1
|
Ganko R, Madhavan A, Hamouda W, Muthu S, Jain A, Yoon ST, El-Rozz H, Cyril D, Pabbruwe M, Tipper JL, Tavakoli J. Spinal implant wear particles: Generation, characterization, biological impacts, and future considerations. iScience 2025; 28:112193. [PMID: 40212584 PMCID: PMC11982499 DOI: 10.1016/j.isci.2025.112193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
The generation of wear debris from orthopedic implants is a known cause of implant failure, particularly in joint replacements. While much research has focused on wear particles from knee and hip implants, spinal implants, such as total disc replacements (TDRs), have received less attention despite their increasing clinical use. Spinal implants face unique biomechanical challenges, including a wider range of motion and higher loads, leading to complex tissue interactions. Studies reveal that TDR wear particles, though similar in size to those from knee implants, cause a stronger immune response, with more macrophages and giant cells found in the surrounding tissue. This may explain the high revision rates seen in spinal surgeries, with some interventions failing in over 30% of cases within 10 years. The younger population undergoing spinal surgery, combined with the productivity losses associated with implant failure, underscores the need for greater understanding. This review discusses recent research on the generation, characterization, and biological impacts of spinal implant wear debris. It draws on retrieval analysis, wear simulation, in vivo models, and a survey conducted with the AO Spine Knowledge Forum Degenerative to assess current clinical practices and highlight gaps in knowledge. Additionally, this critical review explores future strategies to reduce the biological impact of wear particles and improve the safety and longevity of spinal implants through better therapeutics and design innovations. By combining literature and clinical insights, this paper aims to guide future research in addressing the complexities of spinal implant wear and its biological consequences.
Collapse
Affiliation(s)
- Renata Ganko
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Aswini Madhavan
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Waeel Hamouda
- Department of Neurosurgery, Kasr Alainy Faculty of Medicine, Research, and Teaching Hospitals, Cairo University, Cairo, Egypt
- Department of Neurosurgery, Security Forces Hospital, Dammam, Saudi Arabia
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College, Karur, India
- Orthopaedic Research Group, Coimbatore, Tamil Nadu, India
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Amit Jain
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - S. Tim Yoon
- Department of Orthopaedic Surgery, Emory University, Atlanta, GA, USA
| | - Hiba El-Rozz
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Divya Cyril
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Moreica Pabbruwe
- Centre for Implant Retrieval and Analysis, Royal Perth Hospital, Perth, WA, Australia
| | - Joanne L. Tipper
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
- School of Engineering, RMIT University, Melbourne, VIC 3001, Australia
- School of Mechanical Engineering, University of Leeds, Leads, UK
| | - Javad Tavakoli
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
- School of Engineering, RMIT University, Melbourne, VIC 3001, Australia
| |
Collapse
|
2
|
Li X, Shen H, Zhang M, Teissier V, Huang EE, Gao Q, Tsubosaka M, Toya M, Kushioka J, Maduka CV, Contag CH, Chow SKH, Zhang N, Goodman SB. Glycolytic reprogramming in macrophages and MSCs during inflammation. Front Immunol 2023; 14:1199751. [PMID: 37675119 PMCID: PMC10477714 DOI: 10.3389/fimmu.2023.1199751] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/21/2023] [Indexed: 09/08/2023] Open
Abstract
Background Dysregulated inflammation is associated with many skeletal diseases and disorders, such as osteolysis, non-union of fractures, osteonecrosis, osteoarthritis and orthopaedic infections. We previously showed that continuous infusion of lipopolysaccharide (LPS) contaminated polyethylene particles (cPE) caused prolonged inflammation and impaired bone formation. However, the metabolic and bioenergetic processes associated with inflammation of bone are unknown. Mitochondria are highly dynamic organelles that modulate cell metabolism and orchestrate the inflammatory responses that involve both resident and recruited cells. Glycolytic reprogramming, the shift from oxidative phosphorylation (OXPHOS) to glycolysis causes inappropriate cell activation and function, resulting in dysfunctional cellular metabolism. We hypothesized that impaired immunoregulation and bone regeneration from inflammatory states are associated with glycolytic reprogramming and mitochondrial dysfunction in macrophages (Mφ) and mesenchymal stromal cells (MSCs). Methods We used the Seahorse XF96 analyzer and real-time qPCR to study the bioenergetics of Mφ and MSCs exposed to cPE. To understand the oxygen consumption rate (OCR), we used Seahorse XF Cell Mito Stress Test Kit with Seahorse XF96 analyzer. Similarly, Seahorse XF Glycolytic Rate Assay Kit was used to detect the extracellular acidification rate (ECAR) and Seahorse XF Real-Time ATP Rate Assay kit was used to detect the real-time ATP production rates from OXPHOS and glycolysis. Real-time qPCR was performed to analyze the gene expression of key enzymes in glycolysis and mitochondrial biogenesis. We further detected the gene expression of proinflammatory cytokines in Mφ and genes related to cell differentiation in MSC during the challenge of cPE. Results Our results demonstrated that the oxidative phosphorylation of Mφ exposed to cPE was significantly decreased when compared with the control group. We found reduced basal, maximal and ATP-production coupled respiration rates, and decreased proton leak in Mφ during challenge with cPE. Meanwhile, Mφ showed increased basal glycolysis and proton efflux rates (PER) when exposed to cPE. The percentage (%) of PER from glycolysis was higher in Mφ exposed to cPE, indicating that the contribution of the glycolytic pathway to total extracellular acidification was elevated during the challenge of cPE. In line with the results of OCR and ECAR, we found Mφ during cPE challenge showed higher glycolytic ATP (glycoATP) production rates and lower mitochondrial ATP (mitoATP) production rates which is mainly from OXPHOS. Interestingly, MSCs showed enhanced glycolysis during challenge with cPE, but no significant changes in oxygen consumption rates (OCR). In accordance, seahorse assay of real-time ATP revealed glycoATP rates were elevated while mitoATP rates showed no significant differences in MSC during challenge with cPE. Furthermore, Mφ and MSCs exposed to cPE showed upregulated gene expression levels of glycolytic regulators and Mφ exposed to cPE expressed higher levels of pro-inflammatory cytokines. Conclusion This study demonstrated the dysfunctional bioenergetic activity of bone marrow-derived Mφ and MSCs exposed to cPE, which could impair the immunoregulatory properties of cells in the bone niche. The underlying molecular defect related to disordered mitochondrial function could represent a potential therapeutic target during the resolution of inflammation.
Collapse
Affiliation(s)
- Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Orthopaedic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mao Zhang
- Cardiovascular Institute Operations, Stanford University School of Medicine, Stanford, CA, United States
| | - Victoria Teissier
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Ejun Elijah Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Chima V. Maduka
- Departments of Biomedical Engineering and Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Christopher H. Contag
- Departments of Biomedical Engineering and Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
3
|
Kushioka J, Toya M, Shen H, Hirata H, Zhang N, Huang E, Tsubosaka M, Gao Q, Teissier V, Li X, Utsunomiya T, Goodman SB. Therapeutic effects of MSCs, genetically modified MSCs, and NFĸB-inhibitor on chronic inflammatory osteolysis in aged mice. J Orthop Res 2023; 41:1004-1013. [PMID: 36031590 PMCID: PMC9971358 DOI: 10.1002/jor.25434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 02/04/2023]
Abstract
The number of total joint replacements is increasing, especially in elderly patients, and so too are implant-related complications such as prosthesis loosening. Wear particles from the prosthesis induce a chronic inflammatory reaction and subsequent osteolysis, leading to the need for revision surgery. This study investigated the therapeutic effect of NF-ĸB decoy oligodeoxynucleotides (ODN), mesenchymal stem cells (MSCs), and genetically-modified NF-ĸB sensing interleukin-4 over-secreting MSCs (IL4-MSCs) on chronic inflammation in aged mice. The model was generated by continuous infusion of contaminated polyethylene particles into the intramedullary space of the distal femur of aged mice (15-17 months old) for 6 weeks. Local delivery of ODN showed increased bone mineral density (BMD), decreased osteoclast-like cells, increased alkaline phosphatase (ALP)-positive area, and increased M2/M1 macrophage ratio. Local injection of MSCs and IL4-MSCs significantly decreased osteoclast-like cells and increased the M2/M1 ratio, with a greater trend for IL4-MSCs than MSCs. MSCs significantly increased ALP-positive area and BMD values compared with the control. The IL4-MSCs demonstrated higher values for both ALP-positive area and BMD. These findings demonstrated the therapeutic effects of ODN, MSCs, and IL4-MSCs on chronic inflammatory osteolysis in aged mice. The two MSC-based therapies were more effective than ODN in increasing the M2/M1 macrophage ratio, reducing bone resorption, and increasing bone formation. Specifically, MSCs were more effective in increasing bone formation, and IL4-MSCs were more effective in mitigating inflammation. This study suggests potential therapeutic strategies for treating wear particle-associated inflammatory osteolysis after arthroplasty in the elderly.
Collapse
Affiliation(s)
- Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Hirohito Hirata
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Ejun Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Victoria Teissier
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | | | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
4
|
Wu Z, Li X, Chen X, He X, Chen Y, Zhang L, Li Z, Yang M, Yuan G, Shi B, Chen N, Li N, Feng H, Zhou M, Rui G, Xu F, Xu R. Phosphatidyl Inositol 3-Kinase (PI3K)-Inhibitor CDZ173 protects against LPS-induced osteolysis. Front Pharmacol 2023; 13:1021714. [PMID: 36686650 PMCID: PMC9854393 DOI: 10.3389/fphar.2022.1021714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/03/2022] [Indexed: 01/09/2023] Open
Abstract
A major complication of a joint replacement is prosthesis loosening caused by inflammatory osteolysis, leading to the revision of the operation. This is due to the abnormal activation of osteoclast differentiation and function caused by periprosthetic infection. Therefore, targeting abnormally activated osteoclasts is still effective for treating osteolytic inflammatory diseases. CDZ173 is a selective PI3K inhibitor widely used in autoimmune-related diseases and inflammatory diseases and is currently under clinical development. However, the role and mechanism of CDZ173 in osteoclast-related bone metabolism remain unclear. The possibility for treating aseptic prosthesis loosening brought on by inflammatory osteolysis illness can be assessed using an LPS-induced mouse cranial calcium osteolysis model. In this study, we report for the first time that CDZ173 has a protective effect on LPS-induced osteolysis. The data show that this protective effect is due to CDZ173 inhibiting the activation of osteoclasts in vivo. Meanwhile, our result demonstrated that CDZ173 had a significant inhibitory effect on RANKL-induced osteoclasts. Furthermore, using the hydroxyapatite resorption pit assay and podosol actin belt staining, respectively, the inhibitory impact of CDZ173 on bone resorption and osteoclast fusion of pre-OC was determined. In addition, staining with alkaline phosphatase (ALP) and alizarin red (AR) revealed that CDZ173 had no effect on osteoblast development in vitro. Lastly, CDZ173 inhibited the differentiation and function of osteoclasts by weakening the signal axis of PI3K-AKT/MAPK-NFATc1 in osteoclasts. In conclusion, our results highlight the potential pharmacological role of CDZ173 in preventing osteoclast-mediated inflammatory osteolysis and its potential clinical application.
