1
|
Wang J, Yuan B, Yin R, Zhang H. Inflammation Responses to Bone Scaffolds under Mechanical Stimuli in Bone Regeneration. J Funct Biomater 2023; 14:jfb14030169. [PMID: 36976093 PMCID: PMC10059255 DOI: 10.3390/jfb14030169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/05/2023] [Accepted: 03/18/2023] [Indexed: 03/29/2023] Open
Abstract
Physical stimuli play an important role in one tissue engineering. Mechanical stimuli, such as ultrasound with cyclic loading, are widely used to promote bone osteogenesis; however, the inflammatory response under physical stimuli has not been well studied. In this paper, the signaling pathways related to inflammatory responses in bone tissue engineering are evaluated, and the application of physical stimulation to promote osteogenesis and its related mechanisms are reviewed in detail; in particular, how physical stimulation alleviates inflammatory responses during transplantation when employing a bone scaffolding strategy is discussed. It is concluded that physical stimulation (e.g., ultrasound and cyclic stress) helps to promote osteogenesis while reducing the inflammatory response. In addition, apart from 2D cell culture, more consideration should be given to the mechanical stimuli applied to 3D scaffolds and the effects of different force moduli while evaluating inflammatory responses. This will facilitate the application of physiotherapy in bone tissue engineering.
Collapse
Affiliation(s)
- Junjie Wang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Bo Yuan
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Ruixue Yin
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Hongbo Zhang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
2
|
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]
|
3
|
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
|
4
|
Naqvi SM, McNamara LM. Stem Cell Mechanobiology and the Role of Biomaterials in Governing Mechanotransduction and Matrix Production for Tissue Regeneration. Front Bioeng Biotechnol 2020; 8:597661. [PMID: 33381498 PMCID: PMC7767888 DOI: 10.3389/fbioe.2020.597661] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
Mechanobiology has underpinned many scientific advances in understanding how biophysical and biomechanical cues regulate cell behavior by identifying mechanosensitive proteins and specific signaling pathways within the cell that govern the production of proteins necessary for cell-based tissue regeneration. It is now evident that biophysical and biomechanical stimuli are as crucial for regulating stem cell behavior as biochemical stimuli. Despite this, the influence of the biophysical and biomechanical environment presented by biomaterials is less widely accounted for in stem cell-based tissue regeneration studies. This Review focuses on key studies in the field of stem cell mechanobiology, which have uncovered how matrix properties of biomaterial substrates and 3D scaffolds regulate stem cell migration, self-renewal, proliferation and differentiation, and activation of specific biological responses. First, we provide a primer of stem cell biology and mechanobiology in isolation. This is followed by a critical review of key experimental and computational studies, which have unveiled critical information regarding the importance of the biophysical and biomechanical cues for stem cell biology. This review aims to provide an informed understanding of the intrinsic role that physical and mechanical stimulation play in regulating stem cell behavior so that researchers may design strategies that recapitulate the critical cues and develop effective regenerative medicine approaches.
Collapse
Affiliation(s)
- S M Naqvi
- Mechanobiology and Medical Device Research Group, Department of Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - L M McNamara
- Mechanobiology and Medical Device Research Group, Department of Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
5
|
Potekhina Y, Filatova A, Tregubova E, Mokhov D. Mechanosensitivity of Cells and Its Role in the Regulation of Physiological Functions and the Implementation of Physiotherapeutic Effects (Review). Sovrem Tekhnologii Med 2020; 12:77-89. [PMID: 34795996 PMCID: PMC8596276 DOI: 10.17691/stm2020.12.4.10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Indexed: 01/11/2023] Open
Abstract
Regulatory signals in the body are not limited to chemical and electrical ones. There is another type of important signals for cells: those are mechanical signals (coming from the environment or arising from within the body), which have been less known in the literature. The review summarizes new information on the mechanosensitivity of various cells of connective tissue and nervous system. Participation of mechanical stimuli in the regulation of growth, development, differentiation, and functioning of tissues is described. The data focus on bone remodeling, wound healing, neurite growth, and the formation of neural networks. Mechanotransduction, cellular organelles, and mechanosensitive molecules involved in these processes are discussed as well as the role of the extracellular matrix. The importance of mechanical characteristics of cells in the pathogenesis of diseases is highlighted. Finally, the possible role of mechanosensitivity in mediating the physiotherapeutic effects is addressed.
Collapse
Affiliation(s)
- Yu.P. Potekhina
- Professor, Department of Normal Physiology named after N.Y. Belenkov; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - A.I. Filatova
- Student, Faculty of Pediatrics; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - E.S. Tregubova
- Professor, Department of Osteopathy; North-Western State Medical University named after I.I. Mechnikov, 41 Kirochnaya St., Saint Petersburg, 191015, Russia; Associate Professor, Institute of Osteopathy; Saint Petersburg State University, 7/9 Universitetskaya naberezhnaya, Saint Petersburg, 199034, Russia
| | - D.E. Mokhov
- Head of the Department of Osteopathy; North-Western State Medical University named after I.I. Mechnikov, 41 Kirochnaya St., Saint Petersburg, 191015, Russia; Director of the Institute of Osteopathy Saint Petersburg State University, 7/9 Universitetskaya naberezhnaya, Saint Petersburg, 199034, Russia
| |
Collapse
|
6
|
Luo Y, Ding X, Ji H, Li M, Song H, Li S, Wang C, Wu H, Du H. MicroRNA-503-3p affects osteogenic differentiation of human adipose-derived stem cells by regulation of Wnt2 and Wnt7b under cyclic strain. Stem Cell Res Ther 2020; 11:318. [PMID: 32711579 PMCID: PMC7382842 DOI: 10.1186/s13287-020-01842-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/05/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) play a role in regulating osteogenic differentiation (OD) of mesenchymal stem cells by inhibiting mRNAs translation under cyclic strain. miR-503-3p was downregulated in OD of human adipose-derived stem cells (hASCs) in vivo under cyclic strain in our previous study, while it might target the Wnt/β-catenin (W-β) pathway. In this study, we explored miR-503-3p's role in OD of hASCs under cyclic strain. METHODS OD of hASCs was induced by cyclic strain. Bioinformatic and dual luciferase analyses were used to confirm the relationship between Wnt2/Wnt7b and miR-503-3p. Immunofluorescence was used to detect the effect of miR-503-3p on Wnt2/Wnt7b and β-catenin in hASCs transfected with miR-503-3p mimic and inhibitor. Mimic, inhibitor, and small interfering RNA (siRNA) transfected in hASCs to against Wnt2 and Wnt7b. Quantitative real-time PCR (RT-PCR) and western blot were used to examine the OD and W-β pathway at the mRNA and protein levels, respectively. Immunofluorescence was performed to locate β-catenin. ALP activity and calcium were detected by colorimetric assay. RESULTS Results of immunophenotypes by flow cytometry and multi-lineage potential confirmed that the cultured cells were hASCs. Results of luciferase reporter assay indicated that miR-503-3p could regulate the expression levels of Wnt2 and Wnt7b by targeting their respective 3'-untranslated region (UTR). Under cyclic strain, gain- or loss-function of miR-503-3p studies by mimic and inhibitor revealed that decreasing expression of miR-503-3p could significantly bring about promotion of OD of hASCs, whereas increased expression of miR-503-3p inhibited OD. Furthermore, miR-503-3p high-expression reduced the activity of the W-β pathway, as indicated by lowering expression of Wnt2 and Wnt7b, inactive β-catenin in miR-503-3p-treated hASCs. By contrast, miR-503-3p inhibition activated the W-β pathway. CONCLUSIONS Collectively, our findings indicate that miR-503-3p is a negative factor in regulating W-β pathway by Wnt2 and Wnt7b, which inhibit the OD of hASCs under cyclic strain.
Collapse
Affiliation(s)
- Yadong Luo
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Xu Ding
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Huan Ji
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Meng Li
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Haiyang Song
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Sheng Li
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Chenxing Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Heming Wu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Hongming Du
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China. .,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China.
| |
Collapse
|
7
|
|
8
|
Luo Y, Ge R, Wu H, Ding X, Song H, Ji H, Li M, Ma Y, Li S, Wang C, Du H. The osteogenic differentiation of human adipose-derived stem cells is regulated through the let-7i-3p/LEF1/β-catenin axis under cyclic strain. Stem Cell Res Ther 2019; 10:339. [PMID: 31753039 PMCID: PMC6873506 DOI: 10.1186/s13287-019-1470-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023] Open
Abstract
Background The Wnt/β-catenin pathway is involved in the osteogenic differentiation of human adipose-derived stem cells (hASCs) under cyclic strain. Very little is known about the role of microRNAs in these events. Methods Cells were obtained using enzyme digestion methods, and proliferation was detected using Cell Counting Kit 8. Cell cycles and immunophenotypes were detected by flow cytometry. The multilineage potential of hASCs was induced by induction media. Cyclic strain was applied to hASCs (0.5 Hz, 2 h/day, 6 days) to induce osteogenic differentiation and miRNA changes. Bioinformatic and dual-luciferase analyses confirmed lymphoid enhancer factor 1 (LEF1) as a potential target of let-7i-3p. The effect of let-7i-3p on LEF1 in hASCs transfected with a let-7i-3p mimic and inhibitor was analyzed by immunofluorescence. hASCs were transfected with a let-7i-3p mimic, inhibitor, or small interfering RNA (siRNA) against LEF1 and β-catenin. Quantitative real-time PCR (qPCR) and western blotting were performed to examine the osteogenic markers and Wnt/β-catenin pathway at the mRNA and protein levels, respectively. Immunofluorescence and western blotting were performed to confirm the activation of the Wnt/β-catenin pathway. Results Flow cytometry showed that 82.12% ± 5.83% of the cells were in G1 phase and 17.88% ± 2.59% of the cells were in S/G2 phase; hASCs were positive for CD29, CD90, and CD105. hASCs could have the potential for osteogenic, chondrogenic, and adipogenic differentiation. MicroRNA screening via microarray showed that let-7i-3p expression was decreased under cyclic strain. Bioinformatic and dual-luciferase analyses confirmed that LEF1 in the Wnt/β-catenin pathway was the target of let-7i-3p. Under cyclic strain, the osteogenic differentiation of hASCs was promoted by overexpression of LEF1and β-catenin and inhibited by overexpression of let-7i-3p. hASCs were transfected with let-7i-3p mimics and inhibitor. Gain- or loss-of-function analyses of let-7i-3p showed that the osteogenic differentiation of hASCs was promoted by decreased let-7i-3p expression and inhibited by increased let-7i-3p expression. Furthermore, high LEF1 expression inactivated the Wnt/β-catenin pathway in let-7i-3p-enhanced hASCs. In contrast, let-7i-3p inhibition activated the Wnt/β-catenin pathway. Conclusions Let-7i-3p, acting as a negative regulator of the Wnt/β-catenin pathway by targeting LEF1, inhibits the osteogenic differentiation of hASCs under cyclic strain in vitro.
