1
|
Soliman E, Leonard J, Basso EKG, Gershenson I, Ju J, Mills J, de Jager C, Kaloss AM, Elhassanny M, Pereira D, Chen M, Wang X, Theus MH. Efferocytosis is restricted by axon guidance molecule EphA4 via ERK/Stat6/MERTK signaling following brain injury. J Neuroinflammation 2023; 20:256. [PMID: 37941008 PMCID: PMC10633953 DOI: 10.1186/s12974-023-02940-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Efferocytosis is a process that removes apoptotic cells and cellular debris. Clearance of these cells alleviates neuroinflammation, prevents the release of inflammatory molecules, and promotes the production of anti-inflammatory cytokines to help maintain tissue homeostasis. The underlying mechanisms by which this occurs in the brain after injury remain ill-defined. METHODS We used GFP bone marrow chimeric knockout (KO) mice to demonstrate that the axon guidance molecule EphA4 receptor tyrosine kinase is involved in suppressing MERTK in the brain to restrict efferocytosis of resident microglia and peripheral-derived monocyte/macrophages. RESULTS Single-cell RNAseq identified MERTK expression, the primary receptor involved in efferocytosis, on monocytes, microglia, and a subset of astrocytes in the damaged cortex following brain injury. Loss of EphA4 on infiltrating GFP-expressing immune cells improved functional outcome concomitant with enhanced efferocytosis and overall protein expression of p-MERTK, p-ERK, and p-Stat6. The percentage of GFP+ monocyte/macrophages and resident microglia engulfing NeuN+ or TUNEL+ cells was significantly higher in KO chimeric mice. Importantly, mRNA expression of Mertk and its cognate ligand Gas6 was significantly elevated in these mice compared to the wild-type. Analysis of cell-specific expression showed that p-ERK and p-Stat6 co-localized with MERTK-expressing GFP + cells in the peri-lesional area of the cortex following brain injury. Using an in vitro efferocytosis assay, co-culturing pHrodo-labeled apoptotic Jurkat cells and bone marrow (BM)-derived macrophages, we demonstrate that efferocytosis efficiency and mRNA expression of Mertk and Gas6 was enhanced in the absence of EphA4. Selective inhibitors of ERK and Stat6 attenuated this effect, confirming that EphA4 suppresses monocyte/macrophage efferocytosis via inhibition of the ERK/Stat6 pathway. CONCLUSIONS Our findings implicate the ERK/Stat6/MERTK axis as a novel regulator of apoptotic debris clearance in brain injury that is restricted by peripheral myeloid-derived EphA4 to prevent the resolution of inflammation.
Collapse
Affiliation(s)
- Eman Soliman
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - John Leonard
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA
| | | | - Ilana Gershenson
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Jing Ju
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Jatia Mills
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Caroline de Jager
- Translational Biology Medicine and Health Graduate Program, Roanoke, VA, 24001, USA
| | - Alexandra M Kaloss
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Mohamed Elhassanny
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Daniela Pereira
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Michael Chen
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Xia Wang
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, 24061, USA.
- Translational Biology Medicine and Health Graduate Program, Roanoke, VA, 24001, USA.
- Center for Engineered Health, Virginia Tech, Blacksburg, VA, 24061, USA.
- VT-Biomedical Engineering and School of Neuroscience, 970 Washington Street SW, Life Sciences I; Rm 249 (MC0910), Blacksburg, VA, 24061, USA.
| |
Collapse
|
2
|
Xie Y, Peng Y, Fu G, Jin J, Wang S, Li M, Zheng Q, Lyu FJ, Deng Z, Ma Y. Nano wear particles and the periprosthetic microenvironment in aseptic loosening induced osteolysis following joint arthroplasty. Front Cell Infect Microbiol 2023; 13:1275086. [PMID: 37854857 PMCID: PMC10579613 DOI: 10.3389/fcimb.2023.1275086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/05/2023] [Indexed: 10/20/2023] Open
Abstract
Joint arthroplasty is an option for end-stage septic arthritis due to joint infection after effective control of infection. However, complications such as osteolysis and aseptic loosening can arise afterwards due to wear and tear caused by high joint activity after surgery, necessitating joint revision. Some studies on tissue pathology after prosthesis implantation have identified various cell populations involved in the process. However, these studies have often overlooked the complexity of the altered periprosthetic microenvironment, especially the role of nano wear particles in the etiology of osteolysis and aseptic loosening. To address this gap, we propose the concept of the "prosthetic microenvironment". In this perspective, we first summarize the histological changes in the periprosthetic tissue from prosthetic implantation to aseptic loosening, then analyze the cellular components in the periprosthetic microenvironment post prosthetic implantation. We further elucidate the interactions among cells within periprosthetic tissues, and display the impact of wear particles on the disturbed periprosthetic microenvironments. Moreover, we explore the origins of disease states arising from imbalances in the homeostasis of the periprosthetic microenvironment. The aim of this review is to summarize the role of relevant factors in the microenvironment of the periprosthetic tissues, in an attempt to contribute to the development of innovative treatments to manage this common complication of joint replacement surgery.
Collapse
Affiliation(s)
- Yu Xie
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Yujie Peng
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Guangtao Fu
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiewen Jin
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuai Wang
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Mengyuan Li
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Feng-Juan Lyu
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Soliman E, Leonard J, Basso EK, Gershenson I, Ju J, Mills J, Jager C, Kaloss AM, Elhassanny M, Pereira D, Chen M, Wang X, Theus MH. Efferocytosis is restricted by axon guidance molecule EphA4 via ERK/Stat6/Mertk signaling following brain injury. RESEARCH SQUARE 2023:rs.3.rs-3079466. [PMID: 37461720 PMCID: PMC10350120 DOI: 10.21203/rs.3.rs-3079466/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Background Efferocytosis is a process that removes apoptotic cells and cellular debris. Clearance of these cells alleviates neuroinflammation and prevents the release of inflammatory molecules and promotes the production of anti-inflammatory cytokines to help maintain tissue homeostasis. The underlying mechanisms by which this occurs in the brain after injury remains ill-defined. Methods We demonstrate using GFP bone marrow chimeric knockout (KO) mice, that the axon guidance molecule EphA4 receptor tyrosine kinase is involved in suppressing Mertk signaling in the brain to restrict the function of efferocytosis on resident microglia and peripheral-derived monocyte/macrophages. Results Single-cell RNAseq identified Mertk expression, the primary receptor involved in efferocytosis, on monocytes, microglia, and a subset of astrocytes in the damaged cortex following brain injury. Loss of EphA4 on infiltrating GFP-expressing immune cells improved functional outcome concomitant with enhanced efferocytosis, and overall protein expression of p-Mertk, p-ERK, and p-Stat6. The percentage of GFP+ monocyte/macrophages and resident microglia engulfing NeuN+ or TUNEL+ cells was significantly higher in KO chimeric mice. Importantly, mRNA expression of Mertk and its cognate ligand Gas6 was significantly elevated in these mice compared to wild-type. Analysis of cell-specific expression showed that p-ERK and p-Stat6 co-localized with Mertk-expressing GFP + cells in the peri-lesional area of the cortex following brain injury. Using an in vitro efferocytosis assay, co-culturing pHrodo-labeled apoptotic Jurkat cells and bone marrow (BM)-derived macrophages, we demonstrate that efferocytosis efficiency and mRNA expression of Mertk and Gas6 was enhanced in the absence of EphA4. Select inhibitors of ERK and Stat6 attenuated this effect confirming that EphA4 suppresses monocyte/macrophage efferocytosis via inhibition of the ERK/Stat6 pathway. Conclusions Our findings implicate the Mertk/ERK/Stat6 axis as a novel regulator of apoptotic debris clearance in brain injury that is restricted by peripheral myeloid-derived EphA4 to prevent the resolution of inflammation.