Collapse
Affiliation(s)
- Zuoxing Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Xuedong Li
- Department of Medical Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Xiaohui Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Xuemei He
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, China
| | - Yu Chen
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, China
| | - Long Zhang
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, China
| | - Zan Li
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Mengyu Yang
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, China
| | - Guixin Yuan
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, China
| | - Baohong Shi
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, China
| | - Ning Chen
- Department of Endocrinology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Na Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Haotian Feng
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot, China
| | - Mengyu Zhou
- Department of Dentistry, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gang Rui
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Feng Xu
- Department of Subject Planning, Ninth People's Hospital Shanghai, Jiaotong University School of Medicine, Shanghai, China
| | - Ren Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.,Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
5
|
Connors JP, Stelzer JW, Garvin PM, Wellington IJ, Solovyova O. The Role of the Innate Immune System in Wear Debris-Induced Inflammatory Peri-Implant Osteolysis in Total Joint Arthroplasty. Bioengineering (Basel) 2022; 9:764. [PMID: 36550970 PMCID: PMC9774505 DOI: 10.3390/bioengineering9120764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Periprosthetic osteolysis remains a leading complication of total hip and knee arthroplasty, often resulting in aseptic loosening of the implant and necessitating revision surgery. Wear-induced particulate debris is the main cause initiating this destructive process. The purpose of this article is to review recent advances in understanding of how wear debris causes osteolysis, and emergent strategies for the avoidance and treatment of this disease. A strong activator of the peri-implant innate immune this debris-induced inflammatory cascade is dictated by macrophage secretion of TNF-α, IL-1, IL-6, and IL-8, and PGE2, leading to peri-implant bone resorption through activation of osteoclasts and inhibition of osteoblasts through several mechanisms, including the RANK/RANKL/OPG pathway. Therapeutic agents against proinflammatory mediators, such as those targeting tumor necrosis factor (TNF), osteoclasts, and sclerostin, have shown promise in reducing peri-implant osteolysis in vitro and in vivo; however, radiographic changes and clinical diagnosis often lag considerably behind the initiation of osteolysis, making timely treatment difficult. Considerable efforts are underway to develop such diagnostic tools, therapies, and identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- John Patrick Connors
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - John W Stelzer
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - Patrick M Garvin
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - Ian J Wellington
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| | - Olga Solovyova
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06032, USA
| |
Collapse
|
6
|
Novel structural designs of 3D-printed osteogenic graft for rapid angiogenesis. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00212-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
7
|
Zhang N, Utsunomiya T, Lin T, Kohno Y, Ueno M, Maruyama M, Huang E, Rhee C, Yao Z, Goodman SB. Mesenchymal Stem Cells and NF-κB Sensing Interleukin-4 Over-Expressing Mesenchymal Stem Cells Are Equally Effective in Mitigating Particle-Associated Chronic Inflammatory Bone Loss in Mice. Front Cell Dev Biol 2021; 9:757830. [PMID: 34722543 PMCID: PMC8551755 DOI: 10.3389/fcell.2021.757830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/27/2021] [Indexed: 12/25/2022] Open
Abstract
Wear particles from total joint arthroplasties (TJAs) induce chronic inflammation, macrophage infiltration and lead to bone loss by promoting bone destruction and inhibiting bone formation. Inhibition of particle-associated chronic inflammation and the associated bone loss is critical to the success and survivorship of TJAs. The purpose of this study is to test the hypothesis that polyethylene particle induced chronic inflammatory bone loss could be suppressed by local injection of NF-κB sensing Interleukin-4 (IL-4) over-expressing MSCs using the murine continuous polyethylene particle infusion model. The animal model was generated with continuous infusion of polyethylene particles into the intramedullary space of the femur for 6 weeks. Cells were locally injected into the intramedullary space 3 weeks after the primary surgery. Femurs were collected 6 weeks after the primary surgery. Micro-computational tomography (μCT), histochemical and immunohistochemical analyses were performed. Particle-infusion resulted in a prolonged pro-inflammatory M1 macrophage dominated phenotype and a decrease of the anti-inflammatory M2 macrophage phenotype, an increase in TRAP positive osteoclasts, and lower alkaline phosphatase staining area and bone mineral density, indicating chronic particle-associated inflammatory bone loss. Local injection of MSCs or NF-κB sensing IL-4 over-expressing MSCs reversed the particle-associated chronic inflammatory bone loss and facilitated bone healing. These results demonstrated that local inflammatory bone loss can be effectively modulated via MSC-based treatments, which could be an efficacious therapeutic strategy for periprosthetic osteolysis.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Yusuke Kohno
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Ejun Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
8
|
Utsunomiya T, Zhang N, Lin T, Kohno Y, Ueno M, Maruyama M, Huang E, Rhee C, Yao Z, Goodman SB. Suppression of NF-κB-induced chronic inflammation mitigates inflammatory osteolysis in the murine continuous polyethylene particle infusion model. J Biomed Mater Res A 2021; 109:1828-1839. [PMID: 33779115 PMCID: PMC8373802 DOI: 10.1002/jbm.a.37175] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 03/16/2021] [Indexed: 12/18/2022]
Abstract
Wear particle-associated bone loss (periprosthetic osteolysis) constrains the longevity of total joint arthroplasty (TJA). Wear particles induce a prolonged upregulation of nuclear factor kappa B (NF-κB) signaling in macrophages and osteoclasts. Synthetic double-stranded oligodeoxynucleotides (ODNs) can prevent the binding of NF-κB to the promoter regions of targeted genes and inhibit genetic activation. We tested the hypothesis that polyethylene-particle induced chronic inflammatory bone loss could be suppressed by local delivery of NF-κB decoy ODNs in murine in vivo model. Polyethylene particles were continuously infused into the medullary cavity of the distal femur for 6 weeks to induce chronic inflammation, and micro-computational tomography and immunohistochemical analysis were performed. Particle-induced chronic inflammation resulted in lower BMD values, an increase in osteoclastogenesis and nuclear translocation of p65, a prolonged M1 pro-inflammatory macrophage phenotype, and a decrease of M2 anti-inflammatory macrophage phenotype. Delayed timing of local infusion of NF-κB decoy ODN for the last 3 weeks reversed polyethylene-particle associated chronic inflammatory bone loss and facilitated bone healing. This study demonstrated that polyethylene-particle associated chronic inflammatory osteolysis can be effectively modulated via interference with the NF-κB pathway; this minimally invasive intervention could potentially be an efficacious therapeutic strategy for periprosthetic osteolysis after TJA.
Collapse
Affiliation(s)
- Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Yusuke Kohno
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Ejun Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
- Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
9
|
Zhao F, Cang D, Zhang J, Zheng L. Chemerin/ChemR23 signaling mediates the effects of ultra-high molecular weight polyethylene wear particles on the balance between osteoblast and osteoclast differentiation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1149. [PMID: 34430590 PMCID: PMC8350637 DOI: 10.21037/atm-21-2945] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/15/2021] [Indexed: 02/02/2023]
Abstract
Background Ultra-high molecular weight polyethylene (UHMWPE) is one of the favored materials for total joint replacement, but its wear particles cause osteolysis. This study aims to elucidate the signaling that mediates the effects of UHMWPE particles on bone cells. Methods RAW264.7 and MC3T3-E1 cells were treated with UHMWPE particles. Chemerin/ChemR23 signaling was manipulated by either overexpressing Rarres2 and Cmklr1 or silencing Cmklr1. The osteoblast and osteoclast differentiation was evaluated by Alizarin red and TRAP staining, respectively. The expression of osteogenic and osteoclastogenic markers was assessed with quantitative real time PCR and western blot. Results UHMWPE particles upregulated the expression of Rarres2 and Cmklr1 in both osteoblast and osteoclast precursor cells. UHMWPE particles induced osteoclast differentiation while inhibited osteoblast differentiation, and this effect was abrogated by silencing Cmklr1 but augmented by the overexpression of Rarres2 and Cmklr1. Similarly, the expression of osteogenic marker genes was inhibited while that of osteoclastogenic marker genes was activated by UHMWPE particles, and this effect was abolished by silencing Cmklr1 and enhanced by Rarres2 and Cmklr1 overexpression. Conclusions These results demonstrated that chemerin/ChemR23 signaling plays a central role in the effects of UHMWPE particles on the balance of osteogenic and osteoclastogenic differentiation, which changes the course of bone remodeling and eventually results in osteolysis.
Collapse
Affiliation(s)
- Fengchao Zhao
- Department of Orthopedic Surgery, the First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Dingwei Cang
- Department of Orthopedic Surgery, the First People's Hospital of Yancheng City, Yancheng, China
| | - Jianzhi Zhang
- Department of Central Laboratory, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Li Zheng
- Department of Orthopedic Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
10
|
Pajarinen J, Lin T, Nabeshima A, Sato T, Gibon E, Jämsen E, Khan TN, Yao Z, Goodman SB. Interleukin-4 repairs wear particle induced osteolysis by modulating macrophage polarization and bone turnover. J Biomed Mater Res A 2021; 109:1512-1520. [PMID: 33340244 PMCID: PMC8213865 DOI: 10.1002/jbm.a.37142] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/16/2020] [Accepted: 11/28/2020] [Indexed: 01/28/2023]
Abstract
Periprosthetic osteolysis remains as a major complication of total joint replacement surgery. Modulation of macrophage polarization with interleukin-4 (IL-4) has emerged as an effective means to limit wear particle-induced osteolysis. The aim of this study was to evaluate the efficacy of local IL-4 delivery in treating preexisting particle-induced osteolysis. To this end, recently established 8 week modification of murine continuous femoral intramedullary particle infusion model was utilized. Subcutaneous infusion pumps were used to deliver polyethylene (PE) particles into mouse distal femur for 4 weeks to induce osteolysis. IL-4 was then added to the particle infusion for another 4 weeks. This delayed IL-4 treatment (IL-4 Del) was compared to IL-4 delivered continuously (IL-4 Cont) with PE particles from the beginning and to the infusion of particles alone for 8 weeks. Both IL-4 treatments were highly effective in preventing and repairing preexisting particle-induced bone loss as assessed by μCT. Immunofluorescence indicated a significant reduction in the number of F4/80 + iNOS + M1 macrophages and increase in the number of F4/80 + CD206 + M2 macrophages with both IL-4 treatments. Reduction in the number of tartrate resistant acid phosphatase + osteoclasts and increase in the amount of alkaline phosphatase (ALP) + osteoblasts was also observed with both IL-4 treatments likely explaining the regeneration of bone in these samples. Interesting, slightly more bone formation and ALP + osteoblasts were seen in the IL-4 Del group than in the IL-4 Cont group although these differences were not statistically significant. The study is a proof of principle that osteolytic lesions can be repaired via modulation of macrophage polarization.
Collapse
Affiliation(s)
- Jukka Pajarinen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
- Department of Musculoskeletal and Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Medicine, Clinicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tzuhua Lin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Akira Nabeshima
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Taishi Sato
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Emmanuel Gibon
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Eemeli Jämsen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
- Department of Medicine, Clinicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tahsin N. Khan
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Zhenyu Yao
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Stuart B. Goodman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
- Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
11
|
Yamada C, Ho A, Akkaoui J, Garcia C, Duarte C, Movila A. Glycyrrhizin mitigates inflammatory bone loss and promotes expression of senescence-protective sirtuins in an aging mouse model of periprosthetic osteolysis. Biomed Pharmacother 2021; 138:111503. [PMID: 33770668 PMCID: PMC8653540 DOI: 10.1016/j.biopha.2021.111503] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 01/02/2023] Open
Abstract
Although periprosthetic osteolysis induced by wear debris particles is significantly elevated in senior (65+ years old) patients, most of the published pre-clinical studies were performed using young (less than three-month old) mice indicating the critical need to employ experimental models of particle-induced osteolysis involving mice with advanced age. Emerging evidence indicates that currently available antiresorptive bone therapies have serious age-dependent side effects. However, a resurgence of healthcare interest has occurred in glycyrrhizin (GLY), a natural extract from the licorice roots, as alternative sources of drugs for treating inflammatory bone lytic diseases and prevention of cellular senescence. This study investigated the effects of GLY on inflammatory bone loss as well as expression patterns of senescence-associated secretory phenotype and senescence-protective markers using an experimental calvarium osteolytic model induced in aged (twenty-four-month-old) mice by polymethylmethacrylate (PMMA) particles. Our results indicate that local treatment with GLY significantly diminished the size of inflammatory osteolytic lesions in aged mice via the number of CXCR4+OCPs and Tartrate-resistant acid phosphatase positive (TRAP+) osteoclasts. Furthermore, GLY dramatically decreased the amounts of senescence-associated secretory phenotype markers, including pro-inflammatory macrophage migration inhibitory factor (MIF) chemokine, and cathepsins B and K in the bone lesions of aged mice. By contrast, GLY significantly elevated expression patterns of senescence-protective markers, including homeostatic stromal derived factor-1 (SDF-1) chemokine, and sirtuin-1, and sirtuin-6, in the PMMA particle-induced calvarial lesions of aged mice. Collectively, these data suggest that GLY can be used for the development of novel therapies to control bone loss and tissue aging in senior patients with periprosthetic osteolysis.
