1
|
Goto A, Komura S, Kato K, Maki R, Hirakawa A, Aoki H, Tomita H, Taguchi J, Ozawa M, Matsushima T, Kishida A, Kimura T, Asahara H, Imai Y, Yamada Y, Akiyama H. PI3K-Akt signalling regulates Scx-lineage tenocytes and Tppp3-lineage paratenon sheath cells in neonatal tendon regeneration. Nat Commun 2025; 16:3734. [PMID: 40254618 PMCID: PMC12010001 DOI: 10.1038/s41467-025-59010-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
Tendon injuries are frequently occurring disorders; it is clinically important to enhance tendon regeneration and prevent functional impairment post-injury. While tendon injuries in children heal quickly with minimal scarring, those in adults heal slowly and are accompanied by fibrotic scarring. Therefore, investigating the healing mechanisms after tendon injury, and identifying the factors that regulate the inherent regenerative capacity of tendons are promising approaches to promoting tendon regeneration. Here, we identify that the PI3K-Akt signalling pathway is preferentially upregulated in injured neonatal murine Achilles tendons. Inhibition of PI3K-Akt signalling in a neonatal murine Achilles tendon rupture model decreases cell proliferation and migration in both Scx-lineage intrinsic tenocytes and Tppp3-lineage extrinsic paratenon sheath cells. Moreover, the inhibition of PI3K-Akt signalling decreases stemness and promotes mature tenogenic differentiation in both Scx- and Tppp3-lineage cells. Collectively, these results suggest that PI3K-Akt signalling plays a pivotal role in neonatal tendon regeneration.
Collapse
Affiliation(s)
- Atsushi Goto
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Shingo Komura
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, Japan.
| | - Koki Kato
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Rie Maki
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akihiro Hirakawa
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hitomi Aoki
- Department of Stem Cell and Regenerative Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, Japan
| | - Jumpei Taguchi
- Core Laboratory for Developing Advanced Animal Models, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Manabu Ozawa
- Core Laboratory for Developing Advanced Animal Models, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | - Akio Kishida
- Department of Material-Based Medical Engineering, Laboratory for Biomaterials and Bioengineering, Institute of Integrated Research, Institute of Science Tokyo, Tokyo, Japan
| | - Tsuyoshi Kimura
- Materials-based Medical Engineering Laboratory, Department of Biomedical Engineering, Faculty of Life Science, Toyo University, Tokyo, Japan
| | - Hiroshi Asahara
- Department of Systems BioMedicine, Institute of Science Tokyo, Tokyo, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Yasuhiro Yamada
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruhiko Akiyama
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, Japan
| |
Collapse
|
2
|
Ye Y, Huang H, Li H, Wu G. Role of chemical groups in regulating membrane tension of mBMSCs under stretch stimulation. Colloids Surf B Biointerfaces 2025; 252:114644. [PMID: 40132336 DOI: 10.1016/j.colsurfb.2025.114644] [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: 01/28/2025] [Revised: 03/08/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
As a crucial mechanobiological regulator, the tension of the cell membrane plays a vital role in governing cellular adhesion, proliferation, and differentiation processes. Additionally, it displayed a dynamic response to mechanical microenvironmental changes. This research systematically examines the mechanoresponsive behaviors of mouse bone marrow mesenchymal stem cells (mBMSCs) that are cultured on poly(dimethylsiloxane) (PDMS) substrates which are functionalized with methyl (-CH3), amino (-NH2), and carboxyl (-COOH) groups. Under both static and stretching conditions, it is found that compared with the -CH3 surface, static culture on the -NH2 and -COOH functionalized surfaces significantly promotes the proliferation of mBMSCs and upregulates the expression of extracellular matrix adhesion-related genes, especially focal adhesion kinase (FAK) and integrin β1. Morphometric analysis reveals that there are concomitant increases in the cell spreading area and the number of pseudopods on these modified surfaces. Mechanical stretching stimulation not only amplifies these cellular responses but also leads to more uniform FAK distribution. The assessment by atomic force microscopy (AFM) shows that both chemical functionalization (-NH2/-COOH) and stretch stimulation reduce the deformability of the cell membrane, and the -NH2 modification exhibits a greater membrane-stiffening effect than -COOH.
Collapse
Affiliation(s)
- Yunqing Ye
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China
| | - Haoyang Huang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China
| | - Hong Li
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China
| | - Gang Wu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
3
|
Roets B, Abrahamse H, Crous A. Biomaterial Properties and Differentiation Strategies for Tenogenic Differentiation of Mesenchymal Stem Cells. Cells 2025; 14:452. [PMID: 40136701 PMCID: PMC11940850 DOI: 10.3390/cells14060452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Tendinopathy is a prevalent musculoskeletal condition that affects both aging populations and individuals involved in repetitive, high-intensity activities, such as athletes. Current treatment options primarily address symptom management or involve surgery, which carries a significant risk of complications and re-injury. This highlights the need for regenerative medicine approaches that combine stem cells, biomaterials, and growth factors. However, achieving effective tenogenic differentiation remains challenging due to the absence of standardized differentiation protocols. Consequently, a review of existing research has been conducted to identify optimal biomaterial properties and growth factor protocols. Findings suggest that the ideal biomaterial for tenogenic differentiation should feature a 3D structure to preserve tenogenic expression, incorporate a combination of aligned micro- and nanofibers to promote differentiation, and require further investigation into optimal stiffness. Additionally, growth factor protocols should include an induction phase to initiate tenogenic lineage commitment, followed by a maintenance phase to support matrix production and maturation.
Collapse
Affiliation(s)
| | | | - Anine Crous
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa; (B.R.); (H.A.)
| |
Collapse
|
4
|
Matsushima T, Hiroshi A. Molecular mechanisms of mechanosensing and plasticity of tendons and ligaments. J Biochem 2024; 176:263-269. [PMID: 38729213 PMCID: PMC11444931 DOI: 10.1093/jb/mvae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Tendons and ligaments, crucial components of the musculoskeletal system, connect muscles to bones. In the realm of sports, tendons and ligaments are vulnerable tissues, with injuries such as Achilles tendon rupture and anterior cruciate ligament tears directly impacting an athlete's career. Furthermore, repetitive trauma and tissue degeneration can lead to conditions like secondary osteoarthritis, ultimately affecting the overall quality of life. Recent research highlights the pivotal role of mechanical stress in maintaining homeostasis within tendons and ligaments. This review delves into the latest insights on the structure of tendons and ligaments and the plasticity of tendon tissue in response to mechanical loads.
Collapse
Affiliation(s)
- Takahide Matsushima
- Department of Systems BioMedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Asahara Hiroshi
- Department of Systems BioMedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
- Department of Molecular Medicine, Scripps Research, 10550 North Torrey Pines Road, MBB-102, La Jolla, CA 92037, USA
| |
Collapse
|
5
|
Leahy TP, Chenna SS, Soslowsky LJ, Dyment NA. Focal adhesion kinase regulates tendon cell mechanoresponse and physiological tendon development. FASEB J 2024; 38:e70050. [PMID: 39259535 PMCID: PMC11522781 DOI: 10.1096/fj.202400151r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/17/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024]
Abstract
Tendons enable locomotion by transmitting high tensile mechanical forces between muscle and bone via their dense extracellular matrix (ECM). The application of extrinsic mechanical stimuli via muscle contraction is necessary to regulate healthy tendon function. Specifically, applied physiological levels of mechanical loading elicit an anabolic tendon cell response, while decreased mechanical loading evokes a degradative tendon state. Although the tendon response to mechanical stimuli has implications in disease pathogenesis and clinical treatment strategies, the cell signaling mechanisms by which tendon cells sense and respond to mechanical stimuli within the native tendon ECM remain largely unknown. Therefore, we explored the role of cell-ECM adhesions in regulating tendon cell mechanotransduction by perturbing the genetic expression and signaling activity of focal adhesion kinase (FAK) through both in vitro and in vivo approaches. We determined that FAK regulates tendon cell spreading behavior and focal adhesion morphology, nuclear deformation in response to applied mechanical strain, and mechanosensitive gene expression. In addition, our data reveal that FAK signaling plays an essential role in in vivo tendon development and postnatal growth, as FAK-knockout mouse tendons demonstrated reduced tendon size, altered mechanical properties, differences in cellular composition, and reduced maturity of the deposited ECM. These data provide a foundational understanding of the role of FAK signaling as a critical regulator of in situ tendon cell mechanotransduction. Importantly, an increased understanding of tendon cell mechanotransductive mechanisms may inform clinical practice as well as lead to the discovery of diagnostic and/or therapeutic molecular targets.
Collapse
Affiliation(s)
- Thomas P. Leahy
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Srish S. Chenna
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Louis J. Soslowsky
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nathaniel A. Dyment
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Melzer M, Niebert S, Heimann M, Ullm F, Pompe T, Scheiner-Bobis G, Burk J. Differential Smad2/3 linker phosphorylation is a crosstalk mechanism of Rho/ROCK and canonical TGF-β3 signaling in tenogenic differentiation. Sci Rep 2024; 14:10393. [PMID: 38710741 PMCID: PMC11074336 DOI: 10.1038/s41598-024-60717-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 04/26/2024] [Indexed: 05/08/2024] Open
Abstract
The transforming growth factor (TGF)-β3 is a well-known inducer for tenogenic differentiation, signaling via the Smad2/3 pathway. Furthermore, other factors like extracellular matrix or mechanical force can induce tenogenic differentiation and possibly alter the response to TGF-β3 by signaling via the Rho/ROCK pathway. The aim of this study was to investigate the interplay of Rho/ROCK and TGF-β3/Smad signaling in tenogenic differentiation, with the Smad2/3 molecule hypothesized as a possible interface. Cultured as monolayers or on collagen I matrices, mesenchymal stromal cells (MSC) were treated with the ROCK inhibitor Y-27632 (10 µM), TGF-β3 (10 ng/ml) or both combined. Control cells were cultured accordingly, without Y-27632 and/or without TGF-β3. At different time points, MSC were analyzed by real-time RT-PCR, immunofluorescence, and Western blot. Cultivation of MSC on collagen matrices and ROCK inhibition supported tenogenic differentiation and fostered the effect of TGF-β3. The phosphorylation of the linker region of Smad2 was reduced by cultivation on collagen matrices, but not by ROCK inhibition. The latter, however, led to increased phosphorylation of the linker region of Smad3. In conclusion, collagen matrices and the Rho/ROCK signaling pathway influence the TGF-β3/Smad2/3 pathway by regulating different phosphorylation sites of the Smad linker region.
Collapse
Affiliation(s)
- Michaela Melzer
- Equine Clinic (Surgery, Orthopedics), Faculty of Veterinary Medicine, Justus-Liebig-University, 35392, Giessen, Germany
| | - Sabine Niebert
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Manuela Heimann
- Institute of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, Justus-Liebig-University, 35392, Giessen, Germany
| | - Franziska Ullm
- Institute of Biochemistry, Faculty of Life Science, Leipzig University, 04103, Leipzig, Germany
- FILK Freiberg Institute GmbH, 09599, Freiberg, Germany
| | - Tilo Pompe
- Institute of Biochemistry, Faculty of Life Science, Leipzig University, 04103, Leipzig, Germany
| | - Georgios Scheiner-Bobis
- Institute of Biochemistry and Endocrinology, Faculty of Veterinary Medicine, Justus-Liebig-University, 35392, Giessen, Germany
| | - Janina Burk
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Su DB, Zhao ZX, Yin DC, Ye YJ. Promising application of pulsed electromagnetic fields on tissue repair and regeneration. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 187:36-50. [PMID: 38280492 DOI: 10.1016/j.pbiomolbio.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/14/2023] [Accepted: 01/19/2024] [Indexed: 01/29/2024]
Abstract
Tissue repair and regeneration is a vital biological process in organisms, which is influenced by various internal mechanisms and microenvironments. Pulsed electromagnetic fields (PEMFs) are becoming a potential medical technology due to its advantages of effectiveness and non-invasiveness. Numerous studies have demonstrated that PEMFs can stimulate stem cell proliferation and differentiation, regulate inflammatory reactions, accelerate wound healing, which is of great significance for tissue regeneration and repair, providing a solid basis for enlarging its clinical application. However, some important issues such as optimal parameter system and potential deep mechanisms remain to be resolved due to PEMFs window effect and biological complexity. Thus, it is of great importance to comprehensively summarizing and analyzing the literature related to the biological effects of PEMFs in tissue regeneration and repair. This review expounded the biological effects of PEMFs on stem cells, inflammation response, wound healing and musculoskeletal disorders in order to improve the application value of PEMFs in medicine. It is believed that with the continuous exploration of biological effects of PEMFs, it will be applied increasingly widely to tissue repair and other diseases.
