid shear stress (FSS) and hydrostatic pressure (HP). However, current in vitro experiments commonly employ a singular mechanical stimulus to simulate the mechanical environment in vivo. The understanding of the combined effects and mechanisms of multiple mechanical stimuli remains limited. Hence, this study constructed a mechanical stimulation device capable of simultaneously applying FSS and HP to cells. This study investigated the impact of FSS and HP on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and examined the distinctions and interactions between the two mechanisms. The results demonstrated that both FSS and HP individually enhanced the osteogenic differentiation of BMSCs, with a more pronounced effect observed through their combined application. BMSCs responded to external FSS and HP stimulation through the integrin-cytoskeleton and Piezo1 ion channel respectively. This led to the activation of downstream biochemical signals, resulting in the dephosphorylation and nuclear translocation of the intracellular transcription factors Yes Associated Protein 1 (YAP1) and nuclear factor of activated T cells 2 (NFAT2). Activated YAP1 could bind to NFAT2 to enhance transcriptional activity, thereby promoting osteogenic differentiation of BMSCs more effectively. This study highlights the significance of composite mechanical stimulation in BMSCs' osteogenic differentiation, offering guidance for establishing a complex mechanical environment for in vitro functional bone tissue construction.
Collapse
Affiliation(s)
- Yi Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Pan Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Ziyang Jin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Miaomiao Chai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Shuhong Zhang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Henan, People's Republic of China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Henan, People's Republic of China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Yan Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| |
Collapse
|
6
|
Xu X, Guo Y, Liu P, Zhang H, Wang Y, Li Z, Mei Y, Niu L, Liu R. Piezo Mediates the Mechanosensation and Injury-Repair of Pulpo-Dentinal Complex. Int Dent J 2024; 74:71-80. [PMID: 37833209 PMCID: PMC10829354 DOI: 10.1016/j.identj.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 10/15/2023] Open
Abstract
OBJECTIVES The aim of this research was to investigate the functions of Piezo channels in dentin defect, including mechanical signalling and odontoblast responses. METHODS Rat dentin-defect models were constructed, and spatiotemporal expression of Piezo proteins was detected in the pulpo-dentinal complex. Real-time polymerase chain reaction (rtPCR) was used to investigate the functional expression pattern of Piezo channels in odontoblasts. Moreover, RNA interference technology was employed to uncover the underlying mechanisms of the Piezo-driven inflammatory response and repair under fluid shear stress (FSS) conditions in vitro. RESULTS Piezo1 and Piezo2 were found to be widely expressed in the odontoblast layer and dental pulp in the rat dentin-defect model during the end stage of reparative dentin formation. The expression levels of the Piezo1 and Piezo2 genes in MDPC-23 cells were high in the initial stage under FSS loading and then decreased over time. Moreover, the expression trends of inflammatory, odontogenic, and mineralisation genes were generally contrary to those of Piezo1 and Piezo2 over time. After silencing of Piezo1/Piezo2, FSS stimulation resulted in significantly higher expression of inflammatory, odontogenesis, and mineralisation genes in MDPC-23 cells. Finally, the expression of genes involved in the integrin β1/ERK1 and Wnt5b/β-catenin signalling pathways was changed in response to RNA silencing of Piezo1 and Piezo2. CONCLUSIONS Piezo1 and Piezo2 may be involved in regulating the expression of inflammatory and odontogenic genes in odontoblasts stimulated by FSS.
Collapse
Affiliation(s)
- Xiaoqiao Xu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Yi Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Peiqi Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Hui Zhang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, China
| | - Yijie Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Zhen Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Yukun Mei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China
| | - Lin Niu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China; Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
| | - Ruirui Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi' an, China; Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
7
|
Zhou JQ, Wan HY, Wang ZX, Jiang N. Stimulating factors for regulation of osteogenic and chondrogenic differentiation of mesenchymal stem cells. World J Stem Cells 2023; 15:369-384. [PMID: 37342227 PMCID: PMC10277964 DOI: 10.4252/wjsc.v15.i5.369] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/21/2023] [Accepted: 03/29/2023] [Indexed: 05/26/2023] Open
Abstract
Mesenchymal stem cells (MSCs), distributed in many tissues in the human body, are multipotent cells capable of differentiating in specific directions. It is usually considered that the differentiation process of MSCs depends on specialized external stimulating factors, including cell signaling pathways, cytokines, and other physical stimuli. Recent findings have revealed other underrated roles in the differentiation process of MSCs, such as material morphology and exosomes. Although relevant achievements have substantially advanced the applicability of MSCs, some of these regulatory mechanisms still need to be better understood. Moreover, limitations such as long-term survival in vivo hinder the clinical application of MSCs therapy. This review article summarizes current knowledge regarding the differentiation patterns of MSCs under specific stimulating factors.
Collapse
Affiliation(s)
- Jia-Qi Zhou
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Hao-Yang Wan
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Zi-Xuan Wang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Nan Jiang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
8
|
Mao L, Wang L, Xu J, Zou J. The role of integrin family in bone metabolism and tumor bone metastasis. Cell Death Discov 2023; 9:119. [PMID: 37037822 PMCID: PMC10086008 DOI: 10.1038/s41420-023-01417-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023] Open
Abstract
Integrins have been the research focus of cell-extracellular matrix adhesion (ECM) and cytokine receptor signal transduction. They are involved in the regulation of bone metabolism of bone precursor cells, mesenchymal stem cells (MSCs), osteoblasts (OBs), osteoclasts (OCs), and osteocytes. Recent studies expanded and updated the role of integrin in bone metabolism, and a large number of novel cytokines were found to activate bone metabolism pathways through interaction with integrin receptors. Integrins act as transducers that mediate the regulation of bone-related cells by mechanical stress, fluid shear stress (FSS), microgravity, hypergravity, extracellular pressure, and a variety of physical factors. Integrins mediate bone metastasis of breast, prostate, and lung cancer by promoting cancer cell adhesion, migration, and survival. Integrin-mediated targeted therapy showed promising prospects in bone metabolic diseases. This review emphasizes the latest research results of integrins in bone metabolism and bone metastasis and provides a vision for treatment strategies.
Collapse
Affiliation(s)
- Liwei Mao
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China
| | - Lian Wang
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, WA, 6009, Perth, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China.
| |
Collapse
|
9
|
Darshna, Kumar R, Srivastava P, Chandra P. Bioengineering of bone tissues using bioreactors for modulation of mechano-sensitivity in bone. Biotechnol Genet Eng Rev 2023:1-41. [PMID: 36596226 DOI: 10.1080/02648725.2022.2162249] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023]
Abstract
Since the last decade, significant developments have been made in the area of bone tissue engineering associated with the emergence of novel biomaterials as well as techniques of scaffold fabrication. Despite all these developments, the translation from research findings to clinical applications is still very limited. Manufacturing the designed tissue constructs in a scalable manner remains the most challenging aspect. This bottleneck could be overcome by using bioreactors for the manufacture of these tissue constructs. In this review, a current scenario of bone injuries/defects and the cause of the translational gap between laboratory research and clinical use has been emphasized. Furthermore, various bioreactors being used in the area of bone tissue regeneration in recent studies have been highlighted along with their advantages and limitations. A vivid literature survey on the ideal attributes of bioreactors has been accounted, viz. dynamic, versatile, automated, reproducible and commercialization aspects. Additionally, the illustration of computational approaches that should be combined with bone tissue engineering experiments using bioreactors to simulate and optimize cellular growth in bone tissue constructs has also been done extensively.
Collapse
Affiliation(s)
- Darshna
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Rahul Kumar
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Pradeep Srivastava
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| |
Collapse
|
10
|
Ma Q, Miri Z, Haugen HJ, Moghanian A, Loca D. Significance of mechanical loading in bone fracture healing, bone regeneration, and vascularization. J Tissue Eng 2023; 14:20417314231172573. [PMID: 37251734 PMCID: PMC10214107 DOI: 10.1177/20417314231172573] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
In 1892, J.L. Wolff proposed that bone could respond to mechanical and biophysical stimuli as a dynamic organ. This theory presents a unique opportunity for investigations on bone and its potential to aid in tissue repair. Routine activities such as exercise or machinery application can exert mechanical loads on bone. Previous research has demonstrated that mechanical loading can affect the differentiation and development of mesenchymal tissue. However, the extent to which mechanical stimulation can help repair or generate bone tissue and the related mechanisms remain unclear. Four key cell types in bone tissue, including osteoblasts, osteoclasts, bone lining cells, and osteocytes, play critical roles in responding to mechanical stimuli, while other cell lineages such as myocytes, platelets, fibroblasts, endothelial cells, and chondrocytes also exhibit mechanosensitivity. Mechanical loading can regulate the biological functions of bone tissue through the mechanosensor of bone cells intraosseously, making it a potential target for fracture healing and bone regeneration. This review aims to clarify these issues and explain bone remodeling, structure dynamics, and mechano-transduction processes in response to mechanical loading. Loading of different magnitudes, frequencies, and types, such as dynamic versus static loads, are analyzed to determine the effects of mechanical stimulation on bone tissue structure and cellular function. Finally, the importance of vascularization in nutrient supply for bone healing and regeneration was further discussed.
