1
|
von Kroge S, Schmidt C, Butscheidt S, Ohlmeier M, Amling M, Beil FT, Gehrke T, Püschel K, Hahn M, Rolvien T. Allografts promote skeletal regeneration of periprosthetic femoral bone loss. J Orthop Translat 2025; 52:182-191. [PMID: 40330274 PMCID: PMC12053977 DOI: 10.1016/j.jot.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/19/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
Background Periprosthetic bone loss is a common clinical problem in hip arthroplasty that must be addressed during revision surgery to achieve adequate implant stability. Although bone allografts represent the clinical standard among substitute materials used, evidence of their regenerative potential at the microstructural, cellular, and compositional level is lacking. Methods A multiscale imaging approach comprising contact radiography, undecalcified histology, scanning electron microscopy, and nanoindentation was employed on human femoral explants obtained postmortem many years after allograft use during revision surgery. Results The degree of skeletal regeneration through allograft incorporation between host bone and allograft bone was highly dependent on the defect depth (R2 = 0.94, p < 0.001), while no association between the allograft time in situ and incorporation (R2 = 0.06, p = 0.61) was apparent. The host bone-allograft interface showed a high overlap of 4.0 ± 2.9 mm and was characterized by active bone remodelling, as indicated by osteoid accumulation, high abundance of bone cells and vasculature. While bone cement generally limited the incorporation process, the osteocytic canalicular system of the host bone reached the allograft interface to guide bone remodelling. Conclusion This is the first multiscale, histomorphometry-based evaluation of bone allografts used in revision hip arthroplasty for femoral bone loss in humans, demonstrating that they adequately facilitate skeletal regeneration through osteoconduction and subsequent remodelling. The translational potential of this article This study identified the mechanisms and determinants of femoral defect regeneration through allografts on the basis of a unique sample collection. While our results support their favourable clinical outcomes, the scientific basis for incomplete incorporation is also demonstrated.
Collapse
Affiliation(s)
- Simon von Kroge
- Institute of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
- Department of Trauma and Orthopaedic Surgery, Division of Orthopaedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Constantin Schmidt
- Institute of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
- Department of Trauma and Orthopaedic Surgery, Division of Orthopaedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Sebastian Butscheidt
- Institute of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
- Department of Trauma and Orthopaedic Surgery, Division of Orthopaedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Malte Ohlmeier
- Department of Joint Surgery, Helios ENDO-Klinik, Holstenstr. 2, 22767, Hamburg, Germany
| | - Michael Amling
- Institute of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Frank Timo Beil
- Department of Trauma and Orthopaedic Surgery, Division of Orthopaedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Thorsten Gehrke
- Department of Joint Surgery, Helios ENDO-Klinik, Holstenstr. 2, 22767, Hamburg, Germany
| | - Klaus Püschel
- Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Michael Hahn
- Institute of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Tim Rolvien
- Institute of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
- Department of Trauma and Orthopaedic Surgery, Division of Orthopaedics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| |
Collapse
|
2
|
Yuan Z, Lin B, Wang C, Yan Z, Yang F, Su H. Collagen remodeling-mediated signaling pathways and their impact on tumor therapy. J Biol Chem 2025; 301:108330. [PMID: 39984051 PMCID: PMC11957794 DOI: 10.1016/j.jbc.2025.108330] [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: 08/27/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 02/23/2025] Open
Abstract
In addition to their traditional roles in maintaining tissue morphology and organ development, emerging evidence suggests that collagen (COL) remodeling-referring to dynamic changes in the quantity, stiffness, arrangements, cleavage states, and homo-/hetero-trimerization of COLs-serves as a key signaling mechanism that governs tumor growth and metastasis. COL receptors act as switches, linking various forms of COL remodeling to different cell types during cancer progression, including cancer cells, immune cells, and cancer-associated fibroblasts. In this review, we summarize recent findings on the signaling pathways mediated by COL arrangement, cleavage, and trimerization states (both homo- and hetero-), as well as the roles of the primary COL receptors-integrin, DDR1/2, LAIR-1/2, MRC2, and GPVI-in cancer progression. We also discuss the latest therapeutic strategies targeting COL fragments, cancer-associated fibroblasts, and COL receptors, including integrins, DDR1/2, and LAIR1/2. Understanding the pathways modulated by COL remodeling and COL receptors in various pathological contexts will pave the way for developing new precision therapies.
Collapse
Affiliation(s)
- Zihang Yuan
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, MOE Innovation Center for Basic Research in Tumor Immunotherapy, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Bo Lin
- Liver Cancer Institute, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chunlan Wang
- Liver Cancer Institute, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhaoyue Yan
- The Department of Stomatology, Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong, China
| | - Fei Yang
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, MOE Innovation Center for Basic Research in Tumor Immunotherapy, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Hua Su
- Liver Cancer Institute, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Zhang Y, Zhou X, Liu Q, Shen M, Liu Y, Zhang X. Simultaneous co-assembly of collagen and glycosaminoglycans to build a biomimetic extracellular matrix for bone regeneration. Int J Biol Macromol 2024; 279:135535. [PMID: 39349329 DOI: 10.1016/j.ijbiomac.2024.135535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 10/02/2024]
Abstract
Glycosaminoglycans (GAGs), also known as shape modules, are considered junctions that help define the shape of collagen matrix and further promote mineralization during osteogenesis. Many attempts have been made to immobilize GAGs on assembled collagen to modify the latter's surface state. However, it remains unclear how GAGs spontaneously identify collagen molecules during fibrillogenesis in vivo. Understanding the relationship between GAGs and collagen from both the bone physiology and materials science perspectives is of fundamental interest. Here, we introduced hyaluronic acid (HA, a main member of GAGs) during collagen self-assembly, in a process called modification cooperating with self-assembly (MCS). The molecular docking and morphological studies revealed that HA can help define collagen monomer deposition and thus promote fibrillogenesis through steric hindrance or by directly forming hydrogen bonds. Meanwhile, HA acts as a templating chaperone (TC) to increase the local mineral concentration within intrafibrillar channels but does not initiate nucleation, thus improving the crystallinity of formed apatite. The scaffolds synthesized through MCS model significantly improved the physicochemical stability and mechanical strength of collagen-based scaffolds. The optimized scaffolds promoted in-situ osteogenesis by stimulating the osteogenic differentiation of bone mesenchymal stem cells, either in an osteogenic medium, or after implantation into critical calvarial defects. This study provides novel insights towards evolving engineering scaffolds from inert supports to functional substitutes.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Department of Biomaterials, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Xinye Zhou
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Qing Liu
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Minjuan Shen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310003, China.