Collapse
|
4
|
Yang B, Qin Y, Zhang A, Wang P, Jiang H, Shi Y, You G, Shen D, Ni S, Guo L, Liu Y. Circular RNA CircSLC8A1 contributes to osteogenic differentiation in hBMSCs via CircSLC8A1/miR-144-3p/RUNX1 in periprosthetic osteolysis. J Cell Mol Med 2022; 27:189-203. [PMID: 36541023 PMCID: PMC9843530 DOI: 10.1111/jcmm.17633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/03/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022] Open
Abstract
Circular RNAs (circRNAs) are often found in eukaryocyte and have a role in the pathogenesis of a variety of human disorders. Our related research has shown the differential expression of circRNAs in periprosthetic osteolysis (PPOL). However, the involvement of circRNAs in the exact process is yet unknown. CircSLC8A1 expression was evaluated in clinical samples and human bone marrow mesenchymal stem cells (hBMSCs) in this investigation using quantitative real-time PCR. In vitro and in vivo studies were conducted to explicate its functional role and pathway. We demonstrated CircSLC8A1 is involved in PPOL using gain- and loss-of-function methods. The association of CircSLC8A1 and miR-144-3p, along with miR-144-3p and RUNX1, was predicted using bioinformatics. RNA pull-down and luciferase assays confirmed it. The impact of CircSLC8A1 in the PPOL-mouse model was also investigated using adeno-associated virus. CircSLC8A1 was found to be downregulated in PPOL patients' periprosthetic tissues. Overexpression of CircSLC8A1 promoted osteogenic differentiation (OD) and inhibited apoptosis of hBMSCs in vitro. The osteogenic markers of RUNX1, osteopontin (OPN) and osteocalcin (OCN) were significantly upregulated in hBMSCs after miR-144-3p inhibitor was transferred. Mechanistic analysis demonstrated that CircSLC8A1 directly bound to miR-144-3p and participated in PPOL through the miR-144-3p/RUNX1 pathway in hBMSCs. Micro-CT and quantitative analysis showed that CircSLC8A1 markedly inhibited PPOL, and osteogenic markers (RUNX1, OPN and OCN) were significantly increased (P<0.05) in the mice model. Our findings prove that CircSLC8A1 exerted a regulatory role in promoting osteogenic differentiation in hBMSCs, and CircSLC8A1/miR-144-3p/RUNX1 pathway may provide a potential target for prevention of PPOL.
Collapse
Affiliation(s)
- Boning Yang
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Yu Qin
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Ao Zhang
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Penghao Wang
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Hua Jiang
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Yunyi Shi
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Guanchao You
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Dianlin Shen
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Shenghui Ni
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Lei Guo
- Department of Orthopedic Surgery, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| | - Ying Liu
- Department of Nursing, First Affiliated HospitalChina Medical UniversityShenyangLiaoningChina
| |
Collapse
|
5
|
Kim DH, Bae J, Heo JH, Park CH, Kim EB, Lee JH. Nanoparticles as Next-Generation Tooth-Whitening Agents: Progress and Perspectives. ACS NANO 2022; 16:10042-10065. [PMID: 35704786 DOI: 10.1021/acsnano.2c01412] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Whitening agents, such as hydrogen peroxide and carbamide peroxide, are currently used in clinical applications for dental esthetic and dental care. However, the free radicals generated by whitening agents cause pathological damage; therefore, their safety issues remain controversial. Furthermore, whitening agents are known to be unstable and short-lived. Since 2001, nanoparticles (NPs) have been researched for use in tooth whitening. Importantly, nanoparticles not only function as abrasives but also release reactive oxygen species and help remineralization. This review outlines the historical development of several NPs based on their whitening effects and side effects. NPs can be categorized into metals or metal oxides, ceramic particles, graphene oxide, and piezoelectric particles. Moreover, the status quo and future prospects are discussed, and recent progress in the development of NPs and their applications in various fields requiring tooth whitening is examined. This review promotes the research and development of next-generation NPs for use in tooth whitening.
Collapse
Affiliation(s)
- Dai-Hwan Kim
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jina Bae
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jun Hyuk Heo
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Advanced Materials Technology Research Center, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Cheol Hyun Park
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Eun Bi Kim
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jung Heon Lee
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Advanced Materials Technology Research Center, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
6
|
Kwon HK, Dussik CM, Kim SH, Kyriakides TR, Oh I, Lee FY. Treating 'Septic' With Enhanced Antibiotics and 'Arthritis' by Mitigation of Excessive Inflammation. Front Cell Infect Microbiol 2022; 12:897291. [PMID: 35755835 PMCID: PMC9218192 DOI: 10.3389/fcimb.2022.897291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Bacterial infection within the synovial joint, commonly known as septic arthritis, remains a clinical challenge as it presents two concurrent therapeutic goals of reducing bacterial burden and preservation of articular cartilage from destructive host inflammation. We hypothesized that mitigation of MRSA-induced inflammatory signaling could diminish destruction of articular cartilage in the setting of septic arthritis when used in conjunction with antibiotics. Herein, we provide evidence which supports a new therapeutic notion that concurrent antimicrobial therapy to address the 'septic' component of the disease with inflammation mitigation to manage the destructive 'arthritis' component. We established a murine model to mimic septic knee arthritis, as well as a variety of other inflammatory joint conditions. This murine septic arthritis model, in conjunction with in vitro and ex-vivo models, was utilized to characterize the inflammatory profile seen in active septic arthritis, as well as post-antibiotic treatment, via transcriptomic and histologic studies. Finally, we provided the clinical rationale for a novel therapeutic strategy combining enhanced antibiotic treatment with rifampin and adjuvant immunomodulation to inhibit post-infectious, excess chondrolysis and osteolysis. We identified that septic arthritis secondary to MRSA infection in our murine model led to increased articular cartilage damage compared to various types of inflammatory arthritis. The activation of the pERK1/2 signaling pathway, which is implicated with the mounting of an immune response and generation of inflammation, was increased in intracellular MRSA-infected synovial tissue and persisted despite antibiotic treatment. Trametinib, an inhibitor of ERK signaling through suppression of MEK1/2, alleviated the inflammation produced by the addition of intra-articular, heat-killed MRSA. Further, when combined with vancomycin and rifampin, mitigation of inflammation by pERK1/2 targeting improved outcomes for MRSA septic arthritis by conferring chondroprotection to articular cartilage and diminishing inflammatory osteolysis within bone. Our results support a new therapeutic notion that cell/biofilm-penetrating antibiotics alongside adjuvant mitigation of excessive intra-articular inflammation accomplish distinct therapeutic goals: reduction of bacterial burden and preservation of articular cartilage integrity.