Collapse
Affiliation(s)
- Dan-Bo Su
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Zi-Xu Zhao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ya-Jing Ye
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
8
|
Giacomini F, Baião Barata D, Suk Rho H, Tahmasebi Birgani Z, van Blitterswijk C, Giselbrecht S, Truckenmüller R, Habibović P. Microfluidically Aligned Collagen to Maintain the Phenotype of Tenocytes In Vitro. Adv Healthc Mater 2024; 13:e2303672. [PMID: 37902084 PMCID: PMC11468977 DOI: 10.1002/adhm.202303672] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Indexed: 10/31/2023]
Abstract
Tendon is a highly organized tissue that transmits forces between muscle and bone. The architecture of the extracellular matrix of tendon, predominantly from collagen type I, is important for maintaining tenocyte phenotype and function. Therefore, in repair and regeneration of damaged and diseased tendon tissue, it is crucial to restore the aligned arrangement of the collagen type I fibers of the original matrix. To this end, a novel, user-friendly microfluidic piggyback platform is developed allowing the controlled patterned formation and alignment of collagen fibers simply on the bottom of culture dishes. Rat tenocytes cultured on the micropatterns of aligned fibrous collagen exhibit a more elongated morphology. The cells also show an increased expression of tenogenic markers at the gene and protein level compared to tenocytes cultured on tissue culture plastic or non-fibrillar collagen coatings. Moreover, using imprinted polystyrene replicas of aligned collagen fibers, this work shows that the fibrillar structure of collagen per se affects the tenocyte morphology, whereas the biochemical nature of collagen plays a prominent role in the expression of tenogenic markers. Beyond the controlled provision of aligned collagen, the microfluidic platform can aid in developing more physiologically relevant in vitro models of tendon and its regeneration.
Collapse
Affiliation(s)
- Francesca Giacomini
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - David Baião Barata
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
- Instituto de Medicina MolecularFaculdade de MedicinaUniversidade de LisboaAvenida Professor Egas MonizLisbon1649‐028Portugal
| | - Hoon Suk Rho
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Zeinab Tahmasebi Birgani
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Clemens van Blitterswijk
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| |
Collapse
|
9
|
Pampanella L, Petrocelli G, Abruzzo PM, Zucchini C, Canaider S, Ventura C, Facchin F. Cytochalasins as Modulators of Stem Cell Differentiation. Cells 2024; 13:400. [PMID: 38474364 PMCID: PMC10931372 DOI: 10.3390/cells13050400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Regenerative medicine aims to identify new research strategies for the repair and restoration of tissues damaged by pathological or accidental events. Mesenchymal stem cells (MSCs) play a key role in regenerative medicine approaches due to their specific properties, such as the high rate of proliferation, the ability to differentiate into several cell lineages, the immunomodulatory potential, and their easy isolation with minimal ethical issues. One of the main goals of regenerative medicine is to modulate, both in vitro and in vivo, the differentiation potential of MSCs to improve their use in the repair of damaged tissues. Over the years, much evidence has been collected about the ability of cytochalasins, a large family of 60 metabolites isolated mainly from fungi, to modulate multiple properties of stem cells (SCs), such as proliferation, migration, and differentiation, by altering the organization of the cyto- and the nucleo-skeleton. In this review, we discussed the ability of two different cytochalasins, cytochalasins D and B, to influence specific SC differentiation programs modulated by several agents (chemical or physical) or intra- and extra-cellular factors, with particular attention to human MSCs (hMSCs).
Collapse
Affiliation(s)
- Luca Pampanella
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.P.); (G.P.); (P.M.A.); (C.Z.); (F.F.)
| | - Giovannamaria Petrocelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.P.); (G.P.); (P.M.A.); (C.Z.); (F.F.)
| | - Provvidenza Maria Abruzzo
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.P.); (G.P.); (P.M.A.); (C.Z.); (F.F.)
| | - Cinzia Zucchini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.P.); (G.P.); (P.M.A.); (C.Z.); (F.F.)
| | - Silvia Canaider
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.P.); (G.P.); (P.M.A.); (C.Z.); (F.F.)
| | - Carlo Ventura
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.P.); (G.P.); (P.M.A.); (C.Z.); (F.F.)
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) c/o Eldor Lab, Via Corticella 183, 40129 Bologna, Italy
| | - Federica Facchin
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (L.P.); (G.P.); (P.M.A.); (C.Z.); (F.F.)
| |
Collapse
|
10
|
Guo X, Fan A, Qi X, Liu D, Huang J, Lin W. Indoloquinazoline alkaloids suppress angiogenesis and inhibit metastasis of melanoma cells. Bioorg Chem 2023; 141:106873. [PMID: 37734192 DOI: 10.1016/j.bioorg.2023.106873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/10/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023]
Abstract
Metastasis is the leading cause of cancer-related mortality, targeting angiogenesis emerges as a therapeutic strategy for the treatment of melanoma metastasis. Discovery of new antiangiogenic compounds with specific mechanism of action is still desired. In present study, a bioassay-guidance uncovers the EtOAc extract of a marine-derived fungus Aspergillus clavutus LZD32-24 with significant inhibitory activity against the angiogenesis in Tg (fli1a: EGFP) zebrafish model. Extensive chromatographic fractionation led to the isolation of 48 indoloquinazoline alkaloids, including 21 new analogues namely clavutoines A-U (1-21). Their structures were determined by the spectroscopic data, including the ECD, single crystal X-ray diffraction and quantum chemical calculation for the configurational assignments. Among the bioactive analogues, quinadoline B (QB) showed the most efficacy to suppress the zebrafish vascular outgrowth in zebrafish embryos. QB markedly inhibited the migration, invasion and tube formation with weak cytotoxicity in human umbilical vein endothelial cells (HUVECs). Investigation of the mode of action revealed QB suppressed the ROCK/MYPT1/MLC2/coffin and FAK /Src signaling pathways, and subsequently disrupted actin cytoskeletal organization. In addition, QB reduced the number of new vessels sprouting from the ex vivo chick chorioallantoic membrane (CAM), and inhibited the metastasis of B16F10 melanoma cells in lung of C57BL/6 mice through suppressing angiogenesis. These findings suggest that QB is a potential lead for the development of new antiangiogenic agent to inhibit melanoma metastasis.
Collapse
Affiliation(s)
- Xingchen Guo
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, PR China
| | - Aili Fan
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, PR China
| | - Xinyi Qi
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, PR China
| | - Dong Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, PR China
| | - Jian Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, PR China.
| | - Wenhan Lin
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, PR China; Ningbo Institute of Marine Medicine, Peking University, Beijing 100191, PR China.
| |
Collapse
|
11
|
Bakhshandeh B, Sorboni SG, Ranjbar N, Deyhimfar R, Abtahi MS, Izady M, Kazemi N, Noori A, Pennisi CP. Mechanotransduction in tissue engineering: Insights into the interaction of stem cells with biomechanical cues. Exp Cell Res 2023; 431:113766. [PMID: 37678504 DOI: 10.1016/j.yexcr.2023.113766] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Stem cells in their natural microenvironment are exposed to biochemical and biophysical cues emerging from the extracellular matrix (ECM) and neighboring cells. In particular, biomechanical forces modulate stem cell behavior, biological fate, and early developmental processes by sensing, interpreting, and responding through a series of biological processes known as mechanotransduction. Local structural changes in the ECM and mechanics are driven by reciprocal activation of the cell and the ECM itself, as the initial deposition of matrix proteins sequentially affects neighboring cells. Recent studies on stem cell mechanoregulation have provided insight into the importance of biomechanical signals on proper tissue regeneration and function and have shown that precise spatiotemporal control of these signals exists in stem cell niches. Against this background, the aim of this work is to review the current understanding of the molecular basis of mechanotransduction by analyzing how biomechanical forces are converted into biological responses via cellular signaling pathways. In addition, this work provides an overview of advanced strategies using stem cells and biomaterial scaffolds that enable precise spatial and temporal control of mechanical signals and offer great potential for the fields of tissue engineering and regenerative medicine will be presented.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | | | - Nika Ranjbar
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Roham Deyhimfar
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Maryam Sadat Abtahi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mehrnaz Izady
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Navid Kazemi
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Atefeh Noori
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Denmark.
| |
Collapse
|
12
|
Shojaee A. Equine tendon mechanical behaviour: Prospects for repair and regeneration applications. Vet Med Sci 2023; 9:2053-2069. [PMID: 37471573 PMCID: PMC10508504 DOI: 10.1002/vms3.1205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/03/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023] Open
Abstract
Tendons are dense connective tissues that play an important role in the biomechanical function of the musculoskeletal system. The mechanical forces have been implicated in every aspect of tendon biology. Tendon injuries are frequently occurring and their response to treatments is often unsatisfactory. A better understanding of tendon biomechanics and mechanobiology can help develop treatment options to improve clinical outcomes. Recently, tendon tissue engineering has gained more attention as an alternative treatment due to its potential to overcome the limitations of current treatments. This review first provides a summary of tendon mechanical properties, focusing on recent findings of tendon mechanobiological responses. In the next step, we highlight the biomechanical parameters of equine energy-storing and positional tendons. The final section is devoted to how mechanical loading contributes to tenogenic differentiation using bioreactor systems. This study may help develop novel strategies for tendon injury prevention or accelerate and improve tendon healing.
Collapse
Affiliation(s)
- Asiyeh Shojaee
- Division of PhysiologyDepartment of Basic SciencesFaculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| |
Collapse
|
13
|
Safaei S, Sajed R, Shariftabrizi A, Dorafshan S, Saeednejad Zanjani L, Dehghan Manshadi M, Madjd Z, Ghods R. Tumor matrix stiffness provides fertile soil for cancer stem cells. Cancer Cell Int 2023; 23:143. [PMID: 37468874 PMCID: PMC10357884 DOI: 10.1186/s12935-023-02992-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023] Open
Abstract
Matrix stiffness is a mechanical characteristic of the extracellular matrix (ECM) that increases from the tumor core to the tumor periphery in a gradient pattern in a variety of solid tumors and can promote proliferation, invasion, metastasis, drug resistance, and recurrence. Cancer stem cells (CSCs) are a rare subpopulation of tumor cells with self-renewal, asymmetric cell division, and differentiation capabilities. CSCs are thought to be responsible for metastasis, tumor recurrence, chemotherapy resistance, and consequently poor clinical outcomes. Evidence suggests that matrix stiffness can activate receptors and mechanosensor/mechanoregulator proteins such as integrin, FAK, and YAP, modulating the characteristics of tumor cells as well as CSCs through different molecular signaling pathways. A deeper understanding of the effect of matrix stiffness on CSCs characteristics could lead to development of innovative cancer therapies. In this review, we discuss how the stiffness of the ECM is sensed by the cells and how the cells respond to this environmental change as well as the effect of matrix stiffness on CSCs characteristics and also the key malignant processes such as proliferation and EMT. Then, we specifically focus on how increased matrix stiffness affects CSCs in breast, lung, liver, pancreatic, and colorectal cancers. We also discuss how the molecules responsible for increased matrix stiffness and the signaling pathways activated by the enhanced stiffness can be manipulated as a therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Sadegh Safaei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
| | - Roya Sajed
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
| | - Ahmad Shariftabrizi
- Division of Nuclear Medicine, Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Division of Nuclear Medicine, Department of Radiology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Shima Dorafshan
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
| | - Leili Saeednejad Zanjani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Department of Pathology and Genomic Medicine, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Masoumeh Dehghan Manshadi
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
| | - Zahra Madjd
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran.
| | - Roya Ghods
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran.
