101
|
Bi H, Chen X, Gao S, Yu X, Xiao J, Zhang B, Liu X, Dai M. Key Triggers of Osteoclast-Related Diseases and Available Strategies for Targeted Therapies: A Review. Front Med (Lausanne) 2017; 4:234. [PMID: 29326938 PMCID: PMC5742334 DOI: 10.3389/fmed.2017.00234] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 12/04/2017] [Indexed: 01/11/2023] Open
Abstract
Osteoclasts, the only cells with bone resorption functions in vivo, maintain the balance of bone metabolism by cooperating with osteoblasts, which are responsible for bone formation. Excessive activity of osteoclasts causes many diseases such as osteoporosis, periprosthetic osteolysis, bone tumors, and Paget's disease. In contrast, osteopetrosis results from osteoclast deficiency. Available strategies for combating over-activated osteoclasts and the subsequently induced diseases can be categorized into three approaches: facilitating osteoclast apoptosis, inhibiting osteoclastogenesis, and impairing bone resorption. Bisphosphonates are representative molecules that function by triggering osteoclast apoptosis. New drugs, such as tumor necrosis factor and receptor activator of nuclear factor kappa-B ligand (RANKL) inhibitors (e.g., denosumab) have been developed for targeting the receptor activator of nuclear factor kappa-B /RANKL/osteoprotegerin system or CSF-1/CSF-1R axis, which play critical roles in osteoclast formation. Furthermore, vacuolar (H+)-ATPase inhibitors, cathepsin K inhibitors, and glucagon-like peptide 2 impair different stages of the bone resorption process. Recently, significant achievements have been made in this field. The aim of this review is to provide an updated summary of the current progress in research involving osteoclast-related diseases and of the development of targeted inhibitors of osteoclast formation.
Collapse
Affiliation(s)
- Haidi Bi
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Xing Chen
- Department of Orthopaedics, The People's Hospital of Changxing County, Huzhou, China
| | - Song Gao
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Xiaolong Yu
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Jun Xiao
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Bin Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Xuqiang Liu
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Min Dai
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| |
Collapse
|
102
|
Lin T, Pajarinen J, Nabeshima A, Lu L, Nathan K, Jämsen E, Yao Z, Goodman SB. Preconditioning of murine mesenchymal stem cells synergistically enhanced immunomodulation and osteogenesis. Stem Cell Res Ther 2017; 8:277. [PMID: 29212557 PMCID: PMC5719931 DOI: 10.1186/s13287-017-0730-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 12/18/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) are capable of immunomodulation and tissue regeneration, highlighting their potential translational application for treating inflammatory bone disorders. MSC-mediated immunomodulation is regulated by proinflammatory cytokines and pathogen-associated molecular patterns such as lipopolysaccharide (LPS). Previous studies showed that MSCs exposed to interferon gamma (IFN-γ) and the proinflammatory cytokine tumor necrosis factor alpha (TNF-α) synergistically suppressed T-cell activation. Methods In the current study, we developed a novel preconditioning strategy for MSCs using LPS plus TNF-α to optimize the immunomodulating ability of MSCs on macrophage polarization. Results Preconditioned MSCs enhanced anti-inflammatory M2 macrophage marker expression (Arginase 1 and CD206) and decreased inflammatory M1 macrophage marker (TNF-α/IL-1Ra) expression using an in-vitro coculture model. Immunomodulation of MSCs on macrophages was significantly increased compared to the combination of IFN-γ plus TNF-α or single treatment controls. Increased osteogenic differentiation including alkaline phosphate activity and matrix mineralization was only observed in the LPS plus TNF-α preconditioned MSCs. Mechanistic studies showed that increased prostaglandin E2 (PGE2) production was associated with enhanced Arginase 1 expression. Selective cyclooxygenase-2 inhibition by Celecoxib decreased PGE2 production and Arginase 1 expression in cocultured macrophages. Conclusions The novel preconditioned MSCs have increased immunomodulation and bone regeneration potential and could be applied to the treatment of inflammatory bone disorders including periprosthetic osteolysis, fracture healing/nonunions, and osteonecrosis. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0730-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA, 94063, USA
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA, 94063, USA
| | - Akira Nabeshima
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA, 94063, USA
| | - Laura Lu
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA, 94063, USA
| | - Karthik Nathan
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA, 94063, USA
| | - Eemeli Jämsen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA, 94063, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA, 94063, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA, 94063, USA. .,Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
103
|
Jo YY, Kweon H, Kim DW, Baek K, Kim MK, Kim SG, Chae WS, Choi JY, Rotaru H. Bone regeneration is associated with the concentration of tumour necrosis factor-α induced by sericin released from a silk mat. Sci Rep 2017; 7:15589. [PMID: 29138464 PMCID: PMC5686134 DOI: 10.1038/s41598-017-15687-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/31/2017] [Indexed: 01/04/2023] Open
Abstract
To understand the osteogenic effect of the middle layer of the silk cocoon, sericin was examined for its cellular effects associated with tumor necrosis factor-α (TNF-α) signaling in this study. The fragmented sericin proteins in the silk mat were evaluated for the TNF-α expression level in murine macrophages. The concentration of protein released from silk mats was higher in the outermost and the innermost layers than in the middle layers, and the protein released from the silk mat was identified as sericin. The level of TNF-α in murine macrophages was dependent on the applied concentration of sericin, and the expression of genes associated with osteogenesis in osteoblast-like cells was dependent on the applied concentration of TNF-α. In animal experiments, silk mats from the middle layers led to a higher regenerated bone volume than silk mats from the innermost layer or the outermost layer. If TNF-α protein was incorporated into the silk mats from the middle layers, bone regeneration was suppressed compared with unloaded silk mats from the middle layers. Accordingly, silk mats from the silk cocoon can be considered to be a fragmented sericin-secreting carrier, and the level of sericin secretion is associated with TNF-α induction and bone regeneration.
Collapse
Affiliation(s)
- You-Young Jo
- Sericultural and Apicultural Division, National Institute of Agricultural Science, RDA, Wanju, 55365, Republic of Korea
| | - HaeYong Kweon
- Sericultural and Apicultural Division, National Institute of Agricultural Science, RDA, Wanju, 55365, Republic of Korea
| | - Dae-Won Kim
- Department of Oral Biochemistry, College of Dentistry, Gangneung-Wonju National University, Gangneung, 28644, Republic of Korea
| | - Kyunghwa Baek
- Department of Oral Pharmacology, College of Dentistry, Gangneung-Wonju National University, Gangneung, 28644, Republic of Korea
| | - Min-Keun Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung, 28644, Republic of Korea
| | - Seong-Gon Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung, 28644, Republic of Korea.
| | - Weon-Sik Chae
- Analysis Research Division, Daegu Center, Korea Basic Science Institute, Daegu, 41566, Republic of Korea
| | - Je-Yong Choi
- School of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, Skeletal Diseases Analysis Center, Korea Mouse Phenotyping Center (KMPC), Kyungpook National University, Daegu, 41944, Korea.
| | - Horatiu Rotaru
- Department of Cranio-Maxillofacial Surgery, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, 400001, Romania
| |
Collapse
|
104
|
Shen G, Ren H, Qiu T, Zhang Z, Zhao W, Yu X, Huang J, Tang J, Liang D, Yao Z, Yang Z, Jiang X. Mammalian target of rapamycin as a therapeutic target in osteoporosis. J Cell Physiol 2017; 233:3929-3944. [PMID: 28834576 DOI: 10.1002/jcp.26161] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/21/2017] [Indexed: 12/19/2022]
Abstract
The mechanistic target of rapamycin (mTOR) plays a key role in sensing and integrating large amounts of environmental cues to regulate organismal growth, homeostasis, and many major cellular processes. Recently, mounting evidences highlight its roles in regulating bone homeostasis, which sheds light on the pathogenesis of osteoporosis. The activation/inhibition of mTOR signaling is reported to positively/negatively regulate bone marrow mesenchymal stem cells (BMSCs)/osteoblasts-mediated bone formation, adipogenic differentiation, osteocytes homeostasis, and osteoclasts-mediated bone resorption, which result in the changes of bone homeostasis, thereby resulting in or protect against osteoporosis. Given the likely importance of mTOR signaling in the pathogenesis of osteoporosis, here we discuss the detailed mechanisms in mTOR machinery and its association with osteoporosis therapy.
Collapse
Affiliation(s)
- Gengyang Shen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Ren
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Qiu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhida Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhua Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiang Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinjing Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Tang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De Liang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhensong Yao
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhidong Yang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaobing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Laboratory Affiliated to National Key Discipline of Orthopaedic and Traumatology of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
105
|
Lin T, Pajarinen J, Nabeshima A, Córdova LA, Loi F, Gibon E, Lu L, Nathan K, Jämsen E, Yao Z, Goodman SB. Orthopaedic wear particle-induced bone loss and exogenous macrophage infiltration is mitigated by local infusion of NF-κB decoy oligodeoxynucleotide. J Biomed Mater Res A 2017; 105:3169-3175. [PMID: 28782280 DOI: 10.1002/jbm.a.36169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/23/2017] [Accepted: 08/01/2017] [Indexed: 01/02/2023]
Abstract
Excessive production of wear particles from total joint replacements induces chronic inflammation, macrophage infiltration, and consequent bone loss (periprosthetic osteolysis). This inflammation and bone remodeling are critically regulated by the transcription factor NF-κB. We previously demonstrated that inhibition of NF-κB signaling by using the decoy oligodeoxynucleotide (ODN) mitigates polyethylene wear particle-induced bone loss using in vitro and in vivo models. However, the mechanisms of NF-κB decoy ODN action, and in particular its impact on systemic macrophage recruitment, remain unknown. In the current study, this systemic macrophage infiltration was examined in our established murine femoral continuous particle infusion model. RAW264.7 murine macrophages expressing a luciferase reporter gene were injected into the systemic circulation. Quantification of bioluminescence showed that NF-κB decoy ODN reduced the homing of these reporter macrophages into the distal femurs exposed to continuous particle delivery. Particle-induced reduction in bone mineral density at the distal diaphysis of the femur was also mitigated by infusion of decoy ODN. Histological staining showed that the decoy ODN infusion decreased osteoclast and macrophage numbers, but had no significant effects on osteoblasts. Local infusion of NF-κB decoy ODN reduced systemic macrophage infiltration and mitigated particle-induced bone loss, thus providing a potential strategy to treat periprosthetic osteolysis. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 3169-3175, 2017.
Collapse
Affiliation(s)
- Tzuhua Lin
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Jukka Pajarinen
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Akira Nabeshima
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Luis A Córdova
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California.,Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Chile-CONICYT, Santiago, Chile
| | - Florence Loi
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Emmanuel Gibon
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California.,Department of Biomechanics and Bone and Joint Biomaterials Laboratory - UMR CNRS 7052, School of Medicine - Paris7 University, Paris, France
| | - Laura Lu
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Karthik Nathan
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Eemeli Jämsen
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Zhenyu Yao
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Stuart B Goodman
- Departments of Orthopaedic Surgery, Stanford University, Stanford, California.,Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
106
|
Barthes J, Ciftci S, Ponzio F, Knopf-Marques H, Pelyhe L, Gudima A, Kientzl I, Bognár E, Weszl M, Kzhyshkowska J, Vrana NE. Review: the potential impact of surface crystalline states of titanium for biomedical applications. Crit Rev Biotechnol 2017; 38:423-437. [PMID: 28882077 DOI: 10.1080/07388551.2017.1363707] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In many biomedical applications, titanium forms an interface with tissues, which is crucial to ensure its long-term stability and safety. In order to exert control over this process, titanium implants have been treated with various methods that induce physicochemical changes at nano and microscales. In the past 20 years, most of the studies have been conducted to see the effect of topographical and physicochemical changes of titanium surface after surface treatments on cells behavior and bacteria adhesion. In this review, we will first briefly present some of these surface treatments either chemical or physical and we explain the biological responses to titanium with a specific focus on adverse immune reactions. More recently, a new trend has emerged in titanium surface science with a focus on the crystalline phase of titanium dioxide and the associated biological responses. In these recent studies, rutile and anatase are the major two polymorphs used for biomedical applications. In the second part of this review, we consider this emerging topic of the control of the crystalline phase of titanium and discuss its potential biological impacts. More in-depth analysis of treatment-related surface crystalline changes can significantly improve the control over titanium/host tissue interface and can result in considerable decreases in implant-related complications, which is currently a big burden on the healthcare system.
