101
|
Muthukrishnan G, Wallimann A, Rangel-Moreno J, Bentley KLDM, Hildebrand M, Mys K, Kenney HM, Sumrall ET, Daiss JL, Zeiter S, Richards RG, Schwarz EM, Moriarty TF. Humanized Mice Exhibit Exacerbated Abscess Formation and Osteolysis During the Establishment of Implant-Associated Staphylococcus aureus Osteomyelitis. Front Immunol 2021; 12:651515. [PMID: 33815412 PMCID: PMC8012494 DOI: 10.3389/fimmu.2021.651515] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/02/2021] [Indexed: 12/17/2022] Open
Abstract
Staphylococcus aureus is the predominant pathogen causing osteomyelitis. Unfortunately, no immunotherapy exists to treat these very challenging and costly infections despite decades of research, and numerous vaccine failures in clinical trials. This lack of success can partially be attributed to an overreliance on murine models where the immune correlates of protection often diverge from that of humans. Moreover, S. aureus secretes numerous immunotoxins with unique tropism to human leukocytes, which compromises the targeting of immune cells in murine models. To study the response of human immune cells during chronic S. aureus bone infections, we engrafted non-obese diabetic (NOD)-scid IL2Rγnull (NSG) mice with human hematopoietic stem cells (huNSG) and analyzed protection in an established model of implant-associated osteomyelitis. The results showed that huNSG mice have increases in weight loss, osteolysis, bacterial dissemination to internal organs, and numbers of Staphylococcal abscess communities (SACs), during the establishment of implant-associated MRSA osteomyelitis compared to NSG controls (p < 0.05). Flow cytometry and immunohistochemistry demonstrated greater human T cell numbers in infected versus uninfected huNSG mice (p < 0.05), and that T-bet+ human T cells clustered around the SACs, suggesting S. aureus-mediated activation and proliferation of human T cells in the infected bone. Collectively, these proof-of-concept studies underscore the utility of huNSG mice for studying an aggressive form of S. aureus osteomyelitis, which is more akin to that seen in humans. We have also established an experimental system to investigate the contribution of specific human T cells in controlling S. aureus infection and dissemination.
Collapse
Affiliation(s)
- Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | - Alexandra Wallimann
- AO Research Institute Davos, Davos, Switzerland.,Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | | | - Karen Mys
- AO Research Institute Davos, Davos, Switzerland
| | - H Mark Kenney
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | | | - John L Daiss
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | | | | | - Edward M Schwarz
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States.,Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | | |
Collapse
|
102
|
Gimza BD, Cassat JE. Mechanisms of Antibiotic Failure During Staphylococcus aureus Osteomyelitis. Front Immunol 2021; 12:638085. [PMID: 33643322 PMCID: PMC7907425 DOI: 10.3389/fimmu.2021.638085] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus is a highly successful Gram-positive pathogen capable of causing both superficial and invasive, life-threatening diseases. Of the invasive disease manifestations, osteomyelitis or infection of bone, is one of the most prevalent, with S. aureus serving as the most common etiologic agent. Treatment of osteomyelitis is arduous, and is made more difficult by the widespread emergence of antimicrobial resistant strains, the capacity of staphylococci to exhibit tolerance to antibiotics despite originating from a genetically susceptible background, and the significant bone remodeling and destruction that accompanies infection. As a result, there is a need for a better understanding of the factors that lead to antibiotic failure in invasive staphylococcal infections such as osteomyelitis. In this review article, we discuss the different non-resistance mechanisms of antibiotic failure in S. aureus. We focus on how bacterial niche and destructive tissue remodeling impact antibiotic efficacy, the significance of biofilm formation in promoting antibiotic tolerance and persister cell formation, metabolically quiescent small colony variants (SCVs), and potential antibiotic-protected reservoirs within the substructure of bone.
Collapse
Affiliation(s)
- Brittney D Gimza
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James E Cassat
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
103
|
Zhang Y, Wang X, Huang X, Shen L, Zhang L, Shou D, Fan X. Transcriptome sequencing profiling identifies miRNA-331-3p as an osteoblast-specific miRNA in infected bone nonunion. Bone 2021; 143:115619. [PMID: 32858253 DOI: 10.1016/j.bone.2020.115619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 11/25/2022]
Abstract
Bone nonunion caused by bacterial infection accounts for bone fractures, bone trauma and bone transplantation surgeries. Severe consequences include delayed unions and amputation and result in functional limitations, work disability, and poor quality of life. However, the mechanism of bone nonunion remains unknown. In this study, we aimed to screen the miRNA biomarkers of bacterial bone infection and investigated whether miRNAs regulate the osteoblasts and thus contribute to bone nonunion. We established a miRNA-mRNA network based on high-throughput RNA sequencing to compare the model rabbits infected with Staphylococcus aureus with the control rabbits. After validation experiments, miRNA-331-3p and fibroblast growth factor 23 (FGF23) were found to be inversely correlated with the pathways of osteoblast mineralization and pathology of infected bone nonunion. In in vitro experiments, miRNA-331-3p was downregulated and FGF23 was upregulated in lipopolysaccharide (LPS)-induced mouse calvarial osteoblasts. Further studies of the mechanism showed that mutated of putative miRNA-331-3p can bind to FGF23 3'-untranslated region sites. MiRNA-331-3p acted as an osteoblast mineralization promoter by directly targeting FGF23. Downregulation of miRNA-331-3p led to inhibition of osteoblast mineralization by regulating the DKK1/β-catenin mediated signaling. Thus, we established an improved animal model and identified new bone-related biomarkers in the infected bone nonunion. The miRNA-331-3p biomarker was demonstrated to regulate osteoblast mineralization by targeting FGF23. The novel mechanism can be used as potential diagnostic biomarkers and therapeutic targets in the infected bone nonunion and other inflammatory bone disorders.
Collapse
Affiliation(s)
- Yang Zhang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Xuping Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Xiaowen Huang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Lifeng Shen
- Department of Orthopaedic Surgery, Zhejiang Provincial Tongde Hospital, Hangzhou 310012, China
| | - Li Zhang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Dan Shou
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
104
|
Bicer M, Cottrell GS, Widera D. Impact of 3D cell culture on bone regeneration potential of mesenchymal stromal cells. Stem Cell Res Ther 2021; 12:31. [PMID: 33413646 PMCID: PMC7791873 DOI: 10.1186/s13287-020-02094-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022] Open
Abstract
As populations age across the world, osteoporosis and osteoporosis-related fractures are becoming the most prevalent degenerative bone diseases. More than 75 million patients suffer from osteoporosis in the USA, the EU and Japan. Furthermore, it is anticipated that the number of patients affected by osteoporosis will increase by a third by 2050. Although conventional therapies including bisphosphonates, calcitonin and oestrogen-like drugs can be used to treat degenerative diseases of the bone, they are often associated with serious side effects including the development of oesophageal cancer, ocular inflammation, severe musculoskeletal pain and osteonecrosis of the jaw.The use of autologous mesenchymal stromal cells/mesenchymal stem cells (MSCs) is a possible alternative therapeutic approach to tackle osteoporosis while overcoming the limitations of traditional treatment options. However, osteoporosis can cause a decrease in the numbers of MSCs, induce their senescence and lower their osteogenic differentiation potential.Three-dimensional (3D) cell culture is an emerging technology that allows a more physiological expansion and differentiation of stem cells compared to cultivation on conventional flat systems.This review will discuss current understanding of the effects of different 3D cell culture systems on proliferation, viability and osteogenic differentiation, as well as on the immunomodulatory and anti-inflammatory potential of MSCs.
Collapse
Affiliation(s)
- Mesude Bicer
- Stem Cell Biology and Regenerative Medicine Group, Reading School of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, RG6 6AP, UK
| | - Graeme S Cottrell
- Cellular and Molecular Neuroscience, School of Pharmacy, University of Reading, Reading, UK
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, Reading School of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, RG6 6AP, UK.
| |
Collapse
|
105
|
Liu F, Dong J, Zhou D, Zhang Q. Identification of Key Candidate Genes Related to Inflammatory Osteolysis Associated with Vitamin E-Blended UHMWPE Debris of Orthopedic Implants by Integrated Bioinformatics Analysis and Experimental Confirmation. J Inflamm Res 2021; 14:3537-3554. [PMID: 34345178 PMCID: PMC8323865 DOI: 10.2147/jir.s320839] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/12/2021] [Indexed: 02/05/2023] Open
Abstract
PURPOSE This study aims to identify differentially expressed genes (DEGs) in macrophages exposed to ultra-high-molecular-weight polyethylene (UHMWPE) or vitamin E-blended UHMWPE (VE-UHMWPE) particles, thereby providing potential targets for the treatment of inflammatory osteolysis. METHODS The GSE104589 dataset of genome expression in macrophages exposed to UHMWPE and VE-UHMWPE was downloaded from the Gene Expression Omnibus database to identify DEGs. Functional enrichment analysis was performed using DAVID, and the corresponding protein-protein interaction (PPI) network was constructed from the STRING database. Important modules were selected using the molecular complex detection algorithm, and hub genes were identified in cytoHubba. MicroRNAs targeting these DEGs were obtained from the TarBase, miRTarBase, and miRecords databases, while transcription factors (TFs) targeting DEGs were predicted from the ENCODE database. Finally, the top five DEGs were validated by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS A total of 112 DEGs (44 upregulated and 68 downregulated DEGs) were screened. Immune and inflammatory responses were significantly related in gene ontology analysis, and 18 signaling pathways were enriched according to Kyoto Encyclopedia of Genes and Genomes pathway analysis. The PPI network involving 85 nodes and 266 protein pairs indicated that IL1β, CXCL1, ICAM1, CCL5 and CCL4 showed higher degrees. qRT-PCR analysis of the top five DEGs revealed a decreasing trend in the VE-UHMWPE group compared with the UHMWPE group. Key microRNAs (hsa-miR-144, hsa-miR-21, and hsa-miR-221) and TFs (RELA and NFKB1) were predicted to be correlated with the pathogenesis of inflammatory osteolysis through microRNA-TF regulatory network analysis. CONCLUSION The present study helps shed light on the molecular mechanisms underlying the changes in the wear-induced inflammatory process after blending vitamin E with UHMWPE. Hub genes including IL1β, CXCL1, ICAM1, CCL5, and CCL4, key microRNAs (hsa-miR-144, hsa-miR-21, and hsa-miR-221) and TFs (RELA and NFKB1) may serve as prognostic and therapeutic targets of inflammatory osteolysis.
Collapse
Affiliation(s)
- Fanxiao Liu
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Jun Dong
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Dongsheng Zhou
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Qingyu Zhang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
- Correspondence: Qingyu Zhang Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of ChinaTel/Fax +86-0531-68773201 Email
| |
Collapse
|
106
|
Pizzo RA, Patel JN, Viola A, Keller DM, Yoon RS, Liporace FA. Reducing Dislocations of Antibiotic Hip Spacers via Hybrid Cement-screw Constrained Liner Fixation: A Case Series. Hip Pelvis 2020; 32:207-213. [PMID: 33335869 PMCID: PMC7724021 DOI: 10.5371/hp.2020.32.4.207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose Infection following total hip arthroplasty is a challenging and devastating complication. In two-stage revision arthroplasty, antibiotic spacers, although efficacious, can be associated with an unacceptable rate of mechanical complications (e.g., fracture, dislocation). This series describes 15 patients with infected total hip prostheses treated with hybrid cement-screw fixation constrained liner antibiotic spacers to enhance stability and minimize mechanical complications. Materials and Methods All patients with an infected hip prosthesis undergoing two-stage revision arthroplasty at a single academic medical center were identified and screened for inclusion. Clinical and radiographic data including patient demographics and outcome measures were collected and retrospectively analyzed. Results Two patients died of unrelated causes at an average of 6-week postoperatively. Infections in the remaining thirteen patients (100%) were successfully eradicated; all underwent uncomplicated revision arthroplasty at a mean duration of 99.5 days after the placement of the antibiotic spacer. No dislocations, fractures, or other mechanical failures of any spacer were observed in this series. Conclusion The hybrid cement-screw fixation technique for constrained liner antibiotic spacers is a reliable and effective treatment method for eradicating prosthetic joint infections without mechanical complications.
Collapse
Affiliation(s)
- Richard A Pizzo
- Department of Orthopaedic Surgery, Jersey City Medical Center, Jersey City, NJ, USA
| | - Jay N Patel
- Department of Orthopaedic Surgery, Jersey City Medical Center, Jersey City, NJ, USA
| | - Anthony Viola
- Department of Orthopaedic Surgery, Jersey City Medical Center, Jersey City, NJ, USA
| | - David M Keller
- Department of Orthopaedic Surgery, Jersey City Medical Center, Jersey City, NJ, USA
| | - Richard S Yoon
- Department of Orthopaedic Surgery, Jersey City Medical Center, Jersey City, NJ, USA
| | - Frank A Liporace
- Department of Orthopaedic Surgery, Jersey City Medical Center, Jersey City, NJ, USA
| |
Collapse
|
107
|
Tetrandrine Inhibits Titanium Particle-Induced Inflammatory Osteolysis through the Nuclear Factor- κB Pathway. Mediators Inflamm 2020; 2020:1926947. [PMID: 33312069 PMCID: PMC7719528 DOI: 10.1155/2020/1926947] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/03/2020] [Accepted: 11/16/2020] [Indexed: 12/02/2022] Open
Abstract
Peri-implant osteolysis (PIO) and the subsequent aseptic loosening are the main reasons for artificial joint implant failure. Existing methods for treating aseptic loosening are far from satisfactory, necessitating advanced drug exploration. This study is aimed at investigating the effect and underlying mechanism of tetrandrine (Tet) on inflammatory osteolysis. We established a Ti particle-induced inflammatory osteolysis mouse model and administered Tet or an equal volume of phosphate-buffered saline (PBS). Two weeks later, specimens were collected. Histological staining showed that Tet administration inhibited Ti-stimulated osteolysis. Tartrate-resistant acid phosphate (TRAP) staining and transmission electron microscopy (TEM) demonstrated that osteoclast formation was remarkably inhibited in the groups treated with Tet in a dose-dependent manner. In addition, relevant inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6) were also significantly reduced in the calvaria of the Tet-treated groups. Exposure of receptor activator for nuclear factor-κB ligand- (RANKL-) induced bone marrow-derived macrophages (BMMs) and RAW264.7 cells to Tet significantly reduced osteoclast formation, F-actin ring formation, bone resorption, and the expression of relevant genes (matrix metallopeptidase 9 (MMP-9), TRAP, and nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1)) during osteoclastogenesis in vitro. Mechanistic studies using Western blotting demonstrated that Tet inhibited the nuclear factor (NF)-κB signaling pathway by decreasing the phosphorylation of inhibitor of NF-κB α (IκBα) and p65, which play important roles in osteoclast formation. Collectively, our data indicate that Tet suppressed Ti-induced inflammatory osteolysis and osteoclast formation in mice, suggesting that Tet has the potential to be developed to treat and prevent wear particle-induced inflammatory osteolysis.