Collapse
Affiliation(s)
- Yadong Luo
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Oral Disease Key Laboratory of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Ran Ge
- Department of Nuclear Medicine, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, People's Republic of China
| | - Heming Wu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Oral Disease Key Laboratory of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Xu Ding
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Oral Disease Key Laboratory of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Haiyang Song
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Oral Disease Key Laboratory of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Huan Ji
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Oral Disease Key Laboratory of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Meng Li
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Oral Disease Key Laboratory of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Yunan Ma
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Oral Disease Key Laboratory of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Sheng Li
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Oral Disease Key Laboratory of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Chenxing Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Oral Disease Key Laboratory of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Hongming Du
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China. .,Oral Disease Key Laboratory of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China.
| |
Collapse
|
9
|
Nordberg RC, Mellor LF, Krause AR, Donahue HJ, Loboa EG. LRP receptors in chondrocytes are modulated by simulated microgravity and cyclic hydrostatic pressure. PLoS One 2019; 14:e0223245. [PMID: 31584963 PMCID: PMC6777824 DOI: 10.1371/journal.pone.0223245] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 09/17/2019] [Indexed: 01/16/2023] Open
Abstract
Mechanical loading is essential for the maintenance of musculoskeletal homeostasis. Cartilage has been demonstrated to be highly mechanoresponsive, but the mechanisms by which chondrocytes respond to mechanical stimuli are not clearly understood. The goal of the study was to determine how LRP4, LRP5, and LRP6 within canonical Wnt-signaling are regulated in simulated microgravity and cyclic hydrostatic pressure, and to investigate the potential role of LRP 4/5/6 in cartilage degeneration. Rat chondrosacroma cell (RCS) pellets were stimulated using either cyclic hydrostatic pressure (1Hz, 7.5 MPa, 4hr/day) or simulated microgravity in a rotating wall vessel (RWV) bioreactor (11RPM, 24hr/day). LRP4/5/6 mRNA expression was assessed by RT-qPCR and LRP5 protein expression was determined by fluorescent immunostaining. To further evaluate our in vitro findings in vivo, mice were subjected to hindlimb suspension for 14 days and the femoral heads stained for LRP5 expression. We found that, in vitro, LRP4/5/6 mRNA expression is modulated in a time-dependent manner by mechanical stimulation. Additionally, LRP5 protein expression is upregulated in response to both simulated microgravity and cyclic hydrostatic pressure. LRP5 is also upregulated in vivo in the articular cartilage of hindlimb suspended mice. This is the first study to examine how LRP4/5/6, critical receptors within musculoskeletal biology, respond to mechanical stimulation. Further elucidation of this mechanism could provide significant clinical benefit for the identification of pharmaceutical targets for the maintenance of cartilage health.
Collapse
Affiliation(s)
- Rachel C. Nordberg
- College of Engineering, University of Missouri, Columbia, Missouri, United States of America
| | | | - Andrew R. Krause
- Sport Health and Physical Education, Vancouver Island University, Nanaimo, British Columbia, Canada
| | - Henry J. Donahue
- Division of Musculoskeletal Sciences, Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Elizabeth G. Loboa
- College of Engineering, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
10
|
Bodle J, Hamouda MS, Cai S, Williams RB, Bernacki SH, Loboa EG. Primary Cilia Exhibit Mechanosensitivity to Cyclic Tensile Strain and Lineage-Dependent Expression in Adipose-Derived Stem Cells. Sci Rep 2019; 9:8009. [PMID: 31142808 PMCID: PMC6541635 DOI: 10.1038/s41598-019-43351-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Non-motile primary cilia are dynamic cellular sensory structures and are expressed in adipose-derived stem cells (ASCs). We have previously shown that primary cilia are involved in chemically-induced osteogenic differentiation of human ASC (hASCs) in vitro. Further, we have reported that 10% cyclic tensile strain (1 Hz, 4 hours/day) enhances hASC osteogenesis. We hypothesize that primary cilia respond to cyclic tensile strain in a lineage dependent manner and that their mechanosensitivity may regulate the dynamics of signaling pathways localized to the cilium. We found that hASC morphology, cilia length and cilia conformation varied in response to culture in complete growth, osteogenic differentiation, or adipogenic differentiation medium, with the longest cilia expressed in adipogenically differentiating cells. Further, we show that cyclic tensile strain both enhances osteogenic differentiation of hASCs while it suppresses adipogenic differentiation as evidenced by upregulation of RUNX2 gene expression and downregulation of PPARG and IGF-1, respectively. This study demonstrates that hASC primary cilia exhibit mechanosensitivity to cyclic tensile strain and lineage-dependent expression, which may in part regulate signaling pathways localized to the primary cilium during the differentiation process. We highlight the importance of the primary cilium structure in mechanosensing and lineage specification and surmise that this structure may be a novel target in manipulating hASC for in tissue engineering applications.
Collapse
Affiliation(s)
- Josephine Bodle
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, 27695, USA.
| | - Mehdi S Hamouda
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Shaobo Cai
- Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Ramey B Williams
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Susan H Bernacki
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Elizabeth G Loboa
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, 27695, USA.
- College of Engineering at University of Missouri, W1051 Thomas & Nell Lafferre Hall, Columbia, MO, 65211, USA.
| |
Collapse
|
11
|
Mechanical strain promotes osteogenic differentiation of mesenchymal stem cells on TiO2 nanotubes substrate. Biochem Biophys Res Commun 2019; 511:840-846. [DOI: 10.1016/j.bbrc.2019.02.145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 02/27/2019] [Indexed: 12/31/2022]
|
12
|
Huang X, Das R, Patel A, Nguyen TD. Physical Stimulations for Bone and Cartilage Regeneration. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018; 4:216-237. [PMID: 30740512 PMCID: PMC6366645 DOI: 10.1007/s40883-018-0064-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/07/2018] [Indexed: 12/26/2022]
Abstract
A wide range of techniques and methods are actively invented by clinicians and scientists who are dedicated to the field of musculoskeletal tissue regeneration. Biological, chemical, and physiological factors, which play key roles in musculoskeletal tissue development, have been extensively explored. However, physical stimulation is increasingly showing extreme importance in the processes of osteogenic and chondrogenic differentiation, proliferation and maturation through defined dose parameters including mode, frequency, magnitude, and duration of stimuli. Studies have shown manipulation of physical microenvironment is an indispensable strategy for the repair and regeneration of bone and cartilage, and biophysical cues could profoundly promote their regeneration. In this article, we review recent literature on utilization of physical stimulation, such as mechanical forces (cyclic strain, fluid shear stress, etc.), electrical and magnetic fields, ultrasound, shock waves, substrate stimuli, etc., to promote the repair and regeneration of bone and cartilage tissue. Emphasis is placed on the mechanism of cellular response and the potential clinical usage of these stimulations for bone and cartilage regeneration.
Collapse
|
13
|
Chiu CH, Tong YW, Yeh WL, Lei KF, Chen ACY. Self-Renewal and Differentiation of Adipose-Derived Stem Cells (ADSCs) Stimulated by Multi-Axial Tensile Strain in a Pneumatic Microdevice. MICROMACHINES 2018; 9:607. [PMID: 30463251 PMCID: PMC6267491 DOI: 10.3390/mi9110607] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 02/06/2023]
Abstract
Adipose-derived stem cells (ADSCs) were suggested for treating degenerative osteoarthritis, suppressing inflammatory responses, and repairing damaged soft tissues. Moreover, the ADSCs have the potential to undergo self-renewal and differentiate into bone, tendon, cartilage, and ligament. Recently, investigation of the self-renewal and differentiation of the ADSCs has become an attractive area. In this work, a pneumatic microdevice has been developed to study the gene expression of the ADSCs after the stimulation of multi-axial tensile strain. The ADSCs were cultured on the microdevice and experienced multi-axial tensile strain during a three-day culture course. Self-renewal and differentiation abilities were investigated by mRNA expressions of NANOG, sex determining region Y-box 2 (SOX2), octamer-binding transcription factor 4 (OCT4), sex determining region Y-box9 (SOX9), peroxisome proliferator-activated receptor gamma (PPAR-γ), and runt-related transcription factor 2 (RUNX2). The result showed that the genes related self-renewal were significantly up-regulated after the tensile stimulation. Higher proliferation ratio of the ADSCs was also shown by cell viability assay. The microdevice provides a promising platform for cell-based study under mechanical tensile stimulation.
Collapse
Affiliation(s)
- Chih-Hao Chiu
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan.
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| | - Yun-Wen Tong
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan.
| | - Wen-Ling Yeh
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| | - Kin Fong Lei
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan 333, Taiwan.
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| | - Alvin Chao-Yu Chen
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| |
Collapse
|
14
|
Polo-Corrales L, Ramirez-Vick J, Feria-Diaz JJ. Recent Advances in Biophysical stimulation of MSC for bone regeneration. ACTA ACUST UNITED AC 2018. [DOI: 10.17485/ijst/2018/v11i15/121405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
15
|
Nordberg RC, Bodle JC, Loboa EG. Mechanical Stimulation of Adipose-Derived Stem Cells for Functional Tissue Engineering of the Musculoskeletal System via Cyclic Hydrostatic Pressure, Simulated Microgravity, and Cyclic Tensile Strain. Methods Mol Biol 2018; 1773:215-230. [PMID: 29687393 DOI: 10.1007/978-1-4939-7799-4_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
It is critical that human adipose stem cell (hASC) tissue-engineering therapies possess appropriate mechanical properties in order to restore function of the load bearing tissues of the musculoskeletal system. In an effort to elucidate the hASC response to mechanical stimulation and develop mechanically robust tissue engineered constructs, recent research has utilized a variety of mechanical loading paradigms including cyclic tensile strain, cyclic hydrostatic pressure, and mechanical unloading in simulated microgravity. This chapter describes methods for applying these mechanical stimuli to hASC to direct differentiation for functional tissue engineering of the musculoskeletal system.