Collapse
Affiliation(s)
- Julien Barthes
- a Fundamental Research Unit , Protip Medical , Strasbourg , France.,b INSERM, UMR-S 1121 , , "Biomatériaux et Bioingénierie" , Strasbourg Cedex , France
| | - Sait Ciftci
- b INSERM, UMR-S 1121 , , "Biomatériaux et Bioingénierie" , Strasbourg Cedex , France.,c Service ORL , Hopitaux Universitaires de Strasbourg , Strasbourg , France
| | - Florian Ponzio
- b INSERM, UMR-S 1121 , , "Biomatériaux et Bioingénierie" , Strasbourg Cedex , France.,d Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg , Fédération des Matériaux et Nanoscience d'Alsace (FMNA), Faculté de Chirurgie Dentaire , Strasbourg , France
| | - Helena Knopf-Marques
- b INSERM, UMR-S 1121 , , "Biomatériaux et Bioingénierie" , Strasbourg Cedex , France.,d Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg , Fédération des Matériaux et Nanoscience d'Alsace (FMNA), Faculté de Chirurgie Dentaire , Strasbourg , France
| | - Liza Pelyhe
- e Department of Materials Science and Engineering, Faculty of Mechanical Engineering , Budapest University of Technology and Economics , Budapest , Hungary
| | - Alexandru Gudima
- f Medical Faculty Mannheim , Institute of Transfusion Medicine and Immunology, University of Heidelberg , Mannheim , Germany
| | - Imre Kientzl
- e Department of Materials Science and Engineering, Faculty of Mechanical Engineering , Budapest University of Technology and Economics , Budapest , Hungary
| | - Eszter Bognár
- e Department of Materials Science and Engineering, Faculty of Mechanical Engineering , Budapest University of Technology and Economics , Budapest , Hungary.,g MTA-BME Research Group for Composite Science and Technology , Budapest , Hungary
| | - Miklós Weszl
- h Department of Biophysics and Radiation Biology , Semmelweis University , Budapest , Hungary
| | - Julia Kzhyshkowska
- f Medical Faculty Mannheim , Institute of Transfusion Medicine and Immunology, University of Heidelberg , Mannheim , Germany.,i German Red Cross Blood Service Baden-Württemberg-Hessen , Mannheim , Germany
| | - Nihal Engin Vrana
- a Fundamental Research Unit , Protip Medical , Strasbourg , France.,b INSERM, UMR-S 1121 , , "Biomatériaux et Bioingénierie" , Strasbourg Cedex , France
| |
Collapse
|
107
|
Córdova LA, Loi F, Lin TH, Gibon E, Pajarinen J, Nabeshima A, Lu L, Yao Z, Goodman SB. CCL2, CCL5, and IGF-1 participate in the immunomodulation of osteogenesis during M1/M2 transition in vitro. J Biomed Mater Res A 2017; 105:3069-3076. [PMID: 28782174 DOI: 10.1002/jbm.a.36166] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 06/21/2017] [Accepted: 08/01/2017] [Indexed: 12/20/2022]
Abstract
The modulation of macrophage phenotype from pro-inflammatory (M1) to tissue healing (M2) via exogenous addition of interleukin-4 (IL-4) facilitates osteogenesis; however, the molecular mediators underlying this phenomenon remain unknown. This study characterizes the IL-4-dependent paracrine crosstalk between macrophages and osteoprogenitors and its effect on osteogenesis in vitro. Primary murine M1 were co-cultured with MC3T3 cells (M1-MC3T3) in both transwell plates and direct co-cultures. To modulate M1 to M2, M1-MC3T3 were treated with IL-4 (20 ng/mL) at day 3 after seeding (M1 + IL-4-MC3T3). Selected molecular targets were assessed at days 3 and 6 after seeding at protein and mRNA levels. Mineralization was assessed at day 21. Transwell M1 + IL-4-MC3T3 significantly enhanced the secretion of CCL2/MCP-1, IGF-1 and to a lesser degree, CCL5/RANTES at day 6. At day 3, alkaline phosphatase (Alpl) was upregulated in direct M1-MC3T3. At day 6, Smurf2 and Insulin growth factor-1 (IGF-1) were downregulated and upregulated, respectively, in direct M1 + IL-4-MC3T3. Finally, M1 + IL-4-MC3T3 increased bone matrix mineralization compared with MC3T3 cells in transwell, but this was significantly less than M1-MC3T3. Taken together, macrophage subtypes enhanced the osteogenesis in transwell setting and the transition from M1 to M2 was associated with an increase in bone anabolic factors CCL2/MCP-1, CCL5/RANTES and IGF-1 in vitro. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 3069-3076, 2017.
Collapse
Affiliation(s)
- Luis A Córdova
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305.,Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Chile, Independencia, Santiago, 8380000, Chile
| | - Florence Loi
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Tzu-Hua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Emmanuel Gibon
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305.,Biomechanics and Bone & Joint Biomaterials Laboratory, Faculty of Medicine, Paris7 University, Paris, France
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Akira Nabeshima
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Laura Lu
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305.,Department of Bioengineering, Stanford University, Stanford, California, 94305
| |
Collapse
|
108
|
Lee JO, Yang WS, Park JG, Jeong D, Kim HG, Yoon KD, Aravinthan A, Kim JH, Kim E, Cho JY. Src and Syk contribute to the anti-inflammatory activities of Achyranthes aspera ethanolic extract. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:1-7. [PMID: 28502904 DOI: 10.1016/j.jep.2017.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/23/2017] [Accepted: 05/10/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nuclear factor-kappa B (NF-κB) plays pivotal roles in inflammation. Src and Syk are two tyrosine kinases that act upstream of NF-κB signaling. Although Achyranthes aspera L. (A. aspera) has been used as a traditional medicine to treat fevers and inflammatory ailments and heal wounds, the molecular mechanisms of its anti-inflammatory actions are not yet fully understood. MATERIALS AND METHODS In this study, we evaluated the anti-inflammatory effect of A. aspera ethanol extract (Aa-EE). To determine the mechanism by which Aa-EE dampens the inflammatory response, nitric oxide (NO) production and the mRNA expression levels of tumor necrosis factor (TNF)-α and inducible nitric oxide synthase (iNOS) were examined by Griess assay and RT-PCR. Luciferase assays and immunoblotting were also conducted to examine how Aa-EE regulates the NF-κB pathway. RESULTS Aa-EE reduced NO production up to 60% without any cytotoxicity. This extract was found to downregulate the mRNA expression levels of inflammatory genes. Aa-EE blocked NF-κB promoter activity induced by both TNF-α and adaptor molecule MyD88 (about 70% and 40%, respectively). Moreover, nuclear translocation of p65 and IκBα phosphorylation were also inhibited. Furthermore, Aa-EE inactivated two upstream signaling molecules, the Src and Syk kinases. In accordance with these data, the kinase activities of Src and Syk were decreased by 50% and 80%, respectively. The anti-inflammatory action of Aa-EE was also confirmed in a gastritis model. CONCLUSION Our data suggest that Aa-EE targets NF-κB to exert its anti-inflammatory properties by suppressing Src and Syk. Therefore, our study raises the possibility that this extract can be developed as a novel natural anti-inflammatory remedy.
Collapse
Affiliation(s)
- Jeong-Oog Lee
- Department of Aerospace Information Engineering, Bio-Inspired Aerospace Information Laboratory, Konkuk University, Seoul 05029, Republic of Korea
| | - Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Gwang Park
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Deok Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Han Gyung Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kee Dong Yoon
- College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Adithan Aravinthan
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea.
| | - Eunji Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
109
|
Zhao S, Yan L, Li X, Zhang Z, Sun Y, Wang J. Notoginsenoside R1 suppresses wear particle-induced osteolysis and RANKL mediated osteoclastogenesis in vivo and in vitro. Int Immunopharmacol 2017; 47:118-125. [DOI: 10.1016/j.intimp.2017.03.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/05/2017] [Accepted: 03/20/2017] [Indexed: 11/25/2022]
|
110
|
Deng Z, Jin J, Wang Z, Wang Y, Gao Q, Zhao J. The metal nanoparticle-induced inflammatory response is regulated by SIRT1 through NF-κB deacetylation in aseptic loosening. Int J Nanomedicine 2017; 12:3617-3636. [PMID: 28553103 PMCID: PMC5439723 DOI: 10.2147/ijn.s124661] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aseptic loosening is the most common cause of total hip arthroplasty (THA) failure, and osteolysis induced by wear particles plays a major role in aseptic loosening. Various pathways in multiple cell types contribute to the pathogenesis of osteolysis, but the role of Sirtuin 1 (SIRT1), which can regulate inflammatory responses through its deacetylation, has never been investigated. We hypothesized that the downregulation of SIRT1 in macrophages induced by metal nanoparticles was one of the reasons for osteolysis in THA failure. In this study, the expression of SIRT1 was examined in macrophages stimulated with metal nanoparticles from materials used in prosthetics and in specimens from patients suffering from aseptic loosening. To address whether SIRT1 downregulation triggers these inflammatory responses, the effects of the SIRT1 activator resveratrol on the expression of inflammatory cytokines in metal nanoparticle-stimulated macrophages were tested. The results demonstrated that SIRT1 expression was significantly downregulated in metal nanoparticle-stimulated macrophages and clinical specimens of prosthesis loosening. Pharmacological activation of SIRT1 dramatically reduced the particle-induced expression of inflammatory cytokines in vitro and osteolysis in vivo. Furthermore, SIRT1 regulated particle-induced inflammatory responses through nuclear factor kappa B (NF-κB) acetylation. Thus, the results of this study suggest that SIRT1 plays a key role in metal nanoparticle-induced inflammatory responses and that targeting the SIRT1 pathway may lead to novel therapeutic approaches for the treatment of aseptic prosthesis loosening.
Collapse
Affiliation(s)
- Zhantao Deng
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University
- Center for Translational Medicine, Nanjing University Medical School
- Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Jiewen Jin
- Center for Translational Medicine, Nanjing University Medical School
- Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Zhenheng Wang
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University
| | - Yong Wang
- Center for Translational Medicine, Nanjing University Medical School
- Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Qian Gao
- Center for Translational Medicine, Nanjing University Medical School
- Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Jianning Zhao
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University
| |
Collapse
|
111
|
Maycas M, Portolés MT, Matesanz MC, Buendía I, Linares J, Feito MJ, Arcos D, Vallet-Regí M, Plotkin LI, Esbrit P, Gortázar AR. High glucose alters the secretome of mechanically stimulated osteocyte-like cells affecting osteoclast precursor recruitment and differentiation. J Cell Physiol 2017; 232:3611-3621. [PMID: 28138960 DOI: 10.1002/jcp.25829] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 01/27/2017] [Accepted: 01/27/2017] [Indexed: 02/05/2023]
Abstract
Diabetes mellitus (DM) induces bone deterioration, while mechanical stimulation promotes osteocyte-driven bone formation. We aimed to evaluate the interaction of acute exposure (24 h) to high glucose (HG) with both the pro-survival effect conferred to osteocytic MLO-Y4 cells and osteoblastic MC3T3-E1 cells by mechanical stimulation and the interaction of these cells with osteoclast precursor RAW264.7 cells. We found that 24 h of HG (25 mM) pre-exposure prevented both cell survival and ERK and β-catenin nuclear translocation upon mechanical stimulation by fluid flow (FF) (10 min) in both MLO-Y4 and MC3T3-E1 cells. However, migration of RAW 264.7 cells was inhibited by MLO-Y4 cell-conditioned medium (CM), but not by MC3T3-E1 cell-CM, with HG or FF. This inhibitory effect was associated with consistent changes in VEGF, RANTES, MIP-1α, MIP-1β MCP-1, and GM-CSF in MLO-Y4 cell-CM. RAW264.7 proliferation was inhibited by MLO-Y4 CM under static or HG conditions, but it increased by FF-CM with or without HG. In addition, both FF and HG abrogated the capacity of RAW 264.7 cells to differentiate into osteoclasts, but in a different manner. Thus, HG-CM in static condition allowed formation of osteoclast-like cells, which were unable to resorb hydroxyapatite. In contrast, FF-CM prevented osteoclastogenesis even in HG condition. Moreover, HG did not affect basal RANKL or IL-6 secretion or their inhibition induced by FF in MLO-Y4 cells. In conclusion, this in vitro study demonstrates that HG exerts disparate effects on osteocyte mechanotransduction, and provides a novel mechanism by which DM disturbs skeletal metabolism through altered osteocyte-osteoclast communication.
Collapse
Affiliation(s)
- Marta Maycas
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - María Teresa Portolés
- Facultad de Ciencias Químicas, Departamento de Bioquímica y Biología Molecular I, UCM, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - María Concepción Matesanz
- Facultad de Ciencias Químicas, Departamento de Bioquímica y Biología Molecular I, UCM, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Irene Buendía
- IMMA- Facultad de Medicina, Universidad San Pablo CEU, Boadilla del Monte, Madrid, Spain
| | - Javier Linares
- Facultad de Farmacia, Departamento de Química Inorgánica y Bioinorgánica, UCM, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre (i + 12) CIBER-BBN, Madrid, Spain
| | - María José Feito
- Facultad de Farmacia, Departamento de Química Inorgánica y Bioinorgánica, UCM, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre (i + 12) CIBER-BBN, Madrid, Spain
| | - Daniel Arcos
- Facultad de Farmacia, Departamento de Química Inorgánica y Bioinorgánica, UCM, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre (i + 12) CIBER-BBN, Madrid, Spain
| | - María Vallet-Regí
- Facultad de Farmacia, Departamento de Química Inorgánica y Bioinorgánica, UCM, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre (i + 12) CIBER-BBN, Madrid, Spain
| | - Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Roudebush Veterans Administration Medical Center, Indianapolis, Indiana.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana
| | - Pedro Esbrit
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Arancha R Gortázar
- IMMA- Facultad de Medicina, Universidad San Pablo CEU, Boadilla del Monte, Madrid, Spain
| |
Collapse
|
112
|
The regulation of Jmjd3 upon the expression of NF-κB downstream inflammatory genes in LPS activated vascular endothelial cells. Biochem Biophys Res Commun 2017; 485:62-68. [DOI: 10.1016/j.bbrc.2017.02.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 02/05/2017] [Indexed: 12/21/2022]
|
113
|
Urbanski W, Marycz K, Krzak J, Pezowicz C, Dragan SF. Cytokine induction of sol-gel-derived TiO 2 and SiO 2 coatings on metallic substrates after implantation to rat femur. Int J Nanomedicine 2017; 12:1639-1645. [PMID: 28280331 PMCID: PMC5339000 DOI: 10.2147/ijn.s114885] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Material surface is a key determinant of host response on implanted biomaterial. Therefore, modification of the implant surface may optimize implant–tissue reactions. Inflammatory reaction is inevitable after biomaterial implantation, but prolonged inflammation may lead to adverse reactions and subsequent implant failure. Proinflammatory activities of cytokines like interleukin (IL)-1, IL-6, and tumor necrosis factor-alpha (TNF-α) are attractive indicators of these processes and ultimately characterize biocompatibility. The objective of the study was to evaluate local cytokine production after implantation of stainless steel 316L (SS) and titanium alloy (Ti6Al4V) biomaterials coated with titanium dioxide (TiO2) and silica (SiO2) coatings prepared by sol–gel method. Biomaterials were implanted into rat femur and after 12 weeks, bones were harvested. Bone–implant tissue interface was evaluated; immunohistochemical staining was performed to identify IL-6, TNF-α, and Caspase-1. Histomorphometry (AxioVision Rel. 4.6.3 software) of tissue samples was performed in order to quantify the cytokine levels. Both the oxide coatings on SS and Ti6Al4V significantly reduced cytokine production. However, the lowest cytokine levels were observed in TiO2 groups. Cytokine content in uncoated groups was lower in Ti6Al4V than in SS, although coating of either metal reduced cytokine production to similar levels. Sol–gel TiO2 or SiO2 coatings reduced significantly the production of proinflammatory cytokines by local tissues, irrespective of the material used as a substrate, that is, either Ti6Al4V or SS. This suggests lower inflammatory response, which directly points out improvement of materials’ biocompatibility.