| |
Collapse
|
14
|
Karimi E, Vahedi N, Sarbandi RR, Parandakh A, Ganjoury C, Sigaroodi F, Najmoddin N, Tabatabaei M, Tafazzoli-Shadpour M, Ardeshirylajimi A, Khani MM. Nanoscale vibration could promote tenogenic differentiation of umbilical cord mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2023:10.1007/s11626-023-00780-4. [PMID: 37405626 DOI: 10.1007/s11626-023-00780-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/24/2023] [Indexed: 07/06/2023]
Abstract
Regulation of mesenchymal stem cell (MSC) fate for targeted cell therapy applications has been a subject of interest, particularly for tissues such as tendons that possess a marginal regenerative capacity. Control of MSCs' fate into the tendon-specific lineage has mainly been achieved by implementation of chemical growth factors. Mechanical stimuli or 3-dimensional (D) scaffolds have been used as an additional tool for the differentiation of MSCs into tenocytes, but oftentimes, they require a sophisticated bioreactor or a complex scaffold fabrication technique which reduces the feasibility of the proposed method to be used in practice. Here, we used nanovibration to induce the differentiation of MSCs toward the tenogenic fate solely by the use of nanovibration and without the need for growth factors or complex scaffolds. MSCs were cultured on 2D cell culture dishes that were connected to piezo ceramic arrays to apply nanovibration (30-80 nm and 1 kHz frequency) over 7 and 14 d. We observed that nanovibration resulted in significant overexpression of tendon-related markers in both gene expression and protein expression levels, while there was no significant differentiation into adipose and cartilage lineages. These findings could be of assistance in the mechanoregulation of MSCs for stem cell engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Elahe Karimi
- Department of Tissue Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Negin Vahedi
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Reza Ramezani Sarbandi
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Azim Parandakh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Camellia Ganjoury
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faraz Sigaroodi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Najmeh Najmoddin
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Tabatabaei
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | | | - Abdolreza Ardeshirylajimi
- Sina Cell Research and Product Center, Tehran, Iran
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Mohammad-Mehdi Khani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Melzer M, Burk J, Guest DJ, Dudhia J. Influence of Rho/ROCK inhibitor Y-27632 on proliferation of equine mesenchymal stromal cells. Front Vet Sci 2023; 10:1154987. [PMID: 37346276 PMCID: PMC10279950 DOI: 10.3389/fvets.2023.1154987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Mesenchymal stromal cells (MSC) isolated form bone marrow and adipose tissue are the most common cells used for cell therapy of orthopedic diseases. MSC derived from different tissues show differences in terms of their proliferation, differentiation potential and viability in prolonged cell culture. This suggests that there may be subtle differences in intracellular signaling pathways that modulate these cellular characteristics. The Rho/ROCK signaling pathway is essential for many cellular functions. Targeting of this pathway by the ROCK inhibitor Y-27632 has been shown to be beneficial for cell viability and proliferation of different cell types. The aim of this study was to investigate the effects of Rho/ROCK inhibition on equine MSC proliferation using bone marrow-derived MSC (BMSC) and adipose-derived MSC (ASC). Primary ASC and BMSC were stimulated with or without 10 ng/mL TGF-β3 or 10 μM Y-27632, as well as both in combination. Etoposide at 10 μM was used as a positive control for inhibition of cell proliferation. After 48 h of stimulation, cell morphology, proliferation activity and gene expression of cell senescence markers p53 and p21 were assessed. ASC showed a trend for higher basal proliferation than BMSC, which was sustained following stimulation with TGF-β3. This included a higher proliferation with TGF-β3 stimulation compared to Y-27632 stimulation (p < 0.01), but not significantly different to the no treatment control when used in combination. Expression of p21 and p53 was not altered by stimulation with TGF-β3 and/or Y-27632 in either cell type. In summary, the Rho/ROCK inhibitor Y-27632 had no effect on proliferation activity and did not induce cell senescence in equine ASC and BMSC.
Collapse
Affiliation(s)
- Michaela Melzer
- Equine Clinic (Surgery, Orthopedics), Faculty of Veterinary Medicine, Justus Liebig University, Giessen, Germany
| | - Janina Burk
- Equine Clinic (Surgery, Orthopedics), Faculty of Veterinary Medicine, Justus Liebig University, Giessen, Germany
| | - Deborah J. Guest
- Department of Clinical Sciences and Services, Royal Veterinary College, Hertfordshire, United Kingdom
| | - Jayesh Dudhia
- Department of Clinical Sciences and Services, Royal Veterinary College, Hertfordshire, United Kingdom
| |
Collapse
|
16
|
Chretien A, Mabilleau G, Lebacq J, Docquier PL, Behets C. Beneficial Effects of Zoledronic Acid on Tendons of the Osteogenesis Imperfecta Mouse (Oim). Pharmaceuticals (Basel) 2023; 16:832. [PMID: 37375779 DOI: 10.3390/ph16060832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Osteogenesis imperfecta (OI) is a genetic disorder of connective tissue characterized by spontaneous fractures, bone deformities, impaired growth and posture, as well as extra-skeletal manifestations. Recent studies have underlined an impairment of the osteotendinous complex in mice models of OI. The first objective of the present work was to further investigate the properties of tendons in the osteogenesis imperfecta mouse (oim), a model characterized by a mutation in the COL1A2 gene. The second objective was to identify the possible beneficial effects of zoledronic acid on tendons. Oim received a single intravenous injection of zoledronic acid (ZA group) at 5 weeks and were euthanized at 14 weeks. Their tendons were compared with those of untreated oim (oim group) and control mice (WT group) by histology, mechanical tests, western blotting and Raman spectroscopy. The ulnar epiphysis had a significantly lower relative bone surface (BV/TV) in oim than WT mice. The tendon of the triceps brachii was also significantly less birefringent and displayed numerous chondrocytes aligned along the fibers. ZA mice showed an increase in BV/TV of the ulnar epiphysis and in tendon birefringence. The tendon of the flexor digitorum longus was significantly less viscous in oim than WT mice; in ZA-treated mice, there was an improvement of viscoelastic properties, especially in the toe region of stress-strain curve, which corresponds to collagen crimp. The tendons of both oim and ZA groups did not show any significant change in the expression of decorin or tenomodulin. Finally, Raman spectroscopy highlighted differences in material properties between ZA and WT tendons. There was also a significant increase in the rate of hydroxyproline in the tendons of ZA mice compared with oim ones. This study highlighted changes in matrix organization and an alteration of mechanical properties in oim tendons; zoledronic acid treatment had beneficial effects on these parameters. In the future, it will be interesting to better understand the underlying mechanisms which are possibly linked to a greater solicitation of the musculoskeletal system.
Collapse
Affiliation(s)
- Antoine Chretien
- Pole of Morphology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Guillaume Mabilleau
- Univ Angers, Nantes Université, Oniris, Inserm, UMR_S 1229-RMeS, REGOS, SFR ICAT, F-49000 Angers, France
- Centre Hospitalier Universitaire d'Angers, Department of Cell and Tissue Pathology, Bone Pathology Unit, F-49000 Angers, France
| | - Jean Lebacq
- Institute of NeuroScience (IoNS), Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Pierre-Louis Docquier
- Neuromusculoskeletal Lab, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Catherine Behets
- Pole of Morphology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|
17
|
Ambattu LA, Yeo LY. Sonomechanobiology: Vibrational stimulation of cells and its therapeutic implications. BIOPHYSICS REVIEWS 2023; 4:021301. [PMID: 38504927 PMCID: PMC10903386 DOI: 10.1063/5.0127122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/27/2023] [Indexed: 03/21/2024]
Abstract
All cells possess an innate ability to respond to a range of mechanical stimuli through their complex internal machinery. This comprises various mechanosensory elements that detect these mechanical cues and diverse cytoskeletal structures that transmit the force to different parts of the cell, where they are transcribed into complex transcriptomic and signaling events that determine their response and fate. In contrast to static (or steady) mechanostimuli primarily involving constant-force loading such as compression, tension, and shear (or forces applied at very low oscillatory frequencies (≤ 1 Hz) that essentially render their effects quasi-static), dynamic mechanostimuli comprising more complex vibrational forms (e.g., time-dependent, i.e., periodic, forcing) at higher frequencies are less well understood in comparison. We review the mechanotransductive processes associated with such acoustic forcing, typically at ultrasonic frequencies (> 20 kHz), and discuss the various applications that arise from the cellular responses that are generated, particularly for regenerative therapeutics, such as exosome biogenesis, stem cell differentiation, and endothelial barrier modulation. Finally, we offer perspectives on the possible existence of a universal mechanism that is common across all forms of acoustically driven mechanostimuli that underscores the central role of the cell membrane as the key effector, and calcium as the dominant second messenger, in the mechanotransduction process.
Collapse
Affiliation(s)
- Lizebona August Ambattu
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne VIC 3000, Australia
| | - Leslie Y. Yeo
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne VIC 3000, Australia
| |
Collapse
|
18
|
Hallström GF, Jones DL, Locke RC, Bonnevie ED, Kim SY, Laforest L, Garcia DC, Mauck RL. Microenvironmental mechanoactivation through Yap/Taz suppresses chondrogenic gene expression. Mol Biol Cell 2023; 34:ar73. [PMID: 37043309 PMCID: PMC10295477 DOI: 10.1091/mbc.e22-12-0543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/23/2023] [Accepted: 04/06/2023] [Indexed: 04/13/2023] Open
Abstract
Chondrocyte phenotype is preserved when cells are round and the actin cytoskeleton is cortical. Conversely, these cells rapidly dedifferentiate in vitro with increased mechanoactive Rho signaling, which increases cell size and causes large actin stress fiber to form. While the effects of Rho on chondrocyte phenotype are well established, the molecular mechanism is not yet fully elucidated. Yap, a transcriptional coregulator, is regulated by Rho in a mechanotransductive manner and can suppress chondrogenesis in vivo. Here, we sought to elucidate the relationship between mechanoactive Rho and Yap on chondrogenic gene expression. We first show that decreasing mechanoactive state through Rho inhibition results in a broad increase in chondrogenic gene expression. Next, we show that Yap and its coregulator Taz are negative regulators of chondrogenic gene expression, and removal of these factors promotes chondrogenesis even in environments that promote cell spreading. Finally, we establish that Yap/Taz is essential for translating Rho-mediated signals to negatively regulate chondrogenic gene expression, and that its removal negates the effects of increased Rho signaling. Together, these data indicate that Rho is a mechanoregulator of chondrogenic differentiation, and that its impact on chondrogenic expression is exerted principally through mechanically induced translocation and activity of Yap and Taz.
Collapse
Affiliation(s)
- Grey F. Hallström
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Dakota L. Jones
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
| | - Ryan C. Locke
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Edward D. Bonnevie
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Sung Yeon Kim
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Lorielle Laforest
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
| | - Diana Cruz Garcia
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| |
Collapse
|
19
|
Khalifa MO, Moriwaki T, Zhang S, Zhou W, Ito K, Li TS. Negative pressure induces dedifferentiation of hepatocytes via RhoA/ROCK pathway. Biochem Biophys Res Commun 2023; 667:104-110. [PMID: 37210870 DOI: 10.1016/j.bbrc.2023.05.042] [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: 04/21/2023] [Revised: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 05/23/2023]
Abstract
Biomechanical forces are known to regulate the biological behaviors of cells. Although negative pressure has been used for wound healing, it is still unknown about its role in regulating cell plasticity. We investigated whether negative pressure could induce the dedifferentiation of hepatocytes. Using a commercial device, we found that the exposure of primary human hepatocytes to -50 mmHg quickly induced the formation of stress fibers and obviously changed cell morphology in 72 h. Moreover, the exposure of hepatocytes to -50 mmHg significantly upregulated RhoA, ROCK1, and ROCK2 in 1-6 h, and dramatically enhanced the expression of marker molecules on "stemness", such as OCT4, SOX2, KLF4, MYC, NANOG, and CD133 in 6-72 h. However, all these changes in hepatocytes induced by -50 mmHg stimulation were almost abrogated by ROCK inhibitor Y27623. Our data suggest that an appropriate force of negative pressure stimulation can effectively induce the dedifferentiation of hepatocytes via RhoA/ROCK pathway activation.
