1
|
Li Y, Yang Y, Wang X, Li L, Zhou M. Extracellular osmolarity regulates osteoblast migration through the TRPV4-Rho/ROCK signaling. Commun Biol 2025; 8:515. [PMID: 40155775 PMCID: PMC11953337 DOI: 10.1038/s42003-025-07946-8] [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: 09/26/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
For precise bone formation, osteoblasts need to accurately migrate to specific sites guided by various biochemical and mechanical cues. During this migration, fluctuations in extracellular osmolarity may arise from shifts in the surrounding fluid environment. However, as a main regulator of cell morphology and function, whether the extracellular osmolarity change may affect osteoblast migration remains unclear. Here, we provide evidence showing that changes in extracellular osmolarity significantly impact osteoblast migration, with a hypotonic environment enhancing it while a hypertonic environment inhibiting it. Further, our findings reveal that a hypotonic treatment increases intracellular pressure, activating the Transient Receptor Potential Vanilloid 4 (TRPV4) channel. This activation of TRPV4 modulates stress fibers, focal adhesions (FAs), and cell polarity through the Rho/ROCK signaling pathway, ultimately impacting osteoblast migration. Our findings provide valuable insights into the significant influence of extracellular osmolarity on osteoblast migration, which has potential implications for enhancing our understanding of bone remodeling.
Collapse
Affiliation(s)
- Yijie Li
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China
| | - Yanyan Yang
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China
| | - Xiaohuan Wang
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China.
| | - Long Li
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Mouwang Zhou
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, China.
| |
Collapse
|
2
|
Li T, Zhang X, Ma L, Qi X, Wang H, Zhou Q, Sun X, Wang F, Zhao L, Shi W. 3D printing of stiff, tough, and ROS-scavenging nanocomposite hydrogel scaffold for in situ corneal repair. Acta Biomater 2025; 192:189-205. [PMID: 39643222 DOI: 10.1016/j.actbio.2024.12.005] [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: 07/31/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Despite significant advancements in hydrogels in recent years, their application in corneal repair remains limited by several challenges, including unfitted curvatures, inferior mechanical properties, and insufficient reactive oxygen species (ROS)-scavenging activities. To address these issues, this study introduces a 3D-printed corneal scaffold with nanocomposite hydrogel consisting of gelatin methacrylate (GelMA), poly (ethylene glycol) diacrylate (PEGDA), Laponite, and dopamine. GelMA and PEGDA act as matrix materials with photo-crosslinking abilities. As a two-dimensional nanoclay, Laponite enhances the rheological properties of the hydrogel, making it suitable for 3D printing. Dopamine self-polymerizes into polydopamine (PDA), providing the hydrogel with ROS-scavenging activity. The incorporation of Laponite and the synergistic effect of PDA endow the hydrogel with good mechanical properties. In vitro investigations demonstrated the cytocompatibility of GelMA-PEGDA-Laponite-dopamine (GPLD) hydrogel and its ROS-scavenging activity. Furthermore, in vivo experiments using a rabbit model of lamellar keratoplasty showed accelerated corneal re-epithelialization and complete stromal repair after the implantation of the 3D-printed scaffold. Overall, due to its high bioactivity and simple preparation, the 3D-printed scaffold using GPLD hydrogel offers an alternative for corneal repair with potential for clinical translation. STATEMENT OF SIGNIFICANCE: The clinical application of hydrogel corneal scaffolds has been constrained by their inadequate mechanical properties and the complex microenvironment created by elevated levels of ROS post-transplantation. In this study, we developed a kind of nanocomposite hydrogel by integrating Laponite and dopamine into GelMA and PEGDA. This advanced hydrogel was utilized to 3D print a corneal scaffold with high mechanical strength and ROS-scavenging abilities. When applied to a rabbit model of lamellar keratoplasty, the 3D-printed scaffold enabled complete re-epithelialization of the cornea within one week. Three months after surgery, the corneal stroma was fully repaired, and regeneration of corneal nerve fibers was also observed. This 3D-printed scaffold demonstrated exceptional efficacy in repairing corneal defects with potential for clinical translation.
Collapse
Affiliation(s)
- Tan Li
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Xiaoyu Zhang
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China; Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan 250021, China
| | - Li Ma
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Xia Qi
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Hongwei Wang
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Xiuli Sun
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Fuyan Wang
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Long Zhao
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Weiyun Shi
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China; Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan 250021, China.
| |
Collapse
|
3
|
Su P, Hu Y, Li J, Wei D, Fu W. Biodegradable PHBVHHx-PEG/Collagen Hydrogel Scaffolds for Cartilage Repair. Tissue Eng Part A 2025. [PMID: 39745260 DOI: 10.1089/ten.tea.2024.0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2025] Open
Abstract
Recently, there has been increased attention on the treatment of cartilage repair. Overall, we constructed PHBVHHx-COL, a composite hydrogel of PHBVHHx-co-PEG and collagen, and evaluated its cartilage repair efficacy through in vitro and in vivo studies using hydrogel loaded with peripheral blood-derived mesenchymal stem cells (PBMSCs). Rheological properties and compressive mechanical properties of the hydrogels were systematically evaluated. The cytocompatibility of the hydrogels was evaluated using the Cell Counting Kit-8 test, live/dead staining, scratch test, and transwell test. The effect of chondrogenic differentiation of PBMSCs on hydrogels was evaluated using immunofluorescence staining and reverse transcription-polymerase chain reaction. Furthermore, the in vivo cartilage repair ability of the hydrogels was confirmed following in situ injections in rabbit chondral defect models. Finally, the induced polarization of the hydrogel scaffold on macrophages was explored by the expression of CD86 and CD206. In vitro experimental results confirmed that PHBVHHx-COL-gel led to better cell migration, proliferation, and chondrogenic differentiation than PHBVHHx-PEG and COL hydrogels. Hematoxylin and eosin staining indicated that the tissue of the repaired area in the PHBVHHx-COL group was nearly in fusion with the surrounding normal tissue and the reconstruction of subchondral bone was good. Safranin-O staining and COL-2 immunohistochemistry indicated that the tissue of the repaired area in the PHBVHHx-COL group had more cartilage-specific matrix secretion. The PHBVHHx-COL group exhibited more M2 macrophage infiltration and less M1 macrophage presentation than the other groups. This study demonstrated that PHBVHHx-COL scaffolds loaded with PBMSCs significantly promoted the repair of cartilage injury through immune regulation by M2 polarization and could be potential candidates for cartilage tissue engineering.
Collapse
Affiliation(s)
- Peng Su
- Department of Orthopedic Surgery and Orthopedic Research Institute, Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Hand and Foot Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yunan Hu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - DaiXu Wei
- Zigong Psychiatric Research Center, Zigong Institute of Brain Science, Zigong Affiliated Hospital of Southwest Medical University, Zigong, China
- Department of Life Sciences and Medicine, Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Weili Fu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Sports Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Wang X, Komasa S, Tahara Y, Inui S, Matsumoto M, Maekawa K. Novel Injectable Collagen/Glycerol/Pullulan Gel Promotes Osteogenic Differentiation of Mesenchymal Stem Cells and the Repair of Rat Cranial Defects. Gels 2024; 10:775. [PMID: 39727533 DOI: 10.3390/gels10120775] [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: 10/06/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Bone tissue engineering is a technique that simulates the bone tissue microenvironment by utilizing cells, tissue scaffolds, and growth factors. The collagen hydrogel is a three-dimensional network bionic material that has properties and structures comparable to those of the extracellular matrix (ECM), making it an ideal scaffold and drug delivery system for tissue engineering. The clinical applications of this material are restricted due to its low mechanical strength. In this investigation, a collagen-based gel (atelocollagen/glycerol/pullulan [Col/Gly/Pul] gel) that is moldable and injectable with high adhesive qualities was created by employing a straightforward technique that involved the introduction of Gly and Pul. This study aimed to characterize the internal morphology and chemical composition of the Col/Gly/Pul gel, as well as to verify its osteogenic properties through in vivo and in vitro experiments. When compared to a standard pure Col hydrogel, this material is more adaptable to the complexity of the local environment of bone defects and the apposition of irregularly shaped flaws due to its greater mechanical strength, injectability, and moldability. Overall, the Col/Gly/Pul gel is an implant that shows great potential for the treatment of complex bone defects and the enhancement of bone regeneration.
Collapse
Affiliation(s)
- Xin Wang
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1, Kuzuhahanazono-cho, Hirakata-shi 573-1121, Osaka, Japan
| | - Satoshi Komasa
- Department of Oral Health Sciences, Osaka Dental University, 1-4-4, Makino-honmachi, Hirakata-shi 573-1144, Osaka, Japan
| | - Yoshiro Tahara
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe 610-0321, Kyoto, Japan
| | - Shihoko Inui
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1, Kuzuhahanazono-cho, Hirakata-shi 573-1121, Osaka, Japan
| | - Michiaki Matsumoto
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe 610-0321, Kyoto, Japan
| | - Kenji Maekawa
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1, Kuzuhahanazono-cho, Hirakata-shi 573-1121, Osaka, Japan
| |
Collapse
|
5
|
Tang X, Zhou F, Wang S, Wang G, Bai L, Su J. Bioinspired injectable hydrogels for bone regeneration. J Adv Res 2024:S2090-1232(24)00486-7. [PMID: 39505143 DOI: 10.1016/j.jare.2024.10.032] [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: 01/07/2024] [Revised: 09/28/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024] Open
Abstract
The effective regeneration of bone/cartilage defects remains a significant clinical challenge, causing irreversible damage to millions annually.Conventional therapies such as autologous or artificial bone grafting often yield unsatisfactory outcomes, emphasizing the urgent need for innovative treatment methods. Biomaterial-based strategies, including hydrogels and active scaffolds, have shown potential in promoting bone/cartilage regeneration. Among them, injectable hydrogels have garnered substantial attention in recent years on account of their minimal invasiveness, shape adaptation, and controlled spatiotemporal release. This review systematically discusses the synthesis of injectable hydrogels, bioinspired approaches-covering microenvironment, structural, compositional, and bioactive component-inspired strategies-and their applications in various bone/cartilage disease models, highlighting bone/cartilage regeneration from an innovative perspective of bioinspired design. Taken together, bioinspired injectable hydrogels offer promising and feasible solutions for promoting bone/cartilage regeneration, ultimately laying the foundations for clinical applications. Furthermore, insights into further prospective directions for AI in injectable hydrogels screening and organoid construction are provided.
Collapse
Affiliation(s)
- Xuan Tang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an 710000, China
| | - Sicheng Wang
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics Trauma, Shanghai Zhongye Hospital, Shanghai 201900, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Wenzhou Institute of Shanghai University, Wenzhou 325000, China.
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
6
|
Arciero I, Buonvino S, Melino S. Slow H 2S-Releasing Donors and 3D Printable Arrays Cellular Models in Osteo-Differentiation of Mesenchymal Stem Cells for Personalized Therapies. Biomolecules 2024; 14:1380. [PMID: 39595557 PMCID: PMC11592188 DOI: 10.3390/biom14111380] [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: 09/24/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
The effects of the hydrogen sulfide (H2S) slow-releasing donor, named GSGa, a glutathione-conjugate water-soluble garlic extract, on human mesenchymal stem cells (hMSCs) in both bidimensional (2D) and three-dimensional (3D) cultures were investigated, demonstrating increased expression of the antioxidant enzyme HO-1 and decreased expression of the pro-inflammatory cytokine interleukin-6 (IL-6). The administration of the H2S donor can therefore increase the expression of antioxidant enzymes, which may have potential therapeutic applications in osteoarthritis (OA). Moreover, GSGa was able to promote the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), but not of cardiac mesenchymal stem cells (cMSCs) in a 2D culture system. This result highlights the varying sensitivity of hMSCs to the H2S donor GSGa, suggesting that the induction of osteogenic differentiation in stem cells by chemical factors is dependent on the tissue of origin. Additionally, a 3D-printable mesenchymal stem cells-bone matrix array (MSCBM), designed to closely mimic the stiffness of bone tissue, was developed to serve as a versatile tool for evaluating the effects of drugs and stem cells on bone repair in chronic diseases, such as OA. We demonstrated that the osteogenic differentiation process in cMSCs can be induced just by simulating bone stiffness in a 3D system. The expression of osteocalcin, RUNX2, and antioxidant enzymes was also assessed after treating MSCs with GSGa and/or increasing the stiffness of the culture environment. The printability of the array may enable better customization of the cavities, enabling an accurate replication of real bone defects. This could optimize the BM array to mimic bone defects not only in terms of stiffness, but also in terms of shape. This culture system may enable a rapid screening of antioxidant and anti-inflammatory compounds, facilitating a more personalized approach to regenerative therapy.
