1
|
Lokau J, Bollmann M, Garbers Y, Feist E, Lohmann CH, Bertrand J, Garbers C. Transforming growth factor beta induces interleukin-11 expression in osteoarthritis. Cytokine 2025; 187:156863. [PMID: 39879889 DOI: 10.1016/j.cyto.2025.156863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/04/2025] [Accepted: 01/19/2025] [Indexed: 01/31/2025]
Abstract
Interleukin-11 (IL-11) is a member of the IL-6 family of cytokines and possesses both pro- and anti-inflammatory properties. IL-11 activates its target cells via binding to a membrane-bound IL-11R and subsequent formation of a homodimer of the signal-transducing receptor gp130. Thus, the expression pattern of the IL-11R determines which cells can be activated by IL-11. However, knowledge about IL-11 target cells and cells that secrete IL-11 are sparse, and the overall roles of IL-11 in inflammatory diseases are largely unexplored. In this study, we show that high amounts of IL-11 can be detected via ELISA in the synovial fluid of osteoarthritis (OA) patients in comparison to rheumatoid arthritis (RA) patients. Using primary cells and tissue of OA patients, we show that IL-11 is expressed by chondrocytes in cartilage, but not in the synovium. We further identify the cytokine transforming growth factor β 1(TGF-β1) as a potent inducer of IL-11 secretion in both primary chondrocytes and fibroblasts, and TGF-β1 and IL-11 levels correlate significantly in the synovial fluid of OA patients. Using immunohistochemistry, we show that both cartilage and synovium express IL-11R, and the amount of IL-11R is independent of the disease severity. Primary chondrocytes and fibroblasts from OA patients respond to IL-11 stimulation with potent activation of the Jak/STAT3 signaling cascade, suggesting that these cell types are not only the source, but also the targets of IL-11 in OA patients. Our results uncover IL-11 as a potential new target for therapy in OA.
Collapse
Affiliation(s)
- Juliane Lokau
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany; Department of Pathology, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Miriam Bollmann
- Department of Orthopaedic Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Yvonne Garbers
- Faculty of Management, Culture and Technology (Lingen campus), Osnabrück University of Applied Sciences, 49809 Lingen, (Ems), Germany
| | - Eugen Feist
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, and Experimental Rheumatology, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Christoph H Lohmann
- Department of Orthopaedic Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Jessica Bertrand
- Department of Orthopaedic Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany; Department of Pathology, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
2
|
Li B, Shen Y, Liu S, Yuan H, Liu M, Li H, Zhang T, Du S, Liu X. Identification of immune microenvironment subtypes and clinical risk biomarkers for osteoarthritis based on a machine learning model. Front Mol Biosci 2024; 11:1376793. [PMID: 39484639 PMCID: PMC11524973 DOI: 10.3389/fmolb.2024.1376793] [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: 01/26/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024] Open
Abstract
Background Osteoarthritis (OA) is a degenerative disease with a high incidence worldwide. Most affected patients do not exhibit obvious discomfort symptoms or imaging findings until OA progresses, leading to irreversible destruction of articular cartilage and bone. Therefore, developing new diagnostic biomarkers that can reflect articular cartilage injury is crucial for the early diagnosis of OA. This study aims to explore biomarkers related to the immune microenvironment of OA, providing a new research direction for the early diagnosis and identification of risk factors for OA. Methods We screened and downloaded relevant data from the Gene Expression Omnibus (GEO) database, and the immune microenvironment-related genes (Imr-DEGs) were identified using the ImmPort data set by combining weighted coexpression analysis (WGCNA). Functional enrichment of GO and Kyoto Encyclopedia of Genes and Genomes (KEGG) were conducted to explore the correlation of Imr-DEGs. A random forest machine learning model was constructed to analyze the characteristic genes of OA, and the diagnostic significance was determined by the Receiver Operating Characteristic Curve (ROC) curve, with external datasets used to verify the diagnostic ability. Different immune subtypes of OA were identified by unsupervised clustering, and the function of these subtypes was analyzed by gene set enrichment analysis (GSVA). The Drug-Gene Interaction Database was used to explore the relationship between characteristic genes and drugs. Results Single sample gene set enrichment analysis (ssGSEA) revealed that 16 of 28 immune cell subsets in the dataset significantly differed between OA and normal groups. There were 26 Imr-DEGs identified by WGCNA, showing that functional enrichment was related to immune response. Using the random forest machine learning model algorithm, nine characteristic genes were obtained: BLNK (AUC = 0.809), CCL18 (AUC = 0.692), CD74 (AUC = 0.794), CSF1R (AUC = 0.835), RAC2 (AUC = 0.792), INSR (AUC = 0.765), IL11 (AUC = 0.662), IL18 (AUC = 0.699), and TLR7 (AUC = 0.807). A nomogram was constructed to predict the occurrence and development of OA, and the calibration curve confirmed the accuracy of these 9 genes in OA diagnosis. Conclusion This study identified characteristic genes related to the immune microenvironment in OA, providing new insight into the risk factors of OA.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xinwei Liu