Collapse
Affiliation(s)
- Chiaki Yamada
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, United States
| | - Anny Ho
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, United States
| | - Juliet Akkaoui
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, United States
| | - Christopher Garcia
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, United States
| | - Carolina Duarte
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, United States
| | - Alexandru Movila
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, United States.
| |
Collapse
|
12
|
Utsunomiya T, Zhang N, Lin T, Kohno Y, Ueno M, Maruyama M, Rhee C, Huang E, Yao Z, Goodman SB. Different Effects of Intramedullary Injection of Mesenchymal Stem Cells During the Acute vs. Chronic Inflammatory Phase on Bone Healing in the Murine Continuous Polyethylene Particle Infusion Model. Front Cell Dev Biol 2021; 9:631063. [PMID: 33816480 PMCID: PMC8017138 DOI: 10.3389/fcell.2021.631063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/03/2021] [Indexed: 12/23/2022] Open
Abstract
Chronic inflammation is a common feature in many diseases of different organ systems, including bone. However, there are few interventions to mitigate chronic inflammation and preserve host tissue. Previous in vitro studies demonstrated that preconditioning of mesenchymal stem cells (pMSCs) using lipopolysaccharide and tumor necrosis factor-α polarized macrophages from a pro-inflammatory to an anti-inflammatory phenotype and increased osteogenesis compared to unaltered MSCs. In the current study, we investigated the local injection of MSCs or pMSCs during the acute versus chronic inflammatory phase in a murine model of inflammation of bone: the continuous femoral intramedullary polyethylene particle infusion model. Chronic inflammation due to contaminated polyethylene particles decreased bone mineral density and increased osteoclast-like cells positively stained with leukocyte tartrate resistant acid phosphatase (TRAP) staining, and resulted in a sustained M1 pro-inflammatory macrophage phenotype and a decreased M2 anti-inflammatory phenotype. Local injection of MSCs or pMSCs during the chronic inflammatory phase reversed these findings. Conversely, immediate local injection of pMSCs during the acute inflammatory phase impaired bone healing, probably by mitigating the mandatory acute inflammatory reaction. These results suggest that the timing of interventions to facilitate bone healing by modulating inflammation is critical to the outcome. Interventions to facilitate bone healing by modulating acute inflammation should be prudently applied, as this phase of bone healing is temporally sensitive. Alternatively, local injection of MSCs or pMSCs during the chronic inflammatory phase may be a potential intervention to mitigate the adverse effects of contaminated particles on bone.
Collapse
Affiliation(s)
- Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Yusuke Kohno
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Ejun Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
13
|
Tong X, Ganta RR, Liu Z. AMP-activated protein kinase (AMPK) regulates autophagy, inflammation and immunity and contributes to osteoclast differentiation and functionabs. Biol Cell 2020; 112:251-264. [PMID: 32445585 DOI: 10.1111/boc.202000008] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Osteoclasts are multinucleated giant cells, responsible for bone resorption. Osteoclast differentiation and function requires a series of cytokines to remove the old bone, which coordinates with the induction of bone remodelling by osteoblast-mediated bone formation. Studies have demonstrated that AMP-activated protein kinase (AMPK) play a negative regulatory role in osteoclast differentiation and function. Research involving AMPK, a nutrient and energy sensor, has primarily focused on osteoclast differentiation and function; thus, its role in autophagy, inflammation and immunity remains poorly understood. Autophagy is a conservative homoeostatic mechanism of eukaryotic cells, and response to osteoclast differentiation and function; however, how it interacts with inflammation remains unclear. Additionally, based on the regulatory function of different AMPK subunits for osteoclast differentiation and function, its activation is regulated by upstream factors to perform bone metabolism. This review summarises the critical role of AMPK-mediated autophagy, inflammation and immunity by upstream and downstream signalling during receptor activator of nuclear factor kappa-B ligand-induced osteoclast differentiation and function. This pathway may provide therapeutic targets for bone-related diseases, as well as function as a biomarker for bone homoeostasis.
Collapse
Affiliation(s)
- Xishuai Tong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China.,Center of Excellence for Vector-Borne Diseases, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, 66502, USA.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Roman R Ganta
- Center of Excellence for Vector-Borne Diseases, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, 66502, USA
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, Jiangsu, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
| |
Collapse
|
14
|
Hong H, Xu G, Deng H, Zhou X, Liu W, Cui Z. Concentration-Dependent Regulation of TiAl 6V 4 Particles on the Osteogenesis Potential of Human Bone Marrow Mesenchymal Stem Cells. Biol Trace Elem Res 2020; 195:445-453. [PMID: 31486015 DOI: 10.1007/s12011-019-01885-2] [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: 05/13/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
Total joint replacement is one of the most effective treatments for osteoarthritis, while the aseptic loosening of artificial joint is a major complication leading to the joint replacement failure. There are very limited studies about the effects of titanium-alloy particles on the osteogenic differentiation of mesenchymal stem cells. In this study, human bone marrow-derived mesenchymal stem cells (BM-hMSCs) were treated with different concentrations of TiAl6V4 particles. The cell viability was detected by MTT assay, and the cell proliferation was assessed by CKK-8 assay. The early and late stages of osteogenic differentiation were determined by alkaline phosphatase (ALP) and Alizarin Red S (ARS) staining assays. The expression of osteogenic genes and proteins was analyzed by RT-PCR and Western blot. TiAl6V4 particles at high concentration 100 μg/ml inhibited the cell viability of BM-hMSCs. However, TiAl6V4 in the range of 5-50 μg/ml did not show effects neither on the cell viability nor on the cell proliferation of BM-hMSCs. TiAl6V4 particles showed concentration-dependent bidirectional regulations on BM-hMSC osteogenesis. Specifically, TiAl6V4 at 5 μg/ml promoted the osteogenesis of BM-hMSCs, which was suppressed by TiAl6V4 at 50 μg/ml. Further, mechanism study revealed that the regulation of TiAl6V4 on BM-hMSCs was related to Wnt signaling pathway. Given the potential of mesenchymal cells, our study suggested that the minimization of metal use would be an attractive strategy to reduce the joint replacement failure.
Collapse
Affiliation(s)
- Hongxiang Hong
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China
| | - Guanhua Xu
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China
| | - Hongjian Deng
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China
| | - Xiaogang Zhou
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China
| | - Wei Liu
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China
| | - Zhiming Cui
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
15
|
Lei P, Dai Z, Zhang YS, Liu H, Niu W, Li K, Wang L, Hu Y, Xie J. Macrophage inhibits the osteogenesis of fibroblasts in ultrahigh molecular weight polyethylene (UHMWPE) wear particle-induced osteolysis. J Orthop Surg Res 2019; 14:80. [PMID: 30885228 PMCID: PMC6421644 DOI: 10.1186/s13018-019-1119-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 03/06/2019] [Indexed: 12/16/2022] Open
Abstract
Background In the ultrahigh molecular weight polyethylene (UHMWPE) prosthetic environment, fibroblasts affected by wear particles have the capacity of osteogenesis to reduce osteolysis. We aimed to assess the effects of macrophages on the osteogenic capability of fibroblasts treated with UHMWPE wear particles. Methods The effect of different concentrations of UHMWPE (0, 0.01, 0.1, and 1 mg/ml, respectively) on macrophage proliferation were validated by MTT assay to determine the optimum one. The fibroblasts viability was further determined in the co-culture system of UHMWPE particles and macrophage supernatants. The experiment was designed as seven groups: (A) fibroblasts only; (B) fibroblasts + 1 mg/ml UHMWPE particles; and (C1–C5) fibroblasts + 1/16, 1/8, 1/4, 1/2, and 1/1 supernatants of macrophage cultures stimulated by 1 mg/ml UHMWPE particles vs. fibroblast complete media, respectively. Alizarin red staining was used to detect calcium accumulation. The expression levels of osteogenic proteins were detected by Western blot and ELISA, including alkaline phosphatase (ALP) and osteocalcin (OCN). Results The concentration of 0.1 mg/ml was considered as the optimum concentration for macrophage proliferation due to the survival rate and was highest among the four concentrations. Fibroblast viability was better in the group of fibroblasts + 1/16 ratio of macrophage supernatants stimulated by 1 mg/ml of UHMWPE particles than the other groups (1:8, 1:4, 1:2, 1:1). ALP and OCN expressions were significantly decreased in the group of fibroblasts + 1/4, 1/2, and 1/1 supernatants stimulated by 1 mg/ml of UHMWPE particles compared with other groups (1/8, 1/16) and the group of fibroblasts + 1 mg/ml UHMWPE (p < 0.5). Conclusions Macrophages are potentially involved in the periprosthetic osteolysis by reducing the osteogenic capability of fibroblasts treated with wear particles generated from UHMWPE materials in total hip arthroplasty.
Collapse
Affiliation(s)
- Pengfei Lei
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Zixun Dai
- Department of Orthopedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine of Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Yu Shrike Zhang
- Centre for Biomaterials Innovation, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Hua Liu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310058, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, 310058, People's Republic of China.,Harvard-MIT Division of Health Sciences and Technology, Tissue Engineering Lab, Cambridge, USA
| | - Wanting Niu
- VA Boston Healthcare System, West Roxbury, MA, 02132, USA.,Department of Orthopedics, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Kun Li
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Long Wang
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Yihe Hu
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Jie Xie
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
16
|
Zhang Q, Zhang XF. Hyperoside decreases the apoptosis and autophagy rates of osteoblast MC3T3‑E1 cells by regulating TNF‑like weak inducer of apoptosis and the p38mitogen activated protein kinase pathway. Mol Med Rep 2018; 19:41-50. [PMID: 30387825 PMCID: PMC6297762 DOI: 10.3892/mmr.2018.9622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 04/06/2018] [Indexed: 12/20/2022] Open
Abstract
Wear particles generated between the interface of joints and artificial joint replacements are one of the primary causes of aseptic loosening. The aim of the present study was to investigate the influence of titanium (Ti) particles on the apoptosis and autophagy of osteoblasts, and probe into the potential use of hyperoside (Hy) as a protector for osteoblasts in Ti particle-induced injury. MC3T3-E1 cells were divided into control, Ti, Hy-1+Ti and Hy-2+Ti groups. Cell viability was detected using a Cell Counting Kit-8 assay. Apoptosis and autophagy rates were determined using flow cytometry. Expression levels of apoptosis-associated genes, including caspase-3, apoptosis regulator BAX, apoptosis regulator Bcl-2 and cellular tumor antigen p53, in addition to autophagy-associated genes, including Beclin1 and microtubule-associated protein light chain 3 conversion LC3-II/I, were measured using reverse transcription-quantitative polymerase chain reaction and western blotting. Activation of the tumor necrosis factor ligand superfamily member 12 (TWEAK)-mitogen activated protein kinase 11 (p38) mitogen activated protein kinase (MAPK) pathway was observed by western blotting. The present study demonstrated that pretreatment with Hy was able to increase cell viability and proliferation, and decrease apoptosis and autophagy to protect MC3T3-E1 cells against Ti particle-induced damage. Activation of the TWEAK-p38 pathway contributed to the repair processes of treatment with Hy. The present results suggested that Hy protected osteoblasts against Ti particle-induced damage by regulating the TWEAK-p38 pathway, which suggested the potential of Hy as a protective agent for bones.
Collapse
Affiliation(s)
- Qing Zhang
- Division of Hand and Foot Surgery, Department of Orthopedics, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| | - Xiao-Feng Zhang
- Department of Central Pharmacy, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, P.R. China
| |
Collapse
|
17
|
Wang F, Hu Y, He D, Zhou G, Ellis E. Scaffold-free cartilage cell sheet combined with bone-phase BMSCs-scaffold regenerate osteochondral construct in mini-pig model. Am J Transl Res 2018; 10:2997-3010. [PMID: 30416646 PMCID: PMC6220219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 09/10/2018] [Indexed: 06/09/2023]
Abstract
Tissue-engineered condyles provide a promising approach for end-stage osteoarthritis to reconstruct normal physiological structure and function of the temporomandibular joint (TMJ). However, lack of successful biological condyles in large animals restricts clinical translation. Scaffold-free cartilage cell sheets do not contain any polymeric material which potentially risks local nonspecific inflammatory reactions. In this study, we used cartilage cell sheets covering bone marrow mesenchymal stem cells-Polycaprolactone/Hydroxyapatite (BMSCs-PCL/HA) scaffolds (cell sheet group) transplanted subcutaneously and intramuscularly in mini-pigs. In contrast, autogenous chondrocytes were seeded on polyglycolic acid/ polylactic acid (PGA/PLA) scaffolds for 4 and 12 weeks in-vitro pre-cultivation. Then, they were used as a cartilage-phase composition covering BMSCs-PCL/HA scaffolds, then the entirety (biphase scaffold group) was transplanted subcutaneously into mini-pigs. After 12 weeks, the harvested samples were examined histologically. The cartilage layer was evaluated for thickness, glycosaminoglycan (GAG) quantitation, total collagen quantitation and Young's modulus. The biphase scaffold group failed in regeneration, while the cell sheet group regenerated biological condyle with healthy osteochondral construct. The GAG quantitation, total collagen quantitation and Young's modulus of regenerated cartilage was close to those of the natural condyle. Collectively, cartilage cell sheets combined with bone-phase composition had the potential to regenerate biological condylar.