Collapse
|
108
|
TGF-β in the Secretome of Irradiated Peripheral Blood Mononuclear Cells Supports In Vitro Osteoclastogenesis. Int J Mol Sci 2020; 21:ijms21228569. [PMID: 33202935 PMCID: PMC7696998 DOI: 10.3390/ijms21228569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Osteoclastogenesis required for bone remodeling is also a key pathologic mechanism of inflammatory osteolysis being controlled by paracrine factors released from dying cells. The secretome of irradiated, dying peripheral blood mononuclear cells (PBMCs) has a major impact on the differentiation of myeloid cells into dendritic cells, and macrophage polarization. The impact on osteoclastogenesis, however, has not been reported. For this aim, we used murine bone marrow macrophages exposed to RANKL and M-CSF to initiate osteoclastogenesis, with and without the secretome obtained from γ-irradiated PBMCs. We reported that the secretome significantly enhanced in vitro osteoclastogenesis as determined by means of histochemical staining of the tartrate-resistant acid phosphatase (TRAP), as well as the expression of the respective target genes, including TRAP and cathepsin K. Considering that TGF-β enhanced osteoclastogenesis, we confirmed the TGF-β activity in the secretome with a bioassay that was based on the increased expression of IL11 in fibroblasts. Neutralizing TGF-β by an antibody decreased the ability of the secretome to support osteoclastogenesis. These findings suggested that TGF-β released by irradiated PBMCs could enhance the process of osteoclastogenesis in vitro.
Collapse
|
109
|
Kim KM, Son HE, Min HY, Jang WG. Vitexin enhances osteoblast differentiation through phosphorylation of Smad and expression of Runx2 at in vitro and ex vivo. Mol Biol Rep 2020; 47:8809-8817. [PMID: 33118104 DOI: 10.1007/s11033-020-05929-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 10/17/2020] [Indexed: 01/23/2023]
Abstract
Vitexin (apigenin-8-C-d-glucopyranoside) is a flavonoid isolated from natural sources. It has been employed as an anti-oxidant, anti-inflammatory, and anti-cancer agent, and is used as a traditional Chinese medicine to treat a variety of illnesses. The present study investigated the effect of vitexin on osteoblast differentiation of C3H10T1/2 mesenchymal stem cells, MC3T3-E1 preosteoblast, mouse calvarial primary cells, and primary bone marrow stem cells (BMSCs). RT-PCR and quantitative PCR demonstrated that vitexin increased mRNA expression of the osteogenic genes distal-less homeobox 5 (Dlx5) and Runxt-related transcription factor 2 (Runx2). Vitexin also increased the Dlx5 and Runx2 protein levels, Smad1/5/9 phosphorylation, and alkaline phosphatase (ALP) activity. In addition, vitexin increased Runx2-luciferase activity. Moreover, knockdown of Runx2 attenuated the increase in ALP activity induced by vitexin. These results demonstrate that vitexin enhances osteoblast differentiation via Runx2.
Collapse
Affiliation(s)
- Kyeong-Min Kim
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk, 38453, Republic of Korea.,Research a of Anti-Aging, Daegu University, Gyeongbuk, 38453, Republic of Korea
| | - Hyo-Eun Son
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk, 38453, Republic of Korea.,Research a of Anti-Aging, Daegu University, Gyeongbuk, 38453, Republic of Korea
| | - Hyeon-Young Min
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk, 38453, Republic of Korea.,Research a of Anti-Aging, Daegu University, Gyeongbuk, 38453, Republic of Korea
| | - Won-Gu Jang
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk, 38453, Republic of Korea. .,Research a of Anti-Aging, Daegu University, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
110
|
Kuchler U, Heimel P, Stähli A, Josef Strauss F, Luza B, Gruber R. Impact of DBBM Fragments on the Porosity of the Calvarial Bone: A Pilot Study on Mice. MATERIALS 2020; 13:ma13214748. [PMID: 33114211 PMCID: PMC7660694 DOI: 10.3390/ma13214748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/27/2022]
Abstract
Deproteinized bovine bone mineral (DBBM) is brittle and can break into fragments. Here, we examined whether DBBM fragments have an impact on mice calvarial bone during bone augmentation. DBBM was either randomly crushed (DBBM fragments) or left undisturbed (DBBM granules). Then, DBBM fragments or original DBBM granules were placed onto calvarial bone in 20 BALB/c mice. Following random allocation, ten mice received DBBM fragments and ten mice received original DBBM granules. After fourteen days of healing, micro computed tomography (micro-CT) and histological analysis of the augmented sites were performed. The primary outcome was the porosity of the calvarial bone. The micro-CT analysis revealed that DBBM fragments failed to significantly change the porosity of the calvarial bone as compared with original DBBM granules, despite the slightly higher bone resorption in the DBBM fragment group, 10.3% (CI 6.3–11.6) versus 6.1% (CI 4.1–7.8, p = 0.355), respectively. The cortical bone volume was not altered by DBBM fragments as compared with original DBBM granules, i.e., 79.0% (CI 78.9–81.2) versus 81.5% (CI 80.1–83.3, p = 0.357), respectively. The DBBM fragment group revealed similar bone thickness values as compared with the DBBM granules group, i.e., 0.26 mm (CI 0.23–0.29) versus 0.25 mm (CI 0.22–0.27, p = 0.641), respectively. The histological evaluation supported the micro-CT observations, displaying minor signs of porosity and resorption. The particle-size distribution analysis confirmed a shift towards smaller particle sizes in the DBBM fragment group. These findings suggest that DBBM fragments behave similarly to original DBBM granules in terms of bone morphological changes at augmented sites.
Collapse
Affiliation(s)
- Ulrike Kuchler
- Department of Oral Surgery, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria;
| | - Patrick Heimel
- Core Facility Hard Tissue and Biomaterial Research, Karl Donath Laboratory, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (P.H.); (B.L.)
- Ludwig Boltzmann Institute for Clinical and Experimental Traumatology, 1090 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1090 Vienna, Austria
| | - Alexandra Stähli
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (A.S.); (F.J.S.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
| | - Franz Josef Strauss
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (A.S.); (F.J.S.)
- Department of Conservative Dentistry, School of Dentistry, University of Chile, Santiago 8380544, Chile
- Clinic of Reconstructive Dentistry, Center of Dental Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Bernadette Luza
- Core Facility Hard Tissue and Biomaterial Research, Karl Donath Laboratory, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (P.H.); (B.L.)
- Ludwig Boltzmann Institute for Clinical and Experimental Traumatology, 1090 Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (A.S.); (F.J.S.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Correspondence: ; Tel.: +43-699-107-18-472
| |
Collapse
|
111
|
Bangun K, Sukasah CL, Dilogo IH, Indrani DJ, Siregar NC, Pandelaki J, Iskandriati D, Kekalih A, Halim J. Bone Growth Capacity of Human Umbilical Cord Mesenchymal Stem Cells and BMP-2 Seeded Into Hydroxyapatite/Chitosan/Gelatin Scaffold in Alveolar Cleft Defects: An Experimental Study in Goat. Cleft Palate Craniofac J 2020; 58:707-717. [PMID: 34047209 DOI: 10.1177/1055665620962360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE To evaluate bone regeneration in alveolar defects treated with human umbilical cord-derived mesenchymal stem cells (hUCMSCs), hydroxyapatite/chitosan/gelatin (HA/CS/Gel) scaffold, and bone morphogenic protein-2 (BMP-2) in Capra hircus models. DESIGN Randomized posttest-only control group design. SETTING Animal Hospital at Bogor Agricultural Institute. PARTICIPANTS Healthy and equally treated 24 female Capra hircus/goats. INTERVENTION Animals were randomly assigned to 3 experimental group design (iliac crest alveolar bone graft/ICABG [control], HA/Cs/Gel+BMP-2 [Novosys], and HA/Cs/Gel+BMP-2+UCMSCs). Graft materials were implanted in surgically made alveolar defects. MAIN OUTCOME MEASURES Postoperative functional score and operating time were assessed. New bone growth, bone density, inflammatory cells recruitment, and neoangiogenesis were evaluated based on radiological and histological approach at 2 time points, week 4 and 12. Statistical analysis was done between treatment groups. RESULTS Operating time was 34% faster and functional score 94.5% more superior in HA/Cs/Gel+BMP-2+hUCMSC group. Bone growth capacity in HA/Cs/Gel+BMP-2+UCMSCs mimicked ICABG, but ICABG showed possibility of bone loss between week 4 and 12. The HA/Cs/Gel+BMP-2+UCMSCs showed early bone repopulation and unseen inflammatory cells and angiogenesis on week 12. DISCUSSION AND CONCLUSION The HA/Cs/Gel+BMP-2+hUCMSCs were superior in enhancing new bone growth without donor site morbidity compared to ICABG. The presence of hUCMSCs in tissue-engineered alveolar bone graft (ABG), supported with paracrine activity of the resident stem cells, initiated earlier new bone repopulation, and completed faster bone regeneration. The HA/Cs/Gel scaffold seeded with UCMSCs+BMP-2 is a safe substitute of ICABG to close alveolar bone defects suitable for patients with cleft lip, alveolus, and palate.
Collapse
Affiliation(s)
- Kristaninta Bangun
- Department of Plastic and Reconstructive Surgery, Cleft and Craniofacial Center, 364090Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Chaula L Sukasah
- Department of Plastic and Reconstructive Surgery, Cleft and Craniofacial Center, 364090Cipto Mangunkusumo General Hospital, Jakarta, Indonesia.,95338Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Ismail H Dilogo
- Unit Pelayanan Terpadu Teknologi Kedokteran Sel Punca (Stem Cell Research Center), 364090Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Decky J Indrani
- Department of Dental Material Science and Technology, 95338Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Nurjati Chairani Siregar
- 95338Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Anatomical Pathology Department, 364090Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Jacub Pandelaki
- 95338Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Radiology Department of 364090Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Diah Iskandriati
- Primate Research Center of 360976Bogor Agricultural Institute, Bogor, Indonesia
| | - Aria Kekalih
- 95338Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Community Medicine Department, 364090Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Jessica Halim
- 95338Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,364090Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| |
Collapse
|
112
|
Schoon J, Hesse B, Rakow A, Ort MJ, Lagrange A, Jacobi D, Winter A, Huesker K, Reinke S, Cotte M, Tucoulou R, Marx U, Perka C, Duda GN, Geissler S. Metal-Specific Biomaterial Accumulation in Human Peri-Implant Bone and Bone Marrow. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000412. [PMID: 33101844 PMCID: PMC7578891 DOI: 10.1002/advs.202000412] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/09/2020] [Indexed: 05/13/2023]
Abstract
Metallic implants are frequently used in medicine to support and replace degenerated tissues. Implant loosening due to particle exposure remains a major cause for revision arthroplasty. The exact role of metal debris in sterile peri-implant inflammation is controversial, as it remains unclear whether and how metals chemically alter and potentially accumulate behind an insulating peri-implant membrane, in the adjacent bone and bone marrow (BM). An intensively focused and bright synchrotron X-ray beam allows for spatially resolving the multi-elemental composition of peri-implant tissues from patients undergoing revision surgery. In peri-implant BM, particulate cobalt (Co) is exclusively co-localized with chromium (Cr), non-particulate Cr accumulates in the BM matrix. Particles consisting of Co and Cr contain less Co than bulk alloy, which indicates a pronounced dissolution capacity. Particulate titanium (Ti) is abundant in the BM and analyzed Ti nanoparticles predominantly consist of titanium dioxide in the anatase crystal phase. Co and Cr but not Ti integrate into peri-implant bone trabeculae. The characteristic of Cr to accumulate in the intertrabecular matrix and trabecular bone is reproducible in a human 3D in vitro model. This study illustrates the importance of updating the view on long-term consequences of biomaterial usage and reveals toxicokinetics within highly sensitive organs.
