1
|
Wakelin SH, Cobourn KD, Stirrat T, Sayah A, Sage K, Ryaby J, Sandhu FA. Fusion Rate of Biphasic Calcium Phosphate Bone Graft with Needle-Shaped Submicron Surface Topography in Interbody Lumbar Fusion for Degenerative Disc Disease: A Single-Center Retrospective Review. World Neurosurg 2025; 196:123759. [PMID: 39952405 DOI: 10.1016/j.wneu.2025.123759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Calcium phosphate bone grafts are emerging as alternatives to autologous bone grafts in lumbar spinal fusion. This study evaluates the 12-month fusion rate and clinical outcomes of lumbar interbody fusion using synthetic biphasic calcium phosphate with submicron needle-shaped surface topography (BCP<μm). METHODS A retrospective review identified patients who underwent lumbar interbody arthrodesis with BCP<μm, with or without autograft. Fusion was assessed by an independent neuroradiologist using a computed tomography alphanumeric classification based on bridging bone on 12-month postoperative computed tomography. Secondary outcomes included changes in visual analog scale, length of stay, and fusion of all treated levels. RESULTS Fifty-five patients with 93 treated levels were analyzed (average age: 62.78 years; body mass index: 28.73 kg/m2). Patients had an average of 3 comorbidities and a median of 2 levels fused. Procedures included anterior (52 levels), transforaminal (22 levels), and lateral lumbar interbody fusion (19 levels). Fusion occurred in 95.70% of levels without evidence of fixation failure. Fusion rates were unaffected by procedure type (P = 0.965) or supplemental autograft use (P = 1.00). Complications were reported in 4 patients. Six-month postoperative visual analog scale improved by a mean of 25.10 (P < 0.0001). CONCLUSIONS This study demonstrates a high fusion rate (95.70%) and low complication rate in a diverse patient population with multiple comorbidities, suggesting BCP<μm is a viable graft material for lumbar interbody fusion.
Collapse
Affiliation(s)
- Samuel H Wakelin
- Georgetown University School of Medicine, Washington, District of Columbia, USA.
| | - Kelsey D Cobourn
- Department of Neurosurgery, MedStar Georgetown University Hospital, Washington, District of Columbia, USA
| | - Thomas Stirrat
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - Anousheh Sayah
- Department of Neuroradiology, MedStar Georgetown University Hospital, Washington, District of Columbia, USA
| | | | | | - Faheem A Sandhu
- Department of Neurosurgery, MedStar Georgetown University Hospital, Washington, District of Columbia, USA
| |
Collapse
|
2
|
Milborne B, Arjuna A, Islam MT, Arafat A, Layfield R, Thompson A, Ahmed I. Yttrium-Enriched Phosphate Glass-Ceramic Microspheres for Bone Cancer Radiotherapy Treatment. ACS OMEGA 2024; 9:50933-50944. [PMID: 39758667 PMCID: PMC11696403 DOI: 10.1021/acsomega.4c02825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 09/16/2024] [Accepted: 10/14/2024] [Indexed: 01/07/2025]
Abstract
This study presents the development and characterization of high yttrium-content phosphate-based glass-ceramic microspheres for potential applications in bone cancer radiotherapy treatment. The microspheres produced via flame spheroidization, followed by sieving, revealed a lack of aggregation and a narrow size distribution (45-125 μm) achieved across different yttrium oxide to glass ratio samples. Energy dispersive X-ray (EDX) analysis showed a significant increase in yttrium content within the microspheres with increasing yttrium oxide to glass ratio samples, ranging from approximately 1-39 mol % for 10Y-50Y microspheres, respectively. Concurrently, a proportional decrease in the phosphate, calcium, and magnesium content was observed. Further EDX mapping showed a homogeneous distribution of all elements throughout the microspheres, indicating uniform composition. X-ray diffraction profiles confirmed the amorphous nature of the starting P40 glass microspheres, while yttrium-containing microspheres exhibited crystalline peaks corresponding to cubic and hexagonal Y2O3 and Y(PO4) phases, indicating the formation of glass-ceramic materials. Ion release studies revealed the reduction of all ion release rates from yttrium-containing microspheres compared with P40 microspheres. The pH of the surrounding media was also stable at approximately pH 7 over time, highlighting the chemical durability of the microspheres' produced. In vitro cytocompatibility studies demonstrated that both indirect and direct cell culture methods showed favorable cellular responses. The metabolic and alkaline phosphatase activity assays indicated comparable or enhanced cell responses on yttrium-containing microspheres compared to the initial P40 glass microspheres. Overall, these findings showed that significantly high yttrium-content phosphate glass-ceramic microspheres could be produced as versatile biomaterials offering potential applications for combined bone cancer radiotherapy treatment and bone regeneration.
Collapse
Affiliation(s)
- Ben Milborne
- Advanced
Materials Research Group, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Andi Arjuna
- Advanced
Materials Research Group, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, U.K.
- Faculty
of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia
| | - Md Towhidul Islam
- Advanced
Materials Research Group, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Abul Arafat
- School
of Engineering, University of Wolverhampton,
Telford Innovation Campus, Telford TF2 9NT, U.K.
| | - Robert Layfield
- School
of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2UH, U.K.
| | - Alexander Thompson
- Biodiscovery
Institute, Division of Cancer and Stem Cells, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Ifty Ahmed
- Advanced
Materials Research Group, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, U.K.
| |
Collapse
|
3
|
Somasundaram S, D F, Genasan K, Kamarul T, Raghavendran HRB. Implications of Biomaterials and Adipose-Derived Stem Cells in the Management of Calvarial Bone Defects. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024. [DOI: 10.1007/s40883-024-00358-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 08/25/2024] [Accepted: 09/13/2024] [Indexed: 01/03/2025]
|
4
|
Gao Q, Liu J, Wang M, Liu X, Jiang Y, Su J. Biomaterials regulates BMSCs differentiation via mechanical microenvironment. BIOMATERIALS ADVANCES 2024; 157:213738. [PMID: 38154401 DOI: 10.1016/j.bioadv.2023.213738] [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: 10/08/2023] [Revised: 12/11/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
Bone mesenchymal stem cells (BMSCs) are crucial for bone tissue regeneration, the mechanical microenvironment of hard tissues, including bone and teeth, significantly affects the osteogenic differentiation of BMSCs. Biomaterials may mimic the microenvironment of the extracellular matrix and provide mechanical signals to regulate BMSCs differentiation via inducing the secretion of various intracellular factors. Biomaterials direct the differentiation of BMSCs via mechanical signals, including tension, compression, shear, hydrostatic pressure, stiffness, elasticity, and viscoelasticity, which can be transmitted to cells through mechanical signalling pathways. Besides, biomaterials with piezoelectric effects regulate BMSCs differentiation via indirect mechanical signals, such as, electronic signals, which are transformed from mechanical stimuli by piezoelectric biomaterials. Mechanical stimulation facilitates achieving vectored stem cell fate regulation, while understanding the underlying mechanisms remains challenging. Herein, this review summarizes the intracellular factors, including translation factors, epigenetic modifications, and miRNA level, as well as the extracellular factor, including direct and indirect mechanical signals, which regulate the osteogenic differentiation of BMSCs. Besides, this review will also give a comprehensive summary about how mechanical stimuli regulate cellular behaviours, as well as how biomaterials promote the osteogenic differentiation of BMSCs via mechanical microenvironments. The cellular behaviours and activated signal pathways will give more implications for the design of biomaterials with superior properties for bone tissue engineering. Moreover, it will also provide inspiration for the construction of bone organoids which is a useful tool for mimicking in vivo bone tissue microenvironments.