Collapse
Affiliation(s)
- Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| | - Christopher M. Dussik
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| | - Sang-Hun Kim
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Themis R. Kyriakides
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States
| | - Irvin Oh
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| | - Francis Y. Lee
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
7
|
Back J, Nguyen MN, Li L, Lee S, Lee I, Chen F, Gillinov L, Chung YH, Alder KD, Kwon HK, Yu KE, Dussik CM, Hao Z, Flores MJ, Kim Y, Ibe IK, Munger AM, Seo SW, Lee FY. Inflammatory conversion of quiescent osteoblasts by metastatic breast cancer cells through pERK1/2 aggravates cancer-induced bone destruction. Bone Res 2021; 9:43. [PMID: 34588427 PMCID: PMC8481290 DOI: 10.1038/s41413-021-00158-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 03/09/2021] [Accepted: 03/28/2021] [Indexed: 02/08/2023] Open
Abstract
Disruption of bone homeostasis caused by metastatic osteolytic breast cancer cells increases inflammatory osteolysis and decreases bone formation, thereby predisposing patients to pathological fracture and cancer growth. Alteration of osteoblast function induces skeletal diseases due to the disruption of bone homeostasis. We observed increased activation of pERK1/2 in osteolytic breast cancer cells and osteoblasts in human pathological specimens with aggressive osteolytic breast cancer metastases. We confirmed that osteolytic breast cancers with high expression of pERK1/2 disrupt bone homeostasis via osteoblastic ERK1/2 activation at the bone-breast cancer interface. The process of inflammatory osteolysis modulates ERK1/2 activation in osteoblasts and breast cancer cells through dominant-negative MEK1 expression and constitutively active MEK1 expression to promote cancer growth within bone. Trametinib, an FDA-approved MEK inhibitor, not only reduced breast cancer-induced bone destruction but also dramatically reduced cancer growth in bone by inhibiting the inflammatory skeletal microenvironment. Taken together, these findings suggest that ERK1/2 activation in both breast cancer cells and osteoblasts is required for osteolytic breast cancer-induced inflammatory osteolysis and that ERK1/2 pathway inhibitors may represent a promising adjuvant therapy for patients with aggressive osteolytic breast cancers by altering the shared cancer and bone microenvironment.
Collapse
Affiliation(s)
- Jungho Back
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| | - Minh Nam Nguyen
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA ,grid.444808.40000 0001 2037 434XResearch Center for Genetics and Reproductive Health, School of Medicine, Vietnam National University Ho Chi Minh City, Linh Trung Ward, Thu Duc District, Ho Chi Minh City, Vietnam
| | - Lu Li
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA ,grid.415869.7Department of Rehabilitation Medicine, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Saelim Lee
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA ,grid.411982.70000 0001 0705 4288College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Inkyu Lee
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA ,grid.254224.70000 0001 0789 9563Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Fancheng Chen
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA ,grid.11841.3d0000 0004 0619 8943Shanghai Medical College, Fudan University, Shanghai City, China
| | - Lauren Gillinov
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| | - Yeon-Ho Chung
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| | - Kareme D. Alder
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| | - Hyuk-Kwon Kwon
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| | - Kristin E. Yu
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| | - Christopher M. Dussik
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| | - Zichen Hao
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA ,grid.411525.60000 0004 0369 1599Department of Emergency & Trauma, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Michael J. Flores
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| | - Yoseph Kim
- grid.21107.350000 0001 2171 9311Biomedical Engineering, Johns Hopkins University, Baltimore, MD USA
| | - Izuchukwu K. Ibe
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| | - Alana M. Munger
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| | - Sung Wook Seo
- grid.414964.a0000 0001 0640 5613Department of Orthopedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Gangnam-gu Republic of Korea
| | - Francis Y. Lee
- grid.47100.320000000419368710Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT USA
| |
Collapse
|
8
|
Fibroblast-Like-Synoviocytes Mediate Secretion of Pro-Inflammatory Cytokines via ERK and JNK MAPKs in Ti-Particle-Induced Osteolysis. MATERIALS 2020; 13:ma13163628. [PMID: 32824426 PMCID: PMC7476030 DOI: 10.3390/ma13163628] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/08/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022]
Abstract
Biomaterials are designed to replace and augment living tissues in order to provide functional support to skeletal deformities. However, wear debris produced from the interfaces of metal implants initiates inflammatory bone loss, causing periprosthetic osteolysis. Lately, fibroblast-like synoviocytes (FLS) have been shown to play a role in wear-debris-induced osteolysis. Thus, here we have tried to understand the underlying mechanism of FLS involvement in wear-debris-induced osteolysis. Our results demonstrate that the effects of Ti particle (1:100 cell-to-Ti particle ratio) on FLS can induce Cox-2 expression and activate NFkB signaling. Moreover, the mRNA expression of pro-inflammatory cytokines such as IL-6, IL-8, IL-11, IL-1β, and TNFα was found to be elevated. However, among these pro-inflammatory cytokines, the mRNA and protein levels of only IL-6, IL-1β, and TNFα were found to be significantly higher. Ti particles activated extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinases (MAPKs) as an early response in FLS. Co-inhibition of ERK and JNK signaling pathways by their specific inhibitors (PD9805 and SP600125, respectively) resulted in the suppression of mRNA and protein levels of IL-6, IL-1β, and TNFα in FLS. Taken together, targeting ERK and JNK MAPKs in FLS might provide a therapeutic option for reducing the secretion of bone-resorbing pro-inflammatory cytokines, thus preventing periprosthetic osteolysis.
Collapse
|
9
|
Zhang L, Haddouti EM, Welle K, Burger C, Wirtz DC, Schildberg FA, Kabir K. The Effects of Biomaterial Implant Wear Debris on Osteoblasts. Front Cell Dev Biol 2020; 8:352. [PMID: 32582688 PMCID: PMC7283386 DOI: 10.3389/fcell.2020.00352] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Aseptic loosening subsequent to periprosthetic osteolysis is the leading cause for the revision of arthroplasty failure. The biological response of macrophages to wear debris has been well established, however, the equilibrium of bone remodeling is not only dictated by osteoclastic bone resorption but also by osteoblast-mediated bone formation. Increasing evidence shows that wear debris significantly impair osteoblastic physiology and subsequent bone formation. In the present review, we update the current state of knowledge regarding the effect of biomaterial implant wear debris on osteoblasts. The interaction of osteoblasts with osteoclasts and macrophages under wear debris challenge, and potential treatment options targeting osteoblasts are also presented.
Collapse
Affiliation(s)
- Li Zhang
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - El-Mustapha Haddouti
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristian Welle
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Christof Burger
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C Wirtz
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Koroush Kabir
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
10
|
Nishida K, Hasegawa A, Yamasaki S, Uchida R, Ohashi W, Kurashima Y, Kunisawa J, Kimura S, Iwanaga T, Watarai H, Hase K, Ogura H, Nakayama M, Kashiwakura JI, Okayama Y, Kubo M, Ohara O, Kiyono H, Koseki H, Murakami M, Hirano T. Mast cells play role in wound healing through the ZnT2/GPR39/IL-6 axis. Sci Rep 2019; 9:10842. [PMID: 31346193 PMCID: PMC6658492 DOI: 10.1038/s41598-019-47132-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 07/11/2019] [Indexed: 01/08/2023] Open
Abstract
Zinc (Zn) is an essential nutrient and its deficiency causes immunodeficiency and skin disorders. Various cells including mast cells release Zn-containing granules when activated; however, the biological role of the released Zn is currently unclear. Here we report our findings that Zn transporter ZnT2 is required for the release of Zn from mast cells. In addition, we found that Zn and mast cells induce IL-6 production from inflammatory cells such as skin fibroblasts and promote wound healing, a process that involves inflammation. Zn induces the production of a variety of pro-inflammatory cytokines including IL-6 through signaling pathways mediated by the Zn receptor GPR39. Consistent with these findings, wound healing was impaired in mice lacking IL-6 or GPR39. Thus, our results show that Zn and mast cells play a critical role in wound healing through activation of the GPR39/IL-6 signaling axis.