Collapse
Affiliation(s)
- Wiktor Urbanski
- Department of Orthopaedic Surgery and Traumatology, Wroclaw University Hospital
| | - Krzysztof Marycz
- Electron Microscope Laboratory, Wroclaw University of Environmental and Life Sciences
| | - Justyna Krzak
- Institute of Materials Science and Applied Mechanics
| | - Celina Pezowicz
- Division of Biomedical Engineering and Experimental Mechanics, Wroclaw University of Technology, Wroclaw, Poland
| | | |
Collapse
|
114
|
Veronesi F, Tschon M, Fini M. Gene Expression in Osteolysis: Review on the Identification of Altered Molecular Pathways in Preclinical and Clinical Studies. Int J Mol Sci 2017; 18:E499. [PMID: 28245614 PMCID: PMC5372515 DOI: 10.3390/ijms18030499] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 02/06/2023] Open
Abstract
Aseptic loosening (AL) due to osteolysis is the primary cause of joint prosthesis failure. Currently, a second surgery is still the only available treatment for AL, with its associated drawbacks. The present review aims at identifying genes whose expression is altered in osteolysis, and that could be the target of new pharmacological treatments, with the goal of replacing surgery. This review also aims at identifying the molecular pathways altered by different wear particles. We reviewed preclinical and clinical studies from 2010 to 2016, analyzing gene expression of tissues or cells affected by osteolysis. A total of 32 in vitro, 16 in vivo and six clinical studies were included. These studies revealed that genes belonging to both inflammation and osteoclastogenesis pathways are mainly involved in osteolysis. More precisely, an increase in genes encoding for the following factors were observed: Interleukins 6 and 1β (IL16 and β), Tumor Necrosis Factor α (TNFα), nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB), Nuclear factor of activated T-cells, cytoplasmic 1 (NFATC1), Cathepsin K (CATK) and Tartrate-resistant acid phosphatase (TRAP). Titanium (Ti) and Polyethylene (PE) were the most studied particles, showing that Ti up-regulated inflammation and osteoclastogenesis related genes, while PE up-regulated primarily osteoclastogenesis related genes.
Collapse
Affiliation(s)
- Francesca Veronesi
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, via di Barbiano 1/10, 40136 Bologna, Italy.
| | - Matilde Tschon
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, via di Barbiano 1/10, 40136 Bologna, Italy.
| | - Milena Fini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, via di Barbiano 1/10, 40136 Bologna, Italy.
| |
Collapse
|
115
|
Li Y, Li J, Li B, Qin H, Peng X, Zhao Y, Chen Y. Anthocyanin suppresses CoCrMo particle-induced osteolysis by inhibiting IKKα/β mediated NF-κB signaling in a mouse calvarial model. Mol Immunol 2017; 85:27-34. [PMID: 28208071 DOI: 10.1016/j.molimm.2017.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/20/2017] [Accepted: 02/06/2017] [Indexed: 12/23/2022]
Abstract
Wear particle-induced osteolysis and bone resorption have been identified as critical factors of implant failure and total joint revision, in which nuclear factor kappa B (NF-κB) signaling and chronic inflammation have been shown to play key roles. Although anthocyanin is known to have anti-inflammatory function via blocking NF-κB pathway, it is still unclear whether anthocyanin has a protective effect on particle-induced osteolysis. In the present study, we aimed to investigate the detailed effects and the underlying mechanism of anthocyanin on CoCrMo particle-induced osteolysis in a mouse calvavial model. One hundred and twelve male BALB/c mice were divided randomly into four groups: sham group (sham operation and injection with PBS), vehicle group (CoCrMo particle treatment and injection with PBS), low-dose anthocyanin group (CoCrMo particle treatment and injecting anthocyanin with 0.1mg/g/day), and high-dose anthocyanin group (CoCrMo particle treatment and injecting anthocyanin with 0.4mg/g/day). Mice were sacrificed after two weeks, harvesting the calvariae tissue for in depth analysis by micro-CT, histomorphometry, immunohistochemical and molecular biology analysis. As expected, anthocyanin markedly inhibited CoCrMo particle-induced inflammatory infiltration and decreased bone loss in vivo. Anthocyanin also reversed the increase in the ratio of receptor activator of nuclear factor kappa B ligand (RANKL)/osteoproteger (OPG) and suppressed osteoclast formation in CoCrMo particle-stimulated calvaria. Additionally, anthocyanin significantly reduced the expression and secretion of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) in the calvaria of CoCrMo-stimulated mice. Furthermore, we confirmed that anthocyanin attenuated osteolysis by blocking NF-κB pathway via inhibiting inhibitor of nuclear factor kappa-B kinase α/β (IKKα/β) phosphorylation. In conclusion, our study demonstrated that anthocyanin can protect against CoCrMo particle-induced inflammatory osteolysis via inhibiting the IKKα/β-NF-κB pathway, and have a potential therapeutic effect on the treatment of wear particle-induced osteolysis.
Collapse
Affiliation(s)
- Yamin Li
- Department of Orthopaedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 7th Floor Orthopaedic Department, No. 6 Building, No. 600 Yishan Road, Shanghai, China.
| | - Juehong Li
- Department of Orthopaedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 7th Floor Orthopaedic Department, No. 6 Building, No. 600 Yishan Road, Shanghai, China.
| | - Bin Li
- Department of Orthopaedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 7th Floor Orthopaedic Department, No. 6 Building, No. 600 Yishan Road, Shanghai, China.
| | - Hui Qin
- Department of Orthopaedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 7th Floor Orthopaedic Department, No. 6 Building, No. 600 Yishan Road, Shanghai, China.
| | - Xiaochun Peng
- Department of Orthopaedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 7th Floor Orthopaedic Department, No. 6 Building, No. 600 Yishan Road, Shanghai, China.
| | - Yaochao Zhao
- Department of Orthopaedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 7th Floor Orthopaedic Department, No. 6 Building, No. 600 Yishan Road, Shanghai, China.
| | - Yunsu Chen
- Department of Orthopaedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 7th Floor Orthopaedic Department, No. 6 Building, No. 600 Yishan Road, Shanghai, China.
| |
Collapse
|
116
|
Zhang A, Wang K, Ding L, Bao X, Wang X, Qiu X, Liu J. Bay11-7082 attenuates neuropathic pain via inhibition of nuclear factor-kappa B and nucleotide-binding domain-like receptor protein 3 inflammasome activation in dorsal root ganglions in a rat model of lumbar disc herniation. J Pain Res 2017; 10:375-382. [PMID: 28243141 PMCID: PMC5315342 DOI: 10.2147/jpr.s119820] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lumbar disc herniation (LDH) is an important cause of radiculopathy, but the underlying mechanisms are incompletely understood. Many studies suggested that local inflammation, rather than mechanical compression, results in radiculopathy induced by LDH. On the molecular and cellular level, nuclear factor-kappa B (NF-κB) and nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome have been implicated in the regulation of neuroinflammation formation and progression. In this study, the autologous nucleus pulposus (NP) was implanted in the left L5 dorsal root ganglion (DRG) to mimic LDH in rats. We investigated the expression of NF-κB and the components of NLRP3 inflammasome in the DRG neurons in rats. Western blotting and immunofluorescence for the related molecules, including NLRP3, apoptosis-associated speck-like protein containing caspase-1 activator domain (ASC), caspase-1, interleukin (IL)-1β, IL-18, IκBα, p-IκBα, p65, p-p65, and calcitonin gene-related peptide (CGRP) were examined. In the NP-treated group, the activations of NLRP3, ASC, caspase-1, IL-1β, IL-18, p-IκBα, and p-p65 in DRG neurons in rats were elevated at 1 day after surgery, and the peak occurred at 7 days. Treatment with Bay11-7082, an inhibitor of the actions of IKK-β, was able to inhibit expression and activation of the molecules (NLRP3, ASC, caspase-1, IL-1β, IL-18, p-IκBα, and p-p65) and relieve the pain in rats. Our study shows that NF-κB and NLRP3 inflammasome are involved in the maintenance of NP-induced pain, and that Bay11-7082 could alleviate mechanical allodynia and thermal hyperalgesia by inhibiting NF-κB and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Ailiang Zhang
- Spine Surgery, Third Affiliated Hospital of Soochow University, Changzhou, People's Republic of China
| | - Kun Wang
- Spine Surgery, Third Affiliated Hospital of Soochow University, Changzhou, People's Republic of China
| | - Lianghua Ding
- Spine Surgery, Third Affiliated Hospital of Soochow University, Changzhou, People's Republic of China
| | - Xinnan Bao
- Spine Surgery, Third Affiliated Hospital of Soochow University, Changzhou, People's Republic of China
| | - Xuan Wang
- Spine Surgery, Third Affiliated Hospital of Soochow University, Changzhou, People's Republic of China
| | - Xubin Qiu
- Spine Surgery, Third Affiliated Hospital of Soochow University, Changzhou, People's Republic of China
| | - Jinbo Liu
- Spine Surgery, Third Affiliated Hospital of Soochow University, Changzhou, People's Republic of China
| |
Collapse
|
117
|
Lin TH, Gibon E, Loi F, Pajarinen J, Córdova LA, Nabeshima A, Lu L, Yao Z, Goodman SB. Decreased osteogenesis in mesenchymal stem cells derived from the aged mouse is associated with enhanced NF-κB activity. J Orthop Res 2017; 35:281-288. [PMID: 27105133 DOI: 10.1002/jor.23270] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/08/2016] [Indexed: 02/04/2023]
Abstract
Aging is associated with significant bone loss and delayed fracture healing. NF-κB activation is highly correlated with inflammatory-associated bone diseases including infection, wear particle exposure, and chronic inflammation during natural aging processes. The critical roles of NF-κB in both the pro-inflammatory response and osteoclast-mediated bone resorption have been well defined. However, the biological effects of NF-κB activation in mesenchymal stem cell (MSC)-mediated bone formation remain largely unknown. In the current study, bone marrow-MSCs were isolated from young (8 weeks old) and aged (72 weeks old) mice. NF-κB activity in MSCs at basal levels and under different biological conditions were determined by our recently established lentiviral vector-based luciferase reporter assay. We found that NF-κB activity was increased in aged MSCs at basal levels or when exposed to low dose (10 or 100 ng/ml) lipopolysaccharide (LPS); this effect was not seen when the cells were exposed to higher dose (1 μg/ml) LPS. During osteogenesis, NF-κB activity was increased in aged MSCs at weeks 1 and 2, but showed no significant difference at week 3. Both Smurf2 and TAZ, the NF-κB target genes that regulate osteogenic differentiation, were increased in aged MSCs. In addition, the expression of RANKL was dramatically increased, and OPG was decreased in aged MSCs. Our findings suggest that targeting NF-κB activity in MSCs has the potential to modulate aging-associated bone loss, or enhance bone-healing in aged patients. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:281-288, 2017.
Collapse
Affiliation(s)
- Tzu-Hua Lin
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, 300 Pasteur Drive, Palo Alto, California, 94304
| | - Emmanuel Gibon
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, 300 Pasteur Drive, Palo Alto, California, 94304.,Biomecanics and Bone and Joint Biomaterials Laboratory-UMR CNRS 7052, School of Medicine-Paris7 University, Paris, France
| | - Florence Loi
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, 300 Pasteur Drive, Palo Alto, California, 94304
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, 300 Pasteur Drive, Palo Alto, California, 94304
| | - Luis A Córdova
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, 300 Pasteur Drive, Palo Alto, California, 94304.,Faculty of Dentistry, Department of Oral and Maxillofacial Surgery, University of Chile-Conicyt, Santiago, Chile
| | - Akira Nabeshima
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, 300 Pasteur Drive, Palo Alto, California, 94304
| | - Laura Lu
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, 300 Pasteur Drive, Palo Alto, California, 94304
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, 300 Pasteur Drive, Palo Alto, California, 94304
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, 300 Pasteur Drive, Palo Alto, California, 94304.,Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
118
|
Deng Z, Wang Z, Jin J, Wang Y, Bao N, Gao Q, Zhao J. SIRT1 protects osteoblasts against particle-induced inflammatory responses and apoptosis in aseptic prosthesis loosening. Acta Biomater 2017; 49:541-554. [PMID: 27890623 DOI: 10.1016/j.actbio.2016.11.051] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/19/2016] [Accepted: 11/22/2016] [Indexed: 02/07/2023]
Abstract
We hypothesized that SIRT1 downregulation in osteoblasts induced by wear particles was one of the reasons for particle-induced osteolysis (PIO) in total joint arthroplasty failure. In the present study, the expression of SIRT1 was examined in osteoblasts treated with TiAl6V4 particles (TiPs) and CoCrMo particles (CoPs) from materials used in prosthetics and specimens from PIO animal models. To address whether SIRT1 downregulation triggers inflammatory responses and apoptosis in osteoblasts, the effect of a SIRT1 activator, resveratrol on the expression of inflammatory cytokines and apoptosis in particle-treated osteoblasts was tested. The results demonstrated that SIRT1 expression was significantly downregulated in particle-treated osteoblasts and PIO animal models. Both pharmacological activation and overexpression of SIRT1 dramatically reduced the particle-induced expression of inflammatory cytokines and osteoblast apoptosis through NF-κB and p53 signaling, respectively. Furthermore, in PIO animal models, resveratrol significantly reduced the severity of osteolysis. Collectively, the results of the present study indicated that SIRT1 plays a vital role in the pathogenesis of aseptic loosening, and further treatment targeted at SIRT1 possibly lead to novel approaches for prevention of aseptic prosthesis loosening. STATEMENT OF SIGNIFICANCE Aseptic loosening is the most common cause of total hip arthroplasty (THA) and total knee arthroplasty (TKA) failure and revision surgery. However, there is still no effective therapeutic target in the clinical treatment. Besides, the underlying mechanism of aseptic loosening is largely unknown. The result of our study indicated that SIRT1 has the ability to effectively regulate the wear particle-induced inflammatory responses, apoptosis, osteolysis in particle-stimulated osteoblasts and particle-induced osteolysis animal models. Our study provides a potential target for the prevention and treatment of aseptic loosening and further investigated the underlying mechanism of aseptic loosening, which may make contribution to decrease the incidence of THA and TKA failure in the clinical practice.