Collapse
Affiliation(s)
- Qianli Ma
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
- Department of Immunology, School of
Basic Medicine, Fourth Military Medical University, Xi’an, PR China
| | - Zahra Miri
- Department of Materials Engineering,
Isfahan University of Technology, Isfahan, Iran
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
| | - Amirhossein Moghanian
- Department of Materials Engineering,
Imam Khomeini International University, Qazvin, Iran
| | - Dagnjia Loca
- Rudolfs Cimdins Riga Biomaterials
Innovations and Development Centre, Institute of General Chemical Engineering,
Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga,
Latvia
- Baltic Biomaterials Centre of
Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
11
|
Shen Z, Dong W, Chen Z, Chen G, Zhang Y, Li Z, Lin H, Chen H, Huang M, Guo Y, Jiang Z. Total flavonoids of Rhizoma Drynariae enhances CD31 hiEmcn hi vessel formation and subsequent bone regeneration in rat models of distraction osteogenesis by activating PDGF‑BB/VEGF/RUNX2/OSX signaling axis. Int J Mol Med 2022; 50:112. [PMID: 35795995 PMCID: PMC9330352 DOI: 10.3892/ijmm.2022.5167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/19/2020] [Indexed: 11/06/2022] Open
Abstract
Total flavonoids of Rhizoma Drynariae (TFRD), extracted from the kidney‑tonifying Traditional Chinese medicine Rhizoma Drynariae, can be effective in treating osteoporosis, bone fractures and defects. However, the pharmacological effects of TFRD on the specific vessel subtype CD31hiEmcnhi during distraction osteogenesis (DO) remains unclear. The present study aimed to investigate the effects of TFRD on CD31hiEmcnhi vessels in a rat model of DO. In the present study, tibial DO models were established using 60 rats with a distraction rate of 0.2 mm per day for 20 days. Co‑immunofluorescence staining of CD31 and endomucin (Emcn) was conducted to determine CD31hiEmcnhi vessels. Radiographic, angiographic and histological analyses were performed to assess bone and vessel formation. Tube formation, alkaline phosphatase (ALP) and Von Kossa staining assays were performed to test angiogenesis of endothelial precursor cells (EPCs) and osteogenesis of bone marrow‑derived mesenchymal stem cells (BMSCs). Additionally, expression levels of platelet‑derived growth factor (PDGF)‑BB, VEGF, runt‑related transcription factor 2 (RUNX2) and Osterix (OSX) were determined by western blotting and reverse transcription‑quantitative PCR. The in vivo assays demonstrated that TFRD markedly promoted CD31hiEmcnhi vessel formation during DO, whereas PDGF‑BB neutralizing antibody suppressed vessel formation. Furthermore, the ALP, Von Kossa staining and tube formation assays indicated that TFRD notably elevated the angiogenic capacity of EPCs and osteogenic capacity of BMSCs under stress conditions, which was significantly suppressed by blocking PDGF‑BB. The protein and mRNA levels of PDGF‑BB, VEGF, RUNX2 and OSX were upregulated by TFRD, but downregulated by blocking PDGF‑BB. Thus, TFRD could facilitate CD31hiEmcnhi vessel formation and subsequently enhance angiogenic‑osteogenic coupling to regenerate bone defects during DO via the PDGF‑BB/VEGF/RUNX2/OSX signaling axis, which indicated that CD31hiEmcnhi vessels could be a potential novel therapeutic target for DO, and TFRD may represent a promising drug for promoting bone regeneration in DO by increasing CD31hiEmcnhi vessels.
Collapse
Affiliation(s)
- Zhen Shen
- Department of Orthopaedics, Kunming Municipal Hospital of Traditional Chinese Medicine, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, Yunnan 650599, P.R. China
| | - Wei Dong
- Department of Orthopaedics, Kunming Municipal Hospital of Traditional Chinese Medicine, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, Yunnan 650599, P.R. China
| | - Zehua Chen
- Department of Orthopaedics, Kunming Municipal Hospital of Traditional Chinese Medicine, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, Yunnan 650599, P.R. China
| | - Guoqian Chen
- The Fifth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Yan Zhang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, P.R. China
| | - Zige Li
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, P.R. China
| | - Haixiong Lin
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, P.R. China
| | - Huamei Chen
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, P.R. China
| | - Minling Huang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, P.R. China
| | - Ying Guo
- Department of Orthopaedics, Kunming Municipal Hospital of Traditional Chinese Medicine, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, Yunnan 650599, P.R. China
| | - Ziwei Jiang
- Department of Orthopaedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, P.R. China
| |
Collapse
|
12
|
Rayat Pisheh H, Ansari M, Eslami H. How is mechanobiology involved in bone regenerative medicine? Tissue Cell 2022; 76:101821. [DOI: 10.1016/j.tice.2022.101821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/27/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
|
13
|
Mechanical regulation of bone remodeling. Bone Res 2022; 10:16. [PMID: 35181672 PMCID: PMC8857305 DOI: 10.1038/s41413-022-00190-4] [Citation(s) in RCA: 207] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/04/2021] [Accepted: 12/13/2021] [Indexed: 12/17/2022] Open
Abstract
Bone remodeling is a lifelong process that gives rise to a mature, dynamic bone structure via a balance between bone formation by osteoblasts and resorption by osteoclasts. These opposite processes allow the accommodation of bones to dynamic mechanical forces, altering bone mass in response to changing conditions. Mechanical forces are indispensable for bone homeostasis; skeletal formation, resorption, and adaptation are dependent on mechanical signals, and loss of mechanical stimulation can therefore significantly weaken the bone structure, causing disuse osteoporosis and increasing the risk of fracture. The exact mechanisms by which the body senses and transduces mechanical forces to regulate bone remodeling have long been an active area of study among researchers and clinicians. Such research will lead to a deeper understanding of bone disorders and identify new strategies for skeletal rejuvenation. Here, we will discuss the mechanical properties, mechanosensitive cell populations, and mechanotransducive signaling pathways of the skeletal system.