Collapse
Affiliation(s)
- Rachel C Nordberg
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina Chapel Hill, Raleigh, NC, USA
| | - Josie C Bodle
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina Chapel Hill, Raleigh, NC, USA
| | - Elizabeth G Loboa
- College of Engineering, University of Missouri, W1024 Thomas & Nell Lafferre Hall, Columbia, MO, USA.
| |
Collapse
|
16
|
Pham MH, Buser Z, Wang JC, Acosta FL. Low-magnitude mechanical signals and the spine: A review of current and future applications. J Clin Neurosci 2017; 40:18-23. [DOI: 10.1016/j.jocn.2016.12.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 12/27/2016] [Indexed: 01/17/2023]
|
17
|
Virjula S, Zhao F, Leivo J, Vanhatupa S, Kreutzer J, Vaughan TJ, Honkala AM, Viehrig M, Mullen CA, Kallio P, McNamara LM, Miettinen S. The effect of equiaxial stretching on the osteogenic differentiation and mechanical properties of human adipose stem cells. J Mech Behav Biomed Mater 2017; 72:38-48. [PMID: 28448920 DOI: 10.1016/j.jmbbm.2017.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/05/2017] [Accepted: 04/12/2017] [Indexed: 01/22/2023]
Abstract
Although mechanical cues are known to affect stem cell fate and mechanobiology, the significance of such stimuli on the osteogenic differentiation of human adipose stem cells (hASCs) remains unclear. In this study, we investigated the effect of long-term mechanical stimulation on the attachment, osteogenic differentiation and mechanical properties of hASCs. Tailor-made, pneumatic cell stretching devices were used to expose hASCs to cyclic equiaxial stretching in osteogenic medium. Cell attachment and focal adhesions were visualised using immunocytochemical vinculin staining on days 3 and 6, and the proliferation and alkaline phosphatase activity, as a sign of early osteogenic differentiation, were analysed on days 0, 6 and 10. Furthermore, the mechanical properties of hASCs, in terms of apparent Young's modulus and normalised contractility, were obtained using a combination of atomic force microscopy based indentation and computational approaches. Our results indicated that cyclic equiaxial stretching delayed proliferation and promoted osteogenic differentiation of hASCs. Stretching also reduced cell size and intensified focal adhesions and actin cytoskeleton. Moreover, cell stiffening was observed during osteogenic differentiation and especially under mechanical stimulation. These results suggest that cyclic equiaxial stretching modifies cell morphology, focal adhesion formation and mechanical properties of hASCs. This could be exploited to enhance osteogenic differentiation.
Collapse
Affiliation(s)
- Sanni Virjula
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Life Sciences, University of Tampere, Lääkärinkatu 1, 33520 Tampere, Finland; Science Centre, Tampere University Hospital, Biokatu 6, 33520 Tampere, Finland.
| | - Feihu Zhao
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland.
| | - Joni Leivo
- Department of Automation Science and Engineering, BioMediTech, Tampere University of Technology, Korkeakoulunkatu 3, 33720 Tampere, Finland.
| | - Sari Vanhatupa
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Life Sciences, University of Tampere, Lääkärinkatu 1, 33520 Tampere, Finland; Science Centre, Tampere University Hospital, Biokatu 6, 33520 Tampere, Finland.
| | - Joose Kreutzer
- Department of Automation Science and Engineering, BioMediTech, Tampere University of Technology, Korkeakoulunkatu 3, 33720 Tampere, Finland.
| | - Ted J Vaughan
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland.
| | - Anna-Maija Honkala
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Life Sciences, University of Tampere, Lääkärinkatu 1, 33520 Tampere, Finland; Science Centre, Tampere University Hospital, Biokatu 6, 33520 Tampere, Finland.
| | - Marlitt Viehrig
- Department of Automation Science and Engineering, BioMediTech, Tampere University of Technology, Korkeakoulunkatu 3, 33720 Tampere, Finland.
| | - Conleth A Mullen
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland.
| | - Pasi Kallio
- Department of Automation Science and Engineering, BioMediTech, Tampere University of Technology, Korkeakoulunkatu 3, 33720 Tampere, Finland.
| | - Laoise M McNamara
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland.
| | - Susanna Miettinen
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Life Sciences, University of Tampere, Lääkärinkatu 1, 33520 Tampere, Finland; Science Centre, Tampere University Hospital, Biokatu 6, 33520 Tampere, Finland.
| |
Collapse
|
18
|
Nordberg RC, Zhang J, Griffith EH, Frank MW, Starly B, Loboa EG. Electrical Cell-Substrate Impedance Spectroscopy Can Monitor Age-Grouped Human Adipose Stem Cell Variability During Osteogenic Differentiation. Stem Cells Transl Med 2016; 6:502-511. [PMID: 28191763 PMCID: PMC5442814 DOI: 10.5966/sctm.2015-0404] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 07/28/2016] [Indexed: 01/01/2023] Open
Abstract
Human adipose stem cells (hASCs) are an attractive cell source for bone tissue engineering applications. However, a critical issue to be addressed before widespread hASC clinical translation is the dramatic variability in proliferative capacity and osteogenic potential among hASCs isolated from different donors. The goal of this study was to test our hypothesis that electrical cell-substrate impedance spectroscopy (ECIS) could track complex bioimpedance patterns of hASCs throughout proliferation and osteogenic differentiation to better understand and predict variability among hASC populations. Superlots composed of hASCs from young (aged 24-36 years), middle-aged (aged 48-55 years), and elderly (aged 60-81 years) donors were seeded on gold electrode arrays. Complex impedance measurements were taken throughout proliferation and osteogenic differentiation. During osteogenic differentiation, four impedance phases were identified: increase, primary stabilization, drop phase, and secondary stabilization. Matrix deposition was first observed 48-96 hours after the impedance maximum, indicating, for the first time, that ECIS can identify morphological changes that correspond to late-stage osteogenic differentiation. The impedance maximum was observed at day 10.0 in young, day 6.1 in middle-aged, and day 1.3 in elderly hASCs, suggesting that hASCs from younger donors require a longer time to differentiate than do hASCs from older donors, but young hASCs proliferated more and accreted more calcium long-term. This is the first study to use ECIS to predict osteogenic potential of multiple hASC populations and to show that donor age may temporally control onset of osteogenesis. These findings could be critical for development of patient-specific bone tissue engineering and regenerative medicine therapies. Stem Cells Translational Medicine 2017;6:502-511.
Collapse
Affiliation(s)
- Rachel C. Nordberg
- Joint Department of Biomedical Engineering, University of North Carolina Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Jianlei Zhang
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Emily H. Griffith
- Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | - Matthew W. Frank
- Joint Department of Biomedical Engineering, University of North Carolina Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Binil Starly
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Elizabeth G. Loboa
- Joint Department of Biomedical Engineering, University of North Carolina Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
- University of Missouri College of Engineering, Columbia, Missouri, USA
| |
Collapse
|
19
|
Steward AJ, Cole JH, Ligler FS, Loboa EG. Mechanical and Vascular Cues Synergistically Enhance Osteogenesis in Human Mesenchymal Stem Cells. Tissue Eng Part A 2016; 22:997-1005. [PMID: 27392567 DOI: 10.1089/ten.tea.2015.0533] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Development and maintenance of a vascular network are critical for bone growth and homeostasis; strategies that promote vascular function are critical for clinical success of tissue-engineered bone constructs. Co-culture of endothelial cells (ECs) with mesenchymal stem cells (MSCs) and exposure to 10% cyclic tensile strain have both been shown to regulate osteogenesis in isolation, but potential synergistic effects have yet to be explored. The objective of this study was to expose an MSC-EC co-culture to 10% cyclic tensile strain to examine the role of this mechanical stimulus on MSC-EC behavior. We hypothesized that paracrine signaling from ECs would stimulate osteogenesis of MSCs, and exposure to 10% cyclic tensile strain would enhance this anabolic signal. Human umbilical vein ECs and human bone marrow-derived MSCs were either monocultured or co-cultured at a 1:1 ratio in a mixed osteo/angiogenic medium, exposed to 10% cyclic tensile strain at 1 Hz for 4 h/day for 2 weeks, and biochemically and histologically analyzed for endothelial and osteogenic markers. While neither 10% cyclic tensile strain nor co-culture alone had a significant effect on osteogenesis, the concurrent application of strain to an MSC-EC co-culture resulted in a significant increase in calcium accretion and mineral deposition, suggesting that co-culture and strain synergistically enhance osteogenesis. Neither co-culture, 10% cyclic tensile strain, nor a combination of these stimuli affected endothelial markers, indicating that the endothelial phenotype remained stable, but unresponsive to the stimuli evaluated in this study. This study is the first to investigate the role of cyclic tensile strain on the complex interplay between ECs and MSCs in co-culture. The results of this study provide key insights into the synergistic effects of 10% cyclic tensile strain and co-culture on osteogenesis. Understanding mechanobiological factors affecting MSC-EC crosstalk will help enhance strategies for creating vascularized tissues in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Andrew J Steward
- 1 Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University , Raleigh, North Carolina
| | - Jacqueline H Cole
- 1 Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University , Raleigh, North Carolina
| | - Frances S Ligler
- 1 Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University , Raleigh, North Carolina
| | - Elizabeth G Loboa
- 1 Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University , Raleigh, North Carolina.,2 College of Engineering, University of Missouri , Columbia, Missouri
| |
Collapse
|
20
|
Tuin SA, Pourdeyhimi B, Loboa EG. Fabrication of novel high surface area mushroom gilled fibers and their effects on human adipose derived stem cells under pulsatile fluid flow for tissue engineering applications. Acta Biomater 2016; 36:220-30. [PMID: 26992369 DOI: 10.1016/j.actbio.2016.03.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/08/2016] [Accepted: 03/14/2016] [Indexed: 01/20/2023]
Abstract
UNLABELLED The fabrication and characterization of novel high surface area hollow gilled fiber tissue engineering scaffolds via industrially relevant, scalable, repeatable, high speed, and economical nonwoven carding technology is described. Scaffolds were validated as tissue engineering scaffolds using human adipose derived stem cells (hASC) exposed to pulsatile fluid flow (PFF). The effects of fiber morphology on the proliferation and viability of hASC, as well as effects of varied magnitudes of shear stress applied via PFF on the expression of the early osteogenic gene marker runt related transcription factor 2 (RUNX2) were evaluated. Gilled fiber scaffolds led to a significant increase in proliferation of hASC after seven days in static culture, and exhibited fewer dead cells compared to pure PLA round fiber controls. Further, hASC-seeded scaffolds exposed to 3 and 6dyn/cm(2) resulted in significantly increased mRNA expression of RUNX2 after one hour of PFF in the absence of soluble osteogenic induction factors. This is the first study to describe a method for the fabrication of high surface area gilled fibers and scaffolds. The scalable manufacturing process and potential fabrication across multiple nonwoven and woven platforms makes them promising candidates for a variety of applications that require high surface area fibrous materials. STATEMENT OF SIGNIFICANCE We report here for the first time the successful fabrication of novel high surface area gilled fiber scaffolds for tissue engineering applications. Gilled fibers led to a significant increase in proliferation of human adipose derived stem cells after one week in culture, and a greater number of viable cells compared to round fiber controls. Further, in the absence of osteogenic induction factors, gilled fibers led to significantly increased mRNA expression of an early marker for osteogenesis after exposure to pulsatile fluid flow. This is the first study to describe gilled fiber fabrication and their potential for tissue engineering applications. The repeatable, industrially scalable, and versatile fabrication process makes them promising candidates for a variety of scaffold-based tissue engineering applications.