Collapse
Affiliation(s)
- Keigo Nishida
- Laboratory of Immune Regulation, Graduate School of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki-cho, Suzuka, Mie, 513-8670, Japan. .,Laboratory for Homeostatic Network, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.
| | - Aiko Hasegawa
- Laboratory for Homeostatic Network, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.,Department of Pediatrics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Satoru Yamasaki
- Laboratory for Homeostatic Network, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.,Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Ryota Uchida
- Laboratory of Immune Regulation, Graduate School of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki-cho, Suzuka, Mie, 513-8670, Japan
| | - Wakana Ohashi
- Laboratory for Homeostatic Network, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.,Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Toyama, 930-0194, Japan
| | - Yosuke Kurashima
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.,Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, the Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan.,Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.,Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan.,Institute for Global Prominent Research, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California San Diego, 9500 Gilman Dr. MC 0063, San Diego, CA, 92093-0063, United States.,Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085, Japan.,Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Shunsuke Kimura
- Laboratory of Histology and Cytology, Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan.,Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Toshihiko Iwanaga
- Laboratory of Histology and Cytology, Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
| | - Hiroshi Watarai
- Department of Immunology and Stem Cell Biology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan.,International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo (IMSUT), 108-8639, Tokyo, Japan
| | - Hideki Ogura
- Department of Microbiology, Hyogo College of Medicine 1-1, Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Manabu Nakayama
- Laboratory of Medical Omics Research, Department of Frontier Research and Development, Kazusa DNA Research Institute,2-6-7 Kazusa-Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Jun-Ichi Kashiwakura
- Laboratory of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Yoshimichi Okayama
- Allergy and Immunology Project Team, Center for Allergy, Center for Medical Education, Nihon University School of Medicine, 30-1 Oyaguchi Kamicho Itabashi-Ku, Tokyo, 173-8610, Japan
| | - Masato Kubo
- Laboratory for Cytokine Regulation, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.,Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, 2669 Yamazaki, Noda-shi, Chiba, 278-0022, Japan
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, the Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan.,Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan.,Institute for Global Prominent Research, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California San Diego, 9500 Gilman Dr. MC 0063, San Diego, CA, 92093-0063, United States.,Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, 060-815, Japan
| | - Toshio Hirano
- Headquarters, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| |
Collapse
|
11
|
Jagga S, Sharma AR, Bhattacharya M, Chakraborty C, Lee SS. Influence of single nucleotide polymorphisms (SNPs) in genetic susceptibility towards periprosthetic osteolysis. Genes Genomics 2019; 41:1113-1125. [PMID: 31313107 DOI: 10.1007/s13258-019-00845-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/26/2019] [Indexed: 12/13/2022]
Abstract
Wear debris-induced inflammatory osteolysis remains a significant limiting factor for implant replacement surgeries. Hence, a comprehensive understanding of the complex network of cellular and molecular signals leading to these inflammatory responses is required. Both macrophages and monocytes have a critical role in the instigation of the inflammatory reaction to wear debris but differ in the extent to which they induce cytokine expression in patients. Lately, single nucleotide polymorphisms (SNPs) have been associated with genetic susceptibility among individual patients with implant failure. Studies have shown that SNPs in key pro-inflammatory cytokines and their receptors are associated with osteolytic susceptibility. Likewise, SNPs within several genes involved in the regulation of bone turnover have also been found to be associated with wear debris induced osteolysis. It is presumed that SNP variance might play a decisive role in the activation and signaling of macrophages, osteoblasts, chondrocytes, fibroblasts and other cells involved in inflammatory bone loss. Understanding the extent to which SNPs exist among genes that are responsible for inflammatory bone loss may provide potential targets for developing future therapeutic interventions. Herein, we attempt to summarize the various susceptible genes with possible SNP variance that could contribute to the severity of periprosthetic osteolysis in patients with implants.
Collapse
Affiliation(s)
- Supriya Jagga
- Institute for Skeletal Aging and Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, Gangwon-do, 24252, Republic of Korea
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging and Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, Gangwon-do, 24252, Republic of Korea
| | - Manojit Bhattacharya
- Institute for Skeletal Aging and Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, Gangwon-do, 24252, Republic of Korea
| | - Chiranjib Chakraborty
- Institute for Skeletal Aging and Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, Gangwon-do, 24252, Republic of Korea. .,Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Rd, Jagannathpur, Kolkata, West Bengal, 700126, India.
| | - Sang-Soo Lee
- Institute for Skeletal Aging and Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, Gangwon-do, 24252, Republic of Korea.
| |
Collapse
|
12
|
Liu X, Yang H, Liu Y, Jiao Y, Yang L, Wang X, Yu W, Su D, Tian J. Remifentanil upregulates hepatic IL-18 binding protein (IL-18BP) expression through transcriptional control. J Transl Med 2018; 98:1588-1599. [PMID: 30089853 DOI: 10.1038/s41374-018-0111-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/17/2018] [Accepted: 07/01/2018] [Indexed: 01/01/2023] Open
Abstract
Interleukin (IL)-18 plays an important role in liver ischemia/reperfusion (I/R) injury. We have previously demonstrated that remifentanil protects against liver I/R injury by upregulating the hepatic expression of IL-18-binding protein (IL-18BP), a natural IL-18 inhibitor. The current study was performed to further clarify the effects of remifentanil on IL-18BP expression in the liver as well as investigate the underlying mechanisms. In Sprague-Dawley (SD) rats, we demonstrated that remifentanil significantly increased the expression of IL-18BP in normal rat liver tissue over a 24-h time period with maximal expression at 24 h after treatment. The upregulation of remifentanil on IL-18BP expression displayed similar trends in in vitro cellular studies, including mouse primary hepatocytes, normal human hepatocyte LO2, and mouse hepatoma cells Hep1-6. In LO2 cells, preexposure of the cells to remifentanil significantly inhibited IL-18-activated p65 NF-κB phosphorylation, and the inhibition was absent when the cells were transfected with IL-18BP siRNA, indicating the functional effects of IL-18BP induced by remifentanil. Pretreatment with actinomycin D abolished remifentanil-induced upregulation of IL-18BP mRNA, suggesting that the induction occurred at the transcriptional level. This was further supported by the luciferase reporter assay, which demonstrated that remifentanil treatment significantly increased transcription of the IL-18BP promoter. Both western blot analysis and ChIP assays showed that STAT1 and C/EBP β were activated by remifentanil. Furthermore, remifentanil failed to upregulate IL-18BP expression after silencing STAT1 or C/EBP β gene expression. These findings demonstrate that remifentanil could upregulate hepatic IL-18BP expression through transcriptional activation of the IL-18BP promoter, and STAT1 and C/EBP β are two key transcriptional factors involved in this process.
Collapse
Affiliation(s)
- Xiaohua Liu
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Hao Yang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Rd, Shanghai, 200433, China
| | - Yan Liu
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yingfu Jiao
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Liqun Yang
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Xiangrui Wang
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Weifeng Yu
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Diansan Su
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd, Shanghai, 200127, China.
| | - Jie Tian
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd, Shanghai, 200127, China.
| |
Collapse
|
13
|
Liu SB, Lin XP, Xu Y, Shen ZF, Pan WW. DAXX promotes ovarian cancer ascites cell proliferation and migration by activating the ERK signaling pathway. J Ovarian Res 2018; 11:90. [PMID: 30336783 PMCID: PMC6193355 DOI: 10.1186/s13048-018-0462-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/03/2018] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The death-domain-associated protein (DAXX) was originally identified as a protein that binds to the transmembrane death receptor FAS and enhances both FAS-induced and transforming growth factor-β-dependent apoptosis. In a previous study, we found that nude mice injected with DAXX-overexpressing cells (ES-2-DAXX) accumulated large concentrations of first-generation ascites cells (I ascites cells). The role of DAXX in the development of ascites is unknown. The aim of this study was to analyze the effect of DAXX on proliferation and migration of ascites cells in ovarian cancer in vitro and in vivo. METHODS Nude mice were housed in cages with a 14:10 h light:dark cycle; water and food were provided ad libitum. ES-2-DAXX cells (1×106) were injected intraperitoneally into athymic nude mice (8-week-old female mice). After 4 weeks, I ascites cells were collected. The I ascites cells were injected intraperitoneally into athymic nude mice (8-week-old female mice). After 4 weeks, II ascites cells were collected and cultured. Ascites cell survival, migration, and colony formation were measured using colony formation and cell growth assays. Immunofluorescent staining revealed the co-localization of DAXX and promyelocytic leukemia protein (PML) in ascites cell nuclei. Western blotting and immunohistochemistry showed that extracellular signal-related kinase (p-ERK) 1/2 and CEBP-β were highly expressed in tumor tissues formed by II ascites cells. Through immunoprecipitation, we also found that DAXX can interact with CEBP-β. RESULTS DAXX enhanced ascites cell survival, migration, and colony formation. DAXX and PML nuclear foci dramatically increased in a passage-dependent manner in ascites cells, DAXX promoted the tumor growth of ascites cells in vivo, increased ascites cell proliferation in vivo, and enhanced ascites cell survival and migration by activating the ERK signalling pathway and integrating with CEBP-β. CONCLUSIONS DAXX can interact with CEBP-β. DAXX can induce ovarian cancer ascites formation by activating the ERK signal pathway and binding to CEBP-β.