| | - Ying Liu
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China.
| | - Xu Zhang
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China.
| |
Collapse
|
4
|
He X, Yamada M, Watanabe J, Pengyu Q, Chen J, Egusa H. Titanium nanotopography enhances mechano-response of osteocyte three-dimensional network toward osteoblast activation. BIOMATERIALS ADVANCES 2024; 163:213939. [PMID: 38954876 DOI: 10.1016/j.bioadv.2024.213939] [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: 12/21/2023] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
The bone turnover capability influences the acquisition and maintenance of osseointegration. The architectures of osteocyte three-dimensional (3D) networks determine the direction and activity of bone turnover through osteocyte intercellular crosstalk, which exchanges prostaglandins through gap junctions in response to mechanical loading. Titanium nanosurfaces with anisotropically patterned dense nanospikes promote the development of osteocyte lacunar-canalicular networks. We investigated the effects of titanium nanosurfaces on intercellular network development and regulatory capabilities of bone turnover in osteocytes under cyclic compressive loading. MLO-Y4 mouse osteocyte-like cell lines embedded in type I collagen 3D gels on titanium nanosurfaces promoted the formation of intercellular networks and gap junctions even under static culture conditions, in contrast to the poor intercellular connectivity in machined titanium surfaces. The osteocyte 3D network on the titanium nanosurfaces further enhanced gap junction formation after additional culturing under cyclic compressive loading simulating masticatory loading, beyond the degree observed on machined titanium surfaces. A prostaglandin synthesis inhibitor cancelled the dual effects of titanium nanosurfaces and cyclic compressive loading on the upregulation of gap junction-related genes in the osteocyte 3D culture. Supernatants from osteocyte monolayer culture on titanium nanosurfaces promoted osteocyte maturation and intercellular connections with gap junctions. With cyclic loading, titanium nanosurfaces induced expression of the regulatory factors of bone turnover in osteocyte 3D cultures, toward higher osteoblast activation than that observed on machined surfaces. Titanium nanosurfaces with anisotropically patterned dense nanospikes promoted intercellular 3D network development and regulatory function toward osteoblast activation in osteocytes activated by cyclic compressive loading, through intercellular crosstalk by prostaglandin.
Collapse
Affiliation(s)
- Xindie He
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan; Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Masahiro Yamada
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan.
| | - Jun Watanabe
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
| | - Qu Pengyu
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
| | - Jiang Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan; Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan.
| |
Collapse
|
5
|
Finlay M, Hill LA, Neag G, Patel B, Chipara M, Lamont HC, Frost K, Patrick K, Lewis JW, Nicholson T, Edwards J, Jones SW, Grover LM, Naylor AJ. A detailed methodology for a three-dimensional, self-structuring bone model that supports the differentiation of osteoblasts towards osteocytes and the production of a complex collagen-rich mineralised matrix. F1000Res 2024; 12:357. [PMID: 38778815 PMCID: PMC11109547 DOI: 10.12688/f1000research.130779.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 05/25/2024] Open
Abstract
Background There are insufficient in vitro bone models that accommodate long-term culture of osteoblasts and support their differentiation to osteocytes. The increased demand for effective therapies for bone diseases, and the ethical requirement to replace animals in research, warrants the development of such models.Here we present an in-depth protocol to prepare, create and maintain three-dimensional, in vitro, self-structuring bone models that support osteocytogenesis and long-term osteoblast survival (>1 year). Methods Osteoblastic cells are seeded on a fibrin hydrogel, cast between two beta-tricalcium phosphate anchors. Analytical methods optimised for these self-structuring bone model (SSBM) constructs, including RT-qPCR, immunofluorescence staining and XRF, are described in detail. Results Over time, the cells restructure and replace the initial matrix with a collagen-rich, mineralising one; and demonstrate differentiation towards osteocytes within 12 weeks of culture. Conclusions Whilst optimised using a secondary human cell line (hFOB 1.19), this protocol readily accommodates osteoblasts from other species (rat and mouse) and origins (primary and secondary). This simple, straightforward method creates reproducible in vitro bone models that are responsive to exogenous stimuli, offering a versatile platform for conducting preclinical translatable research studies.
Collapse
Affiliation(s)
- Melissa Finlay
- Healthcare Technologies Institute, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Laurence A Hill
- Healthcare Technologies Institute, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Georgiana Neag
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Binal Patel
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Miruna Chipara
- Healthcare Technologies Institute, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Hannah C Lamont
- Healthcare Technologies Institute, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Kathryn Frost
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Kieran Patrick
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Jonathan W Lewis
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Thomas Nicholson
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - James Edwards
- NDORMS, University of Oxford, Oxford, Oxfordshire, OX3 7HE, UK
| | - Simon W Jones
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Liam M Grover
- Healthcare Technologies Institute, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| | - Amy J Naylor
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK
| |
Collapse
|
6
|
Suliman M, Nagasawa M, Al-Omari FA, Uoshima K. The effects of collagen cross-link deficiency on osseointegration process of pure titanium implants. J Prosthodont Res 2024; 68:449-455. [PMID: 37793821 DOI: 10.2186/jpr.jpr_d_22_00249] [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] [Indexed: 10/06/2023]
Abstract
PURPOSE This study aimed to investigate the effect of collagen cross-link deficiency on collagen fiber formation around an implant and its effect on the osseointegration process. METHODS Wistar rats were fed 0.1% beta-aminopropionitrile (BAPN) dissolved in water to induce collagen cross-link deficiency. Custom-made mini-implants with machined surfaces were placed proximal to the tibia. At 1, 2, and 4 weeks postoperatively, the bone area around the implant, bone-implant contact ratio, osteoclast/osteocyte activity, and osseointegration strength were evaluated using histological and immunohistochemical analyses and biomechanical tests. RESULTS Long-term disturbance of collagen cross-link formation in the BAPN group resulted in faster collagen fiber maturation than that in controls, with a defective collagen structure, low bone formation quantity, and low bone-implant contact values. Deficiency of collagen cross-links resulted in increased bone resorption and decreased osteocyte activity. CONCLUSIONS Collagen cross-linking is important for the formation of the collagen matrix, and their deficiency may impair bone activity around implants, affecting the osseointegration process.