Collapse
Affiliation(s)
- Qianmin Gao
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China
| | - Jinlong Liu
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China
| | - Xiangfei Liu
- Department of Orthopedics, Shanghai Zhongye Hospital, NO. 456 Chunlei Road, Shanghai 200941, PR China.
| | - Yingying Jiang
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China; Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, NO.1665 Kongjiang Road, Shanghai 200092, PR China.
| |
Collapse
|
5
|
Ghuloum FI, Stevens LA, Johnson CA, Riobo-Del Galdo NA, Amer MH. Towards modular engineering of cell signalling: Topographically-textured microparticles induce osteogenesis via activation of canonical hedgehog signalling. BIOMATERIALS ADVANCES 2023; 154:213652. [PMID: 37837904 DOI: 10.1016/j.bioadv.2023.213652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/19/2023] [Accepted: 10/03/2023] [Indexed: 10/16/2023]
Abstract
Polymer microparticles possess great potential as functional building blocks for advanced bottom-up engineering of complex tissues. Tailoring the three-dimensional architectural features of culture substrates has been shown to induce osteogenesis in mesenchymal stem cells in vitro, but the molecular mechanisms underpinning this remain unclear. This study proposes a mechanism linking the activation of Hedgehog signalling to the osteoinductive effect of surface-engineered, topographically-textured polymeric microparticles. In this study, mesenchymal progenitor C3H10T1/2 cells were cultured on smooth and dimpled poly(D,l-lactide) microparticles to assess differences in viability, cellular morphology, proliferation, and expression of a range of Hedgehog signalling components and osteogenesis-related genes. Dimpled microparticles induced osteogenesis and activated the Hedgehog signalling pathway relative to smooth microparticles and 2D-cultured controls without the addition of exogenous biochemical factors. We observed upregulation of the osteogenesis markers Runt-related transcription factor2 (Runx2) and bone gamma-carboxyglutamate protein 2 (Bglap2), as well as the Hedgehog signalling components, glioma associated oncogene homolog 1 (Gli1), Patched1 (Ptch1), and Smoothened (Smo). Treatment with the Smo antagonist KAAD-cyclopamine confirmed the involvement of Smo in Gli1 target gene activation, with a significant reduction in the expression of Gli1, Runx2 and Bglap2 (p ≤ 0.001) following KAAD-cyclopamine treatment. Overall, our study demonstrates the role of the topographical microenvironment in the modulation of Hedgehog signalling, highlighting the potential for tailoring substrate topographical design to offer cell-instructive 3D microenvironments. Topographically-textured microparticles allow the modulation of Hedgehog signalling in vitro without adding exogenous biochemical agonists, thereby eliminating potential confounding artefacts in high-throughput drug screening applications.
Collapse
Affiliation(s)
- Fatmah I Ghuloum
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom; Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Lee A Stevens
- Low Carbon Energy and Resources Technologies Research Group, Faculty of Engineering, University of Nottingham, UK
| | - Colin A Johnson
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Natalia A Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom; Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, UK
| | - Mahetab H Amer
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
6
|
LaGuardia JS, Shariati K, Bedar M, Ren X, Moghadam S, Huang KX, Chen W, Kang Y, Yamaguchi DT, Lee JC. Convergence of Calcium Channel Regulation and Mechanotransduction in Skeletal Regenerative Biomaterial Design. Adv Healthc Mater 2023; 12:e2301081. [PMID: 37380172 PMCID: PMC10615747 DOI: 10.1002/adhm.202301081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/20/2023] [Indexed: 06/30/2023]
Abstract
Cells are known to perceive their microenvironment through extracellular and intracellular mechanical signals. Upon sensing mechanical stimuli, cells can initiate various downstream signaling pathways that are vital to regulating proliferation, growth, and homeostasis. One such physiologic activity modulated by mechanical stimuli is osteogenic differentiation. The process of osteogenic mechanotransduction is regulated by numerous calcium ion channels-including channels coupled to cilia, mechanosensitive and voltage-sensitive channels, and channels associated with the endoplasmic reticulum. Evidence suggests these channels are implicated in osteogenic pathways such as the YAP/TAZ and canonical Wnt pathways. This review aims to describe the involvement of calcium channels in regulating osteogenic differentiation in response to mechanical loading and characterize the fashion in which those channels directly or indirectly mediate this process. The mechanotransduction pathway is a promising target for the development of regenerative materials for clinical applications due to its independence from exogenous growth factor supplementation. As such, also described are examples of osteogenic biomaterial strategies that involve the discussed calcium ion channels, calcium-dependent cellular structures, or calcium ion-regulating cellular features. Understanding the distinct ways calcium channels and signaling regulate these processes may uncover potential targets for advancing biomaterials with regenerative osteogenic capabilities.
Collapse
Affiliation(s)
- Jonnby S. LaGuardia
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Kaavian Shariati
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Meiwand Bedar
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Xiaoyan Ren
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Shahrzad Moghadam
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Kelly X. Huang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Wei Chen
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Youngnam Kang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Dean T. Yamaguchi
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Justine C. Lee
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
- Department of Orthopaedic Surgery, Los Angeles, CA, 90095, USA
- UCLA Molecular Biology Institute, Los Angeles, CA, 90095, USA
| |
Collapse
|
7
|
Shi H, Zhou K, Wang M, Wang N, Song Y, Xiong W, Guo S, Yi Z, Wang Q, Yang S. Integrating physicomechanical and biological strategies for BTE: biomaterials-induced osteogenic differentiation of MSCs. Theranostics 2023; 13:3245-3275. [PMID: 37351163 PMCID: PMC10283054 DOI: 10.7150/thno.84759] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/12/2023] [Indexed: 06/24/2023] Open
Abstract
Large bone defects are a major global health concern. Bone tissue engineering (BTE) is the most promising alternative to avoid the drawbacks of autograft and allograft bone. Nevertheless, how to precisely control stem cell osteogenic differentiation has been a long-standing puzzle. Compared with biochemical cues, physicomechanical stimuli have been widely studied for their biosafety and stability. The mechanical properties of various biomaterials (polymers, bioceramics, metal and alloys) become the main source of physicomechanical stimuli. By altering the stiffness, viscoelasticity, and topography of materials, mechanical stimuli with different strengths transmit into precise signals that mediate osteogenic differentiation. In addition, externally mechanical forces also play a critical role in promoting osteogenesis, such as compression stress, tensile stress, fluid shear stress and vibration, etc. When exposed to mechanical forces, mesenchymal stem cells (MSCs) differentiate into osteogenic lineages by sensing mechanical stimuli through mechanical sensors, including integrin and focal adhesions (FAs), cytoskeleton, primary cilium, ions channels, gap junction, and activating osteogenic-related mechanotransduction pathways, such as yes associated proteins (YAP)/TAZ, MAPK, Rho-GTPases, Wnt/β-catenin, TGFβ superfamily, Notch signaling. This review summarizes various biomaterials that transmit mechanical signals, physicomechanical stimuli that directly regulate MSCs differentiation, and the mechanical transduction mechanisms of MSCs. This review provides a deep and broad understanding of mechanical transduction mechanisms and discusses the challenges that remained in clinical translocation as well as the outlook for the future improvements.
Collapse
Affiliation(s)
- Huixin Shi
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Kaixuan Zhou
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Mingfeng Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Ning Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yiping Song
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Wei Xiong
- Department of Plastic Surgery, The First Affiliated Hospital of Medical College of Shihezi University, Shihezi, Xinjiang 832008, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhe Yi
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Qiang Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
| |
Collapse
|
8
|
Wang J, Li H, Fu S, Su Y. Porous BCP ceramics with nanoscale whisker structure accelerate bone regeneration by regulating inflammatory response. BIOMATERIALS ADVANCES 2023; 147:213313. [PMID: 36753873 DOI: 10.1016/j.bioadv.2023.213313] [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: 08/18/2022] [Revised: 01/07/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
Inflammation-induced by biomaterials is a critical event to determine the success and efficiency of tissue repair. Macrophages are a major population that participates the biomaterial induced inflammation. The response of macrophages depends on the characteristics of biomaterials, thus causing a cascade reaction in subsequent biological processes. In this study, porous biphase calcium phosphate (BCP) ceramics with the different surface structures were constructed to compare the effect of surface structure on bone generation potential, and further reveal the inflammation-involved mechanism. Our results demonstrated that macrophages on three ceramics showed distinct morphologies and spreading areas. The nanoscale whisker structure did induce more bone generation in the mice thigh muscle. The in vitro result revealed that nanoscale whisker structure could drive macrophage polarization towards M1-like phenotype, indicated by a higher expression of pro-inflammatory specific markers (iNOS and CCR7), and mass secretion of TNF-α. Further research indicated that additional TNF-α could promote the osteogenic differentiation of mesenchymal stem cells (MSCs). However, excess addition of TNF-α showed an opposite effect on the osteogenic differentiation of MSCs by initiating the NF-κB signaling pathway, which suppresses the osteogenesis process.