Collapse
Affiliation(s)
- Sheng-Bing Liu
- College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - Xue-Ping Lin
- College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - Ying Xu
- College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - Zhong-Fei Shen
- College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - Wei-Wei Pan
- College of Medicine, Jiaxing University, Jiaxing, 314001, China.
| |
Collapse
|
14
|
Ge YW, Liu ZQ, Sun ZY, Yu DG, Feng K, Zhu ZA, Mao YQ. Titanium particle‑mediated osteoclastogenesis may be attenuated via bidirectional ephrin‑B2/eph‑B4 signaling in vitro. Int J Mol Med 2018; 42:2031-2041. [PMID: 30015911 PMCID: PMC6108887 DOI: 10.3892/ijmm.2018.3780] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/06/2018] [Indexed: 11/25/2022] Open
Abstract
The present study investigated the role of bidirectional ephrin-B2/erythropoietin-producing human hepatocellular receptor 4 (ephB4) signaling in the regulation of wear particle-mediated osteoclastogenesis in vitro. Mouse bone marrow macrophages (BMMs) were induced into osteoclasts by receptor activator of nuclear factor-κB ligand (RANKL, 50 ng/ml). EphB4-Fc, an osteoblast membrane surface receptor (4 µg/ml), was used to stimulate the ephrin-B2 ligand of osteoclasts in the presence and absence of titanium (Ti). Tartrate-resistant acid phosphatase (TRAP) staining was used to detect the number of osteoclasts, and phalloidin staining was used to examine the cytoskeletons of the osteoclasts. A bone pit absorption experiment was used to measure osteoclast function. Reverse transcription quantitative polymerase chain reaction and western blot analysis were used to examine osteoclastogenesis. ELISAs were used to detect the production of inflammatory factors. The data demonstrated that Ti significantly promoted the differentiation of BMMs into mature osteoclasts in the presence of RANKL and significantly promoted expression of the ephrin-B2, nuclear factor of activated T-cells 1 (NFATc1), TRAP, Fos proto-oncogene, AP-1 transcription factor subunit (C-FOS), and matrix metalloproteinase 9 (MMP9) genes. Phalloidin and TRAP staining revealed that following the addition of ephB4-Fc, the number, size and cytoskeletal elements of Key words: osteoclasts, osteoblasts, remodeling, ephrin-B2, osteoclastogenesis osteoclasts were significantly decreased compared with those in the titanium particle group without ephB4-Fc. Compared with the titanium particle group, the bone pit absorption experiment revealed significantly decreased absorption pit areas in the titanium particle+ephB4-Fc group. The expression of the NFATc1, TRAP, C-FOS and MMP9 genes was markedly decreased in the ephB4-Fc group; however, the expression of the ephrin-B2 gene was increased compared with the Ti particle group without ephB4-Fc after 5 days. Production of inflammatory cytokines was inhibited by Ti particles through bidirectional signals. Addition of ephB4-Fc inhibited the osteoclast-mediated formation of Ti particles via bidirectional ephrin-B2/ephB4 signaling. Activation of this bidirectional signaling pathway may be a potential clinical treatment for osteolysis surrounding prostheses.
Collapse
Affiliation(s)
- Yu-Wei Ge
- Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Zhi-Qing Liu
- Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Zhen-Yu Sun
- Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - De-Gang Yu
- Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Kai Feng
- Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Zhen-An Zhu
- Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Yuan-Qing Mao
- Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
15
|
Nam JS, Sharma AR, Jagga S, Lee DH, Sharma G, Nguyen LT, Lee YH, Chang JD, Chakraborty C, Lee SS. Suppression of osteogenic activity by regulation of WNT and BMP signaling during titanium particle induced osteolysis. J Biomed Mater Res A 2017; 105:912-926. [PMID: 28076890 DOI: 10.1002/jbm.a.36004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/25/2016] [Accepted: 01/06/2017] [Indexed: 12/12/2022]
Abstract
Periprosthetic osteolysis remains the leading obstacle for total joint replacements. Primarily, it was thought that aseptic loosening is mainly caused by macrophage mediated inflammatory process arising from production of wear debris. The role of osteoclasts and its sequential bone resorption ability has been extensively studied, but little is known about impaired osteogenesis during osteolysis. In the current study, we have tried to delineate the regulatory mechanism of osteogenic signals by Ti particles in osteoprogenitor cells as well its participatory role in wear debris induced osteolysis. Implantation of Ti particles on mice calvaria induced pro-inflammatory response, elevated expression of COX2 and reduced the expression of Osterix. Treatment of Ti particles to MC3T3 E-1 cells displayed decreased osteogenic activity including ALP activity, mineralization and mRNA levels several osteogenic genes. Moreover, the basal activity of WNT and BMP signaling pathways was suppressed in MC3T3 E-1 cells treated with Ti particles. As an early response to Ti particles, MC3T3 E-1 cells showed activation of ERK and JNK. Co-inhibition of ERK and JNK with their specific inhibitors resulted in partial recovery of WNT and BMP signaling activity as well as ALP activity and collagen synthesis. Finally, LiCl mediated activation of WNT signaling pathway demonstrated rescue of Ti particle facilitated suppression of Osterix expression in mice calvaria. Our results provide evidences that WNT signaling pathway is regulated by ERK, JNK, and BMP signaling pathway during wear debris induced inflammatory osteolysis and may be considered as suitable therapeutic targets for the treatment. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 912-926, 2017.
Collapse
Affiliation(s)
- Ju-Suk Nam
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea
| | - Supriya Jagga
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea
| | - Dong-Hyun Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea
| | - Garima Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea
| | - Lich Thi Nguyen
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea
| | - Yeon Hee Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea
| | - Jun-Dong Chang
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea
| | - Chiranjib Chakraborty
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea.,Department of Bio-informatics, School of Computer and Information Sciences, Galgotias University, Greater Noida, 203201, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea
| |
Collapse
|
16
|
Geng H, Chang YN, Bai X, Liu S, Yuan Q, Gu W, Li J, Chen K, Xing G, Xing G. Fullerenol nanoparticles suppress RANKL-induced osteoclastogenesis by inhibiting differentiation and maturation. NANOSCALE 2017; 9:12516-12523. [PMID: 28819671 DOI: 10.1039/c7nr04365a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Bone health requires regulation of homeostatic equilibrium between osteoblasts and osteoclasts. The over-activation of osteoclasts can disrupt bone metabolism, resulting in osteoporosis and other bone-loss diseases. Fullerenol, a polyhydroxy derivative of fullerene, exhibits excellent biocompatibility. Here we show that fullerenol nanoparticles exert two functions: inhibition of osteoclastic differentiation and blockage of pre-osteoclast fusion to restructure osteoclast maturation and function. Experimentally, the nanoparticles reduced pre-osteoclast migration and inhibited ruffled border formation to block their maturation. In addition, fullerenol dose-dependently restricted the differentiation of bone marrow macrophage cells (BMMs) to form osteoclasts following treatment with macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor (NF)-κB (RANKL) to activate NF-κB and mitogen activating protein kinase (MAPK) signaling pathways. It is possible that the very small size of fullerenol allows it to directly cross the cellular membrane to access the cytoplasm and regulate osteoclastogenesis from BMMs. Our results suggest that fullerenol could be used to treat bone-loss diseases such as osteoporosis.