Collapse
Affiliation(s)
- Mubarak Suliman
- Division of Bio-Prosthodontics, Department of Oral Health Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masako Nagasawa
- Division of Bio-Prosthodontics, Department of Oral Health Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Farah A Al-Omari
- Division of Bio-Prosthodontics, Department of Oral Health Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Katsumi Uoshima
- Division of Bio-Prosthodontics, Department of Oral Health Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
7
|
Owen R, Wittkowske C, Lacroix D, Perrault CM, Reilly GC. β-glycerophosphate, not low magnitude fluid shear stress, increases osteocytogenesis in the osteoblast-to-osteocyte cell line IDG-SW3. Connect Tissue Res 2024; 65:313-329. [PMID: 38982804 PMCID: PMC11371265 DOI: 10.1080/03008207.2024.2375065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
AIM As osteoblasts deposit a mineralized collagen network, a subpopulation of these cells differentiates into osteocytes. Biochemical and mechanical stimuli, particularly fluid shear stress (FSS), are thought to regulate this, but their relative influence remains unclear. Here, we assess both biochemical and mechanical stimuli on long-term bone formation and osteocytogenesis using the osteoblast-osteocyte cell line IDG-SW3. METHODS Due to the relative novelty and uncommon culture conditions of IDG-SW3 versus other osteoblast-lineage cell lines, effects of temperature and media formulation on matrix deposition and osteocytogenesis were initially characterized. Subsequently, the relative influence of biochemical (β-glycerophosphate (βGP) and ascorbic acid 2-phosphate (AA2P)) and mechanical stimulation on osteocytogenesis was compared, with intermittent application of low magnitude FSS generated by see-saw rocker. RESULTS βGP and AA2P supplementation were required for mineralization and osteocytogenesis, with 33°C cultures retaining a more osteoblastic phenotype and 37°C cultures undergoing significantly higher osteocytogenesis. βGP concentration positively correlated with calcium deposition, whilst AA2P stimulated alkaline phosphatase (ALP) activity and collagen deposition. We demonstrate that increasing βGP concentration also significantly enhances osteocytogenesis as quantified by the expression of green fluorescent protein linked to Dmp1. Intermittent FSS (~0.06 Pa) rocker had no effect on osteocytogenesis and matrix deposition. CONCLUSIONS This work demonstrates the suitability and ease with which IDG-SW3 can be utilized in osteocytogenesis studies. IDG-SW3 mineralization was only mediated through biochemical stimuli with no detectable effect of low magnitude FSS. Osteocytogenesis of IDG-SW3 primarily occurred in mineralized areas, further demonstrating the role mineralization of the bone extracellular matrix has in osteocyte differentiation.
Collapse
Affiliation(s)
- Robert Owen
- Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Claudia Wittkowske
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Damien Lacroix
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Cecile M. Perrault
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Gwendolen C. Reilly
- Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
| |
Collapse
|
8
|
Korzekwa AJ, Kononiuk A, Kordan W, Orzołek A. Retinoic acid alters metalloproteinase action in red deer antler stem cells. PLoS One 2023; 18:e0287782. [PMID: 37428795 DOI: 10.1371/journal.pone.0287782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 06/13/2023] [Indexed: 07/12/2023] Open
Abstract
Metalloproteinases (MMP)s regulate developmental processes, control angiogenesis and wound healing, participate in the formation of immune receptors, and are expressed in stem cells. Retinoic acid (RA) is a potential modulator of these proteinases. The aim was to determine (1) MMPs' action in antler stem cells (ASCs) before and after differentiation into adipo-, osteo-, and chondrocytes and (2) the effect of RA on modifying MMP action in ASCs. Antler tissue from pedicle was collected approximately 40 days after antler casting, post mortem from healthy breeding five year old males (N = 7). The cells were isolated from the pedicle layer of periosteum after skin separation and cultured. The pluripotency of the ASCs was evaluated by mRNA expression for NANOG, SOX2, and OCT4. ASCs were stimulated with RA (100nM) and differentiated for 14 days. The MMP (1-3) and TIMP(1-3) (tissue inhibitor of MMPs) mRNA expression was determined in the ASCs, their concentrations in the ASCs and the medium after RA stimulation as well as profiles of mRNA expression for MMPs: 1-3 and TIMPs: 1-3 during differentiation of ASC to osteocytes, adipocytes and chondrocytes. RA increased MMP-3 and TIMP-3 mRNA expression and output (P < 0.05) and not influenced on MMP-1 and TIMP-1 mRNA expression and output in ASC (P > 0.05). Depending on differentiation of ASC to osteocytes, adipocytes or chondrocytes, MMPs`and TIMPs`expression profile fluctuates for all studied proteases and its inhibitors. The studies demand continuation considering the role of proteases in stem cells physiology and differentiation. The results may be relevant for the study of cellular processes during the cancerogenesis of tumor stem cells.
Collapse
Affiliation(s)
- Anna J Korzekwa
- Department of Biodiversity Protection (DBP), Institute of Animal Reproduction and Food Research, Polish Academy of Sciences (IAR&FR PAS), Olsztyn, Poland
| | - Anna Kononiuk
- Department of Biodiversity Protection (DBP), Institute of Animal Reproduction and Food Research, Polish Academy of Sciences (IAR&FR PAS), Olsztyn, Poland
| | - Władysław Kordan
- Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Aleksandra Orzołek
- Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
9
|
Luo ZH, Ma JX, Zhang W, Tian AX, Gong SW, Li Y, Lai YX, Ma XL. Alterations in the microenvironment and the effects produced of TRPV5 in osteoporosis. J Transl Med 2023; 21:327. [PMID: 37198647 DOI: 10.1186/s12967-023-04182-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 05/05/2023] [Indexed: 05/19/2023] Open
Abstract
The pathogenesis of osteoporosis involves multiple factors, among which alterations in the bone microenvironment play a crucial role in disrupting normal bone metabolic balance. Transient receptor potential vanilloid 5 (TRPV5), a member of the TRPV family, is an essential determinant of the bone microenvironment, acting at multiple levels to influence its properties. TRPV5 exerts a pivotal influence on bone through the regulation of calcium reabsorption and transportation while also responding to steroid hormones and agonists. Although the metabolic consequences of osteoporosis, such as loss of bone calcium, reduced mineralization capacity, and active osteoclasts, have received significant attention, this review focuses on the changes in the osteoporotic microenvironment and the specific effects of TRPV5 at various levels.