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
3
|
Sims NA, Lévesque JP. Oncostatin M: Dual Regulator of the Skeletal and Hematopoietic Systems. Curr Osteoporos Rep 2024; 22:80-95. [PMID: 38198032 PMCID: PMC10912291 DOI: 10.1007/s11914-023-00837-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 01/11/2024]
Abstract
PURPOSE OF THE REVIEW The bone and hematopoietic tissues coemerge during development and are functionally intertwined throughout mammalian life. Oncostatin M (OSM) is an inflammatory cytokine of the interleukin-6 family produced by osteoblasts, bone marrow macrophages, and neutrophils. OSM acts via two heterodimeric receptors comprising GP130 with either an OSM receptor (OSMR) or a leukemia inhibitory factor receptor (LIFR). OSMR is expressed on osteoblasts, mesenchymal, and endothelial cells and mice deficient for the Osm or Osmr genes have both bone and blood phenotypes illustrating the importance of OSM and OSMR in regulating these two intertwined tissues. RECENT FINDINGS OSM regulates bone mass through signaling via OSMR, adaptor protein SHC1, and transducer STAT3 to both stimulate osteoclast formation and promote osteoblast commitment; the effect on bone formation is also supported by action through LIFR. OSM produced by macrophages is an important inducer of neurogenic heterotopic ossifications in peri-articular muscles following spinal cord injury. OSM produced by neutrophils in the bone marrow induces hematopoietic stem and progenitor cell proliferation in an indirect manner via OSMR expressed by bone marrow stromal and endothelial cells that form hematopoietic stem cell niches. OSM acts as a brake to therapeutic hematopoietic stem cell mobilization in response to G-CSF and CXCR4 antagonist plerixafor. Excessive OSM production by macrophages in the bone marrow is a key contributor to poor hematopoietic stem cell mobilization (mobilopathy) in people with diabetes. OSM and OSMR may also play important roles in the progression of several cancers. It is increasingly clear that OSM plays unique roles in regulating the maintenance and regeneration of bone, hematopoietic stem and progenitor cells, inflammation, and skeletal muscles. Dysregulated OSM production can lead to bone pathologies, defective muscle repair and formation of heterotopic ossifications in injured muscles, suboptimal mobilization of hematopoietic stem cells, exacerbated inflammatory responses, and anti-tumoral immunity. Ongoing research will establish whether neutralizing antibodies or cytokine traps may be useful to correct pathologies associated with excessive OSM production.
Collapse
Affiliation(s)
- Natalie A Sims
- St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC, Australia
- Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
| | - Jean-Pierre Lévesque
- Translational Research Institute, Mater Research Institute - The University of Queensland, 37 Kent Street, Woolloongabba, QLD, Australia.
| |
Collapse
|
4
|
Han Y, Gao H, Gan X, Liu J, Bao C, He C. Roles of IL-11 in the regulation of bone metabolism. Front Endocrinol (Lausanne) 2024; 14:1290130. [PMID: 38352248 PMCID: PMC10862480 DOI: 10.3389/fendo.2023.1290130] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/29/2023] [Indexed: 02/16/2024] Open
Abstract
Bone metabolism is the basis for maintaining the normal physiological state of bone, and imbalance of bone metabolism can lead to a series of metabolic bone diseases. As a member of the IL-6 family, IL-11 acts primarily through the classical signaling pathway IL-11/Receptors, IL-11 (IL-11R)/Glycoprotein 130 (gp130). The regulatory role of IL-11 in bone metabolism has been found earlier, but mainly focuses on the effects on osteogenesis and osteoclasis. In recent years, more studies have focused on IL-11's roles and related mechanisms in different bone metabolism activities. IL-11 regulates osteoblasts, osteoclasts, BM stromal cells, adipose tissue-derived mesenchymal stem cells, and chondrocytes. It's involved in bone homeostasis, including osteogenesis, osteolysis, bone marrow (BM) hematopoiesis, BM adipogenesis, and bone metastasis. This review exams IL-11's role in pathology and bone tissue, the cytokines and pathways that regulate IL-11 expression, and the feedback regulations of these pathways.