Collapse
Affiliation(s)
- Huan Geng
- Department of Orthopedics, General Hospital of Chinese People's Armed Police Forces, Beijing 100039, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Luo B, Gan W, Liu Z, Shen Z, Wang J, Shi R, Liu Y, Liu Y, Jiang M, Zhang Z, Wu Y. Erythropoeitin Signaling in Macrophages Promotes Dying Cell Clearance and Immune Tolerance. Immunity 2016; 44:287-302. [PMID: 26872696 DOI: 10.1016/j.immuni.2016.01.002] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/03/2015] [Accepted: 01/05/2016] [Indexed: 12/19/2022]
Abstract
The failure of apoptotic cell clearance is linked to autoimmune diseases, nonresolving inflammation, and developmental abnormalities; however, pathways that regulate phagocytes for efficient apoptotic cell clearance remain poorly known. Apoptotic cells release find-me signals to recruit phagocytes to initiate their clearance. Here we found that find-me signal sphingosine 1-phosphate (S1P) activated macrophage erythropoietin (EPO) signaling promoted apoptotic cell clearance and immune tolerance. Dying cell-released S1P activated macrophage EPO signaling. Erythropoietin receptor (EPOR)-deficient macrophages exhibited impaired apoptotic cell phagocytosis. EPO enhanced apoptotic cell clearance through peroxisome proliferator activated receptor-γ (PPARγ). Moreover, macrophage-specific Epor(-/-) mice developed lupus-like symptoms, and interference in EPO signaling ameliorated the disease progression in lupus-like mice. Thus, we have identified a pathway that regulates macrophages to clear dying cells, uncovered the priming function of find-me signal S1P, and found a role of the erythropoiesis regulator EPO in apoptotic cell disposal, with implications for harnessing dying cell clearance.
Collapse
Affiliation(s)
- Bangwei Luo
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China
| | - Woting Gan
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China
| | - Zongwei Liu
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China
| | - Zigang Shen
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China
| | - Jinsong Wang
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China
| | - Rongchen Shi
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China
| | - Yuqi Liu
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China
| | - Yu Liu
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China
| | - Man Jiang
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China
| | - Zhiren Zhang
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China.
| | - Yuzhang Wu
- Institute of Immunology of PLA, Third Military Medical University, 30 Gaotanyan Main Street, Chongqing 400038, China.
| |
Collapse
|
20
|
Patel N, Nizami S, Song L, Mikami M, Hsu A, Hickernell T, Chandhanayingyong C, Rho S, Compton JT, Caldwell JM, Kaiser PB, Bai H, Lee HG, Fischer CR, Lee FY. CA-074Me compound inhibits osteoclastogenesis via suppression of the NFATc1 and c-FOS signaling pathways. J Orthop Res 2015; 33:1474-86. [PMID: 25428830 DOI: 10.1002/jor.22795] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 11/24/2014] [Indexed: 02/06/2023]
Abstract
The osteoclast is an integral cell of bone resorption. Since osteolytic disorders hinge on the function and dysfunction of the osteoclast, understanding osteoclast biology is fundamental to designing new therapies that curb osteolytic disorders. The identification and study of lysosomal proteases, such as cathepsins, have shed light on mechanisms of bone resorption. For example, Cathepsin K has already been identified as a collagen degradation protease produced by mature osteoclasts with high activity in the acidic osteoclast resorption pits. Delving into the mechanisms of cathepsins and other osteoclast related compounds provides new targets to explore in osteoclast biology. Through our anti-osteoclastogenic compound screening experiments we encountered a modified version of the Cathepsin B inhibitor CA-074: the cell membrane-permeable CA-074Me (L-3-trans-(Propylcarbamoyl) oxirane-2-carbonyl]-L-isoleucyl-L-proline Methyl Ester). Here we confirm that CA-074Me inhibits osteoclastogenesis in vivo and in vitro in a dose-dependent manner. However, Cathepsin B knockout mice exhibited unaltered osteoclastogenesis, suggesting a more complicated mechanism of action than Cathepsin B inhibition. We found that CA-074Me exerts its osteoclastogenic effect within 24 h of osteoclastogenesis stimulation by suppression of c-FOS and NFATc1 pathways.
Collapse
Affiliation(s)
- Neel Patel
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Saqib Nizami
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Lee Song
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Maya Mikami
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York.,Department of Anesthesiology, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Anny Hsu
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Thomas Hickernell
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | | | - Shim Rho
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Jocelyn T Compton
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York.,Department of Medicine, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Jon-Michael Caldwell
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Philip B Kaiser
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York.,Department of Medicine, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Hanying Bai
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Heon Goo Lee
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Charla R Fischer
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| | - Francis Y Lee
- Department of Orthopaedic Surgery, Columbia University, 650 West 168th Street BB14-1412, NY, 10032, New York
| |
Collapse
|
21
|
Yang M, Liu J, Piao C, Shao J, Du J. ICAM-1 suppresses tumor metastasis by inhibiting macrophage M2 polarization through blockade of efferocytosis. Cell Death Dis 2015; 6:e1780. [PMID: 26068788 PMCID: PMC4669827 DOI: 10.1038/cddis.2015.144] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 04/17/2015] [Accepted: 04/23/2015] [Indexed: 12/14/2022]
Abstract
Efficient clearance of apoptotic cells (efferocytosis) can profoundly influence tumor-specific immunity. Tumor-associated macrophages are M2-polarized macrophages that promote key processes in tumor progression. Efferocytosis stimulates M2 macrophage polarization and contributes to cancer metastasis, but the signaling mechanism underlying this process is unclear. Intercellular cell adhesion molecule-1 (ICAM-1) is a transmembrane glycoprotein member of the immunoglobulin superfamily, which has been implicated in mediating cell–cell interaction and outside-in cell signaling during the immune response. We report that ICAM-1 expression is inversely associated with macrophage infiltration and the metastasis index in human colon tumors by combining Oncomine database analysis and immunohistochemistry for ICAM-1. Using a colon cancer liver metastasis model in ICAM-1-deficient (ICAM-1−/−) mice and their wild-type littermates, we found that loss of ICAM-1 accelerated liver metastasis of colon carcinoma cells. Moreover, ICAM-1 deficiency increased M2 macrophage polarization during tumor progression. We further demonstrated that ICAM-1 deficiency in macrophages led to promotion of efferocytosis of apoptotic tumor cells through activation of the phosphatidylinositol 3 kinase/Akt signaling pathway. More importantly, coculture of ICAM-1−/− macrophages with apoptotic cancer cells resulted in an increase of M2-like macrophages, which was blocked by an efferocytosis inhibitor. Our findings demonstrate a novel role for ICAM-1 in suppressing M2 macrophage polarization via downregulation of efferocytosis in the tumor microenvironment, thereby inhibiting metastatic tumor progression.