Collapse
Affiliation(s)
- Zhi-Heng Luo
- Tianjin Hospital, Tianjin University, Jie Fang Nan Road 406, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, People's Republic of China
| | - Jian-Xiong Ma
- Tianjin Hospital, Tianjin University, Jie Fang Nan Road 406, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, People's Republic of China
| | - Wei Zhang
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xue Yuan Avenue, Shenzhen University Town, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Ai-Xian Tian
- Tianjin Hospital, Tianjin University, Jie Fang Nan Road 406, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, People's Republic of China
| | - Shu-Wei Gong
- Tianjin Hospital, Tianjin University, Jie Fang Nan Road 406, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, People's Republic of China
| | - Yan Li
- Tianjin Hospital, Tianjin University, Jie Fang Nan Road 406, Tianjin, 300211, People's Republic of China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, People's Republic of China
| | - Yu-Xiao Lai
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xue Yuan Avenue, Shenzhen University Town, Shenzhen, 518055, Guangdong, People's Republic of China.
| | - Xin-Long Ma
- Tianjin Hospital, Tianjin University, Jie Fang Nan Road 406, Tianjin, 300211, People's Republic of China.
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, People's Republic of China.
| |
Collapse
|
10
|
3D osteogenic differentiation of human iPSCs reveals the role of TGFβ signal in the transition from progenitors to osteoblasts and osteoblasts to osteocytes. Sci Rep 2023; 13:1094. [PMID: 36658197 PMCID: PMC9852429 DOI: 10.1038/s41598-023-27556-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
Although the formation of bone-like nodules is regarded as the differentiation process from stem cells to osteogenic cells, including osteoblasts and osteocytes, the precise biological events during nodule formation are unknown. Here we performed the osteogenic induction of human induced pluripotent stem cells using a three-dimensional (3D) culture system using type I collagen gel and a rapid induction method with retinoic acid. Confocal and time-lapse imaging revealed the osteogenic differentiation was initiated with vigorous focal proliferation followed by aggregation, from which cells invaded the gel. Invading cells changed their morphology and expressed osteocyte marker genes, suggesting the transition from osteoblasts to osteocytes. Single-cell RNA sequencing analysis revealed that 3D culture-induced cells with features of periosteal skeletal stem cells, some of which expressed TGFβ-regulated osteoblast-related molecules. The role of TGFβ signal was further analyzed in the transition from osteoblasts to osteocytes, which revealed that modulation of the TGFβ signal changed the morphology and motility of cells isolated from the 3D culture, suggesting that the TGFβ signal maintains the osteoblastic phenotype and the transition into osteocytes requires down-regulation of the TGFβ signal.
Collapse
|
11
|
Smout D, Van Craenenbroeck AH, Jørgensen HS, Evenepoel P. MicroRNAs: emerging biomarkers and therapeutic targets of bone fragility in chronic kidney disease. Clin Kidney J 2022; 16:408-421. [PMID: 36865016 PMCID: PMC9972833 DOI: 10.1093/ckj/sfac219] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Indexed: 11/12/2022] Open
Abstract
Bone fragility is highly prevalent, yet underdiagnosed in patients with chronic kidney disease. Incomplete understanding of the pathophysiology and limitations of current diagnostics contribute to therapeutic hesitation, if not nihilism. This narrative review addresses the question of whether microRNAs (miRNAs) may improve therapeutic decision making in osteoporosis and renal osteodystrophy. miRNAs are key epigenetic regulators of bone homeostasis and show promise as both therapeutic targets and as biomarkers, primarily of bone turnover. Experimental studies show that miRNAs are involved in several osteogenic pathways. Clinical studies exploring the usefulness of circulating miRNAs for fracture risk stratification and for guiding and monitoring therapy are few and, so far, provide inconclusive results. Likely, (pre)analytical heterogeneity contributes to these equivocal results. In conclusion, miRNAs are promising in metabolic bone disease, both as a diagnostic tool and as therapeutic targets, but not yet ready for clinical prime time.
Collapse
Affiliation(s)
- Dieter Smout
- Department of Microbiology, Immunology and Transplantation; Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium,Department of Medicine, Division of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Amaryllis H Van Craenenbroeck
- Department of Microbiology, Immunology and Transplantation; Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium,Department of Medicine, Division of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Hanne Skou Jørgensen
- Department of Microbiology, Immunology and Transplantation; Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium,Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
12
|
Xue F, Cornelissen JJ, Yuan Q, Cao S. Delivery of MicroRNAs by plant virus-based nanoparticles to functionally alter the osteogenic differentiation of human mesenchymal stem cells. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.04.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Zhang C, Xia D, Li J, Zheng Y, Weng B, Mao H, Mei J, Wu T, Li M, Zhao J. BMSCs and Osteoblast-Engineered ECM Synergetically Promotes Osteogenesis and Angiogenesis in an Ectopic Bone Formation Model. Front Bioeng Biotechnol 2022; 10:818191. [PMID: 35127662 PMCID: PMC8814575 DOI: 10.3389/fbioe.2022.818191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/04/2022] [Indexed: 12/16/2022] Open
Abstract
Bone mesenchymal stem cells (BMSCs) have been extensively used in bone tissue engineering because of their potential to differentiate into multiple cells, secrete paracrine factors, and attenuate immune responses. Biomaterials are essential for the residence and activities of BMSCs after implantation in vivo. Recently, extracellular matrix (ECM) modification with a favorable regenerative microenvironment has been demonstrated to be a promising approach for cellular activities and bone regeneration. The aim of the present study was to evaluate the effects of BMSCs combined with cell-engineered ECM scaffolds on osteogenesis and angiogenesis in vivo. The ECM scaffolds were generated by osteoblasts on the small intestinal submucosa (SIS) under treatment with calcium (Ca)-enriched medium and icariin (Ic) after decellularization. In a mouse ectopic bone formation model, the SIS scaffolds were demonstrated to reduce the immune response, and lower the levels of immune cells compared with those in the sham group. Ca/Ic-ECM modification inhibited the degradation of the SIS scaffolds in vivo. The generated Ca/Ic-SIS scaffolds ectopically promoted osteogenesis according to the results of micro-CT and histological staining. Moreover, BMSCs on Ca/Ic-SIS further increased the bone volume percentage (BV/TV) and bone density. Moreover, angiogenesis was also enhanced by the Ca/Ic-SIS scaffolds, resulting in the highest levels of neovascularization according to the data ofCD31 staining. In conclusion, osteoblast-engineered ECM under directional induction is a promising strategy to modify biomaterials for osteogenesis and angiogenesis. BMSCs synergetically improve the properties of ECM constructs, which may contribute to the repair of large bone defects.