Collapse
|
14
|
Hao Z, Xu Z, Wang X, Wang Y, Li H, Chen T, Hu Y, Chen R, Huang K, Chen C, Li J. Biophysical Stimuli as the Fourth Pillar of Bone Tissue Engineering. Front Cell Dev Biol 2021; 9:790050. [PMID: 34858997 PMCID: PMC8630705 DOI: 10.3389/fcell.2021.790050] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/26/2021] [Indexed: 01/12/2023] Open
Abstract
The repair of critical bone defects remains challenging worldwide. Three canonical pillars (biomaterial scaffolds, bioactive molecules, and stem cells) of bone tissue engineering have been widely used for bone regeneration in separate or combined strategies, but the delivery of bioactive molecules has several obvious drawbacks. Biophysical stimuli have great potential to become the fourth pillar of bone tissue engineering, which can be categorized into three groups depending on their physical properties: internal structural stimuli, external mechanical stimuli, and electromagnetic stimuli. In this review, distinctive biophysical stimuli coupled with their osteoinductive windows or parameters are initially presented to induce the osteogenesis of mesenchymal stem cells (MSCs). Then, osteoinductive mechanisms of biophysical transduction (a combination of mechanotransduction and electrocoupling) are reviewed to direct the osteogenic differentiation of MSCs. These mechanisms include biophysical sensing, transmission, and regulation. Furthermore, distinctive application strategies of biophysical stimuli are presented for bone tissue engineering, including predesigned biomaterials, tissue-engineered bone grafts, and postoperative biophysical stimuli loading strategies. Finally, ongoing challenges and future perspectives are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenhua Xu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuan Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hanke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Renxin Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kegang Huang
- Wuhan Institute of Proactive Health Management Science, Wuhan, China
| | - Chao Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Orthopedics, Hefeng Central Hospital, Enshi, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Shi F, Xiao D, Zhang C, Zhi W, Liu Y, Weng J. The effect of macropore size of hydroxyapatite scaffold on the osteogenic differentiation of bone mesenchymal stem cells under perfusion culture. Regen Biomater 2021; 8:rbab050. [PMID: 34567788 PMCID: PMC8457200 DOI: 10.1093/rb/rbab050] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/19/2021] [Accepted: 09/02/2021] [Indexed: 11/12/2022] Open
Abstract
Previous studies have proved that dynamic culture could facilitate nutrients transport and apply mechanical stimulation to the cells within three-dimensional scaffolds, thus enhancing the differentiation of stem cells towards the osteogenic phenotype. However, the effects of macropore size on osteogenic differentiation of stem cells under dynamic condition are still unclear. Therefore, the objective of this study was to investigate the effects of macropore size of hydroxyapatite (HAp) scaffolds on osteogenic differentiation of bone mesenchymal stem cells under static and perfusion culture conditions. In vitro cell culture results showed that cell proliferation, alkaline phosphate (ALP) activity, mRNA expression of ALP, collagen-I (Col-I), osteocalcin (OCN) and osteopontin (OPN) were enhanced when cultured under perfusion condition in comparison to static culture. Under perfusion culture condition, the ALP activity and the gene expression of ALP, Col-I, OCN and OPN were enhanced with the macropore size decreasing from 1300 to 800 µm. However, with the further decrease in macropore size from 800 to 500 µm, the osteogenic related gene expression and protein secretion were reduced. Computational fluid dynamics analysis showed that the distribution areas of medium- and high-speed flow increased with the decrease in macropore size, accompanied by the increase of the fluid shear stress within the scaffolds. These results confirm the effects of macropore size on fluid flow stimuli and cell differentiation, and also help optimize the macropore size of HAp scaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- Feng Shi
- Collaboration Innovation Center for Tissue Repair Material Engineering Technology, College of Life Science, China West Normal University, No.1 Shida Road, Nanchong, Sichuan 637002, China.,Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, the Second Clinical College of North Sichuan Medical College, No.97 Renmin South Road, Nanchong, Sichuan 637000, China.,College of Medicine, Southwest Jiaotong University, No.111 North 1st Section of Second Ring Road, Chengdu, Sichuan 610031, China
| | - Dongqin Xiao
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, the Second Clinical College of North Sichuan Medical College, No.97 Renmin South Road, Nanchong, Sichuan 637000, China
| | - Chengdong Zhang
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, the Second Clinical College of North Sichuan Medical College, No.97 Renmin South Road, Nanchong, Sichuan 637000, China.,College of Medicine, Southwest Jiaotong University, No.111 North 1st Section of Second Ring Road, Chengdu, Sichuan 610031, China
| | - Wei Zhi
- College of Medicine, Southwest Jiaotong University, No.111 North 1st Section of Second Ring Road, Chengdu, Sichuan 610031, China
| | - Yumei Liu
- Collaboration Innovation Center for Tissue Repair Material Engineering Technology, College of Life Science, China West Normal University, No.1 Shida Road, Nanchong, Sichuan 637002, China.,College of Environmental Science and Engineering, China West Normal University, No.1 Shida Road, Nanchong, Sichuan 637002, China
| | - Jie Weng
- College of Medicine, Southwest Jiaotong University, No.111 North 1st Section of Second Ring Road, Chengdu, Sichuan 610031, China
| |
Collapse
|
16
|
Manokawinchoke J, Pavasant P, Limjeerajarus CN, Limjeerajarus N, Osathanon T, Egusa H. Mechanical loading and the control of stem cell behavior. Arch Oral Biol 2021; 125:105092. [PMID: 33652301 DOI: 10.1016/j.archoralbio.2021.105092] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/08/2021] [Accepted: 02/21/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Mechanical stimulation regulates many cell responses. The present study describes the effects of different in vitro mechanical stimulation approaches on stem cell behavior. DESIGN The narrative review approach was performed. The articles published in English language that addressed the effects of mechanical force on stem cells were searched on Pubmed and Scopus database. The effects of extrinsic mechanical force on stem cell response was reviewed and discussed. RESULTS Cells sense mechanical stimuli by the function of mechanoreceptors and further transduce force stimulation into intracellular signaling. Cell responses to mechanical stimuli depend on several factors including type, magnitude, and duration. Further, similar mechanical stimuli exhibit distinct cell responses based on numerous factors including cell type and differentiation stage. Various mechanical applications modulate stemness maintenance and cell differentiation toward specific lineages. CONCLUSIONS Mechanical force application modulates stemness maintenance and differentiation. Modification of force regimens could be utilized to precisely control appropriate stem cell behavior toward specific applications.
Collapse
Affiliation(s)
- Jeeranan Manokawinchoke
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan.
| | - Prasit Pavasant
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Chalida Nakalekha Limjeerajarus
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Nuttapol Limjeerajarus
- Research Center for Advanced Energy Technology, Faculty of Engineering, Thai-Nichi Institute of Technology, Bangkok, 10250, Thailand.
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan.
| |
Collapse
|
17
|
Ascolani G, Skerry TM, Lacroix D, Dall'Ara E, Shuaib A. Analysis of mechanotransduction dynamics during combined mechanical stimulation and modulation of the extracellular-regulated kinase cascade uncovers hidden information within the signalling noise. Interface Focus 2021; 11:20190136. [PMID: 33343875 PMCID: PMC7739911 DOI: 10.1098/rsfs.2019.0136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2020] [Indexed: 12/27/2022] Open
Abstract
Osteoporosis is a bone disease characterized by brittle bone and increased fracture incidence. With ageing societies worldwide, the disease presents a high burden on health systems. Furthermore, there are limited treatments for osteoporosis with just two anabolic pharmacological agents approved by the US Food and Drug Administration. Healthy bones are believed to be maintained via an intricate relationship between dual biochemical and mechanical (bio-mechanical) stimulations. It is widely considered that osteoporosis emerges as a result of disturbances to said relationship. The mechanotransduction process is key to this balance, and disruption of its dynamics in bone cells plays a role in osteoporosis development. Nonetheless, the exact details and mechanisms that drive and secure the health of bones are still elusive at the cellular and molecular scales. This study examined the dual modulation of mechanical stimulation and mechanotransduction activation dynamics in an osteoblast (OB). The aim was to find patterns of mechanotransduction dynamics demonstrating a significant change that can be mapped to alterations in the OB responses, specifically at the level of gene expression and osteogenic markers such as alkaline phosphatase. This was achieved using a three-dimensional hybrid multiscale computational model simulating mechanotransduction in the OB and its interaction with the extracellular matrix, combined with a numerical analytical technique. The model and the analysis method predict that within the noise of mechanotransduction, owing to modulation of the bio-mechanical stimulus and consequent gene expression, there are unique events that provide signatures for a shift in the system's dynamics. Furthermore, the study uncovered molecular interactions that can be potential drug targets.
Collapse
Affiliation(s)
- Gianluca Ascolani
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Insigneo Institute of In Silico Medicine, University of Sheffield, Sheffield, UK
| | - Timothy M. Skerry
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Damien Lacroix
- Insigneo Institute of In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Enrico Dall'Ara
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Insigneo Institute of In Silico Medicine, University of Sheffield, Sheffield, UK
| | - Aban Shuaib
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Insigneo Institute of In Silico Medicine, University of Sheffield, Sheffield, UK
| |
Collapse
|
18
|
JIANG M, SHEN Q, ZHOU Y, REN W, CHAI M, ZHOU Y, TAN WS. Fluid shear stress and endothelial cells synergistically promote osteogenesis of mesenchymal stem cells via integrin β1-FAK-ERK1/2 pathway. Turk J Biol 2021; 45:683-694. [PMID: 35068949 PMCID: PMC8733951 DOI: 10.3906/biy-2104-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 10/26/2021] [Indexed: 02/05/2023] Open
Abstract
Prevascularization and mechanical stimulation have been reported as effective methods for the construction of functional bone tissue. However, their combined effects on osteogenic differentiation and its mechanism remain to be explored. Here, the effects of fluid shear stress (FSS) on osteogenic differentiation of rat bone-marrow-derived mesenchymal stem cells (BMSCs) when cocultured with human umbilical vein endothelial cells (HUVECs) were investigated, and underlying signaling mechanisms were further explored. FSS stimulation for 1-4 h/day increased alkaline phosphatase (ALP) activity and calcium deposition in coculture systems and promoted the proliferation of cocultured cells. FSS stimulation for 2 h/day was selected as the optimized protocol according to osteogenesis in the coculture. In this situation, the mRNA levels of ALP, runt-related transcriptional factor 2 (Runx2) and osteocalcin (OCN), and protein levels of OCN and osteopontin (OPN) in BMSCs were upregulated. Furthermore, FSS and coculture with HUVECs synergistically increased integrin β1 expression in BMSCs and further activated focal adhesion kinases (FAKs) and downstream extracellular signal-related kinase (ERK), leading to the enhancement of Runx2 expression. Blocking the phosphorylation of FAK abrogated FSS-induced ERK phosphorylation and inhibited osteogenesis of cocultured BMSCs. These results revealed that FSS and coculture with HUVECs synergistically promotes the osteogenesis of BMSCs, which was mediated by the integrin β1-FAK-ERK signaling pathway.