Collapse
Affiliation(s)
| | | | - Xinling Gan
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | | | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Xu J, Yu L, Liu F, Wan L, Deng Z. The effect of cytokines on osteoblasts and osteoclasts in bone remodeling in osteoporosis: a review. Front Immunol 2023; 14:1222129. [PMID: 37475866 PMCID: PMC10355373 DOI: 10.3389/fimmu.2023.1222129] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 06/12/2023] [Indexed: 07/22/2023] Open
Abstract
The complicated connections and cross talk between the skeletal system and the immune system are attracting more attention, which is developing into the field of Osteoimmunology. In this field, cytokines that are among osteoblasts and osteoclasts play a critical role in bone remodeling, which is a pathological process in the pathogenesis and development of osteoporosis. Those cytokines include the tumor necrosis factor (TNF) family, the interleukin (IL) family, interferon (IFN), chemokines, and so on, most of which influence the bone microenvironment, osteoblasts, and osteoclasts. This review summarizes the effect of cytokines on osteoblasts and osteoclasts in bone remodeling in osteoporosis, aiming to providing the latest reference to the role of immunology in osteoporosis.
Collapse
Affiliation(s)
- Jie Xu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Linxin Yu
- Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Longbiao Wan
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhenhua Deng
- Hubei Provincial Hospital of Traditional Chinese Medicine (TCM), Wuhan, China
| |
Collapse
|
6
|
de Souza PPC, Henning P, Lerner UH. Stimulation of Osteoclast Formation by Oncostatin M and the Role of WNT16 as a Negative Feedback Regulator. Int J Mol Sci 2022; 23:3287. [PMID: 35328707 PMCID: PMC8953253 DOI: 10.3390/ijms23063287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023] Open
Abstract
Oncostatin M (OSM), which belongs to the IL-6 family of cytokines, is the most potent and effective stimulator of osteoclast formation in this family, as assessed by different in vitro assays. Osteoclastogenesis induced by the IL-6 type of cytokines is mediated by the induction and paracrine stimulation of the osteoclastogenic cytokine receptor activator of nuclear factor κ-B ligand (RANKL), expressed on osteoblast cell membranes and targeting the receptor activator of nuclear factor κ-B (RANK) on osteoclast progenitor cells. The potent effect of OSM on osteoclastogenesis is due to an unusually robust induction of RANKL in osteoblasts through the OSM receptor (OSMR), mediated by a JAK-STAT/MAPK signaling pathway and by unique recruitment of the adapter protein Shc1 to the OSMR. Gene deletion of Osmr in mice results in decreased numbers of osteoclasts and enhanced trabecular bone caused by increased trabecular thickness, indicating that OSM may play a role in physiological regulation of bone remodeling. However, increased amounts of OSM, either through administration of recombinant protein or of adenoviral vectors expressing Osm, results in enhanced bone mass due to increased bone formation without any clear sign of increased osteoclast numbers, a finding which can be reconciled by cell culture experiments demonstrating that OSM can induce osteoblast differentiation and stimulate mineralization of bone nodules in such cultures. Thus, in vitro studies and gene deletion experiments show that OSM is a stimulator of osteoclast formation, whereas administration of OSM to mice shows that OSM is not a strong stimulator of osteoclastogenesis in vivo when administered to adult animals. These observations could be explained by our recent finding showing that OSM is a potent stimulator of the osteoclastogenesis inhibitor WNT16, acting in a negative feedback loop to reduce OSM-induced osteoclast formation.