Collapse
Affiliation(s)
- Zhantao Deng
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, PR China; Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu, PR China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, PR China.
| | - Zhenheng Wang
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, PR China.
| | - Jiewen Jin
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu, PR China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, PR China.
| | - Yong Wang
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu, PR China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, PR China.
| | - Nirong Bao
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, PR China.
| | - Qian Gao
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu, PR China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, PR China.
| | - Jianning Zhao
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
119
|
Pajarinen J, Lin TH, Nabeshima A, Jämsen E, Lu L, Nathan K, Yao Z, Goodman SB. Mesenchymal stem cells in the aseptic loosening of total joint replacements. J Biomed Mater Res A 2017; 105:1195-1207. [PMID: 27977880 DOI: 10.1002/jbm.a.35978] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/06/2016] [Indexed: 02/06/2023]
Abstract
Peri-prosthetic osteolysis remains as the main long-term complication of total joint replacement surgery. Research over four decades has established implant wear as the main culprit for chronic inflammation in the peri-implant tissues and macrophages as the key cells mediating the host reaction to implant-derived wear particles. Wear debris activated macrophages secrete inflammatory mediators that stimulate bone resorbing osteoclasts; thus bone loss in the peri-implant tissues is increased. However, the balance of bone turnover is not only dictated by osteoclast-mediated bone resorption but also by the formation of new bone by osteoblasts; under physiological conditions these two processes are tightly coupled. Increasing interest has been placed on the effects of wear debris on the cells of the bone-forming lineage. These cells are derived primarily from multipotent mesenchymal stem cells (MSCs) residing in bone marrow and the walls of the microvasculature. Accumulating evidence indicates that wear debris significantly impairs MSC-to-osteoblast differentiation and subsequent bone formation. In this review, we summarize the current understanding of the effects of biomaterial implant wear debris on MSCs. Emerging treatment options to improve initial implant integration and treat developing osteolytic lesions by utilizing or targeting MSCs are also discussed. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1195-1207, 2017.
Collapse
Affiliation(s)
- Jukka Pajarinen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Tzu-Hua Lin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Akira Nabeshima
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Eemeli Jämsen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California.,Department of Medicine, Clinicum, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Laura Lu
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Karthik Nathan
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Zhenyu Yao
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Stuart B Goodman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
120
|
Zeng X, Zhang Y, Wang S, Wang K, Tao L, Zou M, Chen N, Xu J, Liu S, Li X. Artesunate suppresses RANKL-induced osteoclastogenesis through inhibition of PLCγ1-Ca 2+ –NFATc1 signaling pathway and prevents ovariectomy-induced bone loss. Biochem Pharmacol 2017; 124:57-68. [DOI: 10.1016/j.bcp.2016.10.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/21/2016] [Indexed: 11/29/2022]
|
121
|
Lin TH, Pajarinen J, Lu L, Nabeshima A, Cordova LA, Yao Z, Goodman SB. NF-κB as a Therapeutic Target in Inflammatory-Associated Bone Diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 107:117-154. [PMID: 28215222 DOI: 10.1016/bs.apcsb.2016.11.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Inflammation is a defensive mechanism for pathogen clearance and maintaining tissue homeostasis. In the skeletal system, inflammation is closely associated with many bone disorders including fractures, nonunions, periprosthetic osteolysis (bone loss around orthopedic implants), and osteoporosis. Acute inflammation is a critical step for proper bone-healing and bone-remodeling processes. On the other hand, chronic inflammation with excessive proinflammatory cytokines disrupts the balance of skeletal homeostasis involving osteoblastic (bone formation) and osteoclastic (bone resorption) activities. NF-κB is a transcriptional factor that regulates the inflammatory response and bone-remodeling processes in both bone-forming and bone-resorption cells. In vitro and in vivo evidences suggest that NF-κB is an important potential therapeutic target for inflammation-associated bone disorders by modulating inflammation and bone-remodeling process simultaneously. The challenges of NF-κB-targeting therapy in bone disorders include: (1) the complexity of canonical and noncanonical NF-κB pathways; (2) the fundamental roles of NF-κB-mediated signaling for bone regeneration at earlier phases of tissue damage and acute inflammation; and (3) the potential toxic effects on nontargeted cells such as lymphocytes. Recent developments of novel inhibitors with differential approaches to modulate NF-κB activity, and the controlled release (local) or bone-targeting drug delivery (systemic) strategies, have largely increased the translational application of NF-κB therapy in bone disorders. Taken together, temporal modulation of NF-κB pathways with the combination of recent advanced bone-targeting drug delivery techniques is a highly translational strategy to reestablish homeostasis in the skeletal system.
Collapse
Affiliation(s)
- T-H Lin
- Stanford University, Stanford, CA, United States
| | - J Pajarinen
- Stanford University, Stanford, CA, United States
| | - L Lu
- Stanford University, Stanford, CA, United States
| | - A Nabeshima
- Stanford University, Stanford, CA, United States
| | - L A Cordova
- Stanford University, Stanford, CA, United States; Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Z Yao
- Stanford University, Stanford, CA, United States
| | - S B Goodman
- Stanford University, Stanford, CA, United States.
| |
Collapse
|
122
|
Mouthuy PA, Snelling SJ, Dakin SG, Milković L, Gašparović AČ, Carr AJ, Žarković N. Biocompatibility of implantable materials: An oxidative stress viewpoint. Biomaterials 2016; 109:55-68. [PMID: 27669498 DOI: 10.1016/j.biomaterials.2016.09.010] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/06/2016] [Accepted: 09/13/2016] [Indexed: 12/13/2022]
|
123
|
Sartori M, Vincenzi F, Ravani A, Cepollaro S, Martini L, Varani K, Fini M, Tschon M. RAW 264.7 co-cultured with ultra-high molecular weight polyethylene particles spontaneously differentiate into osteoclasts: an in vitro model of periprosthetic osteolysis. J Biomed Mater Res A 2016; 105:510-520. [PMID: 27667508 DOI: 10.1002/jbm.a.35912] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 09/07/2016] [Accepted: 09/20/2016] [Indexed: 12/19/2022]
Abstract
Wear-particle osteolysis affects prosthesis survival leading to implant loosening up to 70% of revisions. Therapeutic strategies are increasing, however alternative testing methods to experimentally evaluate such treatments are lacking. The aim of this study was to reproduce an in vitro osteolysis model recapitulating the events that, starting from the exposure of macrophages to polyethylene, lead to the establishment of osteoclastogenesis and inflammation. Responses to polyethylene, at 3 and 7 days, in a macrophage cell line, RAW 264.7, were determined by DNA quantification, immunofluorescence, pit assay, gene expression, cytokine production and NF-kB activation. Results showed that 3 days exposure to particles could induce a significant production of Tumor Necrosis Factor alpha (p < 0.0005) and Prostaglandin E2 (p < 0.005) compared to controls. Particles also induced macrophages to spontaneously differentiate into mature and active osteoclasts, in terms of identification of multinucleated cells by Phalloidin staining and by the analysis of osteoclast-specific gene markers. In particular, at 3 days polyethylene induced a significant up-regulation of Nuclear Factor of Activated T-cells, cytoplasmic 1, Receptor Activator of Nuclear factor Kappa-B and Receptor Activator of Nuclear Factor Kappa-B Ligand genes (p < 0.0005) compared to controls. At protein level, the particles induced a significant increase of Receptor Activator of Nuclear Factor Kappa-B Ligand at day 7 over controls (p < 0.0005). Osteoclasts were capable to resorb bone even in absence of differentiating factors. The possible mechanism, beside spontaneous osteoclastogenesis mediated by wear debris, was identified in an autocrine up-regulation of Receptor activator of nuclear factor kappa-B ligand gene expression and protein synthesis. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 510-520, 2017.
Collapse
Affiliation(s)
- M Sartori
- Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, BITTA, Rizzoli Orthopaedic Institute- Research, Innovation and Technology Department (RIT), via di Barbiano 1/10, 40136, Bologna, Italy
| | - F Vincenzi
- Laboratory of Cellular and Molecular Pharmacology Department of Medical Sciences, University of Ferrara, via Fossato di Mortara 17-19, Ferrara, 44121, Italy
| | - A Ravani
- Laboratory of Cellular and Molecular Pharmacology Department of Medical Sciences, University of Ferrara, via Fossato di Mortara 17-19, Ferrara, 44121, Italy
| | - S Cepollaro
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, via di Barbiano 1/10, Bologna, 40136, Italy
| | - L Martini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, via di Barbiano 1/10, Bologna, 40136, Italy
| | - K Varani
- Laboratory of Cellular and Molecular Pharmacology Department of Medical Sciences, University of Ferrara, via Fossato di Mortara 17-19, Ferrara, 44121, Italy
| | - M Fini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, via di Barbiano 1/10, Bologna, 40136, Italy
| | - M Tschon
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, via di Barbiano 1/10, Bologna, 40136, Italy
| |
Collapse
|
124
|
Wang Z, Liu N, Liu K, Zhou G, Gan J, Wang Z, Shi T, He W, Wang L, Guo T, Bao N, Wang R, Huang Z, Chen J, Dong L, Zhao J, Zhang J. Autophagy mediated CoCrMo particle-induced peri-implant osteolysis by promoting osteoblast apoptosis. Autophagy 2016; 11:2358-69. [PMID: 26566231 PMCID: PMC4835204 DOI: 10.1080/15548627.2015.1106779] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Wear particle-induced osteolysis is the leading cause of aseptic loosening, which is the most common reason for THA (total hip arthroplasty) failure and revision surgery. Although existing studies suggest that osteoblast apoptosis induced by wear debris is involved in aseptic loosening, the underlying mechanism linking wear particles to osteoblast apoptosis remains almost totally unknown. In the present study, we investigated the effect of autophagy on osteoblast apoptosis induced by CoCrMo metal particles (CoPs) in vitro and in a calvarial resorption animal model. Our study demonstrated that CoPs stimulated autophagy in osteoblasts and PIO (particle-induced osteolysis) animal models. Both autophagy inhibitor 3-MA (3-methyladenine) and siRNA of Atg5 could dramatically reduce CoPs-induced apoptosis in osteoblasts. Further, inhibition of autophagy with 3-MA ameliorated the severity of osteolysis in PIO animal models. Moreover, 3-MA also prevented osteoblast apoptosis in an antiautophagic way when tested in PIO model. Collectively, these results suggest that autophagy plays a key role in CoPs-induced osteolysis and that targeting autophagy-related pathways may represent a potential therapeutic approach for treating particle-induced peri-implant osteolysis.
Collapse
Affiliation(s)
- Zhenheng Wang
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Naicheng Liu
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Kang Liu
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Gang Zhou
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Jingjing Gan
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Zhenzhen Wang
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Tongguo Shi
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Wei He
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Lintao Wang
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Ting Guo
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Nirong Bao
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China
| | - Rui Wang
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China
| | - Zhen Huang
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Jiangning Chen
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Lei Dong
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Jianning Zhao
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China
| | - Junfeng Zhang
- a Jinling Hospital; Department of Orthopaedics; State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University ; Nanjing , China.,b School of Medicine and School of Life Science; Nanjing University ; Nanjing , China.,c Jiangsu Provincial Laboratory for Nano-Technology; Nanjing University , Nanjing , China
| |
Collapse
|
125
|
Blockade of NF-κB and MAPK pathways by ulinastatin attenuates wear particle-stimulated osteoclast differentiation in vitro and in vivo. Biosci Rep 2016; 36:BSR20160234. [PMID: 27638499 PMCID: PMC5091469 DOI: 10.1042/bsr20160234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/15/2016] [Indexed: 11/17/2022] Open
Abstract
Ulinastatin, a urinary trypsin inhibitor (UTI), is widely used to clinically treat lipopolysaccharide (LPS)-related inflammatory disorders recently. Adherent pathogen-associated molecular patterns (PAMPs), of which LPS is the best-studied and classical endotoxin produced by Gram-negative bacteria, act to increase the biological activity of osteopedic wear particles such as polymethyl-methacrylate (PMMA) and titanium particles in cell culture and animal models of implant loosening. The present study was designed to explore the inhibitory effect of UTI on osteoclastogenesis and inflammatory osteolysis in LPS/PMMA-mediated Raw264.7 cells and murine osteolysis models, and investigate the potential mechanism. The in vitro study was divided into the control group, LPS-induced group, PMMA-stimulated group and UTI-pretreated group. UTI (500 or 5000 units/ml) pretreatment was followed by PMMA (0.5 mg/ml) with adherent LPS. The levels of inflammatory mediators including tumour necrosis factor-α (TNF-α), matrixmetallo-proteinases-9 (MMP-9) and interleukin-6 (IL-6), receptor activation of nuclear factor NF-κB (RANK), and cathepsin K were examined and the amounts of phosphorylated I-κB, MEK, JNK and p38 were measured. In vivo study, murine osteolysis models were divided into the control group, PMMA-induced group and UTI-treated group. UTI (500 or 5000 units/kg per day) was injected intraperitoneally followed by PMMA suspension with adherent LPS (2×108 particles/25 μl) in the UTI-treated group. The thickness of interfacial membrane and the number of infiltrated inflammatory cells around the implants were assessed, and bone mineral density (BMD), trabecular number (Tb.N.), trabecular thickness (Tb.Th.), trabecular separation (Tb.Sp.), relative bone volume over total volume (BV/TV) of distal femur around the implants were calculated. Our results showed that UTI pretreatment suppressed the secretion of proinflammatory cytokines including MMP-9, IL-6, TNF-α, RANK and cathepsin K through down-regulating the activity of nuclear factor kappa B (NF-κB) and MAPKs partly in LPS/PMMA-mediated Raw264.7 cells. Finally, UTI treatment decreased the inflammatory osteolysis reaction in PMMA-induced murine osteolysis models. In conclusion, these results confirm the anti-inflammatory potential of UTI in the prevention of particle disease.