Collapse
Affiliation(s)
- Mingli JIANG
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, ShanghaiChina
| | - Qihua SHEN
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, ShanghaiChina
| | - Yi ZHOU
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, ShanghaiChina
| | - Wenxia REN
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, ShanghaiChina
| | - Miaomiao CHAI
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, ShanghaiChina
| | - Yan ZHOU
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, ShanghaiChina
- * To whom correspondence should be addressed. E-mail: * Correspondence:
| | - Wen-Song TAN
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, ShanghaiChina
| |
Collapse
|
19
|
Shen Z, Chen Z, Li Z, Zhang Y, Jiang T, Lin H, Huang M, Chen H, Feng J, Jiang Z. Total Flavonoids of Rhizoma Drynariae Enhances Angiogenic-Osteogenic Coupling During Distraction Osteogenesis by Promoting Type H Vessel Formation Through PDGF-BB/PDGFR-β Instead of HIF-1α/ VEGF Axis. Front Pharmacol 2020; 11:503524. [PMID: 33328980 PMCID: PMC7729076 DOI: 10.3389/fphar.2020.503524] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 10/29/2020] [Indexed: 01/10/2023] Open
Abstract
Background: Total flavonoids of Rhizoma Drynariae (TFRD), extracted from the kidney-tonifying traditional Chinese medicine Rhizoma Rrynariae, has been proved to be effective in treating osteoporosis, bone fractures and defects. However, pharmacological effects of TFRD on type H vessels, angiogenic-osteogenic coupling in distraction osteogenesis (DO) and the mechanism remain unclear. This study aims at investigating whether type H vessels exist in the DO model, effects of TFRD on angiogenic-osteogenic coupling and further elucidating the underlying mechanism. Methods: Rats models of DO and bone fracture (FR) were established, and then were separately divided into TFRD and control subgroups. Imageological and histological analyses were performed to assess bone and vessel formation. Immunofluorescent staining of CD31 and endomucin (Emcn) was conducted to determine type H vessel formation. Matrigel tube formation, ALP and Alizarin Red S staining assays were performed to test the effects of TFRD on angiogenesis or osteogenesis of endothelial precursor cells (EPCs) or bone marrow-derived mesenchymal stem cells (BMSCs). Additionally, expression levels of HIF-1α, VEGF, PDGF-BB, RUNX2 and OSX were determined by ELISA, qPCR or western blot, respectively. Results: The in vivo results indicated more formed type H vessels in DO groups than in FR groups and TFRD obviously increased the abundance of type H vessels. Moreover, groups with higher abundance of type H vessels showed better angiogenesis and osteogenesis outcomes. Further in vitro experiments showed that TFRD significantly promoted while blocking PDGF-BB remarkably suppressed the angiogenic activity of EPCs under stress conditions. The levels of p-AKT and p-ERK1/2, downstream mediators of the PDGF-BB pathway, were up-regulated by TFRD but blocked by function blocking anti-PDGF-BB antibody. In contrast, the activated AKT and ERK1/2 and corresponding tube formation were not affected by the HIF-1α inhibitor. Besides, blocking PDGF-BB inhibited the osteogenic differentiation of the stretched BMSCs, but TFRD enhanced the osteogenic activity of BMSCs and ameliorated the inhibition, with more calcium nodes, higher ALP activity and mRNA and protein levels of RUNX2 and OSX. Conclusion: Type H vessels exist in the DO model and TFRD enhances angiogenic-osteogenic coupling during DO by promoting type H vessel formation via PDGF-BB/PDGFR-β instead of HIF-1α/VEGF axis.
Collapse
Affiliation(s)
- Zhen Shen
- Department of Orthopaedics, Kunming Municipal Hospital of Traditional Chinese Medicine, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, China
- Department of Orthopaedics, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zehua Chen
- The Fifth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zige Li
- Department of Orthopaedics, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan Zhang
- Department of Orthopaedics, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tao Jiang
- The Fifth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haixiong Lin
- Department of Orthopaedics, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Minling Huang
- Department of Orthopaedics, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huamei Chen
- Department of Orthopaedics, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junjie Feng
- Department of Orthopaedics, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziwei Jiang
- Department of Orthopaedics, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
20
|
Yong KW, Choi JR, Choi JY, Cowie AC. Recent Advances in Mechanically Loaded Human Mesenchymal Stem Cells for Bone Tissue Engineering. Int J Mol Sci 2020; 21:5816. [PMID: 32823645 PMCID: PMC7461207 DOI: 10.3390/ijms21165816] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/06/2020] [Accepted: 08/12/2020] [Indexed: 12/14/2022] Open
Abstract
Large bone defects are a major health concern worldwide. The conventional bone repair techniques (e.g., bone-grafting and Masquelet techniques) have numerous drawbacks, which negatively impact their therapeutic outcomes. Therefore, there is a demand to develop an alternative bone repair approach that can address the existing drawbacks. Bone tissue engineering involving the utilization of human mesenchymal stem cells (hMSCs) has recently emerged as a key strategy for the regeneration of damaged bone tissues. However, the use of tissue-engineered bone graft for the clinical treatment of bone defects remains challenging. While the role of mechanical loading in creating a bone graft has been well explored, the effects of mechanical loading factors (e.g., loading types and regime) on clinical outcomes are poorly understood. This review summarizes the effects of mechanical loading on hMSCs for bone tissue engineering applications. First, we discuss the key assays for assessing the quality of tissue-engineered bone grafts, including specific staining, as well as gene and protein expression of osteogenic markers. Recent studies of the impact of mechanical loading on hMSCs, including compression, perfusion, vibration and stretching, along with the potential mechanotransduction signalling pathways, are subsequently reviewed. Lastly, we discuss the challenges and prospects of bone tissue engineering applications.
Collapse
Affiliation(s)
- Kar Wey Yong
- Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jane Ru Choi
- Department of Mechanical Engineering, University of British Columbia, 2054-6250 Applied Science Lane, Vancouver, BC V6T 1Z4, Canada
- Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Jean Yu Choi
- Ninewells Hospital & Medical School, Dundee, Scotland DD1 5EH, UK; (J.Y.C.); (A.C.C.)
| | - Alistair C. Cowie
- Ninewells Hospital & Medical School, Dundee, Scotland DD1 5EH, UK; (J.Y.C.); (A.C.C.)
| |
Collapse
|
21
|
Nokhbatolfoghahaei H, Bohlouli M, Paknejad Z, R Rad M, M Amirabad L, Salehi-Nik N, Khani MM, Shahriari S, Nadjmi N, Ebrahimpour A, Khojasteh A. Bioreactor cultivation condition for engineered bone tissue: Effect of various bioreactor designs on extra cellular matrix synthesis. J Biomed Mater Res A 2020; 108:1662-1672. [PMID: 32191385 DOI: 10.1002/jbm.a.36932] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 01/18/2023]
Abstract
Dynamic-based systems are bio-designed in order to mimic the micro-environments of the bone tissue. There is limited direct comparison between perfusion and perfusion-rotation forces in designing a bioreactor. Hence, in current study, we aimed to compare given bioreactors for bone regeneration. Two types of bioreactors including rotating & perfusion and perfusion bioreactors were designed. Mesenchymal stem cells derived from buccal fat pad were loaded on a gelatin/β-Tricalcium phosphate scaffold. Cell-scaffold constructs were subjected to different treatment condition and place in either of the bioreactors. Effect of different dynamic conditions on cellular behavior including cell proliferation, cell adhesion, and osteogenic differentiation were assessed. Osteogenic assessment of scaffolds after 24 days revealed that rotating & perfusion bioreactor led to significantly higher expression of OCN and RUNX2 genes and also greater amount of ALP and collagen I protein production compared to static groups and perfusion bioreactor. Observation of cellular sheets which filled the scaffold porosities in SEM images, approved the better cell responses to rotating & perfusion forces of the bioreactor. The outcomes demonstrated that rotating & perfusion bioreactor action on bone regeneration is much preferable than perfusion bioreactor. Therefore, it seems that exertion of multi-stimuli is more effective for bone engineering.