Collapse
|
21
|
Trumbull A, Subramanian G, Yildirim-Ayan E. Mechanoresponsive musculoskeletal tissue differentiation of adipose-derived stem cells. Biomed Eng Online 2016; 15:43. [PMID: 27103394 PMCID: PMC4840975 DOI: 10.1186/s12938-016-0150-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/24/2016] [Indexed: 02/06/2023] Open
Abstract
Musculoskeletal tissues are constantly under mechanical strains within their microenvironment. Yet, little is understood about the effect of in vivo mechanical milieu strains on cell development and function. Thus, this review article outlines the in vivo mechanical environment of bone, muscle, cartilage, tendon, and ligaments, and tabulates the mechanical strain and stress in these tissues during physiological condition, vigorous, and moderate activities. This review article further discusses the principles of mechanical loading platforms to create physiologically relevant mechanical milieu in vitro for musculoskeletal tissue regeneration. A special emphasis is placed on adipose-derived stem cells (ADSCs) as an emerging valuable tool for regenerative musculoskeletal tissue engineering, as they are easily isolated, expanded, and able to differentiate into any musculoskeletal tissue. Finally, it highlights the current state-of-the art in ADSCs-guided musculoskeletal tissue regeneration under mechanical loading.
Collapse
Affiliation(s)
- Andrew Trumbull
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, 43606, USA
| | - Gayathri Subramanian
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, 43606, USA
| | - Eda Yildirim-Ayan
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, 43606, USA. .,Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, OH, 43614, USA.
| |
Collapse
|
22
|
Font Tellado S, Balmayor ER, Van Griensven M. Strategies to engineer tendon/ligament-to-bone interface: Biomaterials, cells and growth factors. Adv Drug Deliv Rev 2015; 94:126-40. [PMID: 25777059 DOI: 10.1016/j.addr.2015.03.004] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/27/2015] [Accepted: 03/07/2015] [Indexed: 02/06/2023]
Abstract
Integration between tendon/ligament and bone occurs through a specialized tissue interface called enthesis. The complex and heterogeneous structure of the enthesis is essential to ensure smooth mechanical stress transfer between bone and soft tissues. Following injury, the interface is not regenerated, resulting in high rupture recurrence rates. Tissue engineering is a promising strategy for the regeneration of a functional enthesis. However, the complex structural and cellular composition of the native interface makes enthesis tissue engineering particularly challenging. Thus, it is likely that a combination of biomaterials and cells stimulated with appropriate biochemical and mechanical cues will be needed. The objective of this review is to describe the current state-of-the-art, challenges and future directions in the field of enthesis tissue engineering focusing on four key parameters: (1) scaffold and biomaterials, (2) cells, (3) growth factors and (4) mechanical stimuli.
Collapse
Affiliation(s)
- Sonia Font Tellado
- Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany.
| | - Elizabeth R Balmayor
- Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany
| | - Martijn Van Griensven
- Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany
| |
Collapse
|
23
|
Nordberg RC, Loboa EG. Our Fat Future: Translating Adipose Stem Cell Therapy. Stem Cells Transl Med 2015; 4:974-9. [PMID: 26185256 DOI: 10.5966/sctm.2015-0071] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/17/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Human adipose stem cells (hASCs) have the potential to treat patients with a variety of clinical conditions. Recent advancements in translational research, regulatory policy, and industry have positioned hASCs on the threshold of clinical translation. We discuss the progress and challenges of bringing adipose stem cell therapy into mainstream clinical use. SIGNIFICANCE This article details the advances made in recent years that have helped move human adipose stem cell therapy toward mainstream clinical use from a translational research, regulatory policy, and industrial standpoint. Four recurrent themes in translational technology as they pertain to human adipose stem cells are discussed: automated closed-system operations, biosensors and real-time monitoring, biomimetics, and rapid manufacturing. In light of recent FDA guidance documents, regulatory concerns about adipose stem cell therapy are discussed. Finally, an update is provided on the current state of clinical trials and the emerging industry that uses human adipose stem cells. This article is expected to stimulate future studies in translational adipose stem cell research.
Collapse
Affiliation(s)
- Rachel C Nordberg
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, and University of North Carolina Chapel Hill, Chapel Hill, North Carolina, USA; Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Elizabeth G Loboa
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, and University of North Carolina Chapel Hill, Chapel Hill, North Carolina, USA; Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
24
|
Damaraju S, Matyas JR, Rancourt DE, Duncan NA. The role of gap junctions and mechanical loading on mineral formation in a collagen-I scaffold seeded with osteoprogenitor cells. Tissue Eng Part A 2015; 21:1720-32. [PMID: 25752490 DOI: 10.1089/ten.tea.2014.0522] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Fracture nonunions represent one of many large bone defects where current treatment strategies fall short in restoring both form and function of the injured tissue. In this case, the use of a tissue-engineered scaffold for promoting bone healing offers an accessible and easy-to-manipulate environment for studying bone formation processes in vitro. We have previously shown that mechanical prestimulation using confined compression of differentiating osteoblasts results in an increase in mineralization formed in a 3D collagen-I scaffold. This study builds on this knowledge by evaluating the short and long-term effects of blocking gap junction-mediated intercellular communication among osteogenic cells on their effectiveness to mineralize collagen-I scaffolds in vitro, and in the presence and absence of mechanical stimulation. In this study, confined compression was applied in conjunction with octanol (a general communication blocker) or 18-α-glycerrhetinic acid (AGA, a specific gap junction blocker) using a modified FlexCell plate to collagen-I scaffolds seeded with murine embryonic stem cells stimulated toward osteoblast differentiation using beta-glycerol phosphate. The activity, presence, and expression of osteoblast cadherin, connexin-43, as well as various pluripotent and osteogenic markers were examined at 5-30 days of differentiation. Fluorescence recovery after photobleaching, immunofluorescence, viability, histology assessments, and reverse-transcriptase polymerase chain reaction assessments revealed that inhibiting communication in this scaffold altered the lineage and function of differentiating osteoblasts. In particular, treatment with communication inhibitors caused reduced mineralization in the matrix, and dissociation between connexin-43 and integrin α5β1. This dissociation was not restored even after long-term recovery. Thus, in order for this scaffold to be considered as an alternative strategy for the repair of large bone defects, cell-cell contacts and cell-matrix interactions must remain intact for osteoblast differentiation and function to be preserved. This study shows that within this 3D scaffold, gap junctions are essential in osteoblast response to mechanical loading, and are essential structures in producing a significant amount and organization of mineralization in the matrix.
Collapse
Affiliation(s)
- Swathi Damaraju
- 1 McCaig Institute for Bone and Joint Health, University of Calgary , Calgary, Canada
| | | | | | | |
Collapse
|
25
|
Lynch ME, Fischbach C. Biomechanical forces in the skeleton and their relevance to bone metastasis: biology and engineering considerations. Adv Drug Deliv Rev 2014; 79-80:119-34. [PMID: 25174311 DOI: 10.1016/j.addr.2014.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 07/30/2014] [Accepted: 08/20/2014] [Indexed: 12/17/2022]
Abstract
Bone metastasis represents the leading cause of breast cancer related-deaths. However, the effect of skeleton-associated biomechanical signals on the initiation, progression, and therapy response of breast cancer bone metastasis is largely unknown. This review seeks to highlight possible functional connections between skeletal mechanical signals and breast cancer bone metastasis and their contribution to clinical outcome. It provides an introduction to the physical and biological signals underlying bone functional adaptation and discusses the modulatory roles of mechanical loading and breast cancer metastasis in this process. Following a definition of biophysical design criteria, in vitro and in vivo approaches from the fields of bone biomechanics and tissue engineering that may be suitable to investigate breast cancer bone metastasis as a function of varied mechano-signaling will be reviewed. Finally, an outlook of future opportunities and challenges associated with this newly emerging field will be provided.
Collapse
Affiliation(s)
- Maureen E Lynch
- Department of Biomedical Engineering, Cornell University, Ithaca, USA
| | - Claudia Fischbach
- Department of Biomedical Engineering, Cornell University, Ithaca, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, USA.