Collapse
Affiliation(s)
- Ilaria Arciero
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy;
| | - Silvia Buonvino
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
| | - Sonia Melino
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
| |
Collapse
|
7
|
Flowers M, Mertens N, Billups A, Ogle BM, Wang C. A Novel Poly(ε-Caprolactone)-Based Photo-Crosslinkable Liquid Copolymer as a Versatile Drug Delivery Platform. Pharmaceutics 2024; 16:1380. [PMID: 39598504 PMCID: PMC11597415 DOI: 10.3390/pharmaceutics16111380] [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: 10/06/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Hydrophobic semi-solid or liquid biodegradable polymers have shown unique advantages as injectable matrices for sustained release of a wide range of drugs. Here we report the design, synthesis, and characterization of a new low-melt liquid copolymer based on poly(ε-caprolactone) (PCL) and establish its utility as a versatile delivery platform. Methods: The copolymer, mPA20, consisting of short PCL blocks connected via acid-labile acetal linkages, was synthesized using a one-pot reaction and its properties were comprehensively characterized. Results: mPA20 is an amorphous, injectable liquid at physiological temperature and can undergo pH-sensitive hydrolytic degradation. mPA20 bearing methacrylate end groups can be photo-crosslinked into solid matrices with tunable mechanical properties. A hydrophobic fluorophore, Nile Red (NR), was solubilized in mPA20 without any solvent. Sustained release of NR into aqueous medium was achieved using mPA20, either as an injectable liquid depot or a photo-crosslinked solid matrix. Further, mPA20 self-emulsified in water to form nanodroplets, which were subsequently photo-crosslinked into nanogels. Both the nanodroplets and nanogels mediated efficient intracellular delivery of NR with no cytotoxicity. Conclusions: mPA20, a new photo-crosslinkable, hydrophobic liquid copolymer with pH-sensitive degradability, is highly adaptable as either an injectable or implantable depot or nanoscale carrier for the controlled release and intracellular delivery of poorly soluble drugs.
Collapse
Affiliation(s)
| | | | | | | | - Chun Wang
- Department of Biomedical Engineering, University of Minnesota, 7-105 Hasselmo Hall, 312 Church Street S.E, Minneapolis, MN 55455, USA; (M.F.); (N.M.); (A.B.); (B.M.O.)
| |
Collapse
|
8
|
Ryu Y, Wague A, Liu X, Feeley BT, Ferguson AR, Morioka K. Cellular signaling pathways in the nervous system activated by various mechanical and electromagnetic stimuli. Front Mol Neurosci 2024; 17:1427070. [PMID: 39430293 PMCID: PMC11486767 DOI: 10.3389/fnmol.2024.1427070] [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: 05/02/2024] [Accepted: 09/25/2024] [Indexed: 10/22/2024] Open
Abstract
Mechanical stimuli, such as stretch, shear stress, or compression, activate a range of biomolecular responses through cellular mechanotransduction. In the nervous system, studies on mechanical stress have highlighted key pathophysiological mechanisms underlying traumatic injury and neurodegenerative diseases. However, the biomolecular pathways triggered by mechanical stimuli in the nervous system has not been fully explored, especially compared to other body systems. This gap in knowledge may be due to the wide variety of methods and definitions used in research. Additionally, as mechanical stimulation techniques such as ultrasound and electromagnetic stimulation are increasingly utilized in psychological and neurorehabilitation treatments, it is vital to understand the underlying biological mechanisms in order to develop accurate pathophysiological models and enhance therapeutic interventions. This review aims to summarize the cellular signaling pathways activated by various mechanical and electromagnetic stimuli with a particular focus on the mammalian nervous system. Furthermore, we briefly discuss potential cellular mechanosensors involved in these processes.
Collapse
Affiliation(s)
- Youngjae Ryu
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Aboubacar Wague
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Xuhui Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Brian T. Feeley
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Adam R. Ferguson
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- Brain and Spinal Injury Center, Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, CA, United States
- San Francisco Veterans Affairs Healthcare System, San Francisco, CA, United States
| | - Kazuhito Morioka
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- Brain and Spinal Injury Center, Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, CA, United States
- Zuckerberg San Francisco General Hospital and Trauma CenterOrthopaedic Trauma Institute, , San Francisco, CA, United States
| |
Collapse
|
9
|
Tan YH, Habing KM, Riesterer JL, Stempinski ES, Lewis SH, Pfeifer CS, Malhotra SV, Nakayama KH. Engineered nanofibrillar collagen with tunable biophysical properties for myogenic, endothelial, and osteogenic cell guidance. Acta Biomater 2024; 186:95-107. [PMID: 39117115 PMCID: PMC11407781 DOI: 10.1016/j.actbio.2024.08.002] [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: 04/15/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
A goal of regenerative engineering is the rational design of materials to restore the structure-function relationships that drive reparative programs in damaged tissues. Despite the widespread use of extracellular matrices for engineering tissues, their application has been limited by a narrow range of tunable features. The primary objective of this study is to develop a versatile platform for evaluating tissue-specific cellular interactions using Type I collagen scaffolds with highly tunable biophysical properties. The kinetics of collagen fibrillogenesis were modulated through a combination of varied shear rate and pH during neutralization, to achieve a broad range of fibril anisotropy, porosity, diameter, and storage modulus. The role that each of these properties play in guiding muscle, bone, and vascular cell types was comprehensively identified, and informed the in vitro generation of three distinct musculoskeletal engineered constructs. Myogenesis was highly regulated by smaller fibrils and larger storage moduli, endothelial inflammatory phenotype was predominantly guided by fibril anisotropy, and osteogenesis was enhanced by highly porous collagen with larger fibrils. This study introduces a novel approach for dynamically modulating Type I collagen materials and provides a robust platform for investigating cell-material interactions, offering insights for the future rational design of tissue-specific regenerative biomaterials. STATEMENT OF SIGNIFICANCE: The biophysical properties of regenerative materials facilitate key cell-substrate interactions that can guide the morphology, phenotype, and biological response of cells. In this study, we describe the fabrication of an engineered collagen hydrogel that can be modified to exhibit control over a wide range of biophysical features, including fibril organization and size, nanoscale porosity, and mechanics. We identified the unique combination of collagen features that optimally promote regenerative muscle, bone, and vascular cell types while also delineating the properties that hinder these same cellular responses. This study presents a highly accessible method to control the biophysical properties of collagen hydrogels that can be adapted for a broad range of tissue engineering and regenerative applications.
Collapse
Affiliation(s)
- Yong How Tan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Krista M Habing
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Jessica L Riesterer
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Erin S Stempinski
- Multiscale Microscopy Core, Oregon Health & Science University, Portland, OR, USA
| | - Steven H Lewis
- Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Carmem S Pfeifer
- Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Sanjay V Malhotra
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Karina H Nakayama
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA; Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
10
|
Jiao H, Lu X, Li Y, Zhang H, Fu Y, Zhong C, Wang Q, Ullah MW, Liu H, Yong YC, Liu J. In situ biomineralization reinforcing anisotropic nanocellulose scaffolds for guiding the differentiation of bone marrow-derived mesenchymal stem cells. Int J Biol Macromol 2024; 274:133515. [PMID: 38944070 DOI: 10.1016/j.ijbiomac.2024.133515] [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: 02/20/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Nanocellulose (NC) is a promising biopolymer for various biomedical applications owing to its biocompatibility and low toxicity. However, it faces challenges in tissue engineering (TE) applications due to the inconsistency of the microenvironment within the NC-based scaffolds with target tissues, including anisotropy microstructure and biomechanics. To address this challenge, a facile swelling-induced nanofiber alignment and a novel in situ biomineralization reinforcement strategies were developed for the preparation of NC-based scaffolds with tunable anisotropic structure and mechanical strength for guiding the differentiation of bone marrow-derived mesenchymal stem cells for potential TE application. The bacterial cellulose (BC) and cellulose nanofibrils (CNFs) based scaffolds with tunable swelling anisotropic index in the range of 10-100 could be prepared by controlling the swelling medium. The in situ biomineralization efficiently reinforced the scaffolds with 2-4 times and 10-20 times modulus increasement for BC and CNFs, respectively. The scaffolds with higher mechanical strength were superior in supporting cell growth and proliferation, suggesting the potential application in TE application. This work demonstrated the feasibility of the proposed strategy in the preparation of scaffolds with mechanical anisotropy to induce cells-directed differentiation for TE applications.
Collapse
Affiliation(s)
- Haixin Jiao
- Biofuels Institute, School of Emergency Management, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Xuechu Lu
- Biofuels Institute, School of Emergency Management, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Yan Li
- Biofuels Institute, School of Emergency Management, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Hongxing Zhang
- Biofuels Institute, School of Emergency Management, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Yinyi Fu
- Biofuels Institute, School of Emergency Management, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | | | - Qianqian Wang
- Biofuels Institute, School of Emergency Management, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Muhammad Wajid Ullah
- Biofuels Institute, School of Emergency Management, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Huan Liu
- Biofuels Institute, School of Emergency Management, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Yang-Chun Yong
- Biofuels Institute, School of Emergency Management, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China; Jiangsu Collaborative Innovation Center of Technology and Material of Water Treatment, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jun Liu
- Biofuels Institute, School of Emergency Management, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China; Jiangsu Collaborative Innovation Center of Technology and Material of Water Treatment, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
11
|
Ma H, Zhang T. Histone demethylase KDM3B mediates matrix stiffness-induced osteogenic differentiation of adipose-derived stem cells. Arch Biochem Biophys 2024; 757:110028. [PMID: 38768746 DOI: 10.1016/j.abb.2024.110028] [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: 09/29/2023] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Biomechanical signals in the extracellular niche are considered promising for programming the lineage specification of stem cells. Recent studies have reported that biomechanics, such as the microstructure of nanomaterials, can induce adipose-derived stem cells (ASCs) to differentiate into osteoblasts, mediating gene regulation at the epigenetic level. Therefore, in this study, transcriptome expression levels of histone demethylases in ASCs were screened after treatment with different matrix stiffnesses, and histone lysine demethylase 3B (KDM3B) was found to promote osteogenic differentiation of ASCs in response to matrix stiffness, indicating a positive modulatory effect on this biological process. ASCs exhibited widespread and polygonal shapes with a distinct bundle-like expression of vinculin parallel to the axial cytoskeleton along the cell margins on the stiff matrix rather than round shapes with a smeared and shorter expression on the soft matrix. Comparatively rigid polydimethylsiloxane material directed ASCs into an osteogenic phenotype in inductive culture media via the upregulation of osteocalcin, alkaline phosphatase, and runt-related transcription factor 2. Treatment with KDM3B-siRNA decreased the expression of osteogenic differentiation markers and impaired mitochondrial dynamics and mitochondrial membrane potential. These results illustrate the critical role of KDM3B in the biomechanics-induced osteogenic commitment of ASCs and provide new avenues for the further application of stem cells as potential therapeutics for bone regeneration.
Collapse
Affiliation(s)
- Huangshui Ma
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
12
|
Buck HV, Stains JP. Osteocyte-mediated mechanical response controls osteoblast differentiation and function. Front Physiol 2024; 15:1364694. [PMID: 38529481 PMCID: PMC10961341 DOI: 10.3389/fphys.2024.1364694] [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: 01/02/2024] [Accepted: 02/29/2024] [Indexed: 03/27/2024] Open
Abstract
Low bone mass is a pervasive global health concern, with implications for osteoporosis, frailty, disability, and mortality. Lifestyle factors, including sedentary habits, metabolic dysfunction, and an aging population, contribute to the escalating prevalence of osteopenia and osteoporosis. The application of mechanical load to bone through physical activity and exercise prevents bone loss, while sufficient mechanical load stimulates new bone mass acquisition. Osteocytes, cells embedded within the bone, receive mechanical signals and translate these mechanical cues into biological signals, termed mechano-transduction. Mechano-transduction signals regulate other bone resident cells, such as osteoblasts and osteoclasts, to orchestrate changes in bone mass. This review explores the mechanisms through which osteocyte-mediated response to mechanical loading regulates osteoblast differentiation and bone formation. An overview of bone cell biology and the impact of mechanical load will be provided, with emphasis on the mechanical cues, mechano-transduction pathways, and factors that direct progenitor cells toward the osteoblast lineage. While there are a wide range of clinically available treatments for osteoporosis, the majority act through manipulation of the osteoclast and may have significant disadvantages. Despite the central role of osteoblasts to the deposition of new bone, few therapies directly target osteoblasts for the preservation of bone mass. Improved understanding of the mechanisms leading to osteoblastogenesis may reveal novel targets for translational investigation.