Collapse
Affiliation(s)
- Mahmoud Osman Khalifa
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan; Department of Anatomy and Embryology, Veterinary Medicine, Aswan University, Aswan, Egypt; Department of Molecular Bone Biology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Takahito Moriwaki
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Shouhua Zhang
- Department of General Surgery, Jiangxi Provincial Children's Hospital, Nanchang, Jiangxi, China
| | - Wei Zhou
- Department of Gastrointestinal Surgery, Jiangxi Provincial Cancer Hospital Nanchang, Jiangxi Province, China
| | - Kosei Ito
- Department of Molecular Bone Biology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| |
Collapse
|
20
|
Xie N, Xiao C, Shu Q, Cheng B, Wang Z, Xue R, Wen Z, Wang J, Shi H, Fan D, Liu N, Xu F. Cell response to mechanical microenvironment cues via Rho signaling: From mechanobiology to mechanomedicine. Acta Biomater 2023; 159:1-20. [PMID: 36717048 DOI: 10.1016/j.actbio.2023.01.039] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/30/2023]
Abstract
Mechanical cues in the cell microenvironment such as those from extracellular matrix properties, stretching, compression and shear stress, play a critical role in maintaining homeostasis. Upon sensing mechanical stimuli, cells can translate these external forces into intracellular biochemical signals to regulate their cellular behaviors, but the specific mechanisms of mechanotransduction at the molecular level remain elusive. As a subfamily of the Ras superfamily, Rho GTPases have been recognized as key intracellular mechanotransduction mediators that can regulate multiple cell activities such as proliferation, migration and differentiation as well as biological processes such as cytoskeletal dynamics, metabolism, and organ development. However, the upstream mechanosensors for Rho proteins and downstream effectors that respond to Rho signal activation have not been well illustrated. Moreover, Rho-mediated mechanical signals in previous studies are highly context-dependent. In this review, we systematically summarize the types of mechanical cues in the cell microenvironment and provide recent advances on the roles of the Rho-based mechanotransduction in various cell activities, physiological processes and diseases. Comprehensive insights into the mechanical roles of Rho GTPase partners would open a new paradigm of mechanomedicine for a variety of diseases. STATEMENT OF SIGNIFICANCE: In this review, we highlight the critical role of Rho GTPases as signal mediators to respond to physical cues in microenvironment. This article will add a distinct contribution to this set of knowledge by intensively addressing the relationship between Rho signaling and mechanobiology/mechanotransduction/mechanomedcine. This topic has not been discussed by the journal, nor has it yet been developed by the field. The comprehensive picture that will develop, from molecular mechanisms and engineering methods to disease treatment strategies, represents an important and distinct contribution to the field. We hope that this review would help researchers in various fields, especially clinicians, oncologists and bioengineers, who study Rho signal pathway and mechanobiology/mechanotransduction, understand the critical role of Rho GTPase in mechanotransduction.
Collapse
Affiliation(s)
- Ning Xie
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Cailan Xiao
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Qiuai Shu
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Bo Cheng
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Ziwei Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Runxin Xue
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Zhang Wen
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Jinhai Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Haitao Shi
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an Shaanxi 710049, China.
| | - Na Liu
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| |
Collapse
|
21
|
Deng R, Kang R, Jin X, Wang Z, Liu X, Wang Q, Xie L. Mechanical stimulation promotes MSCs healing the lesion of intervertebral disc annulus fibrosus. Front Bioeng Biotechnol 2023; 11:1137199. [PMID: 36845186 PMCID: PMC9950411 DOI: 10.3389/fbioe.2023.1137199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/26/2023] [Indexed: 02/12/2023] Open
Abstract
Mesenchymal stem cells (MSCs) and scaffolds offer promising perspectives for annulus fibrosus (AF) repair. The repair effect was linked to features of the local mechanical environment related to the differentiation of MSCs. In this study, we established a Fibrinogen-Thrombin-Genipin (Fib-T-G) gel which is sticky and could transfer strain force from AF tissue to the human mesenchymal stem cells (hMSCs) embedded in the gel. After the Fib-T-G biological gel was injected into the AF fissures, the histology scores of intervertebral disc (IVD) and AF tissue showed that Fib-T-G gel could better repair the AF fissure in caudal IVD of rats, and increase the expression of AF-related proteins including Collagen 1 (COL1), Collagen 2 (COL2) as well as mechanotransduction-related proteins including RhoA and ROCK1. To clarify the mechanism that sticky Fib-T-G gel induces the healing of AF fissures and the differentiation of hMSCs, we further investigated the differentiation of hMSCs under mechanical strain in vitro. It was demonstrated that both AF-specific genes, including Mohawk and SOX-9, and ECM markers (COL1, COL2, aggrecan) of hMSCs were up-regulated in the environment of strain force. Moreover, RhoA/ROCK1 proteins were also found to be significantly up-regulated. In addition, we further -demonstrated that the fibrochondroinductive effect of the mechanical microenvironment process could be significantly blocked or up-regulated by inhibiting the RhoA/ROCK1 pathway or overexpressing RhoA in MSCs, respectively. Summarily, this study will provide a therapeutic alternative to repair AF tears and provide evidence that RhoA/ROCK1 is vital for hMSCs response to mechanical strain and AF-like differentiation.
Collapse
Affiliation(s)
- Rongrong Deng
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ran Kang
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China,Department of Orthopedics, Nanjing Lishui Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China,*Correspondence: Ran Kang, ; Xin Liu, ; Lin Xie,
| | - Xiaoyu Jin
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China,Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Zihan Wang
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xin Liu
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China,*Correspondence: Ran Kang, ; Xin Liu, ; Lin Xie,
| | - Qing Wang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Lin Xie
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China,*Correspondence: Ran Kang, ; Xin Liu, ; Lin Xie,
| |
Collapse
|
22
|
Dede Eren A, Lucassen AWA, Tuvshindorj U, Truckenmüller R, Giselbrecht S, Eren ED, Tas MO, Sudarsanam P, de Boer J. Cells Dynamically Adapt to Surface Geometry by Remodeling Their Focal Adhesions and Actin Cytoskeleton. Front Cell Dev Biol 2022; 10:863721. [PMID: 35721512 PMCID: PMC9203963 DOI: 10.3389/fcell.2022.863721] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/02/2022] [Indexed: 01/16/2023] Open
Abstract
Cells probe their environment and adapt their shape accordingly via the organization of focal adhesions and the actin cytoskeleton. In an earlier publication, we described the relationship between cell shape and physiology, for example, shape-induced differentiation, metabolism, and proliferation in mesenchymal stem cells and tenocytes. In this study, we investigated how these cells organize their adhesive machinery over time when exposed to microfabricated surfaces of different topographies and adhesive island geometries. We further examined the reciprocal interaction between stress fiber and focal adhesion formation by pharmacological perturbations. Our results confirm the current literature that spatial organization of adhesive sites determines the ability to form focal adhesions and stress fibers. Therefore, cells on roughened surfaces have smaller focal adhesion and fewer stress fibers. Our results further highlight the importance of integrin-mediated adhesion in the adaptive properties of cells and provide clear links to the development of bioactive materials.
Collapse
Affiliation(s)
- Aysegul Dede Eren
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Amy W. A. Lucassen
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Urandelger Tuvshindorj
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - E. Deniz Eren
- Laboratory of Physical Chemistry, Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mehmet Orhan Tas
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Phanikrishna Sudarsanam
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- *Correspondence: Jan de Boer,
| |
Collapse
|
23
|
Rigiracciolo DC, Nohata N, Lappano R, Cirillo F, Talia M, Adame-Garcia SR, Arang N, Lubrano S, De Francesco EM, Belfiore A, Gutkind JS, Maggiolini M. Focal Adhesion Kinase (FAK)-Hippo/YAP transduction signaling mediates the stimulatory effects exerted by S100A8/A9-RAGE system in triple-negative breast cancer (TNBC). JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:193. [PMID: 35655319 PMCID: PMC9164429 DOI: 10.1186/s13046-022-02396-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/17/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Understanding the intricate signaling network involved in triple-negative breast cancer (TNBC) represents a challenge for developing novel therapeutic approaches. Here, we aim to provide novel mechanistic insights on the function of the S100A8/A9-RAGE system in TNBC. METHODS TNM plot analyzer, Kaplan-Meier plotter, Meta-analysis, GEPIA2 and GOBO publicly available datasets were used to evaluate the clinical significance of S100A8/A9 and expression levels of S100A8/A9, RAGE and Filamin family members in breast cancer (BC) subtypes. METABRIC database and Cox proportional hazard model defined the clinical impact of high RAGE expression in BC patients. Multiple bioinformatics programs identified the main enriched pathways within high RAGE expression BC cohorts. By lentiviral system, TNBC cells were engineered to overexpress RAGE. Western blotting, immunofluorescence, nucleus/cytoplasm fractionation, qRT-PCR, gene silencing and luciferase experiments were performed to identify signal transduction mediators engaged by RAGE upon stimulation with S100A8/A9 in TNBC cells. Proliferation, colony formation and transwell migration assays were carried out to evaluate the growth and migratory capacity of TNBC cells. Statistical analysis was performed by ANOVA and independent t-tests. RESULTS We found a remarkable high expression of S100A8 and S100A9 in BC, particularly in HER2-positive and TNBC, with the latter associated to worst clinical outcomes. In addition, high RAGE expression correlated with a poor overall survival in BC. Next, we determined that the S100A8/A9-RAGE system triggers FAK activation by engaging a cytoskeleton mechanosensing complex in TNBC cells. Through bioinformatics analysis, we identified the Hippo pathway as the most enriched in BC patients expressing high RAGE levels. In accordance with these data, we demonstrated the involvement of S100A8/A9-RAGE-FAK signaling in the control of Hippo/YAP activities, and we established the crucial contribution of RAGE-FAK-YAP circuitry in the growth and migratory effects initiated by S100A8/A9 in TNBC cells. CONCLUSIONS The present study provides novel mechanistic insights on RAGE actions in TNBC. Moreover, our findings suggest that RAGE-FAK-YAP transduction pathway could be exploited as a druggable system halting the aggressive TNBC subtype.
Collapse
Affiliation(s)
- Damiano Cosimo Rigiracciolo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.,Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | | | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Marianna Talia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | | | - Nadia Arang
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Simone Lubrano
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | | | - Antonino Belfiore
- Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | - J Silvio Gutkind
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA. .,Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA.
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
| |
Collapse
|
24
|
Donderwinkel I, Tuan RS, Cameron NR, Frith JE. Tendon tissue engineering: Current progress towards an optimized tenogenic differentiation protocol for human stem cells. Acta Biomater 2022; 145:25-42. [PMID: 35470075 DOI: 10.1016/j.actbio.2022.04.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/10/2022] [Accepted: 04/18/2022] [Indexed: 12/19/2022]
Abstract
Tendons are integral to our daily lives by allowing movement and locomotion but are frequently injured, leading to patient discomfort and impaired mobility. Current clinical procedures are unable to fully restore the native structure of the tendon, resulting in loss of full functionality, and the weakened tissue following repair often re-ruptures. Tendon tissue engineering, involving the combination of cells with biomaterial scaffolds to form new tendon tissue, holds promise to improve patient outcomes. A key requirement for efficacy in promoting tendon tissue formation is the optimal differentiation of the starting cell populations, most commonly adult tissue-derived mesenchymal stem/stromal cells (MSCs), into tenocytes, the predominant cellular component of tendon tissue. Currently, a lack of consensus on the protocols for effective tenogenic differentiation is hampering progress in tendon tissue engineering. In this review, we discuss the current state of knowledge regarding human stem cell differentiation towards tenocytes and tendon tissue formation. Tendon development and healing mechanisms are described, followed by a comprehensive overview of the current protocols for tenogenic differentiation, including the effects of biochemical and biophysical cues, and their combination, on tenogenesis. Lastly, a synthesis of the key features of these protocols is used to design future approaches. The holistic evaluation of current knowledge should facilitate and expedite the development of efficacious stem cell tenogenic differentiation protocols with future impact in tendon tissue engineering. STATEMENT OF SIGNIFICANCE: The lack of a widely-adopted tenogenic differentiation protocol has been a major hurdle in the tendon tissue engineering field. Building on current knowledge on tendon development and tendon healing, this review surveys peer-reviewed protocols to present a holistic evaluation and propose a pathway to facilitate and expedite the development of a consensus protocol for stem cell tenogenic differentiation and tendon tissue engineering.