Collapse
Affiliation(s)
- Janosch Schoon
- Julius Wolff InstituteCharité – Universitätsmedizin BerlinBerlin13353Germany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of HealthBerlin10178Germany
- Berlin‐Brandenburg School for Regenerative TherapiesCharité – Universitätsmedizin BerlinBerlin13353Germany
| | - Bernhard Hesse
- Xploraytion GmbHBerlin10625Germany
- European Synchrotron Radiation FacilityGrenoble38000France
| | - Anastasia Rakow
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of HealthBerlin10178Germany
- Center for Musculoskeletal SurgeryCharité – Universitätsmedizin BerlinBerlin10117Germany
| | - Melanie J. Ort
- Julius Wolff InstituteCharité – Universitätsmedizin BerlinBerlin13353Germany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of HealthBerlin10178Germany
- Berlin‐Brandenburg School for Regenerative TherapiesCharité – Universitätsmedizin BerlinBerlin13353Germany
| | - Adrien Lagrange
- Xploraytion GmbHBerlin10625Germany
- Department of Materials Science and EngineeringInstitute of Materials Science and TechnologiesTechnische Universität BerlinBerlin10623Germany
| | - Dorit Jacobi
- Julius Wolff InstituteCharité – Universitätsmedizin BerlinBerlin13353Germany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of HealthBerlin10178Germany
| | | | - Katrin Huesker
- Endocrinology and Immunology DepartmentInstitute for Medical DiagnosticsBerlin12247Germany
| | - Simon Reinke
- Julius Wolff InstituteCharité – Universitätsmedizin BerlinBerlin13353Germany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of HealthBerlin10178Germany
| | - Marine Cotte
- European Synchrotron Radiation FacilityGrenoble38000France
- CNRSLaboratoire d'archéologie moléculaire et structuraleLAMSSorbonne UniversitéParis75005France
| | - Remi Tucoulou
- European Synchrotron Radiation FacilityGrenoble38000France
| | | | - Carsten Perka
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of HealthBerlin10178Germany
- Berlin‐Brandenburg School for Regenerative TherapiesCharité – Universitätsmedizin BerlinBerlin13353Germany
- Center for Musculoskeletal SurgeryCharité – Universitätsmedizin BerlinBerlin10117Germany
| | - Georg N. Duda
- Julius Wolff InstituteCharité – Universitätsmedizin BerlinBerlin13353Germany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of HealthBerlin10178Germany
- Berlin‐Brandenburg School for Regenerative TherapiesCharité – Universitätsmedizin BerlinBerlin13353Germany
| | - Sven Geissler
- Julius Wolff InstituteCharité – Universitätsmedizin BerlinBerlin13353Germany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of HealthBerlin10178Germany
- Berlin‐Brandenburg School for Regenerative TherapiesCharité – Universitätsmedizin BerlinBerlin13353Germany
| |
Collapse
|
113
|
Multitasking by the OC Lineage during Bone Infection: Bone Resorption, Immune Modulation, and Microbial Niche. Cells 2020; 9:cells9102157. [PMID: 32987689 PMCID: PMC7598711 DOI: 10.3390/cells9102157] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 01/18/2023] Open
Abstract
Bone infections, also known as infectious osteomyelitis, are accompanied by significant inflammation, osteolysis, and necrosis. Osteoclasts (OCs) are the bone-resorbing cells that work in concert with osteoblasts and osteocytes to properly maintain skeletal health and are well known to respond to inflammation by increasing their resorptive activity. OCs have typically been viewed merely as effectors of pathologic bone resorption, but recent evidence suggests they may play an active role in the progression of infections through direct effects on pathogens and via the immune system. This review discusses the host- and pathogen-derived factors involved in the in generation of OCs during infection, the crosstalk between OCs and immune cells, and the role of OC lineage cells in the growth and survival of pathogens, and highlights unanswered questions in the field.
Collapse
|
114
|
Tang W, Xiao L, Ge G, Zhong M, Zhu J, Qin J, Feng C, Zhang W, Bai J, Zhu X, Wei M, Geng D, Wang Z. Puerarin inhibits titanium particle-induced osteolysis and RANKL-induced osteoclastogenesis via suppression of the NF-κB signaling pathway. J Cell Mol Med 2020; 24:11972-11983. [PMID: 32896108 PMCID: PMC7578865 DOI: 10.1111/jcmm.15821] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023] Open
Abstract
Osteolysis around the prosthesis and subsequent aseptic loosening are the main causes of prosthesis failure. Inflammation due to wear particles and osteoclast activation are the key factors in osteolysis and are also potential targets for the treatment of osteolysis. However, it is not clear whether puerarin can inhibit chronic inflammation and alleviate osteolysis. In this study, we investigated the effect of puerarin on Ti particle-induced inflammatory osteolysis in vivo in rat femoral models and in vitro in receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast activation models. Our in vivo results showed that puerarin significantly inhibited Ti particle-induced osteolysis and the expression of matrix metallopeptidase 9 (MMP-9), nuclear factor of activated T cells 1 (NFATc1), tumour necrosis factor (TNF)-α and interleukin (IL)-6. In vitro, puerarin prevented RANKL-induced osteoclast differentiation, bone resorption and F-actin ring formation in a concentration-dependent manner. Furthermore, puerarin decreased the phosphorylation of p65 and prevented p65 moving from the cytoplasm to the nucleus. Puerarin also reduced the expression of osteoclast-specific factors and inhibited the inflammatory response. In conclusion, our study proves that puerarin can block the NF-κB signalling pathway to inhibit osteoclast activation and inflammatory processes, which provides a new direction for the treatment of osteolysis-related diseases.
Collapse
Affiliation(s)
- Wenkai Tang
- Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Long Xiao
- Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China.,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China.,Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Gaoran Ge
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mengdan Zhong
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China.,Department of Endocrinology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Jie Zhu
- Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China.,Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Jialin Qin
- Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Chencheng Feng
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China.,Department of Endocrinology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Wenhao Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiaxiang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Minggang Wei
- Traditional Chinese Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhirong Wang
- Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China.,Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| |
Collapse
|
115
|
Qian Z, Zhong Z, Ni S, Li D, Zhang F, Zhou Y, Kang Z, Qian J, Yu B. Cytisine attenuates bone loss of ovariectomy mouse by preventing RANKL-induced osteoclastogenesis. J Cell Mol Med 2020; 24:10112-10127. [PMID: 32790170 PMCID: PMC7520284 DOI: 10.1111/jcmm.15622] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/07/2020] [Accepted: 06/23/2020] [Indexed: 12/22/2022] Open
Abstract
Postmenopausal Osteoporosis (PMOP) is oestrogen withdrawal characterized of much production and activation by osteoclast in the elderly female. Cytisine is a quinolizidine alkaloid that comes from seeds or other plants of the Leguminosae (Fabaceae) family. Cytisine has been shown several potential pharmacological functions. However, its effects on PMOP remain unknown. This study designed to explore whether Cytisine is able to suppress RANKL-induced osteoclastogenesis and prevent the bone loss induced by oestrogen deficiency in ovariectomized (OVX) mice. In this study, we investigated the effect of Cytisine on RAW 264.7 cells and bone marrow monocytes (BMMs) derived osteoclast culture system in vitro and observed the effect of Cytisine on ovariectomized (OVX) mice model to imitate postmenopausal osteoporosis in vivo. We found that Cytisine inhibited F-actin ring formation and tartrate-resistant acid phosphatase (TRAP) staining in dose-dependent ways, as well as bone resorption by pit formation assays. For molecular mechanism, Cytisine suppressed RANK-related trigger RANKL by phosphorylation JNK/ERK/p38-MAPK, IκBα/p65-NF-κB, and PI3K/AKT axis and significantly inhibited these signalling pathways. However, the suppression of PI3K-AKT-NFATc1 axis was rescued by AKT activator SC79. Meanwhile, Cytisine inhibited RANKL-induced RANK-TRAF6 association and RANKL-related gene and protein markers such as NFATc1, Cathepsin K, MMP-9 and TRAP. Our study indicated that Cytisine could suppress bone loss in OVX mouse through inhibited osteoclastogenesis. All data provide the evidence that Cytisine may be a promising agent in the treatment of osteoclast-related diseases such as osteoporosis.
Collapse
Affiliation(s)
- Zhi Qian
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
- Department of Orthopaedic SurgeryZhangye People's Hospital affiliated to Hexi UniversityZhangye CityChina
| | - Zeyuan Zhong
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Shuo Ni
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Dejian Li
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Fangxue Zhang
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Ying Zhou
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Zhanrong Kang
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Jun Qian
- Department of Orthopaedic SurgeryZhangye People's Hospital affiliated to Hexi UniversityZhangye CityChina
| | - Baoqing Yu
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| |
Collapse
|
116
|
Wang Q, Ge G, Liang X, Bai J, Wang W, Zhang W, Zheng K, Yang S, Wei M, Yang H, Xu Y, Liu B, Geng D. Punicalagin ameliorates wear-particle-induced inflammatory bone destruction by bi-directional regulation of osteoblastic formation and osteoclastic resorption. Biomater Sci 2020; 8:5157-5171. [PMID: 32840273 DOI: 10.1039/d0bm00718h] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Periprosthetic osteolysis (PPO) and subsequent aseptic loosening are the main causes of implant failure and revision surgery. Emerging evidence has suggested that wear-particle-induced chronic inflammation, osteoblast inhibition and osteoclast formation at the biointerface of implant materials are responsible for PPO. Punicalagin (PCG), a polyphenolic compound molecularly extracted from pomegranate rinds, plays a critical role in antioxidant, anticancer and anti-inflammatory activities. However, whether PCG could attenuate chronic inflammation and bone destruction at sites of titanium (Ti)-particle-induced osteolysis remains to be determined. In this study, we explored the effect of PCG on Ti-particle-induced osteolysis in vivo and osteoblast and osteoclast differentiation in vitro. We found that PCG could relieve wear-particle-induced bone destruction in a murine calvarial osteolysis model by increasing bone formation activity and suppressing bone resorption activity. PCG treatment also reduced the Ti-particle-induced inflammatory response in vivo and vitro. In addition, we also observed that PCG promotes osteogenic differentiation of MC3T3-E1 cells under inflammatory conditions and inhibits RANKL-induced osteoclast formation of bone marrow-derived macrophages (BMMs). Meanwhile, the induction of the RANKL to OPG ratio was reversed by PCG treatment in vivo and in vitro, which demonstrated that PCG could also indirectly inhibit osteoclastogenesis. Collectively, our findings suggest that PCG represents a potential approach for the treatment of wear-particle-induced inflammatory osteolysis.
Collapse
Affiliation(s)
- Qing Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Amini A, Namvarpour Z, Namvarpour M, Raoofi A. Risperidone accelerates bone loss in rats with autistic-like deficits induced by maternal lipopolysaccharides exposure. Life Sci 2020; 258:118197. [PMID: 32781059 DOI: 10.1016/j.lfs.2020.118197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/22/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
AIMS Patients with neurodevelopmental disorders, usually suffer from bone diseases. Many studies have revealed a higher risk of fracture after atypical antipsychotic drug Risperidone (RIS) treatment, which is usually used to treat such disorders. It remains debatable whether neurodevelopmental disorders by itself are the cause of bone diseases or pharmacotherapy may be the reason. MATERIALS AND METHODS This study attempts to evaluate the biomechanical, histological, stereological, and molecular properties of bones in the offspring of Lipopolysaccharide (LPS) and saline-treated mothers that received saline, drug vehicle or the atypical antipsychotic drug risperidone (RIS) at different days of postnatal development. After postnatal drug treatment, animals were assessed for autistic-like behaviors. Then their bones were taken for evaluations. RESULTS Maternal LPS exposure resulted in deficits in all behavioral tests and RIS ameliorated these behaviors (p < 0.01& p < 0.05). The administration of LPS and RIS individually led to a significant decrease in the biomechanical parameters such as bone stiffness, strength and the energy used to fracture of bone. The numerical density of osteocalcin-positive cells were significantly decreased in these groups. These rats also had decreased RUNX2 and osteocalcin gene expression. When LPS rats were treated with RIS, these conditions were accelerated (p < 0.001). DISCUSSIONS The results of our preclinical study, consistent with previous studies in animals, explore that autistic-like deficits induced by prenatal exposure to LPS, can reduce bone stability and bone mass similar to those observed in neurodevelopmental disorders, and, for the first time, reveal that this condition worsened when these animals were treated with RIS.
Collapse
Affiliation(s)
- Abdollah Amini
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Namvarpour
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute for Cognitive Science Studies (ICSS), Tehran, Iran.
| | - Mozhdeh Namvarpour
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Raoofi
- Leishmaniasis Research Center, Department of Anatomy, Sabzevar University of Medical Sciences, Sabzevar, Iran
| |
Collapse
|
118
|
Zhu X, Zhang Y, Yang H, He F, Lin J. Melatonin suppresses Ti-particle-induced inflammatory osteolysis via activation of the Nrf2/Catalase signaling pathway. Int Immunopharmacol 2020; 88:106847. [PMID: 32771943 DOI: 10.1016/j.intimp.2020.106847] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/27/2020] [Accepted: 07/26/2020] [Indexed: 12/15/2022]
Abstract
Aseptic loosening induced by osteolysis is recognized as a late complication of joint replacement. Osteoclasts stimulated by Titanium (Ti) nanoparticles play a critical role in periprosthetic osteolysis. Emerging evidence indicates that melatonin, a hormone primarily synthesized by the pineal gland, has been shown an inhibitory effect on osteoclast formation. However, it is unclear whether melatonin could suppress Ti-particle-induced osteoclastogenesis and what the underlying mechanisms were involved in. Herein, we aimed to investigate the effect of melatonin on osteoclast differentiation and osteolysis stimulated by Ti particles. Our results showed that the in vitro osteoclastogenesis of mouse bone marrow monocytes (BMMs) stimulated by Ti particles was suppressed by melatonin treatments in a dose-dependent manner. Further experiments revealed that melatonin up-regulated the expression of the nuclear factor erythroid 2-related factor 2 (Nrf2) and catalase (CAT) at both the mRNA and protein levels. The role of the Nrf2/CAT signaling pathway was confirmed by the fact that silencing the expression of NRF2 by small interfering RNA (siRNA) counteracted the anti-osteolysis effects of melatonin. Furthermore, in vivo intraperitoneal injection of melatonin successfully attenuated periprosthetic osteolysis induced by Ti particles in a murine calvarial model. Our findings demonstrate that melatonin is a promising therapeutic agent for treating periprosthetic osteolysis by inhibiting the Ti-particle-stimulated osteoclastogenesis via activation of the Nrf2/Catalase signaling pathway.