Collapse
Affiliation(s)
| | - Joseph Paul Stains
- School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
13
|
Kosmidis Papadimitriou A, Chong SW, Shen Y, Lee OS, Knowles TPJ, Grover LM, Vigolo D. Fabrication of gradient hydrogels using a thermophoretic approach in microfluidics. Biofabrication 2024; 16:025023. [PMID: 38377611 DOI: 10.1088/1758-5090/ad2b05] [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: 10/17/2023] [Accepted: 02/20/2024] [Indexed: 02/22/2024]
Abstract
The extracellular matrix presents spatially varying physical cues that can influence cell behavior in many processes. Physical gradients within hydrogels that mimic the heterogenous mechanical microenvironment are useful to study the impact of these cues on cellular responses. Therefore, simple and reliable techniques to create such gradient hydrogels are highly desirable. This work demonstrates the fabrication of stiffness gradient Gellan gum (GG) hydrogels by applying a temperature gradient across a microchannel containing hydrogel precursor solution. Thermophoretic migration of components within the precursor solution generates a concentration gradient that mirrors the temperature gradient profile, which translates into mechanical gradients upon crosslinking. Using this technique, GG hydrogels with stiffness gradients ranging from 20 to 90 kPa over 600µm are created, covering the elastic moduli typical of moderately hard to hard tissues. MC3T3 osteoblast cells are then cultured on these gradient substrates, which exhibit preferential migration and enhanced osteogenic potential toward the stiffest region on the gradient. Overall, the thermophoretic approach provides a non-toxic and effective method to create hydrogels with defined mechanical gradients at the micron scale suitable forin vitrobiological studies and potentially tissue engineering applications.
Collapse
Affiliation(s)
| | - Shin Wei Chong
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Yi Shen
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
- The University of Sydney, School of Chemical and Biomolecular Engineering, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Oisin Stefan Lee
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW 2006, Australia
| | - Tuomas P J Knowles
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Liam M Grover
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Daniele Vigolo
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
- The University of Sydney, School of Biomedical Engineering, Sydney, NSW 2006, Australia
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
14
|
Bontempi M, Marchiori G, Petretta M, Capozza R, Grigolo B, Giavaresi G, Gambardella A. Nanomechanical Mapping of Three Dimensionally Printed Poly-ε-Caprolactone Single Microfibers at the Cell Scale for Bone Tissue Engineering Applications. Biomimetics (Basel) 2023; 8:617. [PMID: 38132556 PMCID: PMC10742115 DOI: 10.3390/biomimetics8080617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Poly-ε-caprolactone (PCL) has been widely used in additive manufacturing for the construction of scaffolds for bone tissue engineering. However, its use is limited by its lack of bioactivity and inability to induce cell adhesion, hence limiting bone tissue regeneration. Biomimicry is strongly influenced by the dynamics of cell-substrate interaction. Thus, characterizing scaffolds at the cell scale could help to better understand the relationship between surface mechanics and biological response. We conducted atomic force microscopy-based nanoindentation on 3D-printed PCL fibers of ~300 µm thickness and mapped the near-surface Young's modulus at loading forces below 50 nN. In this non-disruptive regime, force mapping did not show clear patterns in the spatial distribution of moduli or a relationship with the topographic asperities within a given region. Remarkably, we found that the average modulus increased linearly with the logarithm of the strain rate. Finally, a dependence of the moduli on the history of nanoindentation was demonstrated on locations of repeated nanoindentations, likely due to creep phenomena capable of hindering viscoelasticity. Our findings can contribute to the rational design of scaffolds for bone regeneration that are capable of inducing cell adhesion and proliferation. The methodologies described are potentially applicable to various tissue-engineered biopolymers.
Collapse
Affiliation(s)
- Marco Bontempi
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (M.B.); (G.M.); (G.G.)
| | - Gregorio Marchiori
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (M.B.); (G.M.); (G.G.)
| | - Mauro Petretta
- REGENHU SA, Z.I Du Vivier 22, CH-1690 Villaz-St-Pierre, Switzerland;
| | - Rosario Capozza
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3DW, UK;
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
| | - Gianluca Giavaresi
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (M.B.); (G.M.); (G.G.)
| | - Alessandro Gambardella
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (M.B.); (G.M.); (G.G.)
| |
Collapse
|
15
|
Hao S, Wang M, Yin Z, Jing Y, Bai L, Su J. Microenvironment-targeted strategy steers advanced bone regeneration. Mater Today Bio 2023; 22:100741. [PMID: 37576867 PMCID: PMC10413201 DOI: 10.1016/j.mtbio.2023.100741] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Treatment of large bone defects represents a great challenge in orthopedic and craniomaxillofacial surgery. Traditional strategies in bone tissue engineering have focused primarily on mimicking the extracellular matrix (ECM) of bone in terms of structure and composition. However, the synergistic effects of other cues from the microenvironment during bone regeneration are often neglected. The bone microenvironment is a sophisticated system that includes physiological (e.g., neighboring cells such as macrophages), chemical (e.g., oxygen, pH), and physical factors (e.g., mechanics, acoustics) that dynamically interact with each other. Microenvironment-targeted strategies are increasingly recognized as crucial for successful bone regeneration and offer promising solutions for advancing bone tissue engineering. This review provides a comprehensive overview of current microenvironment-targeted strategies and challenges for bone regeneration and further outlines prospective directions of the approaches in construction of bone organoids.
Collapse
Affiliation(s)
- Shuyue Hao
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, China
| |
Collapse
|
16
|
Moore ER, Maridas DE, Gamer L, Chen G, Burton K, Rosen V. A periosteum-derived cell line to study the role of BMP/TGFβ signaling in periosteal cell behavior and function. Front Physiol 2023; 14:1221152. [PMID: 37799511 PMCID: PMC10547901 DOI: 10.3389/fphys.2023.1221152] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/05/2023] [Indexed: 10/07/2023] Open
Abstract
The periosteum is a thin tissue surrounding each skeletal element that contains stem and progenitor cells involved in bone development, postnatal appositional bone growth, load-induced bone formation, and fracture repair. BMP and TGFβ signaling are important for periosteal activity and periosteal cell behavior, but thorough examination of the influence of these pathways on specific cell populations resident in the periosteum is lacking due to limitations associated with primary periosteal cell isolations and in vitro experiments. Here we describe the generation of a novel periosteum-derived clonal cell (PDC) line from postnatal day 14 mice and use it to examine periosteal cell behavior in vitro. PDCs exhibit key characteristics of periosteal cells observed during skeletal development, maintenance, and bone repair. Specifically, PDCs express established periosteal markers, can be expanded in culture, demonstrate the ability to differentiate into chondrocytes, osteoblasts, and adipocytes, and exhibit an osteogenic response to physical stimulation. PDCs also engage in BMP and/or TGFβ signaling when treated with the activating ligands BMP2 and TGFβ-1, and in response to mechanical stimulation via fluid shear. We believe that this PDC line will be useful for large-scale, long-term experiments that were not feasible when using primary periosteal cells. Anticipated future uses include advancing our understanding of the signaling interactions that occur during appositional bone growth and fracture repair and developing drug screening platforms to discover novel growth and fracture healing factors.
Collapse
Affiliation(s)
- Emily R. Moore
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, United States
| | | | | | | | | | | |
Collapse
|
17
|
Gao S, Chen B, Gao M, Xu Y, Yang X, Yang C, Pan S. Substrate Stiffness of Bone Microenvironment Controls Functions of Pre-Osteoblasts and Fibroblasts In Vitro. Biomimetics (Basel) 2023; 8:344. [PMID: 37622949 PMCID: PMC10452586 DOI: 10.3390/biomimetics8040344] [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: 05/29/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
The formation of bone in a bone defect is accomplished by osteoblasts, while the over activation of fibroblasts promotes fibrosis. However, it is not clear how the extracellular matrix stiffness of the bone-regeneration microenvironment affects the function of osteoblasts and fibroblasts. This study aim to investigate the effect of bone-regeneration microenvironment stiffness on cell adhesion, cell proliferation, cell differentiation, synthesizing matrix ability and its potential mechanisms in mechanotransduction, in pre-osteoblasts and fibroblasts. Polyacrylamide substrates mimicking the matrix stiffness of different stages of the bone-healing process (15 kPa, mimic granulation tissue; 35 kPa, mimic osteoid; 150 kPa, mimic calcified bone matrix) were prepared. Mouse pre-osteoblasts MC3T3-E1 and mouse fibroblasts NIH3T3 were plated on three types of substrates, respectively. There were significant differences in the adhesion of pre-osteoblasts and fibroblasts on different polyacrylamide substrates. Runx2 expression increased with increasing substrate stiffness in pre-osteoblasts, while no statistical differences were found in the Acta2 expression in fibroblasts on three substrates. OPN expression in pre-osteoblasts, as well as Fn1 and Col1a1 expression in fibroblasts, decreased with increasing stiffness. The difference between the cell traction force generated by pre-osteoblasts and fibroblasts on substrates was also found. Our results indicated that substrate stiffness is a potent regulator of pre-osteoblasts and fibroblasts with the ability of promoting osteogenic differentiation of pre-osteoblasts, while having no effect on myofibroblast differentiation of fibroblasts.
Collapse
Affiliation(s)
- Shenghan Gao
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Central Laboratory, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Bo Chen
- Department of Implantology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Min Gao
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No. 22, Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Yue Xu
- Institute of Biomechanics and Medical Engineering, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
| | - Xueyi Yang
- Institute of Biomechanics and Medical Engineering, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
| | - Chun Yang
- Institute of Biomechanics and Medical Engineering, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
| | - Shaoxia Pan
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Central Laboratory, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| |
Collapse
|
18
|
Kutluk H, Bastounis EE, Constantinou I. Integration of Extracellular Matrices into Organ-on-Chip Systems. Adv Healthc Mater 2023; 12:e2203256. [PMID: 37018430 PMCID: PMC11468608 DOI: 10.1002/adhm.202203256] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/20/2023] [Indexed: 04/07/2023]
Abstract
The extracellular matrix (ECM) is a complex, dynamic network present within all tissues and organs that not only acts as a mechanical support and anchorage point but can also direct fundamental cell behavior, function, and characteristics. Although the importance of the ECM is well established, the integration of well-controlled ECMs into Organ-on-Chip (OoC) platforms remains challenging and the methods to modulate and assess ECM properties on OoCs remain underdeveloped. In this review, current state-of-the-art design and assessment of in vitro ECM environments is discussed with a focus on their integration into OoCs. Among other things, synthetic and natural hydrogels, as well as polydimethylsiloxane (PDMS) used as substrates, coatings, or cell culture membranes are reviewed in terms of their ability to mimic the native ECM and their accessibility for characterization. The intricate interplay among materials, OoC architecture, and ECM characterization is critically discussed as it significantly complicates the design of ECM-related studies, comparability between works, and reproducibility that can be achieved across research laboratories. Improving the biomimetic nature of OoCs by integrating properly considered ECMs would contribute to their further adoption as replacements for animal models, and precisely tailored ECM properties would promote the use of OoCs in mechanobiology.
Collapse
Affiliation(s)
- Hazal Kutluk
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| | - Effie E. Bastounis
- Institute of Microbiology and Infection Medicine (IMIT)Eberhard Karls University of TübingenAuf der Morgenstelle 28, E872076TübingenGermany
- Cluster of Excellence “Controlling Microbes to Fight Infections” EXC 2124Eberhard Karls University of TübingenAuf der Morgenstelle 2872076TübingenGermany
| | - Iordania Constantinou
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| |
Collapse
|
19
|
Bessot A, Gunter J, Waugh D, Clements JA, Hutmacher DW, McGovern J, Bock N. GelMA and Biomimetic Culture Allow the Engineering of Mineralized, Adipose, and Tumor Tissue Human Microenvironments for the Study of Advanced Prostate Cancer In Vitro and In Vivo. Adv Healthc Mater 2023; 12:e2201701. [PMID: 36708740 PMCID: PMC11469108 DOI: 10.1002/adhm.202201701] [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: 07/11/2022] [Revised: 12/21/2022] [Indexed: 01/30/2023]
Abstract
Increasing evidence shows bone marrow (BM)-adipocytes as a potentially important contributor in prostate cancer (PCa) bone metastases. However, a lack of relevant models has prevented the full understanding of the effects of human BM-adipocytes in this microenvironment. It is hypothesized that the combination of tunable gelatin methacrylamide (GelMA)-based hydrogels with the biomimetic culture of human cells would offer a versatile 3D platform to engineer human bone tumor microenvironments containing BM-adipocytes. Human osteoprogenitors, adipocytes, and PCa cells are individually cultured in vitro in GelMA hydrogels, leading to mineralized, adipose, and PCa tumor 3D microtissues, respectively. Osteoblast mineralization and tumor spheroid formation are tailored by hydrogel stiffness with lower stiffnesses correlating with increased mineralization and tumor spheroid size. Upon coculture with tumor cells, BM-adipocytes undergo morphological changes and delipidation, suggesting reciprocal interactions between the cell types. When brought in vivo, the mineralized and adipose microtissues successfully form a humanized fatty bone microenvironment, presenting, for the first time, with human adipocytes. Using this model, an increase in tumor burden is observed when human adipocytes are present, suggesting that adipocytes support early bone tumor growth. The advanced platform presented here combines natural aspects of the microenvironment with tunable properties useful for bone tumor research.