Collapse
Affiliation(s)
- Jing Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Huishan Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Shijia Fu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yangyang Su
- State Key Laboratory of Solidification Processing, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
9
|
Fainor M, Mahindroo S, Betz KR, Augustin J, Smith HE, Mauck RL, Gullbrand SE. A Tunable Calcium Phosphate Coating to Drive in vivo Osseointegration of Composite Engineered Tissues. Cells Tissues Organs 2023; 212:383-398. [PMID: 36966531 PMCID: PMC10616759 DOI: 10.1159/000528965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/30/2022] [Indexed: 06/18/2023] Open
Abstract
Varying degrees of hydroxyapatite (HA) surface functionalization have been implicated as the primary driver of differential osteogenesis observed in infiltrating cells. The ability to reliably create spatially controlled areas of mineralization in composite engineered tissues is of growing interest in the field, and the use of HA-functionalized biomaterials may provide a robust solution to this challenge. In this study, we successfully fabricated polycaprolactone salt-leached scaffolds with two levels of a biomimetic calcium phosphate coating to examine their effects on MSC osteogenesis. Longer duration coating in simulated body fluid (SBF) led to increased HA crystal nucleation within scaffold interiors as well as more robust HA crystal formation on scaffold surfaces. Ultimately, the increased surface stiffness of scaffolds coated in SBF for 7 days in comparison to scaffolds coated in SBF for 1 day led to more robust osteogenesis of MSCs in vitro without the assistance of osteogenic signaling molecules. This study also demonstrated that the use of SBF-based HA coatings can promote higher levels of osteogenesis in vivo. Finally, when incorporated as the endplate region of a larger tissue-engineered intervertebral disc replacement, HA coating did not induce mineralization in or promote cell migration out of neighboring biomaterials. Overall, these results verified tunable biomimetic HA coatings as a promising biomaterial modification to promote discrete regions of mineralization within composite engineered tissues.
Collapse
Affiliation(s)
- Matthew Fainor
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, (PA,) USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
| | - Sonal Mahindroo
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
- Department of Biology, St. Bonaventure University, St. Bonaventure, (NY,) USA
| | - Kerri R. Betz
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
- Department of Biology, Widener University, Chester, (PA,) USA
| | - Janai Augustin
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
- Department of Biomedical Engineering, City College of New York, New York City, (NY,) USA
| | - Harvey E. Smith
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, (PA,) USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
| | - Robert L. Mauck
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, (PA,) USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, (PA,) USA
| | - Sarah E. Gullbrand
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, (PA,) USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, (PA,) USA
| |
Collapse
|
10
|
Nie Z, Hu Z, Guo X, Xiao Y, Liu X, de Bruijn JD, Bao C, Yuan H. Genesis of osteoclasts on calcium phosphate ceramics and their role in material-induced bone formation. Acta Biomater 2023; 157:625-638. [PMID: 36371003 DOI: 10.1016/j.actbio.2022.11.005] [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: 07/10/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 11/10/2022]
Abstract
Innate immune responses play important roles in material-induced bone formation and such roles were further explored in the current study with an emphasis on M2 macrophages and osteoclastogenesis. With the presence of M-CSF and RANKL, M0 macrophages from FVB mouse bone marrow-derived monocytes (BMMs) fused to osteoclasts with both M2 marker and osteoclast marker at day 5, and such osteoclast formation at day 5 was enhanced when the cells were treated with IL-4 at day 3. With IL-4 treatment alone for 24 h, M0 polarized into M2 macrophages. Conditioned medium of M2 macrophages enhanced osteogenic differentiation of MC3T3-E1 (pre-osteoblasts) while osteoclast conditioned medium enhanced osteogenic differentiation of CRL-12424 (osteogenic precursors). TCPs (a typical osteoinductive material) supported M2 macrophage polarization at day 4 and osteoclast formation at day 5, while TCPb (a typical non-osteoinductive material) was less effective. Moreover, osteoclasts formed on TCPs produced osteogenic factors including S1P, Wnt10B and BMP-6, resulting osteogenic differentiation of CRL-12424 cells. Similar to in vitro testing, TCPs favored M2 macrophage polarization followed by the formation of osteoclasts in vivo, as compared to TCPb. The overall data provided evidence of a coupling between M2 macrophages, osteoclasts and material-induced bone formation: osteoclasts formed from M2 macrophages secrete osteogenic cytokines to induce osteogenic differentiation of osteogenic precursor cells to finally form bone. The current findings outlined a biological mechanism of material-induced bone formation and further rationalized the use of osteoinductive materials for bone regeneration. STATEMENT OF SIGNIFICANCE: This paper provides evidence for finding out the relationship between M2 macrophages, osteoclasts and osteogenesis in material-induced bone formation. It suggested that osteoinductive materials enhanced macrophage polarization to M2 macrophages which fuses to osteoclasts, osteoclasts subsequently secret osteogenic cytokines to differentiate finally osteogenic precursors to form bone in osteoinductive materials. The data supports scientifically the superiority of osteoinductive materials for bone regeneration in clinics.
Collapse
Affiliation(s)
- Zhangling Nie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Zhiqiao Hu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Xiaodong Guo
- National Center of Stomatology & National Clinical Research Center for Oral Disease, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yu Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Xian Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Joost D de Bruijn
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, MB Bilthoven 3723, the Netherlands; School of Engineering and Materials Science, Queen Mary University of London, UK
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China.
| | - Huipin Yuan
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, MB Bilthoven 3723, the Netherlands; Huipin Yuan's Lab, Sichuan, China.
| |
Collapse
|
11
|
Galván-Chacón V, de Melo Pereira D, Vermeulen S, Yuan H, Li J, Habibović P. Decoupling the role of chemistry and microstructure in hMSCs response to an osteoinductive calcium phosphate ceramic. Bioact Mater 2023; 19:127-138. [PMID: 35475029 PMCID: PMC9014318 DOI: 10.1016/j.bioactmat.2022.03.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- V.P. Galván-Chacón
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, the Netherlands
| | - D. de Melo Pereira
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, the Netherlands
| | - S. Vermeulen
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, the Netherlands
| | - H. Yuan
- Kuros Biosciences BV, 3723 MB, Bilthoven, the Netherlands
| | - J. Li
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, the Netherlands
| | - P. Habibović
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, the Netherlands
- Corresponding author. Maastricht University, MERLN Institute, Universiteitsingel 40, 6229ER, Maastricht, the Netherlands.
| |
Collapse
|
12
|
Calcium Phosphate-Based Biomaterials for Bone Repair. J Funct Biomater 2022; 13:jfb13040187. [PMID: 36278657 PMCID: PMC9589993 DOI: 10.3390/jfb13040187] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
Traumatic, tumoral, and infectious bone defects are common in clinics, and create a big burden on patient's families and society. Calcium phosphate (CaP)-based biomaterials have superior properties and have been widely used for bone defect repair, due to their similarities to the inorganic components of human bones. The biological performance of CaPs, as a determining factor for their applications, are dependent on their physicochemical properties. Hydroxyapatite (HAP) as the most thermally stable crystalline phase of CaP is mostly used in the form of ceramics or composites scaffolds with polymers. Nanostructured CaPs with large surface areas are suitable for drug/gene delivery systems. Additionally, CaP scaffolds with hierarchical nano-/microstructures have demonstrated excellent ability in promoting bone regeneration. This review focuses on the relationships and interactions between the physicochemical/biological properties of CaP biomaterials and their species, sizes, and morphologies in bone regeneration, including synthesis strategies, structure control, biological behavior, and the mechanisms of CaP in promoting osteogenesis. This review will be helpful for scientists and engineers to further understand CaP-based biomaterials (CaPs), and be useful in developing new high-performance biomaterials for bone repair.