| |
Collapse
|
26
|
Huisman E, Lu A, McCormack RG, Scott A. Enhanced collagen type I synthesis by human tenocytes subjected to periodic in vitro mechanical stimulation. BMC Musculoskelet Disord 2014; 15:386. [PMID: 25414072 PMCID: PMC4256895 DOI: 10.1186/1471-2474-15-386] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 11/06/2014] [Indexed: 11/10/2022] Open
Abstract
Background Mechanical stimulation (e.g. slow heavy loading) has proven beneficial in the rehabilitation of chronic tendinopathy, however the optimal parameters of stimulation have not been experimentally determined. In this study of mechanically stimulated human tenocytes, the influence of rest insertion and cycle number on (1) the protein and mRNA levels of type I and III collagen; (2) the mRNA levels of transforming growth factor beta (TGFB1) and scleraxis (SCXA); and (3) tenocyte morphology, were assessed. Methods Human hamstring tenocytes were mechanically stimulated using a Flexcell® system. The stimulation regimens were 1) continuous and 2) rest-inserted cyclic equiaxial strain at a frequency of 0.1 Hz for 100 or 1000 cycles. Data were normalized to unstimulated (non-stretched) control groups for every experimental condition. qPCR was performed to determine relative mRNA levels and quantitative immunocytochemistry image analysis was used to assess protein levels and cell morphology. Results Collagen type I mRNA level and pro-collagen protein levels were higher in tenocytes that were subjected to rest-inserted mechanical stimulation, compared to continuous stretching (p < 0.05). Rest insertion and increased cycle number also had significant positive effects on the levels of mRNA for TGFB1 and SCXA (p < 0.05). There was no direct relation between cell morphology and gene expression, however mechanical stimulation, overall, induced a metabolically active tenocyte phenotype as evidenced by cells that on average demonstrated a decreased major-minor axis ratio (p < 0.05) with greater branching (p < 0.01). Conclusions The incorporation of rest periods in a mechanical stretching regimen results in greater collagen type I synthesis. This knowledge may be beneficial in refining rehabilitation protocols for tendon injury. Electronic supplementary material The online version of this article (doi:10.1186/1471-2474-15-386) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | - Alex Scott
- Department of Physical Therapy, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
27
|
Steward AJ, Kelly DJ. Mechanical regulation of mesenchymal stem cell differentiation. J Anat 2014; 227:717-31. [PMID: 25382217 DOI: 10.1111/joa.12243] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2014] [Indexed: 12/18/2022] Open
Abstract
Biophysical cues play a key role in directing the lineage commitment of mesenchymal stem cells or multipotent stromal cells (MSCs), but the mechanotransductive mechanisms at play are still not fully understood. This review article first describes the roles of both substrate mechanics (e.g. stiffness and topography) and extrinsic mechanical cues (e.g. fluid flow, compression, hydrostatic pressure, tension) on the differentiation of MSCs. A specific focus is placed on the role of such factors in regulating the osteogenic, chondrogenic, myogenic and adipogenic differentiation of MSCs. Next, the article focuses on the cellular components, specifically integrins, ion channels, focal adhesions and the cytoskeleton, hypothesized to be involved in MSC mechanotransduction. This review aims to illustrate the strides that have been made in elucidating how MSCs sense and respond to their mechanical environment, and also to identify areas where further research is needed.
Collapse
Affiliation(s)
- Andrew J Steward
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, USA
| | - Daniel J Kelly
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
28
|
Mathieu PS, Bodle JC, Loboa EG. Primary cilium mechanotransduction of tensile strain in 3D culture: Finite element analyses of strain amplification caused by tensile strain applied to a primary cilium embedded in a collagen matrix. J Biomech 2014; 47:2211-7. [PMID: 24831236 DOI: 10.1016/j.jbiomech.2014.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 03/28/2014] [Accepted: 04/05/2014] [Indexed: 01/22/2023]
Abstract
Human adipose-derived stem cells (hASC) exhibit multilineage differentiation potential with lineage specification that is dictated by both the chemical and mechanical stimuli to which they are exposed. We have previously shown that 10% cyclic tensile strain increases hASC osteogenesis and cell-mediated calcium accretion. We have also recently shown that primary cilia are present on hASC and that chemically-induced lineage specification of hASC concurrently results in length and conformation changes of the primary cilia. Further, we have observed cilia length changes in hASC cultured within a collagen I gel in response to 10% cyclic tensile strain. We therefore hypothesize that primary cilia may play a key mechanotransduction role for hASC exposed to tensile strain. The goal of this study was to use finite element analysis (FEA) to determine strains occurring within the ciliary membrane in response to 10% tensile strain applied parallel, or perpendicular, to cilia orientation. To elucidate the mechanical environment experienced by the cilium, several lengths were modeled and evaluated based on cilia lengths measured on hASC grown under varied culture conditions. Principal tensile strains in both hASC and ciliary membranes were calculated using FEA, and the magnitude and location of maximum principal tensile strain determined. We found that maximum principal tensile strain was concentrated at the base of the cilium. In the linear elastic model, applying strain perpendicular to the cilium resulted in maximum strains within the ciliary membrane from 150% to 200%, while applying strain parallel to the cilium resulted in much higher strains, approximately 400%. In the hyperelastic model, applying strain perpendicular to the cilium resulted in maximum strains within the ciliary membrane around 30%, while applying strain parallel to the cilium resulted in much higher strains ranging from 50% to 70%. Interestingly, FEA results indicated that primary cilium length was not directly related to ciliary membrane strain. Rather, it appears that cilium orientation may be more important than cilium length in determining sensitivity of hASC to tensile strain. This is the first study to model the effects of tensile strain on the primary cilium and provides newfound insight into the potential role of the primary cilium as a mechanosensor, particularly in tensile strain and potentially a multitude of other mechanical stimuli beyond fluid shear.
Collapse
Affiliation(s)
- Pattie S Mathieu
- Joint Department of Biomedical Engineering at North Carolina State University and University of North Carolina-Chapel Hill, Engineering Building III (EB3) 4208B, Box 7115, NCSU Campus, Raleigh, NC 27695, USA
| | - Josephine C Bodle
- Joint Department of Biomedical Engineering at North Carolina State University and University of North Carolina-Chapel Hill, Engineering Building III (EB3) 4208B, Box 7115, NCSU Campus, Raleigh, NC 27695, USA
| | - Elizabeth G Loboa
- Joint Department of Biomedical Engineering at North Carolina State University and University of North Carolina-Chapel Hill, Engineering Building III (EB3) 4208B, Box 7115, NCSU Campus, Raleigh, NC 27695, USA; Department of Materials Science & Engineering, North Carolina State University, USA.
| |
Collapse
|
29
|
Yang P, Huang X, Wang C, Dang X, Wang K. Repair of bone defects using a new biomimetic construction fabricated by adipose-derived stem cells, collagen I, and porous beta-tricalcium phosphate scaffolds. Exp Biol Med (Maywood) 2013; 238:1331-43. [PMID: 24157587 DOI: 10.1177/1535370213505827] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Adipose derived stem cells (ASCs) with multilineage differentiation capacities have been demonstrated as an alternative cell candidate for in vitro and in vivo bone regeneration. This suggests that they may be a potential candidate to repair the bone defects. We attempted to demonstrate the use of new biomimetic constructions of undifferentiated rabbit adipose-derived stem cells (rASCs) with fully interconnected porous beta-tricalcium phosphate (β-TCP) scaffolds encapsulated by collagen I hydrogel in the regeneration of a critical-sized defect of rabbit radii. Critical-sized defects in the left radii of rabbits were prepared and inserted with rASCs/collagen I/β-TCP scaffold composites or collagen I/β-TCP scaffold composites. The results were evaluated by histology, radiographs, micro-CT, Emission Computed Tomography (ECT), fluorochrome labeling, western blot, and mechanical testing at 4, 8, and 12 weeks postsurgery. Twelve weeks after implantation, the defects were almost completely repaired as confirmed by the presence of the cortical bone and medullary cavity, which was evaluated through radiologic, histologic, and biomechanical examination. Biodegradation of the biomaterials may be attributed to extracellular liquid dissolution together with cell-mediated phagocytosis. Our study shows that a greater number of rASCs in the porous β-TCP scaffold encapsulated by collagen I gel enhanced osteogenesis in critical-sized defects. We hope to garner new insight into the engineering of rASCs-based bone tissue for clinical application.
Collapse
Affiliation(s)
- Pei Yang
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710004, China
| | | | | | | | | |
Collapse
|
30
|
Charoenpanich A, Wall ME, Tucker CJ, Andrews DMK, Lalush DS, Dirschl DR, Loboa EG. Cyclic tensile strain enhances osteogenesis and angiogenesis in mesenchymal stem cells from osteoporotic donors. Tissue Eng Part A 2013; 20:67-78. [PMID: 23927731 DOI: 10.1089/ten.tea.2013.0006] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We have shown that the uniaxial cyclic tensile strain of magnitude 10% promotes and enhances osteogenesis of human mesenchymal stem cells (hMSC) and human adipose-derived stem cells (hASC) from normal, nonosteoporotic donors. In the present study, MSC from osteoporotic donors were analyzed for changes in mRNA expression in response to 10% uniaxial tensile strain to identify potential mechanisms underlying the use of this mechanical loading paradigm for prevention and treatment of osteoporosis. Human MSC isolated from three female, postmenopausal osteoporotic donors were analyzed for their responses to mechanical loading using microarray analysis of over 47,000 gene probes. Human MSC were seeded in three-dimensional collagen type I constructs to mimic the organic extracellular matrix of bone and 10% uniaxial cyclic tensile strain was applied to promote osteogenesis. Seventy-nine genes were shown to be regulated within hMSC from osteoporotic donors in response to 10% cyclic tensile strain. Upregulation of six genes were further confirmed with real-time RT-PCR: jun D proto-oncogene (JUND) and plasminogen activator, urokinase receptor (PLAUR), two genes identified as potential key molecules from network analysis; phosphoinositide-3-kinase, catalytic, delta polypeptide (PIK3CD) and wingless-type MMTV integration site family, member 5B (WNT5B), two genes with known importance in bone biology; and, PDZ and LIM domain 4 (PDLIM4) and vascular endothelial growth factor A (VEGFA), two genes that we have previously shown are significantly regulated in hASC in response to this mechanical stimulus. Function analysis indicated that 10% cyclic tensile strain induced expression of genes associated with cell movement, cell proliferation, and tissue development, including development in musculoskeletal and cardiovascular systems. Our results demonstrate that hMSC from aged, osteoporotic donors are capable of enhanced osteogenic differentiation in response to 10% cyclic tensile strain with significant increases in the expression of genes associated with enhanced cell proliferation, musculoskeletal development, and angiogenesis. Surprisingly, cyclic tensile strain of magnitude 10% not only enhanced osteogenesis in hMSC from osteoporotic donors, but also enhanced expression of angiogenic factors. Better understanding and methodologies to promote osteogenesis in hMSC from elderly, osteoporotic donors may greatly facilitate achieving long-term success in bone regeneration and functional bone tissue engineering for this ever-growing patient population.
Collapse
Affiliation(s)
- Adisri Charoenpanich
- 1 Joint Department of Biomedical Engineering at University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, and North Carolina State University , Raleigh, North Carolina
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
The aortic valve is highly responsive to cyclical and continuous mechanical forces, at the macroscopic and cellular levels. In this report, we delineate mechanokinetics (effects of mechanical inputs on the cells) and mechanodynamics (effects of cells and pathologic processes on the mechanics) of the aortic valve, with a particular focus on how mechanical inputs synergize with the inflammatory cytokine and other biomolecular signaling to contribute to the process of aortic valve calcification.