Collapse
Affiliation(s)
- Pedro P. C. de Souza
- The Innovation in Biomaterials Laboratory, School of Dentistry, Federal University of Goiás, Goiânia 74690-900, Brazil;
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| | - Ulf H. Lerner
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| |
Collapse
|
7
|
Henning P, Movérare-Skrtic S, Westerlund A, Chaves de Souza PP, Floriano-Marcelino T, Nilsson KH, El Shahawy M, Ohlsson C, Lerner UH. WNT16 is Robustly Increased by Oncostatin M in Mouse Calvarial Osteoblasts and Acts as a Negative Feedback Regulator of Osteoclast Formation Induced by Oncostatin M. J Inflamm Res 2021; 14:4723-4741. [PMID: 34566421 PMCID: PMC8457865 DOI: 10.2147/jir.s323435] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/02/2021] [Indexed: 01/22/2023] Open
Abstract
Background Bone loss is often observed adjacent to inflammatory processes. The WNT signaling pathways have been implicated as novel regulators of both immune responses and bone metabolism. WNT16 is important for cortical bone mass by inhibiting osteoclast differentiation, and we have here investigated the regulation of WNT16 by several members of the pro-inflammatory gp130 cytokine family. Methods The expression and regulation of Wnt16 in primary murine cells were studied by qPCR, scRNAseq and in situ hybridization. Signaling pathways were studied by siRNA silencing. The importance of oncostatin M (OSM)-induced WNT16 expression for osteoclastogenesis was studied in cells from Wnt16-deficient and wild-type mice. Results We found that IL-6/sIL-6R and OSM induce the expression of Wnt16 in primary mouse calvarial osteoblasts, with OSM being the most robust stimulator. The induction of Wnt16 by OSM was dependent on gp130 and OSM receptor (OSMR), and downstream signaling by the SHC1/STAT3 pathway, but independent of ERK. Stimulation of the calvarial cells with OSM resulted in enhanced numbers of mature, oversized osteoclasts when cells were isolated from Wnt16 deficient mice compared to cells from wild-type mice. OSM did not affect Wnt16 mRNA expression in bone marrow cell cultures, explained by the finding that Wnt16 and Osmr are expressed in distinctly different cells in bone marrow, nor was osteoclast differentiation different in OSM-stimulated bone marrow cell cultures isolated from Wnt16−/- or wild-type mice. Furthermore, we found that Wnt16 expression is substantially lower in cells from bone marrow compared to calvarial osteoblasts. Conclusion These findings demonstrate that OSM is a robust stimulator of Wnt16 mRNA in calvarial osteoblasts and that WNT16 acts as a negative feedback regulator of OSM-induced osteoclast formation in the calvarial bone cells, but not in the bone marrow.
Collapse
Affiliation(s)
- Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sofia Movérare-Skrtic
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Westerlund
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pedro Paulo Chaves de Souza
- The Innovation in Biomaterials Laboratory, School of Dentistry, Federal University of Goiás, Goiânia, Brazil.,Department of Physiology and Pathology, São Paulo State University (UNESP), School of Dentistry, Araraquara, Brazil
| | - Thais Floriano-Marcelino
- Department of Physiology and Pathology, São Paulo State University (UNESP), School of Dentistry, Araraquara, Brazil
| | - Karin H Nilsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maha El Shahawy
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Oral Biology, Faculty of Dentistry, Minia University, Minia, 61511, Egypt
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf H Lerner
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Influences of the IL-6 cytokine family on bone structure and function. Cytokine 2021; 146:155655. [PMID: 34332274 DOI: 10.1016/j.cyto.2021.155655] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 01/12/2023]
Abstract
The IL-6 family of cytokines comprises a large group of cytokines that all act via the formation of a signaling complex that includes the glycoprotein 130 (gp130) receptor. Despite this, many of these cytokines have unique roles that regulate the activity of bone forming osteoblasts, bone resorbing osteoclasts, bone-resident osteocytes, and cartilage cells (chondrocytes). These include specific functions in craniofacial development, longitudinal bone growth, and the maintenance of trabecular and cortical bone structure, and have been implicated in musculoskeletal pathologies such as craniosynostosis, osteoporosis, rheumatoid arthritis, osteoarthritis, and heterotopic ossifications. This review will work systematically through each member of this family and provide an overview and an update on the expression patterns and functions of each of these cytokines in the skeleton, as well as their negative feedback pathways, particularly suppressor of cytokine signaling 3 (SOCS3). The specific cytokines described are interleukin 6 (IL-6), interleukin 11 (IL-11), oncostatin M (OSM), leukemia inhibitory factor (LIF), cardiotrophin 1 (CT-1), ciliary neurotrophic factor (CNTF), cardiotrophin-like cytokine factor 1 (CLCF1), neuropoietin, humanin and interleukin 27 (IL-27).