Collapse
Affiliation(s)
- Feiyu Wang
- Department of Oral Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of StomatologyShanghai 200011, PR China
- Department of Stomatology, Shanghai Tenth People’s Hospital of Tongji UniversityShanghai 200072, PR China
| | - Yihui Hu
- Department of Oral Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of StomatologyShanghai 200011, PR China
| | - Dongmei He
- Department of Oral Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of StomatologyShanghai 200011, PR China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, PR China
| | - Edward Ellis
- Department of Oral and Maxillofacial Surgery, University of Texas Health Science Center at San AntonioTX, USA
| |
Collapse
|
18
|
Philbrick KA, Branscum AJ, Wong CP, Turner RT, Iwaniec UT. Leptin Increases Particle-Induced Osteolysis in Female ob/ob Mice. Sci Rep 2018; 8:14790. [PMID: 30287858 PMCID: PMC6172200 DOI: 10.1038/s41598-018-33173-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 09/17/2018] [Indexed: 11/09/2022] Open
Abstract
Particles generated from wear of prosthesis joint bearing surfaces induce inflammation-mediated periprosthetic bone resorption (osteolysis). Morbidly obese leptin-deficient ob/ob mice are resistant to polyethylene particle-induced bone loss, suggesting that leptin, a hormone produced by adipocytes that circulates in concentrations proportional to total body adiposity, increases osteolysis. To confirm that particles induce less osteolysis in leptin-deficient mice after controlling for cold stress (room temperature)-induced bone loss, ob/ob mice on a C57BL/6 (B6) background and colony B6 wildtype (WT) mice housed at thermoneutral temperature were randomized to control or particle treatment groups (N = 5/group). Polyethylene particles were implanted over calvaria and mice sacrificed 2 weeks later. Compared to particle-treated WT mice, particle-treated ob/ob mice had lower osteolysis score, less infiltration of immune cells, and less woven bone formation. To determine the role of leptin in particle-induced osteolysis, ob/ob mice were randomized into one of 4 groups (n = 6-8/group): (1) control, (2) particles, (3) particles + continuous leptin (osmotic pump, 6 μg/d), or (4) particles + intermittent leptin (daily injection, 40 μg/d). Leptin treatment increased particle-induced osteolysis in ob/ob mice, providing evidence that the adpiokine may play a role in inflammation-driven bone loss. Additional research is required to determine whether altering leptin levels within the physiological range results in corresponding changes in polyethylene-particle-induced osteolysis.
Collapse
Affiliation(s)
- Kenneth A Philbrick
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Adam J Branscum
- Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Carmen P Wong
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Russell T Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA.,Center for Healthy Aging Research, Oregon State University, Corvallis, OR, 97331, USA
| | - Urszula T Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA. .,Center for Healthy Aging Research, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
19
|
Polyethylene particles inserted over calvarium induce cancellous bone loss in femur in female mice. Bone Rep 2018; 9:84-92. [PMID: 30094298 PMCID: PMC6073052 DOI: 10.1016/j.bonr.2018.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/13/2018] [Accepted: 07/02/2018] [Indexed: 12/29/2022] Open
Abstract
Focal bone resorption (osteolysis) induced by wear particles contributes to long-term orthopedic joint failure. However, the impact of focal osteolysis on remote skeletal sites has received less attention. The goal of this study was to determine the effects of polyethylene particles placed over calvaria on representative axial and appendicular skeletal sites in female mice. Because recent work has identified housing temperature as an important biological variable in mice, response to particle treatment was measured in animals housed at room (22 °C) and thermoneutral (32 °C) temperature. Osteolysis was evident in skeletal tissue adjacent to particle insertion. In addition, cancellous bone loss was observed in distal femur metaphysis. The bone loss was associated with lower osteoblast-lined perimeter and lower mineralizing perimeter in distal femur, lower osteocalcin gene expression in tibia, and lower serum osteocalcin, suggesting the response was due, at least in part, to reduced bone formation. Mild cold stress induced by sub-thermoneutral housing resulted in cancellous bone loss in distal femur and lumbar vertebra but did not influence skeletal response to particles. In summary, the results indicate that focal inflammation induced by polyethylene particles has the potential to result in systemic bone loss. This is significant because bone loss is a risk factor for fracture. Wear particles contribute to orthopedic joint failure by promoting focal inflammation- mediated osteolysis. Here we investigated effects of polyethylene particles placed over calvaria on remote skeletal sites in female mice. Particles placed over calvaria resulted in focal inflammation and cancellous bone loss in distal femur metaphysis. Lower osteoblast-lined perimeter suggests reduced bone formation contributed to bone loss in distal femur. Our results suggest focal inflammation induced by polyethylene particles has the potential to result in systemic bone loss.
Collapse
|
20
|
Production of GFP and Luciferase-Expressing Reporter Macrophages for In Vivo Bioluminescence Imaging. Methods Mol Biol 2018. [PMID: 29858786 DOI: 10.1007/978-1-4939-7860-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Macrophages have emerged as crucial regulators of tissue homeostasis, inflammation, and tissue regeneration. In vivo bioluminescence imaging could offer a powerful tool to study many poorly understood aspects of macrophage biology. Thus, we recently developed a straightforward method for the production of large numbers of green fluorescent protein (GFP) and firefly luciferase (fLUC)-expressing reporter macrophages for various in vivo bioluminescence imaging applications. Lentivirus vector containing the GFP/fLUC reporter gene is produced and mouse bone marrow macrophages are isolated following established protocols. Macrophages are then exposed to the lentivirus in the presence of 10 μM cyclosporine for 24 h. After a 24-h recovery period, the transduction is repeated. Three days after the second infection the cells are ready to be used in vivo. Following this cyclosporine-mediated double infection strategy up to 60% of the macrophages express GFP in flow cytometry. The macrophages maintain their ability to polarize to M1 and M2 phenotypes and, when injected to the systemic circulation of a mouse model, reporter cells are both easily detectable with BLI and migrate to a local site of inflammation. These GFP/fLUC-expressing reporter macrophages could prove to be useful tools to study the role of macrophages in health and disease.
Collapse
|
21
|
Yan Z, Tian X, Zhu J, Lu Z, Yu L, Zhang D, Liu Y, Yang C, Zhu Q, Cao X. Metformin suppresses UHMWPE particle-induced osteolysis in the mouse calvaria by promoting polarization of macrophages to an anti-inflammatory phenotype. Mol Med 2018; 24:20. [PMID: 30134793 PMCID: PMC6016863 DOI: 10.1186/s10020-018-0013-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/06/2018] [Indexed: 03/02/2023] Open
Abstract
Background Implant failure remains a major obstacle to successful treatment via TJA. Periprosthetic osteolysis and aseptic loosening are considered as proof of wear debris-induced disruption of local regulatory mechanisms related to excessive bone resorption associated with osteolysis and the damage at the bone-prosthesis interface. Therefore, there is an immediate need to explore strategies for limiting and curing periprosthetic osteolysis and aseptic loosening. Methods We analyzed the in vitro cytokine production by primary mouse bone marrow macrophages (BMMs) that were exposed to ultra-high molecular weight polyethylene (UHMWPE) particles and treated with metformin at different concentrations with or without 5-aminoimidazole-4-carboxamide ribonucleoside to activate or inhibit AMPK. A mouse calvarial model was used to examine the in vivo effects of metformin on UHMWPE particle-induced osteolysis. Results With particles, primary mouse BMMs secreted more pro-inflammatory cytokines tumor necrosis factor-α and interleukin (IL)-6. Treatment with metformin inhibited these variations and promoted the release of cytokine IL-10 with anti-inflammatory capability. In vivo, metformin reduced the production of pro-inflammatory cytokines, osteoclastogenesis, and osteolysis, increasing IL-10 production. Metformin also promoted the polarization of macrophages to an anti-inflammatory phenotype in vivo via AMPK activation. Discussion A crucial point in limiting and correcting the periprosthetic osteolysis and aseptic loosening is the inhibition of inflammatory factor production and osteoclast activation induced by activated macrophages. The ability of metformin to attenuate osteolysis induced in mouse calvaria by the particles was related to a reduction in osteoclast number and polarization of macrophages to an anti-inflammatory functional phenotype. Conclusions Metformin could limit the osteolysis induced by implant debris. Therefore, we hypothesized that metformin could be a potential drug for osteolysis induced by implant debris. Electronic supplementary material The online version of this article (10.1186/s10020-018-0013-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhao Yan
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaoxi Tian
- Emergency department of Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Jinyu Zhu
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zifan Lu
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an, 710032, China
| | - Lifeng Yu
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Dawei Zhang
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yanwu Liu
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chongfei Yang
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Qingsheng Zhu
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xiaorui Cao
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
22
|
Micrometer-Sized Titanium Particles Induce Aseptic Loosening in Rabbit Knee. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5410875. [PMID: 29651439 PMCID: PMC5831897 DOI: 10.1155/2018/5410875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/06/2018] [Accepted: 01/14/2018] [Indexed: 11/26/2022]
Abstract
Wear debris induced aseptic loosening is the leading cause of total knee arthroplasty (TKA) failure. The complex mechanism of aseptic loosening has been a major issue for introducing effective prevention and treatment methods, so a simplified yet efficient rabbit model was established to address this concern with the use of micrometer-sized titanium particles. 20 New Zealand white rabbits were selected and divided into two groups (control = 10, study = 10). A TKA surgery was then performed for each of them, with implantation of a titanium rod prosthesis which was coated evenly with micrometer-sized titanium in the study group and nothing in the control group, into right femoral medullary cavity. After 12 weeks, all the animals were euthanized and X-ray analyses, H&E staining, Goldner Masson trichrome staining, Von Kossa staining, PCR, and Western blotting of some specific mRNAs and proteins in the interface membrane tissues around the prosthesis were carried out. The implantation of a titanium rod prosthesis coated with 20 μm titanium particles into the femoral medullary cavity of rabbits caused continuous titanium particle stimulation around the prosthesis, effectively inducing osteolysis and aseptic loosening. Titanium particle-induced macrophages produce multiple inflammatory factors able to activate osteoclast differentiation through the OPG/RANKL/RANK signaling pathway, resulting in osteolysis while suppressing the function of osteoblasts and reducing bone ingrowth around the prosthesis. This model simulated the implantation and loosening process of an artificial prosthesis, which is an ideal etiological model to study the aseptic prosthetic loosening.
Collapse
|
23
|
Affiliation(s)
- Jukka Pajarinen
- a Department of Orthopaedic Surgery , Stanford University , Stanford , CA , USA
| | - Jiri Gallo
- b Department of Orthopaedics , Faculty of Medicine and Dentistry, Palacky University Olomouc , Czech Republic
| | - Michiaki Takagi
- c Department of Orthopaedic Surgery , Yamagata University Faculty of Medicine , Yamagata City , Yamagata , Japan
| | - Stuart B Goodman
- a Department of Orthopaedic Surgery , Stanford University , Stanford , CA , USA
| | | |
Collapse
|
24
|
Immunomodulation of Biomaterials by Controlling Macrophage Polarization. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:197-206. [PMID: 30471034 DOI: 10.1007/978-981-13-0445-3_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Macrophages are key players in innate immune responses to foreign substances. They participate in the phagocytosis of biomaterial-derived particles, angiogenesis, recruitment of fibroblasts, and formation of granulation tissues. Most macrophage functions are achieved through the release of various cytokines and chemokines; the release profile of cytokines is dependent on the phenotype of macrophages, namely proinflammatory M1 or antiinflammatory M2. M1 and M2 macrophages coexist during an inflammatory phase, and the M1/M2 ratio is considered to be an important factor for wound-healing or tissue regeneration. This ratio depends on the chemical and physical properties of biomaterials. To obtain a favorable foreign body reaction to biomaterials, the phenotypes of the macrophages can be modulated by cytokines, antibodies, small chemicals, and microRNAs. Geometrical surface fabrication of biomaterials can also be used for modulating the phenotype of macrophages.