Collapse
|
32
|
Worton LE, Ausk BJ, Downey LM, Bain SD, Gardiner EM, Srinivasan S, Gross TS, Kwon RY. Systems-based identification of temporal processing pathways during bone cell mechanotransduction. PLoS One 2013; 8:e74205. [PMID: 24040202 PMCID: PMC3770665 DOI: 10.1371/journal.pone.0074205] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/26/2013] [Indexed: 11/25/2022] Open
Abstract
Bone has long been established to be a highly mechanosensitive tissue. When subjected to mechanical loading, bone exhibits profoundly different anabolic responses depending on the temporal pattern in which the stimulus is applied. This phenomenon has been termed temporal processing, and involves complex signal amplification mechanisms that are largely unidentified. In this study, our goal was to characterize transcriptomic perturbations arising from the insertion of intermittent rest periods (a temporal variation with profound effects on bone anabolism) in osteoblastic cells subjected to fluid flow, and assess the utility of these perturbations to identify signaling pathways that are differentially activated by this temporal variation. At the level of the genome, we found that the common and differential alterations in gene expression arising from the two flow conditions were distributionally distinct, with the differential alterations characterized by many small changes in a large number of genes. Using bioinformatics analysis, we identified distinct up- and down-regulation transcriptomic signatures associated with the insertion of rest intervals, and found that the up-regulation signature was significantly associated with MAPK signaling. Confirming the involvement of the MAPK pathway, we found that the insertion of rest intervals significantly elevated flow-induced p-ERK1/2 levels by enabling a second spike in activity that was not observed in response to continuous flow. Collectively, these studies are the first to characterize distinct transcriptomic perturbations in bone cells subjected to continuous and intermittent stimulation, and directly demonstrate the utility of systems-based transcriptomic analysis to identify novel acute signaling pathways underlying temporal processing in bone cells.
Collapse
Affiliation(s)
- Leah E. Worton
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Brandon J. Ausk
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Leah M. Downey
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Steven D. Bain
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Edith M. Gardiner
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Sundar Srinivasan
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Ted S. Gross
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Ronald Y. Kwon
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
33
|
Grottkau BE, Lin Y. Osteogenesis of Adipose-Derived Stem Cells. Bone Res 2013; 1:133-45. [PMID: 26273498 DOI: 10.4248/br201302003] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 04/12/2013] [Indexed: 01/01/2023] Open
Abstract
Current treatment options for skeletal repair, including immobilization, rigid fixation, alloplastic materials and bone grafts, have significant limitations. Bone tissue engineering offers a promising method for the repair of bone deficieny caused by fractures, bone loss and tumors. The use of adipose derived stem cells (ASCs) has received attention because of the self-renewal ability, high proliferative capacity and potential of osteogenic differentiation in vitro and in vivo studies of bone regeneration. Although cell therapies using ASCs are widely promising in various clinical fields, no large human clinical trials exist for bone tissue engineering. The aim of this review is to introduce how they are harvested, examine the characterization of ASCs, to review the mechanisms of osteogenic differentiation, to analyze the effect of mechanical and chemical stimuli on ASC osteodifferentiation, to summarize the current knowledge about usage of ASC in vivo studies and clinical trials, and finally to conclude with a general summary of the field and comments on its future direction.
Collapse
Affiliation(s)
- Brian E Grottkau
- Department of Orthopaedic Surgery, MassGeneral Hospital for Children and the Pediatric Orthopaedic Laboratory for Tissue Engineering and Regenerative Medicine, Harvard Medical School , Boston, Massachusetts, USA
| | - Yunfeng Lin
- Department of Orthopaedic Surgery, MassGeneral Hospital for Children and the Pediatric Orthopaedic Laboratory for Tissue Engineering and Regenerative Medicine, Harvard Medical School , Boston, Massachusetts, USA
| |
Collapse
|
34
|
Bodle JC, Rubenstein CD, Phillips ME, Bernacki SH, Qi J, Banes AJ, Loboa EG. Primary cilia: the chemical antenna regulating human adipose-derived stem cell osteogenesis. PLoS One 2013; 8:e62554. [PMID: 23690943 PMCID: PMC3656889 DOI: 10.1371/journal.pone.0062554] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 03/23/2013] [Indexed: 02/07/2023] Open
Abstract
Adipose-derived stem cells (ASC) are multipotent stem cells that show great potential as a cell source for osteogenic tissue replacements and it is critical to understand the underlying mechanisms of lineage specification. Here we explore the role of primary cilia in human ASC (hASC) differentiation. This study focuses on the chemosensitivity of the primary cilium and the action of its associated proteins: polycystin-1 (PC1), polycystin-2 (PC2) and intraflagellar transport protein-88 (IFT88), in hASC osteogenesis. To elucidate cilia-mediated mechanisms of hASC differentiation, siRNA knockdown of PC1, PC2 and IFT88 was performed to disrupt cilia-associated protein function. Immunostaining of the primary cilium structure indicated phenotypic-dependent changes in cilia morphology. hASC cultured in osteogenic differentiation media yielded cilia of a more elongated conformation than those cultured in expansion media, indicating cilia-sensitivity to the chemical environment and a relationship between the cilium structure and phenotypic determination. Abrogation of PC1, PC2 and IFT88 effected changes in both hASC proliferation and differentiation activity, as measured through proliferative activity, expression of osteogenic gene markers, calcium accretion and endogenous alkaline phosphatase activity. Results indicated that IFT88 may be an early mediator of the hASC differentiation process with its knockdown increasing hASC proliferation and decreasing Runx2, alkaline phosphatase and BMP-2 mRNA expression. PC1 and PC2 knockdown affected later osteogenic gene and end-product expression. PC1 knockdown resulted in downregulation of alkaline phosphatase and osteocalcin gene expression, diminished calcium accretion and reduced alkaline phosphatase enzymatic activity. Taken together our results indicate that the structure of the primary cilium is intimately associated with the process of hASC osteogenic differentiation and that its associated proteins are critical players in this process. Elucidating the dynamic role of the primary cilium and its associated proteins will help advance the application of hASC in generating autologous tissue engineered therapies in critical defect bone injuries.
Collapse
Affiliation(s)
- Josephine C. Bodle
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
| | - Candace D. Rubenstein
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
| | - Michelle E. Phillips
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
| | - Susan H. Bernacki
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
| | - Jie Qi
- Flexcell International Corporation, Hillsborough, North Carolina, United States of America
| | - Albert J. Banes
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
- Flexcell International Corporation, Hillsborough, North Carolina, United States of America
| | - Elizabeth G. Loboa
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Materials Science and Engineering, North Carolina State University, Raleigh North Carolina, United States of America
| |
Collapse
|
35
|
|
36
|
Subramony SD, Dargis BR, Castillo M, Azeloglu EU, Tracey MS, Su A, Lu HH. The guidance of stem cell differentiation by substrate alignment and mechanical stimulation. Biomaterials 2012; 34:1942-53. [PMID: 23245926 DOI: 10.1016/j.biomaterials.2012.11.012] [Citation(s) in RCA: 191] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 11/10/2012] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSC) represent a promising and clinically relevant cell source for tissue engineering applications. As such, guiding MSCs toward specific lineages and maintaining these phenotypes have been particularly challenging as the contributions of mechanical, chemical and structural cues to the complex differentiation process are largely unknown. To fully harness the potential of MSCs for regenerative medicine, a systematic investigation into the individual and combined effects of these stimuli is needed. In addition, unlike chemical stimulation, for which temporal and concentration gradients are difficult to control, mechanical stimulation and scaffold-based cues may be relatively more biomimetic and can be applied with greater control to ensure fidelity in MSC differentiation. The objective of this study is to investigate the role of nanofiber matrix alignment and mechanical stimulation on MSC differentiation, focusing on elucidating the relative contribution of each parameter in guided regeneration of functional connective tissues. It is observed that nanofiber alignment directs MSC response to physiological loading and that fibroblastic differentiation requires a combination of physiologically-relevant cell-material interactions in conjunction with mechanical stimulation. Importantly, the results of this study reveal that systemic and readily controllable cues, such as scaffold alignment and optimized mechanical stimulation, are sufficient to drive MSC differentiation, without the need for additional chemical stimuli. Moreover, these findings yield a set of fundamental design rules that can be readily applied to connective tissue regeneration strategies.
Collapse
Affiliation(s)
- Siddarth D Subramony
- Biomaterials and Interface Tissue Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Huang G, Wang L, Wang S, Han Y, Wu J, Zhang Q, Xu F, Lu TJ. Engineering three-dimensional cell mechanical microenvironment with hydrogels. Biofabrication 2012; 4:042001. [PMID: 23164720 DOI: 10.1088/1758-5082/4/4/042001] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cell mechanical microenvironment (CMM) significantly affects cell behaviors such as spreading, migration, proliferation and differentiation. However, most studies on cell response to mechanical stimulation are based on two-dimensional (2D) planar substrates, which cannot mimic native three-dimensional (3D) CMM. Accumulating evidence has shown that there is a significant difference in cell behavior in 2D and 3D microenvironments. Among the materials used for engineering 3D CMM, hydrogels have gained increasing attention due to their tunable properties (e.g. chemical and mechanical properties). In this paper, we provide an overview of recent advances in engineering hydrogel-based 3D CMM. Effects of mechanical cues (e.g. hydrogel stiffness and externally induced stress/strain in hydrogels) on cell behaviors are described. A variety of approaches to load mechanical stimuli in 3D hydrogel-based constructs are also discussed.
Collapse
Affiliation(s)
- Guoyou Huang
- Biomedical Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Park SH, Sim WY, Min BH, Yang SS, Khademhosseini A, Kaplan DL. Chip-based comparison of the osteogenesis of human bone marrow- and adipose tissue-derived mesenchymal stem cells under mechanical stimulation. PLoS One 2012; 7:e46689. [PMID: 23029565 PMCID: PMC3460891 DOI: 10.1371/journal.pone.0046689] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Accepted: 09/06/2012] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue-derived stem cells (ASCs) are considered as an attractive stem cell source for tissue engineering and regenerative medicine. We compared human bone marrow-derived mesenchymal stem cells (hMSCs) and hASCs under dynamic hydraulic compression to evaluate and compare osteogenic abilities. A novel micro cell chip integrated with microvalves and microscale cell culture chambers separated from an air-pressure chamber was developed using microfabrication technology. The microscale chip enables the culture of two types of stem cells concurrently, where each is loaded into cell culture chambers and dynamic compressive stimulation is applied to the cells uniformly. Dynamic hydraulic compression (1 Hz, 1 psi) increased the production of osteogenic matrix components (bone sialoprotein, oateopontin, type I collagen) and integrin (CD11b and CD31) expression from both stem cell sources. Alkaline phosphatase and Alrizarin red staining were evident in the stimulated hMSCs, while the stimulated hASCs did not show significant increases in staining under the same stimulation conditions. Upon application of mechanical stimulus to the two types of stem cells, integrin (β1) and osteogenic gene markers were upregulated from both cell types. In conclusion, stimulated hMSCs and hASCs showed increased osteogenic gene expression compared to non-stimulated groups. The hMSCs were more sensitive to mechanical stimulation and more effective towards osteogenic differentiation than the hASCs under these modes of mechanical stimulation.