Collapse
|
9
|
Persson E, Souza PPC, Floriano-Marcelino T, Conaway HH, Henning P, Lerner UH. Activation of Shc1 Allows Oncostatin M to Induce RANKL and Osteoclast Formation More Effectively Than Leukemia Inhibitory Factor. Front Immunol 2019; 10:1164. [PMID: 31191537 PMCID: PMC6547810 DOI: 10.3389/fimmu.2019.01164] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/08/2019] [Indexed: 11/16/2022] Open
Abstract
Background and Purpose: The gp130 family of cytokines signals through receptors dimerizing with the gp130 subunit. Downstream signaling typically activates STAT3 but also SHP2/Ras/MAPK pathways. Oncostatin M (OSM) is a unique cytokine in this family since the receptor (OSMR) activates a non-redundant signaling pathway by recruitment of the adapter Shc1. We have studied the functional relevance of Shc1 for OSM-induced bone resorption. Experimental Approach: Osteoblasts were stimulated with OSM and STAT3 and Shc1 activations were studied using real-time PCR and Western blots. The role of STAT3 and Shc1 for OSM-induced RANKL expression and osteoclast formation was studied by silencing their mRNA expressions. Effects of OSM were compared to those of the closely related cytokine leukemia inhibitory factor (LIF). Key Results: OSM, but not LIF, induced the mRNA and protein expression of Shc1 and activated phosphorylation of Shc1 in the osteoblasts. Silencing of Shc1 decreased OSM-induced activation of STAT3 and RANKL expression. Silencing of STAT3 had no effect on activation of Shc1, but prevented the OSM-mediated increase of RANKL expression. Silencing of either Shc1 or STAT3 in osteoblasts decreased formation of osteoclasts in OSM-stimulated co-cultures of osteoblasts and macrophages. In agreement with these observations, OSM was a more potent and robust stimulator than LIF of RANKL formation and bone resorption in mouse calvariae and osteoclast formation in bone marrow cultures. Conclusions and Implications: Activation of the Shc1-dependent STAT3 signaling is crucial for OSM-induced osteoclast formation. Inhibition of Shc1 is a potential mechanism to specifically inhibit OSM-induced bone resorption.
Collapse
Affiliation(s)
- Emma Persson
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - Pedro P C Souza
- Bone Biology Research Group, Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil.,School of Dentistry, Federal University of Goiás, Goiânia, Brazil
| | - Thais Floriano-Marcelino
- Bone Biology Research Group, Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Howard Herschel Conaway
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf H Lerner
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden.,Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
10
|
Wojdasiewicz P, Poniatowski ŁA, Nauman P, Mandat T, Paradowska-Gorycka A, Romanowska-Próchnicka K, Szukiewicz D, Kotela A, Kubaszewski Ł, Kotela I, Kurkowska-Jastrzębska I, Gasik R. Cytokines in the pathogenesis of hemophilic arthropathy. Cytokine Growth Factor Rev 2018; 39:71-91. [DOI: 10.1016/j.cytogfr.2017.11.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 11/09/2017] [Indexed: 01/26/2023]
|
11
|
Mechanism of prostaglandin E 2-induced transcriptional up-regulation of Oncostatin-M by CREB and Sp1. Biochem J 2018; 475:477-494. [PMID: 29269396 DOI: 10.1042/bcj20170545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 01/15/2023]
Abstract
Oncostatin-M (OSM) is a pleotropic cytokine belonging to the interleukin-6 family. Differential expression of OSM in response to varying stimuli and exhibiting repertoire of functions in different cells renders it challenging to study the mechanism of its expression. Prostaglandin E2 (PGE2) transcriptionally increased osm levels. In silico studies of ∼1 kb upstream of osm promoter region yielded the presence of CRE (cyclic AMP response element)-like sites at the distal end (CREosm). Deletion and point mutation of CREosm clearly indicated that this region imparted an important role in PGE2-mediated transcription. Nuclear protein(s) from PGE2-treated U937 cells, bound to this region, was identified as CRE-binding protein (CREB). CREB was phosphorylated on treatment and was found to be directly associated with CREosm The presence of cofactors p300 and CREB-binding protein in the complex was confirmed. A marked decrease in CREB phosphorylation, binding and transcriptional inhibition on treatment with PKA (protein kinase A) inhibitor, H89 (N-[2-[[3-(4-bromophenyl)-2-propenyl]amino]ethyl]-5-soquinolinesulfonamide), revealed the role of phosphorylated CREB in osm transcription. Additionally, other nuclear protein(s) were specifically associated with the proximal GC region (GCosm) post PGE2 treatment, later confirmed to be specificity protein 1 (Sp1). Interestingly, Sp1 bound to the proximal osm promoter was found to be associated with phospho-CREB-p300 complex bound to the distal osm promoter. Knockdown of Sp1 abrogated the expression and functionality of OSM. Thus, the present study conclusively proves that these transcription factors, bound at the distal and proximal promoter elements are found to associate with each other in a DNA-dependent manner and both are responsible for the PGE2-mediated transcriptional up-regulation of Oncostatin-M.