Collapse
|
13
|
Guimarães CF, Marques AP, Reis RL. Pushing the Natural Frontier: Progress on the Integration of Biomaterial Cues toward Combinatorial Biofabrication and Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105645. [PMID: 35419887 DOI: 10.1002/adma.202105645] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/14/2022] [Indexed: 06/14/2023]
Abstract
The engineering of fully functional, biological-like tissues requires biomaterials to direct cellular events to a near-native, 3D niche extent. Natural biomaterials are generally seen as a safe option for cell support, but their biocompatibility and biodegradability can be just as limited as their bioactive/biomimetic performance. Furthermore, integrating different biomaterial cues and their final impact on cellular behavior is a complex equation where the outcome might be very different from the sum of individual parts. This review critically analyses recent progress on biomaterial-induced cellular responses, from simple adhesion to more complex stem cell differentiation, looking at the ever-growing possibilities of natural materials modification. Starting with a discussion on native material formulation and the inclusion of cell-instructive cues, the roles of shape and mechanical stimuli, the susceptibility to cellular remodeling, and the often-overlooked impact of cellular density and cell-cell interactions within constructs, are delved into. Along the way, synergistic and antagonistic combinations reported in vitro and in vivo are singled out, identifying needs and current lessons on the development of natural biomaterial libraries to solve the cell-material puzzle efficiently. This review brings together knowledge from different fields envisioning next-generation, combinatorial biomaterial development toward complex tissue engineering.
Collapse
Affiliation(s)
- Carlos F Guimarães
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra P Marques
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
14
|
Bohner M, Maazouz Y, Ginebra MP, Habibovic P, Schoenecker JG, Seeherman H, van den Beucken JJ, Witte F. Sustained local ionic homeostatic imbalance caused by calcification modulates inflammation to trigger heterotopic ossification. Acta Biomater 2022; 145:1-24. [PMID: 35398267 DOI: 10.1016/j.actbio.2022.03.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022]
Abstract
Heterotopic ossification (HO) is a condition triggered by an injury leading to the formation of mature lamellar bone in extraskeletal soft tissues. Despite being a frequent complication of orthopedic and trauma surgery, brain and spinal injury, the etiology of HO is poorly understood. The aim of this study is to evaluate the hypothesis that a sustained local ionic homeostatic imbalance (SLIHI) created by mineral formation during tissue calcification modulates inflammation to trigger HO. This evaluation also considers the role SLIHI could play for the design of cell-free, drug-free osteoinductive bone graft substitutes. The evaluation contains five main sections. The first section defines relevant concepts in the context of HO and provides a summary of proposed causes of HO. The second section starts with a detailed analysis of the occurrence and involvement of calcification in HO. It is followed by an explanation of the causes of calcification and its consequences. This allows to speculate on the potential chemical modulators of inflammation and triggers of HO. The end of this second section is devoted to in vitro mineralization tests used to predict the ectopic potential of materials. The third section reviews the biological cascade of events occurring during pathological and material-induced HO, and attempts to propose a quantitative timeline of HO formation. The fourth section looks at potential ways to control HO formation, either acting on SLIHI or on inflammation. Chemical, physical, and drug-based approaches are considered. Finally, the evaluation finishes with a critical assessment of the definition of osteoinduction. STATEMENT OF SIGNIFICANCE: The ability to regenerate bone in a spatially controlled and reproducible manner is an essential prerequisite for the treatment of large bone defects. As such, understanding the mechanism leading to heterotopic ossification (HO), a condition triggered by an injury leading to the formation of mature lamellar bone in extraskeletal soft tissues, would be very useful. Unfortunately, the mechanism(s) behind HO is(are) poorly understood. The present study reviews the literature on HO and based on it, proposes that HO can be caused by a combination of inflammation and calcification. This mechanism helps to better understand current strategies to prevent and treat HO. It also shows new opportunities to improve the treatment of bone defects in orthopedic and dental procedures.
Collapse
|
15
|
Vermeulen S, Birgani ZT, Habibovic P. Biomaterial-induced pathway modulation for bone regeneration. Biomaterials 2022; 283:121431. [DOI: 10.1016/j.biomaterials.2022.121431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022]
|
16
|
Guo X, Li M, Qi W, Bai H, Nie Z, Hu Z, Xiao Y, de Bruijn JD, Bao C, Yuan H. Serial cellular events in bone formation initiated by calcium phosphate ceramics. Acta Biomater 2021; 134:730-743. [PMID: 34303865 DOI: 10.1016/j.actbio.2021.07.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 02/05/2023]
Abstract
To better understand the biological mechanisms triggered by osteoinductive materials in vivo, we evaluated the timeline of cellular responses to osteoinductive materials subcutaneously implanted in FVB mice. More F4/80-positive macrophages were present in osteoinductive tri-CaP ceramic (TCP) with submicron surface topography (TCPs) than non-osteoinductive TCP with micron surface topography (TCPb) at week 1. Moreover, TCPs (but not TCPb) significantly enhanced osteoclastogenesis, and induced macrophages to polarize from M1 to M2 in the first week. The time sequence and relevance of macrophages and osteoclasts responses involved in bone formation was then evaluated through peri-implant injection of specific chemicals in mice implanted with osteoinductive TCPs. Day-1 injection of clodronate liposomes (LipClod) depleted macrophages, inhibited macrophage polarization to M2, blocked osteoclastogenesis and bone formation, while the day-6 injection was less effective. Anti-RANKL antibody (aRANKL) did not affect macrophage colonization but inhibited osteoclastogenesis. Injection of aRANKL before week 2 aborted bone formation in TCPs, while injection at week 4 partially inhibited bone formation. The overall data show that following ectopic implantation, osteoinductive materials allow macrophage colonization in hours to days, macrophage polarization to M2 in days (within 7 days), osteoclastogenesis in weeks (e.g. in 2 weeks) and bone formation thereafter (after 4 weeks). The serial cellular events verified herein bring a new insight on material-induced bone formation and pave the way to further explore the mechanisms triggered by osteoinductive materials. STATEMENT OF SIGNIFICANCE: A series of key cellular events triggered by osteoinductive calcium phosphate ceramic was revealed: macrophages colonized within hours to days, polarization of M2 macrophages occurred within 7 days, osteoclastogenesis mainly occurred in weeks (e.g. in 2 weeks) and bone formation finally arose thereafter (after 4 weeks). Moreover, such time sequence of cellular events was confirmed with specific chemicals (clodronate liposomes and anti-RANKL antibody). The findings verified herein bring a new insight on material-induced bone formation and pave the way to further explore the mechanisms triggered by osteoinductive materials.
Collapse
Affiliation(s)
- Xiaodong Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Mingzheng Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 400015, China
| | - Wenting Qi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Hetian Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Zhangling Nie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Zhiqiao Hu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Yu Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China
| | - Joost D de Bruijn
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, MB Bilthoven 3723, the Netherlands; School of Engineering and Materials Science, Queen Mary University of London, UK
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, Sichuan 610041, China.
| | - Huipin Yuan
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, MB Bilthoven 3723, the Netherlands; Huipin Yuan's Lab, Sichuan, China.