Collapse
Affiliation(s)
- Agathe Bessot
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
- Australian Prostate Cancer Research Centre ‐ Queensland (APCRC‐Q)QUTBrisbaneQLD4102Australia
- Centre for Biomedical TechnologiesQUTBrisbaneQLD4000Australia
- Max Planck Queensland CentreBrisbaneQLD4059Australia
| | - Jennifer Gunter
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
- Australian Prostate Cancer Research Centre ‐ Queensland (APCRC‐Q)QUTBrisbaneQLD4102Australia
- Centre for Genomics and Personalised HealthQUTBrisbaneQLD4102Australia
| | - David Waugh
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
| | - Judith A. Clements
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
- Australian Prostate Cancer Research Centre ‐ Queensland (APCRC‐Q)QUTBrisbaneQLD4102Australia
| | - Dietmar W. Hutmacher
- School of MechanicalMedical and Process EngineeringEngineering FacultyQUTBrisbaneQLD4000Australia
- Max Planck Queensland CentreBrisbaneQLD4059Australia
| | - Jacqui McGovern
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
- Centre for Biomedical TechnologiesQUTBrisbaneQLD4000Australia
- Max Planck Queensland CentreBrisbaneQLD4059Australia
| | - Nathalie Bock
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
- Australian Prostate Cancer Research Centre ‐ Queensland (APCRC‐Q)QUTBrisbaneQLD4102Australia
- Centre for Biomedical TechnologiesQUTBrisbaneQLD4000Australia
- Max Planck Queensland CentreBrisbaneQLD4059Australia
| |
Collapse
|
20
|
Peng L, Wu F, Cao M, Li M, Cui J, Liu L, Zhao Y, Yang J. Effects of different physical factors on osteogenic differentiation. Biochimie 2023; 207:62-74. [PMID: 36336107 DOI: 10.1016/j.biochi.2022.10.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 10/11/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Osteoblasts are essential for bone formation and can perceive external mechanical stimuli, which are translated into biochemical responses that ultimately alter cell phenotypes and respond to environmental stimuli, described as mechanical transduction. These cells actively participate in osteogenesis and the formation and mineralisation of the extracellular bone matrix. This review summarises the basic physiological and biological mechanisms of five different physical stimuli, i.e. light, electricity, magnetism, force and sound, to induce osteogenesis; further, it summarises the effects of changing culture conditions on the morphology, structure and function of osteoblasts. These findings may provide a theoretical basis for further studies on bone physiology and pathology at the cytological level and will be useful in the clinical application of bone formation and bone regeneration technology.
Collapse
Affiliation(s)
- Li Peng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Fanzi Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, China
| | - Mengjiao Cao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, China
| | - Mengxin Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, China
| | - Jingyao Cui
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, China
| | - Lijia Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, China
| | - Yun Zhao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China.
| | - Jing Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, China.
| |
Collapse
|
21
|
Zheng X, Wang W, Chen S, Zuo B, Li J. Transplanted mesenchymal stromal cells are unable to migrate to the bone surface and subsequently improve osteogenesis in glucocorticoid-induced osteoporosis. Cytotherapy 2023; 25:472-482. [PMID: 36863932 DOI: 10.1016/j.jcyt.2023.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/14/2022] [Accepted: 01/07/2023] [Indexed: 03/03/2023]
Abstract
Long-term or high-dose use of glucocorticoids causes bone loss and low bone formation. We previously demonstrated that dexamethasone (Dex) administration caused the shifted differentiation balance of mesenchymal stromal cells (MSCs) to favor adipogenic lineage over osteoblastic lineage, which is one of the key mechanisms for Dex-induced osteoporosis (DIO). These findings indicate that supplementing functional allogeneic MSCs could be a therapeutic strategy for DIO. Here, we found that transplanting MSCs by intramedullary injection had little effect in promoting new bone formation. Fluorescent-labeled lineage tracing revealed that 1 week after transplantation, green fluorescent protein (GFP)-MSCs were found to migrate to the bone surface (BS) in control mice but not in DIO mice. As expected, GFP-MSCs on the BS were mostly Runx2-positive; however, GFP-MSCs located away from the BS failed to differentiate into osteoblasts. We further discovered that the levels of transforming growth factor beta 1 (TGF-β1), one of the main chemokines for MSC migration, is significantly decreased in the bone marrow fluid of DIO mice, which is insufficient to direct MSC migration. Mechanistically, Dex inhibits TGF-β1 expression by down-regulating its promoter activity, which decreases bone matrix-deposited TGF-β1 as well as active TGF-β1 released during osteoclast-mediated bone resorption. This study indicates that blocking MSC migration in osteoporotic BM contributes to bone loss and suggests that MSC mobilization to the BS may be a promising target for treating osteoporosis.
Collapse
Affiliation(s)
- Xueling Zheng
- Department of Cell Biology, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Wanyuji Wang
- Department of Cell Biology, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Sisi Chen
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Zuo
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiao Li
- Department of Cell Biology, Zunyi Medical University, Zunyi, Guizhou Province, China.
| |
Collapse
|
22
|
Meyer N, Bax DV, Beck J, Cameron RE, Best SM. Adjusting the physico-chemical properties of collagen scaffolds to accommodate primary osteoblasts and endothelial cells. Regen Biomater 2023; 10:rbad015. [PMID: 36937897 PMCID: PMC10019812 DOI: 10.1093/rb/rbad015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/08/2022] [Accepted: 01/09/2023] [Indexed: 03/12/2023] Open
Abstract
Collagen-based biomaterials are used widely as tissue engineering scaffolds because of their excellent bioactivity and their similarity to the natural ECM. The regeneration of healthy bone tissue requires simultaneous support for both osteoblasts and, where angiogenesis is intended, endothelial cells. Hence it is important to tailor carefully the biochemical and structural characteristics of the scaffold to suit the needs of each cell type. This work describes for the first time a systematic study to gain insight into the cell type-specific response of primary human osteoblast (hOBs) and human dermal microvascular endothelial cells (HDMECs) to insoluble collagen-based biomaterials. The behaviour was evaluated on both 2D films and 3D scaffolds, produced using freeze-drying. The collagen was cross-linked at various EDC/NHS concentrations and mono-cultured with hOBs and HDMECs to assess the effect of architectural features and scaffold stabilization on cell behaviour. It was observed that 3D scaffolds cross-linked at 30% of the standard conditions in literature offered an optimal combination of mechanical stiffness and cellular response for both cell types, although endothelial cells were more sensitive to the degree of cross-linking than hOBs. Architectural features have a time-dependent impact on the cell migration profile, with alignment being the most influential parameter overall.
Collapse
Affiliation(s)
- Nima Meyer
- Department of Materials Science and Metallurgy, University of Cambridge, Cambridge CB3 0FS, UK
| | | | - Jochen Beck
- Geistlich Pharma AG, Product Development, Wolhusen, Switzerland
| | - Ruth E Cameron
- Department of Materials Science and Metallurgy, University of Cambridge, Cambridge CB3 0FS, UK
| | - Serena M Best
- Department of Materials Science and Metallurgy, University of Cambridge, Cambridge CB3 0FS, UK
| |
Collapse
|
23
|
Erenay B, Sağlam ASY, Garipcan B, Jandt KD, Odabaş S. Bone surface mimicked PDMS membranes stimulate osteoblasts and calcification of bone matrix. BIOMATERIALS ADVANCES 2022; 142:213170. [PMID: 36341745 DOI: 10.1016/j.bioadv.2022.213170] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
Cellular microenvironments play a crucial role in cell behavior. In addition to the biochemical cues present in the microenvironments, biophysical and biomechanical properties on surfaces have an impact on cellular functionality and eventually cellular fate. Effects of surface topography on cell behavior are being studied extensively in the literature. However, these studies often try to replicate topographical features of tissue surfaces by using techniques such as chemical etching, photolithography, and electrospinning, which may result in the loss of crucial micro- and nano- features on the tissue surfaces such as bone. This study investigates the topographical effects of bone surface by transferring its surface features onto polydimethylsiloxane (PDMS) membranes using soft lithography from a bovine femur. Our results have shown that major features on bone surfaces were successfully transferred onto PDMS using soft lithography. Osteoblast proliferation and calcification of bone matrix have significantly increased along with osteoblast-specific differentiation and maturation markers such as osteocalcin (OSC), osterix (OSX), collagen type I alpha 1 chain (COL1A1), and alkaline phosphatase (ALP) on bone surface mimicked (BSM) PDMS membranes in addition to a unidirectional alignment of osteoblast cells compared to plain PDMS surfaces. This presented bone surface mimicking method can provide a versatile native-like platform for further investigation of intracellular pathways regarding osteoblast growth and differentiation.
Collapse
Affiliation(s)
- Berkay Erenay
- Biomimetics and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Turkey
| | - Atiye Seda Yar Sağlam
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara 06500, Turkey
| | - Bora Garipcan
- Biomimetics and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Turkey
| | - Klaus D Jandt
- Chair of Materials Science, Otto Schott Institute of Materials Research, Friedrich Schiller University, Jena 07743, Germany.
| | - Sedat Odabaş
- Biomaterials and Tissue Engineering Laboratory (BteLAB), Faculty of Science, Department of Chemistry, Ankara University, 06560, Turkey; Interdisciplinary Research Unit for Advanced Materials (INTRAM), Ankara University, Ankara 06560, Turkey.
| |
Collapse
|
24
|
Tsuji N, Sakamoto T, Hoshi K, Hikita A. Spatiotemporal Analysis of Osteoblast Morphology and Wnt Signal‐Induced Osteoblast Reactivation during Bone Modeling in Vitro. JBMR Plus 2022; 6:e10689. [PMID: 36398107 PMCID: PMC9664540 DOI: 10.1002/jbm4.10689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
Bone nodule formation by differentiating osteoblasts is considered an in vitro model that mimics bone modeling. However, the details of osteoblast behavior and matrix production during bone nodule formation are poorly understood. Here, we present a spatiotemporal analysis system for evaluating osteoblast morphology and matrix production during bone modeling in vitro via two-photon microscopy. Using this system, a change in osteoblast morphology from cuboidal to flat was observed during the formation of mineralized nodules, and this change was quantified. Areas with high bone formation were densely populated with cuboidal osteoblasts, which were characterized by blebs, protruding structures on their cell membranes. Cuboidal osteoblasts with blebs were highly mobile, and osteoblast blebs exhibited a polar distribution. Furthermore, mimicking romosozumab treatment, when differentiated flattened osteoblasts were stimulated with BIO, a GSK3β inhibitor, they were reactivated to acquire a cuboidal morphology with blebs on their membranes and produced more matrix than nonstimulated cells. Our analysis system is a powerful tool for evaluating the cell morphology and function of osteoblasts during bone modeling. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Naoki Tsuji
- Department of Sensory and Motor System Medicine, Graduate School of Medicine The University of Tokyo Tokyo Japan
| | - Tomoaki Sakamoto
- Department of Tissue Engineering The University of Tokyo Hospital Tokyo Japan
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Graduate School of Medicine The University of Tokyo Tokyo Japan
- Department of Tissue Engineering The University of Tokyo Hospital Tokyo Japan
| | - Atsuhiko Hikita
- Department of Tissue Engineering The University of Tokyo Hospital Tokyo Japan
| |
Collapse
|
25
|
Deering J, Lin DSY, D'Elia A, Zhang B, Grandfield K. Fabrication of succinate-alginate xerogel films for in vitro coupling of osteogenesis and neovascularization. BIOMATERIALS ADVANCES 2022; 141:213122. [PMID: 36162345 DOI: 10.1016/j.bioadv.2022.213122] [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: 04/22/2022] [Revised: 08/30/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
The osseointegration of metallic implants is reliant on a cascade of molecular interactions and the delivery of macromolecules to the implant environment that occurs before substantial bone formation. Early blood vessel formation is a requisite first step in the healing timeline for osteoid formation, where vascular development can be accelerated as a result of controlled hypoxic conditioning. In this study, alginate-derived xerogel films containing varied concentrations of disodium succinate salt which has been shown to induce pseudohypoxia (short-term hypoxic effects while maintaining an oxygenated environment) were developed. Xerogels were characterized for their morphology, succinate release over time and cellular response with osteoblast-mimicking Saos-2 and human umbilical vein endothelial cells (HUVEC). Scanning electron microscopy revealed a multiscale topography that may favour osseointegration and alamarBlue assays indicated no cytotoxic effects during in vitro proliferation of Saos-2 cells. pH measurements of eluted succinate reach 95 % of peak value after 7 h of immersion for all gels containing 10 mM of succinate or less, and 60 % within the first 40 min. In vitro exposure of HUVECs to succinate-conditioned media increased the net concentration of total proteins measured by bicinchoninic acid (BCA) assay and maintains stable vascular endothelial growth factor (VEGF) and extracellular platelet-derived growth factor (PDGF) for vessel formation through comparison of enzyme-linked immunosorbent assays (ELISAs) of the culture media and cell lysate. Tube formation assays also showed a sustained increase in tube diameter across the first 48 h of HUVEC culture when succinate concentrations of 1 and 10 μM in the xerogel. Overall, the succinate-alginate films serve as a prospective organic coating for bone-interfacing implant materials which may induce temporary pseudohypoxic conditions favourable for early angiogenesis and bone regeneration in vivo at succinate concentrations of 1 or 10 μM.