Collapse
|
12
|
Torossian F, Guerton B, Anginot A, Alexander KA, Desterke C, Soave S, Tseng HW, Arouche N, Boutin L, Kulina I, Salga M, Jose B, Pettit AR, Clay D, Rochet N, Vlachos E, Genet G, Debaud C, Denormandie P, Genet F, Sims NA, Banzet S, Levesque JP, Lataillade JJ, Le Bousse-Kerdilès MC. Macrophage-derived oncostatin M contributes to human and mouse neurogenic heterotopic ossifications. JCI Insight 2017; 2:96034. [PMID: 29093266 DOI: 10.1172/jci.insight.96034] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/26/2017] [Indexed: 02/04/2023] Open
Abstract
Neurogenic heterotopic ossification (NHO) is the formation of ectopic bone generally in muscles surrounding joints following spinal cord or brain injury. We investigated the mechanisms of NHO formation in 64 patients and a mouse model of spinal cord injury-induced NHO. We show that marrow from human NHOs contains hematopoietic stem cell (HSC) niches, in which mesenchymal stromal cells (MSCs) and endothelial cells provide an environment supporting HSC maintenance, proliferation, and differentiation. The transcriptomic signature of MSCs from NHOs shows a neuronal imprinting associated with a molecular network required for HSC support. We demonstrate that oncostatin M (OSM) produced by activated macrophages promotes osteoblastic differentiation and mineralization of human muscle-derived stromal cells surrounding NHOs. The key role of OSM was confirmed using an experimental model of NHO in mice defective for the OSM receptor (OSMR). Our results provide strong evidence that macrophages contribute to NHO formation through the osteogenic action of OSM on muscle cells within an inflammatory context and suggest that OSM/OSMR could be a suitable therapeutic target. Altogether, the evidence of HSCs in ectopic bones growing at the expense of soft tissue in spinal cord/brain-injured patients indicates that inflammation and muscle contribute to HSC regulation by the brain-bone-blood triad.