Collapse
|
126
|
Tondera C, Hauser S, Krüger-Genge A, Jung F, Neffe AT, Lendlein A, Klopfleisch R, Steinbach J, Neuber C, Pietzsch J. Gelatin-based Hydrogel Degradation and Tissue Interaction in vivo: Insights from Multimodal Preclinical Imaging in Immunocompetent Nude Mice. Theranostics 2016; 6:2114-2128. [PMID: 27698944 PMCID: PMC5039684 DOI: 10.7150/thno.16614] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/05/2016] [Indexed: 12/11/2022] Open
Abstract
Hydrogels based on gelatin have evolved as promising multifunctional biomaterials. Gelatin is crosslinked with lysine diisocyanate ethyl ester (LDI) and the molar ratio of gelatin and LDI in the starting material mixture determines elastic properties of the resulting hydrogel. In order to investigate the clinical potential of these biopolymers, hydrogels with different ratios of gelatin and diisocyanate (3-fold (G10_LNCO3) and 8-fold (G10_LNCO8) molar excess of isocyanate groups) were subcutaneously implanted in mice (uni- or bilateral implantation). Degradation and biomaterial-tissue-interaction were investigated in vivo (MRI, optical imaging, PET) and ex vivo (autoradiography, histology, serum analysis). Multimodal imaging revealed that the number of covalent net points correlates well with degradation time, which allows for targeted modification of hydrogels based on properties of the tissue to be replaced. Importantly, the degradation time was also dependent on the number of implants per animal. Despite local mechanisms of tissue remodeling no adverse tissue responses could be observed neither locally nor systemically. Finally, this preclinical investigation in immunocompetent mice clearly demonstrated a complete restoration of the original healthy tissue.
Collapse
Affiliation(s)
- Christoph Tondera
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany
- Technische Universität Dresden, Department of Chemistry and Food Chemistry, Dresden, Germany
| | - Sandra Hauser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany
| | - Anne Krüger-Genge
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Friedrich Jung
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Helmholtz Virtual Institute “Multifunctional Biomaterials for Medicine”, Teltow and Berlin
| | - Axel T. Neffe
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Helmholtz Virtual Institute “Multifunctional Biomaterials for Medicine”, Teltow and Berlin
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Helmholtz Virtual Institute “Multifunctional Biomaterials for Medicine”, Teltow and Berlin
| | - Robert Klopfleisch
- Freie Universität Berlin, Institute of Veterinary Pathology, Berlin, Germany
| | - Jörg Steinbach
- Technische Universität Dresden, Department of Chemistry and Food Chemistry, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany
- Technische Universität Dresden, Department of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
127
|
NF-κB decoy oligodeoxynucleotide mitigates wear particle-associated bone loss in the murine continuous infusion model. Acta Biomater 2016; 41:273-81. [PMID: 27260104 DOI: 10.1016/j.actbio.2016.05.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 05/25/2016] [Accepted: 05/30/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Total joint replacement is a cost-effective surgical procedure for patients with end-stage arthritis. Wear particle-induced chronic inflammation is associated with the development of periprosthetic osteolysis. Modulation of NF-κB signaling in macrophages, osteoclasts, and mesenchymal stem cells could potentially mitigate this disease. In the current study, we examined the effects of local delivery of decoy NF-κB oligo-deoxynucleotide (ODN) on wear particle-induced bone loss in a murine continuous femoral particle infusion model. Ultra-high molecular weight polyethylene particles (UHMWPE) with or without lipopolysaccharide (LPS) were infused via osmotic pumps into hollow titanium rods placed in the distal femur of mice for 4weeks. Particle-induced bone loss was evaluated by μCT, and immunohistochemical analysis of sections from the femur. Particle infusion alone resulted in reduced bone mineral density and trabecular bone volume fraction in the distal femur. The decoy ODN reversed the particle-associated bone volume fraction loss around the implant, irrespective of the presence of LPS. Particle-infusion with LPS increased bone mineral density in the distal femur compared with particle-infusion alone. NF-κB decoy ODN reversed or further increased the bone mineral density in the femur (3-6mm from the distal end) exposed to particles alone or particles plus LPS. NF-κB decoy ODN also inhibited macrophage infiltration and osteoclast number, but had no significant effects on osteoblast numbers in femurs exposed to wear particles and LPS. Our study suggests that targeting NF-κB activity via local delivery of decoy ODN has great potential to mitigate wear particle-induced osteolysis. STATEMENT OF SIGNIFICANCE Total joint replacement is a cost-effective surgical procedure for patients with end-stage arthritis. Chronic inflammation is crucial for the development of wear particle-associated bone loss. Modulation of NF-κB signaling in macrophages (pro-inflammatory cells), osteoclasts (bone-resorbing cells), and osteoblasts (bone-forming cells) could potentially mitigate this disease. Here we demonstrated that local delivery of decoy NF-κB oligo-deoxynucleotide (ODN) mitigated ultra-high molecular weight polyethylene (UHMWPE) wear particle induced bone loss in a clinically relevant murine model. The protective effects of decoy ODN was associated with reduced macrophage infiltration and osteoclast activation, but had no significant effects on osteoblast numbers. Our study suggests that targeting NF-κB activity via local delivery of decoy ODN has great potential to mitigate wear particle-induced bone loss.
Collapse
|
128
|
Gibon E, Loi F, Córdova LA, Pajarinen J, Lin T, Lu L, Nabeshima A, Yao Z, Goodman SB. Aging Affects Bone Marrow Macrophage Polarization: Relevance to Bone Healing. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2016; 2:98-104. [PMID: 28138512 PMCID: PMC5270653 DOI: 10.1007/s40883-016-0016-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/06/2016] [Indexed: 12/23/2022]
Abstract
Macrophages are an important component of the inflammatory cascade by initiating and modulating the processes leading to tissue regeneration and bone healing. Depending on the local environment, macrophages can be polarized into M1 (pro-inflammatory) or M2 (anti-inflammatory) phenotypes. In order to assess the effects of aging on macrophage function, bone marrow macrophage polarization using primary bone marrow macrophages (BMMs) from young (8 weeks old) and aged (72 weeks old) wild-type male C57BL/6J mice was analyzed. Fluorescence-activated cell sorting (FACS) analysis (CD11b, iNOS, CD206), qRT-PCR (iNOS, TNF-α, CD206, Arginase 1), and ELISA (TNF-α, IL-1ra) were performed to compare the M1 and M2 phenotypic markers in young and aged mouse macrophages. Once M1 and M2 macrophage phenotypes were confirmed, the results showed that TNF-α mRNA was significantly upregulated in aged M1s after interferon gamma (INF-γ) exposure. Arginase 1 and CD206 mRNA expression were still upregulated with IL4 stimulation in aged macrophages, but to a lesser extend than those from younger animals. TNF-α secretion was also significantly increased in aged M1s compared to young M1s, following lipopolysaccharide (LPS) exposure. However, the IL-1ra secretion did not increase accordingly in aged mice. The results demonstrate that, compared to younger animals, aging of bone marrow derived macrophages increases the resting levels of oxidative stress, and the ratios of pro- to anti-inflammatory markers. These age-related changes in macrophage polarization may explain in part the attenuated response to adverse stimuli and delay in processes such as fracture healing seen in the elderly. LAY SUMMARY Bone healing is a complex process that involves both biological and mechanical factors. Macrophages are key cells that regulate the events involved in bone healing, especially the initial inflammatory phase. In this biological cascade of events, macrophages present as different functional phenotypes including uncommitted (M0), pro-inflammatory (M1), and anti-inflammatory (M2), a process called macrophage polarization. A clear understanding of the effects of aging on macrophage polarization is critical to modulating adverse events such as fractures, atraumatic bone loss, and tissue regeneration in an aging population.
Collapse
Affiliation(s)
- E Gibon
- Department of Orthopaedic Surgery, Stanford University, R116, 300 Pasteur Drive, Stanford, CA 94305, USA; Laboratoire de Biomécanique et Biomatériaux Ostéo-Articulaires -UMR CNRS 7052, Faculté de Médecine - Université Paris7, 10 avenue de Verdun, 75010 Paris, France; Department of Orthopaedic Surgery, Hopital Cochin, APHP, Université Paris5, 27 rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - F Loi
- Department of Orthopaedic Surgery, Stanford University, R116, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Luis A Córdova
- Department of Orthopaedic Surgery, Stanford University, R116, 300 Pasteur Drive, Stanford, CA 94305, USA; Department of Oral and Maxillofacial Surgery, University of Chile-Conicyt, Santiago, Chile
| | - J Pajarinen
- Department of Orthopaedic Surgery, Stanford University, R116, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - T Lin
- Department of Orthopaedic Surgery, Stanford University, R116, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - L Lu
- Department of Orthopaedic Surgery, Stanford University, R116, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - A Nabeshima
- Department of Orthopaedic Surgery, Stanford University, R116, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Z Yao
- Department of Orthopaedic Surgery, Stanford University, R116, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, R116, 300 Pasteur Drive, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA; Department of Orthopaedic Surgery and (by courtesy) Bioengineering, Stanford University Medical Center Outpatient Center, 450 Broadway St., M/C 6342, Redwood City, CA 94063, USA
| |
Collapse
|
129
|
Loi F, Córdova LA, Pajarinen J, Lin TH, Yao Z, Goodman SB. Inflammation, fracture and bone repair. Bone 2016; 86:119-30. [PMID: 26946132 PMCID: PMC4833637 DOI: 10.1016/j.bone.2016.02.020] [Citation(s) in RCA: 754] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/30/2015] [Accepted: 02/29/2016] [Indexed: 12/20/2022]
Abstract
The reconstitution of lost bone is a subject that is germane to many orthopedic conditions including fractures and non-unions, infection, inflammatory arthritis, osteoporosis, osteonecrosis, metabolic bone disease, tumors, and periprosthetic particle-associated osteolysis. In this regard, the processes of acute and chronic inflammation play an integral role. Acute inflammation is initiated by endogenous or exogenous adverse stimuli, and can become chronic in nature if not resolved by normal homeostatic mechanisms. Dysregulated inflammation leads to increased bone resorption and suppressed bone formation. Crosstalk among inflammatory cells (polymorphonuclear leukocytes and cells of the monocyte-macrophage-osteoclast lineage) and cells related to bone healing (cells of the mesenchymal stem cell-osteoblast lineage and vascular lineage) is essential to the formation, repair and remodeling of bone. In this review, the authors provide a comprehensive summary of the literature related to inflammation and bone repair. Special emphasis is placed on the underlying cellular and molecular mechanisms, and potential interventions that can favorably modulate the outcome of clinical conditions that involve bone repair.
Collapse
Affiliation(s)
- Florence Loi
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Luis A Córdova
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA; Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Chile, Sergio Livingstone Polhammer 943, Independencia, 8380000 Santiago, Chile.
| | - Jukka Pajarinen
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Tzu-hua Lin
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Zhenyu Yao
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Stuart B Goodman
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA; 300 Pasteur Drive, Edwards Building, Room R114, Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
130
|
Guo W, Guan X, Pan X, Sun X, Wang F, Ji Y, Huang P, Deng Y, Zhang Q, Han Q, Yi P, Namaka M, Liu Y, Deng Y, Li X. Post-Natal Inhibition of NF-κB Activation Prevents Renal Damage Caused by Prenatal LPS Exposure. PLoS One 2016; 11:e0153434. [PMID: 27073902 PMCID: PMC4830567 DOI: 10.1371/journal.pone.0153434] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 03/29/2016] [Indexed: 02/05/2023] Open
Abstract
Prenatal exposure to an inflammatory stimulus has been shown to cause renal damage in offspring. Our present study explored the role of intra-renal NF-κB activation in the development of progressive renal fibrosis in offspring that underwent prenatal exposure to an inflammatory stimulus. Time-dated pregnant rats were treated with saline (control group) or 0.79 mg/kg lipopolysaccharide (LPS) through intra-peritoneal injection on gestational day 8, 10 and 12. At the age of 7 weeks, offspring from control or LPS group were treated with either tap water (Con+Ve or LPS+Ve group) or pyrollidine dithiocarbamate (PDTC, 120mg/L), a NF-κB inhibitor, via drinking water starting (Con+PDTC or LPS+PDTC group), respectively, till the age of 20 or 68 weeks. The gross structure of kidney was assessed by hematoxylin-eosin, periodic acid–Schiff staining and Sirius red staining. The expression levels of TNF-α, IL-6, α-smooth muscle actin (α-SMA) and renin-angiotensin system (RAS) genes were determined by real time polymerase chain reaction and/or immunohistochemical staining. Our data showed that post-natal persistent PDTC administration efficiently repressed intra-renal NF-κB activation, TNF-α and IL-6 expression. Post-natal PDTC also prevented intra-renal glycogen deposition and collagenous fiber generation as evident by the reduced expression of collagen III and interstitial α-SMA in offspring of prenatal LPS exposure. Furthermore, post-natal PDTC administration reversed the intra-renal renin-angiotensin system (RAS) over-activity in offspring of prenatal LPS exposure. In conclusion, prenatal inflammatory exposure results in offspring’s intra-renal NF-κB activation along with inflammation which cross-talked with excessive RAS activation that caused exacerbation of renal fibrosis and dysfunction in the offspring. Thus, early life prevention of NF-κB activation may be a potential preventive strategy for chronic renal inflammation and progressive renal damage.