Collapse
Affiliation(s)
- Chi Zhang
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
- Medical Research Center, Ningbo City First Hospital, Ningbo, China
| | - Dongdong Xia
- Orthopedic Department, Ningbo City First Hospital, Ningbo, China
| | - Jiajing Li
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Yanan Zheng
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Bowen Weng
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Haijiao Mao
- Department of Orthopaedic Surgery, the Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Jing Mei
- Medical Research Center, Ningbo City First Hospital, Ningbo, China
| | - Tao Wu
- Cardiovascular Center, the Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Mei Li
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
- Ningbo Institute of Medical Sciences, Ningbo, China
- *Correspondence: Mei Li, ; Jiyuan Zhao,
| | - Jiyuan Zhao
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
- *Correspondence: Mei Li, ; Jiyuan Zhao,
| |
Collapse
|
14
|
Liu X, Bai M, Sun Y, Hu X, Wang C, Xie J, Ye L. FGF7-induced E11 facilitates cell-cell communication through connexin43. Int J Biol Sci 2021; 17:3862-3874. [PMID: 34671204 PMCID: PMC8495393 DOI: 10.7150/ijbs.65240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/26/2021] [Indexed: 02/05/2023] Open
Abstract
Fibroblast growth factors (FGFs) include a large family of growth factors that play a critical role in maintaining bone homeostasis, but the specific role of its members such as FGF7 does not well understand. Osteoblasts are a kind of major cells essential for bone formation. Osteoblasts interact with one another to create the unique structure of osteons. The well-connected osteons constitute the cortical bone. As an early osteocyte marker that triggers actin cytoskeleton dynamics, E11 is essential for osteoblasts' dendrites formation. However, the upstream which regulates E11 is mainly unknown. The purpose of this study was to examine the influence of FGF7 on the expression and the distribution of E11 in osteoblasts, which mediated osteoblasts' processes formation and gap junctional intercellular communication (GJIC) partly through connexin43 (Cx43). We first demonstrated that FGF7 increased the expression of E11 in osteoblasts. We then showed that FGF7 promoted osteoblasts' dendrites elongation and functional gap junctions formation. Furthermore, E11 interacted directly with Cx43 in primary osteoblasts. MAPK pathway and PI3K-AKT pathway were involved in the effect of FGF7. Our results shed light on the unique role of FGF7 on osteoblasts, which may indicate that FGF7 plays a more significant role in the later stages of bone development and homeostasis.
Collapse
Affiliation(s)
- Xiaoyu Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Mingru Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yimin Sun
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xuchen Hu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chenglin Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
15
|
Wu Y, Zhou L, Li Y, Lou X. Osteoblast-derived extracellular matrix coated PLLA/silk fibroin composite nanofibers promote osteogenic differentiation of bone mesenchymal stem cells. J Biomed Mater Res A 2021; 110:525-534. [PMID: 34494712 DOI: 10.1002/jbm.a.37302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/08/2021] [Accepted: 08/16/2021] [Indexed: 11/09/2022]
Abstract
Poly-L-lactic acid (PLLA) is one of the most commonly used synthetic materials for regenerative medicine, and silk fibroin (SF) is a natural protein with excellent biocompatibility. Combination of PLLA and SF in a proper proportion by electrospinning may generate composite nanofibers that could meet the requirements of scaffolding in bone tissue engineering. The application of PLLA/SF nanofibrous scaffold for osteogenesis is well established in vitro and in vivo. However, PLLA/SF nanofibrous scaffold does not have an ideal ability to promote cell adhesion, proliferation, and differentiation. Extracellular matrix (ECM) plays a critical role in modulating cellular behavior. However, the role of combination of natural ECM with nanofibrous scaffold in regulating osteogenic differentiation is unclear. In this study, we aimed to develop a novel composite PLLA/SF nanofibrous scaffold coated with osteoblast-derived extracellular matrix (O-ECM/PLLA/SF) and analyze the effects of the modified scaffold on osteogenic differentiation of BMSCs. The surface structural features and compositions of the O-ECM/PLLA/SF scaffold were characterized by SEM and immunofluorescence staining. The capacities of the O-ECM/PLLA/SF scaffold to induce osteogenic differentiation of BMSCs were investigated by alkaline phosphatase (ALP) and alizarin red staining (ARS). The results showed BMSCs cultured on O-ECM/PLLA/SF scaffold significantly increased osteogenic differentiation compared with cells cultured individually on a scaffold or O-ECM. Collectively, these findings indicate that O-ECM-coated nanofibrous scaffold can be a promising strategy for osteogenic differentiation of BMSCs, opening a new possibility of utilizing composite scaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- Yunliang Wu
- College of Chemical Engineering & Biotechnology, Donghua University, Shanghai, China
| | - Lei Zhou
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuexia Li
- College of Chemical Engineering & Biotechnology, Donghua University, Shanghai, China
| | - Xiangxin Lou
- College of Chemical Engineering & Biotechnology, Donghua University, Shanghai, China
| |
Collapse
|
16
|
Hosseinpour S, Cao Y, Liu J, Xu C, Walsh LJ. Efficient transfection and long-term stability of rno-miRNA-26a-5p for osteogenic differentiation by large pore sized mesoporous silica nanoparticles. J Mater Chem B 2021; 9:2275-2284. [PMID: 33606863 DOI: 10.1039/d0tb02756a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MicroRNA (miRNA) based therapy for bone repair has shown promising results for regulating stem cell proliferation and differentiation, an efficient and stable vector for delivery of microRNA delivery is needed. The present study explored the stability and functionality of lyophilized mesoporous silica nanoparticles with core-cone structure and coated with polyethylenimine (MSN-CC-PEI) as a system for delivering Rattus norvegicus (rno)-miRNA-26a-5p into rat marrow mesenchymal cells (rBMSCs) to promote their osteogenic differentiation. We assessed the cellular uptake and transfection efficiency of nanoparticles loaded with labelled miRNA using confocal laser scanning microscopy and flow cytometry, and the cell viability using the MTT assay. The expression levels of osteogenic genes after one and two weeks were analysed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Extracellular matrix deposition and mineralization at 3 weeks were evaluated using Picro Sirius red and Alizarin red staining. We also assessed the performance of the delivery system after long term storage, by freeze drying rno-miRNA-26a-5p@MSN-CC-PEI with 5% trehalose and keeping them at -30 °C for 3 and 6 months. Osteogenic differentiation, matrix deposition, and mineralization were all significantly increased by rno-miRNA-26a-5p. In addition, this enhancement was not significantly altered by lyophilization and storage. Overall, these findings support the concept of MSN-CC-PEI as a delivery system for gene therapy. The complex of rno-miRNA-26a-5p@MSN-CC-PEI could efficiently transfect rBMSCs and enhance their osteogenic differentiation. In addition, the lyophilized complexes remain functional after 6 months of storage.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, The University of Queensland, Herston QLD 4006, Australia.