Collapse
|
25
|
Mahon OR, O'Hanlon S, Cunningham CC, McCarthy GM, Hobbs C, Nicolosi V, Kelly DJ, Dunne A. Orthopaedic implant materials drive M1 macrophage polarization in a spleen tyrosine kinase- and mitogen-activated protein kinase-dependent manner. Acta Biomater 2018; 65:426-435. [PMID: 29104084 DOI: 10.1016/j.actbio.2017.10.041] [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: 06/29/2017] [Revised: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 12/17/2022]
Abstract
Total joint replacements (TJR) are costly procedures required to relieve pain and restore function in patients suffering from end-stage arthritis. Despite great progress in the development and durability of TJRs, the generation of prosthesis-associated wear particles over time leads to an inflammatory cascade which culminates in periprosthetic osteolysis. Studies suggest that wear particles drive the polarization/differentiation of immature macrophages towards a pro-inflammatory M1 phenotype rather than an anti-inflammatory M2 phenotype associated with normal bone and wound healing. This, in turn, contributes to the initiation of peri-implant inflammation. As a result, modulating M1 macrophage cytokine production has been recognised as a viable therapeutic option. The aim of this study was to examine the impact of hydroxyapatite (HA) and poly(methyl methacrylate) (PMMA) particles on human macrophage polarization by comparing their effect on M1/M2-associated gene expression using real-time PCR. Furthermore, using immunoblotting to assess kinase activation, we sought to identify the intracellular signalling molecules activated by PMMA/HA particles and to determine whether pharmacological blockade of these molecules impacts on macrophage phenotype and cytokine production as measured by ELISA. We report that wear particles preferentially polarize macrophages towards an M1 phenotype, an effect that is dependent on activation of the membrane proximal kinase, Syk and members of the mitogen-activated protein kinase (MAPK) family of signalling molecules. Pre-treatment of macrophages with Syk inhibitors (R788/piceatannol) or MAPK inhibitors (SB203580 and PD98059), not only prevents M1 polarization, but also attenuates production of key pro-inflammatory mediators that have been specifically implicated in periprosthetic osteolysis and osteoclast differentiation. STATEMENT OF SIGNIFICANCE It is now well established that wear-debris particles from implanted materials drive deleterious inflammatory responses which can eventually lead to implant loosening. In this study, we provide further insight into the specific cellular pathways activated by wear particles in primary human immune cells. We demonstrate that PMMA bone cement and hydroxyapatite, a commonly used biomaterial, drive the polarization of macrophages towards an inflammatory phenotype and identify the specific signalling molecules that are activated in this process. Pre-treatment of macrophages with pharmacological inhibitors of these molecules in turn prevents macrophage polarization and dampens inflammatory cytokine production. Hence these signalling molecules represent potential therapeutic targets to treat or possibly prevent particulate induced osteolysis.
Collapse
|
26
|
Moran MM, Wilson BM, Ross RD, Virdi AS, Sumner DR. Arthrotomy-based preclinical models of particle-induced osteolysis: A systematic review. J Orthop Res 2017; 35:2595-2605. [PMID: 28548682 PMCID: PMC5702596 DOI: 10.1002/jor.23619] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 05/24/2017] [Indexed: 02/04/2023]
Abstract
We completed a systematic literature review of in vivo animal models that use arthrotomy-based methods to study particle-induced peri-implant osteolysis. The purpose of the review was to characterize the models developed to date, to determine the questions addressed, to assess scientific rigor and transparency, and to identify gaps in knowledge. We probed three literature databases (Medline, Embase, and Scopus) and found 77 manuscripts that fit the search parameters. In the most recent 10 years, researchers mainly used rat and mouse models, whereas in the previous 20 years, large animal, canine, and rabbit models were more common. The studies have demonstrated several pathophysiology pathways, including macrophage migration, particle phagocytosis, increased local production of cytokines and lysosomal enzymes, elevated bone resorption, and suppressed bone formation. The effect of variation in particle characteristics and concentration received limited attention with somewhat mixed findings. Particle contamination by endotoxin was shown to exacerbate peri-implant osteolysis. The possibility of early diagnosis was demonstrated through imaging and biomarker approaches. Several studies showed that both local and systemic delivery of bisphosphonates inhibits the development of particle-induced osteolysis. Other methods of inhibiting osteolysis include the use of anabolic agents and altering the implant design. Few studies examined non-surgical rescue of loosened implants, with conflicting results with alendronate. We found that the manuscripts often lacked the methodological detail now advocated by the ARRIVE guidelines, suggesting that improvement in reporting would be useful to maximize rigor and transparency. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2595-2605, 2017.
Collapse
Affiliation(s)
- Meghan M. Moran
- Department of Anatomy and Cell Biology, Rush University Medical Center
| | | | - Ryan D. Ross
- Department of Anatomy and Cell Biology, Rush University Medical Center
| | - Amarjit S. Virdi
- Department of Anatomy and Cell Biology, Rush University Medical Center
| | | |
Collapse
|
27
|
Pajarinen J, Nabeshima A, Lin TH, Sato T, Gibon E, Jämsen E, Lu L, Nathan K, Yao Z, Goodman SB. * Murine Model of Progressive Orthopedic Wear Particle-Induced Chronic Inflammation and Osteolysis. Tissue Eng Part C Methods 2017; 23:1003-1011. [PMID: 28978284 DOI: 10.1089/ten.tec.2017.0166] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Periprosthetic osteolysis and subsequent aseptic loosening of total joint replacements are driven by byproducts of wear released from the implant. Wear particles cause macrophage-mediated inflammation that culminates with periprosthetic bone loss. Most current animal models of particle-induced osteolysis are based on the acute inflammatory reaction induced by wear debris, which is distinct from the slowly progressive clinical scenario. To address this limitation, we previously developed a murine model of periprosthetic osteolysis that is based on slow continuous delivery of wear particles into the murine distal femur over a period of 4 weeks. The particle delivery was accomplished by using subcutaneously implanted osmotic pumps and tubing, and a hollow titanium rod press-fit into the distal femur. In this study, we report a modification of our prior model in which particle delivery is extended to 8 weeks to better mimic the progressive development of periprosthetic osteolysis and allow the assessment of interventions in a setting where the chronic particle-induced osteolysis is already present at the initiation of the treatment. Compared to 4-week samples, extending the particle delivery to 8 weeks significantly exacerbated the local bone loss observed with μCT and the amount of both peri-implant F4/80+ macrophages and tartrate-resistant acid phosphatase-positive osteoclasts detected with immunohistochemical and histochemical staining. Furthermore, systemic recruitment of reporter macrophages to peri-implant tissues observed with bioluminescence imaging continued even at the later stages of particle-induced inflammation. This modified model system could provide new insights into the mechanisms of chronic inflammatory bone loss and be particularly useful in assessing the efficacy of treatments in a setting that resembles the clinical scenario of developing periprosthetic osteolysis more closely than currently existing model systems.
Collapse
Affiliation(s)
- Jukka Pajarinen
- 1 Department of Orthopaedic Surgery, Stanford University School of Medicine , Redwood City, California
| | - Akira Nabeshima
- 1 Department of Orthopaedic Surgery, Stanford University School of Medicine , Redwood City, California
| | - Tzu-Hua Lin
- 1 Department of Orthopaedic Surgery, Stanford University School of Medicine , Redwood City, California
| | - Taishi Sato
- 1 Department of Orthopaedic Surgery, Stanford University School of Medicine , Redwood City, California
| | - Emmanuel Gibon
- 1 Department of Orthopaedic Surgery, Stanford University School of Medicine , Redwood City, California
| | - Eemeli Jämsen
- 1 Department of Orthopaedic Surgery, Stanford University School of Medicine , Redwood City, California
| | - Laura Lu
- 1 Department of Orthopaedic Surgery, Stanford University School of Medicine , Redwood City, California
| | - Karthik Nathan
- 1 Department of Orthopaedic Surgery, Stanford University School of Medicine , Redwood City, California
| | - Zhenyu Yao
- 1 Department of Orthopaedic Surgery, Stanford University School of Medicine , Redwood City, California
| | - Stuart B Goodman
- 1 Department of Orthopaedic Surgery, Stanford University School of Medicine , Redwood City, California.,2 Department of Bioengineering, Stanford University School of Medicine , Redwood City, California
| |
Collapse
|
28
|
Zhang J, Zheng X, Zhao F, Liu X, Pang Y, Cheng Q, Wang Y, Zhu Y, Zhang Y. UHMWPE wear particles and dendritic cells promote osteoclastogenesis of RAW264.7 cells through RANK-activated NF-κB/MAPK/AKT pathways. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:9400-9408. [PMID: 31966812 PMCID: PMC6965995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/19/2017] [Indexed: 06/10/2023]
Abstract
Dendritic cells (DCs) were shown to enhance UHMWPE particle promoted osteoclastogenesis in RAW264.7 macrophages. This study aimed to elucidate the signaling network mediating the osteoclastogenic effects of UHMWPE particles and DCs. RAW264.7 cells were induced with UHMWPE particles and /or DC2.4 cells. The expression of Rank was silenced by shRNA. The activation of p38MAPK, AKT, and NF-κB was inhibited specific inhibitors. The osteoclasts were assessed by TRAP staining. The expression of osteoclastogenic genes and activation of p38MAPK, AKT, and NF-κB were analyzed by qPCR and/or Western blot. UHMWPE particles and DC2.4 cells cooperatively induced RAW264.7 macrophages to differentiate into osteoclasts, activated p38MAPK, AKT, and NF-κB pathways, and upregulated Nfatc1 and osteoclast markers, which was abolished by silencing Rank or inhibited by inhibitors of p38MAPK, AKT, and NF-κB in RAW264.7 cells. These data demonstrated the complexity of signaling network and cell-cell interactions involved in UHMWPE particles-induced osteolysis.
Collapse
Affiliation(s)
- Jianzhi Zhang
- Central Laboratory, Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Xin Zheng
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Fengchao Zhao
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Xiaoyun Liu
- Central Laboratory, Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Yong Pang
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Qi Cheng
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Yi Wang
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Yi Zhu
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Yan Zhang
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| |
Collapse
|
29
|
Amirhosseini M, Andersson G, Aspenberg P, Fahlgren A. Mechanical instability and titanium particles induce similar transcriptomic changes in a rat model for periprosthetic osteolysis and aseptic loosening. Bone Rep 2017; 7:17-25. [PMID: 28795083 PMCID: PMC5544474 DOI: 10.1016/j.bonr.2017.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 01/30/2023] Open
Abstract
Wear debris particles released from prosthetic bearing surfaces and mechanical instability of implants are two main causes of periprosthetic osteolysis. While particle-induced loosening has been studied extensively, mechanisms through which mechanical factors lead to implant loosening have been less investigated. This study compares the transcriptional profiles associated with osteolysis in a rat model for aseptic loosening, induced by either mechanical instability or titanium particles. Rats were exposed to mechanical instability or titanium particles. After 15 min, 3, 48 or 120 h from start of the stimulation, gene expression changes in periprosthetic bone tissue was determined by microarray analysis. Microarray data were analyzed by PANTHER Gene List Analysis tool and Ingenuity Pathway Analysis (IPA). Both types of osteolytic stimulation led to gene regulation in comparison to unstimulated controls after 3, 48 or 120 h. However, when mechanical instability was compared to titanium particles, no gene showed a statistically significant difference (fold change ≥ ± 1.5 and adjusted p-value ≤ 0.05) at any time point. There was a remarkable similarity in numbers and functional classification of regulated genes. Pathway analysis showed several inflammatory pathways activated by both stimuli, including Acute Phase Response signaling, IL-6 signaling and Oncostatin M signaling. Quantitative PCR confirmed the changes in expression of key genes involved in osteolysis observed by global transcriptomics. Inflammatory mediators including interleukin (IL)-6, IL-1β, chemokine (C-C motif) ligand (CCL)2, prostaglandin-endoperoxide synthase (Ptgs)2 and leukemia inhibitory factor (LIF) showed strong upregulation, as assessed by both microarray and qPCR. By investigating genome-wide expression changes we show that, despite the different nature of mechanical implant instability and titanium particles, osteolysis seems to be induced through similar biological and signaling pathways in this rat model for aseptic loosening. Pathways associated to the innate inflammatory response appear to be a major driver for osteolysis. Our findings implicate early restriction of inflammation to be critical to prevent or mitigate osteolysis and aseptic loosening of orthopedic implants.