Collapse
Affiliation(s)
- Frédéric Torossian
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Bernadette Guerton
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Adrienne Anginot
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Kylie A Alexander
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | | | - Sabrina Soave
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Hsu-Wen Tseng
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nassim Arouche
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Laetitia Boutin
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Irina Kulina
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Marjorie Salga
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia.,Université de Versailles Saint-Quentin-en-Yvelines, Evolution of neuromuscular diseases: innovative concepts and practices, Inserm U1179, Montigny le Bretonneux, France
| | - Beulah Jose
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Allison R Pettit
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Denis Clay
- UMS33, Paris 11 University, Paul Brousse Hospital, Villejuif, France
| | - Nathalie Rochet
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, France
| | - Erica Vlachos
- Service de Médecine Physique et de Réadaptation, Paris 12 University, Garches, France
| | - Guillaume Genet
- Service de Médecine Physique et de Réadaptation, Paris 12 University, Garches, France
| | - Charlotte Debaud
- Université de Versailles Saint-Quentin-en-Yvelines, Evolution of neuromuscular diseases: innovative concepts and practices, Inserm U1179, Montigny le Bretonneux, France.,Service de Médecine Physique et de Réadaptation, Paris 12 University, Garches, France
| | - Philippe Denormandie
- Service de Médecine Physique et de Réadaptation, Paris 12 University, Garches, France
| | - François Genet
- Université de Versailles Saint-Quentin-en-Yvelines, Evolution of neuromuscular diseases: innovative concepts and practices, Inserm U1179, Montigny le Bretonneux, France.,Service de Médecine Physique et de Réadaptation, Paris 12 University, Garches, France
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research and Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, Victoria, Australia
| | - Sébastien Banzet
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France.,Centre de Transfusion Sanguine des Armées, L'Institut de Recherche Biomédicale des Armées, Clamart, France
| | - Jean-Pierre Levesque
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Jean-Jacques Lataillade
- Inserm UMR-S-MD1197, Paris 11 University, Paul Brousse Hospital, Villejuif, France.,Centre de Transfusion Sanguine des Armées, L'Institut de Recherche Biomédicale des Armées, Clamart, France
| | | |
Collapse
|
13
|
Wu R, Wang W, Huang G, Mao X, Chen Y, Tang Q, Liao L. Endothelin‑1 induces oncostatin M expression in osteoarthritis osteoblasts by trans‑activating the oncostatin M gene promoter via Ets‑1. Mol Med Rep 2016; 13:3559-66. [PMID: 26934912 DOI: 10.3892/mmr.2016.4960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 01/21/2016] [Indexed: 11/05/2022] Open
Abstract
Oncostatin M (OSM) contributes to cartilage degeneration in osteoarthritis (OA) and was demonstrated to be expressed in OA osteoblasts. Endothelin‑1 (ET‑1) is implicated in the degradation of OA articular cartilage, and osteoblast proliferation and bone development. In the present study, the effects of ET‑1 on OSM expression in human OA osteoblasts were investigated, to the best of our knowledge, for the first time. Primary human OA osteoblasts were treated with ET‑1 (1, 5, 10, 20 and 30 nM) for 0.5, 1, 2, 3 and 4 h with or without the selective ETA receptor (ETAR) antagonist, BQ123, ETB receptor antagonist, BQ788 or the phosphatidylinositol 3‑kinase (PI3K) inhibitor, BKM120. ET‑1 treatment induced OSM mRNA expression, and the intracellular and secreted protein levels of OA osteoblasts in a dose‑dependent manner. This effect was suppressed by BQ123 and BKM120, but not BQ788 administration. In combination with electrophoretic mobility shift assays, deletional and mutational analyses on the activity of a human OSM promoter/luciferase reporter demonstrated that ET‑1 induced OSM expression in OA osteoblasts by trans‑activating the OSM gene promoter through specific binding of Ets‑1 to an Ets‑1 binding site in the OSM promoter in an ETAR‑ and PI3K‑dependent manner. Furthermore, ET‑1 treatment increased the expression of Ets‑1 in a dose‑dependent manner, however the knockdown of Ets‑1 suppressed the ET1‑induced expression of OSM in OA osteoblasts. In conclusion, the present study demonstrated that ET‑1 induces the expression of OSM in OA osteoblasts by trans‑activating the OSM gene promoter primarily through increasing the expression level of Ets‑1 in an ETAR‑ and PI3K‑dependent manner. The current study suggested novel insights into the mechanistic role of ET‑1 in the pathophysiology of OA.