| | - Yuxue Cao
- School of Dentistry, The University of Queensland, Herston QLD 4006, Australia.
| | - Jingyu Liu
- Taiyuan University of Technology, Taiyuan, 030024, China
| | - Chun Xu
- School of Dentistry, The University of Queensland, Herston QLD 4006, Australia.
| | - Laurence J Walsh
- School of Dentistry, The University of Queensland, Herston QLD 4006, Australia.
| |
Collapse
|
17
|
Mähr M, Blouin S, Behanova M, Misof BM, Glorieux FH, Zwerina J, Rauch F, Hartmann MA, Fratzl-Zelman N. Increased Osteocyte Lacunae Density in the Hypermineralized Bone Matrix of Children with Osteogenesis Imperfecta Type I. Int J Mol Sci 2021; 22:ijms22094508. [PMID: 33925942 PMCID: PMC8123504 DOI: 10.3390/ijms22094508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 12/23/2022] Open
Abstract
Osteocytes are terminally differentiated osteoblasts embedded within the bone matrix and key orchestrators of bone metabolism. However, they are generally not characterized by conventional bone histomorphometry because of their location and the limited resolution of light microscopy. OI is characterized by disturbed bone homeostasis, matrix abnormalities and elevated bone matrix mineralization density. To gain further insights into osteocyte characteristics and bone metabolism in OI, we evaluated 2D osteocyte lacunae sections (OLS) based on quantitative backscattered electron imaging in transiliac bone biopsy samples from children with OI type I (n = 19) and age-matched controls (n = 24). The OLS characteristics were related to previously obtained, re-visited histomorphometric parameters. Moreover, we present pediatric bone mineralization density distribution reference data in OI type I (n = 19) and controls (n = 50) obtained with a field emission scanning electron microscope. Compared to controls, OI has highly increased OLS density in cortical and trabecular bone (+50.66%, +61.73%; both p < 0.001), whereas OLS area is slightly decreased in trabecular bone (−10.28%; p = 0.015). Correlation analyses show a low to moderate, positive association of OLS density with surface-based bone formation parameters and negative association with indices of osteoblast function. In conclusion, hyperosteocytosis of the hypermineralized OI bone matrix associates with abnormal bone cell metabolism and might further impact the mechanical competence of the bone tissue.
Collapse
Affiliation(s)
- Matthias Mähr
- 1st Medical Department, Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, Hanusch Hospital, 1140 Vienna, Austria; (M.M.); (S.B.); (M.B.); (B.M.M.); (J.Z.); (M.A.H.)
| | - Stéphane Blouin
- 1st Medical Department, Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, Hanusch Hospital, 1140 Vienna, Austria; (M.M.); (S.B.); (M.B.); (B.M.M.); (J.Z.); (M.A.H.)
| | - Martina Behanova
- 1st Medical Department, Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, Hanusch Hospital, 1140 Vienna, Austria; (M.M.); (S.B.); (M.B.); (B.M.M.); (J.Z.); (M.A.H.)
| | - Barbara M. Misof
- 1st Medical Department, Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, Hanusch Hospital, 1140 Vienna, Austria; (M.M.); (S.B.); (M.B.); (B.M.M.); (J.Z.); (M.A.H.)
| | - Francis H. Glorieux
- Genetics Unit, Shriners Hospital for Children and McGill University, Montreal, ON H4A 0A9, Canada; (F.H.G.); (F.R.)
| | - Jochen Zwerina
- 1st Medical Department, Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, Hanusch Hospital, 1140 Vienna, Austria; (M.M.); (S.B.); (M.B.); (B.M.M.); (J.Z.); (M.A.H.)
| | - Frank Rauch
- Genetics Unit, Shriners Hospital for Children and McGill University, Montreal, ON H4A 0A9, Canada; (F.H.G.); (F.R.)
| | - Markus A. Hartmann
- 1st Medical Department, Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, Hanusch Hospital, 1140 Vienna, Austria; (M.M.); (S.B.); (M.B.); (B.M.M.); (J.Z.); (M.A.H.)
| | - Nadja Fratzl-Zelman
- 1st Medical Department, Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, Hanusch Hospital, 1140 Vienna, Austria; (M.M.); (S.B.); (M.B.); (B.M.M.); (J.Z.); (M.A.H.)
- Correspondence: ; Tel.: +43-5-9393-55770
| |
Collapse
|
18
|
Chen K, Zhou Q, Kang H, Yan Y, Qian N, Li C, Wang F, Yang K, Deng L, Qi J. High Mineralization Capacity of IDG-SW3 Cells in 3D Collagen Hydrogel for Bone Healing in Estrogen-Deficient Mice. Front Bioeng Biotechnol 2020; 8:864. [PMID: 32984264 PMCID: PMC7488085 DOI: 10.3389/fbioe.2020.00864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/06/2020] [Indexed: 01/10/2023] Open
Abstract
Tissue engineering with 3D scaffold is a simple and effective method for bone healing after large-scale bone loss. So far, bone marrow-derived mesenchymal stem cells (BMSCs) are mostly used in the treatment of bone healing in animal models due to their self-renewal capability and osteogenic potential. Due to the fact that the main functional cells in promoting osteoid mineralization and bone remodeling were osteocytes, we chose an osteoblast-to-osteocyte transition cell line, IDG-SW3, which are not proliferative under physiological conditions, and compared the healing capability of these cells to that of BMSCs in bone defect. In vitro, IDG-SW3 cells revealed a stronger mineralization capacity when grown in 3D collagen gel, compared to that of BMSCs. Although both BMSC and IDG-SW3 can generate stable calcium-phosphate crystal similar to hydroxyapatite (HA), the content was much more enriched in IDG-SW3-mixed collagen gel. Moreover, the osteoclasts co-cultured with IDG-SW3-mixed collagen gel were easier to be activated, indicating that the IDG-SW3 grafting could promote the bone remodeling more efficiently in vivo. Last, in order to reduce the self-healing capability, we assessed the healing capability between the IDG-SW3 cells and BMSCs in osteoporotic mice. We found that the collagen hydrogel mixed with IDG-SW3 cells has a better healing pattern than what was seen in hydrogel mixed with BMSCs. Therefore, these results demonstrated that by promoting osteoblast-to-osteocyte transition, the therapeutic effect of BMSCs in bone defect repair could be improved.