Collapse
Affiliation(s)
- M Yang
- 1] Beijing Anzhen Hospital, Capital Medical University, Beijing, China [2] Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing collaborative innovative research center for cardiovascular diseases, Beijing, China
| | - J Liu
- 1] Beijing Anzhen Hospital, Capital Medical University, Beijing, China [2] Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing collaborative innovative research center for cardiovascular diseases, Beijing, China
| | - C Piao
- 1] Beijing Anzhen Hospital, Capital Medical University, Beijing, China [2] Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing collaborative innovative research center for cardiovascular diseases, Beijing, China
| | - J Shao
- Second Affiliated Hospital to Nanchang University, Jiangxi 330006, China
| | - J Du
- 1] Beijing Anzhen Hospital, Capital Medical University, Beijing, China [2] Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing collaborative innovative research center for cardiovascular diseases, Beijing, China
| |
Collapse
|
22
|
Muehlschlegel JD, Christodoulou DC, McKean D, Gorham J, Mazaika E, Heydarpour M, Lee G, DePalma SR, Perry TE, Fox AA, Shernan SK, Seidman CE, Aranki SF, Seidman JG, Body SC. Using next-generation RNA sequencing to examine ischemic changes induced by cold blood cardioplegia on the human left ventricular myocardium transcriptome. Anesthesiology 2015; 122:537-50. [PMID: 25581909 PMCID: PMC4333028 DOI: 10.1097/aln.0000000000000582] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND The exact mechanisms that underlie the pathological processes of myocardial ischemia in humans are unclear. Cardiopulmonary bypass with cardioplegic arrest allows the authors to examine the whole transcriptional profile of human left ventricular myocardium at baseline and after exposure to cold cardioplegia-induced ischemia as a human ischemia model. METHODS The authors obtained biopsies from 45 patients undergoing aortic valve replacement surgery at baseline and after an average of 79 min of cold cardioplegic arrest. Samples were RNA sequenced and analyzed with the Partek Genomics Suite (Partek Inc., St. Louis, MO) for differential expression. Ingenuity Pathway Analysis (Ingenuity Systems, Redwood City, CA) and Biobase ExPlain (Biobase GmbH, Wolfenbuettel, Germany) systems were used for functional and pathway analyses. RESULTS Of the 4,098 genes with a mean expression value greater than 5, 90% were down-regulated and 9.1% were up-regulated. Of those, 1,241 were significantly differentially expressed. Gene ontology analysis revealed significant down-regulation in immune inflammatory response and complement activation categories and highly consistent was the down-regulation of intelectin 1, proteoglycan, and secretory leukocyte peptidase inhibitor. Up-regulated genes of interest were FBJ murine osteosarcoma viral oncogene homolog and the hemoglobin genes hemoglobin α1 (HBA1) and hemoglobin β. In addition, analysis of transcription factor-binding sites revealed interesting targets in factors regulating reactive oxygen species production, apoptosis, immunity, cytokine production, and inflammatory response. CONCLUSIONS The authors have shown that the human left ventricle exhibits significant changes in gene expression in response to cold cardioplegia-induced ischemia during cardiopulmonary bypass, which provides great insight into the pathophysiology of ventricular ischemia, and thus, may help guide efforts to reduce myocardial damage during surgery.
Collapse
Affiliation(s)
- Jochen D Muehlschlegel
- From the Brigham and Women's Hospital, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Medical School, Boston, Massachusetts (J.D.M., M.H., S.K.S., S.C.B.); Department of Genetics, Harvard Medical School, Boston, Massachusetts (D.C.C., D.M., J.G., E.M., S.R.D., J.G.S.); Harvard Medical School, Boston, Massachusetts (G.L.); Northwest Anesthesia, Abbott Northwestern Hospital and Minneapolis Heart Institute, Minneapolis, Minnesota (T.E.P.); Department of Anesthesiology, University of Texas Southwestern Medical Center, Dallas, Texas (A.A.F.); Howard Hughes Medical Institute, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (C.E.S.); and Brigham and Women's Hospital, Division of Cardiac Surgery, Department of Surgery, Harvard Medical School, Boston, Massachusetts (S.F.A.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Orthopaedic implant failure: aseptic implant loosening–the contribution and future challenges of mouse models in translational research. Clin Sci (Lond) 2014; 127:277-93. [DOI: 10.1042/cs20130338] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aseptic loosening as a result of wear debris is considered to be the main cause of long-term implant failure in orthopaedic surgery and improved biomaterials for bearing surfaces decreases significantly the release of micrometric wear particles. Increasingly, in-depth knowledge of osteoimmunology highlights the role of nanoparticles and ions released from some of these new bearing couples, opening up a new era in the comprehension of aseptic loosening. Mouse models have been essential in the progress made in the early comprehension of pathophysiology and in testing new therapeutic agents for particle-induced osteolysis. However, despite this encouraging progress, there is still no valid clinical alternative to revision surgery. The present review provides an update of the most commonly used bearing couples, the current concepts regarding particle–cell interactions and the approaches used to study the biology of periprosthetic osteolysis. It also discusses the contribution and future challenges of mouse models for successful translation of the preclinical progress into clinical applications.
Collapse
|
24
|
Liu Y, Zhu Z, Cai H, Liu Q, Zhou H, Zhu Z. SKI-II reverses the chemoresistance of SGC7901/DDP gastric cancer cells. Oncol Lett 2014; 8:367-373. [PMID: 24959278 PMCID: PMC4063656 DOI: 10.3892/ol.2014.2083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 07/26/2013] [Indexed: 01/25/2023] Open
Abstract
Cisplatin is frequently used in treating gastric cancers; however, acquired resistance to the drug often reduces the efficacy of therapy. The present study analyzed the efficacy of the combination of 4-[4-(4-chloro-phenyl)-thiazol-2-ylamino]-phenol (SKI-II) and cisplatin [cis-diamminedichloroplatinum (II); DDP] on the gastric cancer SGC7901/DDP cell line. The results revealed that SKI-II and DDP had a clear synergistic effect. Glutathione (GSH) and glutathione S-transferase (GST) levels decreased significantly subsequent to the cells being treated with the combination of DDP and SKI-II compared with the cells that were treated with DDP or SKI-II alone. Phosphorylated extracellular-signal-regulated kinase (p-ERK) and phosphorylated c-Jun N-terminal kinase (p-JNK) expression levels also decreased following treatment with SKI-II. The results suggested that SKI-II is able to reverse the drug resistance in human gastric carcinoma cells and enhance the antitumor effect of DDP through the ras/mitogen-activated protein kinase (MAPK) proliferation pathway.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Zuan Zhu
- Department of Gastroenterology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Hongxing Cai
- Department of Forensic Medicine, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Qinghua Liu
- Department of Pathology, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Honglian Zhou
- Department of Gastroenterology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Zhengqiu Zhu
- Department of Medical Oncology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
25
|
Zhao Y, Wang MY, Hao K, Chen XQ, Du JZ. CRHR1 mediates p53 transcription induced by high altitude hypoxia through ERK 1/2 signaling in rat hepatic cells. Peptides 2013; 44:8-14. [PMID: 23538210 DOI: 10.1016/j.peptides.2013.03.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 01/14/2023]
Abstract
We have previously reported that hypoxia activates corticotrophin-releasing hormone (CRH) and the expression of its type-1 receptor (CRHR1) and induces disorders of the brain-endocrine-immune network. p53 is activated by hypoxia and involved in tumorigenesis and apoptosis. Whether CRHR1 regulates p53 transactivation to further influence apoptotic genes remains unclear. Here, we showed that hypoxia at a simulated altitude of 5km or 7km for 8 and 24h increased p53 protein and mRNA, and reduced apoptotic bax and IGFBP3 gene expression while upregulating the cell-arrest gene p21 for 8h in rat liver cells. The upregulation of p53 mRNA and downregulation of bax mRNA induced by hypoxia were blocked by pretreatment with the specific CRHR1 antagonist CP-154,526, but the downregulation of IGFBP3 and upregulation of p21 mRNA were not. Furthermore, CRH stimulated p53 mRNA via the ERK 1/2 pathway in the BRL-3A cell line and this was blocked by the ERK 1/2 antagonist U0126. These data provide novel evidence that the CRHR1-triggered ERK 1/2 pathway is involved in the activation of p53 and suppression of the apoptotic bax gene by hypoxia in rat liver.