Collapse
|
25
|
Guo X, Wang X, Tang H, Ren Y, Li D, Yi B, Zhang Y. Engineering a Mechanoactive Fibrous Substrate with Enhanced Efficiency in Regulating Stem Cell Tenodifferentiation. ACS APPLIED MATERIALS & INTERFACES 2022; 14:23219-23231. [PMID: 35544769 DOI: 10.1021/acsami.2c04294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Electrospun-aligned fibers in ultrathin fineness have previously demonstrated a limited capacity in driving stem cells to differentiate into tendon-like cells. In view of the tendon's mechanoactive nature, endowing such aligned fibrous structure with mechanoactivity to exert in situ mechanical stimulus by itself, namely, without any forces externally applied, is likely to potentiate its efficiency of tenogenic induction. To test this hypothesis, in this study, a shape-memory-capable poly(l-lactide-co-caprolactone) (PLCL) copolymer was electrospun into aligned fibrous form followed by a "stretching-recovery" shape-programming procedure to impart shape memory capability. Thereafter, in the absence of tenogenic supplements, human adipose-derived stem cells (ADSCs) were cultured on the programmed fibrous substrates for a duration of 7 days, and the effects of constrained recovery resultant stress-stiffening on cell morphology, proliferation, and tenogenic differentiation were examined. The results indicate that the in situ enacted mechanical stimulus due to shape memory effect (SME) did not have adverse influence on cell viability and proliferation, but significantly promoted cellular elongation along the direction of fiber alignment. Moreover, it revealed that tendon-specific protein markers such as tenomodulin (TNMD) and tenascin-C (TNC) and gene expression of scleraxis (SCX), TNMD, TNC, and collagen I (COL I) were significantly upregulated on the mechanoactive fibrous substrate with higher recovery stress compared to the counterparts. Mechanistically, the Rho/ROCK signaling pathway was identified to be involved in the substrate self-actuation-induced enhancement in tenodifferentiation. Together, these results suggest that constrained shape recovery stress may be employed as an innovative loading modality to regulate the stem cell tenodifferentiation by presenting the fibrous substrate with an aligned tendon-like topographical cue and an additional mechanoactivity. This newly demonstrated paradigm in modulating stem cell tenodifferentiation may improve the efficacy of tendon tissue engineering strategy for tendon healing and regeneration.
Collapse
Affiliation(s)
- Xuran Guo
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
| | - Xianliu Wang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
| | - Han Tang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
| | - Yajuan Ren
- Longhua Hospital affiliated to the Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Donghong Li
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
| | - Bingcheng Yi
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital affiliated to the Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yanzhong Zhang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital affiliated to the Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| |
Collapse
|
26
|
He P, Ruan D, Huang Z, Wang C, Xu Y, Cai H, Liu H, Fei Y, Heng BC, Chen W, Shen W. Comparison of Tendon Development Versus Tendon Healing and Regeneration. Front Cell Dev Biol 2022; 10:821667. [PMID: 35141224 PMCID: PMC8819183 DOI: 10.3389/fcell.2022.821667] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/07/2022] [Indexed: 12/27/2022] Open
Abstract
Tendon is a vital connective tissue in human skeletal muscle system, and tendon injury is very common and intractable in clinic. Tendon development and repair are two closely related but still not fully understood processes. Tendon development involves multiple germ layer, as well as the regulation of diversity transcription factors (Scx et al.), proteins (Tnmd et al.) and signaling pathways (TGFβ et al.). The nature process of tendon repair is roughly divided in three stages, which are dominated by various cells and cell factors. This review will describe the whole process of tendon development and compare it with the process of tendon repair, focusing on the understanding and recent advances in the regulation of tendon development and repair. The study and comparison of tendon development and repair process can thus provide references and guidelines for treatment of tendon injuries.
Collapse
Affiliation(s)
- Peiwen He
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Zizhan Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Canlong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Yiwen Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Honglu Cai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Hengzhi Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Yang Fei
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School of Stomatology, Bejing, China
| | - Weishan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Weishan Chen, ; Weiliang Shen,
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
- *Correspondence: Weishan Chen, ; Weiliang Shen,
| |
Collapse
|
27
|
He Q, Lin J, Zhou F, Cai D, Yan Y, Shan Y, Zhang S, Li T, Yao X, Ouyang H. “Musical dish” efficiently induces osteogenic differentiation of mesenchymal stem cells through music derived micro‐stretch with variable frequency. Bioeng Transl Med 2022; 7:e10291. [PMID: 35600662 PMCID: PMC9115692 DOI: 10.1002/btm2.10291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/01/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022] Open
Abstract
Nonuniform microstretching (NUMS) naturally occurs in real bone tissues in vivo, but its profound effects have not been identified yet. In order to explore the biological effects of NUMS and static stretch (uniform stretch [US]) on cells, a new “musical dish” device was developed. Musical signal was used to provide NUMS to cells. More stress fibers, arranging along the long axis of cells, were formed throughout the cells under NUMS, compared with US and untreated control group, although cell morphology did not show any alteration. Whole transcriptome sequencing revealed enhanced osteogenic differentiation of cells after NUMS treatment. Cells in the NUMS group showed a higher expression of bone‐related genes, while genes related to stemness and other lineages were down‐regulated. Our results give insights into the biological effects of NUMS and US on stem cell osteogenic differentiation, suggesting beneficial effects of micromechanical stimulus for osteogenesis. The newly developed device provides a basis for the development of NUMS derived rehabilitation technology to promote bone healing.
Collapse
Affiliation(s)
- Qiulin He
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Zhejiang University‐University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine Hangzhou China
| | - Junxin Lin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Zhejiang University‐University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine Hangzhou China
| | - Fanghao Zhou
- Center for X‐Mechanics, Department of Engineering Mechanics Zhejiang University Hangzhou China
| | - Dandan Cai
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Zhejiang University‐University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine Hangzhou China
| | - Yiyang Yan
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Zhejiang University‐University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine Hangzhou China
| | - Yejie Shan
- Center for X‐Mechanics, Department of Engineering Mechanics Zhejiang University Hangzhou China
| | - Shufang Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Zhejiang University‐University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine Hangzhou China
- China Orthopedic Regenerative Medicine Group (CORMed) Hangzhou China
| | - Tiefeng Li
- Center for X‐Mechanics, Department of Engineering Mechanics Zhejiang University Hangzhou China
| | - Xudong Yao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine Yiwu China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Department of Sports Medicine Zhejiang University School of Medicine Hangzhou China
- Zhejiang University‐University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine Hangzhou China
- China Orthopedic Regenerative Medicine Group (CORMed) Hangzhou China
| |
Collapse
|
28
|
Rho/ROCK Inhibition Promotes TGF- β3-Induced Tenogenic Differentiation in Mesenchymal Stromal Cells. Stem Cells Int 2021; 2021:8284690. [PMID: 34659420 PMCID: PMC8519677 DOI: 10.1155/2021/8284690] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/04/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stromal cells (MSC) represent a promising therapeutic tool for tendon regeneration. Their tenogenic differentiation is crucial for tissue engineering approaches and may support their beneficial effects after cell transplantation in vivo. The transforming growth factor (TGF)-β, signalling via intracellular Smad molecules, is a potent paracrine mediator of tenogenic induction. Moreover, scaffold topography or tendon matrix components induced tenogenesis via activation of the Rho/ROCK cascade, which, however, is also involved in pathological adaptations in extracellular matrix pathologies. The aim of this study was to investigate the interplay of Rho/ROCK and TGF-β3/Smad signalling in tenogenic differentiation in both human and equine MSC. Primary equine and human MSC isolated from adipose tissue were cultured as monolayers or on tendon-derived decellularized scaffolds to evaluate the influence of the ROCK inhibitor Y-27632 on TGF-β3-induced tenogenic differentiation. The MSC were incubated with and without TGF-β3 (10 ng/ml), Y-27632 (10 μM), or both. On day 1 and day 3, the signalling pathway of TGF-β and the actin cytoskeleton were visualized by Smad 2/3 and phalloidin staining, and gene expression of signalling molecules and tendon markers was assessed. ROCK inhibition was confirmed by disruption of the actin cytoskeleton. Activation of Smad 2/3 with nuclear translocation was evident upon TGF-β3 stimulation. Interestingly, this effect was most pronounced with additional ROCK inhibition in both species (p < 0.05 in equine MSC). In line with that, the tendon marker scleraxis showed the strongest upregulation when TGF-β3 and ROCK inhibition were combined (p < 0.05 in human MSC). The regulation pattern of tendon extracellular matrix components and the signalling molecules TGF-β3 and Smad 8 showed differences between human and equine MSC. The obtained results showed that ROCK inhibition promotes the TGF-β3/Smad 2/3 axis, with possible implications for future MSC priming regimes in tendon therapy.
Collapse
|
29
|
Zhang D, Zhang R, Song X, Yan KC, Liang H. Uniaxial Cyclic Stretching Promotes Chromatin Accessibility of Gene Loci Associated With Mesenchymal Stem Cells Morphogenesis and Osteogenesis. Front Cell Dev Biol 2021; 9:664545. [PMID: 34307349 PMCID: PMC8294092 DOI: 10.3389/fcell.2021.664545] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/28/2021] [Indexed: 01/08/2023] Open
Abstract
It has been previously demonstrated that uniaxial cyclic stretching (UCS) induces differentiation of mesenchymal stem cells (MSCs) into osteoblasts in vitro. It is also known that interactions between cells and external forces occur at various aspects including cell–matrix, cytoskeleton, nucleus membrane, and chromatin. However, changes in chromatin landscape during this process are still not clear. The present study was aimed to determine changes of chromatin accessibility under cyclic stretch. The influence of cyclic stretching on the morphology, proliferation, and differentiation of hMSCs was characterized. Changes of open chromatin sites were determined by assay for transposase accessible chromatin with high-throughput sequencing (ATAC-seq). Our results showed that UCS induced cell reorientation and actin stress fibers realignment, and in turn caused nuclear reorientation and deformation. Compared with unstrained group, the expression of osteogenic and chondrogenic marker genes were the highest in group of 1 Hz + 8% strain; this condition also led to lower cell proliferation rate. Furthermore, there were 2022 gene loci with upregulated chromatin accessibility in 1 Hz + 8% groups based on the analysis of chromatin accessibility. These genes are associated with regulation of cell morphogenesis, cell–substrate adhesion, and ossification. Signaling pathways involved in osteogenic differentiation were found in up-regulated GO biological processes. These findings demonstrated that UCS increased the openness of gene loci associated with regulation of cell morphogenesis and osteogenesis as well as the corresponding transcription activities. Moreover, the findings also connect the changes in chromatin accessibility with cell reorientation, nuclear reorientation, and deformation. Our study may provide reference for directed differentiation of stem cells induced by mechanical microenvironments.
Collapse
Affiliation(s)
- Duo Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Ran Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Xiaoyuan Song
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Karen Chang Yan
- Mechanical Engineering and Biomedical Engineering, The College of New Jersey, Ewing Township, NJ, United States
| | - Haiyi Liang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| |
Collapse
|
30
|
Sankar D, Mony U, Rangasamy J. Combinatorial effect of plasma treatment, fiber alignment and fiber scale of poly (ε-caprolactone)/collagen multiscale fibers in inducing tenogenesis in non-tenogenic media. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112206. [PMID: 34225858 DOI: 10.1016/j.msec.2021.112206] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022]
Abstract
Tendon being a hypocellular, low vascularized tissue often requires assistance for restoration after complete tear. Tendon tissue engineering aims in the development of suitable scaffold that could support the regeneration of tendon after damage. The success of such scaffolds is dependent on its integration with the native tissue which in turn is influenced by the cell-material interaction. In this work aligned poly(ε-caprolactone)/collagen (PCL/collagen) multiscale fibers were developed and plasma treatment using argon, nitrogen and its combination was accessed for inducing tenogenic differentiation in mesenchymal stem cells. The developed fibers mimicked tendon extracellular matrix (ECM) which upon plasma treatment maintained moderate hydrophilicity. Oxygen and nitrogen containing groups were observed to be incorporated after argon and nitrogen treatment respectively. Statistically significant (p < 0.001) enhancement was observed in average and root mean square (RMS) roughness after plasma treatment with the maximum in argon treated fibers. Vitronectin was competitively (statistically significant, p < 0.05) adsorbed after argon and combination treatment whereas nitrogen treatment led to the competitive adsorption of fibronectin (statistically significant, p < 0.05). Human mesenchymal stem cells (hMSCs) showed enhanced proliferation and attachment on plasma treated fibers. Increased porosity due to the presence of sacrificial collagen nanofibers improved cell infiltration which was further enhanced upon plasma treatment. RhoA activation was observed (statistically significant, p < 0.05) on aligned PCL/collagen multiscale fibers and PCL microfibers, which proved its impact on tenogenic differentiation. Further enhancement in rhoA expression was observed on argon (p < 0.01) and combination plasma (p < 0.05) treated fibers. Tenogenic differentiation of hMSCs was enhanced (statistically significant) on argon plasma treated aligned fibers which was confirmed by the expression of scleraxis, mohawk (early markers) and tenomodulin (late marker) at protein level and mohawk, collagen I, collagen III (early markers), thrombospondin 4 and tenascin C (late markers) at gene level. Thus argon plasma treatment on aligned fibers is an effective method to induce tenogenesis even in non-tenogenic media.