| |
Collapse
|
17
|
Guan Z, Chen S, Pan F, Fan L, Sun D. Effects of Gene Delivery Approaches on Differentiation Potential and Gene Function of Mesenchymal Stem Cells. IEEE Trans Biomed Eng 2021; 69:83-95. [PMID: 34101578 DOI: 10.1109/tbme.2021.3087129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Introduction of a gene to mesenchymal stem cells (MSCs) is a well-known strategy to purposely manipulate the cell fate and further enhance therapeutic performance in cell-based therapy. Viral and chemical approaches for gene delivery interfere with differentiation potential. Although microinjection as a physical delivery method is commonly used for transfection, its influence on MSC cell fate is not fully understood. The current study aimed to evaluate the effects of four nonviral gene delivery methods on stem cell multi-potency. The four delivery methods are robotic microinjection, polyethylenimine (PEI), cationic liposome (cLipo), and calcium phosphate nanoparticles (CaP). Among the four methods, microinjection has exhibited the highest transfection efficiency of ~60%, while the three others showed lower efficiency of 10-25%. Robotic microinjection preserved fibroblast-like cell morphology, stress fibre intactness, and mature focal adhesion complex, while PEI caused severe cytotoxicity. No marked differentiation bias was observed after microinjection and cLipo treatment. By contrast, CaP-treated MSCs exhibited excessive osteogenesis, while PEI-treated MSCs showed excessive adipogenesis. Robotic microinjection system was used to inject the CRISPR/Cas9-encoding plasmid to knock out PPAR gene in MSCs, and the robotic microinjection did not interfere with PPAR function in differentiation commitment. Meanwhile, the bias in osteo-adipogenic differentiation exhibited in CaP and PEI-treated MSCs after PPAR knockout via chemical carriers. Our results indicate that gene delivery vehicles variously disturb MSCs differentiation and interfere with exogenous gene function. Our findings further suggest that robotic microinjection offers a promise of generating genetically modified MSCs without disrupting stem cell multi-potency and therapeutic gene function.
Collapse
|
18
|
Li M, Guo X, Qi W, Wu Z, de Bruijn JD, Xiao Y, Bao C, Yuan H. Macrophage polarization plays roles in bone formation instructed by calcium phosphate ceramics. J Mater Chem B 2021; 8:1863-1877. [PMID: 32067012 DOI: 10.1039/c9tb02932j] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To investigate the roles of macrophages in material-instructed bone formation, two calcium phosphate (TCP) ceramics with the same chemistry but various scales of surface topography were employed in this study. After being implanted subcutaneously in FVB mice for 8 weeks, TCPs (TCP ceramics with submicron surface topography) gave rise to bone formation, while TCPb (TCP ceramics with micron surface topography) did not, showing the crucial role of surface topography scale in material-instructed bone formation. Depletion of macrophages with liposomal clodronate (LipClod) blocked such bone formation instructed by TCPs, confirming the role of macrophages in material-instructed bone formation. Macrophage cells (i.e. RAW 264.7 cells) cultured on TCPs in vitro polarized to tissue repair macrophages as evidenced by gene expression and cytokine production, while polarizing to pro-inflammatory macrophages on TCPb. Submicron surface topography of TCP ceramics directed macrophage polarization via PI3K/AKT pathways with the synergistic regulation of integrin β1. Finally, the tissue repair macrophage polarization on TCPs resulted in osteogenic differentiation of mesenchymal stem cells in vitro. At early implantation in FVB mice, TCPs recruited more macrophages which polarized towards tissue repair macrophages with time. The present data demonstrate the important roles of macrophage polarization in bone formation instructed by calcium phosphate ceramics.
Collapse
Affiliation(s)
- Mingzheng Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, 610041, Sichuan, China.
| | - Xiaodong Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, 610041, Sichuan, China.
| | - Wenting Qi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, 610041, Sichuan, China.
| | - Zhenzhen Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, 610041, Sichuan, China.
| | - Joost D de Bruijn
- School of Engineering and Materials Science, Queen Mary University of London, UK
| | - Yu Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, 610041, Sichuan, China.
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Rd, Chengdu, 610041, Sichuan, China.
| | - Huipin Yuan
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, 3723 MB Bilthoven, The Netherlands and MERLN Institute, Maastricht University, The Netherlands
| |
Collapse
|
19
|
Charbonnier B, Hadida M, Marchat D. Additive manufacturing pertaining to bone: Hopes, reality and future challenges for clinical applications. Acta Biomater 2021; 121:1-28. [PMID: 33271354 DOI: 10.1016/j.actbio.2020.11.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/06/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
For the past 20 years, the democratization of additive manufacturing (AM) technologies has made many of us dream of: low cost, waste-free, and on-demand production of functional parts; fully customized tools; designs limited by imagination only, etc. As every patient is unique, the potential of AM for the medical field is thought to be considerable: AM would allow the division of dedicated patient-specific healthcare solutions entirely adapted to the patients' clinical needs. Pertinently, this review offers an extensive overview of bone-related clinical applications of AM and ongoing research trends, from 3D anatomical models for patient and student education to ephemeral structures supporting and promoting bone regeneration. Today, AM has undoubtably improved patient care and should facilitate many more improvements in the near future. However, despite extensive research, AM-based strategies for bone regeneration remain the only bone-related field without compelling clinical proof of concept to date. This may be due to a lack of understanding of the biological mechanisms guiding and promoting bone formation and due to the traditional top-down strategies devised to solve clinical issues. Indeed, the integrated holistic approach recommended for the design of regenerative systems (i.e., fixation systems and scaffolds) has remained at the conceptual state. Challenged by these issues, a slower but incremental research dynamic has occurred for the last few years, and recent progress suggests notable improvement in the years to come, with in view the development of safe, robust and standardized patient-specific clinical solutions for the regeneration of large bone defects.
Collapse
|
20
|
Zhang H, Liu K, Lu M, Liu L, Yan Y, Chu Z, Ge Y, Wang T, Qiu J, Bu S, Tang C. Micro/nanostructured calcium phytate coating on titanium fabricated by chemical conversion deposition for biomedical application. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111402. [PMID: 33255005 DOI: 10.1016/j.msec.2020.111402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/26/2020] [Accepted: 08/11/2020] [Indexed: 01/01/2023]
Abstract
A bioactive micro/nanostructured calcium phytate coating was successfully prepared on titanium surfaces by chemical conversion deposition, mainly through hydrothermal treatment of a mixed solution of phytic acid and saturated calcium hydroxide solution. Ultraviolet radiation was carried out to improve the adhesion of the coating to the titanium substrate. Pure titanium with a sandblasted/acid-etched surface was used as the control group. The topography and chemical composition of the modified surfaces were characterized by scanning electron microscopy (SEM), atomic force microscopy (AFM), X-ray photoelectron spectroscopy (XPS), energy-dispersive X-ray spectroscopy (EDX), and static water contact angle measurement. A pull-off test was performed to measure the coating-to-substrate adhesion strength. Bovine serum albumin was used as a model to study the protein adsorption effect. Cells were cultured on titanium surfaces for 7 days in osteogenic differentiation medium, then the osteoblast compatibility in vitro were explored by alkaline phosphatase and alizarin red staining. After 1, 2, 4 and 8 wks of immediate implantation of titanium implants into the mandibles of New Zealand white rabbits, biological effects in vivo were researched by microcomputed tomography analysis and histological evaluation. The results indicated that the roughness and hydrophilicity of the modified surfaces with micro/nanostructure remarkably increased compared to those of the control group. The pull-off test showed the average adhesion strength at the coating-substrate interface to be higher than 13.56 ± 1.71 MPa. In addition, approximately 4.41 mg/L calcium ion was released from the calcium phytate micro/nano coatings to the local environment after 48 h of immersion. More importantly, the micro/nanostructure titanium substrates significantly promoted cellular differentiation in vitro and in vivo. After 8 wks, the bone implant contact ratio (BIC, %) of the modified implants was higher than that of the control group, at 94.09 ± 0.55% and 86.18 ± 1.99% (p < 0.05). Overall, this study provided new insights into the factors promoting early osseointegration of titanium alloys, which had great potential not only for dental implants but also for various other biomaterial applications.