Collapse
Affiliation(s)
- Yang Zhao
- Division of Neurobiology and Physiology, Department of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | | | | | | | | |
Collapse
|
26
|
Lee HG, Hsu A, Goto H, Nizami S, Lee JH, Cadet ER, Tang P, Shaji R, Chandhanayinyong C, Kweon SH, Oh DS, Tawfeek H, Lee FY. Aggravation of inflammatory response by costimulation with titanium particles and mechanical perturbations in osteoblast- and macrophage-like cells. Am J Physiol Cell Physiol 2012; 304:C431-9. [PMID: 23255578 DOI: 10.1152/ajpcell.00202.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The interface between bone tissue and metal implants undergoes various types of mechanical loading, such as strain, compression, fluid pressure, and shear stress, from daily activities. Such mechanical perturbations create suboptimal environments at the host bone-implant junction, causing an accumulation of wear particles and debilitating osseous integration, potentially leading to implant failure. While many studies have focused on the effect of particles on macrophages or osteoprogenitor cells, differential and combined effects of mechanical perturbations and particles on such cell types have not been extensively studied. In this study, macrophages and osteoprogenitor cells were subjected to physiological and superphysiological mechanical stimuli in the presence and absence of Ti particles with the aim of simulating various microenvironments of the host bone-implant junction. Macrophages and osteoprogenitor cells were capable of engulfing Ti particles through actin remodeling and also exhibited changes in mRNA levels of proinflammatory cytokines under certain conditions. In osteoprogenitor cells, superphysiological strain increased proinflammatory gene expression; in macrophages, such mechanical perturbations did not affect gene expression. We confirmed that this phenomenon in osteoprogenitor cells occurred via activation of the ERK1/2 signaling pathway as a result of damage to the cytoplasmic membrane. Furthermore, AZD6244, a clinically relevant inhibitor of the ERK1/2 pathway, mitigated particle-induced inflammatory gene expression in osteoprogenitor cells and macrophages. This study provides evidence of more inflammatory responses under mechanical strains in osteoprogenitor cells than macrophages. Phagocytosis of particles and mechanical perturbation costimulate the ERK1/2 pathway, leading to expression of proinflammatory genes.
Collapse
Affiliation(s)
- Heon Goo Lee
- Center for Orthopaedic Research, Columbia Orthopaedics, Department of Orthopaedic Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kim HG, Han EH, Jang WS, Choi JH, Khanal T, Park BH, Tran TP, Chung YC, Jeong HG. Piperine inhibits PMA-induced cyclooxygenase-2 expression through downregulating NF-κB, C/EBP and AP-1 signaling pathways in murine macrophages. Food Chem Toxicol 2012; 50:2342-8. [DOI: 10.1016/j.fct.2012.04.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 04/02/2012] [Accepted: 04/14/2012] [Indexed: 10/28/2022]
|
28
|
Chandhanayingyong C, Kim Y, Staples JR, Hahn C, Lee FY. MAPK/ERK Signaling in Osteosarcomas, Ewing Sarcomas and Chondrosarcomas: Therapeutic Implications and Future Directions. Sarcoma 2012; 2012:404810. [PMID: 22577336 PMCID: PMC3345255 DOI: 10.1155/2012/404810] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 01/09/2012] [Indexed: 01/08/2023] Open
Abstract
The introduction of cytotoxic chemotherapeutic drugs in the 1970's improved the survival rate of patients with bone sarcomas and allowed limb salvage surgeries. However, since the turn of the century, survival data has plateaued for a subset of metastatic, nonresponding osteo, and/or Ewing sarcomas. In addition, most high-grade chondrosarcoma does not respond to current chemotherapy. With an increased understanding of molecular pathways governing oncogenesis, modern targeted therapy regimens may enhance the efficacy of current therapeutic modalities. Mitogen-Activated Protein Kinases (MAPK)/Extracellular-Signal-Regulated Kinases (ERK) are key regulators of oncogenic phenotypes such as proliferation, invasion, angiogenesis, and inflammatory responses; which are the hallmarks of cancer. Consequently, MAPK/ERK inhibitors have emerged as promising therapeutic targets for certain types of cancers, but there have been sparse reports in bone sarcomas. Scattered papers suggest that MAPK targeting inhibits proliferation, local invasiveness, metastasis, and drug resistance in bone sarcomas. A recent clinical trial showed some clinical benefits in patients with unresectable or metastatic osteosarcomas following MAPK/ERK targeting therapy. Despite in vitro proof of therapeutic concept, there are no sufficient in vivo or clinical data available for Ewing sarcomas or chondrosarcomas. Further experimental and clinical trials are awaited in order to bring MAPK targeting into a clinical arena.
Collapse
Affiliation(s)
- Chandhanarat Chandhanayingyong
- Center for Orthopedic Research (COR), Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
- Department of Orthopedic Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Yuhree Kim
- Center for Orthopedic Research (COR), Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| | - J. Robert Staples
- Center for Orthopedic Research (COR), Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| | - Cody Hahn
- Center for Orthopedic Research (COR), Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| | - Francis Youngin Lee
- Center for Orthopedic Research (COR), Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| |
Collapse
|
29
|
Lee SS, Sharma AR, Choi BS, Jung JS, Chang JD, Park S, Salvati EA, Purdue EP, Song DK, Nam JS. The effect of TNFα secreted from macrophages activated by titanium particles on osteogenic activity regulated by WNT/BMP signaling in osteoprogenitor cells. Biomaterials 2012; 33:4251-63. [PMID: 22436801 DOI: 10.1016/j.biomaterials.2012.03.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/03/2012] [Indexed: 12/22/2022]
Abstract
Wear particles are the major cause of osteolysis associated with failure of implant following total joint replacement. During this pathologic process, activated macrophages mediate inflammatory responses to increase osteoclastogenesis, leading to enhanced bone resorption. In osteolysis caused by wear particles, osteoprogenitors present along with macrophages at the implant interface may play significant roles in bone regeneration and implant osteointegration. Although the direct effects of wear particles on osteoblasts have been addressed recently, the role of activated macrophages in regulation of osteogenic activity of osteoblasts has scarcely been studied. In the present study, we examined the molecular communication between macrophages and osteoprogenitor cells that may explain the effect of wear particles on impaired bone forming activity in inflammatory bone diseases. It has been demonstrated that conditioned medium of macrophages challenged with titanium particles (Ti CM) suppresses early and late differentiation markers of osteoprogenitors, including alkaline phosphatase (ALP) activity, collagen synthesis, matrix mineralization and expression of osteocalcin and Runx2. Moreover, bone forming signals such as WNT and BMP signaling pathways were inhibited by Ti CM. Interestingly, TNFα was identified as a predominant factor in Ti CM to suppress osteogenic activity as well as WNT and BMP signaling activity. Furthermore, Ti CM or TNFα induces the expression of sclerostin (SOST) which is able to inhibit WNT and BMP signaling pathways. It was determined that over-expression of SOST suppressed ALP activity, whereas the inhibition of SOST by siRNA partially restored the effect of Ti CM on ALP activity. This study highlights the role of activated macrophages in regulation of impaired osteogenic activity seen in inflammatory conditions and provides a potential mechanism for autocrine regulation of WNT and BMP signaling mediated by TNFα via induction of SOST in osteprogenitor cells.
Collapse
Affiliation(s)
- Sang-Soo Lee
- Infectious Disease Medical Research Center & Department of Pharmacology, College of Medicine, Hallym University, Chucheon, Gangwon-do 200-702, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|