Collapse
Affiliation(s)
- Mehdi Amirhosseini
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
- Corresponding author.
| | - Göran Andersson
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Per Aspenberg
- Orthopedics, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Anna Fahlgren
- Division of Cell Biology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
30
|
Wang F, Hu Y, He D, Zhou G, Yang X, Ellis E. Regeneration of subcutaneous tissue-engineered mandibular condyle in nude mice. J Craniomaxillofac Surg 2017; 45:855-861. [DOI: 10.1016/j.jcms.2017.03.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 03/20/2017] [Accepted: 03/28/2017] [Indexed: 10/19/2022] Open
|
31
|
Schipper ON, Haddad SL, Pytel P, Zhou Y. Histological Analysis of Early Osteolysis in Total Ankle Arthroplasty. Foot Ankle Int 2017; 38:351-359. [PMID: 28367690 DOI: 10.1177/1071100716682333] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND The purpose of this study was to perform a histological comparative analysis of tibiotalar joint samples taken from areas of osteolysis adjacent to total ankle arthroplasties vs control synovial specimens to determine the reaction to and presence of polyethylene (PE) particles. METHODS A total of 57 pathology samples were identified in the osteolysis group, while 11 were identified in the control group. For each sample, hematoxylin and eosin, Oil Red O (ORO), and macrophage marker CD163-stained slides were created. Polarized light and ORO stain were used to identify PE particles. The presence of metal particles and giant cell reaction to PE particles were also scored. RESULTS Macrophages, PE particles, metallosis, and foreign body giant cell reaction scores were significantly higher in the osteolysis group compared with the control group. In the osteolysis group, ORO staining was positive in 93% (53/57), birefringent material was present in 96.5% (55/57), and macrophage infiltrates were present in 96.5% (55/57). Foreign body giant cell reaction with giant cells surrounding PE particles was present in 49.1% (28/57) of osteolytic specimens. The presence of foreign body giant cell reaction was associated with significantly higher macrophage, ORO, and polarizable material scores. The average time to surgery for osteolysis from the index ankle replacement was 6.0 (range, 0-15) years for the 57 patients in the osteolysis group. CONCLUSION This study is the largest ankle arthroplasty histological analysis to show that areas of osteolysis consist of abundant polyethylene wear particles, present both intracellularly and extracellularly. Furthermore, these areas were associated with a CD163+ macrophage infiltrate and frequently a foreign body reaction with giant cells engulfing PE particles. It is likely that implant wear particles play a significant role in osteolysis based on the histopathology. LEVEL OF EVIDENCE Level III, retrospective comparative series.
Collapse
Affiliation(s)
| | | | - Peter Pytel
- 3 Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Ying Zhou
- 4 Center for Clinical and Research Informatics, NorthShore University HealthSystem Research Institute, Evanston IL, USA
| |
Collapse
|
32
|
Utomo L, Boersema GSA, Bayon Y, Lange JF, van Osch GJVM, Bastiaansen-Jenniskens YM. In vitro modulation of the behavior of adhering macrophages by medications is biomaterial-dependent. ACTA ACUST UNITED AC 2017; 12:025006. [PMID: 28267684 DOI: 10.1088/1748-605x/aa5cbc] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
After implantation of a biomaterial, an inflammatory response involving macrophages is induced. The behavior of macrophages depends on their phenotype, and by directing macrophage polarization unwanted effects may be avoided. In this study, the possibility to modulate the behavior of macrophages activated by biomaterials was assessed in an in vitro model. Primary human monocytes were seeded on polyethylene terephthalate, polypropylene and polylactic acid yarns, and treated with medications frequently used by patients: rapamycin, dexamethasone, celecoxib or pravastatin. Modulation of the adhering macrophages with rapamycin resulted in a generally pro-inflammatory effect. Dexamethasone caused an overall anti-inflammatory effect on the macrophages cultured on either material, while celecoxib only affected macrophages adhering to polyethylene terephthalate and polylactic acid. Pravastatin increased the pro-inflammatory genes of macrophages cultured on polypropylene and polylactic acid. Pairwise comparison revealed that macrophages adhering to polylactic acid seemed to be more susceptible to phenotype modulation than when adhering to polypropylene or polyethylene terephthalate. The data show that macrophages activated by the biomaterials can be modulated, yet the degree of the modulatory capacity depends on the type of material. Combined, this model provides insights into the possibility of using a medication in combination with a biomaterial to direct macrophage behavior and thereby possibly avoid unwanted effects after implantation.
Collapse
Affiliation(s)
- Lizette Utomo
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | | | | | | | | | | |
Collapse
|
33
|
Hallab NJ, Jacobs JJ. Chemokines Associated with Pathologic Responses to Orthopedic Implant Debris. Front Endocrinol (Lausanne) 2017; 8:5. [PMID: 28154552 PMCID: PMC5243846 DOI: 10.3389/fendo.2017.00005] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 01/06/2017] [Indexed: 01/19/2023] Open
Abstract
Despite the success in returning people to health saving mobility and high quality of life, the over 1 million total joint replacements implanted in the US each year are expected to eventually fail after approximately 15-25 years of use, due to slow progressive subtle inflammation to implant debris compromising the bone implant interface. This local inflammatory pseudo disease state is primarily caused by implant debris interaction with innate immune cells, i.e., macrophages. This implant debris can also activate an adaptive immune reaction giving rise to the concept of implant-related metal sensitivity. However, a consensus of studies agree the dominant form of this response is due to innate reactivity by macrophages to implant debris danger signaling (danger-associated molecular pattern) eliciting cytokine-based and chemokine inflammatory responses. This review covers implant debris-induced release of the cytokines and chemokines due to activation of the innate (and the adaptive) immune system and how this leads to subsequent implant failure through loosening and osteolysis, i.e., what is known of central chemokines (e.g., IL-8, monocyte chemotactic protein-1, MIP-1, CCL9, CCL10, CCL17, and CCL22) associated with implant debris reactivity as related to the innate immune system activation/cytokine expression, e.g., danger signaling (e.g., IL-1β, IL-18, IL-33, etc.), toll-like receptor activation (e.g., IL-6, tumor necrosis factor α, etc.), bone catabolism (e.g., TRAP5b), and hypoxia responses (HIF-1α). More study is needed, however, to fully understand these interactions to effectively counter cytokine- and chemokine-based orthopedic implant-related inflammation.
Collapse
Affiliation(s)
- Nadim J. Hallab
- Department of Orthopedics, Rush University Medical Center, Chicago, IL, USA
| | - Joshua J. Jacobs
- Department of Orthopedics, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
34
|
Qin CQ, Huang DS, Zhang C, Song B, Huang JB, Ding Y. Lentivirus-mediated short hairpin RNA interference targeting TNF-alpha in macrophages inhibits particle-induced inflammation and osteolysis in vitro and in vivo. BMC Musculoskelet Disord 2016; 17:431. [PMID: 27756280 PMCID: PMC5069821 DOI: 10.1186/s12891-016-1290-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 10/11/2016] [Indexed: 11/23/2022] Open
Abstract
Background Aseptic loosening is a significant impediment to joint implant longevity. Prosthetic wear particles are postulated to play a central role in the onset and progression of periprosthetic osteolysis, leading to aseptic loosening of the prosthesis. Methods We investigated the inhibitory effects of a lentivirus-mediated short hairpin RNA that targets the TNF-alpha gene on the particle-induced inflammatory and osteolytic changes via macrophages both in vitro and in vivo. An siRNA sequence targeting the mouse TNF-alpha gene from four candidates, transcribed in vitro, was screened and identified. A lentivirus vector expressing short hairpin RNA (shRNA) was then constructed in order to facilitate efficient expression of TNF-alpha-siRNA. Lentivirus-mediated shRNA was transduced into cells of the mouse macrophage line RAW 264.7. Ceramic and titanium particles were introduced 24 h after lentivirus transduction to stimulate cells. TNF-alpha expression, represented by both mRNA and protein levels, was quantified with real-time PCR and ELISA at all time intervals. Lentivirus-mediated shRNA suspension was locally administered into the murine calvarial model, followed by local injection of particles. A multi-slice spiral CT scan was used to evaluate the osteolysis of the calvaria by detecting the width of the cranial sutures. Results Macrophages developed pseudopods when co-cultured with particles. Lentivirus-mediated shRNA was shown to effectively inhibit the expression of TNF-alpha at both the mRNA and protein levels in RAW 264.7. The multi-slice spiral CT scan showed that the lentivirus-mediated shRNA significantly suppressed osteolysis of mouse calvaria. Conclusions Our investigation highlighted the results that lentivirus-mediated shRNA targeting the TNF-alpha gene successfully inhibited particle-induced inflammatory and osteolytic changes both in vitro and in vivo. Therefore, lentivirus-mediated gene therapy may provide a novel therapeutic approach to aseptic joint loosening.
Collapse
Affiliation(s)
- Chu-Qiang Qin
- Department of Orthopaedic Surgery, The Memorial Hospital of Sun Yat-Sen University, 107 Yanjiangxi Road, 510120, Guangzhou, China
| | - Dong-Sheng Huang
- Department of Orthopaedic Surgery, The Memorial Hospital of Sun Yat-Sen University, 107 Yanjiangxi Road, 510120, Guangzhou, China
| | - Chi Zhang
- Department of Orthopaedic Surgery, The Memorial Hospital of Sun Yat-Sen University, 107 Yanjiangxi Road, 510120, Guangzhou, China
| | - Bin Song
- Department of Orthopaedic Surgery, The Memorial Hospital of Sun Yat-Sen University, 107 Yanjiangxi Road, 510120, Guangzhou, China
| | - Jian-Bin Huang
- Department of Orthopaedic Surgery, The Memorial Hospital of Sun Yat-Sen University, 107 Yanjiangxi Road, 510120, Guangzhou, China
| | - Yue Ding
- Department of Orthopaedic Surgery, The Memorial Hospital of Sun Yat-Sen University, 107 Yanjiangxi Road, 510120, Guangzhou, China.
| |
Collapse
|
35
|
She C, Shi GL, Xu W, Zhou XZ, Li J, Tian Y, Li J, Li WH, Dong QR, Ren PG. Effect of low-dose X-ray irradiation and Ti particles on the osseointegration of prosthetic. J Orthop Res 2016; 34:1688-1696. [PMID: 26826053 DOI: 10.1002/jor.23179] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/28/2016] [Indexed: 02/04/2023]
Abstract
Low-dose irradiation (LDI) exhibits a positive effect on osteoblasts and inhibitory effect of inflammation. Here, we test the hypothesis that LDI can promote osseointegration and inhibit the inflammatory membrane formation in the presence of titanium (Ti) particles. Endotoxin-free titanium particles were injected into rabbit, prior to the insertion of a Ti6-Al-4-V sticks pre-coated with hydroxyapatite. Two days after operation, both distal femurs of the animal were exposed to 0.5 Gy X-ray irradiation. All ani-mals were euthanized 8 weeks after the operation. The PINP concentration was determined at day 0, 2, 4, and 8 weeks after operation. Trabecular morphology around the implants 8 weeks after operation was assessed using micro-CT, then the maximum push out force of simples was assessed using biomechanics test. Five samples in each group were chosen for bone histomorphology study without decalcification 8 weeks after operation. The results confirmed that the LDI can significantly improve ingrowth of bone into the prosthetic interface and stability of the prosthesis when there was no wear particles. Although promotion effects for bone formation induced by LDI can be counteracted by wear particles, LDI can significantly inhibit the interface membrane formation around the implant induced by wear particles. Based on these results, we conclude that LDI may be useful for enhancing the stability of prosthesis when there are no wear particles and for inhibiting the interface membrane formation during the early stage of aseptic loosening in the presence of wear particles. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1688-1696, 2016.