Collapse
Affiliation(s)
- Xu Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Yazhong Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215007, China.
| | - Jun Lin
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
119
|
Chen YJ, Wang X, Zhang HY, Du YN, Jin XJ, Zhang ZF. Inverse association between bone mineral density and fibrinogen in menopausal women. Climacteric 2020; 24:146-150. [PMID: 32602367 DOI: 10.1080/13697137.2020.1776249] [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: 10/24/2022]
Abstract
OBJECTIVE Inflammatory diseases are risk factors for osteoporosis. We aimed to explore whether fibrinogen, which is linked to chronic inflammation, is associated with bone mineral density (BMD) in menopausal women. METHODS In this cross-sectional study, we analyzed 339 menopausal women from Zhejiang Province between January 2016 and October 2019. Linear regression analysis was performed to assess the relationship between fibrinogen and BMD. RESULTS Significant inverse association was observed between the serum fibrinogen level and BMD in menopausal women. The mean BMD in each quartile of fibrinogen level was 0.901, 0.897, 0.892, and 0.855 g/cm2, respectively (p = 0.027). After adjusting for age, body mass index, metabolic profiles, blood inflammatory factors, and serum levels of estradiol, calcium, phosphorus, and alkaline phosphatase, fibrinogen levels remained significantly associated with BMD (regression coefficients for quartiles 1-3 vs. quartile 4 were 0.046, 0.027, and 0.036, respectively; p for trend <0.05). CONCLUSIONS Higher fibrinogen levels were associated with lower BMD in menopausal women, which was independent of age, body mass index, estradiol, and other factors. Therefore, serum fibrinogen can be used as a new predictor of reduced BMD in menopausal women.
Collapse
Affiliation(s)
- Y J Chen
- Department of fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - X Wang
- Nanjing Medical University, Nanjing, China
| | - H Y Zhang
- Department of Obstetrics and Gynecology, Hangzhou Women's Hospital, Hangzhou, China
| | - Y N Du
- Nanjing Medical University, Nanjing, China
| | - X J Jin
- Department of Obstetrics and Gynecology, Hangzhou Women's Hospital, Hangzhou, China
| | - Z F Zhang
- Department of fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.,Nanjing Medical University, Nanjing, China.,Department of Obstetrics and Gynecology, Hangzhou Women's Hospital, Hangzhou, China
| |
Collapse
|
120
|
Abstract
Osteomyelitis, or inflammation of bone, is most commonly caused by invasion of bacterial pathogens into the skeleton. Bacterial osteomyelitis is notoriously difficult to treat, in part because of the widespread antimicrobial resistance in the preeminent etiologic agent, the Gram-positive bacterium Staphylococcus aureus Bacterial osteomyelitis triggers pathological bone remodeling, which in turn leads to sequestration of infectious foci from innate immune effectors and systemically delivered antimicrobials. Treatment of osteomyelitis therefore typically consists of long courses of antibiotics in conjunction with surgical debridement of necrotic infected tissues. Even with these extreme measures, many patients go on to develop chronic infection or sustain disease comorbidities. A better mechanistic understanding of how bacteria invade, survive within, and trigger pathological remodeling of bone could therefore lead to new therapies aimed at prevention or treatment of osteomyelitis as well as amelioration of disease morbidity. In this minireview, we highlight recent developments in our understanding of how pathogens invade and survive within bone, how bacterial infection or resulting innate immune responses trigger changes in bone remodeling, and how model systems can be leveraged to identify new therapeutic targets. We review the current state of osteomyelitis epidemiology, diagnostics, and therapeutic guidelines to help direct future research in bacterial pathogenesis.
Collapse
|
121
|
Wang Y, Lin Y, Cheng C, Chen P, Zhang P, Wu H, Li K, Deng Y, Qian J, Zhang X, Yu B. NF-κB/TWIST1 Mediates Migration and Phagocytosis of Macrophages in the Mice Model of Implant-Associated Staphylococcus aureus Osteomyelitis. Front Microbiol 2020; 11:1301. [PMID: 32595631 PMCID: PMC7304240 DOI: 10.3389/fmicb.2020.01301] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus (S. aureus) infection-induced osteomyelitis is a great challenge in clinic treatment. Identification of the essential genes and biological processes that are specifically changed in mononuclear cells at an early stage of S. aureus osteomyelitis is of great clinical significance. Based on transcriptional dataset GSE16129 available publicly, a bioinformatic analysis was performed to identify the differentially expressed genes of osteomyelitis caused by S. aureus infection. ERBB2, TWIST1, and NANOG were screened out as the most valuable osteomyelitis-related genes (OMRGs). A mice model of implant-associated S. aureus osteomyelitis was used to verify the above genes. We found significantly up-regulated expression of TWIST1 in macrophages and accumulation of macrophages around the infected implant. Meanwhile, S. aureus infection increased the expression of TWIST1, MMP9, and MMP13, and stimulated the migration and phagocytosis function of Raw 264.7 cells. Additionally, knock-down of the expression of TWIST1 by siRNA could significantly down-regulate MMP9 and MMP13 and suppress the migration and phagocytosis ability of macrophages in response to S. aureus infection. Furthermore, we found that NF-κB signaling was activated in Raw 264.7 cells by S. aureus and that inhibition of NF-κB signaling by Bay11-7082 blocked the expression of TWIST1, MMP9, and MMP13 as well as cell migration and phagocytosis evoked by S. aureus. Our findings demonstrate that NF-κB/TWIST1 is necessary for migration and phagocytosis of macrophages in response to S. aureus infection. Our study highlights the essential role of NF-κB/TWIST1 in early innate immune response to S. aureus infection in bone.
Collapse
Affiliation(s)
- Yutian Wang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yihuang Lin
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Caiyu Cheng
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pengyu Chen
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ping Zhang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hangtian Wu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kaiqun Li
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Deng
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jikun Qian
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xianrong Zhang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
122
|
Shang N, Wu J. Egg-Derived Tripeptide IRW Attenuates LPS-Induced Osteoclastogenesis in RAW 264.7 Macrophages via Inhibition of Inflammatory Responses and NF-κB/MAPK Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:6132-6141. [PMID: 32383875 DOI: 10.1021/acs.jafc.0c01159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Excessive bone resorption, because of increased osteoclastic activity, is a key underlying cause of osteolytic disorders. Lipopolysaccharide (LPS) is a potent factor to stimulate osteoclastic activity by inducing inflammatory stress. An egg-derived tripeptide IRW (Ile-Arg-Trp) was previously shown to exert anti-inflammatory activity. The overall objective of this study was to investigate the effect of IRW on inhibiting LPS-induced osteoclastogenesis and inflammatory bone resorption in the mouse macrophage RAW 264.7 cells. IRW (25 and 50 μM) significantly inhibited the LPS-induced osteoclast formation and resorptive activity. Meanwhile, IRW significantly suppressed the LPS-induced expression of TNF-α, IL-6, iNOS, COXII, NO, and PGE2. Furthermore, IRW regulated a group of osteoclastogenesis-associated factors (TRAF6, c-Fos, NFATc1, and cathepsin K) because of the inhibition of LPS-activated NF-κB and MAPK pathways. In conclusion, our study suggested the ability of IRW to prevent LPS-induced inflammatory bone resorption activity via the inhibition of inflammatory responses and the activation of osteoclastogenesis-associated signaling pathways.
Collapse
Affiliation(s)
- Nan Shang
- Dept. of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jianping Wu
- Dept. of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
123
|
Yu B, Bai J, Shi J, Shen J, Guo X, Liu Y, Ge G, Lin J, Tao Y, Yang H, Xu Y, Qu Q, Geng D. MiR-106b inhibition suppresses inflammatory bone destruction of wear debris-induced periprosthetic osteolysis in rats. J Cell Mol Med 2020; 24:7490-7503. [PMID: 32485091 PMCID: PMC7339204 DOI: 10.1111/jcmm.15376] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/10/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022] Open
Abstract
Aseptic loosening caused by periprosthetic osteolysis (PPO) is the main reason for the primary artificial joint replacement. Inhibition of inflammatory osteolysis has become the main target of drug therapy for prosthesis loosening. MiR‐106b is a newly discovered miRNA that plays an important role in tumour biology, inflammation and the regulation of bone mass. In this study, we analysed the in vivo effect of miR‐106b on wear debris‐induced PPO. A rat implant loosening model was established. The rats were then administrated a lentivirus‐mediated miR‐106b inhibitor, miR‐106b mimics or an equivalent volume of PBS by tail vein injection. The expression levels of miR‐106b were analysed by real‐time PCR. Morphological changes in the distal femurs were assessed via micro‐CT and histopathological analysis, and cytokine expression levels were examined via immunohistochemical staining and ELISA. The results showed that treatment with the miR‐106b inhibitor markedly suppressed the expression of miR‐106b in distal femur and alleviated titanium particle‐induced osteolysis and bone loss. Moreover, the miR‐106b inhibitor decreased TRAP‐positive cell numbers and suppressed osteoclast formation, in addition to promoting the activity of osteoblasts and increasing bone formation. MiR‐106b inhibition also significantly regulated macrophage polarization and decreased the inflammatory response as compared to the control group. Furthermore, miR‐106b inhibition blocked the activation of the PTEN/PI3K/AKT and NF‐κB signalling pathways. Our findings indicated that miR‐106b inhibition suppresses wear particles‐induced osteolysis and bone destruction and thus may serve as a potential therapy for PPO and aseptic loosening.
Collapse
Affiliation(s)
- Binqing Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Shi
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jining Shen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaobin Guo
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gaoran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiayi Lin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunxia Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiuxia Qu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
124
|
Chen Y, Wang Y, Tang R, Yang J, Dou C, Dong Y, Sun D, Zhang C, Zhang L, Tang Y, Dai Q, Luo F, Xu J, Dong S. Dendritic cells-derived interferon-λ1 ameliorated inflammatory bone destruction through inhibiting osteoclastogenesis. Cell Death Dis 2020; 11:414. [PMID: 32488049 PMCID: PMC7265503 DOI: 10.1038/s41419-020-2612-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 01/08/2023]
Abstract
Bone infection contributing to inflammatory osteolysis is common in orthopedic surgery. The dynamic balance between bone formation and bone resorption is destroyed due to excessive osteoclast fusion and differentiation, which results in severe bone matrix loss. Many therapeutic approaches that restrain osteoclast formation and function act as efficient ways to prevent inflammatory bone erosion. We have demonstrated for the first time that dendritic cells-derived interferon-λ1 (IFN-λ1) inhibited inflammatory bone destruction in vivo and explored its underlying mechanisms on osteoclast formation in vitro. We found that IFN-λ1 was highly expressed in infectious bone tissue compared with that of non-infectious bone tissue. Additionally, dendritic cells marker genes such as CD80, CD86, and CD1a were higher expressed in infectious bone tissue than that of non-infectious bone tissue. Dendritic cells that were pretreated with LPS showed high expression of IFN-λ1. Moreover, conditioned medium of LPS-pretreated dendritic cells significantly inhibited osteoclast differentiation, as determined by TRAP staining assay. This suppressive effect was reversed by adding an IFN-λ1 monoclonal antibody. It was also investigated whether exogenous IFN-λ1 restrained osteoclastogenesis, bone resorption, F-actin ring formation, osteoclast-specific gene expression, release of pro-inflammatory cytokines, and translocation of p65 and NFATc1 by preventing the NF-κB signaling pathway and NLRP3 inflammasome formation, as well as by inducing the JAK-STAT signaling pathways in vitro. In vivo study indicated that IFN-λ1 prevents lipopolysaccharide (LPS)-induced inflammatory bone destruction by inhibiting excessive osteoclast fusion and bone resorption activity. In conclusion, our findings confirmed that dendritic cells-derived IFN-λ1 could attenuate osteoclast formation and bone resorptive activity in vitro and in vivo. These novel findings pave the way for the use of exogenous IFN-λ1 as a potential therapeutic treatment for excessive osteoclast-related diseases, such as inflammatory osteolysis, by regulating osteoclastogenesis to maintain the dynamic balance between bone formation and bone resorption.
Collapse
Affiliation(s)
- Yueqi Chen
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yiran Wang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Ruohui Tang
- Department of Emergency, Daping Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Jing Yang
- Department of Emergency, Daping Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Ce Dou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yutong Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Dong Sun
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Chengmin Zhang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Lincheng Zhang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yong Tang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Qijie Dai
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Fei Luo
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| | - Shiwu Dong
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| |
Collapse
|
125
|
Yoo HJ, Hwang WC, Min DS. Targeting of Phospholipase D1 Ameliorates Collagen-Induced Arthritis via Modulation of Treg and Th17 Cell Imbalance and Suppression of Osteoclastogenesis. Int J Mol Sci 2020; 21:ijms21093230. [PMID: 32370217 PMCID: PMC7247592 DOI: 10.3390/ijms21093230] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
Phospholipase D1 (PLD1) plays a crucial role in various inflammatory and autoimmune diseases. Rheumatoid arthritis (RA) is a chronic and systemic autoimmune disease. However, the role of PLD1 in the pathogenesis of RA remains unknown. Here, we first investigated the role and effects of PLD1 in collagen-induced arthritis (CIA) and found that genetic and pharmacological inhibition of PLD1 in DBA1/J mice with CIA reduced the incidence of CIA, decreased the clinical score, and abrogated disease symptoms including infiltration of leukocytes, synovial inflammation, bone erosion, and cartilage destruction. Moreover, ablation and inhibition of PLD1 suppressed the production of type II collagen-specific IgG2a autoantibody and proinflammatory cytokines, accompanied by an increase in the regulatory T (Treg) cell population and a decrease in the Th17 cell population in CIA mice. The PLD1 inhibitor also promoted differentiation of Treg cells and suppressed differentiation of Th17 cells in vitro. Furthermore, the PLD1 inhibitor attenuated pathologic bone destruction in CIA mice by suppressing osteoclastogenesis and bone resorption. Thus, our findings indicate that the targeting of PLD1 can ameliorate CIA by modulating the imbalance of Treg and Th17 cells and suppressing osteoclastogenesis, which might be a novel strategy to treat autoimmune diseases, such as RA.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/prevention & control
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/prevention & control
- Benzimidazoles/pharmacology
- Cell Differentiation/drug effects
- Cell Differentiation/immunology
- Cytokines/blood
- Disease Models, Animal
- Knee Joint/drug effects
- Knee Joint/metabolism
- Knee Joint/pathology
- Male
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Osteogenesis/drug effects
- Osteogenesis/genetics
- Phospholipase D/antagonists & inhibitors
- Phospholipase D/genetics
- Phospholipase D/metabolism
- Piperidines/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
- X-Ray Microtomography
Collapse
Affiliation(s)
- Hyun Jung Yoo
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46241, Korea; (H.J.Y.); (W.C.H.)
| | - Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46241, Korea; (H.J.Y.); (W.C.H.)