Collapse
Affiliation(s)
- Deepthi Sankar
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Ullas Mony
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India.
| | - Jayakumar Rangasamy
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India.
| |
Collapse
|
31
|
Wu S, Liu J, Qi Y, Cai J, Zhao J, Duan B, Chen S. Tendon-bioinspired wavy nanofibrous scaffolds provide tunable anisotropy and promote tenogenesis for tendon tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112181. [PMID: 34082981 DOI: 10.1016/j.msec.2021.112181] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/23/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023]
Abstract
The development of tendon-biomimetic nanofibrous scaffolds with mesenchymal stem cells may represent a promising strategy to improve the unsatisfactory outcomes of traditional treatments in tendon repair. In the present study, the nanofibrous scaffolds comprised of poly(p-dioxanone) (PPDO) and silk fibroin (SF) composites were fabricated by using electrospinning technique and subsequent thermal ethanol treatment. The PPDO/SF composite scaffolds presented parallel fiber arrangement with crimped features and nonlinear mechanical properties, which mimic the structure-function relationship of native tendon tissue mechanics. We demonstrated that the fiber crimp degree and mechanical properties of as-prepared PPDO/SF wavy nanofibrous scaffolds (WNSs) could be tunable by adjusting the mass ratio of PPDO/SF. The biological tests revealed that the addition of SF obviously promoted the cell adhesion, proliferation, and phenotypic maintenance of human tenocytes on the WNSs. A preliminary study on the subcutaneous implantation showed that the PPDO/SF WNSs notably decreased the inflammatory response compared with pure PPDO WNSs. More importantly, a combination of growth factor induction and mechanical stimulation was found to notably enhance the tenogenic differentiation of human adipose derived mesenchymal stem cells on the PPDO/SF WNSs by upregulating the expressions of tendon-associated protein and gene markers. Overall, this study demonstrated that our PPDO/SF WNSs could provide a beneficial microenvironment for various cell activities, making them an attractive candidate for tendon tissue engineering research.
Collapse
Affiliation(s)
- Shaohua Wu
- College of Textiles & Clothing, Qingdao University, Qingdao, China.
| | - Jiao Liu
- College of Textiles & Clothing, Qingdao University, Qingdao, China
| | - Ye Qi
- College of Textiles & Clothing, Qingdao University, Qingdao, China
| | - Jiangyu Cai
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Shaojuan Chen
- College of Textiles & Clothing, Qingdao University, Qingdao, China.
| |
Collapse
|
32
|
Yang L, Pijuan-Galito S, Rho HS, Vasilevich AS, Eren AD, Ge L, Habibović P, Alexander MR, de Boer J, Carlier A, van Rijn P, Zhou Q. High-Throughput Methods in the Discovery and Study of Biomaterials and Materiobiology. Chem Rev 2021; 121:4561-4677. [PMID: 33705116 PMCID: PMC8154331 DOI: 10.1021/acs.chemrev.0c00752] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 02/07/2023]
Abstract
The complex interaction of cells with biomaterials (i.e., materiobiology) plays an increasingly pivotal role in the development of novel implants, biomedical devices, and tissue engineering scaffolds to treat diseases, aid in the restoration of bodily functions, construct healthy tissues, or regenerate diseased ones. However, the conventional approaches are incapable of screening the huge amount of potential material parameter combinations to identify the optimal cell responses and involve a combination of serendipity and many series of trial-and-error experiments. For advanced tissue engineering and regenerative medicine, highly efficient and complex bioanalysis platforms are expected to explore the complex interaction of cells with biomaterials using combinatorial approaches that offer desired complex microenvironments during healing, development, and homeostasis. In this review, we first introduce materiobiology and its high-throughput screening (HTS). Then we present an in-depth of the recent progress of 2D/3D HTS platforms (i.e., gradient and microarray) in the principle, preparation, screening for materiobiology, and combination with other advanced technologies. The Compendium for Biomaterial Transcriptomics and high content imaging, computational simulations, and their translation toward commercial and clinical uses are highlighted. In the final section, current challenges and future perspectives are discussed. High-throughput experimentation within the field of materiobiology enables the elucidation of the relationships between biomaterial properties and biological behavior and thereby serves as a potential tool for accelerating the development of high-performance biomaterials.
Collapse
Affiliation(s)
- Liangliang Yang
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sara Pijuan-Galito
- School
of Pharmacy, Biodiscovery Institute, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Hoon Suk Rho
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Aliaksei S. Vasilevich
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aysegul Dede Eren
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lu Ge
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Pamela Habibović
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Morgan R. Alexander
- School
of Pharmacy, Boots Science Building, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Jan de Boer
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aurélie Carlier
- Department
of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick van Rijn
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Qihui Zhou
- Institute
for Translational Medicine, Department of Stomatology, The Affiliated
Hospital of Qingdao University, Qingdao
University, Qingdao 266003, China
| |
Collapse
|
33
|
Vermeulen S, Roumans N, Honig F, Carlier A, Hebels DG, Eren AD, Dijke PT, Vasilevich A, de Boer J. Mechanotransduction is a context-dependent activator of TGF-β signaling in mesenchymal stem cells. Biomaterials 2020; 259:120331. [DOI: 10.1016/j.biomaterials.2020.120331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/15/2020] [Accepted: 08/13/2020] [Indexed: 02/08/2023]
|
34
|
Wang HN, Huang YC, Ni GX. Mechanotransduction of stem cells for tendon repair. World J Stem Cells 2020; 12:952-965. [PMID: 33033557 PMCID: PMC7524696 DOI: 10.4252/wjsc.v12.i9.952] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/06/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Tendon is a mechanosensitive tissue that transmits force from muscle to bone. Physiological loading contributes to maintaining the homeostasis and adaptation of tendon, but aberrant loading may lead to injury or failed repair. It is shown that stem cells respond to mechanical loading and play an essential role in both acute and chronic injuries, as well as in tendon repair. In the process of mechanotransduction, mechanical loading is detected by mechanosensors that regulate cell differentiation and proliferation via several signaling pathways. In order to better understand the stem-cell response to mechanical stimulation and the potential mechanism of the tendon repair process, in this review, we summarize the source and role of endogenous and exogenous stem cells active in tendon repair, describe the mechanical response of stem cells, and finally, highlight the mechanotransduction process and underlying signaling pathways.
Collapse
Affiliation(s)
- Hao-Nan Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Yong-Can Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Guo-Xin Ni
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China.
| |
Collapse
|
35
|
The Application of Mechanical Stimulations in Tendon Tissue Engineering. Stem Cells Int 2020; 2020:8824783. [PMID: 33029149 PMCID: PMC7532391 DOI: 10.1155/2020/8824783] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 12/18/2022] Open
Abstract
Tendon injury is the most common disease in the musculoskeletal system. The current treatment methods have many limitations, such as poor therapeutic effects, functional loss of donor site, and immune rejection. Tendon tissue engineering provides a new treatment strategy for tendon repair and regeneration. In this review, we made a retrospective analysis of applying mechanical stimulation in tendon tissue engineering, and its potential as a direction of development for future clinical treatment strategies. For this purpose, the following topics are discussed; (1) the context of tendon tissue engineering and mechanical stimulation; (2) the applications of various mechanical stimulations in tendon tissue engineering, as well as their inherent mechanisms; (3) the application of magnetic force and the synergy of mechanical and biochemical stimulation. With this, we aim at clarifying some of the main questions that currently exist in the field of tendon tissue engineering and consequently gain new knowledge that may help in the development of future clinical application of tissue engineering in tendon injury.
Collapse
|
36
|
Chu J, Lu M, Pfeifer CG, Alt V, Docheva D. Rebuilding Tendons: A Concise Review on the Potential of Dermal Fibroblasts. Cells 2020; 9:E2047. [PMID: 32911760 PMCID: PMC7563185 DOI: 10.3390/cells9092047] [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: 06/29/2020] [Revised: 08/26/2020] [Accepted: 09/02/2020] [Indexed: 12/26/2022] Open
Abstract
Tendons are vital to joint movement by connecting muscles to bones. Along with an increasing incidence of tendon injuries, tendon disorders can burden the quality of life of patients or the career of athletes. Current treatments involve surgical reconstruction and conservative therapy. Especially in the elderly population, tendon recovery requires lengthy periods and it may result in unsatisfactory outcome. Cell-mediated tendon engineering is a rapidly progressing experimental and pre-clinical field, which holds great potential for an alternative approach to established medical treatments. The selection of an appropriate cell source is critical and remains under investigation. Dermal fibroblasts exhibit multiple similarities to tendon cells, suggesting they may be a promising cell source for tendon engineering. Hence, the purpose of this review article was in brief, to compare tendon to dermis tissues, and summarize in vitro studies on tenogenic differentiation of dermal fibroblasts. Furthermore, analysis of an open source Gene Expression Omnibus (GEO) data repository was carried out, revealing great overlap in the molecular profiles of both cell types. Lastly, a summary of in vivo studies employing dermal fibroblasts in tendon repair as well as pilot clinical studies in this area is included. Altogether, dermal fibroblasts hold therapeutic potential and are attractive cells for rebuilding injured tendons.
Collapse
Affiliation(s)
- Jin Chu
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, 93053 Regensburg, Germany; (J.C.); (C.G.P.); (V.A.)
| | - Ming Lu
- Department of Orthopaedic Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116023, China;
| | - Christian G. Pfeifer
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, 93053 Regensburg, Germany; (J.C.); (C.G.P.); (V.A.)
- Department of Trauma Surgery, University Regensburg Medical Centre, 93053 Regensburg, Germany
| | - Volker Alt
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, 93053 Regensburg, Germany; (J.C.); (C.G.P.); (V.A.)
- Department of Trauma Surgery, University Regensburg Medical Centre, 93053 Regensburg, Germany
| | - Denitsa Docheva
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, 93053 Regensburg, Germany; (J.C.); (C.G.P.); (V.A.)
| |
Collapse
|
37
|
Liu R, Zhang S, Chen X. Injectable hydrogels for tendon and ligament tissue engineering. J Tissue Eng Regen Med 2020; 14:1333-1348. [PMID: 32495524 DOI: 10.1002/term.3078] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/06/2020] [Accepted: 05/17/2020] [Indexed: 01/14/2023]
Abstract
The problem of tendon and ligament (T/L) regeneration in musculoskeletal diseases has long constituted a major challenge. In situ injection of formable biodegradable hydrogels, however, has been demonstrated to treat T/L injury and reduce patient suffering in a minimally invasive manner. An injectable hydrogel is more suitable than other biological materials due to the special physiological structure of T/L. Most other materials utilized to repair T/L are cell-based, growth factor-based materials, with few material properties. In addition, the mechanical property of the gel cannot reach the normal T/L level. This review summarizes advances in natural and synthetic polymeric injectable hydrogels for tissue engineering in T/L and presents prospects for injectable and biodegradable hydrogels for its treatment. In future T/L applications, it is necessary develop an injectable hydrogel with mechanics, tissue damage-specific binding, and disease response. Simultaneously, the advantages of various biological materials must be combined in order to achieve personalized precision therapy.
Collapse
Affiliation(s)
- Richun Liu
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Shichen Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao Chen
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
38
|
Chen J, Backman LJ, Zhang W, Ling C, Danielson P. Regulation of Keratocyte Phenotype and Cell Behavior by Substrate Stiffness. ACS Biomater Sci Eng 2020; 6:5162-5171. [PMID: 33455266 DOI: 10.1021/acsbiomaterials.0c00510] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Corneal tissue engineering is an alternative way to solve the problem of lack of corneal donor tissue in corneal transplantation. Keratocytes with a normal phenotype and function in tissue-engineered cornea would be critical for corneal regeneration. Although the role of extracellular/substrate material stiffness is well-known for the regulation of the cell phenotype and cell behavior in many different cell types, its effects in keratocyte culture have not yet been thoroughly studied. This project studied the effect of substrate stiffness on the keratocyte phenotype marker expression and typical cell behavior (cell adhesion, proliferation, and migration), and the possible mechanisms involved. Human primary keratocytes were cultured on tissue culture plastic (TCP, ∼106 kPa) or on plates with the stiffness equivalent of physiological human corneal stroma (25 kPa) or vitreous body (1 kPa). The expression of keratocyte phenotype markers, cell adhesion, proliferation, and migration were compared. The results showed that the stiffness of the substrate material regulates the phenotype marker expression and cell behavior of cultured keratocytes. Physiological corneal stiffness (25 kPa) superiorly preserved the cell phenotype when compared to the TCP and 1 kPa group. Keratocytes had a larger cell area when cultured on 25 kPa plates as compared to on TCP. Treatment of cells with NSC 23766 (Rac1 inhibitor) mimicked the response in the cell phenotype and behavior seen in the transition from soft materials to stiff materials, including the cytoskeletal structure, expression of keratocyte phenotype markers, and cell behavior. In conclusion, this study shows that substrate stiffness regulates the cell phenotype marker expression and cell behavior of keratocytes by Rac1-mediated cytoskeletal reorganization. This knowledge contributes to the development of corneal tissue engineering.