Collapse
Affiliation(s)
- Sang-Hyug Park
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
- Department of Biomedical Engineering, Jungwon University, Goesan-eup, Chungbuk, Korea
| | - Woo Young Sim
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Byoung-Hyun Min
- Department of Orthopeadic Surgery, Medical School, Ajou University, Youngtong-Gu, Suwon, Korea
- Department of Molecular Science and Technology, Ajou University, Youngtong-Gu, Suwon, Korea
| | - Sang Sik Yang
- Department of Electrical and Computer Engineering, Ajou University, Youngtong-Gu, Suwon, Korea
| | - Ali Khademhosseini
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, United States of America
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
39
|
Mathieu PS, Loboa EG. Cytoskeletal and focal adhesion influences on mesenchymal stem cell shape, mechanical properties, and differentiation down osteogenic, adipogenic, and chondrogenic pathways. TISSUE ENGINEERING PART B-REVIEWS 2012; 18:436-44. [PMID: 22741572 DOI: 10.1089/ten.teb.2012.0014] [Citation(s) in RCA: 282] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Mesenchymal stem cells (MSCs) hold great potential for regenerative medicine and tissue-engineering applications. They have multipotent differentiation capabilities and have been shown to differentiate down various lineages, including osteoblasts, adipocytes, chondrocytes, myocytes, and possibly neurons. The majority of approaches to control the MSC fate have been via the use of chemical factors in the form of growth factors within the culture medium. More recently, it has been understood that mechanical forces play a significant role in regulating MSC fate. We and others have shown that mechanical stimuli can control MSC lineage specification. The cytoskeleton is known to play a large role in mechanotransduction, and a growing number of studies are showing that it can also contribute to MSC differentiation. This review analyzes the significant contribution of actin and integrin distribution, and the smaller role of microtubules, in regulating MSC fate. Osteogenic differentiation is more prevalent in MSCs with a stiff, spread actin cytoskeleton and greater numbers of focal adhesions. Both adipogenic differentiation and chondrogenic differentiation are encouraged when MSCs have a spherical morphology associated with a dispersed actin cytoskeleton with few focal adhesions. Different mechanical stimuli can be implemented to alter these cytoskeletal patterns and encourage MSC differentiation to the desired lineage.
Collapse
Affiliation(s)
- Pattie S Mathieu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | | |
Collapse
|
40
|
Review of biophysical factors affecting osteogenic differentiation of human adult adipose-derived stem cells. Biophys Rev 2012; 5:11-28. [PMID: 28510177 DOI: 10.1007/s12551-012-0079-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Accepted: 03/15/2012] [Indexed: 12/11/2022] Open
Abstract
Developing bone is subject to the control of a broad variety of influences in vivo. For bone repair applications, in vitro osteogenic assays are routinely used to test the responses of bone-forming cells to drugs, hormones, and biomaterials. Results of these assays are used to predict the behavior of bone-forming cells in vivo. Stem cell research has shown promise for enhancing bone repair. In vitro osteogenic assays to test the bone-forming response of stem cells typically use chemical solutions. Stem cell in vitro osteogenic assays often neglect important biophysical cues, such as the forces associated with regular weight-bearing exercise, which promote bone formation. Incorporating more biophysical cues that promote bone formation would improve in vitro osteogenic assays for stem cells. Improved in vitro osteogenic stimulation opens opportunities for "pre-conditioning" cells to differentiate towards the desired lineage. In this review, we explore the role of select biophysical factors-growth surfaces, tensile strain, fluid flow and electromagnetic stimulation-in promoting osteogenic differentiation of stem cells from human adipose. Emphasis is placed on the potential for physical microenvironment manipulation to translate tissue engineering and stem cell research into widespread clinical usage.
Collapse
|
41
|
Du HM, Zheng XH, Wang LY, Tang W, Liu L, Jing W, Lin YF, Tian WD, Long J. The osteogenic response of undifferentiated human adipose-derived stem cells under mechanical stimulation. Cells Tissues Organs 2012; 196:313-324. [PMID: 22584128 DOI: 10.1159/000335905] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2011] [Indexed: 01/06/2023] Open
Abstract
The purpose of this study was to investigate the osteogenic response of human adipose-derived stem cells (hASCs) under mechanical and/or chemical stimulation. hASCs were divided into three groups. In group A, the cells were cultured without any stimulation, in group B, the cells were induced with chemical stimulation, and in group C, the cells were induced with a combination of chemical stimulation and stretch loading. Stretch loading and chemical stimulation were applied using a four-point bending apparatus (0.5 Hz, 2,000 µε, 2 h/day) and osteogenic differentiation medium, respectively. At the 1st, 2nd, 3rd, 5th and 7th day following initiation of stretch loading, we detected alkaline phosphatase activity, mRNA expression (RUNX2, ALPL, osteonectin, osteopontin and type I collagen) and protein expression (RUNX2 and osteopontin) by colorimetric assay, real-time PCR and Western blot methods, respectively. Alkaline phosphatase activity, mRNA expression and protein expression all increased in groups B and C along with the culture time, but were observed to be downregulated by the 7th day in group C (p < 0.05). Compared to group A, most of the above markers were significantly higher in groups B and C (p < 0.05). All of the above markers in group C were higher than those in group B before the 5th day (p < 0.05), except at the 1st day. These results indicated that stretch loading promoted osteogenic differentiation of hASCs and that the combination of mechanical and chemical stimulation could enhance the osteogenic capability up to the 5th day relative to chemical stimulation alone.
Collapse
Affiliation(s)
- Hong-ming Du
- The State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Riehl BD, Park JH, Kwon IK, Lim JY. Mechanical stretching for tissue engineering: two-dimensional and three-dimensional constructs. TISSUE ENGINEERING PART B-REVIEWS 2012; 18:288-300. [PMID: 22335794 DOI: 10.1089/ten.teb.2011.0465] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mechanical cell stretching may be an attractive strategy for the tissue engineering of mechanically functional tissues. It has been demonstrated that cell growth and differentiation can be guided by cell stretch with minimal help from soluble factors and engineered tissues that are mechanically stretched in bioreactors may have superior organization, functionality, and strength compared with unstretched counterparts. This review explores recent studies on cell stretching in both two-dimensional (2D) and three-dimensional (3D) setups focusing on the applications of stretch stimulation as a tool for controlling cell orientation, growth, gene expression, lineage commitment, and differentiation and for achieving successful tissue engineering of mechanically functional tissues, including cardiac, muscle, vasculature, ligament, tendon, bone, and so on. Custom stretching devices and lab-specific mechanical bioreactors are described with a discussion on capabilities and limitations. While stretch mechanotransduction pathways have been examined using 2D stretch, studying such pathways in physiologically relevant 3D environments may be required to understand how cells direct tissue development under stretch. Cell stretch study using 3D milieus may also help to develop tissue-specific stretch regimens optimized with biochemical feedback, which once developed will provide optimal tissue engineering protocols.
Collapse
Affiliation(s)
- Brandon D Riehl
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, USA
| | | | | | | |
Collapse
|
43
|
Charoenpanich A, Wall ME, Tucker CJ, Andrews DMK, Lalush DS, Loboa EG. Microarray analysis of human adipose-derived stem cells in three-dimensional collagen culture: osteogenesis inhibits bone morphogenic protein and Wnt signaling pathways, and cyclic tensile strain causes upregulation of proinflammatory cytokine regulators and angiogenic factors. Tissue Eng Part A 2011; 17:2615-27. [PMID: 21767168 DOI: 10.1089/ten.tea.2011.0107] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human adipose-derived stem cells (hASC) have shown great potential for bone tissue engineering. However, the molecular mechanisms underlying this potential are not yet known, in particular the separate and combined effects of three-dimensional (3D) culture and mechanical loading on hASC osteogenesis. Mechanical stimuli play a pivotal role in bone formation, remodeling, and fracture repair. To further understand hASC osteogenic differentiation and response to mechanical stimuli, gene expression profiles of proliferating or osteogenically induced hASC in 3D collagen I culture in the presence and absence of 10% uniaxial cyclic tensile strain were examined using microarray analysis. About 847 genes and 95 canonical pathways were affected during osteogenesis of hASC in 3D culture. Pathway analysis indicated the potential roles of Wnt/β-catenin signaling, bone morphogenic protein (BMP) signaling, platelet-derived growth factor (PDGF) signaling, and insulin-like growth factor 1 (IGF-1) signaling in hASC during osteogenic differentiation. Application of 10% uniaxial cyclic tensile strain suggested synergistic effects of strain with osteogenic differentiation media on hASC osteogenesis as indicated by significantly increased calcium accretion of hASC. There was no significant further alteration in the four major pathways (Wnt/β-catenin, BMP, PDGF, and IGF-1). However, 184 transcripts were affected by 10% cyclic tensile strain. Function and network analysis of these transcripts suggested that 10% cyclic tensile strain may play a role during hASC osteogenic differentiation by upregulating two crucial factors in bone regeneration: (1) proinflammatory cytokine regulators interleukin 1 receptor antagonist and suppressor of cytokine signaling 3; (2) known angiogenic inductors fibroblast growth factor 2, matrix metalloproteinase 2, and vascular endothelial growth factor A. This is the first study to investigate the effects of both 3D culture and mechanical load on hASC osteogenic differentiation. A complete microarray analysis investigating both the separate effect of soluble osteogenic inductive factors and the combined effects of chemical and mechanical stimulation was performed on hASC undergoing osteogenic differentiation. We have identified specific genes and pathways associated with mechanical response and osteogenic potential of hASC, thus providing significant information toward improved understanding of our use of hASC for functional bone tissue engineering applications.