- College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Do Sik Min
- College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Correspondence: ; Tel.: +82-32-749-4522
| |
Collapse
|
126
|
Yang X, Chen S, Liu X, Yu M, Liu X. Drug Delivery Based on Nanotechnology for Target Bone Disease. Curr Drug Deliv 2020; 16:782-792. [PMID: 31530265 DOI: 10.2174/1567201816666190917123948] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/03/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023]
Abstract
Bone diseases are a serious problem in modern human life. With the coming acceleration of global population ageing, this problem will become more and more serious. Due to the specific physiological characteristics and local microenvironment of bone tissue, it is difficult to deliver drugs to the lesion site. Therefore, the traditional orthopedic medicine scheme has the disadvantages of high drug frequency, large dose and relatively strong side effects. How to target deliver drugs to the bone tissue or even target cells is the focus of the development of new drugs. Nano drug delivery system with a targeting group can realize precise delivery of orthopedic drugs and effectively reduce the systemic toxicity. In addition, the application of bone tissue engineering scaffolds and biomedical materials to realize in situ drug delivery also are research hotspot. In this article, we briefly review the application of nanotechnology in targeted therapies for bone diseases.
Collapse
Affiliation(s)
- Xiaosong Yang
- Orthopedic Department, Peking University Third Hospital, Beijing 100191, China
| | - Shizhu Chen
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Xiao Liu
- Orthopedic Department, Peking University Third Hospital, Beijing 100191, China
| | - Miao Yu
- Orthopedic Department, Peking University Third Hospital, Beijing 100191, China
| | - Xiaoguang Liu
- Orthopedic Department, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
127
|
Egawa S, Hirai K, Matsumoto R, Yoshii T, Yuasa M, Okawa A, Sugo K, Sotome S. Efficacy of Antibiotic-Loaded Hydroxyapatite/Collagen Composites Is Dependent on Adsorbability for Treating Staphylococcus aureus Osteomyelitis in Rats. J Orthop Res 2020; 38:843-851. [PMID: 31691335 PMCID: PMC7155147 DOI: 10.1002/jor.24507] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 10/25/2019] [Indexed: 02/04/2023]
Abstract
Osteomyelitis remains one of the most challenging disorders for orthopedic doctors despite the advancement of therapeutic techniques. The purpose of this study was to investigate the feasibility of local antibiotic administration using hydroxyapatite/collagen (HAp/Col) as a drug delivery system. We hypothesized that higher adsorbability of antibiotics onto HAp/Col will result in more efficacious activity and therefore, treatment of osteomyelitis. Eight antibiotics were examined in this study: amikacin, cefazolin, cefotiam, daptomycin, minocycline, piperacillin, teicoplanin, and vancomycin. Aligning with their adsorbability onto HAp/Col, minocycline, teicoplanin, and vancomycin showed antibacterial effects up to 14 days after subcutaneous implantation in Wistar rats; while antibiotics with reduced adsorbability (cefazolin, cefotiam, piperacillin) had diminished antibacterial effects. Furthermore, when implanted into a rat femur, vancomycin levels from the Hap/Col were detected in the medullary space above the minimum inhibitory concentration for Staphylococcus aureus for 7 days, while cefazolin levels were undetectable. Aligning with these results, implantation of Hap/Col impregnated with vancomycin to the femur in an acute osteomyelitis rat model had a greater therapeutic effect than cefazolin, as measured by the number of bacteria, the extent of bone destruction, and bone regeneration. These results indicated that the adsorbability of antibiotics onto their carrier is important when locally administered and that HAp/Col scaffolds might be a useful antibiotic delivery system for osteomyelitis. © 2019 The Authors. Journal of Orthopaedic Research® published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society J Orthop Res 38:843-851, 2020.
Collapse
Affiliation(s)
- Satoru Egawa
- Department of Orthopaedic and Spinal Surgery, Graduate SchoolTokyo Medical and Dental University1‐5‐45 Yushima, Bunkyo‐kuTokyo113‐8519Japan
| | - Keigo Hirai
- Department of Orthopaedic and Spinal Surgery, Graduate SchoolTokyo Medical and Dental University1‐5‐45 Yushima, Bunkyo‐kuTokyo113‐8519Japan
| | - Rempei Matsumoto
- Department of Orthopaedic and Spinal Surgery, Graduate SchoolTokyo Medical and Dental University1‐5‐45 Yushima, Bunkyo‐kuTokyo113‐8519Japan
| | - Toshitaka Yoshii
- Department of Orthopaedic and Spinal Surgery, Graduate SchoolTokyo Medical and Dental University1‐5‐45 Yushima, Bunkyo‐kuTokyo113‐8519Japan
| | - Masato Yuasa
- Department of Orthopaedic and Spinal Surgery, Graduate SchoolTokyo Medical and Dental University1‐5‐45 Yushima, Bunkyo‐kuTokyo113‐8519Japan
| | - Atsushi Okawa
- Department of Orthopaedic and Spinal Surgery, Graduate SchoolTokyo Medical and Dental University1‐5‐45 Yushima, Bunkyo‐kuTokyo113‐8519Japan
| | - Ken Sugo
- HOYA Technosurgical Corporation1‐1‐110, Tsutsujigaoka Akishima‐shiTokyo196‐0012Japan
| | - Shinichi Sotome
- Department of Orthopaedic Research and Development, Graduate SchoolTokyo, Medical and Dental University1‐5‐45 Yushima, Bunkyo‐kuTokyo113‐8519Japan
| |
Collapse
|
128
|
Iyer S, Melendez-Suchi C, Han L, Baldini G, Almeida M, Jilka RL. Elevation of the unfolded protein response increases RANKL expression. FASEB Bioadv 2020; 2:207-218. [PMID: 32259048 PMCID: PMC7133738 DOI: 10.1096/fba.2019-00032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/12/2019] [Accepted: 01/17/2020] [Indexed: 02/06/2023] Open
Abstract
Increased production of the osteoclastogenic cytokine RANKL is a common feature of pathologic bone loss, but the underlying cause of this increase is poorly understood. The unfolded protein response (UPR) is activated in response to accumulation of misfolded proteins in the endoplasmic reticulum (ER). Failure to resolve misfolding results in excess UPR signaling that stimulates cytokine production and cell death. We therefore investigated whether RANKL is one of the cytokines stimulated in response to elevated UPR in bone cells. Pharmacologic induction of UPR with tunicamycin (Tm)-stimulated RANKL expression in cultures of primary osteoblastic cells and in osteoblast and osteocyte cell lines. Pharmacologic inhibition of the UPR blunted Tm-induced RANKL production. Silencing Edem1 or Sel1l, proteins that aid in degradation of misfolded proteins, also induced UPR and increased RANKL mRNA. Moreover, Tm or hypoxia increased RANKL and bone resorption in cultures of neonatal murine calvaria. Administration of Tm to adult mice caused dilation of ER in osteoblasts and osteocytes, elevated the UPR, and increased RANKL expression and osteoclast number. These findings support the hypothesis that excessive UPR signaling stimulates the expression of RANKL by osteoblasts and osteocytes, and thereby facilitates excessive bone resorption and bone loss in pathologic conditions.
Collapse
Affiliation(s)
- Srividhya Iyer
- Department of Orthopaedic Surgery University of Arkansas Medical Sciences Little Rock AR USA
| | | | - Li Han
- Division of Endocrinology and Metabolism Center for Osteoporosis and Metabolic Bone Diseases University of Arkansas Medical Sciences Little Rock AR USA
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology University of Arkansas Medical Sciences Little Rock AR USA
| | - Maria Almeida
- Department of Orthopaedic Surgery University of Arkansas Medical Sciences Little Rock AR USA
- Division of Endocrinology and Metabolism Center for Osteoporosis and Metabolic Bone Diseases University of Arkansas Medical Sciences Little Rock AR USA
| | - Robert L Jilka
- Division of Endocrinology and Metabolism Center for Osteoporosis and Metabolic Bone Diseases University of Arkansas Medical Sciences Little Rock AR USA
- Central Arkansas Veterans Healthcare System Little Rock AR USA
| |
Collapse
|
129
|
Kamiya Y, Kikuchi T, Goto H, Okabe I, Takayanagi Y, Suzuki Y, Sawada N, Okabe T, Suzuki Y, Kondo S, Hayashi JI, Mitani A. IL-35 and RANKL Synergistically Induce Osteoclastogenesis in RAW264 Mouse Monocytic Cells. Int J Mol Sci 2020; 21:ijms21062069. [PMID: 32197293 PMCID: PMC7139320 DOI: 10.3390/ijms21062069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 02/07/2023] Open
Abstract
Interleukin (IL)-35 is an immunosuppressive cytokine mainly produced by regulatory T cells. IL-35 mediates immunological functions by suppressing the inflammatory immune response. However, the role of IL-35 in bone-destructive diseases remains unclear, especially in terms of osteoclastogenesis. Therefore, the current study investigated the synergistic effect of IL-35 on osteoclastogenesis that is involved the pathogeneses of periodontitis and rheumatoid arthritis. Osteoclastic differentiation and osteoclastogenesis of RAW264 (RAW) cells induced by receptor activator of nuclear factor (NF)-κB ligand (RANKL) and IL-35 were evaluated by tartrate-resistant acid phosphate staining, hydroxyapatite resorption assays, and quantitative polymerase chain reaction. The effect of IL-35 on RANKL-stimulated signaling pathways was assessed by Western blot analysis. Costimulation of RAW cells by RANKL and IL-35 induced osteoclastogenesis significantly compared with stimulation by RANKL alone. Phosphorylations of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase tended to be increased by RANKL and IL-35 compared with RANKL or IL-35 alone. Additionally, the osteoclastogenesis induced by RANKL and IL-35 was suppressed by inhibition of ERK. In this study, IL-35 and RANKL induced osteoclastogenesis synergistically. Previous reports have shown that IL-35 suppresses the differentiation of osteoclasts. Therefore, IL-35 might play dual roles of destruction and protection in osteoclastogenesis.
Collapse
|
130
|
Relucenti M, Miglietta S, Bove G, Donfrancesco O, Battaglione E, Familiari P, Barbaranelli C, Covelli E, Barbara M, Familiari G. SEM BSE 3D Image Analysis of Human Incus Bone Affected by Cholesteatoma Ascribes to Osteoclasts the Bone Erosion and VpSEM dEDX Analysis Reveals New Bone Formation. SCANNING 2020; 2020:9371516. [PMID: 32158510 PMCID: PMC7048945 DOI: 10.1155/2020/9371516] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 01/22/2020] [Indexed: 06/10/2023]
Abstract
Bone erosion is considered a typical characteristic of advanced or complicated cholesteatoma (CHO), although it is still a matter of debate if bone erosion is due to osteoclast action, being the specific literature controversial. The purpose of this study was to apply a novel scanning characterization approach, the BSE 3D image analysis, to study the pathological erosion on the surface of human incus bone involved by CHO, in order to definitely assess the eventual osteoclastic resorptive action. To do this, a comparison of BSE 3D image of resorption lacunae (resorption pits) from osteoporotic human femur neck (indubitably of osteoclastic origin) with that of the incus was performed. Surface parameters (area, mean depth, and volume) were calculated by the software Hitachi MountainsMap© from BSE 3D-reconstructed images; results were then statistically analyzed by SPSS statistical software. Our findings showed that no significant differences exist between the two groups. This quantitative approach implements the morphological characterization, allowing us to state that surface erosion of the incus is due to osteoclast action. Moreover, our observation and processing image workflow are the first in the literature showing the presence not only of bone erosion but also of matrix vesicles releasing their content on collagen bundles and self-immuring osteocytes, all markers of new bone formation on incus bone surface. On the basis of recent literature, it has been hypothesized that inflammatory environment induced by CHO may trigger the osteoclast activity, eliciting bone erosion. The observed new bone formation probably takes place at a slower rate in respect to the normal bone turnover, and the process is uncoupled (as recently demonstrated for several inflammatory diseases that promote bone loss) thus resulting in an overall bone loss. Novel scanning characterization approaches used in this study allowed for the first time the 3D imaging of incus bone erosion and its quantitative measurement, opening a new era of quantitative SEM morphology.