Collapse
Affiliation(s)
- Joseph Deering
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, Canada
| | - Dawn S Y Lin
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Andrew D'Elia
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, Canada
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada; School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
| | - Kathryn Grandfield
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, Canada; School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada; Brockhouse Institute for Materials Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
26
|
Buj-Corral I, Sanz-Fraile H, Ulldemolins A, Tejo-Otero A, Domínguez-Fernández A, Almendros I, Otero J. Characterization of 3D Printed Metal-PLA Composite Scaffolds for Biomedical Applications. Polymers (Basel) 2022; 14:polym14132754. [PMID: 35808799 PMCID: PMC9268876 DOI: 10.3390/polym14132754] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/27/2022] [Accepted: 07/01/2022] [Indexed: 01/06/2023] Open
Abstract
Three-dimensional printing is revolutionizing the development of scaffolds due to their rapid-prototyping characteristics. One of the most used techniques is fused filament fabrication (FFF), which is fast and compatible with a wide range of polymers, such as PolyLactic Acid (PLA). Mechanical properties of the 3D printed polymeric scaffolds are often weak for certain applications. A potential solution is the development of composite materials. In the present work, metal-PLA composites have been tested as a material for 3D printing scaffolds. Three different materials were tested: copper-filled PLA, bronze-filled PLA, and steel-filled PLA. Disk-shaped samples were printed with linear infill patterns and line spacing of 0.6, 0.7, and 0.8 mm, respectively. The porosity of the samples was measured from cross-sectional images. Biocompatibility was assessed by culturing Human Bone Marrow-Derived Mesenchymal Stromal on the surface of the printed scaffolds. The results showed that, for identical line spacing value, the highest porosity corresponded to bronze-filled material and the lowest one to steel-filled material. Steel-filled PLA polymers showed good cytocompatibility without the need to coat the material with biomolecules. Moreover, human bone marrow-derived mesenchymal stromal cells differentiated towards osteoblasts when cultured on top of the developed scaffolds. Therefore, it can be concluded that steel-filled PLA bioprinted parts are valid scaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- Irene Buj-Corral
- Department of Mechanical Engineering, School of Engineering of Barcelona (ETSEIB), Universitat Politècnica de Catalunya, Av. Diagonal 647, 08028 Barcelona, Spain; (A.T.-O.); (A.D.-F.)
- Correspondence: (I.B.-C.); (J.O.)
| | - Héctor Sanz-Fraile
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (H.S.-F.); (A.U.); (I.A.)
| | - Anna Ulldemolins
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (H.S.-F.); (A.U.); (I.A.)
| | - Aitor Tejo-Otero
- Department of Mechanical Engineering, School of Engineering of Barcelona (ETSEIB), Universitat Politècnica de Catalunya, Av. Diagonal 647, 08028 Barcelona, Spain; (A.T.-O.); (A.D.-F.)
| | - Alejandro Domínguez-Fernández
- Department of Mechanical Engineering, School of Engineering of Barcelona (ETSEIB), Universitat Politècnica de Catalunya, Av. Diagonal 647, 08028 Barcelona, Spain; (A.T.-O.); (A.D.-F.)
| | - Isaac Almendros
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (H.S.-F.); (A.U.); (I.A.)
- CIBER de Enfermedades Respiratorias, 28029 Madrid, Spain
| | - Jorge Otero
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (H.S.-F.); (A.U.); (I.A.)
- CIBER de Enfermedades Respiratorias, 28029 Madrid, Spain
- Correspondence: (I.B.-C.); (J.O.)
| |
Collapse
|
27
|
Effect on Rheological Properties and 3D Printability of Biphasic Calcium Phosphate Microporous Particles in Hydrocolloid-Based Hydrogels. Gels 2022; 8:gels8010028. [PMID: 35049563 PMCID: PMC8774978 DOI: 10.3390/gels8010028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/04/2022] Open
Abstract
The production of patient-specific bone substitutes with an exact fit through 3D printing is emerging as an alternative to autologous bone grafting. To the success of tissue regeneration, the material characteristics such as porosity, stiffness, and surface topography have a strong influence on the cell–material interaction and require significant attention. Printing a soft hydrocolloid-based hydrogel reinforced with irregularly-shaped microporous biphasic calcium phosphate (BCP) particles (150–500 µm) is an alternative strategy for the acquisition of a complex network with good mechanical properties that could fulfill the needs of cell proliferation and regeneration. Three well-known hydrocolloids (sodium alginate, xanthan gum, and gelatin) have been combined with BCP particles to generate stable, homogenous, and printable solid dispersions. Through rheological assessment, it was determined that the crosslinking time, printing process parameters (infill density percentage and infill pattern), as well as BCP particle size and concentration all influence the stiffness of the printed matrices. Additionally, the swelling behavior on fresh and dehydrated 3D-printed structures was investigated, where it was observed that the BCP particle characteristics influenced the constructs’ water absorption, particle diffusion out of the matrix and degradability.
Collapse
|
28
|
Wang Q, Xie J, Zhou C, Lai W. Substrate stiffness regulates the differentiation profile and functions of osteoclasts via cytoskeletal arrangement. Cell Prolif 2021; 55:e13172. [PMID: 34953003 PMCID: PMC8780927 DOI: 10.1111/cpr.13172] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 02/05/2023] Open
Abstract
Objectives Aging and common diseases alter the stiffness of bone tissue, causing changes to the microenvironment of the mechanosensitive bone cells. Osteoclasts, the sole bone‐resorbing cells, play a vital role in bone remodeling. This study was performed to elucidate the mechanism through which osteoclasts sense and react to substrate stiffness signals. Materials and methods We fabricated polydimethylsiloxane (PDMS) substrates of different stiffness degrees for osteoclast formation progressed from osteoclast precursors including bone marrow‐derived macrophages (BMMs) and RAW264.7 monocytes. Osteoclast differentiation in response to the stiffness signals was determined by examining the cell morphology, fusion/fission activities, transcriptional profile, and resorption function. Cytoskeletal changes and mechanosensitive adhesion molecules were also assessed. Results Stiffer PDMS substrates accelerated osteoclast differentiation, firstly observed by variations in their morphology and fusion/fission activities. Upregulation of canonical osteoclast markers (Nfatc1, Acp5, Ctsk, Camk2a, Mmp9, Rela, and Traf6) and the fusion master regulator DC‐stamp were detected on stiffer substrates, with similar increases in their bone resorption functions. Additionally, the activation of cytoskeleton‐associated adhesion molecules, including fibronectin and integrin αvβ3, followed by biochemical signaling cascades of paxillin, FAK, PKC, and RhoA, was detected on the stiffer substrates. Conclusions This is the first study to provide evidence proving that extracellular substrate stiffness is a strong determinant of osteoclast differentiation and functions. Higher stiffness upregulated the differentiation profile and activity of osteoclasts, revealing the mechanical regulation of osteoclast activity in bone homeostasis and diseases.
Collapse
Affiliation(s)
- Qingxuan Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenli Lai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Panda AK, Basu B. Functionalized Fluoropolymer-Compatibilized Elastomeric Bilayer Composites for Osteochondral Repair: Unraveling the Role of Substrate Stiffness and Functionalities. ACS APPLIED BIO MATERIALS 2021; 4:8543-8558. [PMID: 35005914 DOI: 10.1021/acsabm.1c01021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The osteochondral lesions and osteoarthritis-related complications continue to be clinically relevant challenges to be addressed by the biomaterials community. Hydrogel-based scaffolds have been widely investigated to enhance osteochondral regeneration, but the inferior mechanical properties together with poor functional stability are the major constraints in their clinical translation. The development of osteochondral implants with natural tissue-mimicking mechanical properties remains largely unexplored. In this perspective, the present study demonstrates a strategy to develop a bilayer osteochondral implant with an elastically stiff composite (poly(vinylidene difluoride)-reinforced BaTiO3, PVDF/BT) and elastically compliant composite (maleic anhydride-functionalized PVDF/thermoplastic polyurethane/BaTiO3, m-PVDF/TPU/BT). The compositional variation in polymer composites allowed the elastic modulus of the hybrid bilayer construct to vary from ∼2 GPa to ∼90 MPa, which enabled a better understanding of the substrate-stiffness-dependent cellular behavior and maturation of preosteoblasts and chondrocytes. The cellular functionalities on PVDF-based polymer matrices have been benchmarked against ultrahigh-molecular-weight polyethylene (UHMWPE), which is clinically used for a wide spectrum of orthopedic applications. The increased alkaline phosphatase (ALP) activity, collagen synthesis, and matrix mineralization confirmed the early differentiation of preosteoblasts on the PVDF/BT matrix with subchondral bone-like mechanical properties. On the contrary, the upregulated chondrogenic functionalities were recorded on m-PVDF/TPU/BT with an elevated level of collagen content, glycosaminoglycans, and proteoglycans. Emphasis has been laid on probing the regulation of the osteochondral behavior using tailored substrate stiffness and functionalities using compatibilized fluoropolymer-based elastomeric composites. Taken together, the results of this work conclusively establish the efficacy of the hybrid bilayer composite with natural tissue-mimicking mechanical properties for the functional repair of osteochondral defects.
Collapse
Affiliation(s)
- Asish Kumar Panda
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore, Bangalore 560012, India
| | - Bikramjit Basu
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore, Bangalore 560012, India.,Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, Bangalore 560012, India
| |
Collapse
|
30
|
Kamaraj M, Sreevani G, Prabusankar G, Rath SN. Mechanically tunable photo-cross-linkable bioinks for osteogenic differentiation of MSCs in 3D bioprinted constructs. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112478. [PMID: 34857263 DOI: 10.1016/j.msec.2021.112478] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 11/24/2022]
Abstract
3D bioprinting technique renders a plausible solution to tissue engineering applications, mainly bone tissue regeneration, which could provide the microenvironment with desired physical, chemical, and mechanical properties. However, the mechanical and structural stability of current natural polymers is a critical issue in the fabrication of bone tissue-engineered scaffolds. To overcome these issues, we have developed 3D bioprintable semi-synthetic polymers derived from natural (sodium alginate, A) and synthetic (polyethylene glycol, PEG) biopolymers. In order to enhance the cross-linking properties and biocompatibility, we have functionalized these polymers with acrylate and methacrylate chemical moieties. These selected combination of natural and synthetic polymers improved the mechanical strength due to the synergistic effect of covalent as well as ionic bond formation in the hydrogel system, which is evident from the tested tensile data. Further, the feasibility of 3D bioprinting of acrylate and methacrylate functionalized PEG and hydrogels have been tested for the biocompatibility of the fabricated structures with human umbilical cord mesenchymal stem cells (UMSCs). Further, these bioprinted scaffolds were investigated for osteogenic differentiation of UMSCs in two types of culture conditions: namely, i) with osteoinduction media (with OIM), ii) without osteoinduction media (w/o OIM). We have examined the osteoinductivity of scaffolds with the activity of alkaline phosphatase (ALP) content, and significant changes in the ALP activity was observed with the stiffness of developed materials. The extent osteogenic differentiation was observed by alizarin red staining and reverse transcription PCR analysis. Elevated levels of ALP, RUNX2 and COL1 gene expression has been observed in without OIM samples on week 1 and week 3. Further, our study showed that the synthesized alginate methacrylate (AMA) without osteoinduction supplement with young's modulus of 0.34 MPa has a significant difference in ALP quantity and gene expression over the other reported literature. Thus, this work plays a pivotal role in the development of 3D bioprintable and photo-cross-linkable hydrogels in osteogenic differentiation of mesenchymal stem cells.