Collapse
Affiliation(s)
- Kaizhe Chen
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Zhou
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Kang
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yufei Yan
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Niandong Qian
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changwei Li
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Wang
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Yang
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lianfu Deng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Qi
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Bone, a Secondary Growth Site of Breast and Prostate Carcinomas: Role of Osteocytes. Cancers (Basel) 2020; 12:cancers12071812. [PMID: 32640686 PMCID: PMC7408809 DOI: 10.3390/cancers12071812] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Bone is the primarily preferred site for breast and prostate cancer to metastasize. Bone metastases are responsible for most deaths related to breast and prostate cancer. The bone's particular microenvironment makes it conducive for the growth of cancer cells. Studies on bone metastasis have focused on the interaction between cancer cells and the bone microenvironment. Osteocytes, the most common cell type of bone tissue, have received little attention in bone metastasis, although they are master signal sensors, integrators, and skeleton transducers. They play an important role in regulating bone mass by acting on both osteoblasts and osteoclasts, through the release of proteins such as sclerostin, Dickkopf-1 (DKK-1), and fibroblast growth factor 23 (FGF23). Osteocytes have been extensively re-evaluated, in light of their multiple functions: with different experimental approaches, it has been shown that, indeed, osteocytes are actively involved in the colonization of bone tissue by cancer cells. The present review focuses on recent research on the role that osteocytes play in bone metastasis of breast and prostate cancers. Moreover, the studies here summarized open up perspectives for new therapeutic approaches focused on modulating the activity of osteocytes to improve the condition of the bone metastatic patients. A better understanding of the complex interactions between cancer cells and bone-resident cells is indispensable for identifying potential therapeutic targets to stop tumor progression and prevent bone metastases.
Collapse
|
20
|
Hardy E, Fernandez-Patron C. Destroy to Rebuild: The Connection Between Bone Tissue Remodeling and Matrix Metalloproteinases. Front Physiol 2020; 11:47. [PMID: 32116759 PMCID: PMC7013034 DOI: 10.3389/fphys.2020.00047] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
Bone is a dynamic organ that undergoes constant remodeling, an energetically costly process by which old bone is replaced and localized bone defects are repaired to renew the skeleton over time, thereby maintaining skeletal health. This review provides a general overview of bone’s main players (bone lining cells, osteocytes, osteoclasts, reversal cells, and osteoblasts) that participate in bone remodeling. Placing emphasis on the family of extracellular matrix metalloproteinases (MMPs), we describe how: (i) Convergence of multiple protease families (including MMPs and cysteine proteinases) ensures complexity and robustness of the bone remodeling process, (ii) Enzymatic activity of MMPs affects bone physiology at the molecular and cellular levels and (iii) Either overexpression or deficiency/insufficiency of individual MMPs impairs healthy bone remodeling and systemic metabolism. Today, it is generally accepted that proteolytic activity is required for the degradation of bone tissue in osteoarthritis and osteoporosis. However, it is increasingly evident that inactivating mutations in MMP genes can also lead to bone pathology including osteolysis and metabolic abnormalities such as delayed growth. We argue that there remains a need to rethink the role played by proteases in bone physiology and pathology.
Collapse
Affiliation(s)
| | - Carlos Fernandez-Patron
- Department of Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
21
|
Guder C, Gravius S, Burger C, Wirtz DC, Schildberg FA. Osteoimmunology: A Current Update of the Interplay Between Bone and the Immune System. Front Immunol 2020; 11:58. [PMID: 32082321 PMCID: PMC7004969 DOI: 10.3389/fimmu.2020.00058] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Immunology, already a discipline in its own right, has become a major part of many different medical fields. However, its relationship to orthopedics and trauma surgery has unfortunately, and perhaps unjustly, been developing rather slowly. Discoveries in recent years have emphasized the immense breadth of communication and connection between both systems and, importantly, the highly promising therapeutic opportunities. Recent discoveries of factors originally assigned to the immune system have now also been shown to have a significant impact on bone health and disease, which has greatly changed how we approach treatment of bone pathologies. In case of bone fracture, immune cells, especially macrophages, are present throughout the whole healing process, assure defense against pathogens and discharge a complex variety of effectors to regulate bone modeling. In rheumatoid arthritis and osteoporosis, the immune system contributes to the formation of the pathological and chronic conditions. Fascinatingly, prosthesis failure is not at all solely a mechanical problem of improper strain but works in conjunction with an active contribution of the immune system as a reaction to irritant debris from material wear. Unraveling conjoined mechanisms of the immune and osseous systems heralds therapeutic possibilities for ailments of both. Contemplation of the bone as merely an unchanging support pillar is outdated and obsolete. Instead it is mandatory that this highly diverse network be incorporated in our understanding of the immune system and hematopoiesis.