Collapse
Affiliation(s)
- Hanieh Nokhbatolfoghahaei
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Bohlouli
- Student Research Committee, Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahrasadat Paknejad
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam R Rad
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila M Amirabad
- School of Dentistry, Marquette University, Milwaukee, Wisconsin, USA
| | - Nasim Salehi-Nik
- Department of Biomechanical Engineering, Faulty of Engineering Technology, University of Twente, Enschede, The Netherlands
| | - Mohammad M Khani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shayan Shahriari
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nasser Nadjmi
- The Team for Cleft and Craniofacial Anomalies, Oral and Maxillofacial Surgery, University of Antwerp, Antwerp, Belgium
| | - Adel Ebrahimpour
- Department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Khojasteh
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Ascolani G, Skerry TM, Lacroix D, Dall'Ara E, Shuaib A. Revealing hidden information in osteoblast's mechanotransduction through analysis of time patterns of critical events. BMC Bioinformatics 2020; 21:114. [PMID: 32183690 PMCID: PMC7079370 DOI: 10.1186/s12859-020-3394-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 02/04/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mechanotransduction in bone cells plays a pivotal role in osteoblast differentiation and bone remodelling. Mechanotransduction provides the link between modulation of the extracellular matrix by mechanical load and intracellular activity. By controlling the balance between the intracellular and extracellular domains, mechanotransduction determines the optimum functionality of skeletal dynamics. Failure of this relationship was suggested to contribute to bone-related diseases such as osteoporosis. RESULTS A hybrid mechanical and agent-based model (Mech-ABM), simulating mechanotransduction in a single osteoblast under external mechanical perturbations, was utilised to simulate and examine modulation of the activation dynamics of molecules within mechanotransduction on the cellular response to mechanical stimulation. The number of molecules and their fluctuations have been analysed in terms of recurrences of critical events. A numerical approach has been developed to invert subordination processes and to extract the direction processes from the molecular signals in order to derive the distribution of recurring events. These predict that there are large fluctuations enclosing information hidden in the noise which is beyond the dynamic variations of molecular baselines. Moreover, studying the system under different mechanical load regimes and altered dynamics of feedback loops, illustrate that the waiting time distributions of each molecule are a signature of the system's state. CONCLUSIONS The behaviours of the molecular waiting times change with the changing of mechanical load regimes and altered dynamics of feedback loops, presenting the same variation of patterns for similar interacting molecules and identifying specific alterations for key molecules in mechanotransduction. This methodology could be used to provide a new tool to identify potent molecular candidates to modulate mechanotransduction, hence accelerate drug discovery towards therapeutic targets for bone mass upregulation.
Collapse
Affiliation(s)
- Gianluca Ascolani
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Insigneo Institute of In Silico Medicine, University of Sheffield, Sheffield, UK
| | - Timothy M Skerry
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Damien Lacroix
- Insigneo Institute of In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Enrico Dall'Ara
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
- Insigneo Institute of In Silico Medicine, University of Sheffield, Sheffield, UK
| | - Aban Shuaib
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK.
- Insigneo Institute of In Silico Medicine, University of Sheffield, Sheffield, UK.
| |
Collapse
|
23
|
Zerdoum AB, Fowler EW, Jia X. Induction of Fibrogenic Phenotype in Human Mesenchymal Stem Cells by Connective Tissue Growth Factor in a Hydrogel Model of Soft Connective Tissue. ACS Biomater Sci Eng 2019; 5:4531-4541. [PMID: 33178886 PMCID: PMC7654958 DOI: 10.1021/acsbiomaterials.9b00425] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Scar formation is the typical endpoint of wound healing in adult mammalian tissues. An overactive or prolonged fibrogenic response following injury leads to excessive deposition of fibrotic proteins that promote tissue contraction and scar formation. Although well-defined in the dermal tissue, the progression of fibrosis is less explored in other connective tissues, such as the vocal fold. To establish a physiologically relevant 3D model of loose connective tissue fibrosis, we have developed a synthetic extracellular matrix using hyaluronic acid (HA) and peptidic building blocks carrying complementary functional groups. The resultant network was cell adhesive and protease degradable, exhibiting viscoelastic properties similar to the human vocal fold. Human mesenchymal stem cells (hMSCs) were encapsulated in the HA matrix as single cells or multicellular aggregates and cultured in pro-fibrotic media containing connective tissue growth factor (CTGF) for up to 21 days. hMSCs treated with CTGF-supplemented media exhibited an increased expression of fibrogenic markers and ECM proteins associated with scarring. Incorporation of α-smooth muscle actin into F-actin stress fibers was also observed. Furthermore, CTGF treatment increased the migratory capacity of hMSCs as compared to the CTGF-free control groups, indicative of the development of a myofibroblast phenotype. Addition of an inhibitor of the mitogen-activated protein kinase (MAPK) pathway attenuated cellular expression of fibrotic markers and related ECM proteins. Overall, this study demonstrates that CTGF promotes the development of a fibrogenic phenotype in hMSCs encapsulated within an HA matrix and that the MAPK pathway is a potential target for future therapeutic endeavors towards limiting scar formation in loose connective tissues.
Collapse
Affiliation(s)
- Aidan B. Zerdoum
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Eric W. Fowler
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Xinqiao Jia
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA
| |
Collapse
|
24
|
Zhao H, Sun J, Shao J, Zou Z, Qiu X, Wang E, Wu G. Glucose Transporter 1 Promotes the Malignant Phenotype of Non-Small Cell Lung Cancer through Integrin β1/Src/FAK Signaling. J Cancer 2019; 10:4989-4997. [PMID: 31598171 PMCID: PMC6775508 DOI: 10.7150/jca.30772] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 06/23/2019] [Indexed: 12/25/2022] Open
Abstract
Background: Glucose transporter 1 (GLUT1) is the main factor of Warburg effect, which is associated with poor prognosis in many tumors. However, the underlying molecular mechanism of GLUT1 in the progression of non-small cell lung cancer (NSCLC) is unclear. Methods: We used quantitative real-time PCR to detect GLUT1 mRNA expression in bronchial brushing samples and performed Western Blot and biological behavior testing to check the effect of GLUT1 on NSCLC cell proliferation, migration, invasion and apoptosis. Results: We found that the C(t) normalized value of GLUT1 in malignant bronchial brushing samples was significantly higher than that in benign samples (P<0.05). GLUT1 significantly increased the expressions of cyclin A, cyclin D1, cyclin E, cyclin dependent kinase 2 (CDK2), CDK4, CDK6 and matrix metalloproteinase 2 (MMP2), but decreased the expressions of p53 and p130 in NSCLC cells. The biological behavior testing indicated that GLUT1 enhanced NSCLC cell proliferation, invasion and migration but inhibited cell apoptosis. In addition, GLUT1 upregulated the expression of integrin β1 and promoted the phosphorylation of focal adhesion kinase (FAK, phosphorylation at Tyr576/577) and Src (Src phosphorylation at Tyr530). siRNA knock down of integrin β1 expression suppressed GLUT1 induced NSCLC cell biological behavior, as well as the phosphorylation of FAK and Src. Conclusion: Taken together, our data confirms that GLUT1 promotes the malignant phenotype of NSCLC through integrin β1/Src/FAK signaling, which provides a new therapeutic target for the treatment and research of lung cancer.