Collapse
Affiliation(s)
- Michela Relucenti
- Department SAIMLAL Section of Human Anatomy, Laboratory of Electron Microscopy “Pietro M. Motta”, Sapienza University of Rome, Via Alfonso Borelli 50, 00161 Rome, Italy
| | - Selenia Miglietta
- Department SAIMLAL Section of Human Anatomy, Laboratory of Electron Microscopy “Pietro M. Motta”, Sapienza University of Rome, Via Alfonso Borelli 50, 00161 Rome, Italy
| | - Gabriele Bove
- Department SAIMLAL Section of Human Anatomy, Laboratory of Electron Microscopy “Pietro M. Motta”, Sapienza University of Rome, Via Alfonso Borelli 50, 00161 Rome, Italy
| | - Orlando Donfrancesco
- Department SAIMLAL Section of Human Anatomy, Laboratory of Electron Microscopy “Pietro M. Motta”, Sapienza University of Rome, Via Alfonso Borelli 50, 00161 Rome, Italy
| | - Ezio Battaglione
- Department SAIMLAL Section of Human Anatomy, Laboratory of Electron Microscopy “Pietro M. Motta”, Sapienza University of Rome, Via Alfonso Borelli 50, 00161 Rome, Italy
| | - Pietro Familiari
- Department NESMOS, Neurosurgery Unit, Sapienza University of Rome, Via di Grottarossa 1039, 00189 Rome, Italy
| | - Claudio Barbaranelli
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
| | - Edoardo Covelli
- Department NESMOS, ENT Unit, Sapienza University of Rome, Via di Grottarossa 1039, 00189 Rome, Italy
| | - Maurizio Barbara
- Department NESMOS, ENT Unit, Sapienza University of Rome, Via di Grottarossa 1039, 00189 Rome, Italy
| | - Giuseppe Familiari
- Department SAIMLAL Section of Human Anatomy, Laboratory of Electron Microscopy “Pietro M. Motta”, Sapienza University of Rome, Via Alfonso Borelli 50, 00161 Rome, Italy
| |
Collapse
|
131
|
Song M, Lin X, Zhao J, Wang X, Jiao H, Li H, Sun S, Lin H. High frequency vaccination-induced immune stress reduces bone strength with the involvement of activated osteoclastogenesis in layer pullets. Poult Sci 2020; 99:734-743. [PMID: 32029158 PMCID: PMC7587667 DOI: 10.1016/j.psj.2019.12.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/02/2019] [Indexed: 12/29/2022] Open
Abstract
In poultry production, vaccination is an effective measure to protect chickens from diseases. Vaccination, however, is a stressor that may induce stress responses that interfere with the growth and development of chickens. The interaction between the skeletal and immune systems on bone quality has gained more attention. In the present study, the influence of high frequency vaccinations on the bone development of layer pullets was investigated. Thirty 35-day-old SPF White Leghorn layer pullets were obtained and randomly subjected to the following treatments: vaccinated against Newcastle disease (ND) with LoSota vaccine once at 35-day-old (V1, control); 4 times at 35, 49, 63, and 77 d of age (V4); and 7 times at 35, 42, 49, 56, 63, 70, and 77 d of age (V7). The body weight and organ index of the spleen, thymus, and tibia were recorded. The antibody titer and serum and the tibia calcium and phosphorus concentrations were measured. The transcription levels of the IL-6, IL-17, TNF-α, receptor activator of NF-κB ligand (RANKL), and osteoprotegerin (OPG) genes were determined in spleen, thymus, and the tibia. The results showed that V7 decreased body weight and increased the ND antibody titer, compared to V1-chickens. The expression levels of IL-6, IL-17, and TNF-α were upregulated in spleen, thymus, and the tibia of V7 chickens. In the tibia, RANKL was upregulated, while OPG was downregulated by V7 treatment. The results indicate that high frequency vaccination induces immune stress and impairs bone development. The results suggest that the augmented cytokine expression in immune organs and the tibia is associated with activation of the OPG/RANKL pathway, which, in turn, enhances osteoclastogenesis. The appropriate frequency of vaccination should support optimal bone development and full immunoprotection in layer pullets.
Collapse
Affiliation(s)
- Mengze Song
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, P. R. China 271018
| | - Xiaoyan Lin
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, P. R. China 271018
| | - Jingpeng Zhao
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, P. R. China 271018
| | - Xiaojuan Wang
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, P. R. China 271018
| | - Hongchao Jiao
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, P. R. China 271018
| | - Haifang Li
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, P. R. China 271018
| | - Shuhong Sun
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, P. R. China 271018
| | - Hai Lin
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, P. R. China 271018.
| |
Collapse
|
132
|
Liu L, Zhou M, Zhu R, Zhou J, Ni L, Wang Z, Liu N, Zhu F, Shi T, Deng Z, Wang Y, Tian Y, Li R, Yang H, Wang Z, Jiang J, Xu Y. Hydrogen sulfide protects against particle-induced inflammatory response and osteolysis via SIRT1 pathway in prosthesis loosening. FASEB J 2020; 34:3743-3754. [PMID: 31943384 DOI: 10.1096/fj.201900393rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 11/16/2019] [Accepted: 12/19/2019] [Indexed: 01/01/2023]
Abstract
Wear debris-induced osteolysis and ensuing aseptic loosening is the main cause of implant failure and revision surgery. Wear debris-induced inflammatory response plays key roles in peri-implant osteolysis. Recently, substantial of evidence suggests that hydrogen sulfide (H2 S), the third gasotransmitter, is a critical player regulating inflammation. However, the role and therapeutic potential of H2 S in wear debris-induced inflammation and osteolysis remains to be defined. In the present study, we investigated the effect of H2 S on wear debris-induced pro-inflammatory cytokines expression and osteolysis in vitro and in vivo. With a slow-releasing H2 S donor GYY4137, our study demonstrated that H2 S attenuated wear debris-induced osteolysis and osteoclastogenesis in murine calvaria resorption models. The expression of tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) that stimulated by wear particles were significantly reduced by GYY4137. Further, the level of sirtuin 1 (SIRT1), which possesses anti-inflammation property, was examined in vivo and in macrophages. And we found that wear debris decreased the expression of SIRT1. Cotreated macrophages with GYY4137 in part reversed the decline of SIRT1. More importantly, with the SIRT1 recombinant lentivirus and small interfering RNAs (siRNA) against SIRT1, our data indicated that SIRT1 mediated the inhibitory effects of GYY4137 on wear debris-induced inflammation. Collectively, these results suggested that exogenous H2 S production (via H2 S donors) may represent a potential approach for the treatment of wear particle-induced osteolysis.
Collapse
Affiliation(s)
- Lei Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China.,Department of Orthopedics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China.,The Affiliated Yixing Clinical School of Medical School of Yangzhou University, Yixing, China
| | - Ming Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China.,Department of Orthopedics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Ruofu Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Jun Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Li Ni
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Zhidong Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Naicheng Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Feng Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow, Soochow, China
| | - Zhantao Deng
- Division of Orthopedics Surgery, Department of Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yong Wang
- Department of Orthopedics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Yixing Tian
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Rongqun Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Zhenheng Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Jiannong Jiang
- Department of Orthopedics, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China.,The Affiliated Yixing Clinical School of Medical School of Yangzhou University, Yixing, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow, China
| |
Collapse
|
133
|
Maximova N, Zennaro F, Gregori M, Boz G, Zanon D, Mbalaviele G. Hematopoietic stem cell transplantation-induced bone remodeling in autosomal recessive osteopetrosis: Interaction between skeleton and hematopoietic and sensory nervous systems. Bone 2020; 130:115144. [PMID: 31706050 DOI: 10.1016/j.bone.2019.115144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Autosomal recessive osteopetrosis (ARO) is a rare congenital disorder of defective bone resorption. The inability of osteoclasts to resorb bone compromises the development of bone marrow cavity, and ultimately, leads to defective hematopoiesis and death within the first decade. The only curative treatment currently available for certain forms of ARO is hematopoietic stem cell transplantation (HSCT). Infants over ten months of age suffering from ARO are defined as patients with advanced disease; HSCT to these patients is associated with high risk of transplant-related mortality (TRM). Because of the extreme variability of ARO clinical phenotypes, the most reliable predictive factor of TRM and graft failure risk is the residual bone marrow space volume. CASE REPORT We report clinical and radiological outcomes of one patient affected by ARO and treated with HSCT at advance stage of the disease. We describe the anomalies in various tissues, including bone marrow and bones at the moment of the diagnosis and document their gradual disappearance after HSCT until their complete resolution based on magnetic resonance imaging (MRI) observations. We provided radiological images of the cranial vault bone structure modifications, correlating the radiological appearance of the optical canals and nerves and of the cerebellum with the neurological manifestations of the disease. CONCLUSIONS Our results demonstrate that MRI is a highly sensitive technique that provides excellent images of bone marrow space before and after HSCT without exposing children to ionizing radiation. MRI also permits us to evaluate post-transplant skeletal remodeling and the deriving changes in the hematopoietic and sensory system.
Collapse
Affiliation(s)
- Natalia Maximova
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, via dell'Istria 65/1, 34137 Trieste, Italy.
| | - Floriana Zennaro
- Hôpitaux Pédiatriques de Nice, CHU Lenval, 57 Avenue de la Californie, 06200 Nice, France
| | - Massimo Gregori
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, via dell'Istria 65/1, 34137 Trieste, Italy.
| | - Giulia Boz
- University of Cagliari, Cittadella Universitaria di Monserrato, S. P. Monserrato Sestu Km 0.700 CA, 09042 Monserrato, Cagliari, Italy
| | - Davide Zanon
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, via dell'Istria 65/1, 34137 Trieste, Italy.
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave, CB 8301, St. Louis, MO 63110, USA.
| |
Collapse
|
134
|
Characterization and immunogenicity of bone marrow-derived mesenchymal stem cells under osteoporotic conditions. SCIENCE CHINA-LIFE SCIENCES 2019; 63:429-442. [PMID: 31879847 DOI: 10.1007/s11427-019-1555-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 09/28/2019] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are characterized by their multilineage potential and low immunogenicity. However, the properties of MSCs under pathological conditions are unclear. The current study investigated the differentiation potential and immunological characteristics of bone marrow-derived MSCs from ovariectomized-osteoporotic rats (OP-BMSCs). Although the expression of cell morphology- and stemness-related surface markers was similar between OP-BMSCs and BMSCs from healthy rats (H-BMSCs), the proliferation rate was significantly decreased compared with that of H-BMSCs. Regarding multilineage potential, osteogenesis and chondrogenesis abilities of OP-BMSCs decreased, but the adipogenesis ability was significantly enhanced compared with that of H-BMSCs. As expected, decreased osteogenesis following osteogenic induction resulted in reduced expression of β-catenin, osteocalcin, and runt-related transcription factor 2 in OP-BMSCs. Remarkably, the expression of the co-stimulatory proteins CD40 and CD80 was significantly higher, whereas the expression of the negative co-stimulatory molecule programmed cell death ligand 1 was significantly lower in the OP-BMSCs than that in H-BMSCs. Consequently, H-BMSCs inhibited the proliferation and secretion of inflammatory cytokines from anti-CD3 antibody-activated T cells, whereas OP-BMSCs did not. These results indicate that decreased osteogenesis and increased immunogenicity of OP-BMSCs contribute to bone loss in osteoporosis.
Collapse
|
135
|
Hasegawa T, Kikuta J, Sudo T, Matsuura Y, Matsui T, Simmons S, Ebina K, Hirao M, Okuzaki D, Yoshida Y, Hirao A, Kalinichenko VV, Yamaoka K, Takeuchi T, Ishii M. Identification of a novel arthritis-associated osteoclast precursor macrophage regulated by FoxM1. Nat Immunol 2019; 20:1631-1643. [DOI: 10.1038/s41590-019-0526-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 09/24/2019] [Indexed: 12/12/2022]
|
136
|
Medhat D, Rodríguez CI, Infante A. Immunomodulatory Effects of MSCs in Bone Healing. Int J Mol Sci 2019; 20:ijms20215467. [PMID: 31684035 PMCID: PMC6862454 DOI: 10.3390/ijms20215467] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are capable of differentiating into multilineage cells, thus making them a significant prospect as a cell source for regenerative therapy; however, the differentiation capacity of MSCs into osteoblasts seems to not be the main mechanism responsible for the benefits associated with human mesenchymal stem cells hMSCs when used in cell therapy approaches. The process of bone fracture restoration starts with an instant inflammatory reaction, as the innate immune system responds with cytokines that enhance and activate many cell types, including MSCs, at the site of the injury. In this review, we address the influence of MSCs on the immune system in fracture repair and osteogenesis. This paradigm offers a means of distinguishing target bone diseases to be treated with MSC therapy to enhance bone repair by targeting the crosstalk between MSCs and the immune system.
Collapse
Affiliation(s)
- Dalia Medhat
- Medical Biochemistry Department, National Research Centre, Dokki, Giza 12622, Egypt.
| | - Clara I Rodríguez
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Bizkaia, Spain.
| | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Bizkaia, Spain.
| |
Collapse
|
137
|
Wang C, Mbalaviele G. Role of APD-Ribosylation in Bone Health and Disease. Cells 2019; 8:cells8101201. [PMID: 31590342 PMCID: PMC6829334 DOI: 10.3390/cells8101201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022] Open
Abstract
The transfer of adenosine diphosphate (ADP)-ribose unit(s) from nicotinamide adenine dinucleotide (NAD+) to acceptor proteins is known as ADP-ribosylation. This post-translational modification (PTM) unavoidably alters protein functions and signaling networks, thereby impacting cell behaviors and tissue outcomes. As a ubiquitous mechanism, ADP-ribosylation affects multiple tissues, including bones, as abnormal ADP-ribosylation compromises bone development and remodeling. In this review, we describe the effects of ADP-ribosylation in bone development and maintenance, and highlight the underlying mechanisms.