Collapse
Affiliation(s)
- Meenakshi Kamaraj
- Regenerative Medicine and Stem cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Gaddamedi Sreevani
- Regenerative Medicine and Stem cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Ganesan Prabusankar
- Organometallic Chemistry Laboratory, Department of Chemistry, Indian Institute of Technology Hyderabad, Telangana, India
| | - Subha Narayan Rath
- Regenerative Medicine and Stem cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India.
| |
Collapse
|
31
|
Subbiah R, Balbinot GDS, Athirasala A, Collares FM, Sereda G, Bertassoni LE. Nanoscale mineralization of cell-laden methacrylated gelatin hydrogels using calcium carbonate-calcium citrate core-shell microparticles. J Mater Chem B 2021; 9:9583-9593. [PMID: 34779469 DOI: 10.1039/d1tb01673c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Conventional biomaterials developed for bone regeneration fail to fully recapitulate the nanoscale structural organization and complex composition of the native bone microenvironment. Therefore, despite promoting osteogenic differentiation of stem cells, they fall short of providing the structural, biochemical, and mechanical stimuli necessary to drive osteogenesis for bone regeneration and function. To address this, we have recently developed a novel strategy to engineer bone-like tissue using a biomimetic approach to achieve rapid and controlled nanoscale mineralization of a cell-laden matrix in the presence of osteopontin, a non-collagenous protein, and a supersaturated solution of calcium and phosphate medium. Here, we build on this approach to engineer bone regeneration scaffolds comprising methacrylated gelatin (GelMA) hydrogels incorporated with calcium citrate core-shell microparticles as a sustained and reliable source of calcium ions for in situ mineralization. We demonstrate successful biomineralization of GelMA hydrogels by embedded calcium carbonate-calcium citrate core-shell microparticles with the resultant mineral chemistry, structure, and organization reminiscent of that of native bone. The biomimetic mineralization was further shown to promote osteogenic differentiation of encapsulated human mesenchymal stem cells even in the absence of other exogenous osteogenic induction factors. Ultimately, by combining the superior biological response engendered by biomimetic mineralization with the intrinsic tissue engineering advantages offered by GelMA, such as biocompatibility, biodegradability, and printability, we envision that our system offers great potential for bone regeneration efforts.
Collapse
Affiliation(s)
- Ramesh Subbiah
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
| | - Gabriela de Souza Balbinot
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Dental Materials, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Avathamsa Athirasala
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
| | - Fabricio Mezzomo Collares
- Department of Dental Materials, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Grigoriy Sereda
- Department of Chemistry, University of South Dakota, Vermillion, SD 57069, USA.
| | - Luiz E Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Center for Regenerative Medicine, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA.
| |
Collapse
|
32
|
Petzold J, Gentleman E. Intrinsic Mechanical Cues and Their Impact on Stem Cells and Embryogenesis. Front Cell Dev Biol 2021; 9:761871. [PMID: 34820380 PMCID: PMC8606660 DOI: 10.3389/fcell.2021.761871] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
Although understanding how soluble cues direct cellular processes revolutionised the study of cell biology in the second half of the 20th century, over the last two decades, new insights into how mechanical cues similarly impact cell fate decisions has gained momentum. During development, extrinsic cues such as fluid flow, shear stress and compressive forces are essential for normal embryogenesis to proceed. Indeed, both adult and embryonic stem cells can respond to applied forces, but they can also detect intrinsic mechanical cues from their surrounding environment, such as the stiffness of the extracellular matrix, which impacts differentiation and morphogenesis. Cells can detect changes in their mechanical environment using cell surface receptors such as integrins and focal adhesions. Moreover, dynamic rearrangements of the cytoskeleton have been identified as a key means by which forces are transmitted from the extracellular matrix to the cell and vice versa. Although we have some understanding of the downstream mechanisms whereby mechanical cues are translated into changes in cell behaviour, many of the signalling pathways remain to be defined. This review discusses the importance of intrinsic mechanical cues on adult cell fate decisions, the emerging roles of cell surface mechano-sensors and the cytoskeleton in enabling cells to sense its microenvironment, and the role of intracellular signalling in translating mechanical cues into transcriptional outputs. In addition, the contribution of mechanical cues to fundamental processes during embryogenesis such as apical constriction and convergent extension is discussed. The continued development of tools to measure the biomechanical properties of soft tissues in vivo is likely to uncover currently underestimated contributions of these cues to adult stem cell fate decisions and embryogenesis, and may inform on regenerative strategies for tissue repair.
Collapse
Affiliation(s)
- Jonna Petzold
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
33
|
A regulatory role of circRNA-miRNA-mRNA network in osteoblast differentiation. Biochimie 2021; 193:137-147. [PMID: 34742858 DOI: 10.1016/j.biochi.2021.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 02/07/2023]
Abstract
Osteoblast differentiation is an important process in skeletal development and bone remodelling. Serious bone diseases occur from any delay, defect, or imbalance in osteoblastic differentiation. Non-coding RNAs (ncRNAs) play a regulatory role in controlling the expression of proteins under physiological and pathological conditions via inhibiting mRNA translation or degrading mRNA. Circular RNAs (circRNAs) and microRNAs (miRNAs) are the long and small ncRNAs, respectively, which have been reported to regulate the expression of osteoblast marker genes directly or indirectly. Also, recent studies identified the regulatory mechanisms involving the crosstalk among circRNAs, miRNAs, and mRNAs during osteoblast differentiation. Understanding these regulatory mechanisms behind osteoblastic differentiation would help to diagnose or treat bone and bone-related disorders. Hence, the current review comprehensively discussed the regulatory relationship of circRNAs, miRNAs and mRNAs, and their functional role as circRNA-miRNA-mRNA axis in osteoblast differentiation.
Collapse
|
34
|
Yamada S, Yassin MA, Weigel T, Schmitz T, Hansmann J, Mustafa K. Surface activation with oxygen plasma promotes osteogenesis with enhanced extracellular matrix formation in three-dimensional microporous scaffolds. J Biomed Mater Res A 2021; 109:1560-1574. [PMID: 33675166 DOI: 10.1002/jbm.a.37151] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022]
Abstract
Various types of synthetic polyesters have been developed as biomaterials for tissue engineering. These materials commonly possess biodegradability, biocompatibility, and formability, which are preferable properties for bone regeneration. The major challenge of using synthetic polyesters is the result of low cell affinity due to their hydrophobic nature, which hinders efficient cell seeding and active cell dynamics. To improve wettability, plasma treatment is widely used in industry. Here, we performed surface activation with oxygen plasma to hydrophobic copolymers, poly(l-lactide-co-trimethylene carbonate), which were shaped in 2D films and 3D microporous scaffolds, and then we evaluated the resulting surface properties and the cellular responses of rat bone marrow stem cells (rBMSC) to the material. Using scanning electron microscopy and Fourier-transform infrared spectroscopy, we demonstrated that short-term plasma treatment increased nanotopographical surface roughness and wettability with minimal change in surface chemistry. On treated surfaces, initial cell adhesion and elongation were significantly promoted, and seeding efficiency was improved. In an osteoinductive environment, rBMSC on plasma-treated scaffolds exhibited accelerated osteogenic differentiation with osteogenic markers including RUNX2, osterix, bone sialoprotein, and osteocalcin upregulated, and a greater amount of collagen matrix and mineral deposition were found. This study shows the utility of plasma surface activation for polymeric scaffolds in bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Mohammed A Yassin
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Tobias Weigel
- Chair of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
- Translational Center Regenerative Therapies, Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
| | - Tobias Schmitz
- Chair of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
| | - Jan Hansmann
- Chair of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
- Translational Center Regenerative Therapies, Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
- Department Electrical Engineering, University for Applied Sciences Würzburg/Schweinfurt, Schweinfurt, Germany
| | - Kamal Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
35
|
El-Rashidy AA, El Moshy S, Radwan IA, Rady D, Abbass MMS, Dörfer CE, Fawzy El-Sayed KM. Effect of Polymeric Matrix Stiffness on Osteogenic Differentiation of Mesenchymal Stem/Progenitor Cells: Concise Review. Polymers (Basel) 2021; 13:2950. [PMID: 34502988 PMCID: PMC8434088 DOI: 10.3390/polym13172950] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 01/23/2023] Open
Abstract
Mesenchymal stem/progenitor cells (MSCs) have a multi-differentiation potential into specialized cell types, with remarkable regenerative and therapeutic results. Several factors could trigger the differentiation of MSCs into specific lineages, among them the biophysical and chemical characteristics of the extracellular matrix (ECM), including its stiffness, composition, topography, and mechanical properties. MSCs can sense and assess the stiffness of extracellular substrates through the process of mechanotransduction. Through this process, the extracellular matrix can govern and direct MSCs' lineage commitment through complex intracellular pathways. Hence, various biomimetic natural and synthetic polymeric matrices of tunable stiffness were developed and further investigated to mimic the MSCs' native tissues. Customizing scaffold materials to mimic cells' natural environment is of utmost importance during the process of tissue engineering. This review aims to highlight the regulatory role of matrix stiffness in directing the osteogenic differentiation of MSCs, addressing how MSCs sense and respond to their ECM, in addition to listing different polymeric biomaterials and methods used to alter their stiffness to dictate MSCs' differentiation towards the osteogenic lineage.
Collapse
Affiliation(s)
- Aiah A. El-Rashidy
- Biomaterials Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt;
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
| | - Sara El Moshy
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Israa Ahmed Radwan
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Dina Rady
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Marwa M. S. Abbass
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany;
| | - Karim M. Fawzy El-Sayed
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany;
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
36
|
Calcium silicate-based cements cause environmental stiffness and show diverse potential to induce osteogenesis in human osteoblastic cells. Sci Rep 2021; 11:16784. [PMID: 34408247 PMCID: PMC8373887 DOI: 10.1038/s41598-021-96353-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 08/09/2021] [Indexed: 01/12/2023] Open
Abstract
Calcium silicate-based cements differ markedly in their radiopacifiers and the presence of calcium sulfate, aluminates, carbonates and other components that can affect their biological properties. This study aimed to compare the biological properties of six calcium silicate cements in human osteoblastic cell culture (Saos-2 cells): Bio-C Repair (Bio-C), PBS HP (PBS-HP), Biodentine (Biodentine), MTA Repair HP (MTA-HP), NeoMTA Plus (NeoMTA-P), and ProRoot MTA (ProRoot). After exposure to these materials, the cells were analyzed by MTT, wound healing, cell migration, and alkaline phosphatase activity (ALP) assays, real-time PCR (qPCR) analysis of the osteogenesis markers (osteocalcin or bone gamma-carboxyglutamate protein, BGLAP; alkaline phosphatase, ALPL; bone sialoprotein or secreted phosphoprotein 1, BNSP), and alizarin red staining (ARS). Curiously, the migration rates were low 24-48 h after exposure to the materials, despite the cells showing ideal rates of viability. The advanced and intermediate cell differentiation markers BGLAP and BNSP were overexpressed in the Bio-C, MTA-HP, and ProRoot groups. Only the Biodentine group showed ALPL overexpression, a marker of initial differentiation. However, the enzymatic activity was high in all groups except Biodentine. The mineralization area was significantly large in the NeoMTA-P, ProRoot, PBS-HP, MTA-HP, and Bio-C groups. The results showed that cellular environmental stiffness, which impairs cell mobility and diverse patterns of osteogenesis marker expression, is a consequence of cement exposure. Environmental stiffness indicates chemical and physical stimuli in the microenvironment; for instance, the release of cement compounds contributes to calcium phosphate matrix formation with diverse stiffnesses, which could be essential or detrimental for the migration and differentiation of osteoblastic cells. Cells exposed to Bio-C, PBS-HP, ProRoot, NeoMTA-P, and MTA-HP seemed to enter the advanced or intermediate differentiation phases early, which is indicative of the diverse potential of cements to induce osteogenesis. Cements that quickly stimulate osteoblast differentiation may be ideal for reparative and regenerative purposes since they promptly lead to dentin or bone deposition.
Collapse
|
37
|
Stiffness Regulates the Morphology, Adhesion, Proliferation, and Osteogenic Differentiation of Maxillary Schneiderian Sinus Membrane-Derived Stem Cells. Stem Cells Int 2021; 2021:8868004. [PMID: 34306097 PMCID: PMC8285206 DOI: 10.1155/2021/8868004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 05/06/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022] Open
Abstract
Recent studies, which aim to optimize maxillary sinus augmentation, have paid significant attention exploring osteogenic potential of maxillary Schneiderian sinus membrane-derived cells (MSSM-derived cells). However, it remains unclear that how MSSM-derived cells could respond to niche's biomechanical properties. Herein, this study investigated the possible effects of substrate stiffness on rMSSM-derived stem cell fate. Initially, rMSSM-derived stem cells with multiple differentiation potential were successfully obtained. We then fabricated polyacrylamide substrates with varied stiffness ranging from 13 to 68 kPa to modulate the mechanical environment of rMSSM-derived stem cells. A larger cell spreading area and increased proliferation of rMSSM-derived stem cells were found on the stiffer substrates. Similarly, cells became more adhesive as their stiffness increased. Furthermore, the higher stiffness facilitated osteogenic differentiation of rMSSM-derived stem cells. Overall, our results indicated that increase in stiffness could mediate behaviors of rMSSM-derived stem cells, which may serve as a guide in future research to design novel biomaterials for maxillary sinus augmentation.
Collapse
|
38
|
Osmond MJ, Krebs MD. Tunable chitosan-calcium phosphate composites as cell-instructive dental pulp capping agents. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1450-1465. [PMID: 33941040 DOI: 10.1080/09205063.2021.1925390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Dental cavities are the most prevalent, preventable disease worldwide providing a need for robust treatment options to restore both the form and function of decaying teeth. Here is a presentation of a possible regenerative pulp capping agent that can serve to restore tooth function while regenerating healthy dentin tissue over a long period of time. To achieve this goal a material needs to crosslink quickly, be structurally rigid, and support the proliferation and differentiation of stem cells contained within the dental pulp. In this study, calcium phosphate nanoparticles were embedded in polymer hydrogels of carboxymethyl-chitosan and a diglycidyl ether. The particle size, surface, and mechanical properties of these materials were characterized. These composites have moduli up to 3 MPa, support the culture of dental pulp stem cells more than 3 weeks and exhibit osteogenic potential even without osteogenic media. These composites demonstrate a promising potential as the next generation of pulp capping materials.