Collapse
Affiliation(s)
- Hao Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Department of Dental Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China; Department of Stomatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Kun Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Department of Dental Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China; Department of Implantology, Hefei Stomatological Hospital, Hefei Clinical School of Stomatology, Anhui Medical University, Hefei 230001, China
| | - Mengmeng Lu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Department of Dental Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lin Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Department of Dental Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yanzhe Yan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
| | - Zhuangzhuang Chu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
| | - Yuran Ge
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
| | - Tao Wang
- College of Materials Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Jing Qiu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Department of Dental Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shoushan Bu
- Department of Stomatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunbo Tang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China; Department of Dental Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
21
|
Upadhyai P, Guleria VS, Udupa P. Characterization of primary cilia features reveal cell-type specific variability in in vitro models of osteogenic and chondrogenic differentiation. PeerJ 2020; 8:e9799. [PMID: 32884864 PMCID: PMC7444507 DOI: 10.7717/peerj.9799] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Primary cilia are non-motile sensory antennae present on most vertebrate cell surfaces. They serve to transduce and integrate diverse external stimuli into functional cellular responses vital for development, differentiation and homeostasis. Ciliary characteristics, such as length, structure and frequency are often tailored to distinct differentiated cell states. Primary cilia are present on a variety of skeletal cell-types and facilitate the assimilation of sensory cues to direct skeletal development and repair. However, there is limited knowledge of ciliary variation in response to the activation of distinct differentiation cascades in different skeletal cell-types. C3H10T1/2, MC3T3-E1 and ATDC5 cells are mesenchymal stem cells, preosteoblast and prechondrocyte cell-lines, respectively. They are commonly employed in numerous in vitro studies, investigating the molecular mechanisms underlying osteoblast and chondrocyte differentiation, skeletal disease and repair. Here we sought to evaluate the primary cilia length and frequencies during osteogenic differentiation in C3H10T1/2 and MC3T3-E1 and chondrogenic differentiation in ATDC5 cells, over a period of 21 days. Our data inform on the presence of stable cilia to orchestrate signaling and dynamic alterations in their features during extended periods of differentiation. Taken together with existing literature these findings reflect the occurrence of not only lineage but cell-type specific variation in ciliary attributes during differentiation. These results extend our current knowledge, shining light on the variabilities in primary cilia features correlated with distinct differentiated cell phenotypes. It may have broader implications in studies using these cell-lines to explore cilia dependent cellular processes and treatment modalities for skeletal disorders centered on cilia modulation.
Collapse
Affiliation(s)
- Priyanka Upadhyai
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Vishal Singh Guleria
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Prajna Udupa
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
22
|
Abstract
The modification of implant devices with biocompatible coatings has become necessary as a consequence of premature loosening of prosthesis. This is caused mainly by chronic inflammation or allergies that are triggered by implant wear, production of abrasion particles, and/or release of metallic ions from the implantable device surface. Specific to the implant tissue destination, it could require coatings with specific features in order to provide optimal osseointegration. Pulsed laser deposition (PLD) became a well-known physical vapor deposition technology that has been successfully applied to a large variety of biocompatible inorganic coatings for biomedical prosthetic applications. Matrix assisted pulsed laser evaporation (MAPLE) is a PLD-derived technology used for depositions of thin organic material coatings. In an attempt to surpass solvent related difficulties, when different solvents are used for blending various organic materials, combinatorial MAPLE was proposed to grow thin hybrid coatings, assembled in a gradient of composition. We review herein the evolution of the laser technological process and capabilities of growing thin bio-coatings with emphasis on blended or multilayered biomimetic combinations. These can be used either as implant surfaces with enhanced bioactivity for accelerating orthopedic integration and tissue regeneration or combinatorial bio-platforms for cancer research.
Collapse
|
23
|
Xiao D, Zhang J, Zhang C, Barbieri D, Yuan H, Moroni L, Feng G. The role of calcium phosphate surface structure in osteogenesis and the mechanisms involved. Acta Biomater 2020; 106:22-33. [PMID: 31926336 DOI: 10.1016/j.actbio.2019.12.034] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/11/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Calcium phosphate (CaP) ceramics have been widely used for bone regeneration because of their ability to induce osteogenesis. Surface properties, including chemical composition and surface structure, are known to play a crucial role in osteoconduction and osteoinduction. This review systematically analyzes the effects of surface properties, in particular the surface structure, of CaP scaffolds on cell behavior and new bone formation. We also summarize the possible signaling pathways involved in the osteogenic differentiation of bone-related cells when cultured on surfaces with various structures in vitro. The significant immune response initiated by surface structure involved in osteogenic differentiation of cells is also discussed in this review. Taken together, the new biological principle for advanced biomaterials is not only to directly stimulate osteogenic differentiation of bone-related cells but also to modulate the immune response in vivo. Although the reaction mechanism responsible for bone formation induced by CaP surface structure is not clear yet, the insights on surface structure-mediated osteogenic differentiation and osteoimmunomodulation could aid the optimization of CaP-based biomaterials for bone regeneration. STATEMENT OF SIGNIFICANCE: CaP ceramics have similar inorganic composition with natural bone, which have been widely used for bone tissue scaffolds. CaP themselves are not osteoinductive; however, osteoinductive properties could be introduced to CaP materials by surface engineering. This paper systematically summarizes the effects of surface properties, especially surface structure, of CaP scaffolds on bone formation. Additionally, increasing evidence has proved that the bone healing process is not only affected by the osteogenic differentiation of bone-related cells, but also relevant to the the cooperation of immune system. Thus, we further review the possible signaling pathways involved in the osteogenic differentiation and immune response of cells cultured on scaffold surface. These insights into surface structure-mediated osteogenic differentiation and osteoimmunomodulated-based strategy could aid the optimization of CaP-based biomaterials.
Collapse
|
24
|
Hou W, Fu H, Liu X, Duan K, Lu X, Lu M, Sun T, Guo T, Weng J. Cation Channel Transient Receptor Potential Vanilloid 4 Mediates Topography-Induced Osteoblastic Differentiation of Bone Marrow Stem Cells. ACS Biomater Sci Eng 2019; 5:6520-6529. [PMID: 33417804 DOI: 10.1021/acsbiomaterials.9b01237] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Micro/nanotopographies (MNTs) have been reported to enhance the osseointegration of biomaterials and modulate cell functions, but the underlying mechanisms are incompletely understood. We hypothesized that transient receptor potential vanilloid 4 (TRPV4) may mediate the topographically induced osteoblastic differentiation of bone marrow stem cells (BMSCs) by regulating the NFATc1 and Wnt/β-catenin signaling. To test this hypothesis, murine BMSCs were cultured on polished titanium (Ti) discs (PT) and Ti discs carrying titania nanotubes (i.e., MNTs) with diameters of ∼30 and ∼100 nm (termed TNT-30 and TNT-100, respectively). It was found that the MNTs (in particular TNT-100) promoted the expression and activation of TRPV4. Inhibition of TRPV4 in BMSCs cultured on TNT-100 reduced the expression of osteoblastic genes and the gene expression and protein levels of NFATc1 and Wnt3a/β-catenin and also decreased nuclear translocation of NFATc1 and β-catenin (all vs uninhibited BMSCs). Conversely, activation of TRPV4 in BMSCs cultured on PT increased the expression of the osteoblastic gene and the gene expression and protein level of NFATc1 and Wnt3a/β-catenin and also enhanced the nuclear translocation of NFATc1 and β-catenin (all vs unactivated BMSCs). These differences suggest that the MNTs promoted TRPV4 expression and activation to enhance intracellular Ca2+, which further increased the nuclear translocation of NFATc1 and stimulated the Wnt/β-catenin signaling, thus leading to upregulated expression of osteoblastic genes. These results indicate TRPV4 to be a mediator in MNT-induced osteoblastic differentiation of BMSCs.
Collapse
|
25
|
Abstract
Primary cilia project in a single copy from the surface of most vertebrate cell types; they detect and transmit extracellular cues to regulate diverse cellular processes during development and to maintain tissue homeostasis. The sensory capacity of primary cilia relies on the coordinated trafficking and temporal localization of specific receptors and associated signal transduction modules in the cilium. The canonical Hedgehog (HH) pathway, for example, is a bona fide ciliary signalling system that regulates cell fate and self-renewal in development and tissue homeostasis. Specific receptors and associated signal transduction proteins can also localize to primary cilia in a cell type-dependent manner; available evidence suggests that the ciliary constellation of these proteins can temporally change to allow the cell to adapt to specific developmental and homeostatic cues. Consistent with important roles for primary cilia in signalling, mutations that lead to their dysfunction underlie a pleiotropic group of diseases and syndromic disorders termed ciliopathies, which affect many different tissues and organs of the body. In this Review, we highlight central mechanisms by which primary cilia coordinate HH, G protein-coupled receptor, WNT, receptor tyrosine kinase and transforming growth factor-β (TGFβ)/bone morphogenetic protein (BMP) signalling and illustrate how defects in the balanced output of ciliary signalling events are coupled to developmental disorders and disease progression.