Collapse
Affiliation(s)
- Wei Guo
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Department of Pharmacy, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiao Guan
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Xiaodong Pan
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Xiongshan Sun
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Fangjie Wang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Yan Ji
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Pei Huang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Yafei Deng
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Qi Zhang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Qi Han
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, Daping Hospital, Third Military Medical University, Chongqing 400038, China
| | - Michael Namaka
- Colleges of Pharmacy and Medicine, University of Manitoba, Apotex Center 750, McDermot Avenue, Winnipeg, R3E 0T5, MB, Canada
- Joint Laboratory of Biological Psychiatry between Shantou University Medical College and the College of Medicine University of Manitoba, Shantou 515063, China
| | - Ya Liu
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- * E-mail: (YCD); (YL); (XL)
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- * E-mail: (YCD); (YL); (XL)
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
- * E-mail: (YCD); (YL); (XL)
| |
Collapse
|
131
|
Zhou G, Loppnow H, Groth T. A macrophage/fibroblast co-culture system using a cell migration chamber to study inflammatory effects of biomaterials. Acta Biomater 2015; 26:54-63. [PMID: 26292266 DOI: 10.1016/j.actbio.2015.08.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/22/2015] [Accepted: 08/16/2015] [Indexed: 01/05/2023]
Abstract
Chronic inflammatory reactions hamper the use of biomaterials after implantation. Thus, the aim of the study was to develop a novel predictive in vitro macrophage/fibroblast co-culture model based on cell migration chambers that allows a timely and locally controlled interaction of both cell types to study the inflammatory responses of biomaterials in vitro. Here, self-assembled monolayers (SAMs) with different wettability and charge properties were used as model biomaterials on which co-cultures were established by use of fence chambers having internal and external compartments. This allowed establishing separated and mixed co-cultures of both cell types before and after removal of the chamber, respectively. The key advantages of this novel co-culture model included not only to establish a timely-resolved study of cytokine release, but also the ability to assess individual macrophage migration in both macrophage mono-cultures and co-cultures. All inflammatory reactions in terms of macrophage adhesion, macrophage migration, foreign body giant cell (FBGC) formation, β1 integrin expression and pro-inflammatory cytokine production were found strongly surface property dependent. The results show that the hydrophobic CH3 surface caused the strongest inflammatory reactions, whereas the hydrophilic/anionic COOH surface caused the least inflammatory response, indicating low and high biocompatibility of the surfaces, respectively. Most importantly, we found that both macrophage motility and directional movement were increased in the presence of fibroblasts in co-cultures compared with macrophage mono-cultures. Overall, the novel co-culture system provides access to a range of parameters for studying inflammatory reactions and reveals how material surface properties affect the inflammatory responses.
Collapse
|
132
|
Scislowska-Czarnecka A, Szmigiel D, Genet M, Dupont-Gillain C, Pamula E, Kolaczkowska E. Oxygen plasma surface modification augments poly(L-lactide-co-glycolide) cytocompatibility toward osteoblasts and minimizes immune activation of macrophages. J Biomed Mater Res A 2015; 103:3965-77. [DOI: 10.1002/jbm.a.35509] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 05/06/2015] [Accepted: 05/19/2015] [Indexed: 11/07/2022]
Affiliation(s)
| | - Dariusz Szmigiel
- Division of Silicon Microsystem and Nanostructure Technology; Institute of Electron Technology; Warsaw Poland
| | - Michel Genet
- Division of Bio & Soft Matter; Université Catholique De Louvain, Institute of Condensed Matter and Nanosciences; Louvain-la-Neuve 1348 Belgium
| | - Christine Dupont-Gillain
- Division of Bio & Soft Matter; Université Catholique De Louvain, Institute of Condensed Matter and Nanosciences; Louvain-la-Neuve 1348 Belgium
| | - Elzbieta Pamula
- Faculty of Materials Science and Ceramics, Department of Biomaterials; AGH University of Science and Technology; Krakow 30-059 Poland
| | - Elzbieta Kolaczkowska
- Department of Evolutionary Immunobiology; Jagiellonian University, Institute of Zoology; Krakow Poland
| |
Collapse
|
133
|
Wang Z, Liu N, Shi T, Zhou G, Wang Z, Gan J, Guo T, Qian H, Bao N, Zhao J. ER Stress Mediates TiAl6V4 Particle-Induced Peri-Implant Osteolysis by Promoting RANKL Expression in Fibroblasts. PLoS One 2015; 10:e0137774. [PMID: 26366858 PMCID: PMC4569331 DOI: 10.1371/journal.pone.0137774] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/20/2015] [Indexed: 12/12/2022] Open
Abstract
Wear particle-induced osteolysis is a major cause of aseptic loosening, which is one of the most common reasons for total hip arthroplasty (THA) failure. Previous studies have shown that the synovial fibroblasts present in the periprosthetic membrane are important targets of wear debris during osteolysis. However, the interaction mechanisms between the wear debris and fibroblasts remain largely unknown. In the present study, we investigated the effect of ER (endoplasmic reticulum) stress induced by TiAl6V4 particles (TiPs) in human synovial fibroblasts and calvarial resorption animal models. The expression of ER stress markers, including IRE1-α, GRP78/Bip and CHOP, were determined by western blot in fibroblasts that had been treated with TiPs for various times and concentration. To address whether ER stress was involved in the expression of RANKL, the effects of ER stress blockers (including 4-PBA and TUDCA) on the expression of RANKL in TiPs-treated fibroblasts were examined by real-time PCR, western blot and ELISA. Osteoclastogenesis was assessed by tartrate resistant acid phosphatase (TRAP) staining. Our study demonstrated that ER stress markers were markedly upregulated in TiPs-treated fibroblasts. Blocking ER stress significantly reduced the TiPs-induced expression of RANKL both in vitro and in vivo. Moreover, the inhibition of ER stress ameliorated wear particle-induced osteolysis in animal models. Taken together, these results suggested that the expression of RANKL induced by TiPs was mediated by ER stress in fibroblasts. Therefore, down regulating the ER stress of fibroblasts represents a potential therapeutic approach for wear particle-induced periprosthetic osteolysis.
Collapse
Affiliation(s)
- Zhenheng Wang
- Jinling Hospital, Department of Orthopaedics, School of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, China
| | - Naicheng Liu
- Jinling Hospital, Department of Orthopaedics, School of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, China
| | - Tongguo Shi
- Jinling Hospital, Department of Orthopaedics, School of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, China
| | - Gang Zhou
- Jinling Hospital, Department of Orthopaedics, School of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, China
| | - Zhenzhen Wang
- Jinling Hospital, Department of Orthopaedics, School of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, China
| | - Jingjing Gan
- Jinling Hospital, Department of Orthopaedics, School of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, China
| | - Ting Guo
- Jinling Hospital, Department of Orthopaedics, School of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, China
| | - Hongbo Qian
- Jinling Hospital, Department of Orthopaedics, School of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, China
- * E-mail: (HQ); (NB); (JZ)
| | - Nirong Bao
- Jinling Hospital, Department of Orthopaedics, School of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, China
- * E-mail: (HQ); (NB); (JZ)
| | - Jianning Zhao
- Jinling Hospital, Department of Orthopaedics, School of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, China
- * E-mail: (HQ); (NB); (JZ)
| |
Collapse
|
134
|
Sato T, Pajarinen J, Lin TH, Tamaki Y, Loi F, Egashira K, Yao Z, Goodman SB. NF-κB decoy oligodeoxynucleotide inhibits wear particle-induced inflammation in a murine calvarial model. J Biomed Mater Res A 2015; 103:3872-8. [PMID: 26123702 DOI: 10.1002/jbm.a.35532] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/08/2015] [Accepted: 06/23/2015] [Indexed: 12/23/2022]
Abstract
Wear particles induce periprosthetic inflammation and osteolysis through activation of nuclear factor kappa B (NF-κB), which up-regulates the downstream target gene expression for proinflammatory cytokines in macrophages. It was hypothesized that direct suppression of NF-κB activity in the early phases of this disorder could be a therapeutic strategy for preventing the inflammatory response to wear particles, potentially mitigating osteolysis. NF-κB activity can be suppressed via competitive binding with double stranded NF-κB decoy oligodeoxynucleotides (ODNs) that blocks this transcription factor from binding to the promoter regions of targeted genes. In this murine calvarial study, clinically relevant polyethylene particles (PEs) with/without ODN were subcutaneously injected over the calvarial bone. In the presence of PE particles, macrophages migrated to the inflammatory site and induced tumor necrosis factor alpha (TNF-α) and receptor activator of nuclear factor kappa B ligand (RANKL) expression, resulting in an increase in the number of osteoclasts. Local injections of ODN mitigated the expression of TNF-α, RANKL, and induced the expression of two anti-inflammatory, antiresorptive cytokines: interleukin-1 receptor antagonist and osteoprotegerin. Local intervention with NF-κB decoy ODN in early cases of particle-induced inflammation in which the prosthesis is still salvageable may potentially preserve periprosthetic bone stock.
Collapse
Affiliation(s)
- Taishi Sato
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Tzu-hua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Yasunobu Tamaki
- Department of Orthopaedic Surgery, Yamagata University School of Medicine, Yamagata, Japan
| | - Florence Loi
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Kensuke Egashira
- Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University, Fukuoka, Japan
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California.,Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
135
|
Kzhyshkowska J, Gudima A, Riabov V, Dollinger C, Lavalle P, Vrana NE. Macrophage responses to implants: prospects for personalized medicine. J Leukoc Biol 2015; 98:953-62. [PMID: 26168797 DOI: 10.1189/jlb.5vmr0415-166r] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/15/2015] [Indexed: 01/08/2023] Open
Abstract
Implants, transplants, and implantable biomedical devices are mainstream solutions for a wide variety of human pathologies. One of the persistent problems around nondegradable metallic and polymeric implants is failure of macrophages to resolve the inflammation and their tendency to stay in a state, named "frustrated phagocytosis." During the initial phase, proinflammatory macrophages induce acute reactions to trauma and foreign materials, whereas tolerogenic anti-inflammatory macrophages control resolution of inflammation and induce the subsequent healing stage. However, implanted materials can induce a mixed pro/anti-inflammatory phenotype, supporting chronic inflammatory reactions accompanied by microbial contamination and resulting in implant failure. Several materials based on natural polymers for improved interaction with host tissue or surfaces that release anti-inflammatory drugs/bioactive agents have been developed for implant coating to reduce implant rejection. However, no definitive, long-term solution to avoid adverse immune responses to the implanted materials is available to date. The prevention of implant-associated infections or chronic inflammation by manipulating the macrophage phenotype is a promising strategy to improve implant acceptance. The immunomodulatory properties of currently available implant coatings need to be improved to develop personalized therapeutic solutions. Human primary macrophages exposed to the implantable materials ex vivo can be used to predict the individual's reactions and allow selection of an optimal coating composition. Our review describes current understanding of the mechanisms of macrophage interactions with implantable materials and outlines the prospects for use of human primary macrophages for diagnostic and therapeutic approaches to personalized implant therapy.