Collapse
Affiliation(s)
- Huanyu Zhao
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Jian Sun
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China.,Guangzhou DaAn Clinical Laboratory Center, No. 74 Zhongshan Er Road, Guangzhou, 510000, China
| | - Jianshuang Shao
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Zifang Zou
- Department of Chest Surgery, The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Xueshan Qiu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Enhua Wang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| | - Guangping Wu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, China
| |
Collapse
|
25
|
Zhang L, Wang Y, Zhou N, Feng Y, Yang X. Cyclic tensile stress promotes osteogenic differentiation of adipose stem cells via ERK and p38 pathways. Stem Cell Res 2019; 37:101433. [PMID: 31005788 DOI: 10.1016/j.scr.2019.101433] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/03/2019] [Accepted: 04/06/2019] [Indexed: 02/05/2023] Open
Abstract
The present study aimed to elucidate whether extracellular signal-regulated kinases 1/2 (ERK1/2) and p38 mitogen-activated protein kinases pathways participate in the transduction of mechanical stretch exerted on adipose stem cells (ASCs) into intracellular osteogenic signals, and if so whether both pathways have time-dependent feature. Rat ASCs were cultured in osteogenic medium for 72 h and assigned into three sets, namely ERK1/2 inhibitor treated set, p38 inhibitor treated set, and the control set. After inhibitor treatment, all cells were subjected to cyclic stretch(2000 με, 1 Hz) on a four-point bending mechanical loading device. Protein and mRNA samples were acquired at six time points: 0, 15 min, 30 min, 1 h, 2 h and 6 h. Western blot showed phosphorylation level of ERK1/2 was elevated by cyclic tensile stress at all time points, while p38 at 15 min, 30 min and 1 h, and the elevation can be completely blocked by corresponding inhibitors. The treatment by ERK1/2 inhibitor was shown to antagonize the up-regulation of osteogenic genes bone morphogenetic protein 2 (BMP-2) and runt-related transcription factor 2 (Runx2) by mechanical stretch at 15 min and 6 h, whereas p38 inhibitor took effect at 15 min only. The results suggested both ERK and p38 could be positive mediators of stretch-induced osteogenic differentiation of ASCs, and ERK stimulate the stretch-induced osteogenic differentiation at both early and late stages while p38 responds to mechanical stretch in a more rapid fashion.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yingkai Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Nan Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuzhang Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xingmei Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
26
|
Alteration of mesenchymal stem cells polarity by laminar shear stimulation promoting β-catenin nuclear localization. Biomaterials 2019; 190-191:1-10. [DOI: 10.1016/j.biomaterials.2018.10.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 10/19/2018] [Indexed: 12/28/2022]
|
27
|
Birru B, Mekala NK, Parcha SR. Improved osteogenic differentiation of umbilical cord blood MSCs using custom made perfusion bioreactor. Biomed J 2018; 41:290-297. [PMID: 30580792 PMCID: PMC6306301 DOI: 10.1016/j.bj.2018.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 06/07/2018] [Accepted: 07/20/2018] [Indexed: 12/22/2022] Open
Abstract
Background 3D cell culture is an appropriate method to develop engineered bone tissue, where different bioreactors have been designed to mitigate the challenges in 3D culture. Currently, we tailored a perfusion reactor to witness human mesenchymal stem cells (MSCs) proliferation and differentiation over polylactic acid-polyethylene glycol (PLA/PEG) composite scaffolds. Methods The composite scaffolds with different weight ratios of PLA and PEG were prepared using solvent casting-particulate leaching technique. Human umbilcal card blood MSCs were cultured under dynamic and static conditions to elucidate the role of dynamic fluid flow in osteogenesis of MSCs. Results The human MSCs distribution over the scaffolds was confirmed with fluorescent microscopy. Alkaline phosphatase (ALP), calcium mineralization, and collagen formation were found to be higher in PLA90 scaffolds than PLA100 and PLA75. PLA90 scaffolds with better cell adhesion/proliferartion were considered for bioreactor studies and they exhibited enhanced ALP, Ca+2 mineralization and collagen formation under dynamic perfusion than static culture. We further confirmed our observation by looking at expression levels of osteogenic marker (Runx2 and osteonectin) in differentiated MSCs subjected to perfusion culture compared to static culture. Conclusion The results of the current investigation once again proves that dynamic perfusion cultures improve the osteogenic differentiation of MSCs over hybrid polymer scaffolds (PLA90) for effective bone regeneration.
Collapse
Affiliation(s)
- Bhaskar Birru
- Department of Biotechnology, National Institute of Technology Warangal, TS, India
| | | | | |
Collapse
|
28
|
Song J, Ye B, Liu H, Bi R, Zhang N, Hu J, Luo E. Fak-Mapk, Hippo and Wnt signalling pathway expression and regulation in distraction osteogenesis. Cell Prolif 2018; 51:e12453. [PMID: 29504176 PMCID: PMC6528869 DOI: 10.1111/cpr.12453] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 01/27/2018] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES To investigate the mechanism of mechanical stimulation in bone formation and regeneration during distraction osteogenesis. MATERIALS AND METHODS In this study, microarray technology was used to investigate the time course of bone-related molecular changes in distraction osteogenesis in rats. Real-time PCR and Western-blot analyses were used to confirm the expression of genes identified in microarrays. Meanwhile, we used a lentivirus vector to inhibit Fak expression, in order to identify the osteogenic effect of Fak and Fak-Mapk pathway during distraction osteogenesis. RESULTS Several components of the Wnt and Hippo pathways were found to be up- or down-regulated during distraction osteogenesis by microarray. Meanwhile, it was found that Fak, Src, Raf-1, Erk1, Jnk and p38-Mapk were up-regulated during gradual distraction, compared with consolidation. To further determine whether Fak-Mapk pathway played an important role in distraction osteogenesis, Fak was disrupted with a lentivirus vector. The expressions levels of p-Fak, p-Erk1/2, p-JNK and p-p38Mapk were decreased. Meanwhile, a poor early and late osteogenesis effect was found in the shRNA-Fak group. CONCLUSION It was inferred that the mechanical stimulus induces increased expression of Fak and activates Fak-Mapk pathway, by activation of Erk, Jnk and p38-Mapk pathway, and that Fak at least, in part, plays an important role in maintaining osteogenic effect by activating Fak-Mapk pathway during distraction osteogenesis.
Collapse
Affiliation(s)
- Jian Song
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Bin Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ruiye Bi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Nian Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Jing Hu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
29
|
The Tenascin-C-Derived Peptide VSWRAPTA Promotes Neuronal Branching Via Transcellular Activation of the Focal Adhesion Kinase (FAK) and the ERK1/2 Signaling Pathway In Vitro. Mol Neurobiol 2018; 56:632-647. [DOI: 10.1007/s12035-018-1108-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/03/2018] [Indexed: 12/27/2022]
|
30
|
Ratushnyy AY, Buravkova LB. Expression of focal adhesion genes in mesenchymal stem cells under simulated microgravity. DOKL BIOCHEM BIOPHYS 2018; 477:354-356. [PMID: 29297120 DOI: 10.1134/s1607672917060035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Indexed: 11/22/2022]
Abstract
The expression of 84 focal adhesion genes of multipotent mesenchymal stromal cells (MMSCs) after 96-h microgravity simulation at 3D clinorotation was studied. The upregulation of ITGA6, ITGA7, BCAR1, GRB2, CAV1, and DIAPH1 and the downregulation of ITGA11, ITGAV, ITGB1, PTEN, PTK2 (FAK), ARHGAP5, DOCK1, ROCK2, and AKT3 was found. These changes at the transcriptional level may be a cause of the reduction of the osteogenic potential of MMSCs and their ability to migration and adhesion in microgravity.
Collapse
Affiliation(s)
- A Yu Ratushnyy
- Institute for Biomedical Problems, Russian Academy of Sciences, Khoroshevskoe sh. 76a, Moscow, 123007, Russia
| | - L B Buravkova
- Institute for Biomedical Problems, Russian Academy of Sciences, Khoroshevskoe sh. 76a, Moscow, 123007, Russia.
| |
Collapse
|
31
|
Liu L, Luo Q, Sun J, Wang A, Shi Y, Ju Y, Morita Y, Song G. Decreased nuclear stiffness via FAK-ERK1/2 signaling is necessary for osteopontin-promoted migration of bone marrow-derived mesenchymal stem cells. Exp Cell Res 2017; 355:172-181. [PMID: 28392353 DOI: 10.1016/j.yexcr.2017.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 04/01/2017] [Accepted: 04/05/2017] [Indexed: 10/19/2022]
Abstract
Migration of bone marrow-derived mesenchymal stem cells (BMSCs) plays an important role in many physiological and pathological settings, including wound healing. During the migration of BMSCs through interstitial tissues, the movement of the nucleus must be coordinated with the cytoskeletal dynamics, which in turn affects the cell migration efficiency. Our previous study indicated that osteopontin (OPN) significantly promotes the migration of rat BMSCs. However, the nuclear behaviors and involved molecular mechanisms in OPN-mediated BMSC migration are largely unclear. In the present study, using an atomic force microscope (AFM), we found that OPN could decrease the nuclear stiffness of BMSCs and reduce the expression of lamin A/C, which is the main determinant of nuclear stiffness. Increased lamin A/C expression attenuates BMSC migration by increasing nuclear stiffness. Decreased lamin A/C expression promotes BMSC migration by decreasing nuclear stiffness. Furthermore, OPN promotes BMSC migration by diminishing lamin A/C expression and decreasing nuclear stiffness via the FAK-ERK1/2 signaling pathway. This study provides strong evidence for the role of nuclear mechanics in BMSC migration as well as new insight into the molecular mechanisms of OPN-promoted BMSC migration.
Collapse
Affiliation(s)
- Lingling Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, People's Republic of China.
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, People's Republic of China.
| | - Jinghui Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, People's Republic of China.
| | - Aoli Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, People's Republic of China.
| | - Yisong Shi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, People's Republic of China.
| | - Yang Ju
- Department of Mechanical Science and Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Yasuyuki Morita
- Department of Mechanical Science and Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, People's Republic of China.
| |
Collapse
|
32
|
Hadden WJ, Choi YS. The extracellular microscape governs mesenchymal stem cell fate. J Biol Eng 2016; 10:16. [PMID: 27895704 PMCID: PMC5117578 DOI: 10.1186/s13036-016-0037-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/09/2016] [Indexed: 12/15/2022] Open
Abstract
Each cell forever interacts with its extracellular matrix (ECM); a stem cell relies on this interaction to guide differentiation. The stiffness, nanotopography, protein composition, stress and strain inherent to any given ECM influences stem cell lineage commitment. This interaction is dynamic, multidimensional and reciprocally evolving through time, and from this concerted exchange the macroscopic tissues that comprise living organisms are formed. Mesenchymal stem cells can give rise to bone, cartilage, tendon and muscle; thus attempts to manipulate their differentiation must heed the physical properties of incredibly complex native microenvironments to realize regenerative goals.