Collapse
Affiliation(s)
- Chun Wang
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
138
|
Raynaud-Messina B, Verollet C, Maridonneau-Parini I. The osteoclast, a target cell for microorganisms. Bone 2019; 127:315-323. [PMID: 31233933 DOI: 10.1016/j.bone.2019.06.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/11/2019] [Accepted: 06/21/2019] [Indexed: 02/02/2023]
Abstract
Bone is a highly adaptive tissue with regenerative properties that is subject to numerous diseases. Infection is one of the causes of altered bone homeostasis. Bone infection happens subsequently to bone surgery or to systemic spreading of microorganisms. In addition to osteoblasts, osteoclasts (OCs) also constitute cell targets for pathogens. OCs are multinucleated cells that have the exclusive ability to resorb bone mineral tissue. However, the OC is much more than a bone eater. Beyond its role in the control of bone turnover, the OC is an immune cell that produces and senses inflammatory cytokines, ingests microorganisms and presents antigens. Today, increasing evidence shows that several pathogens use OC as a host cell to grow, generating debilitating bone defects. In this review, we exhaustively inventory the bacteria and viruses that infect OC and report the present knowledge in this topic. We point out that most of the microorganisms enhance the bone resorption activity of OC. We notice that pathogen interactions with the OC require further investigation, in particular to validate the OC as a host cell in vivo and to identify the cellular mechanisms involved in altered bone resorption. Thus, we conclude that the OC is a new cell target for pathogens; this new research area paves the way for new therapeutic strategies in the infections causing bone defects.
Collapse
Affiliation(s)
- Brigitte Raynaud-Messina
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina
| | - Christel Verollet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina.
| |
Collapse
|
139
|
Alippe Y, Mbalaviele G. Omnipresence of inflammasome activities in inflammatory bone diseases. Semin Immunopathol 2019; 41:607-618. [PMID: 31520179 PMCID: PMC6814643 DOI: 10.1007/s00281-019-00753-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/29/2019] [Indexed: 12/17/2022]
Abstract
The inflammasomes are intracellular protein complexes that are assembled in response to a variety of perturbations including infections and injuries. Failure of the inflammasomes to rapidly clear the insults or restore tissue homeostasis can result in chronic inflammation. Recurring inflammation is also provoked by mutations that cause the constitutive assembly of the components of these protein platforms. Evidence suggests that chronic inflammation is a shared mechanism in bone loss associated with aging, dysregulated metabolism, autoinflammatory, and autoimmune diseases. Mechanistically, inflammatory mediators promote bone resorption while suppressing bone formation, an imbalance which over time leads to bone loss and increased fracture risk. Thus, while acute inflammation is important for the maintenance of bone integrity, its chronic state damages this tissue. In this review, we discuss the role of the inflammasomes in inflammation-induced osteolysis.
Collapse
Affiliation(s)
- Yael Alippe
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA.
| |
Collapse
|
140
|
Jeong HM, Kim DJ. Bone Diseases in Patients with Chronic Liver Disease. Int J Mol Sci 2019; 20:E4270. [PMID: 31480433 PMCID: PMC6747370 DOI: 10.3390/ijms20174270] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 08/25/2019] [Accepted: 08/28/2019] [Indexed: 02/07/2023] Open
Abstract
Osteoporosis is a frequently observed complication in patients with chronic liver disease, particularly liver cirrhosis and cholestatic liver diseases. In addition, osteoporosis is critical in patients receiving a liver transplant. Nevertheless, few studies have evaluated bone diseases in patients with more frequently observed chronic liver disease, such as chronic viral hepatitis, nonalcoholic fatty liver disease and alcoholic liver disease. Osteoporosis is a disease caused by an imbalance in the activities of osteoblasts and osteoclasts. Over the last few decades, many advances have improved our knowledge of the pathogenesis of osteoporosis. Importantly, activated immune cells affect the progression of osteoporosis, and chronic inflammation may exert an additional effect on the existing pathophysiology of osteoporosis. The microbiota of the intestinal tract may also affect the progression of bone loss in patients with chronic liver disease. Recently, studies regarding the effects of chronic inflammation on dysbiosis in bone diseases have been conducted. However, mechanisms underlying osteoporosis in patients with chronic liver disease are complex and precise mechanisms remain unknown. The following special considerations in patients with chronic liver disease are reviewed: bone diseases in patients who underwent a liver transplant, the association between chronic hepatitis B virus infection treatment and bone diseases, the association between sarcopenia and bone diseases in patients with chronic liver disease, and the association between chronic liver disease and avascular necrosis of the hip. Few guidelines are currently available for the management of low bone mineral density or bone diseases in patients with chronic liver disease. Due to increased life expectancy and therapeutic advances in chronic liver disease, the importance of managing osteoporosis and other bone diseases in patients with chronic liver disease is expected to increase. Consequently, specific guidelines need to be established in the near future.
Collapse
Affiliation(s)
- Hae Min Jeong
- Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, Chuncheon, Gangwon-do 24253, Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Gangwon-do 24253, Korea
| | - Dong Joon Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Gangwon-do 24253, Korea.
- Department of Internal Medicine, Hallym University College of Medicine, Seoul 05355, Korea.
| |
Collapse
|
141
|
Dyskova T, Kriegova E, Slobodova Z, Zehnalova S, Kudelka M, Schneiderova P, Fillerova R, Gallo J. Inflammation time-axis in aseptic loosening of total knee arthroplasty: A preliminary study. PLoS One 2019; 14:e0221056. [PMID: 31469844 PMCID: PMC6716666 DOI: 10.1371/journal.pone.0221056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/29/2019] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Aseptic loosening (AL) is the most frequent long-term reason for revision of total knee arthroplasty (TKA) affecting about 15-20% patients within 20 years after the surgery. Although there is a solid body of evidence about the crucial role of inflammation in the AL pathogenesis, scared information on inflammation signature and its time-axis in tissues around TKA exists. DESIGN The inflammation protein signatures in pseudosynovial tissues collected at revision surgery from patients with AL (AL, n = 12) and those with no clinical/radiographic signs of AL (non-AL, n = 9) were investigated by Proximity Extension Assay (PEA)-Immunoassay and immunohistochemistry. RESULTS AL tissues had elevated levels of TNF-family members sTNFR2, TNFSF14, sFasL, sBAFF, cytokines/chemokines IL8, CCL2, IL1RA/IL36, sIL6R, and growth factors sAREG, CSF1, comparing to non-AL. High interindividual variability in protein levels was evident particularly in non-AL. Levels of sTNFR2, sBAFF, IL8, sIL6R, and MPO discriminated between AL and non-AL and were associated with the time from index surgery, suggesting the cumulative character of inflammatory osteolytic response to prosthetic byproducts. The source of elevated inflammatory molecules was macrophages and multinucleated osteoclast-like cells in AL and histiocytes and osteoclast-like cells in non-AL tissues, respectively. All proteins were present in higher levels in osteoclast-like cells than in macrophages. CONCLUSIONS Our study revealed a differential inflammation signature between AL and non-AL stages of TKA. It also highlighted the unique patient's response to TKA in non-AL stages. Further confirmation of our preliminary results on a larger cohort is needed. Analysis of the time-axis of processes ongoing around TKA implantation may help to understand the mechanisms driving periprosthetic bone resorption needed for diagnostic/preventative strategies.
Collapse
Affiliation(s)
- Tereza Dyskova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Eva Kriegova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Zuzana Slobodova
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Sarka Zehnalova
- Department of Computer Science, Faculty of Electrical Engineering and Computer Science, VSB-Technical University of Ostrava, Ostrava, Czech Republic
| | - Milos Kudelka
- Department of Computer Science, Faculty of Electrical Engineering and Computer Science, VSB-Technical University of Ostrava, Ostrava, Czech Republic
| | - Petra Schneiderova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Regina Fillerova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jiri Gallo
- Department of Orthopaedics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| |
Collapse
|
142
|
Arciola CR, Campoccia D, Montanaro L. Implant infections: adhesion, biofilm formation and immune evasion. Nat Rev Microbiol 2019; 16:397-409. [PMID: 29720707 DOI: 10.1038/s41579-018-0019-y] [Citation(s) in RCA: 1145] [Impact Index Per Article: 190.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Medical device-associated infections account for a large proportion of hospital-acquired infections. A variety of opportunistic pathogens can cause implant infections, depending on the type of the implant and on the anatomical site of implantation. The success of these versatile pathogens depends on rapid adhesion to virtually all biomaterial surfaces and survival in the hostile host environment. Biofilm formation on implant surfaces shelters the bacteria and encourages persistence of infection. Furthermore, implant-infecting bacteria can elude innate and adaptive host defences as well as biocides and antibiotic chemotherapies. In this Review, we explore the fundamental pathogenic mechanisms underlying implant infections, highlighting orthopaedic implants and Staphylococcus aureus as a prime example, and discuss innovative targets for preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy. .,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - Davide Campoccia
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Lucio Montanaro
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
143
|
Loin J, Kün-Darbois JD, Guillaume B, Badja S, Libouban H, Chappard D. Maxillary sinus floor elevation using Beta-Tricalcium-Phosphate (beta-TCP) or natural bone: same inflammatory response. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:97. [PMID: 31440845 DOI: 10.1007/s10856-019-6299-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 08/02/2019] [Indexed: 06/10/2023]
Abstract
Sinus elevation is a common procedure to increase bone volume in the atrophic maxilla to allow placement of dental implants. Autogenous bone is the gold standard but is limited in quantity and causes morbidity at the donor site. β-TCP is a synthetic biomaterial commonly used in that purpose. It appears to induce a poor inflammatory response. This study aimed to evaluate the degree of edema of the sinus mucosa after sinus lift surgery according to the type of biomaterial. Forty sinuses (20 patients) were included retrospectively and divided into 2 groups according to the biomaterial that was used: synthetic biomaterial (BTCP group), natural bone (BONE group). A control group (CTRL group) was constituted by the non-grafted maxillary sinuses. Twelve measurements per sinus were realized on pre- and post-operative computed tomography and averaged to provide the sinus membrane thickness value (SM.Th). SM.Th was thicker post-operatively in the BTCP and BONE groups in comparison with the CTRL group and in comparison with pre-operative measurements. No difference was found post operatively between the BTCP and BONE groups. We found that a synthetic biomaterial (β-TCP) induced the same degree of edema, and thus of inflammation, as natural bone. It constitutes therefore an interesting alternative to autogenous bone for maxillary sinus lifts.
Collapse
Affiliation(s)
- Justine Loin
- Service de Chirurgie Maxillo-faciale et stomatologie, CHU d'Angers, 4 rue Larrey, 49933, Angers Cedex, France
| | - Jean-Daniel Kün-Darbois
- Service de Chirurgie Maxillo-faciale et stomatologie, CHU d'Angers, 4 rue Larrey, 49933, Angers Cedex, France.
- Groupe Etudes Remodelage Osseux et bioMatériaux, GEROM, EA 4658, SFR 4208, Univ-Angers, IRIS-IBS Institut de Biologie en Santé, CHU d'Angers, 49933, Angers Cedex, France.
| | - Bernard Guillaume
- Groupe Etudes Remodelage Osseux et bioMatériaux, GEROM, EA 4658, SFR 4208, Univ-Angers, IRIS-IBS Institut de Biologie en Santé, CHU d'Angers, 49933, Angers Cedex, France
- Centre Français d'Implantologie CFI, 6 rue de Rome, 75008, Paris, France
| | - Smail Badja
- Service de Chirurgie Maxillo-faciale et stomatologie, CHU d'Angers, 4 rue Larrey, 49933, Angers Cedex, France
| | - Hélène Libouban
- Groupe Etudes Remodelage Osseux et bioMatériaux, GEROM, EA 4658, SFR 4208, Univ-Angers, IRIS-IBS Institut de Biologie en Santé, CHU d'Angers, 49933, Angers Cedex, France
| | - Daniel Chappard
- Groupe Etudes Remodelage Osseux et bioMatériaux, GEROM, EA 4658, SFR 4208, Univ-Angers, IRIS-IBS Institut de Biologie en Santé, CHU d'Angers, 49933, Angers Cedex, France
| |
Collapse
|
144
|
Aseptic Ligatures Induce Marginal Peri-Implant Bone Loss-An 8-Week Trial in Rabbits. J Clin Med 2019; 8:jcm8081248. [PMID: 31426572 PMCID: PMC6723089 DOI: 10.3390/jcm8081248] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/23/2022] Open
Abstract
The clinical value of ligature-induced experimental peri-implantitis studies has been questioned due to the artificial nature of the model. Despite repeated claims that ligatures of silk, cotton and other materials may not induce bone resorption by themselves; a recent review showed that the tissue reaction toward them has not been investigated. Hence, the current study aimed to explore the hard and soft tissue reactions toward commonly used ligature materials. A total of 60 dental implants were inserted into the femur (n = 20) and tibia (n = 40) of 10 rabbits. The femoral implants were ligated with sterile 3-0 braided silk in one leg and sterile cotton retraction chord in the other leg. The tibial implants were ligated with silk or left as non-ligated controls. All wounds were closed in layers. After a healing time of 8 weeks, femoral (silk versus cotton) and proximal tibial (silk versus non-ligated control) implants were investigated histologically. Distal tibial (silk versus non-ligated control) implants were investigated with real time polymerase chain reaction (qPCR). The distance from the implant-top to first bone contact point was longer for silk ligated implants compared to non-ligated controls (p = 0.007), but did not vary between cotton and silk. The ligatures triggered an immunological reaction with cell infiltrates in close contact with the ligature materials, adjacent soft tissue encapsulation and bone resorption. qPCR further demonstrated an upregulated immune response toward the silk ligatures compared to non-ligated controls. Silk and cotton ligatures provoke foreign body reactions of soft tissue encapsulation type and bone resorption around implants in the absence of plaque.