Collapse
Affiliation(s)
- Matthew J Osmond
- Department of Chemical & Biological Engineering, Colorado School of Mines, Golden, CO, USA
| | - Melissa D Krebs
- Department of Chemical & Biological Engineering, Colorado School of Mines, Golden, CO, USA
| |
Collapse
|
39
|
Osteogenic Potential of Mesenchymal Stem Cells from Adipose Tissue, Bone Marrow and Hair Follicle Outer Root Sheath in a 3D Crosslinked Gelatin-Based Hydrogel. Int J Mol Sci 2021; 22:ijms22105404. [PMID: 34065598 PMCID: PMC8161179 DOI: 10.3390/ijms22105404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 11/23/2022] Open
Abstract
Bone transplantation is regarded as the preferred therapy to treat a variety of bone defects. Autologous bone tissue is often lacking at the source, and the mesenchymal stem cells (MSCs) responsible for bone repair mechanisms are extracted by invasive procedures. This study explores the potential of autologous mesenchymal stem cells derived from the hair follicle outer root sheath (MSCORS). We demonstrated that MSCORS have a remarkable capacity to differentiate in vitro towards the osteogenic lineage. Indeed, when combined with a novel gelatin-based hydrogel called Osteogel, they provided additional osteoinductive cues in vitro that may pave the way for future application in bone regeneration. MSCORS were also compared to MSCs from adipose tissue (ADMSC) and bone marrow (BMMSC) in a 3D Osteogel model. We analyzed gel plasticity, cell phenotype, cell viability, and differentiation capacity towards the osteogenic lineage by measuring alkaline phosphatase (ALP) activity, calcium deposition, and specific gene expression. The novel injectable hydrogel filled an irregularly shaped lesion in a porcine wound model displaying high plasticity. MSCORS in Osteogel showed a higher osteo-commitment in terms of calcium deposition and expression dynamics of OCN, BMP2, and PPARG when compared to ADMSC and BMMSC, whilst displaying comparable cell viability and ALP activity. In conclusion, autologous MSCORS combined with our novel gelatin-based hydrogel displayed a high capacity for differentiation towards the osteogenic lineage and are acquired by non-invasive procedures, therefore qualifying as a suitable and expandable novel approach in the field of bone regeneration therapy.
Collapse
|
40
|
Chen Y, Jia D, Wang Q, Sun Y, Rao Z, Lei X, Zhao J, Zeng K, Xu Z, Ming J. Promotion of the anticancer activity of curcumin based on a metal-polyphenol networks delivery system. Int J Pharm 2021; 602:120650. [PMID: 33957265 DOI: 10.1016/j.ijpharm.2021.120650] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/14/2021] [Accepted: 04/22/2021] [Indexed: 12/30/2022]
Abstract
Curcumin (Cur), a hydrophobic active pharmaceutical ingredient with high anticancer activity, has poor water solubility and low bioavailability. Although many delivery systems have been developed to improve their bioavailability, some limitation such as low drug loading efficiency and poor stability are still remained. The metal-polyphenol networks (MPNs) delivery system designed in this subject solved above problems and effectively improved the anticancer activity of Cur. The synthesized Cur@EGCG-Fe(III) is consisting of epigallocatechin gallate (EGCG), iron chloride (FeCl3) and Cur, and the well-designed structure endow Cur@EGCG-Fe(III) high loading efficiency, good water solubility and stability. After the Cur@EGCG-Fe(III) nanoparticles were internalized by MCF-7 cells, the Cur could be released in endo/lysosomal microenvironment (pH = 5.0), and the Cur delivery in the deep tumor could be realized. The distribution of Cur@EGCG-Fe(III) in MCF-7 cells was analyzed by laser confocal, and Cur@EGCG-Fe(III) could effectively deliver more Cur into MCF-7 cells in comparison with free Cur. In addition, the results of flow cytometry and western blot further indicated that Cur@EGCG-Fe(III) had a stronger ability to induce apoptosis than free Cur. Transwell cell migration and invasion experiments showed that Cur and EGCG-Fe(III) had a synergistic effect in inhibiting MCF-7 cell migration and invasion. In vitro hemolysis and in vivo experiments showed that the Cur@EGCG-Fe(III) had negligible effect on the blood environment and a great tumor-inhibition efficacy, indicating that the MPNs delivery system had a good blood compatibility and antitumor activity. Our results indicated that MPNs-coated Cur nanoparticle could be a new form of Cur delivery system for anticancer application.
Collapse
Affiliation(s)
- Yuanyuan Chen
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Die Jia
- School of Materials and Energy, Southwest University, Chongqing 400715, People's Republic of China
| | - Qiming Wang
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Yueru Sun
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Zhenan Rao
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Xiaojuan Lei
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Jichun Zhao
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China
| | - Kaifang Zeng
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China; Research Center of Food Storage & Logistics, Southwest University, Chongqing 400715, People's Republic of China
| | - Zhigang Xu
- School of Materials and Energy, Southwest University, Chongqing 400715, People's Republic of China
| | - Jian Ming
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China; Research Center of Food Storage & Logistics, Southwest University, Chongqing 400715, People's Republic of China.
| |
Collapse
|
41
|
Gwon Y, Park S, Kim W, Han T, Kim H, Kim J. Radially patterned transplantable biodegradable scaffolds as topographically defined contact guidance platforms for accelerating bone regeneration. J Biol Eng 2021; 15:12. [PMID: 33752709 PMCID: PMC7986475 DOI: 10.1186/s13036-021-00263-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The healing of large critical-sized bone defects remains a clinical challenge in modern orthopedic medicine. The current gold standard for treating critical-sized bone defects is autologous bone graft; however, it has critical limitations. Bone tissue engineering has been proposed as a viable alternative, not only for replacing the current standard treatment, but also for producing complete regeneration of bone tissue without complex surgical treatments or tissue transplantation. In this study, we proposed a transplantable radially patterned scaffold for bone regeneration that was defined by capillary force lithography technology using biodegradable polycaprolactone polymer. RESULTS The radially patterned transplantable biodegradable scaffolds had a radial structure aligned in a central direction. The radially aligned pattern significantly promoted the recruitment of host cells and migration of osteoblasts into the defect site. Furthermore, the transplantable scaffolds promoted regeneration of critical-sized bone defects by inducing cell migration and differentiation. CONCLUSIONS Our findings demonstrated that topographically defined radially patterned transplantable biodegradable scaffolds may have great potential for clinical application of bone tissue regeneration.
Collapse
Affiliation(s)
- Yonghyun Gwon
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sunho Park
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Woochan Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Taeseong Han
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyoseong Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jangho Kim
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea.
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
42
|
Hsu FY, Chen JJ, Sung WC, Hwang PA. Preparation of a Fucoidan-Grafted Hyaluronan Composite Hydrogel for the Induction of Osteoblast Differentiation in Osteoblast-Like Cells. MATERIALS (BASEL, SWITZERLAND) 2021; 14:1168. [PMID: 33801348 PMCID: PMC7958341 DOI: 10.3390/ma14051168] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 11/17/2022]
Abstract
A suitable bone substitute is necessary in bone regenerative medicine. Hyaluronan (HA) has excellent biocompatibility and biodegradability and is widely used in tissue engineering. Additionally, research on fucoidan (Fu), a fucose- and sulfate-rich polysaccharide from brown seaweed, for the promotion of bone osteogenic differentiation has increased exponentially. In this study, HA and Fu were functionalized by grafting methacrylic groups onto the backbone of the chain. Methacrylate-hyaluronan (MHA) and methacrylate-fucoidan (MFu) were characterized by FTIR and 1H NMR spectroscopy to confirm functionalization. The degrees of methacrylation (DMs) of MHA and MFu were 9.2% and 98.6%, respectively. Furthermore, we evaluated the mechanical properties of the hydrogels formed from mixtures of photo-crosslinkable MHA (1%) with varying concentrations of MFu (0%, 0.5%, and 1%). There were no changes in the hardness values of the hydrogels, but the elastic modulus decreased upon the addition of MFu, and these mechanical properties were not significantly different with or without preosteoblastic MG63 cell culture for up to 28 days. Furthermore, the cell morphologies and viabilities were not significantly different after culture with the MHA, MHA-MFu0.5, or MHA-MFu1.0 hydrogels, but the specific activity and mineralization of alkaline phosphatase (ALP) were significantly higher in the MHA-MFu1.0 hydrogel group compared to the other hydrogels. Hence, MHA-MFu composite hydrogels are potential bone graft materials that can provide a flexible structure and favorable niche for inducing bone osteogenic differentiation.
Collapse
Affiliation(s)
- Fu-Yin Hsu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City 20224, Taiwan; (F.-Y.H.); (J.-J.C.)
| | - Jheng-Jie Chen
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City 20224, Taiwan; (F.-Y.H.); (J.-J.C.)
| | - Wen-Chieh Sung
- Department of Food Science, National Taiwan Ocean University, Keelung City 20224, Taiwan;
| | - Pai-An Hwang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City 20224, Taiwan; (F.-Y.H.); (J.-J.C.)
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung City 20224, Taiwan
| |
Collapse
|
43
|
Molecular Mechanisms of Topography Sensing by Osteoblasts: An Update. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11041791] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Bone is a specialized tissue formed by different cell types and a multiscale, complex mineralized matrix. The architecture and the surface chemistry of this microenvironment can be factors of considerable influence on cell biology, and can affect cell proliferation, commitment to differentiation, gene expression, matrix production and/or composition. It has been shown that osteoblasts encounter natural motifs in vivo, with various topographies (shapes, sizes, organization), and that cell cultures on flat surfaces do not reflect the total potential of the tissue. Therefore, studies investigating the role of topographies on cell behavior are important in order to better understand the interaction between cells and surfaces, to improve osseointegration processes in vivo between tissues and biomaterials, and to find a better topographic surface to enhance bone repair. In this review, we evaluate the main available data about surface topographies, techniques for topographies’ production, mechanical signal transduction from surfaces to cells and the impact of cell–surface interactions on osteoblasts or preosteoblasts’ behavior.
Collapse
|
44
|
Neubauer VJ, Döbl A, Scheibel T. Silk-Based Materials for Hard Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2021; 14:674. [PMID: 33535662 PMCID: PMC7867174 DOI: 10.3390/ma14030674] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023]
Abstract
Hard tissues, e.g., bone, are mechanically stiff and, most typically, mineralized. To design scaffolds for hard tissue regeneration, mechanical, physico-chemical and biological cues must align with those found in the natural tissue. Combining these aspects poses challenges for material and construct design. Silk-based materials are promising for bone tissue regeneration as they fulfill several of such necessary requirements, and they are non-toxic and biodegradable. They can be processed into a variety of morphologies such as hydrogels, particles and fibers and can be mineralized. Therefore, silk-based materials are versatile candidates for biomedical applications in the field of hard tissue engineering. This review summarizes silk-based approaches for mineralized tissue replacements, and how to find the balance between sufficient material stiffness upon mineralization and cell survival upon attachment as well as nutrient supply.
Collapse
Affiliation(s)
- Vanessa J. Neubauer
- Lehrstuhl Biomaterialien, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Prof.-Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany; (V.J.N.); (A.D.)
| | - Annika Döbl
- Lehrstuhl Biomaterialien, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Prof.-Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany; (V.J.N.); (A.D.)
| | - Thomas Scheibel
- Lehrstuhl Biomaterialien, Fakultät für Ingenieurwissenschaften, Universität Bayreuth, Prof.-Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany; (V.J.N.); (A.D.)
- Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayerisches Polymerinstitut (BPI), Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuther Materialzentrum (BayMAT), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| |
Collapse
|
45
|
Areevijit K, Dhanesuan N, Luckanagul JA, Rungsiyanont S. Biocompatibility study of modified injectable hyaluronic acid hydrogel with mannitol/BSA to alveolar bone cells. J Biomater Appl 2020; 35:1294-1303. [PMID: 33148100 DOI: 10.1177/0885328220971746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The quality and quantity of bone are crucial to the success of dental implant treatment. Recently, bone grafting materials have reached some limitations. This study aimed to evaluate the biocompatibility of novel drug delivery material, injectable methacrylated hyaluronic acid hydrogel incorporated with different ratios of mannitol and BSA (Man/BSA MeHA), to human alveolar bone cells. The three-dimensionally encapsulated cell culture was evaluated with the resazurin cell viability test, alkaline phosphatase activity assay, immunohistochemistry test for collagen type-I synthesis, and cell morphology. The results showed that the encapsulated cells were viable in all four ratios of Man/BSA MeHA hydrogel and the average metabolic rate was not less than the control group. The morphology test showed round shape cells at the upper portion of the hydrogel and fibroblast-like or polygonal shape at the lower portion of hydrogel next to the culture plate. All four groups could express enzyme alkaline phosphatase and collagen type-I. In conclusion, four ratios of Man/BSA MeHA hydrogel were biocompatible with primary human alveolar bone cells.