Collapse
Affiliation(s)
- Julia Kzhyshkowska
- *Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany; Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia; Protip SAS, Strasbourg, France; Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, Strasbourg, France; and Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France
| | - Alexandru Gudima
- *Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany; Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia; Protip SAS, Strasbourg, France; Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, Strasbourg, France; and Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France
| | - Vladimir Riabov
- *Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany; Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia; Protip SAS, Strasbourg, France; Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, Strasbourg, France; and Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France
| | - Camille Dollinger
- *Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany; Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia; Protip SAS, Strasbourg, France; Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, Strasbourg, France; and Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France
| | - Philippe Lavalle
- *Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany; Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia; Protip SAS, Strasbourg, France; Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, Strasbourg, France; and Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France
| | - Nihal Engin Vrana
- *Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany; Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia; Protip SAS, Strasbourg, France; Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, Strasbourg, France; and Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
136
|
Qiu L, Xu R, Wang S, Li S, Sheng H, Wu J, Qu Y. Honokiol ameliorates endothelial dysfunction through suppression of PTX3 expression, a key mediator of IKK/IκB/NF-κB, in atherosclerotic cell model. Exp Mol Med 2015; 47:e171. [PMID: 26138903 PMCID: PMC4525296 DOI: 10.1038/emm.2015.37] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/04/2015] [Accepted: 03/11/2015] [Indexed: 12/13/2022] Open
Abstract
Pentraxin 3 (PTX3) was identified as a marker of the inflammatory response and overexpressed in various tissues and cells related to cardiovascular disease. Honokiol, an active component isolated from the Chinese medicinal herb Magnolia officinalis, was shown to have a variety of pharmacological activities. In the present study, we aimed to investigate the effects of honokiol on palmitic acid (PA)-induced dysfunction of human umbilical vein endothelial cells (HUVECs) and to elucidate potential regulatory mechanisms in this atherosclerotic cell model. Our results showed that PA significantly accelerated the expression of PTX3 in HUVECs through the IκB kinase (IKK)/IκB/nuclear factor-κB (NF-κB) pathway, reduced cell viability, induced cell apoptosis and triggered the inflammatory response. Knockdown of PTX3 supported cell growth and prevented apoptosis by blocking PA-inducted nitric oxide (NO) overproduction. Honokiol significantly suppressed the overexpression of PTX3 in PA-inducted HUVECs by inhibiting IκB phosphorylation and the expression of two NF-κB subunits (p50 and p65) in the IKK/IκB/NF-κB signaling pathway. Furthermore, honokiol reduced endothelial cell injury and apoptosis by regulating the expression of inducible NO synthase and endothelial NO synthase, as well as the generation of NO. Honokiol showed an anti-inflammatory effect in PA-inducted HUVECs by significantly inhibiting the generation of interleukin-6 (IL-6), IL-8 and monocyte chemoattractant protein-1. In summary, honokiol repaired endothelial dysfunction by suppressing PTX3 overexpression in an atherosclerotic cell model. PTX3 may be a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Ling Qiu
- Geriatrics Department, Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Rong Xu
- Geriatrics Department, Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Siyang Wang
- Geriatrics Department, Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Shuijun Li
- Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Hongguang Sheng
- Endocrinology Department, Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Jiaxi Wu
- Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Yi Qu
- Geriatrics Department, Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| |
Collapse
|
137
|
Cang D, Guo K, Zhao F. Dendritic cells enhance UHMWPE wear particle-induced osteoclast differentiation of macrophages. J Biomed Mater Res A 2015; 103:3349-54. [PMID: 25808788 DOI: 10.1002/jbm.a.35459] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 03/03/2015] [Accepted: 03/13/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Dingwei Cang
- Xuzhou Medical College; Xuzhou Jiangsu 221004 People's Republic of China
| | - Kaijin Guo
- The Affiliate Hospital of Xuzhou Medical College; Xuzhou Jiangsu 221004 China
| | - Fengchao Zhao
- The Affiliate Hospital of Xuzhou Medical College; Xuzhou Jiangsu 221004 China
| |
Collapse
|
138
|
Endoplasmic reticulum stress-mediated inflammatory signaling pathways within the osteolytic periosteum and interface membrane in particle-induced osteolysis. Cell Tissue Res 2015; 363:427-47. [PMID: 26004143 PMCID: PMC4735257 DOI: 10.1007/s00441-015-2205-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/23/2015] [Indexed: 12/29/2022]
Abstract
Aseptic loosening secondary to periprosthetic inflammatory osteolysis results from the biological response to wear particles and is a leading cause of arthroplasty failure. The origin of this inflammatory response remains unclear. We aim to validate the definite link between endoplasmic reticulum (ER) stress and particle-induced inflammatory signaling pathways in periprosthetic osteolysis. We examine the histopathologic changes of osteolysis and the expression of specific biomarkers for ER-stress-mediated inflammatory signaling pathways (IRE1α, GRP78/Bip, c-Fos, NF-κB, ROS and Ca(2+)). Moreover, pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) and osteoclastogenic molecules (VEGF, OPG, RANKL and M-CSF) were assessed in clinical interface membranes and murine periosteum tissues. We found wear particles to be capable of inducing ER stress in macrophages within clinical osteolytic interface membranes and murine osteolytic periosteum tissues and to be associated with the inflammatory response and osteoclastogenesis. Blocking ER stress with sodium 4-phenylbutyrate (4-PBA) results in a dramatic amelioration of particle-induced osteolysis and a significant reduction of ER-stress intensity. Simultaneously, this ER-stress blocker also lessens inflammatory cell infiltration, diminishes the capability of osteoclastogenesis and reduces the inflammatory response by lowering IRE1α, GRP78/Bip, c-Fos, NF-κB, ROS and Ca(2+) levels. Thus, ER stress plays an important role in particle-induced inflammatory osteolysis and osteoclastogenic reactions. The pharmacological targeting of ER-stress-mediated inflammatory signaling pathways might be an appealing approach for alleviating or preventing particle-induced osteolysis in at-risk patients.
Collapse
|
139
|
Bhardwaj G, Yazici H, Webster TJ. Reducing bacteria and macrophage density on nanophase hydroxyapatite coated onto titanium surfaces without releasing pharmaceutical agents. NANOSCALE 2015; 7:8416-8427. [PMID: 25876524 DOI: 10.1039/c5nr00471c] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Reducing bacterial density on titanium implant surfaces has been a major concern because of the increasing number of nosocomial infections. Controlling the inflammatory response post implantation has also been an important issue for medical devices due to the detrimental effects of chronic inflammation on device performance. It has recently been demonstrated that manipulating medical device surface properties including chemistry, roughness and wettability can control both infection and inflammation. Here, we synthesized nanophase (that is, materials with one dimension in the nanoscale) hydroxyapatite coatings on titanium to reduce bacterial adhesion and inflammatory responses (as measured by macrophage functions) and compared such results to bare titanium and plasma sprayed hydroxyapatite titanium coated surfaces used clinically today. This approach is a pharmaceutical-free approach to inhibit infection and inflammation due to the detrimental side effects of any drug released in the body. Here, nanophase hydroxyapatite was synthesized in sizes ranging from 110-170 nm and was subsequently coated onto titanium samples using electrophoretic deposition. Results indicated that smaller nanoscale hydroxyapatite features on titanium surfaces alone decreased bacterial attachment in the presence of gram negative (P. aeruginosa), gram positive (S. aureus) and ampicillin resistant gram-negative (E. coli) bacteria as well as were able to control inflammatory responses; properties which should lead to their further investigation for improved medical applications.
Collapse
Affiliation(s)
- Garima Bhardwaj
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA.
| | | | | |
Collapse
|
140
|
Córdova LA, Trichet V, Escriou V, Rosset P, Amiaud J, Battaglia S, Charrier C, Berreur M, Brion R, Gouin F, Layrolle P, Passuti N, Heymann D. Inhibition of osteolysis and increase of bone formation after local administration of siRNA-targeting RANK in a polyethylene particle-induced osteolysis model. Acta Biomater 2015; 13:150-8. [PMID: 25462844 DOI: 10.1016/j.actbio.2014.10.042] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 10/25/2014] [Accepted: 10/28/2014] [Indexed: 01/24/2023]
Abstract
Receptor activator of nuclear factor kappa-B (RANK) and RANK-ligand are relevant targets for the treatment of polyethylene particle-induced osteolysis. This study assessed the local administration of siRNA, targeting both human RANK and mouse Rank transcripts in a mouse model. Four groups of mice were implanted with polyethylene (PE) particles in the calvaria and treated locally with 2.5, 5 and 10 μg of RANK siRNA or a control siRNA delivered by the cationic liposome DMAPAP/DOPE. The tissues were harvested at day 9 after surgery and evaluated by micro-computed tomography, tartrate-resistant acid phosphatase (TRAP) immunohistochemistry for macrophages and osteoblasts, and gene relative expression of inflammatory and osteolytic markers. 10 μg of RANK siRNA exerted a protective effect against PE particle-induced osteolysis, decreasing the bone loss and the osteoclastogenesis, demonstrated by the significant increase in the bone volume (P<0.001) and by the reduction in both the number of TRAP(+) cells and osteoclast activity (P<0.01). A bone anabolic effect demonstrated by the formation of new trabecular bone was confirmed by the increased immunopositive staining for osteoblast-specific proteins. In addition, 5 and 10 μg of RANK siRNA downregulated the expression of pro-inflammatory cytokines (P<0.01) without depletion of macrophages. Our findings show that RANK siRNA delivered locally by a synthetic vector may be an effective approach for reducing osteolysis and may even stimulate bone formation in aseptic loosening of prosthetic implants.
Collapse
Affiliation(s)
- L A Córdova
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; Department of Oral and Maxillofacial Surgery, San Borja Arriaran University Hospital, Faculty of Dentistry, University of Chile-CONICYT, Sergio Livingstone Polhammer 943, Independencia, Santiago, Chile.
| | - V Trichet
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France
| | - V Escriou
- UTCBS CNRS UMR 8258 INSERM UMR-S 1022, 4 avenue de l'Observatoire, 75006 Paris, France
| | - P Rosset
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; Tours University Hospital, François Rabelais, 37044 Tours Cedex 9, France
| | - J Amiaud
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France
| | - S Battaglia
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France
| | - C Charrier
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France
| | - M Berreur
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France
| | - R Brion
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; Nantes University Hospital, 1 place Alexis-Ricordeau, 44093 Nantes Cedex 1, France
| | - F Gouin
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; Nantes University Hospital, 1 place Alexis-Ricordeau, 44093 Nantes Cedex 1, France
| | - P Layrolle
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France
| | - N Passuti
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; Nantes University Hospital, 1 place Alexis-Ricordeau, 44093 Nantes Cedex 1, France
| | - D Heymann
- INSERM, UMR 957, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; University of Nantes, Nantes Atlantique Universities, Laboratory of Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France; Nantes University Hospital, 1 place Alexis-Ricordeau, 44093 Nantes Cedex 1, France
| |
Collapse
|
141
|
Lin TH, Sato T, Barcay KR, Waters H, Loi F, Zhang R, Pajarinen J, Egashira K, Yao Z, Goodman SB. NF-κB decoy oligodeoxynucleotide enhanced osteogenesis in mesenchymal stem cells exposed to polyethylene particle. Tissue Eng Part A 2015; 21:875-83. [PMID: 25518013 DOI: 10.1089/ten.tea.2014.0144] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Excessive generation of wear particles after total joint replacement may lead to local inflammation and periprosthetic osteolysis. Modulation of the key transcription factor NF-κB in immune cells could potentially mitigate the osteolytic process. We previously showed that local delivery of ultrahigh-molecular-weight polyethylene (UHMWPE) particles recruited osteoprogenitor cells and reduced osteolysis. However, the biological effects of modulating the NF-κB signaling pathway on osteoprogenitor/mesenchymal stem cells (MSCs) remain unclear. Here we showed that decoy oligodeoxynucleotide (ODN) increased cell viability when primary murine MSCs were exposed to UHMWPE particles, but had no effects on cellular apoptosis. Decoy ODN increased transforming growth factor-beta 1 (TGF-β1) and osteoprotegerin (OPG) in MSCs exposed to UHMWPE particles. Mechanistic studies showed that decoy ODN upregulated OPG expression through a TGF-β1-dependent pathway. By measuring the alkaline phosphatase activity, osteocalcin levels, Runx2 and osteopontin expression, and performing a bone mineralization assay, we found that decoy ODN increased MSC osteogenic ability when the cells were exposed to UHMWPE particles. Furthermore, the cellular response to decoy ODN and UHMWPE particles with regard to cell phenotype, cell viability, and osteogenic ability was confirmed using primary human MSCs. Our results suggest that modulation of wear particle-induced inflammation by NF-κB decoy ODN had no adverse effects on MSCs and may potentially further mitigate periprosthetic osteolysis by protecting MSC viability and osteogenic ability.
Collapse
Affiliation(s)
- Tzu-Hua Lin
- 1 Department of Orthopaedic Surgery, Stanford University , Stanford, California
| | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Wear Particles Impair Antimicrobial Activity Via Suppression of Reactive Oxygen Species Generation and ERK1/2 Phosphorylation in Activated Macrophages. Inflammation 2015; 38:1289-96. [DOI: 10.1007/s10753-014-0099-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
143
|
Choy J, Albers CE, Siebenrock KA, Dolder S, Hofstetter W, Klenke FM. Incorporation of RANKL promotes osteoclast formation and osteoclast activity on β-TCP ceramics. Bone 2014; 69:80-8. [PMID: 25245204 DOI: 10.1016/j.bone.2014.09.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 09/11/2014] [Accepted: 09/12/2014] [Indexed: 01/01/2023]
Abstract
β-Tricalcium phosphate (β-TCP) ceramics are approved for the repair of osseous defects. In large defects, however, the substitution of the material by authentic bone is inadequate to provide sufficient long-term mechanical stability. We aimed to develop composites of β-TCP ceramics and receptor activator of nuclear factor κ-B ligand (RANKL) to enhance the formation of osteoclasts and promote cell mediated calcium phosphate resorption. RANKL was adsorbed superficially onto β-TCP ceramics or incorporated into a crystalline layer of calcium phosphate by the use of a co-precipitation technique. Murine osteoclast precursors were seeded onto the ceramics. After 15 days, the formation of osteoclasts was quantified cytologically and colorimetrically with tartrate-resistant acidic phosphatase (TRAP) staining and TRAP activity measurements, respectively. Additionally, the expression of transcripts encoding the osteoclast gene products cathepsin K, calcitonin receptor, and of the sodium/hydrogen exchanger NHA2 were quantified by real-time PCR. The activity of newly formed osteoclasts was evaluated by means of a calcium phosphate resorption assay. Superficially adsorbed RANKL did not induce the formation of osteoclasts on β-TCP ceramics. When co-precipitated onto β-TCP ceramics RANKL supported the formation of mature osteoclasts. The development of osteoclast lineage cells was further confirmed by the increased expression of cathepsin K, calcitonin receptor, and NHA2. Incorporated RANKL stimulated the cells to resorb crystalline calcium phosphate. Our in vitro study shows that RANKL incorporated into β-TCP ceramics induces the formation of active, resorbing osteoclasts on the material surface. Once formed, osteoclasts mediate the release of RANKL thereby perpetuating their differentiation and activation. In vivo, the stimulation of osteoclast-mediated resorption may contribute to a coordinated sequence of material resorption and bone formation. Further in vivo studies are needed to confirm the current in vitro findings.