Collapse
Affiliation(s)
- William J Hadden
- University of Sydney Medical School & Kolling Institute of Medical Research, Sydney, NSW Australia
| | - Yu Suk Choi
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Entrance 2, Hackett Dr, M309, Level 1, Crawley, WA 6009 Australia
| |
Collapse
|
33
|
Lynch ME, Chiou AE, Lee MJ, Marcott SC, Polamraju PV, Lee Y, Fischbach C. Three-Dimensional Mechanical Loading Modulates the Osteogenic Response of Mesenchymal Stem Cells to Tumor-Derived Soluble Signals. Tissue Eng Part A 2016; 22:1006-15. [PMID: 27401765 DOI: 10.1089/ten.tea.2016.0153] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Dynamic mechanical loading is a strong anabolic signal in the skeleton, increasing osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BM-MSCs) and increasing the bone-forming activity of osteoblasts, but its role in bone metastatic cancer is relatively unknown. In this study, we integrated a hydroxyapatite-containing three-dimensional (3D) scaffold platform with controlled mechanical stimulation to investigate the effects of cyclic compression on the interplay between breast cancer cells and BM-MSCs as it pertains to bone metastasis. BM-MSCs cultured within mineral-containing 3D poly(lactide-co-glycolide) (PLG) scaffolds differentiated into mature osteoblasts, and exposure to tumor-derived soluble factors promoted this process. When BM-MSCs undergoing osteogenic differentiation were exposed to conditioned media collected from mechanically loaded breast cancer cells, their gene expression of osteopontin was increased. This was further enhanced when mechanical compression was simultaneously applied to BM-MSCs, leading to more uniformly deposited osteopontin within scaffold pores. These results suggest that mechanical loading of 3D scaffold-based culture models may be utilized to evaluate the role of physiologically relevant physical cues on bone metastatic breast cancer. Furthermore, our data imply that cyclic mechanical stimuli within the bone microenvironment modulate interactions between tumor cells and BM-MSCs that are relevant to bone metastasis.
Collapse
Affiliation(s)
- Maureen E Lynch
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York.,2 Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst , Amherst, Massachusetts
| | - Aaron E Chiou
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Min Joon Lee
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Stephen C Marcott
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Praveen V Polamraju
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Yeonkyung Lee
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Claudia Fischbach
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University , Ithaca, New York.,3 Kavli Institute at Cornell for Nanoscale Science, Cornell University , Ithaca, New York
| |
Collapse
|
34
|
Guo R, Lu S, Merkel AR, Sterling JA, Guelcher SA. Substrate Modulus Regulates Osteogenic Differentiation of Rat Mesenchymal Stem Cells through Integrin β1 and BMP Receptor Type IA. J Mater Chem B 2016; 4:3584-3593. [PMID: 27551426 PMCID: PMC4991780 DOI: 10.1039/c5tb02747k] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Osteoblast differentiation of mesenchymal stem cells is regulated by both soluble factor (e.g., bone morphogenetic proteins (BMP)) and mechanically transduced signaling, but the mechanisms have only been partially elucidated. In this study, physical association of BMP Receptor I (BMPRI) with integrin β1 sub-unit (Iβ1) was hypothesized to mediate osteoblast differentiation of rat bone marrow-derived mesenchymal stem cells (MSCs) on bone-like substrates. The effects of substrate modulus on osteoblast differentiation of MSCs were investigated for 2D poly(ester urethane) films with moduli varying from 5 - 266 MPa, which spans the range from collagen fibrils to trabecular bone. SMAD1/5 and p44/42 MAPK signaling, expression of markers of osteoblast differentiation, and matrix mineralization increased with increasing substrate modulus. The effects of substrate modulus on osteoblast differentiation were mediated by Iβ1, which was also expressed at higher levels on increasingly rigid substrates. Förster resonance energy transfer (FRET) and immunoprecipitation (IP) experiments showed that physical association of Iβ1 and BMP Receptor I (BMRPRI) increased with substrate modulus, resulting in activation of the BMP signaling pathway. Thus, these studies showed that integrin and BMP signaling converge to regulate osteoblast differentiation of MSCs, which may potentially guide the design of scaffolds and rhBMP-2 delivery systems for bone regeneration.
Collapse
Affiliation(s)
- R Guo
- Department of Chemical and BIomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - S Lu
- Department of Chemical and BIomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - A R Merkel
- Department of Veterans Affairs: Tennessee Valley Healthcare System, Nashville, TN 37212, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - J A Sterling
- Department of Veterans Affairs: Tennessee Valley Healthcare System, Nashville, TN 37212, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - S A Guelcher
- Department of Chemical and BIomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
35
|
Kleinhans C, Mohan RR, Vacun G, Schwarz T, Haller B, Sun Y, Kahlig A, Kluger P, Finne-Wistrand A, Walles H, Hansmann J. A perfusion bioreactor system efficiently generates cell-loaded bone substitute materials for addressing critical size bone defects. Biotechnol J 2015; 10:1727-38. [PMID: 26011163 PMCID: PMC4744951 DOI: 10.1002/biot.201400813] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/20/2015] [Accepted: 05/19/2015] [Indexed: 12/28/2022]
Abstract
Critical size bone defects and non‐union fractions are still challenging to treat. Cell‐loaded bone substitutes have shown improved bone ingrowth and bone formation. However, a lack of methods for homogenously colonizing scaffolds limits the maximum volume of bone grafts. Additionally, therapy robustness is impaired by heterogeneous cell populations after graft generation. Our aim was to establish a technology for generating grafts with a size of 10.5 mm in diameter and 25 mm of height, and thus for grafts suited for treatment of critical size bone defects. Therefore, a novel tailor‐made bioreactor system was developed, allowing standardized flow conditions in a porous poly(L‐lactide‐co‐caprolactone) material. Scaffolds were seeded with primary human mesenchymal stem cells derived from four different donors. In contrast to static experimental conditions, homogenous cell distributions were accomplished under dynamic culture. Additionally, culture in the bioreactor system allowed the induction of osteogenic lineage commitment after one week of culture without addition of soluble factors. This was demonstrated by quantitative analysis of calcification and gene expression markers related to osteogenic lineage. In conclusion, the novel bioreactor technology allows efficient and standardized conditions for generating bone substitutes that are suitable for the treatment of critical size defects in humans.
Collapse
Affiliation(s)
- Claudia Kleinhans
- Institute for Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany.,Department of Orthopedics, Medical University Graz, Graz, Austria
| | - Ramkumar Ramani Mohan
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany.,Fraunhofer Institute for Interfacial Engineering and Biotechnology, Stuttgart, Germany Department
| | - Gabriele Vacun
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, Stuttgart, Germany Department
| | - Thomas Schwarz
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, Stuttgart, Germany Department
| | | | - Yang Sun
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Alexander Kahlig
- Institute for Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Petra Kluger
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, Stuttgart, Germany Department
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Heike Walles
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany.,Fraunhofer Institute for Interfacial Engineering and Biotechnology, Stuttgart, Germany Department
| | - Jan Hansmann
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany. .,Fraunhofer Institute for Interfacial Engineering and Biotechnology, Stuttgart, Germany Department.
| |
Collapse
|
36
|
RhoGTPases as key players in mammalian cell adaptation to microgravity. BIOMED RESEARCH INTERNATIONAL 2015; 2015:747693. [PMID: 25649831 PMCID: PMC4310447 DOI: 10.1155/2015/747693] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 08/14/2014] [Accepted: 09/09/2014] [Indexed: 01/03/2023]
Abstract
A growing number of studies are revealing that cells reorganize their cytoskeleton when exposed to conditions of microgravity. Most, if not all, of the structural changes observed on flown cells can be explained by modulation of RhoGTPases, which are mechanosensitive switches responsible for cytoskeletal dynamics control. This review identifies general principles defining cell sensitivity to gravitational stresses. We discuss what is known about changes in cell shape, nucleus, and focal adhesions and try to establish the relationship with specific RhoGTPase activities. We conclude by considering the potential relevance of live imaging of RhoGTPase activity or cytoskeletal structures in order to enhance our understanding of cell adaptation to microgravity-related conditions.