Collapse
Affiliation(s)
- Jialin Chen
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China.,Department of Integrative Medical Biology, Anatomy, Umeå University, Umeå SE-901 87, Sweden.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210009, China
| | - Ludvig J Backman
- Department of Integrative Medical Biology, Anatomy, Umeå University, Umeå SE-901 87, Sweden.,Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, Umeå SE-901 87, Sweden
| | - Wei Zhang
- Department of Integrative Medical Biology, Anatomy, Umeå University, Umeå SE-901 87, Sweden.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210009, China.,Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chen Ling
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Patrik Danielson
- Department of Integrative Medical Biology, Anatomy, Umeå University, Umeå SE-901 87, Sweden.,Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå SE-901 87, Sweden
| |
Collapse
|
39
|
Mat Afandi MA, Maarof M, Chowdhury SR, Bt Hj Idrus R. Synergistic Effect of Laminin and Epidermal Growth Factor on Biological and Morphological Properties of Co-Cultured Myoblasts and Fibroblasts. Tissue Eng Regen Med 2020; 17:835-845. [PMID: 32767029 DOI: 10.1007/s13770-020-00283-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/02/2020] [Accepted: 07/05/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND One of the long-standing problems of myoblasts in vitro expansion is slow cell migration and this causes fibroblast population to exceed myoblasts. In this study, we investigated the synergistic effect of laminin and epidermal growth factor (EGF) on co-cultured myoblasts and fibroblasts for cell attachment, proliferation and migration. METHODS Skeletal human muscle cells were cultured in four different conditions; control, EGF, laminin (Lam) and laminin EGF (Lam + EGF). Using live imaging system, their cellular properties; attachment, migration and growth were exposed to Rho kinase inhibitor, Y-27632, and EGF-receptor (EGF-R) inhibitor, gefitinib were measured. RESULTS Myoblast migration and proliferation was enhanced significantly by synergistic stimulation of laminin and EGF (0.61 ± 0.14 µm/min, 0.008 ± 0.001 h-1) compare to that by EGF alone (0.26 ± 0.13 µm/min, 0.004 ± 0.0009 h-1). However, no changes in proliferation and migration were observed for fibroblasts among the culture conditions. Inhibition of Rho kinase resulted in the increase of the myoblast migration on the laminin-coated surface with EGF condition (0.64 ± 0.18 µm/min). Compared to the untreated conditions, myoblasts cultured on the laminin-coated surface and EGF demonstrated elongated morphology, and average cell length increase significantly. In contrast, inhibition of EGF-R resulted in the decrease of myoblast migration on the laminin coated surface with EGF supplemented condition (0.43 ± 0.05 µm/min) in comparison to the untreated control (0.53 ± 0.05 µm/min). CONCLUSION Laminin and EGF preferentially enhance the proliferation and migration of myoblasts, and Rho kinase and EGF-R play a role in this synergistic effect. These results will be beneficial for the propagation of skeletal muscle cells for clinical applications.
Collapse
Affiliation(s)
- Mohd Asyraf Mat Afandi
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Manira Maarof
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia
| | - S R Chowdhury
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Ruszymah Bt Hj Idrus
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia.
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
40
|
Selig M, Lauer JC, Hart ML, Rolauffs B. Mechanotransduction and Stiffness-Sensing: Mechanisms and Opportunities to Control Multiple Molecular Aspects of Cell Phenotype as a Design Cornerstone of Cell-Instructive Biomaterials for Articular Cartilage Repair. Int J Mol Sci 2020; 21:E5399. [PMID: 32751354 PMCID: PMC7432012 DOI: 10.3390/ijms21155399] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/23/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Since material stiffness controls many cell functions, we reviewed the currently available knowledge on stiffness sensing and elucidated what is known in the context of clinical and experimental articular cartilage (AC) repair. Remarkably, no stiffness information on the various biomaterials for clinical AC repair was accessible. Using mRNA expression profiles and morphology as surrogate markers of stiffness-related effects, we deduced that the various clinically available biomaterials control chondrocyte (CH) phenotype well, but not to equal extents, and only in non-degenerative settings. Ample evidence demonstrates that multiple molecular aspects of CH and mesenchymal stromal cell (MSC) phenotype are susceptible to material stiffness, because proliferation, migration, lineage determination, shape, cytoskeletal properties, expression profiles, cell surface receptor composition, integrin subunit expression, and nuclear shape and composition of CHs and/or MSCs are stiffness-regulated. Moreover, material stiffness modulates MSC immuno-modulatory and angiogenic properties, transforming growth factor beta 1 (TGF-β1)-induced lineage determination, and CH re-differentiation/de-differentiation, collagen type II fragment production, and TGF-β1- and interleukin 1 beta (IL-1β)-induced changes in cell stiffness and traction force. We then integrated the available molecular signaling data into a stiffness-regulated CH phenotype model. Overall, we recommend using material stiffness for controlling cell phenotype, as this would be a promising design cornerstone for novel future-oriented, cell-instructive biomaterials for clinical high-quality AC repair tissue.
Collapse
Affiliation(s)
- Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Jasmin C. Lauer
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Melanie L. Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| |
Collapse
|
41
|
Jaiswal D, Yousman L, Neary M, Fernschild E, Zolnoski B, Katebifar S, Rudraiah S, Mazzocca AD, Kumbar SG. Tendon tissue engineering: biomechanical considerations. Biomed Mater 2020; 15:052001. [DOI: 10.1088/1748-605x/ab852f] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
42
|
Tendon and ligament mechanical loading in the pathogenesis of inflammatory arthritis. Nat Rev Rheumatol 2020; 16:193-207. [PMID: 32080619 DOI: 10.1038/s41584-019-0364-x] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Abstract
Mechanical loading is an important factor in musculoskeletal health and disease. Tendons and ligaments require physiological levels of mechanical loading to develop and maintain their tissue architecture, a process that is achieved at the cellular level through mechanotransduction-mediated fine tuning of the extracellular matrix by tendon and ligament stromal cells. Pathological levels of force represent a biological (mechanical) stress that elicits an immune system-mediated tissue repair pathway in tendons and ligaments. The biomechanics and mechanobiology of tendons and ligaments form the basis for understanding how such tissues sense and respond to mechanical force, and the anatomical extent of several mechanical stress-related disorders in tendons and ligaments overlaps with that of chronic inflammatory arthritis in joints. The role of mechanical stress in 'overuse' injuries, such as tendinopathy, has long been known, but mechanical stress is now also emerging as a possible trigger for some forms of chronic inflammatory arthritis, including spondyloarthritis and rheumatoid arthritis. Thus, seemingly diverse diseases of the musculoskeletal system might have similar mechanisms of immunopathogenesis owing to conserved responses to mechanical stress.
Collapse
|
43
|
Su X, Wang J, Kang H, Bao G, Liu L. Effects of dynamic radial tensile stress on fibrocartilage differentiation of bone marrow mesenchymal stem cells. Biomed Eng Online 2020; 19:8. [PMID: 32024525 PMCID: PMC7003351 DOI: 10.1186/s12938-020-0751-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/24/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Uniaxial/biaxial tensile stress has been employed to induce chondrocyte differentiation of mesenchymal stem cells. However, the effects of radial tensile stimuli on differentiation of MSCs into fibrocartilage remain unclear. RESULTS It was found that induced bone marrow mesenchymal stem cells (BMSCs) were not only similar to TMJ disc cells in morphology, but also could synthesize type I collagen (Col I), a small amount of type II collagen (Col II) and glycosaminoglycans (GAGs). The synthesis of Col I significantly increased while that of Col II gradually decreased with increasing tensile strength. The ratio of Col I to Col II was 1.8 to 1 and 2 to 1 in the 10% and 15% stretching groups, respectively. The gene expression of Col I and GAGs was significantly upregulated, whereas that of Col II was downregulated. However, the higher tensile stimulation (15%) promoted the synthesis of α-smooth muscle actin (α-SMA). Too much α-SMA is not conducive to constructing engineered tissue. CONCLUSION Therefore, the 10% radial tensile stimulus was the optimal strength for inducing the BMSCs to differentiate into fibrochondrocytes of the temporomandibular joint (TMJ) disc. This work provided a novel approach for inducing BMSCs to differentiate into fibrochondrocytes.
Collapse
Affiliation(s)
- Xuelian Su
- College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.,Key Lab of Oral Diseases of Gansu Province, Northwest Minzu University, Lanzhou, People's Republic of China.,Key Lab of Stomatology of State Ethnic Affairs Commission, Northwest Minzu University, Lanzhou, People's Republic of China.,Key Laboratory of Mechanics on Disaster and Environment in Western China, The Ministry of Education of China, Lanzhou University, Lanzhou, People's Republic of China.,Department of Prosthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| | - Jizeng Wang
- College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China. .,Key Laboratory of Mechanics on Disaster and Environment in Western China, The Ministry of Education of China, Lanzhou University, Lanzhou, People's Republic of China.
| | - Hong Kang
- College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.,Key Laboratory of Mechanics on Disaster and Environment in Western China, The Ministry of Education of China, Lanzhou University, Lanzhou, People's Republic of China
| | - Guangjie Bao
- Key Lab of Oral Diseases of Gansu Province, Northwest Minzu University, Lanzhou, People's Republic of China.,Key Lab of Stomatology of State Ethnic Affairs Commission, Northwest Minzu University, Lanzhou, People's Republic of China.,Department of Prosthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| | - Lin Liu
- Key Lab of Oral Diseases of Gansu Province, Northwest Minzu University, Lanzhou, People's Republic of China.,Key Lab of Stomatology of State Ethnic Affairs Commission, Northwest Minzu University, Lanzhou, People's Republic of China.,Department of Prosthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
44
|
Katoh K. FAK-Dependent Cell Motility and Cell Elongation. Cells 2020; 9:cells9010192. [PMID: 31940873 PMCID: PMC7017285 DOI: 10.3390/cells9010192] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/02/2020] [Accepted: 01/08/2020] [Indexed: 12/20/2022] Open
Abstract
Fibroblastic cells show specific substrate selectivity for typical cell–substrate adhesion. However, focal adhesion kinase (FAK) contributes to controlling the regulation of orientation and polarity. When fibroblasts attach to micropatterns, tyrosine-phosphorylated proteins and FAK are both detected along the inner border between the adhesive micropatterns and the nonadhesive glass surface. FAK likely plays important roles in regulation of cell adhesion to the substrate, as FAK is a tyrosine-phosphorylated protein that acts as a signal transduction molecule at sites of cell–substrate attachment, called focal adhesions. FAK has been suggested to play a role in the attachment of cells at adhesive micropatterns by affecting cell polarity. Therefore, the localization of FAK might play a key role in recognition of the border of the cell with the adhesive micropattern, thus regulating cell polarity and the cell axis. This review discusses the regulation and molecular mechanism of cell proliferation and cell elongation by FAK and its associated signal transduction proteins.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology Tsukuba-city, Ibaraki, Japan
| |
Collapse
|
45
|
Monemian Esfahani A, Rosenbohm J, Reddy K, Jin X, Bouzid T, Riehl B, Kim E, Lim JY, Yang R. Tissue Regeneration from Mechanical Stretching of Cell-Cell Adhesion. Tissue Eng Part C Methods 2019; 25:631-640. [PMID: 31407627 PMCID: PMC6859692 DOI: 10.1089/ten.tec.2019.0098] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/05/2019] [Indexed: 01/09/2023] Open
Abstract
Cell-cell adhesion complexes are macromolecular adhesive organelles that integrate cells into tissues. This mechanochemical coupling in cell-cell adhesion is required for a large number of cell behaviors, and perturbations of the cell-cell adhesion structure or related mechanotransduction pathways can lead to critical pathological conditions such as skin and heart diseases, arthritis, and cancer. Mechanical stretching has been a widely used method to stimulate the mechanotransduction process originating from the cell-cell adhesion and cell-extracellular matrix (ECM) complexes. These studies aimed to reveal the biophysical processes governing cell proliferation, wound healing, gene expression regulation, and cell differentiation in various tissues, including cardiac, muscle, vascular, and bone. This review explores techniques in mechanical stretching in two-dimensional settings with different stretching regimens on different cell types. The mechanotransduction responses from these different cell types will be discussed with an emphasis on their biophysical transformations during mechanical stretching and the cross talk between the cell-cell and cell-ECM adhesion complexes. Therapeutic aspects of mechanical stretching are reviewed considering these cellular responses after the application of mechanical forces, with a focus on wound healing and tissue regeneration. Impact Statement Mechanical stretching has been proposed as a therapeutic option for tissue regeneration and wound healing. It has been accepted that mechanotransduction processes elicited by mechanical stretching govern cellular response and behavior, and these studies have predominantly focused on the cell-extracellular matrix (ECM) sites. This review serves the mechanobiology community by shifting the focus of mechanical stretching effects from cell-ECM adhesions to the less examined cell-cell adhesions, which we believe play an equally important role in orchestrating the response pathways.