Collapse
Affiliation(s)
- Chang She
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Jiangsu, Suzhou, China
| | - Gao-Long Shi
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Jiangsu, Suzhou, China
| | - Wei Xu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Jiangsu, Suzhou, China
| | - Xiao-Zhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Jiangsu, Suzhou, China
| | - Jian Li
- Department of Translational Medicine R&D Center, Shenzhen Institute of Advanced Technology, CAS, Guangdong, Shenzhen, China
| | - Ye Tian
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soo-chow University, Jiangsu, Suzhou, China
| | - Jian Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Jiangsu, Suzhou, China
| | - Wei-Hao Li
- Department of Radiotherapy and Oncology, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Qi-Rong Dong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Jiangsu, Suzhou, China.
| | - Pei-Gen Ren
- Department of Translational Medicine R&D Center, Shenzhen Institute of Advanced Technology, CAS, Guangdong, Shenzhen, China.
| |
Collapse
|
36
|
NF-κB decoy oligodeoxynucleotide mitigates wear particle-associated bone loss in the murine continuous infusion model. Acta Biomater 2016; 41:273-81. [PMID: 27260104 DOI: 10.1016/j.actbio.2016.05.038] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 05/25/2016] [Accepted: 05/30/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Total joint replacement is a cost-effective surgical procedure for patients with end-stage arthritis. Wear particle-induced chronic inflammation is associated with the development of periprosthetic osteolysis. Modulation of NF-κB signaling in macrophages, osteoclasts, and mesenchymal stem cells could potentially mitigate this disease. In the current study, we examined the effects of local delivery of decoy NF-κB oligo-deoxynucleotide (ODN) on wear particle-induced bone loss in a murine continuous femoral particle infusion model. Ultra-high molecular weight polyethylene particles (UHMWPE) with or without lipopolysaccharide (LPS) were infused via osmotic pumps into hollow titanium rods placed in the distal femur of mice for 4weeks. Particle-induced bone loss was evaluated by μCT, and immunohistochemical analysis of sections from the femur. Particle infusion alone resulted in reduced bone mineral density and trabecular bone volume fraction in the distal femur. The decoy ODN reversed the particle-associated bone volume fraction loss around the implant, irrespective of the presence of LPS. Particle-infusion with LPS increased bone mineral density in the distal femur compared with particle-infusion alone. NF-κB decoy ODN reversed or further increased the bone mineral density in the femur (3-6mm from the distal end) exposed to particles alone or particles plus LPS. NF-κB decoy ODN also inhibited macrophage infiltration and osteoclast number, but had no significant effects on osteoblast numbers in femurs exposed to wear particles and LPS. Our study suggests that targeting NF-κB activity via local delivery of decoy ODN has great potential to mitigate wear particle-induced osteolysis. STATEMENT OF SIGNIFICANCE Total joint replacement is a cost-effective surgical procedure for patients with end-stage arthritis. Chronic inflammation is crucial for the development of wear particle-associated bone loss. Modulation of NF-κB signaling in macrophages (pro-inflammatory cells), osteoclasts (bone-resorbing cells), and osteoblasts (bone-forming cells) could potentially mitigate this disease. Here we demonstrated that local delivery of decoy NF-κB oligo-deoxynucleotide (ODN) mitigated ultra-high molecular weight polyethylene (UHMWPE) wear particle induced bone loss in a clinically relevant murine model. The protective effects of decoy ODN was associated with reduced macrophage infiltration and osteoclast activation, but had no significant effects on osteoblast numbers. Our study suggests that targeting NF-κB activity via local delivery of decoy ODN has great potential to mitigate wear particle-induced bone loss.
Collapse
|
37
|
A review of UHMWPE wear-induced osteolysis: the role for early detection of the immune response. Bone Res 2016; 4:16014. [PMID: 27468360 PMCID: PMC4941197 DOI: 10.1038/boneres.2016.14] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 05/08/2016] [Accepted: 05/13/2016] [Indexed: 12/15/2022] Open
Abstract
In a world where increasing joint arthroplasties are being performed on increasingly younger patients, osteolysis as the leading cause of failure after total joint arthroplasty (TJA) has gained considerable attention. Ultra-high molecular weight polyethylene wear-induced osteolysis is the process by which prosthetic debris mechanically released from the surface of prosthetic joints induces an immune response that favors bone catabolism, resulting in loosening of prostheses with eventual failure or fracture. The immune response initiated is innate in that it is nonspecific and self-propagating, with monocytic cells and osteoclasts being the main effectors. To date, detecting disease early enough to implement effective intervention without unwanted systemic side effects has been a major barrier. These barriers can be overcome using newer in vivo imaging techniques and modules linked with fluorescence and/or chemotherapies. We discuss the pathogenesis of osteolysis, and provide discussion of the challenges with imaging and therapeutics. We describe a positron emission tomography imaging cinnamoyl-Phe-(D)-Leu-Phe-(D)-Leu-Phe-Lys module, specific to macrophages, which holds promise in early detection of disease and localization of treatment. Further research and increased collaboration among therapeutic and three-dimensional imaging researchers are essential in realizing a solution to clinical osteolysis in TJA.
Collapse
|
38
|
Brulefert K, Córdova LA, Brulin B, Faucon A, Hulin P, Nedellec S, Gouin F, Passuti N, Ishow E, Heymann D. Pro-osteoclastic in vitro effect of Polyethylene-like nanoparticles: Involvement in the pathogenesis of implant aseptic loosening. J Biomed Mater Res A 2016; 104:2649-57. [PMID: 27254768 DOI: 10.1002/jbm.a.35803] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/07/2016] [Accepted: 03/16/2016] [Indexed: 12/21/2022]
Abstract
Polyethylene micro-sized wear particles released from orthopedic implants promote inflammation and osteolysis; however, less is known about the bioactivity of polyethylene nanosized wear particles released from the last generation of polymer-bearing surfaces. We aim to assess the internalization of fluorescent polyethylene-like nanoparticles by both human macrophages and osteoclasts and also, to determine their effects in osteoclastogenesis in vitro. Human macrophages and osteoclasts were incubated with several ratios of fluorescent polyethylene-like nanoparticles between 1 and 72 h, and 4 h, 2, 4, 6, and 9 days, respectively. The internalization of nanoparticles was quantified by flow cytometry and followed by both confocal and video time-lapse microscopy. Osteoclast differentiation and activity was semiquantified by tartrate-resistant acid phosphatase (TRAP) staining, TRAP mRNA relative expression, and pit resorption assay, respectively. Macrophages, osteoclast precursors and mature osteoclasts internalized nanoparticles in a dose- and time-dependent manner and maintained their resorptive activity. In addition, nanoparticles significantly increased the osteoclastogenesis as shown by upregulation of the TRAP expressing cell number. We conclude that polyethylene-like nanosized wear particles promote osteoclast differentiation without alteration of bone resorptive activity of mature osteoclasts and they could be considered as important actors in periprosthetic osteolysis of the last new generation of polymer-bearing surfaces. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2649-2657, 2016.
Collapse
Affiliation(s)
- Kevin Brulefert
- Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, INSERM, UMR 957, Nantes, 44035, France.,University of Nantes, Nantes Atlantique Universities, Nantes, France.,Nantes University Hospital, Nantes, France
| | - Luis A Córdova
- Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, INSERM, UMR 957, Nantes, 44035, France. .,University of Nantes, Nantes Atlantique Universities, Nantes, France. .,Department of Oral and Maxillofacial Surgery-Faculty of Dentistry, University of Chile-Conicyt, Santiago, Chile.
| | - Bénédicte Brulin
- Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, INSERM, UMR 957, Nantes, 44035, France.,University of Nantes, Nantes Atlantique Universities, Nantes, France
| | - Adrien Faucon
- CEISAM-UMR CNRS 6230, University of Nantes, Nantes, France
| | - Philipe Hulin
- Nantes University Hospital, Nantes, France.,MicroPICell Platform, SFR Santé François Bonamy, INSERM, UMS 016-UMS CNRS 3556, Nantes, France
| | - Steven Nedellec
- Nantes University Hospital, Nantes, France.,MicroPICell Platform, SFR Santé François Bonamy, INSERM, UMS 016-UMS CNRS 3556, Nantes, France
| | - François Gouin
- Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, INSERM, UMR 957, Nantes, 44035, France.,University of Nantes, Nantes Atlantique Universities, Nantes, France.,Nantes University Hospital, Nantes, France
| | - Norbert Passuti
- Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, INSERM, UMR 957, Nantes, 44035, France.,University of Nantes, Nantes Atlantique Universities, Nantes, France.,Nantes University Hospital, Nantes, France
| | - Eléna Ishow
- CEISAM-UMR CNRS 6230, University of Nantes, Nantes, France
| | - Dominique Heymann
- Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, INSERM, UMR 957, Nantes, 44035, France.,University of Nantes, Nantes Atlantique Universities, Nantes, France.,Nantes University Hospital, Nantes, France.,Department of Oncology and Metabolism, University of Sheffield, The Medical School, Sheffield, England, United Kingdom
| |
Collapse
|
39
|
Sato T, Pajarinen J, Behn A, Jiang X, Lin TH, Loi F, Yao Z, Egashira K, Yang F, Goodman SB. The effect of local IL-4 delivery or CCL2 blockade on implant fixation and bone structural properties in a mouse model of wear particle induced osteolysis. J Biomed Mater Res A 2016; 104:2255-62. [PMID: 27114284 DOI: 10.1002/jbm.a.35759] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/25/2016] [Accepted: 04/21/2016] [Indexed: 12/21/2022]
Abstract
Modulation of macrophage polarization and prevention of CCL2-induced macrophage chemotaxis are emerging strategies to reduce wear particle induced osteolysis and aseptic total joint replacement loosening. In this study, the effect of continuous IL-4 delivery or bioactive implant coating that constitutively releases a protein inhibitor of CCL2 signaling (7ND) on particle induced osteolysis were studied in the murine continuous femoral intramedullary particle infusion model. Polyethylene particles with or without IL-4 were infused into mouse distal femurs implanted with hollow titanium rods using subcutaneous infusion pumps. In another experimental group, particles were infused into the femur through a 7ND coated rod. After 4 weeks, fixation of the implant was assessed using a pullout test. The volume of trabecular bone and the geometry of the local cortical bone were assessed by µCT and the corresponding structural properties of the cortical bone determined by torsional testing. Continuous IL-4 delivery led to increased trabecular bone volume as well as enhanced local bone geometry and structural properties, while 7ND implant coating did not have effect on these parameters. The results suggest that local IL-4 treatment is a promising strategy to mitigate wear particle induced osteolysis. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2255-2262, 2016.
Collapse
Affiliation(s)
- Taishi Sato
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Anthony Behn
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Xinyi Jiang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Tzu-Hua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Florence Loi
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Kensuke Egashira
- Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California.,Department of Bioengineering, Stanford University, Stanford, California
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California.,Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
40
|
Gibon E, Córdova LA, Lu L, Lin TH, Yao Z, Hamadouche M, Goodman SB. The biological response to orthopedic implants for joint replacement. II: Polyethylene, ceramics, PMMA, and the foreign body reaction. J Biomed Mater Res B Appl Biomater 2016; 105:1685-1691. [PMID: 27080740 DOI: 10.1002/jbm.b.33676] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/02/2016] [Accepted: 03/20/2016] [Indexed: 11/10/2022]
Abstract
Novel evidence-based prosthetic designs and biomaterials facilitate the performance of highly successful joint replacement (JR) procedures. To achieve this goal, constructs must be durable, biomechanically sound, and avoid adverse local tissue reactions. Different biomaterials such as metals and their alloys, polymers, ceramics, and composites are currently used for JR implants. This review focuses on (1) the biological response to the different biomaterials used for TJR and (2) the chronic inflammatory and foreign-body response induced by byproducts of these biomaterials. A homeostatic state of bone and surrounding soft tissue with current biomaterials for JR can be achieved with mechanically stable, infection free and intact (as opposed to the release of particulate or ionic byproducts) implants. Adverse local tissue reactions (an acute/chronic inflammatory reaction, periprosthetic osteolysis, loosening and subsequent mechanical failure) may evolve when the latter conditions are not met. This article (Part 2 of 2) summarizes the biological response to the non-metallic materials commonly used for joint replacement including polyethylene, ceramics, and polymethylmethacrylate (PMMA), as well as the foreign body reaction to byproducts of these materials. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1685-1691, 2017.