Collapse
Affiliation(s)
- John Choy
- Group for Bone Biology and Orthopedic Research, Department of Clinical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Christoph E Albers
- Department of Orthopaedic Surgery, Inselspital, Bern University Hospital, CH-3010 Bern, Switzerland
| | - Klaus A Siebenrock
- Department of Orthopaedic Surgery, Inselspital, Bern University Hospital, CH-3010 Bern, Switzerland
| | - Silvia Dolder
- Group for Bone Biology and Orthopedic Research, Department of Clinical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Wilhelm Hofstetter
- Group for Bone Biology and Orthopedic Research, Department of Clinical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Frank M Klenke
- Department of Orthopaedic Surgery, Inselspital, Bern University Hospital, CH-3010 Bern, Switzerland.
| |
Collapse
|
144
|
Pajarinen J, Lin TH, Sato T, Yao Z, Goodman SB. Interaction of Materials and Biology in Total Joint Replacement - Successes, Challenges and Future Directions. J Mater Chem B 2014; 2:7094-7108. [PMID: 25541591 PMCID: PMC4273175 DOI: 10.1039/c4tb01005a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Total joint replacement (TJR) has revolutionized the treatment of end-stage arthritic disorders. This success is due, in large part, to a clear understanding of the important interaction between the artificial implant and the biology of the host. All surgical procedures in which implants are placed in the body evoke an initial inflammatory reaction, which generally subsides over several weeks. Thereafter, a series of homeostatic events occur leading to progressive integration of the implant within bone and the surrounding musculoskeletal tissues. The eventual outcome of the operation is dependent on the characteristics of the implant, the precision of the surgical technique and operative environment, and the biological milieu of the host. If these factors and events are not optimal, adverse events can occur such as the development of chronic inflammation, progressive bone loss due to increased production of degradation products from the implant (periprosthetic osteolysis), implant loosening or infection. These complications can lead to chronic pain and poor function of the joint reconstruction, and may necessitate revision surgery or removal of the prosthesis entirely. Recent advances in engineering, materials science, and the immunological aspects associated with orthopaedic implants have fostered intense research with the hope that joint replacements will last a lifetime, and facilitate pain-free, normal function.
Collapse
Affiliation(s)
- J Pajarinen
- Department of Orthopaedic Surgery, Orthopaedic Surgery Laboratories, Stanford University, Stanford, CA, USA
| | - T-H Lin
- Department of Orthopaedic Surgery, Orthopaedic Surgery Laboratories, Stanford University, Stanford, CA, USA
| | - T Sato
- Department of Orthopaedic Surgery, Orthopaedic Surgery Laboratories, Stanford University, Stanford, CA, USA
| | - Z Yao
- Department of Orthopaedic Surgery, Orthopaedic Surgery Laboratories, Stanford University, Stanford, CA, USA
| | - S B Goodman
- Department of Orthopaedic Surgery, Orthopaedic Surgery Laboratories, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Orthopaedic Surgery Laboratories, Stanford University, Stanford, CA, USA
| |
Collapse
|
145
|
Comparison of periprosthetic tissues in knee and hip joints: differential expression of CCL3 and DC-STAMP in total knee and hip arthroplasty and similar cytokine profiles in primary knee and hip osteoarthritis. Osteoarthritis Cartilage 2014; 22:1851-60. [PMID: 25151085 DOI: 10.1016/j.joca.2014.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 07/11/2014] [Accepted: 08/03/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To identify expression profiles (EP) associated with aseptic loosening of total knee arthroplasty (TKA) and to compare them with EP observed in total hip arthroplasty (THA), and primary knee and hip osteoarthritis (OA). DESIGN Gene EP of TNF, IL-6, IL-8, CHIT1, BMP4, CCL3, CCL18, MMP9, RANKL, OPG, DC-STAMP and SOCS3 were assessed using quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) on tissues retrieved from patients with aseptically failed TKA (n = 21), THA (n = 41) and primary knee (n = 20) and hip (n = 17) OA. Immunohistochemistry was applied to localize the proteins. RESULTS When compared to knee OA, the pseudosynovial tissue in TKA exhibit (1) elevation of alternative macrophage activation marker (CHIT1), chemokine (IL-8), and a proteolytic enzyme (MMP9); (2) downregulation of pro-inflammatory cytokine (TNF), osteoclastic regulator (OPG) and a stimulator of bone formation (BMP4); (3) no difference in IL-6, CCL3, CCL18, RANKL, DC-STAMP and SOCS3. The EP in TKA differed from EP in aseptically failed THA by lower CCL3 and DC-STAMP mRNA and protein expression. EP of all studied inflammatory and osteoclastogenic molecules were similar in knee and hip OA. CONCLUSIONS Comparing to OA, aseptic loosening of TKA is associated with upregulated expression of CHIT1, IL-8 and MMP9, dysregulated RANKL:OPG ratio and low levels of inflammatory cytokines. Similar cytokine profiles were associated with primary knee and hip OA. Further research is required to explain the differences in CCL3 and DC-STAMP expression between failed TKA and THA.
Collapse
|
146
|
Lin TH, Goodman SB. Suppression of NF-κB signaling mitigates polyethylene wear particle-induced inflammatory response. INFLAMMATION AND CELL SIGNALING 2014; 1. [PMID: 26052541 DOI: 10.14800/ics.223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In end-stage arthritis patients, total joint replacement is a very effective surgical procedure. Nevertheless, the high revision rate after surgery remains a major concern. The wear particles generated from biomaterial-induced tissue responses may lead to chronic inflammation and local bone destruction (periprosthetic osteolysis). Several important signaling pathways are involved in wear particles induced inflammatory reactions, including the transcription factor NF-κB. We recently reported that RAW264.7 macrophage cell exposure to ultra-high molecular weight polyethylene (UHMWPE) particles significantly increased the NF-κB activity in a generated NF-κB responsive luciferase reporter cell clone. The NF-κB activity induced by UHMWPE particles in a mouse RAW264.7 macrophage cell line, bone marrow derived macrophages, and human THP1 macrophage cell line, were suppressed by double strand decoy oligodeoxynucleotide (ODN) containing an NF-κB binding element. Macrophages exposure to UHMWPE particles with or without endotoxin induced pro-inflammatory cytokine and chemokine expression including TNF-α, MCP1, MIP1α, and others. Finally, the decoy ODN significantly suppressed the induced cytokine and chemokine expression in both murine and human macrophages, consequently reducing macrophage recruitment by cellular conditioned medium exposed to wear particles. These findings suggest that local suppression of inflammatory cytokine production via inhibition of NF-κB activity with decoy ODN in total joint replacement patients could potentially be an effective strategy to alleviate wear particle-induced chronic inflammation.
Collapse
Affiliation(s)
- Tzu-Hua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA ; Department of Bioengineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
147
|
Drynda A, Singh G, Buchhorn GH, Awiszus F, Ruetschi M, Feuerstein B, Kliche S, Lohmann CH. Metallic wear debris may regulate CXCR4 expression in vitro and in vivo. J Biomed Mater Res A 2014; 103:1940-8. [PMID: 25205627 DOI: 10.1002/jbm.a.35330] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/25/2014] [Accepted: 09/05/2014] [Indexed: 11/05/2022]
Abstract
CXCR4, the chemokine receptor for CXCL12, also known as SDF-1 (stromal cell derived factor-1), has been shown to play a pivotal role in bone metastasis, inflammatory, and autoimmune conditions but has not been investigated in periprosthetic osteolysis. We co-cultured osteoblast-like cells with increasing concentrations of metallic (Co-35Ni-20Cr-10Mo and Co-28Cr-6Mo) and Co-ions simulating wear debris. Real-time polymerase chain reaction (RT-PCR) and Western blotting were used to quantify gene and protein expression of CXCR4. The expression of tumor necrosis factor-alpha (TNF-α) and the effects of AMD3100 (bicyclam) on both CXCR4 and TNF-α expression among these cells was investigated. RT-PCR showed an increase in CXCR4 mRNA (7.5-fold for MG63 and 4.0-fold for SaOs-2 cells) among cells co-cultured with metal alloy particles. Western blotting showed a time-dependent increase in protein expression of CXCR4. The attempted blockade of CXCR4 by its known competitive receptor agonist AMD3100 led to a significant inhibition TNF-α mRNA expression. Immunohistochemistry showed CXCR4 positivity among patients with failed metal-on-metal hip replacements and radiographic evidence of osteolysis. Our data collectively suggest that the CXCR4 chemokine is upregulated in a dose- and time-dependent manner in the presence of metallic wear debris.
Collapse
Affiliation(s)
- Andreas Drynda
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Yu M, Zhou K, Li Z, Zhang D. Preparation, characterization and in vitro gentamicin release of porous HA microspheres. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2014; 45:306-12. [PMID: 25491833 DOI: 10.1016/j.msec.2014.08.075] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 07/15/2014] [Accepted: 08/31/2014] [Indexed: 01/28/2023]
Abstract
Hydroxyapatite (HA) microspheres with high porosities were successfully obtained using an improved ice-templated spray drying (ITSD) technique for drug delivery applications. Pore structures and pore sizes of microspheres have great impact on drug loading and release kinetics. Therefore, solvent types, polyvinyl alcohol (PVA) contents and solid loadings of suspensions were adjusted to control the pore structures and pore sizes. Microspheres with interconnected pore networks and aligned pore structures were obtained using camphene-based and tert-butyl alcohol (TBA)-based suspensions, respectively. With the increase of PVA contents in suspensions, the growth of sintering neck became more obvious and the surface of HA particles became smoother. The inner pore structures of microspheres transformed from uniformly distributed cellular pores to three-dimensional interconnected pore networks, with the increase of solid loadings in suspensions. Gentamicin was successfully loaded into porous HA microspheres. The drug loading percentage increased from 40.59 to 49.82% with the increase of porosity of HA microspheres. The release percentage during the initial 18 h increased from 48.72 to 65.68% with the transformation of pore structures from independent cellular pores (main diameter~3 μm) to three-dimensional interconnected pore networks (main diameter>3 μm).
Collapse
Affiliation(s)
- Min Yu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan 410083, PR China
| | - Kechao Zhou
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan 410083, PR China
| | - Zhiyou Li
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan 410083, PR China
| | - Dou Zhang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan 410083, PR China.
| |
Collapse
|
149
|
Lin TH, Yao Z, Sato T, Keeney M, Li C, Pajarinen J, Yang F, Egashira K, Goodman SB. Suppression of wear-particle-induced pro-inflammatory cytokine and chemokine production in macrophages via NF-κB decoy oligodeoxynucleotide: a preliminary report. Acta Biomater 2014; 10:3747-55. [PMID: 24814879 DOI: 10.1016/j.actbio.2014.04.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/01/2014] [Accepted: 04/29/2014] [Indexed: 12/28/2022]
Abstract
Total joint replacement (TJR) is very cost-effective surgery for end-stage arthritis. One important goal is to decrease the revision rate, mainly because TJR has been extended to younger patients. Continuous production of ultra-high molecular weight polyethylene (UHMWPE) wear particles induces macrophage infiltration and chronic inflammation, which can lead to periprosthetic osteolysis. Targeting individual pro-inflammatory cytokines directly has not reversed the osteolytic process in clinical trials, owing to compensatory up-regulation of other pro-inflammatory factors. It is hypothesized that targeting the important transcription factor NF-κB could mitigate the inflammatory response to wear particles, potentially diminishing osteolysis. In the current study, NF-κB activity in mouse RAW 264.7 and human THP1 macrophage cell lines, as well as primary mouse and human macrophages, was suppressed via competitive binding with double strand decoy oligodeoxynucleotide (ODN) containing an NF-κB binding element. It was found that macrophage exposure to UHMWPE particles induced multiple pro-inflammatory cytokine and chemokine expression, including TNF-α, MCP1, MIP1α and others. Importantly, the decoy ODN significantly suppressed the induced cytokine and chemokine expression in both murine and human macrophages, and resulted in suppression of macrophage recruitment. The strategic use of decoy NF-κB ODN, delivered locally, could potentially diminish particle-induced periprosthetic osteolysis.
Collapse
|
150
|
Tian B, Jiang T, Shao Z, Zhai Z, Li H, Fan Q, Liu X, Ouyang Z, Tang T, Jiang Q, Zheng M, Dai K, Qin A, Yu Y, Zhu Z. The prevention of titanium-particle-induced osteolysis by OA-14 through the suppression of the p38 signaling pathway and inhibition of osteoclastogenesis. Biomaterials 2014; 35:8937-50. [PMID: 25086794 DOI: 10.1016/j.biomaterials.2014.06.055] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 06/29/2014] [Indexed: 12/29/2022]
Abstract
Wear-particle-induced osteolysis leads to prosthesis loosening, which is one of the most common causes of joint-implant failure, a problem that must be fixed using revision surgery. Thus, a potential treatment for prosthetic loosening is focused on inhibiting osteoclastic bone resorption, which prevents wear-particle-induced osteolysis. In this study, we synthesized a compound named OA-14 (N-(3- (dodecylcarbamoyl)phenyl)-1H-indole-2-carboxamide) and examined how OA-14 affects titanium (Ti)-particle-induced osteolysis and osteoclastogenesis. We report that OA-14 treatment protected against Ti-particle-induced osteolysis in a mouse calvarial model. Interestingly, the number of tartrate-resistant acid phosphatase-positive osteoclasts decreased after treatment with OA-14 in vivo, which suggested that OA-14 inhibits osteoclast formation. To test this hypothesis, we conducted in vitro studies, and our results revealed that OA-14 markedly diminished osteoclast differentiation and osteoclast-specific gene expression in a dose- and time-dependent manner. Moreover, OA-14 suppressed osteoclastic bone resorption and F-actin ring formation. Furthermore, we determined that OA-14 inhibited osteoclastogenesis by specifically blocking the p38-Mitf-c-fos-NFATc1 signaling cascade induced by RANKL (ligand of receptor activator of nuclear factor κB). Collectively, our results suggest that the compound OA-14 can be safely used for treating particle-induced peri-implant osteolysis and other diseases caused by excessive osteoclast formation and function.
Collapse
Affiliation(s)
- Bo Tian
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tao Jiang
- The Center of Diagnosis and Treatment for Joint Disease, Drum Tower Clinical Medical College of Nanjing Medical University, Jiangsu, PR China
| | - Zhanying Shao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Zanjing Zhai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Haowei Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qiming Fan
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Xuqiang Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhengxiao Ouyang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qing Jiang
- The Center of Diagnosis and Treatment for Joint Disease, Drum Tower Clinical Medical College of Nanjing Medical University, Jiangsu, PR China
| | - Minghao Zheng
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, Australia
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - An Qin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, Australia.
| | - Yongping Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China.
| | - Zhenan Zhu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|