Collapse
|
145
|
Zaiss MM, Hall C, McGowan NWA, Babb R, Devlia V, Lucas S, Meghji S, Henderson B, Bozec A, Schett G, David JP, Panayi GS, Grigoriadis AE, Corrigall VM. Binding Immunoglobulin Protein (BIP) Inhibits TNF-α-Induced Osteoclast Differentiation and Systemic Bone Loss in an Erosive Arthritis Model. ACR Open Rheumatol 2019; 1:382-393. [PMID: 31777818 PMCID: PMC6857990 DOI: 10.1002/acr2.11060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/11/2019] [Indexed: 12/15/2022] Open
Abstract
Objective The association between inflammation and dysregulated bone remodeling is apparent in rheumatoid arthritis and is recapitulated in the human tumor necrosis factor transgenic (hTNFtg) mouse model. We investigated whether extracellular binding immunoglobulin protein (BiP) would protect the hTNFtg mouse from both inflammatory arthritis as well as extensive systemic bone loss and whether BiP had direct antiosteoclast properties in vitro. Methods hTNFtg mice received a single intraperitoneal administration of BiP at onset of arthritis. Clinical disease parameters were measured weekly. Bone analysis was performed by microcomputed tomography and histomorphometry. Mouse bone marrow macrophage and human peripheral blood monocyte precursors were used to study the direct effect of BiP on osteoclast differentiation and function in vitro. Monocyte and osteoclast signaling was analyzed by Western blotting, flow cytometry, and imaging flow cytometry. Results BiP-treated mice showed reduced inflammation and cartilage destruction, and histomorphometric analysis revealed a decrease in osteoclast number with protection from systemic bone loss. Abrogation of osteoclast function was also observed in an ex vivo murine calvarial model. BiP inhibited differentiation of osteoclast precursors and prevented bone resorption by mature osteoclasts in vitro. BiP also induced downregulation of CD115/c-Fms and Receptor Activator of NF-κB (RANK) messenger RNA and protein, causing reduced phosphorylation of the p38 mitogen-activated protein kinases, extracellular signal-regulated kinases 1/2 and p38, with suppression of essential osteoclast transcription factors, c-Fos and NFATc1. BiP directly inhibited TNF-α- or Receptor Activator of NF-κB Ligand (RANKL)-induced NF-κB nuclear translocation in THP-1 monocytic cells and preosteoclasts by the canonical and noncanonical pathways. Conclusion BiP combines an anti-inflammatory function with antiosteoclast activity, which establishes it as a potential novel therapeutic for inflammatory disorders associated with bone loss.
Collapse
Affiliation(s)
- Mario M Zaiss
- Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen Erlangen Germany
| | | | | | | | | | - Sébastien Lucas
- Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen Erlangen Germany
| | - Sajeda Meghji
- UCL-Eastman Dental Institute University College London London UK
| | - Brian Henderson
- UCL-Eastman Dental Institute University College London London UK
| | - Aline Bozec
- Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen Erlangen Germany
| | - Georg Schett
- Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen Erlangen Germany
| | - Jean-Pierre David
- Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany, and Institute of Osteology and Biomechanics (IOBM) University Medical Center Hamburg-Eppendorf Hamburg Germany
| | | | | | | |
Collapse
|
146
|
Li L, Rao S, Cheng Y, Zhuo X, Deng C, Xu N, Zhang H, Yang L. Microbial osteoporosis: The interplay between the gut microbiota and bones via host metabolism and immunity. Microbiologyopen 2019; 8:e00810. [PMID: 31001921 PMCID: PMC6692530 DOI: 10.1002/mbo3.810] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 01/15/2023] Open
Abstract
The complex relationship between intestinal microbiota and host is a novel field in recent years. A large number of studies are being conducted on the relationship between intestinal microbiota and bone metabolism. Bone metabolism consisted of bone absorption and formation exists in the whole process of human growth and development. The nutrient components, inflammatory factors, and hormone environment play important roles in bone metabolism. Recently, intestinal microbiota has been found to influence bone metabolism via influencing the host metabolism, immune function, and hormone secretion. Here, we searched relevant literature on Pubmed and reviewed the effect of intestinal microbiota on bone metabolism through the three aspects, which may provide new ideas and targets for the clinical treatment of osteoporosis.
Collapse
Affiliation(s)
- Lishan Li
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Shitao Rao
- School of Biomedical SciencesCUHKShatin, N.THong Kong SARChina
| | - Yanzhen Cheng
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xiaoyun Zhuo
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Caihong Deng
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ningning Xu
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Hua Zhang
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Li Yang
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
147
|
Yu J, Canalis E. The Hajdu Cheney mutation sensitizes mice to the osteolytic actions of tumor necrosis factor α. J Biol Chem 2019; 294:14203-14214. [PMID: 31371452 DOI: 10.1074/jbc.ra119.009824] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/29/2019] [Indexed: 11/06/2022] Open
Abstract
Hajdu Cheney syndrome (HCS) is characterized by craniofacial developmental abnormalities, acro-osteolysis, and osteoporosis and is associated with gain-of-NOTCH2 function mutations. A mouse model of HCS termed Notch2tm1.1Ecan harboring a mutation in exon 34 of Notch2 replicating the one found in HCS was used to determine whether the HCS mutation sensitizes the skeleton to the osteolytic effects of tumor necrosis factor α (TNFα). TNFα injected over the calvarial vault caused a greater increase in osteoclast number, osteoclast surface, and eroded surface in Notch2tm1.1Ecan mice compared with littermate WT controls. Accordingly, the effect of TNFα on osteoclastogenesis was greatly enhanced in cultures of bone marrow-derived macrophages (BMMs) from Notch2tm1.1Ecan mice when compared with the activity of TNFα in control cultures. TNFα induced the expression of Notch2 and Notch2 mutant mRNA by ∼2-fold, possibly amplifying the NOTCH2-dependent induction of osteoclastogenesis. The effect of TNFα on osteoclastogenesis in Notch2tm1.1Ecan mutants depended on NOTCH2 activation because it was reversed by anti-NOTCH2 negative regulatory region and anti-jagged 1 antibodies. The inactivation of Hes1 prevented the TNFα effect on osteoclastogenesis in the context of the Notch2tm1.1Ecan mutation. In addition, the induction of Il1b, but not of Tnfa and Il6, mRNA by TNFα was greater in Notch2tm1.1Ecan BMMs than in control cells, possibly contributing to the actions of TNFα and NOTCH2 on osteoclastogenesis. In conclusion, the HCS mutation enhances TNFα-induced osteoclastogenesis and the inflammatory bone-resorptive response possibly explaining the acro-osteolysis observed in affected individuals.
Collapse
Affiliation(s)
- Jungeun Yu
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut 06030.,UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut 06030
| | - Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut 06030 .,UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut 06030.,Department of Medicine, UConn Health, Farmington, Connecticut 06030
| |
Collapse
|
148
|
Hoppstädter J, Ammit AJ. Role of Dual-Specificity Phosphatase 1 in Glucocorticoid-Driven Anti-inflammatory Responses. Front Immunol 2019; 10:1446. [PMID: 31316508 PMCID: PMC6611420 DOI: 10.3389/fimmu.2019.01446] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022] Open
Abstract
Glucocorticoids (GCs) potently inhibit pro-inflammatory responses and are widely used for the treatment of inflammatory diseases, such as allergies, autoimmune disorders, and asthma. Dual-specificity phosphatase 1 (DUSP1), also known as mitogen-activated protein kinase (MAPK) phosphatase-1 (MKP-1), exerts its effects by dephosphorylation of MAPKs, i.e., extracellular-signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK). Endogenous DUSP1 expression is tightly regulated at multiple levels, involving both transcriptional and post-transcriptional mechanisms. DUSP1 has emerged as a central mediator in the resolution of inflammation, and upregulation of DUSP1 by GCs has been suggested to be a key mechanism of GC actions. In this review, we discuss the impact of DUSP1 on the efficacy of GC-mediated suppression of inflammation and address the underlying mechanisms.
Collapse
Affiliation(s)
- Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | - Alaina J Ammit
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia.,Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
149
|
Abu-Amer W, Arra M, Clohisy JCF, Abu-Amer Y, Swarnkar G. Targeting vascular endothelial growth factor ameliorates PMMA-particles induced inflammatory osteolysis in murine calvaria. Bone 2019; 123:86-91. [PMID: 30904629 PMCID: PMC6491226 DOI: 10.1016/j.bone.2019.03.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/07/2019] [Accepted: 03/19/2019] [Indexed: 12/14/2022]
Abstract
Cytokines and growth factors mediate inflammatory osteolysis in response to particles released from bone implants. However, the mechanism by which this process develops is not entirely clear. Blood vessels and related factors may be required to deliver immune cells and soluble factors to the injury site. Therefore, in the current study we investigated if, vascular endothelial growth factor (VEGF), which is required for angiogenesis, mediates polymethylmethacrylate (PMMA) particles-induced osteolysis. Using bone marrow derived macrophages (BMMs) and ST2 stromal cell line, we show that PMMA particles increase VEGF expression. Further, using a murine calvarial osteolysis model, we found that PMMA injection over calvaria induce significant increase in VEGF expression as well as new vessel formation, represented by von Willebrand factor (vWF) staining. Co-treatment using a VEGF-neutralizing antibody abrogated expression of vWF, indicating decreased angiogenesis. Finally, VEGF neutralizing antibody reduced expression of Tumor necrosis factor (TNF) and decreased osteoclastogenesis induced by PMMA particles in calvariae. This work highlights the significance of angiogenesis, specifically VEGF, as key driver of PMMA particle-induced inflammatory osteolysis, inhibition of which attenuates this response.
Collapse
Affiliation(s)
- Wahid Abu-Amer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, United States of America
| | - Manoj Arra
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, United States of America
| | - John C F Clohisy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, United States of America
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, United States of America; Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110, United States of America; Shriners Hospital for Children, St. Louis, MO 63110, United States of America
| | - Gaurav Swarnkar
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, United States of America.
| |
Collapse
|
150
|
Terkawi MA, Kadoya K, Takahashi D, Tian Y, Hamasaki M, Matsumae G, Alhasan H, Elmorsy S, Uetsuki K, Onodera T, Takahata M, Iwasaki N. Identification of IL-27 as potent regulator of inflammatory osteolysis associated with vitamin E-blended ultra-high molecular weight polyethylene debris of orthopedic implants. Acta Biomater 2019; 89:242-251. [PMID: 30880234 DOI: 10.1016/j.actbio.2019.03.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/11/2019] [Accepted: 03/13/2019] [Indexed: 12/18/2022]
Abstract
Vitamin E-blended ultra-high molecular weight polyethylene (VE-UHMWPE) is a newly introduced material for prosthetic components that has proven a better mechanical performance with lesser adverse cellular responses than conventional polyethylene in experimental animal models. However, the mechanisms by which VE-UHMWPE particles trigger a reduced osteolytic activity are unclear and remain to be investigated. Therefore, the current study aims at exploring a possible anti-osteolytic mechanism associated with VE-UHMWPE particles. Transcriptional profiling and bioinformatic analyses of human macrophages stimulated by VE-UHMWPE particles revealed a distinct transcriptional program from macrophages stimulated with UHMWPE particles. Out of the up-regulated genes, IL-27 was found to be significantly elevated in macrophages cultured with VE-UHMWPE particles as compared to these with UHMWPE particles (p = 0.0084). Furthermore, we studied the potential anti-osteolytic function of IL-27 in osteolysis murine model. Interestingly, administration of recombinant IL-27 onto calvariae significantly alleviated osteolytic lesions triggered by UHMWPE particles (p = 0.0002). Likewise, IL-27 inhibited differentiation of osteoclasts (p = 0.0116) and reduced inflammatory response (p < 0.0001) elicited by conventional UHMWPE particles in vitro. This is the first study demonstrating the involvement of IL-27 in macrophage response to VE-UHMWPE particles and its regulatory role in osteolysis. Our data highlight a novel therapeutic agent for treatment of inflammatory osteolysis induced by polyethylene debris. STATEMENT OF SIGNIFICANCE: Aseptic loosening due to inflammatory osteolysis remains the major cause of arthroplasty failure and represents a substantial economic burden worldwide. Ideal approach to prevent this failure should be directed to minimize inflammatory response triggered by wear particles at the site of implant. Understanding the mechanism by which VE-UHMWPE particles triggers lesser cellular responses and reduced osteolysis as compared to conventional UHMWPE particles may aid in discovery of regulatory factors. In the current study, we reported that IL-27 is a potent regulator of inflammatory osteolysis involved in the reduced biologic activities and osteolytic potentials associated with VE-UHMWPE particles. Initiating the production IL-27 in vivo after total joint arthroplasties might be a novel strategy to prolong the life-spam of implant.
Collapse
Affiliation(s)
- Mohamad Alaa Terkawi
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; Global Institution for Collaborative Research and Education (GI-CoRE), Frontier Research Center for Advanced Material and Life Science Bldg No 2, Hokkaido University, Japan.
| | - Ken Kadoya
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan
| | - Daisuke Takahashi
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan.
| | - Yuan Tian
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan
| | - Masanari Hamasaki
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan
| | - Gen Matsumae
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan
| | - Hend Alhasan
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan
| | - Sameh Elmorsy
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; Department of Orthopedic Surgery, Beni-Suef University, Faculty of Medicine, Mokbel 62511, Beni-Suef, Egypt
| | - Keita Uetsuki
- R&D Center, Teijin Nakashima Medical Co., Ltd., 5322, Haga, Kita-ku, Okayama 701-1221, Japan
| | - Tomohiro Onodera
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; Global Institution for Collaborative Research and Education (GI-CoRE), Frontier Research Center for Advanced Material and Life Science Bldg No 2, Hokkaido University, Japan
| | - Masahiko Takahata
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Hokkaido University, Faculty of Medicine and Graduate School of Medicine, Kita-15, Nish-7, Kita-ku, Sapporo 060-8638, Japan; Global Institution for Collaborative Research and Education (GI-CoRE), Frontier Research Center for Advanced Material and Life Science Bldg No 2, Hokkaido University, Japan
| |
Collapse
|