Collapse
|
26
|
Lu B, Zhu DY, Yin JH, Xu H, Zhang CQ, Ke QF, Gao YS, Guo YP. Incorporation of cerium oxide in hollow mesoporous bioglass scaffolds for enhanced bone regeneration by activating the ERK signaling pathway. Biofabrication 2019; 11:025012. [PMID: 30754024 DOI: 10.1088/1758-5090/ab0676] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hierarchically porous structures and bioactive compositions of artificial biomaterials play a positive role in bone defect healing and new bone regeneration. Herein, cerium oxide nanoparticles-modified bioglass (Ce-BG) scaffolds were firstly constructed by the incorporation of hollow mesoporous Ce-BG microspheres in CTS via a freeze-drying technology. The interconnected macropores in Ce-BG scaffolds facilitated the in-growth of bone cells/tissues from material surfaces into the interiors, while the hollow cores and mesopore shells in Ce-BG microspheres provides more active sites for bone mineralization. The cerium oxide nanoparticles in the scaffolds rapidly promoted the proliferation and osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs), as confirmed by the up-regulation of osteogenesis-related markers such as OCN, ALP and COL-1. The enhanced osteoinductivity of Ce-BG scaffolds was mainly related to the activated ERK pathway, and it was blocked by adding a selective ERK1/2 inhibitor (SCH772984). In vivo rat cranial defect models revealed that Ce-BG scaffolds accelerated collagen deposition, osteoblast formation and bone regeneration as compared to BG scaffolds. The exciting results demonstrated that the synergistic effects between hierarchically porous structures and cerium oxide nanoparticles contributed to osteogenic ability, and hollow mesoporous Ce-BG scaffolds would be a novel platform for bone regeneration.
Collapse
Affiliation(s)
- Bin Lu
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Huang J, Chen Y, Tang C, Fei Y, Wu H, Ruan D, Paul ME, Chen X, Yin Z, Heng BC, Chen W, Shen W. The relationship between substrate topography and stem cell differentiation in the musculoskeletal system. Cell Mol Life Sci 2019; 76:505-521. [PMID: 30390116 PMCID: PMC11105278 DOI: 10.1007/s00018-018-2945-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/15/2018] [Accepted: 10/12/2018] [Indexed: 12/11/2022]
Abstract
It is well known that biomaterial topography can exert a profound influence on various cellular functions such as migration, polarization, and adhesion. With the development and refinement of manufacturing technology, much research has recently been focused on substrate topography-induced cell differentiation, particularly in the field of tissue engineering. Even without biological and chemical stimuli, the differentiation of stem cells can also be initiated by various biomaterials with different topographic features. However, the underlying mechanisms of this biological phenomenon remain elusive. During the past few decades, many researchers have demonstrated that cells can sense the topography of materials through the assembly and polymerization of membrane proteins. Following the activation of RHO, TGF-b or FAK signaling pathways, cells can be induced into various differentiation states. But these signaling pathways often coincide with canonical mechanical transduction pathways, and no firm conclusion has been reached among researchers in this field on topography-specific signaling pathways. On the other hand, some substrate topographies are reported to have the ability to inhibit differentiation and maintain the 'stemness' of stem cells. In this review, we will summarize the role of topography in musculoskeletal system regeneration and explore possible topography-related signaling pathways involved in cell differentiation.
Collapse
Affiliation(s)
- Jiayun Huang
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang, 310000, China
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China
- Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang, 310000, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Yangwu Chen
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang, 310000, China
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China
- Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang, 310000, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Chenqi Tang
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang, 310000, China
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China
- Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang, 310000, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Yang Fei
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, China
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Haoyu Wu
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang, 310000, China
- Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang, 310000, China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, China
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Maswikiti Ewetse Paul
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang, 310000, China
- Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang, 310000, China
| | - Xiao Chen
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang, 310000, China
- Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang, 310000, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Zi Yin
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang, 310000, China
- Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang, 310000, China
| | - Boon Chin Heng
- Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Weishan Chen
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, China
| | - Weiliang Shen
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, 310009, China.
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Zhejiang, 310000, China.
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China.
- Department of Sports Medicine, School of Medicine, Zhejiang University, Zhejiang, 310000, China.
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China.
| |
Collapse
|
28
|
Nucleic acids and analogs for bone regeneration. Bone Res 2018; 6:37. [PMID: 30603226 PMCID: PMC6306486 DOI: 10.1038/s41413-018-0042-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/19/2018] [Accepted: 11/26/2018] [Indexed: 02/07/2023] Open
Abstract
With the incidence of different bone diseases increasing, effective therapies are needed that coordinate a combination of various technologies and biological materials. Bone tissue engineering has also been considered as a promising strategy to repair various bone defects. Therefore, different biological materials that can promote stem cell proliferation, migration, and osteoblastic differentiation to accelerate bone tissue regeneration and repair have also become the focus of research in multiple fields. Stem cell therapy, biomaterial scaffolds, and biological growth factors have shown potential for bone tissue engineering; however, off-target effects and cytotoxicity have limited their clinical use. The application of nucleic acids (deoxyribonucleic acid or ribonucleic acid) and nucleic acid analogs (peptide nucleic acids or locked nucleic acids), which are designed based on foreign genes or with special structures, can be taken up by target cells to exert different effects such as modulating protein expression, replacing a missing gene, or targeting specific gens or proteins. Due to some drawbacks, nucleic acids and nucleic acid analogs are combined with various delivery systems to exert enhanced effects, but current studies of these molecules have not yet satisfied clinical requirements. In-depth studies of nucleic acid or nucleic acid analog delivery systems have been performed, with a particular focus on bone tissue regeneration and repair. In this review, we mainly introduce delivery systems for nucleic acids and nucleic acid analogs and their applications in bone repair and regeneration. At the same time, the application of conventional scaffold materials for the delivery of nucleic acids and nucleic acid analogs is also discussed. Used with an appropriate delivery system, nucleic acids and nucleic acid analogs have excellent potential for bone repair and regeneration. Owing to various challenges with bone tissue regeneration, current research is largely focused on gene therapy, which employs genes to treat or prevent disease, and such new materials as nucleic acids (DNA and RNA) and nucleic acid analogs (compounds structurally similar to naturally occurring nucleic acids). A team headed by Yunfeng Lin at Sichuan University, China conducted a review of delivery systems for nucleic acids and nucleic acid analogs and their application in bone repair and regeneration. The authors identified the use of biomaterial scaffolds (which mimic living tissue) as one of the most important research areas for gene therapy, and that strategy has proven effective with all types of bone regeneration and repair.