Collapse
|
37
|
Abstract
Bones adapt to accommodate the physical forces they experience through changes in architecture and mass. Stem cells differentiate into bone-forming osteoblasts, and mechanical stimulation is involved in this process. Various studies have applied controlled mechanical stimulation to stem cells and investigated the effects on osteogenic lineage commitment. These studies demonstrate that physical stimuli can induce osteogenic lineage commitment. Tension, fluid shear stress, substrate material properties, and cell shape are all factors that influence osteogenic differentiation. In particular, the level of tension is important. Also, rigid substrates with stiffness similar to collagenous bone induce osteogenic differentiation, while softer substrates induce other lineages. Finally, cells allowed to adhere over a larger area are able to differentiate towards the osteogenic lineage while cells adhering to a smaller area are restricted to the adipogenic lineage. Stem cells are able to sense their mechanical environments through various mechanosensors, including the cytoskeleton, focal adhesions, and primary cilia. The cytoskeleton provides a structural frame for the cell, and myosin interacts with actin to generate cytoskeletal tension, which is important for mechanically induced osteogenesis of stem cells. Adapter proteins link the cytoskeleton to integrins, which attach the cell to the substrate, forming a focal adhesion. A variety of signaling proteins are also associated with focal adhesions. Forces are transmitted to the substrate at these sites, and an intact focal adhesion is important for mechanically induced osteogenesis. The primary cilium is a single, immotile, antenna-like structure that extends from the cell into the extracellular space. It has emerged as an important signaling center, acting as a microdomain to facilitate biochemical signaling. Mechanotransduction is the process by which physical stimuli are converted into biochemical responses. When potential mechanosensors are disrupted, the activities of components of mechanotransduction pathways are also inhibited, preventing mechanically induced osteogenesis. Calcium, mitogen-activated protein kinase/extracellular signal-regulated kinase, Wnt, Yes-associated protein/transcriptional coactivator with PDZ-binding motif and RhoA/Rho kinase signaling are some of the mechanotransduction pathways proposed to be important. In this review, types of mechanical stimuli, mechanosensors, and key pathways involved in mechanically induced osteogenesis of stem cells are discussed.
Collapse
|
38
|
Ji J, Sun W, Wang W, Munyombwe T, Yang XB. The effect of mechanical loading on osteogenesis of human dental pulp stromal cells in a novel in vitro model. Cell Tissue Res 2014; 358:123-33. [PMID: 24916612 DOI: 10.1007/s00441-014-1907-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 04/28/2014] [Indexed: 12/13/2022]
Abstract
Tooth loss often results in alveolar bone resorption because of lack of mechanical stimulation. Thus, the mechanism of mechanical loading on stem cell osteogenesis is crucial for alveolar bone regeneration. We have investigated the effect of mechanical loading on osteogenesis in human dental pulp stromal cells (hDPSCs) in a novel in vitro model. Briefly, 1 × 10(7) hDPSCs were seeded into 1 ml 3% agarose gel in a 48-well-plate. A loading tube was then placed in the middle of the gel to mimic tooth-chewing movement (1 Hz, 3 × 30 min per day, n = 3). A non-loading group was used as a control. At various time points, the distribution of live/dead cells within the gel was confirmed by fluorescence markers and confocal microscopy. The correlation and interaction between the factors (e.g. force, time, depth and distance) were statistically analysed. The samples were processed for histology and immunohistochemistry. After 1-3 weeks of culture in the in-house-designed in vitro bioreactor, fluorescence imaging confirmed that additional mechanical loading increased the viable cell numbers over time as compared with the control. Cells of various phenotypes formed different patterns away from the reaction tube. The cells in the middle part of the gel showed enhanced alkaline phosphatase staining at week 1 but reduced staining at weeks 2 and 3. Additional loading enhanced Sirius Red and type I collagen staining compared with the control. We have thus successfully developed a novel in-house-designed in vitro bioreactor mimicking the biting force to enhance hDPSC osteogenesis in an agarose scaffold and to promote bone formation and/or prevent bone resorption.
Collapse
Affiliation(s)
- Jun Ji
- Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | | | | | | | | |
Collapse
|
39
|
Arfat Y, Xiao WZ, Iftikhar S, Zhao F, Li DJ, Sun YL, Zhang G, Shang P, Qian AR. Physiological effects of microgravity on bone cells. Calcif Tissue Int 2014; 94:569-79. [PMID: 24687524 DOI: 10.1007/s00223-014-9851-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 03/12/2014] [Indexed: 01/07/2023]
Abstract
Life on Earth developed under the influence of normal gravity (1g). With evidence from previous studies, scientists have suggested that normal physiological processes, such as the functional integrity of muscles and bone mass, can be affected by microgravity during spaceflight. During the life span, bone not only develops as a structure designed specifically for mechanical tasks but also adapts for efficiency. The lack of weight-bearing forces makes microgravity an ideal physical stimulus to evaluate bone cell responses. One of the most serious problems induced by long-term weightlessness is bone mineral loss. Results from in vitro studies that entailed the use of bone cells in spaceflights showed modification in cell attachment structures and cytoskeletal reorganization, which may be involved in bone loss. Humans exposed to microgravity conditions experience various physiological changes, including loss of bone mass, muscle deterioration, and immunodeficiency. In vitro models can be used to extract valuable information about changes in mechanical stress to ultimately identify the different pathways of mechanotransduction in bone cells. Despite many in vivo and in vitro studies under both real microgravity and simulated conditions, the mechanism of bone loss is still not well defined. The objective of this review is to summarize the recent research on bone cells under microgravity conditions based on advances in the field.
Collapse
Affiliation(s)
- Yasir Arfat
- Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Faculty of Life Sciences, Northwestern Polytechnical University, 127 Youyi Xilu, Xi'an, 710072, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hamamura K, Zhang P, Zhao L, Shim JW, Chen A, Dodge TR, Wan Q, Shih H, Na S, Lin CC, Sun HB, Yokota H. Knee loading reduces MMP13 activity in the mouse cartilage. BMC Musculoskelet Disord 2013; 14:312. [PMID: 24180431 PMCID: PMC3924329 DOI: 10.1186/1471-2474-14-312] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 10/18/2013] [Indexed: 11/17/2022] Open
Abstract
Background Moderate loads with knee loading enhance bone formation, but its effects on the maintenance of the knee are not well understood. In this study, we examined the effects of knee loading on the activity of matrix metalloproteinase13 (MMP13) and evaluated the role of p38 MAPK and Rac1 GTPase in the regulation of MMP13. Methods Knee loading (0.5–3 N for 5 min) was applied to the right knee of surgically-induced osteoarthritis (OA) mice as well as normal (non-OA) mice, and MMP13 activity in the femoral cartilage was examined. The sham-loaded knee was used as a non-loading control. We also employed primary non-OA and OA human chondrocytes as well as C28/I2 chondrocyte cells, and examined MMP13 activity and molecular signaling in response to shear at 2–20 dyn/cm2. Results Daily knee loading at 1 N for 2 weeks suppressed cartilage destruction in the knee of OA mice. Induction of OA elevated MMP13 activity and knee loading at 1 N suppressed this elevation. MMP13 activity was also increased in primary OA chondrocytes, and this increase was attenuated by applying shear at 10 dyn/cm2. Load-driven reduction in MMP13 was associated with a decrease in the phosphorylation level of p38 MAPK (p-p38) and NFκB (p-NFκB). Molecular imaging using a fluorescence resonance energy transfer (FRET) technique showed that Rac1 activity was reduced by shear at 10 dyn/cm2 and elevated by it at 20 dyn/cm2. Silencing Rac1 GTPase significantly reduced MMP13 expression and p-p38 but not p-NFκB. Transfection of a constitutively active Rac1 GTPase mutant increased MMP13 activity, while a dominant negative mutant decreased it. Conclusions Knee loading reduces MMP13 activity at least in part through Rac1-mediated p38 MAPK signaling. This study suggests the possibility of knee loading as a therapy not only for strengthening bone but also preventing tissue degradation of the femoral cartilage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, SL220C, 723 West Michigan Street, Indianapolis, IN 46202, USA.
| |
Collapse
|
41
|
New insights into adhesion signaling in bone formation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 305:1-68. [PMID: 23890379 DOI: 10.1016/b978-0-12-407695-2.00001-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mineralized tissues that are protective scaffolds in the most primitive species have evolved and acquired more specific functions in modern animals. These are as diverse as support in locomotion, ion homeostasis, and precise hormonal regulation. Bone formation is tightly controlled by a balance between anabolism, in which osteoblasts are the main players, and catabolism mediated by the osteoclasts. The bone matrix is deposited in a cyclic fashion during homeostasis and integrates several environmental cues. These include diffusible elements that would include estrogen or growth factors and physicochemical parameters such as bone matrix composition, stiffness, and mechanical stress. Therefore, the microenvironment is of paramount importance for controlling this delicate equilibrium. Here, we provide an overview of the most recent data highlighting the role of cell-adhesion molecules during bone formation. Due to the very large scope of the topic, we focus mainly on the role of the integrin receptor family during osteogenesis. Bone phenotypes of some deficient mice as well as diseases of human bones involving cell adhesion during this process are discussed in the context of bone physiology.
Collapse
|