Collapse
Affiliation(s)
- Emmanuel Gibon
- Department of Orthopedic Surgery, Stanford University, Stanford, California.,Laboratoire de Biomécanique et Biomatériaux Ostéo-Articulaires-UMR CNRS 7052, Faculté de Médecine-Université Paris 7, Paris, France.,Department of Orthopedic Surgery, Hopital Cochin, APHP, Paris, France
| | - Luis A Córdova
- Department of Orthopedic Surgery, Stanford University, Stanford, California.,Department of Oral and Maxillofacial Surgery, University of Chile-Conicyt, Santiago, Chile
| | - Laura Lu
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Tzu-Hua Lin
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Zhenyu Yao
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Moussa Hamadouche
- Laboratoire de Biomécanique et Biomatériaux Ostéo-Articulaires-UMR CNRS 7052, Faculté de Médecine-Université Paris 7, Paris, France.,Department of Orthopedic Surgery, Hopital Cochin, APHP, Paris, France
| | - Stuart B Goodman
- Department of Orthopedic Surgery, Stanford University, Stanford, California.,Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
41
|
Vasconcelos DM, Santos SG, Lamghari M, Barbosa MA. The two faces of metal ions: From implants rejection to tissue repair/regeneration. Biomaterials 2016; 84:262-275. [DOI: 10.1016/j.biomaterials.2016.01.046] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/20/2022]
|
42
|
Pajarinen J, Lin TH, Sato T, Loi F, Yao Z, Konttinen YT, Goodman SB. Establishment of Green Fluorescent Protein and Firefly Luciferase Expressing Mouse Primary Macrophages for In Vivo Bioluminescence Imaging. PLoS One 2015; 10:e0142736. [PMID: 26555613 PMCID: PMC4640705 DOI: 10.1371/journal.pone.0142736] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 10/26/2015] [Indexed: 12/23/2022] Open
Abstract
Macrophages play a key role in tissue homeostasis as well as in a range of pathological conditions including atherosclerosis, cancer, and autoimmunity. Many aspects of their in vivo behavior are, however, poorly understood. Bioluminescence imaging (BLI) with green fluorescent protein (GFP) and firefly luciferase (FLUC) labelled autologous reporter macrophages could potentially offer a powerful tool to study macrophage biology, but this approach has been hindered by the relative difficulty of efficient gene transfer into primary macrophages. Here we describe a straightforward method for producing large numbers of GFP/FLUC expressing mouse primary macrophages utilizing lentivirus vector, cyclosporine, and a double infection strategy. Using this method we achieved up to 60% of macrophages to express GFP with correspondingly high FLUC signal. When injected into the circulation using a mouse model of local biomaterial induced inflammation and osteolysis, macrophages were initially detectable within the lungs, followed by systemic homing to the local area of chronic inflammation in the distal femur. In addition, transduced macrophages maintained their ability to assume M1 and M2 phenotypes although the GFP/FLUC expression was altered by the polarizing signals. These reporter macrophages could prove to be valuable tools to study the role of macrophages in health and disease.
Collapse
Affiliation(s)
- Jukka Pajarinen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Tzu-hua Lin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Taishi Sato
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Florence Loi
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Zhenyu Yao
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Yrjö T. Konttinen
- Department of Medicine, Institute of Clinical Medicine, University of Helsinki, Helsinki, Finland
| | - Stuart B. Goodman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
- * E-mail:
| |
Collapse
|
43
|
Gallo J, Raska M, Konttinen YT, Nich C, Goodman SB. Innate immunity sensors participating in pathophysiology of joint diseases: a brief overview. J Long Term Eff Med Implants 2015; 24:297-317. [PMID: 25747032 DOI: 10.1615/jlongtermeffmedimplants.2014010825] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The innate immune system consists of functionally specialized "modules" that are activated in response to a particular set of stimuli via sensors located on the surface or inside the tissue cells. These cells screen tissues for a wide range of exogenous and endogenous danger/damage-induced signals with the aim to reject or tolerate them and maintain tissue integrity. In this line of thinking, inflammation evolved as an adaptive tool for restoring tissue homeostasis. A number of diseases are mediated by a maladaptation of the innate immune response, perpetuating chronic inflammation and tissue damage. Here, we review recent evidence on the cross talk between innate immune sensors and development of rheumatoid arthritis, osteoarthritis, and aseptic loosening of total joint replacements. In relation to the latter topic, there is a growing body of evidence that aseptic loosening and periprosthetic osteolysis results from long-term maladaptation of periprosthetic tissues to the presence of by-products continuously released from an artificial joint.
Collapse
Affiliation(s)
- Jiri Gallo
- Department of Orthopedics, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc 775 20, Czech Republic
| | - Milan Raska
- Department of Immunology, Faculty of Medicine & Dentistry, Palacky University, Hnevotinska 3, 775 15 Olomouc, Czech Republic
| | - Yrjo T Konttinen
- Department of Clinical Medicine, University of Helsinki and ORTON Orthopaedic Hospital of the Invalid Foundation, Helsinki, Finland
| | - Christophe Nich
- Laboratoire de Biomecanique et Biomateriaux Osteo-Articulaires - UMR CNRS 7052, Faculte de Medecine - Universite Paris 7, Paris, France; Department of Orthopaedic Surgery, European Teaching Hospital, Assistance Publique - Hopitaux de Paris
| | - Stuart B Goodman
- Department of Orthopaedic Surgery Stanford University Medical Center Redwood City, CA
| |
Collapse
|
44
|
Cang D, Guo K, Zhao F. Dendritic cells enhance UHMWPE wear particle-induced osteoclast differentiation of macrophages. J Biomed Mater Res A 2015; 103:3349-54. [PMID: 25808788 DOI: 10.1002/jbm.a.35459] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 03/03/2015] [Accepted: 03/13/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Dingwei Cang
- Xuzhou Medical College; Xuzhou Jiangsu 221004 People's Republic of China
| | - Kaijin Guo
- The Affiliate Hospital of Xuzhou Medical College; Xuzhou Jiangsu 221004 China
| | - Fengchao Zhao
- The Affiliate Hospital of Xuzhou Medical College; Xuzhou Jiangsu 221004 China
| |
Collapse
|
45
|
The pathology of orthopedic implant failure is mediated by innate immune system cytokines. Mediators Inflamm 2014; 2014:185150. [PMID: 24891761 PMCID: PMC4033543 DOI: 10.1155/2014/185150] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 03/25/2014] [Accepted: 04/08/2014] [Indexed: 12/22/2022] Open
Abstract
All of the over 1 million total joint replacements implanted in the US each year are expected to eventually fail after 15–25 years of use, due to slow progressive subtle inflammation at the bone implant interface. This inflammatory disease state is caused by implant debris acting, primarily, on innate immune cells, that is, macrophages. This slow progressive pathological bone loss or “aseptic loosening” is a potentially life-threatening condition due to the serious complications in older people (>75 yrs) of total joint replacement revision surgery. In some people implant debris (particles and ions from metals) can influence the adaptive immune system as well, giving rise to the concept of metal sensitivity. However, a consensus of studies agrees that the dominant form of this response is due to innate reactivity by macrophages to implant debris where both danger (DAMP) and pathogen (PAMP) signalling elicit cytokine-based inflammatory responses. This paper discusses implant debris induced release of the cytokines and chemokines due to activation of the innate (and the adaptive) immune system and the subsequent formation of osteolysis. Different mechanisms of implant-debris reactivity related to the innate immune system are detailed, for example, danger signalling (e.g., IL-1β, IL-18, IL-33, etc.), toll-like receptor activation (e.g., IL-6, TNF-α, etc.), apoptosis (e.g., caspases 3–9), bone catabolism (e.g., TRAP5b), and hypoxia responses (Hif1-α). Cytokine-based clinical and basic science studies are in progress to provide diagnosis and therapeutic intervention strategies.
Collapse
|
46
|
Orthopaedic implant failure: aseptic implant loosening–the contribution and future challenges of mouse models in translational research. Clin Sci (Lond) 2014; 127:277-93. [DOI: 10.1042/cs20130338] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aseptic loosening as a result of wear debris is considered to be the main cause of long-term implant failure in orthopaedic surgery and improved biomaterials for bearing surfaces decreases significantly the release of micrometric wear particles. Increasingly, in-depth knowledge of osteoimmunology highlights the role of nanoparticles and ions released from some of these new bearing couples, opening up a new era in the comprehension of aseptic loosening. Mouse models have been essential in the progress made in the early comprehension of pathophysiology and in testing new therapeutic agents for particle-induced osteolysis. However, despite this encouraging progress, there is still no valid clinical alternative to revision surgery. The present review provides an update of the most commonly used bearing couples, the current concepts regarding particle–cell interactions and the approaches used to study the biology of periprosthetic osteolysis. It also discusses the contribution and future challenges of mouse models for successful translation of the preclinical progress into clinical applications.
Collapse
|
47
|
Periprosthetic wear particle migration and distribution modelling and the implication for osteolysis in cementless total hip replacement. J Mech Behav Biomed Mater 2014; 32:225-244. [DOI: 10.1016/j.jmbbm.2014.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 01/03/2014] [Accepted: 01/08/2014] [Indexed: 11/19/2022]
|
48
|
Goodman SB, Gibon E, Pajarinen J, Lin TH, Keeney M, Ren PG, Nich C, Yao Z, Egashira K, Yang F, Konttinen YT. Novel biological strategies for treatment of wear particle-induced periprosthetic osteolysis of orthopaedic implants for joint replacement. J R Soc Interface 2014; 11:20130962. [PMID: 24478281 DOI: 10.1098/rsif.2013.0962] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Wear particles and by-products from joint replacements and other orthopaedic implants may result in a local chronic inflammatory and foreign body reaction. This may lead to persistent synovitis resulting in joint pain and swelling, periprosthetic osteolysis, implant loosening and pathologic fracture. Strategies to modulate the adverse effects of wear debris may improve the function and longevity of joint replacements and other orthopaedic implants, potentially delaying or avoiding complex revision surgical procedures. Three novel biological strategies to mitigate the chronic inflammatory reaction to orthopaedic wear particles are reported. These include (i) interference with systemic macrophage trafficking to the local implant site, (ii) modulation of macrophages from an M1 (pro-inflammatory) to an M2 (anti-inflammatory, pro-tissue healing) phenotype in the periprosthetic tissues, and (iii) local inhibition of the transcription factor nuclear factor kappa B (NF-κB) by delivery of an NF-κB decoy oligodeoxynucleotide, thereby interfering with the production of pro-inflammatory mediators. These three approaches have been shown to be viable strategies for mitigating the undesirable effects of wear particles in preclinical studies. Targeted local delivery of specific biologics may potentially extend the lifetime of orthopaedic implants.
Collapse
Affiliation(s)
- S B Goodman
- Department of Orthopaedic Surgery, Stanford University, , Stanford, CA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Antonios JK, Yao Z, Li C, Rao AJ, Goodman SB. Macrophage polarization in response to wear particles in vitro. Cell Mol Immunol 2013; 10:471-82. [PMID: 24013843 PMCID: PMC3818297 DOI: 10.1038/cmi.2013.39] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 06/22/2013] [Accepted: 07/15/2013] [Indexed: 01/23/2023] Open
Abstract
Total joint replacement is a highly successful surgical procedure for treatment of patients with disabling arthritis and joint dysfunction. However, over time, with high levels of activity and usage of the joint, implant wear particles are generated from the articulating surfaces. These wear particles can lead to activation of an inflammatory reaction, and subsequent bone resorption around the implant (periprosthetic osteolysis). Cells of the monocyte/macrophage lineage orchestrate this chronic inflammatory response, which is dominated by a pro-inflammatory (M1) macrophage phenotype rather than an anti-inflammatory pro-tissue healing (M2) macrophage phenotype. While it has been shown that interleukin-4 (IL-4) selectively polarizes macrophages towards an M2 anti-inflammatory phenotype which promotes bone healing, rather than inflammation, little is known about the time course in which this occurs or conditions in which repolarization through IL-4 is most effective. The goal of this work was to study the time course of murine macrophage polarization and cytokine release in response to challenge with combinations of polymethyl methacrylate (PMMA) particles, lipopolysaccharide (LPS) and IL-4 in vitro. Treatment of particle-challenged monocyte/macrophages with IL-4 led to an initial suppression of pro-inflammatory cytokines and inducible nitric oxide synthase (iNOS) production and subsequent polarization into an M2 anti-inflammatory phenotype. This result was optimized when IL-4 was delivered before PMMA particle challenge, to an M1 phenotype rather than to uncommitted (M0) macrophages. The effects of this polarization were sustained over a 5-day time course. Polarization of M1 macrophages into an M2 phenotype may be a strategy to mitigate wear particle associated periprosthetic osteolysis.
Collapse
|
50
|
Abstract
The field of anatomic pathology has changed significantly over the last decades and, as a result of the technological developments in molecular pathology and genetics, has had increasing pressures put on it to become quantitative and to provide more information about protein expression on a cellular level in tissue sections. Multispectral imaging (MSI) has a long history as an advanced imaging modality and has been used for over a decade now in pathology to improve quantitative accuracy, enable the analysis of multicolor immunohistochemistry, and drastically reduce the impact of contrast-robbing tissue autofluorescence common in formalin-fixed, paraffin-embedded tissues. When combined with advanced software for the automated segmentation of different tissue morphologies (eg, tumor vs stroma) and cellular and subcellular segmentation, MSI can enable the per-cell quantitation of many markers simultaneously. This article covers the role that MSI has played in anatomic pathology in the analysis of formalin-fixed, paraffin-embedded tissue sections, discusses the technological aspects of why MSI has been adopted, and provides a review of the literature of the application of MSI in anatomic pathology.
Collapse
|