Collapse
Affiliation(s)
- Kwanhatai Areevijit
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, Srinakharinwirot University, Bangkok, Thailand
| | - Nirada Dhanesuan
- Department of Stomatology, Faculty of Dentistry, Srinakharinwirot University, Bangkok, Thailand
| | - Jittima Amie Luckanagul
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences/Biomaterial Engineering for Medical and Health Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Sorasun Rungsiyanont
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, Srinakharinwirot University, Bangkok, Thailand
| |
Collapse
|
46
|
Mieczkowska A, Millar P, Chappard D, Gault VA, Mabilleau G. Dapagliflozin and Liraglutide Therapies Rapidly Enhanced Bone Material Properties and Matrix Biomechanics at Bone Formation Site in a Type 2 Diabetic Mouse Model. Calcif Tissue Int 2020; 107:281-293. [PMID: 32642787 DOI: 10.1007/s00223-020-00720-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/29/2020] [Indexed: 12/16/2022]
Abstract
The aim of this study is to compare head-to-head the effects of dapagliflozin and liraglutide on bone strength and bone material properties in a pre-clinical model of diabetes-obesity. Combined low-dose streptozotocin and high fat feeding were employed in mice to promote obesity, insulin resistance, and hyperglycaemia. Mice were administered daily for 28 days with saline vehicle, 1 mg/kg dapagliflozin or 25 nmol/kg liraglutide. Bone strength was assessed by three-point bending and nanoindentation. Bone material properties were investigated by Fourier transform infrared microspectroscopy/imaging. Although diabetic controls presented with dramatic reductions in mechanical strength, no deterioration of bone microarchitecture was apparent. At the tissue level, significant alterations in phosphate/amide ratio, carbonate/phosphate ratio, tissue water content, crystal size index, collagen maturity and collagen glycation were observed and linked to alteration of matrix biomechanics. Dapagliflozin and liraglutide failed to improve bone strength by 3-point bending or bone microarchitecture during the 28-day-treatment period. At bone formation site, dapagliflozin enhanced phosphate/amide ratio, mineral maturity, and reduced tissue water content, crystal size index, and collagen glycation. Liraglutide had significant effects on phosphate/amide ratio, tissue water content, crystal size index, mature collagen crosslinks, collagen maturity, and collagen glycation. At bone formation site, both drugs modulated matrix biomechanics. This study highlighted that these two molecules are effective in improving bone material properties and modulating matrix biomechanics at bone formation site. This study also highlighted that the resulting effects on bone material properties are not identical between dapagliflozin and liraglutide and not only mediated by lower blood glucose.
Collapse
Affiliation(s)
- Aleksandra Mieczkowska
- Groupe Etude Remodelage Osseux et biomatériaux, GEROM, UPRES EA 4658, SFR ICAT 4208, Institut de Biologie en Santé, UNIV Angers, 4 rue larrey, 49933, Angers Cedex 09, France
| | - Paul Millar
- School of Biomedical Sciences, University of Ulster, Coleraine, Northern Ireland, UK
| | - Daniel Chappard
- Groupe Etude Remodelage Osseux et biomatériaux, GEROM, UPRES EA 4658, SFR ICAT 4208, Institut de Biologie en Santé, UNIV Angers, 4 rue larrey, 49933, Angers Cedex 09, France
- Service Commun d'Imagerie et Analyses Microscopiques, SCIAM, SFR ICAT 4208, Institut de Biologie en Santé, UNIV Angers, Angers, France
- Bone Pathology Unit, CHU Angers, 49933, Angers Cedex, France
| | - Victor A Gault
- School of Biomedical Sciences, University of Ulster, Coleraine, Northern Ireland, UK
| | - Guillaume Mabilleau
- Groupe Etude Remodelage Osseux et biomatériaux, GEROM, UPRES EA 4658, SFR ICAT 4208, Institut de Biologie en Santé, UNIV Angers, 4 rue larrey, 49933, Angers Cedex 09, France.
- Service Commun d'Imagerie et Analyses Microscopiques, SCIAM, SFR ICAT 4208, Institut de Biologie en Santé, UNIV Angers, Angers, France.
- Bone Pathology Unit, CHU Angers, 49933, Angers Cedex, France.
| |
Collapse
|
47
|
Palomino-Durand C, Lopez M, Marchandise P, Martel B, Blanchemain N, Chai F. Chitosan/Polycyclodextrin (CHT/PCD)-Based Sponges Delivering VEGF to Enhance Angiogenesis for Bone Regeneration. Pharmaceutics 2020; 12:pharmaceutics12090784. [PMID: 32825081 PMCID: PMC7557476 DOI: 10.3390/pharmaceutics12090784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/11/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Vascularization is one of the main challenges in bone tissue engineering (BTE). In this study, vascular endothelial growth factor (VEGF), known for its angiogenic effect, was delivered by our developed sponge, derived from a polyelectrolyte complexes hydrogel between chitosan (CHT) and anionic cyclodextrin polymer (PCD). This sponge, as a scaffold for growth factor delivery, was formed by freeze-drying a homogeneous CHT/PCD hydrogel, and thereafter stabilized by a thermal treatment. Microstructure, water-uptake, biodegradation, mechanical properties, and cytocompatibility of sponges were assessed. VEGF-delivery following incubation in medium was then evaluated by monitoring the VEGF-release profile and its bioactivity. CHT/PCD sponge showed a porous (open porosity of 87.5%) interconnected microstructure with pores of different sizes (an average pore size of 153 μm), a slow biodegradation (12% till 21 days), a high water-uptake capacity (~600% in 2 h), an elastic property under compression (elastic modulus of compression 256 ± 4 kPa), and a good cytocompatibility in contact with osteoblast and endothelial cells. The kinetic release of VEGF was found to exert a pro-proliferation and a pro-migration effect on endothelial cells, which are two important processes during scaffold vascularization. Hence, CHT/PCD sponges were promising vehicles for the delivery of growth factors in BTE.
Collapse
Affiliation(s)
- Carla Palomino-Durand
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
| | - Marco Lopez
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
| | - Pierre Marchandise
- ULR 4490–MABLab–Adiposité Médullaire et Os, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France;
- ULR 4490–MABLab–Adiposité Médullaire et Os, Univ. Littoral Côte d’Opale, 62200 Boulogne-sur-Mer, France
| | - Bernard Martel
- UMR 8207, UMET—Unité Matériaux et Transformations, Centre National de la Recherche Scientifique (CNRS), Institut National de la Recherche Agronomique (INRA), Ecole Nationale Supérieure de Chimie de Lille (ENSCL), University of Lille, 59655 Lille, France;
| | - Nicolas Blanchemain
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
| | - Feng Chai
- U1008 Controlled Drug Delivery Systems and Biomaterials, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Lille (CHU Lille), University of Lille, 59000 Lille, France; (C.P.-D.); (M.L.); (N.B.)
- Correspondence: ; Tel.: +33-320-626-997
| |
Collapse
|
48
|
Bai M, Cai L, Li X, Ye L, Xie J. Stiffness and topography of biomaterials dictate cell-matrix interaction in musculoskeletal cells at the bio-interface: A concise progress review. J Biomed Mater Res B Appl Biomater 2020; 108:2426-2440. [PMID: 32027091 DOI: 10.1002/jbm.b.34575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/30/2019] [Accepted: 01/19/2020] [Indexed: 02/05/2023]
Abstract
Mutually interacted musculoskeletal tissues work together within the physiological environment full of varieties of external stimulus. Consistent with the locomotive function of the tissues, musculoskeletal cells are remarkably mechanosensitive to the physical cues. Signals like extracellular matrix (ECM) stiffness, topography, and geometry can be sensed and transduced into intracellular signaling cascades to trigger a series of cell responses, including cell adhesion, cell phenotype maintenance, cytoskeletal reconstruction, and stem cell differentiation (Du et al., 2011; Murphy et al., 2014; Lv et al., 2015; Kim et al., 2016; Kumar et al., 2017). With the development of tissue engineering and regenerative medicine, the potent effects of ECM physical properties on cell behaviors at the cell-matrix interface are drawing much attention. To mimic the interaction between cell and its ECM physical properties, developing advanced biomaterials with desired characteristics which could achieve the biointerface between cells and the surrounded matrix close to the physiological conditions becomes a great hotspot. In this review, based on the current publications in the field of biointerfaces, we systematically summarized the significant roles of stiffness and topography on musculoskeletal cell behaviors. We hope to shed light on the importance of physical cues in musculoskeletal tissue engineering and provide up to date strategies towards the natural or artificial replication of physiological microenvironment.
Collapse
Affiliation(s)
- Mingru Bai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linyi Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Janßen S, Gach S, Kant S, Aveic S, Rütten S, Olschok S, Reisgen U, Fischer H. Enhanced osteogenic differentiation of human mesenchymal stromal cells as response to periodical microstructured Ti6Al4V surfaces. J Biomed Mater Res B Appl Biomater 2020; 108:2218-2226. [PMID: 31981406 DOI: 10.1002/jbm.b.34559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 12/10/2019] [Accepted: 01/06/2020] [Indexed: 12/31/2022]
Abstract
Titanium-based alloys, for example, Ti6Al4V, are frequently employed for load-bearing orthopedic and dental implants. Growth of new bone tissue and therefore osseointegration can be promoted by the implant's microtopography, which can lead to improved long-term stability of the implant. This study investigates the effect that an organized, periodical microstructure produced by an electron beam (EB) technique has on the viability, morphology, and osteogenic differentiation capacity of human mesenchymal stromal cells (hMSC) in vitro. The technique generates topographical features of 20 μm in height with varying distances of 80-240 μm. Applied alterations of the surface roughness and local alloy composition do not impair hMSC viability (>94%) or proliferation. A favorable growth of hMSC onto the structure peaks and well-defined focal adhesions of the analyzed cells to the electron beam microstructured surfaces is verified. The morphological adaptation of hMSC to the underlying topography is detected using a three-dimensional (3D) visualization. In addition to the morphological changes, an increase in the expression of osteogenic markers such as osteocalcin (up to 17-fold) and osteoprotegerin (up to sixfold) is observed. Taken together, these results imply that the proposed periodical microstucturing method could potentially accelerate and enhance osseointegration of titanium-based bone implants.
Collapse
Affiliation(s)
- Simon Janßen
- Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Stefan Gach
- Welding and Joining Institute, RWTH Aachen University, Aachen, Germany
| | - Sebastian Kant
- Molecular and Cellular Anatomy, RWTH Aachen University Hospital, Aachen, Germany
| | - Sanja Aveic
- Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany.,Neuroblastoma Laboratory, Paediatric Research Institute Città della Speranza, Padova, Italy
| | - Stephan Rütten
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Simon Olschok
- Welding and Joining Institute, RWTH Aachen University, Aachen, Germany
| | - Uwe Reisgen
- Welding and Joining Institute, RWTH Aachen University, Aachen, Germany
| | - Horst Fischer
- Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
50
|
Delgado-Ruiz RA, Calvo-Guirado JL, Romanos GE. Effects of occlusal forces on the peri-implant-bone interface stability. Periodontol 2000 2019; 81:179-193. [PMID: 31407438 DOI: 10.1111/prd.12291] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The occlusal forces and their influence on the initiation of peri-implant bone loss or their relationship with peri-implantitis have created discussion during the past 30 years given the discrepancies observed in clinical, animal, and finite element analysis studies. Beyond these contradictions, in the case of an osseointegrated implant, the occlusal forces can influence the implant-bone interface and the cells responsible for the bone remodeling in different ways that may result in the maintenance or loss of the osseointegration. This comprehensive review focuses on the information available about the forces transmitted through the implant-crown system to the implant-bone interface and the mechano-transduction phenomena responsible for the bone cells' behavior and their interactions. Knowledge of the basic molecular biology of the peri-implant bone would help clinicians to understand the complex phenomenon of occlusal forces and their effects on the implant-bone interface, and would allow better control of the negative effects of mechanical stresses, leading to therapy with fewer risks and complications.
Collapse
Affiliation(s)
- Rafael Arcesio Delgado-Ruiz
- Department of Prosthodontics and Digital Technology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Jose Luis Calvo-Guirado
- International Dentistry Research Cathedra, Faculty of Medicine and Dentistry, Universidad Catolica San Antonio De Murcia (UCAM), Murcia, Spain
| | - Georgios E Romanos
- Department of Periodontology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA.,Department of Oral Surgery and Implant Dentistry, Johann Wolfgang Goethe University, Frankfurt, Germany
| |
Collapse
|