Collapse
Affiliation(s)
- Amir Monemian Esfahani
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Jordan Rosenbohm
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Keerthana Reddy
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Xiaowei Jin
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Tasneem Bouzid
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Brandon Riehl
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Eunju Kim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska
| |
Collapse
|
46
|
Kim JY, Jun JH, Park SY, Yang SW, Bae SH, Kim GJ. Dynamic Regulation of miRNA Expression by Functionally Enhanced Placental Mesenchymal Stem Cells PromotesHepatic Regeneration in a Rat Model with Bile Duct Ligation. Int J Mol Sci 2019; 20:ijms20215299. [PMID: 31653075 PMCID: PMC6862171 DOI: 10.3390/ijms20215299] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Placenta-derived mesenchymal stem cells (PD-MSCs) were highlighted as therapeutic sources in several degenerative diseases. Recently, microRNAs (miRNAs)were found to mediate one of the therapeutic mechanisms of PD-MSCs in regenerative medicine. To enhance the therapeutic effects of PD-MSCs, we established functionally enhanced PD-MSCs with phosphatase of regenerating liver-1 overexpression (PRL-1(+)). However, the profile and functions of miRNAs induced by PRL-1(+) PD-MSCs in a rat model with hepatic failure prepared by bile duct ligation (BDL) remained unclear. Hence, the objectives of the present study were to analyze the expression of miRNAs and investigate their therapeutic mechanisms for hepatic regeneration via PRL-1(+) in a rat model with BDL. We selected candidate miRNAs based on microarray analysis. Under hypoxic conditions, compared with migrated naïve PD-MSCs, migrated PRL-1(+) PD-MSCs showed improved integrin-dependent migration abilitythrough Ras homolog (RHO) family-targeted miRNA expression (e.g., hsa-miR-30a-5p, 340-5p, and 146a-3p). Moreover, rno-miR-30a-5p and 340-5p regulated engraftment into injured rat liver by transplantedPRL-1(+) PD-MSCs through the integrin family. Additionally, an increase inplatelet-derived growth factor receptor A (PDGFRA) by suppressing rno-miR-27a-3p improved vascular structure in rat liver tissues after PRL-1(+) PD-MSC transplantation. Furthermore, decreased rno-miR-122-5p was significantly correlated with increased proliferation of hepatocytes in liver tissues by PRL-1(+) PD-MSCs byactivating the interleukin-6 (IL-6) signaling pathway through the repression of rno-miR-21-5p. Taken together, these findings improve the understandingof therapeutic mechanisms based on miRNA-mediated stem-cell therapy in liver diseases.
Collapse
Affiliation(s)
- Jae Yeon Kim
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea.
| | - Ji Hye Jun
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea.
| | - Soo Young Park
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea.
| | - Seong Wook Yang
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120749, Korea.
| | - Si Hyun Bae
- Department of Internal Medicine, Catholic University Medical College, Seoul 03312, Korea.
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea.
| |
Collapse
|
47
|
Chang W, Callan KT, Dragoo JL. The Behavior of Tendon Progenitor Cells from Tendinopathic Tendons: Implications for Treatment. Tissue Eng Part A 2019; 26:38-46. [PMID: 31111771 DOI: 10.1089/ten.tea.2019.0042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tendinopathy remains a significant clinical challenge. Although there is some evidence that leukocyte-rich platelet-rich plasma can improve the symptoms of tendinopathy, more efficacious treatments will be required in the future to improve probability of successfully resolving this condition in athletes. Because optimal treatments are not currently available, there is a need to better understand the pathology of tendinopathy from the perspective of tendon progenitor cells (TPCs). TPCs isolated from normal and tendinopathy donors were characterized by their stem cell properties and proliferation capacities, along with their ability to become tenocytes under mechanical loading. The results showed a significant 2.6-fold increase in the viable cell population in tendinopathy versus normal donors. Although the percentage of self-renewing cells was similar, the total number of TPCs in tendinopathy was significantly higher (1.6-fold) than normal TPCs based on the colony formation assays. In contrast, TPCs from tendinopathy tissue showed significantly lower cellular proliferation rate by cumulative population doublings. Next, the expanded TPCs from both tissues successfully demonstrated the trilineage differentiation capabilities with specific gene markers, staining, and biochemical assays. To induce tenogenic differentiation, stretchable silicone wells were designed and fabricated, plus the creation of an adaptor platform used on a syringe pump for mechanical stretch. This economic design provided the adequate cyclic loading to drive tenogenic differentiation. With these devices, the stretch duration was optimized and showed the significant increase in scleraxis (SCX) and tenomodulin (TNMD) expression at 2.60 (fold change) and 3.86 (fold change in logarithm), respectively, by reverse transcription-quantitative polymerase chain reaction in normal TPCs after stretch. This assay also demonstrated the widespread cell reorientation following stretch in normal TPCs. In contrast, the mechanical loading did not increase the SCX gene expression; TNMD expression remained undetectable, and cell realignment was significantly less in tendinopathy TPCs. In addition, western blot analysis confirmed the elevated TNMD protein expression in normal TPCs following stretch and the lack of expression in tendinopathy TPCs. In summary, tendinopathy TPCs were unable to differentiate into tenocytes following mechanical stretch. Future studies may aim to reprogram tendinopathy TPCs to allow tenogenic induction. Impact Statement This article presents a model to distinguish between normal and tendinopathy progenitor cell behavior, which reveals insight into the pathophysiology of tendinopathy. With the design of a platform adaptor, mechanical stretch was applied to tendon progenitor cells (TPCs) that promoted tenogenic differentiation. This design provided programmable features for more flexible application with low cost. These devices successfully stimulated tenogenic differentiation of TPCs from normal, but not tendinopathic tendons under cyclic stretch. The scientific method provided in this article will allow testing of biologics, exosomes, and other treatment strategies to derive new, more efficient treatment of tendinopathy in the future.
Collapse
Affiliation(s)
- Wenteh Chang
- Department of Orthopaedic Surgery, Stanford University, Redwood City, California
| | - Kylie T Callan
- Department of Orthopaedic Surgery, Stanford University, Redwood City, California
| | - Jason L Dragoo
- Department of Orthopaedic Surgery, Stanford University, Redwood City, California
| |
Collapse
|
48
|
Necula MG, Mazare A, Ion RN, Ozkan S, Park J, Schmuki P, Cimpean A. Lateral Spacing of TiO 2 Nanotubes Modulates Osteoblast Behavior. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2956. [PMID: 31547276 PMCID: PMC6766216 DOI: 10.3390/ma12182956] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 01/06/2023]
Abstract
Titanium dioxide (TiO2) nanotube coated substrates have revolutionized the concept of implant in a number of ways, being endowed with superior osseointegration properties and local drug delivery capacity. While accumulating reports describe the influence of nanotube diameter on cell behavior, little is known about the effects of nanotube lateral spacing on cells involved in bone regeneration. In this context, in the present study the MC3T3-E1 murine pre-osteoblast cells behavior has been investigated by using TiO2 nanotubes of ~78 nm diameter and lateral spacing of 18 nm and 80 nm, respectively. Both nanostructured surfaces supported cell viability and proliferation in approximately equal extent. However, obvious differences in the cell spreading areas, morphologies, the organization of the actin cytoskeleton and the pattern of the focal adhesions were noticed. Furthermore, investigation of the pre-osteoblast differentiation potential indicated a higher capacity of larger spacing nanostructure to enhance the expression of the alkaline phosphatase, osteopontin and osteocalcin osteoblast specific markers inducing osteogenic differentiation. These findings provide the proof that lateral spacing of the TiO2 nanotube coated titanium (Ti) surfaces has to be considered in designing bone implants with improved biological performance.
Collapse
Affiliation(s)
- Madalina Georgiana Necula
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania.
| | - Anca Mazare
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany.
| | - Raluca Nicoleta Ion
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania.
| | - Selda Ozkan
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany.
| | - Jung Park
- Division of Molecular Pediatrics, Department of Pediatrics, University Hospital Erlangen, 91054 Erlangen, Germany.
| | - Patrik Schmuki
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany.
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania.
| |
Collapse
|
49
|
Fu X, Liu G, Halim A, Ju Y, Luo Q, Song AG. Mesenchymal Stem Cell Migration and Tissue Repair. Cells 2019; 8:E784. [PMID: 31357692 PMCID: PMC6721499 DOI: 10.3390/cells8080784] [Citation(s) in RCA: 605] [Impact Index Per Article: 100.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/13/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multilineage cells with the ability to self-renew and differentiate into a variety of cell types, which play key roles in tissue healing and regenerative medicine. Bone marrow-derived mesenchymal stem cells (BMSCs) are the most frequently used stem cells in cell therapy and tissue engineering. However, it is prerequisite for BMSCs to mobilize from bone marrow and migrate into injured tissues during the healing process, through peripheral circulation. The migration of BMSCs is regulated by mechanical and chemical factors in this trafficking process. In this paper, we review the effects of several main regulatory factors on BMSC migration and its underlying mechanism; discuss two critical roles of BMSCs-namely, directed differentiation and the paracrine function-in tissue repair; and provide insight into the relationship between BMSC migration and tissue repair, which may provide a better guide for clinical applications in tissue repair through the efficient regulation of BMSC migration.
Collapse
Affiliation(s)
- Xiaorong Fu
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China
| | - Ge Liu
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China
| | - Alexander Halim
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China
| | - Yang Ju
- Department of Mechanical Science and Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Qing Luo
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China
| | - And Guanbin Song
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China.
| |
Collapse
|
50
|
Wang W, Xu X, Li Z, Kratz K, Ma N, Lendlein A. Modulating human mesenchymal stem cells using poly(n-butyl acrylate) networks in vitro with elasticity matching human arteries. Clin Hemorheol Microcirc 2019; 71:277-289. [PMID: 30530970 DOI: 10.3233/ch-189418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Non-swelling hydrophobic poly(n-butyl acrylate) network (cPnBA) is a candidate material for synthetic vascular grafts owing to its low toxicity and tailorable mechanical properties. Mesenchymal stem cells (MSCs) are an attractive cell type for accelerating endothelialization because of their superior anti-thrombosis and immune modulatory function. Further, they can differentiate into smooth muscle cells or endothelial-like cells and secret pro-angiogenic factors such as vascular endothelial growth factor (VEGF). MSCs are sensitive to the substrate mechanical properties, with the alteration of their major cellular behavior and functions as a response to substrate elasticity. Here, we cultured human adipose-derived mesenchymal stem cells (hADSCs) on cPnBAs with different mechanical properties (cPnBA250, Young's modulus (E) = 250 kPa; cPnBA1100, E = 1100 kPa) matching the elasticity of native arteries, and investigated their cellular response to the materials including cell attachment, proliferation, viability, apoptosis, senescence and secretion. The cPnBA allowed high cell attachment and showed negligible cytotoxicity. F-actin assembly of hADSCs decreased on cPnBA films compared to classical tissue culture plate. The difference of cPnBA elasticity did not show dramatic effects on cell attachment, morphology, cytoskeleton assembly, apoptosis and senescence. Cells on cPnBA250, with lower proliferation rate, had significantly higher VEGF secretion activity. These results demonstrated that tuning polymer elasticity to regulate human stem cells might be a potential strategy for constructing stem cell-based artificial blood vessels.
Collapse
Affiliation(s)
- Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Zhengdong Li
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Helmholtz Virtual Institute "Multifunctional Biomaterials for Medicine", Teltow, Germany
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Helmholtz Virtual Institute "Multifunctional Biomaterials for Medicine", Teltow, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Helmholtz Virtual Institute "Multifunctional Biomaterials for Medicine", Teltow, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| |
Collapse
|