Collapse
|
29
|
Baumgartner W, Otto L, Hess SC, Stark WJ, Märsmann S, Bürgisser GM, Calcagni M, Cinelli P, Buschmann J. Cartilage/bone interface fabricated under perfusion: Spatially organized commitment of adipose‐derived stem cells without medium supplementation. J Biomed Mater Res B Appl Biomater 2018; 107:1833-1843. [DOI: 10.1002/jbm.b.34276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/27/2018] [Accepted: 10/17/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Walter Baumgartner
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Lukas Otto
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Samuel C. Hess
- Institute for Chemical‐ and BioengineeringDepartment of Chemistry and Applied Biosciences ETH Zürich, Zürich Switzerland
| | - Wendelin J. Stark
- Institute for Chemical‐ and BioengineeringDepartment of Chemistry and Applied Biosciences ETH Zürich, Zürich Switzerland
| | - Sonja Märsmann
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
- Division of Trauma SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | | | - Maurizio Calcagni
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Paolo Cinelli
- Division of Trauma SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Johanna Buschmann
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| |
Collapse
|
30
|
Duan R, Barbieri D, Luo X, Weng J, Bao C, de Bruijn JD, Yuan H. Variation of the bone forming ability with the physicochemical properties of calcium phosphate bone substitutes. Biomater Sci 2018; 6:136-145. [PMID: 29147713 DOI: 10.1039/c7bm00717e] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Because of their bioactive properties and chemical similarity to the inorganic component of bone, calcium phosphate (CaP) materials are widely used for bone regeneration. Six commercially available CaP bone substitutes (Bio-Oss, Actifuse, Bi-Ostetic, MBCP, Vitoss and chronOs) as well as two tricalcium phosphate (TCP) ceramics with either a micron-scale (TCP-B) or submicron-scale (TCP-S) surface structure are characterized and their bone forming potential is evaluated in a canine ectopic implantation model. After 12 weeks of implantation in the paraspinal muscle of four beagles, sporadic bone (0.1 ± 0.1%) is observed in two Actifuse implants (2/4), limited bone (2.1 ± 1.4%) in four MBCP implants (4/4) and abundant bone (21.6 ± 4.5%) is formed in all TCP-S implants (4/4). Bone is not observed in any of the Bio-Oss, Bi-Ostetic, Vitoss, chronOs and TCP-B implants (0/4). When correlating the bone forming potential with the physicochemical properties of each material, we observe that the physical characteristics (e.g. grain size and micropore size at the submicron scale) might be the dominant trigger of material directed bone formation via specific mechanotransduction, instead of protein adsorption, surface mineralization and calcium ion release.
Collapse
Affiliation(s)
- Rongquan Duan
- Biomaterials Science and Technology, MIRA Institute, University of Twente, 7500 AE, Enschede, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
31
|
Mechanical loading induces primary cilia disassembly in tendon cells via TGFβ and HDAC6. Sci Rep 2018; 8:11107. [PMID: 30038235 PMCID: PMC6056413 DOI: 10.1038/s41598-018-29502-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/04/2018] [Indexed: 11/24/2022] Open
Abstract
This study used isolated human tenocytes to test the hypothesis that cyclic mechanical strain directly stimulates primary cilia disassembly, and to elucidate the mechanisms involved. Cells were seeded onto flexible membranes and strained at 0–3%; 1 Hz, for up to 24 hours. Cilia length and prevalence progressively reduced with increasing strain duration but showed full recovery within 2 hours of strain removal. The response to loading was not influenced by actin organisation as seen in other cell types. However, the loading response could be recreated by treatment with TGFβ. Furthermore, treatment with the HDAC6 inhibitor Tubacin, or a TGFβ receptor inhibitor both prevented strain induced cilia disassembly. These data are the first to describe primary cilia expression in isolated tenocytes, showing that mechanical strain regulates cilia expression independent of changes in tendon extracellular matrix. Furthermore, we show that cilia disassembly is mediated by the activation of TGFβ receptors leading to activation of HDAC6. Previous studies have shown that cilia are required for TGFβ signalling and that tendon mechanosignalling is mediated by TGFβ. The present study therefore suggests a novel feedback mechanism whereby cilia disassembly inhibits prolonged TGFβ activation in response to continuous cyclic loading.
Collapse
|
32
|
Ghayor C, Weber FE. Osteoconductive Microarchitecture of Bone Substitutes for Bone Regeneration Revisited. Front Physiol 2018; 9:960. [PMID: 30072920 PMCID: PMC6060436 DOI: 10.3389/fphys.2018.00960] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/29/2018] [Indexed: 12/11/2022] Open
Abstract
In the last three decades, all efforts in bone tissue engineering were driven by the dogma that the ideal pore size in bone substitutes lies between 0.3 and 0.5 mm in diameter. Newly developed additive manufacturing methodologies for ceramics facilitate the total control over pore size, pore distribution, bottleneck size, and bottleneck distribution. Therefore, this appears to be the method of choice with which to test the aforementioned characteristics of an ideal bone substitute. To this end, we produced a library of 15 scaffolds with diverse defined pore/bottleneck dimensions and distributions, tested them in vivo in a calvarial bone defect model in rabbits, and assessed the clinically most relevant parameters: defect bridging and bony regenerated area. Our in vivo data revealed that the ideal pore/bottleneck dimension for bone substitutes is in the range of 0.7-1.2 mm, and appears therefore to be twofold to fourfold more extended than previously thought. Pore/bottleneck dimensions of 1.5 and 1.7 mm perform significantly worse and appear unsuitable in bone substitutes. Thus, our results set the ideal range of pore/bottleneck dimensions and are likely to have a significant impact on the microarchitectural design of future bone substitutes for use in orthopedic, trauma, cranio-maxillofacial and oral surgery.
Collapse
Affiliation(s)
- Chafik Ghayor
- Oral Biotechnology and Bioengineering, Department of Cranio-Maxillofacial and Oral Surgery, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| | - Franz E. Weber
- Oral Biotechnology and Bioengineering, Department of Cranio-Maxillofacial and Oral Surgery, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
33
|
Baumgartner W, Schneider I, Hess SC, Stark WJ, Märsmann S, Brunelli M, Calcagni M, Cinelli P, Buschmann J. Cyclic uniaxial compression of human stem cells seeded on a bone biomimetic nanocomposite decreases anti-osteogenic commitment evoked by shear stress. J Mech Behav Biomed Mater 2018; 83:84-93. [DOI: 10.1016/j.jmbbm.2018.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/16/2018] [Accepted: 04/03/2018] [Indexed: 01/01/2023]
|
34
|
Morthorst SK, Christensen ST, Pedersen LB. Regulation of ciliary membrane protein trafficking and signalling by kinesin motor proteins. FEBS J 2018; 285:4535-4564. [PMID: 29894023 DOI: 10.1111/febs.14583] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/09/2018] [Accepted: 06/11/2018] [Indexed: 12/14/2022]
Abstract
Primary cilia are antenna-like sensory organelles that regulate a substantial number of cellular signalling pathways in vertebrates, both during embryonic development as well as in adulthood, and mutations in genes coding for ciliary proteins are causative of an expanding group of pleiotropic diseases known as ciliopathies. Cilia consist of a microtubule-based axoneme core, which is subtended by a basal body and covered by a bilayer lipid membrane of unique protein and lipid composition. Cilia are dynamic organelles, and the ability of cells to regulate ciliary protein and lipid content in response to specific cellular and environmental cues is crucial for balancing ciliary signalling output. Here we discuss mechanisms involved in regulation of ciliary membrane protein trafficking and signalling, with main focus on kinesin-2 and kinesin-3 family members.
Collapse
|
35
|
Wheatley DN. The primary cilium - once a "rudimentary" organelle that is now a ubiquitous sensory cellular structure involved in many pathological disorders. J Cell Commun Signal 2018; 12:211-216. [PMID: 29218455 PMCID: PMC5842197 DOI: 10.1007/s12079-017-0436-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 11/16/2017] [Indexed: 11/26/2022] Open
Abstract
This article looks mostly at the steps that have led to the primary cilium finding its place in our understanding of cell biology, developmental biology, and medical syndromes due to its aberrations. It is a personal account that stresses, if nothing else, the value of the adage "stick to your guns". My obsession with this organelle, following on from fascination with the centriole, has led to a whole career devoted to determining the nature and role of primary cilia in basic cell biology, which has proved much more important than had been appreciated for almost a century. They are heavily involved in very many aspects of cell physiology that have much wider implications with regard to human biology and probably throughout the animal kingdom. That aberrations, to the surprise of many researchers in their structure or functioning has led to their being implicated or perhaps deeply involved in an extraordinary range of medical conditions. This invitation allows me to raise crucial questions that need answers regarding the regulation of their genesis, their cache of both intracellular and extracellular signal, and their association with a multitude of development processes from embryo to adult status.
Collapse
Affiliation(s)
- Denys N Wheatley
- BioMedES, Leggat, Keithall, Inverurie, AB51 0LX, Aberdeenshire, UK.
| |
Collapse
|