Collapse
Affiliation(s)
- Christian Guder
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Sascha Gravius
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany.,Department of Orthopedics and Trauma Surgery, University Medical Center Mannheim of University Heidelberg, Mannheim, Germany
| | - Christof Burger
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C Wirtz
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
22
|
Yan J, Lu X, Zhu X, Hu X, Wang L, Qian J, Zhang F, Liu M. Effects of miR-26a on Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells by a Mesoporous Silica Nanoparticle - PEI - Peptide System. Int J Nanomedicine 2020; 15:497-511. [PMID: 32158207 PMCID: PMC6986258 DOI: 10.2147/ijn.s228797] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/28/2019] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION RNA-based therapy for bone repair and regeneration is a highly safe and effective approach, which has been extensively investigated in recent years. However, the molecular stability of RNA agents still remains insufficient for clinical application. High porosity, tunable size, and ideal biodegradability and biosafety are a few of the characters of mesoporous silicon nanoparticles (MSNs) that render them a promising biomaterial carrier for RNA treatment. MATERIALS AND METHODS In this study, a novel miR-26a delivery system was constructed based on MSNs. Next, we assessed the miRNA protection of the delivery vehicles. Then, rat bone marrow mesenchymal stem cells (rBMSCs) were incubated with the vectors, and the transfection efficiency, cellular uptake, and effects on cell viability and osteogenic differentiation were evaluated. RESULTS The results demonstrated that the vectors protected miR-26a from degradation in vitro and delivered it into the cytoplasm. A relatively low concentration of the delivery systems significantly increased osteogenic differentiation of rBMSCs. CONCLUSION The vectors constructed in our study provide new methods and strategies for the delivery of microRNAs in bone tissue engineering.
Collapse
Affiliation(s)
- Jia Yan
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing210029, People’s Republic of China
| | - Xiaoli Lu
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing210029, People’s Republic of China
| | - Xinchen Zhu
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing210029, People’s Republic of China
| | - Xiaokun Hu
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing210029, People’s Republic of China
| | - Lili Wang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing210029, People’s Republic of China
| | - Jun Qian
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing210029, People’s Republic of China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing210029, People’s Republic of China
| | - Mei Liu
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing210029, People’s Republic of China
| |
Collapse
|
23
|
Shiozawa Y. The Roles of Bone Marrow-Resident Cells as a Microenvironment for Bone Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1226:57-72. [PMID: 32030676 DOI: 10.1007/978-3-030-36214-0_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
It has been appreciated that the cross talk between bone metastatic cancer cells and bone marrow microenvironment influence one another to worsen bone metastatic disease progression. Bone marrow contains various cell types, including (1) cells of mesenchymal origin (e.g., osteoblasts, osteocytes, and adipocytes), (2) cells of hematopoietic origin (e.g., osteoclast and immune cells), and (3) others (e.g., endothelial cells and nerves). The recent studies have enabled us to discover many important cancer-derived factors responsible for the development of bone metastasis. However, many critical questions regarding the roles of bone microenvironment in bone metastatic progression remain elusive. To answer these questions, a deeper understanding of the cross talk between bone metastatic cancer and bone marrow microenvironment is clearly warranted.
Collapse
Affiliation(s)
- Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC, USA.
| |
Collapse
|
24
|
Effects of ionizing radiation on woven bone: influence on the osteocyte lacunar network, collagen maturation, and microarchitecture. Clin Oral Investig 2019; 24:2763-2771. [PMID: 31732880 DOI: 10.1007/s00784-019-03138-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/24/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Evaluate the effects of ionizing radiation on microarchitecture, the osteocyte lacunar network, and collagen maturity in a bone repair site. MATERIALS AND METHODS Bone defects were created on tibias of 20 New Zealand rabbits. After 2 weeks, the animals were randomly divided into (n = 10) NoIr (nonirradiated group) and Ir (irradiated group). In the Ir, the animals received single-dose irradiation of 30 Gy on the tibia and were euthanized after 2 weeks. Bone microarchitecture parameters were analyzed by using micro-CT, and the osteocyte lacunar network, bone matrix, and collagen maturation by histomorphometric analysis. The data were analyzed using unpaired Student's t test (α = 0.05). RESULTS Trabecular thickness in Ir was lower than that in NoIr (P = 0.028). No difference was found for bone volume fraction and bone area. Lacunae filled with osteocytes were more numerous (P < 0.0001) in NoIr (2.6 ± 0.6) than in Ir (1.97 ± 0.53). Empty lacunae were more prevalent (P < 0.003) in Ir (0.14 ± 0.10) than in NoIr (0.1 ± 0.1). The mean osteocyte lacunae size was higher (P < 0.01) in Ir (15.4 ± 4.41) than in NoIr (12.7 ± 3.7). Picrosirius red analysis showed more (P < 0.05) mature collagen in NoIr (29.0 ± 5.3) than in Ir (23.4 ± 4.5). Immature collagen quantification revealed no difference between groups. CONCLUSIONS Ionizing radiation compromised bone formation and an impairment in bone repair in irradiated woven bone was observed. CLINICAL RELEVANCE Before radiotherapy, patients usually need surgical intervention, which may be better performed, if clinicians understand the repair process in irradiated bone, using novel approaches for treating these individuals.
Collapse
|
25
|
|
26
|
Liang X, Gao J, Xu W, Wang X, Shen Y, Tang J, Cui S, Yang X, Liu Q, Yu L, Ding J. Structural mechanics of 3D-printed poly(lactic acid) scaffolds with tetragonal, hexagonal and wheel-like designs. Biofabrication 2019; 11:035009. [DOI: 10.1088/1758-5090/ab0f59] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
27
|
Lee J, Byun H, Madhurakkat Perikamana SK, Lee S, Shin H. Current Advances in Immunomodulatory Biomaterials for Bone Regeneration. Adv Healthc Mater 2019; 8:e1801106. [PMID: 30328293 DOI: 10.1002/adhm.201801106] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/19/2018] [Indexed: 12/14/2022]
Abstract
Biomaterials with suitable surface modification strategies are contributing significantly to the rapid development of the field of bone tissue engineering. Despite these encouraging results, utilization of biomaterials is poorly translated to human clinical trials potentially due to lack of knowledge about the interaction between biomaterials and the body defense mechanism, the "immune system". The highly complex immune system involves the coordinated action of many immune cells that can produce various inflammatory and anti-inflammatory cytokines. Besides, bone fracture healing initiates with acute inflammation and may later transform to a regenerative or degenerative phase mainly due to the cross-talk between immune cells and other cells in the bone regeneration process. Among various immune cells, macrophages possess a significant role in the immune defense, where their polarization state plays a key role in the wound healing process. Growing evidence shows that the macrophage polarization state is highly sensitive to the biomaterial's physiochemical properties, and advances in biomaterial research now allow well controlled surface properties. This review provides an overview of biomaterial-mediated modulation of the immune response for regulating key bone regeneration events, such as osteogenesis, osteoclastogenesis, and inflammation, and it discusses how these strategies can be utilized for future bone tissue engineering applications.
Collapse
Affiliation(s)
- Jinkyu Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Hayeon Byun
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | | | - Sangmin Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| |
Collapse
|