Collapse
Affiliation(s)
- Adisri Charoenpanich
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695-7115, USA
| | | | | | | | | | | |
Collapse
|
44
|
Xiang X, Zhao J, Xu G, Li Y, Zhang W. mTOR and the differentiation of mesenchymal stem cells. Acta Biochim Biophys Sin (Shanghai) 2011; 43:501-10. [PMID: 21642276 DOI: 10.1093/abbs/gmr041] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The mammalian target of rapamycin (mTOR), an evolutionarily conserved serine-threonine protein kinase, belongs to the phosphoinositide 3-kinase (PI3K)-related kinase family, which contains a lipid kinase-like domain within their C-terminal region. Recent studies have revealed that mTOR as a critical intracellular molecule can sense the extracellular energy status and regulate the cell growth and proliferation in a variety of cells and tissues. This review summarizes our current understanding about the effects of mTOR on cell differentiation and tissue development, with an emphasis on the lineage determination of mesenchymal stem cells. mTOR can promote adipogenesis in white adipocytes, brown adipocytes, and muscle satellite cells, while rapamycin inhibits the adipogenic function of mTOR. mTOR signaling may function to affect osteoblast proliferation and differentiation, however, rapamycin has been reported to either inhibit or promote osteogenesis. Although the precise mechanism remains unclear, mTOR is indispensable for myogenesis. Depending on the cell type, rapamycin has been reported to inhibit, promote, or have no effect on myogenesis.
Collapse
Affiliation(s)
- Xinxin Xiang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Department of Physiology and Pathophysiology, Health Science Center, Peking University, Beijing, China
| | | | | | | | | |
Collapse
|
45
|
Bodle JC, Hanson AD, Loboa EG. Adipose-derived stem cells in functional bone tissue engineering: lessons from bone mechanobiology. TISSUE ENGINEERING PART B-REVIEWS 2011; 17:195-211. [PMID: 21338267 DOI: 10.1089/ten.teb.2010.0738] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
This review aims to highlight the current and significant work in the use of adipose-derived stem cells (ASC) in functional bone tissue engineering framed through the bone mechanobiology perspective. Over a century of work on the principles of bone mechanosensitivity is now being applied to our understanding of bone development. We are just beginning to harness that potential using stem cells in bone tissue engineering. ASC are the primary focus of this review due to their abundance and relative ease of accessibility for autologous procedures. This article outlines the current knowledge base in bone mechanobiology to investigate how the knowledge from this area has been applied to the various stem cell-based approaches to engineering bone tissue constructs. Specific emphasis is placed on the use of human ASC for this application.
Collapse
Affiliation(s)
- Josephine C Bodle
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695-7115, USA
| | | | | |
Collapse
|
46
|
Ferdous Z, Jo H, Nerem RM. Differences in valvular and vascular cell responses to strain in osteogenic media. Biomaterials 2011; 32:2885-93. [PMID: 21284997 DOI: 10.1016/j.biomaterials.2011.01.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 01/12/2011] [Indexed: 10/18/2022]
Abstract
Calcification is the primary cause of failure of bioprosthetic and tissue-engineered vascular and valvular grafts. We used tissue-engineered collagen gels containing human aortic smooth muscle cells (HASMC) and human aortic valvular interstitial cells (HAVIC) as a model to investigate cell-mediated differences in early markers of calcification. The HASMCs and HAVICs were isolated from non-sclerotic human tissues. After 21 days of culture in either regular or osteogenic media with or without 10% cyclic strain at 1 Hz, the collagen gels were assessed for DNA content, collagen I, matrix metalloproteinase (MMP)-2 and glycosaminoglycan (GAG) content. The collagen gels containing HASMCs contained significantly greater amounts of collagen I and GAG compared to HAVICs. Although strain increased MMP-2 activity for both cell types, this trend was significant (p ≤ 0.05) only for HAVICs. Cultured gels were also assessed for osteogenic markers calcium content, alkaline phosphatase (ALP), and Runx2 and were present at greater amounts in gels containing HASMCs than HAVICs. Calcium content, Runx2 expression, and ALP activity were also modulated by mechanical strain. The results indicate that cell-mediated differences exist between the vascular and valvular calcification processes. Further investigation is necessary for improved understanding and to detect biomarkers for early detection or prevention of these diseases.
Collapse
Affiliation(s)
- Zannatul Ferdous
- Institute of Bioengineering and Biosciences, Georgia Institute of Technology, 315 Ferst Drive, Mail code 0363, Atlanta, GA 30332, USA.
| | | | | |
Collapse
|
47
|
Delaine-Smith RM, Reilly GC. The effects of mechanical loading on mesenchymal stem cell differentiation and matrix production. VITAMINS AND HORMONES 2011; 87:417-80. [PMID: 22127254 DOI: 10.1016/b978-0-12-386015-6.00039-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells or stromal cells (MSCs) have the potential to be used therapeutically in tissue engineering and regenerative medicine to replace or restore the function of damaged tissues. Therefore, considerable effort has been ongoing in the research community to optimize culture conditions for predifferentiation of MSCs. All mesenchymal tissues are subjected to mechanical forces in vivo and all fully differentiated mesenchymal lineage cells respond to mechanical stimulation in vivo and in vitro. Therefore, it is not surprising that MSCs are highly mechanosensitive. We present a summary of current methods of mechanical stimulation of MSCs and an overview of the outcomes of the different mechanical culture techniques tested. Tissue engineers and stem cell researchers should be able to harness this mechanosensitivity to modulate MSC differentiation and matrix production; however, more research needs to be undertaken to understand the complex interactions between the mechanosensitive and biochemically stimulated differentiation pathways.
Collapse
Affiliation(s)
- Robin M Delaine-Smith
- The Kroto Research Institute, Department of Materials Science and Engineering, University of Sheffield, Sheffield, United Kingdom
| | | |
Collapse
|
48
|
Marvel S, Okrasinski S, Bernacki SH, Loboa E, Dayton PA. The development and validation of a LIPUS system with preliminary observations of ultrasonic effects on human adult stem cells. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2010; 57:1977-1984. [PMID: 20875987 DOI: 10.1109/tuffc.2010.1645] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
To study the potential effects of low-intensity pulsed ultrasound (LIPUS) on cell response in vitro, the ability to alter LIPUS parameters is required. However, commercial LIPUS systems have very little control over parameter selection. In this study, a custom LIPUS system was designed and validated by exploring the effects of using different pulse repetition frequency (PRF) parameters on human adipose derived adult stem cells (hASCs) and bone marrow derived mesenchymal stem cells (hMSCs), two common stem cell sources for creating bone constructs in vitro. Changing the PRF was found to affect cellular response to LIPUS stimulation for both cell types. Proliferation of LIPUS-stimulated cells was found to decrease for hASCs by d 7 for all three groups compared with unstimulated control cells (P = 0.008, 0.011, 0.014 for 1 Hz, 100 Hz and 1 kHz PRF, respectively) and for hMSCs by d 14 (donor 1: P = 0.0005, 0.0002, 0.0003; donor 2: P = 0.0003, 0.0002, 0.0001; for PRFs of 1 Hz, 100 Hz, and 1 kHz, respectively). Additionally, LIPUS was shown to strongly accelerate osteogenic differentiation of hASCs based on amount of calcium accretion normalized by total DNA (P = 0.003, 0.001, 0.003, and 0.032 between control/100 Hz, control/1 kHz, 1 Hz/1 kHz, and 100 Hz/1 kHz pulse repetition frequencies, respectively). These findings promote the study of using LIPUS to induce osteogenic differentiation and further encourage the exploration of LIPUS parameter optimization. The custom LIPUS system was successfully designed to allow extreme parameter variation, specifically PRF, and encourages further studies.
Collapse
Affiliation(s)
- Skylar Marvel
- University of North Carolina at Chapel Hill, Joint Department of Biomedical Engineering, Chapel Hill, NC, USA
| | | | | | | | | |
Collapse
|
49
|
2010 Nicolas Andry Award: Multipotent adult stem cells from adipose tissue for musculoskeletal tissue engineering. Clin Orthop Relat Res 2010; 468:2530-40. [PMID: 20625952 PMCID: PMC2919887 DOI: 10.1007/s11999-010-1410-9] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Accepted: 05/18/2010] [Indexed: 01/31/2023]
Abstract
BACKGROUND Cell-based therapies such as tissue engineering provide promising therapeutic possibilities to enhance the repair or regeneration of damaged or diseased tissues but are dependent on the availability and controlled manipulation of appropriate cell sources. QUESTIONS/PURPOSES The goal of this study was to test the hypothesis that adult subcutaneous fat contains stem cells with multilineage potential and to determine the influence of specific soluble mediators and biomaterial scaffolds on their differentiation into musculoskeletal phenotypes. METHODS We reviewed recent studies showing the stem-like characteristics and multipotency of adipose-derived stem cells (ASCs), and their potential application in cell-based therapies in orthopaedics. RESULTS Under controlled conditions, ASCs show phenotypic characteristics of various cell types, including chondrocytes, osteoblasts, adipocytes, neuronal cells, or muscle cells. In particular, the chondrogenic differentiation of ASCs can be induced by low oxygen tension, growth factors such as bone morphogenetic protein-6 (BMP-6), or biomaterial scaffolds consisting of native tissue matrices derived from cartilage. Finally, focus is given to the development of a functional biomaterial scaffold that can provide ASC-based constructs with mechanical properties similar to native cartilage. CONCLUSIONS Adipose tissue contains an abundant source of multipotent progenitor cells. These cells show cell surface marker profiles and differentiation characteristics that are similar to but distinct from other adult stem cells, such as bone marrow mesenchymal stem cells (MSCs). CLINICAL RELEVANCE The availability of an easily accessible and reproducible cell source may greatly facilitate the development of new cell-based therapies for regenerative medicine applications in the musculoskeletal system.
Collapse
|
50
|
Abstract
Bone marrow-derived multipotent stem and stromal cells (MSCs) are likely candidates for cell-based therapies for various conditions including skeletal disease. Advancement of these therapies will rely on an ability to identify, isolate, manipulate, and deliver stem cells in a safe and effective manner. Although it is clear that physical signals affect tissue morphogenesis, stem cell differentiation, and healing processes, integration of mechanically induced signaling events remain obscure. Mechanisms underlying sensation and interpretation of mechanical signals by stem cells are the focus of intense study. External mechanical signals have the ability to activate osteogenic signaling pathways in MSCs including Wnt, Ror2, and Runx2. It is also clear that intracellular tensile forces resulting from cell-extracellular matrix interactions play a critical role in MSC regulation. Further work is required to determine the precise role that mechanical forces play in stem cell function.
Collapse
Affiliation(s)
- Alesha B Castillo
- Bone and Joint Rehabilitation Research and Development Center, VA Palo Alto Health Care System, 3801 Miranda Ave, Mail Stop: 153, Palo Alto, CA, 94304, USA.
| | | |
Collapse
|