Collapse
Affiliation(s)
- Ren Wu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Wanchun Wang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Guoliang Huang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Xinzhan Mao
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - You Chen
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Qi Tang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Lele Liao
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
14
|
Su CM, Chiang YC, Huang CY, Hsu CJ, Fong YC, Tang CH. Osteopontin Promotes Oncostatin M Production in Human Osteoblasts: Implication of Rheumatoid Arthritis Therapy. THE JOURNAL OF IMMUNOLOGY 2015; 195:3355-64. [PMID: 26304992 DOI: 10.4049/jimmunol.1403191] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 07/23/2015] [Indexed: 12/21/2022]
Abstract
Accumulating evidence indicates that subchondral bone might play an essential role in rheumatoid arthritis (RA). Osteopontin (OPN) induces the production of an important proinflammatory cytokine involved in the pathogenesis of RA. This study evaluated the activation of oncostatin M (OSM) by OPN in human primary osteoblasts to understand RA pathogenesis and characterized the intracellular signaling pathways involved in this activation. Quantitative PCR, ELISA, and Western blot results indicated that stimulation of human primary osteoblasts with OPN induces OSM expression through αvβ3 integrin/c-Src/platelet-derived growth factor receptor transactivation/MEK/ERK. Treatment of osteoblasts with OPN also increased c-Jun phosphorylation, AP-1 luciferase activity, and c-Jun binding to the AP-1 element on the OSM promoter, as demonstrated using chromatin immunoprecipitation assay. Moreover, inhibition of OPN expression using lentiviral-OPN short hairpin RNA resulted in the amelioration of articular swelling, cartilage erosion, and OSM expression in the ankle joint of mice with collagen-induced arthritis as shown using microcomputed tomography and immunohistochemistry staining. Our results imply that OSM expression in osteoblasts increases in response to OPN-induced inflammation in vitro. Finally, lentiviral-OPN short hairpin RNA ameliorates the inflammatory response and bone destruction in mice with collagen-induced arthritis. Therefore, OPN may be a potential therapeutic target for RA.
Collapse
Affiliation(s)
- Chen-Ming Su
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China 322100; Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan 40466
| | - Yi-Chun Chiang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan 40466
| | - Chun-Yin Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan 40466; Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yun-Lin County, Taiwan 65142
| | - Chin-Jung Hsu
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan 40466; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan 40466
| | - Yi-Chin Fong
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan 40466; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan 40466
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan 40466; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan 40466; and Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan 40466
| |
Collapse
|
15
|
NI J, YUAN XM, YAO Q, PENG LB. OSM is overexpressed in knee osteoarthritis and Notch signaling is involved in the effects of OSM on MC3T3-E1 cell proliferation and differentiation. Int J Mol Med 2015; 35:1755-60. [DOI: 10.3892/ijmm.2015.2168] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/23/2015] [Indexed: 11/05/2022] Open
|
16
|
|
17
|
TLR signaling that induces weak inflammatory response and SHIP1 enhances osteogenic functions. Bone Res 2014; 2:14031. [PMID: 26273527 PMCID: PMC4472124 DOI: 10.1038/boneres.2014.31] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/12/2014] [Accepted: 09/03/2014] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptor (TLR)-mediated inflammatory response could negatively affect bone metabolism. In this study, we determined how osteogenesis is regulated during inflammatory responses that are downstream of TLR signaling. Human primary osteoblasts were cultured in collagen gels. Pam3CSK4 (P3C) and Escherichia coli lipopolysaccharide (EcLPS) were used as TLR2 and TLR4 ligand respectively. Porphyromonas gingivalis LPS having TLR2 activity with either TLR4 agonism (Pg1690) or TLR4 antagonism (Pg1449) and mutant E. coli LPS (LPxE/LPxF/WSK) were used. IL-1β, SH2-containing inositol phosphatase-1 (SHIP1) that has regulatory roles in osteogenesis, alkaline phosphatase and mineralization were analyzed. 3α-Aminocholestane (3AC) was used to inhibit SHIP1. Our results suggest that osteoblasts stimulated by P3C, poorly induced IL-1β but strongly upregulated SHIP1 and enhanced osteogenic mediators. On the contrary, EcLPS significantly induced IL-1β and osteogenic mediators were not induced. While Pg1690 downmodulated osteogenic mediators, Pg1449 enhanced osteogenic responses, suggesting that TLR4 signaling annuls osteogenesis even with TLR2 activity. Interestingly, mutant E. coli LPS that induces weak inflammation upregulated osteogenesis, but SHIP1 was not induced. Moreover, inhibiting SHIP1 significantly upregulated TLR2-mediated inflammatory response and downmodulated osteogenesis. In conclusion, these results suggest that induction of weak inflammatory response through TLR2 (with SHIP1 activity) and mutant TLR4 ligands could enhance osteogenesis.
Collapse
|
18
|
Yang WH, Tsai CH, Fong YC, Huang YL, Wang SJ, Chang YS, Tang CH. Leptin induces oncostatin M production in osteoblasts by downregulating miR-93 through the Akt signaling pathway. Int J Mol Sci 2014; 15:15778-90. [PMID: 25198901 PMCID: PMC4200751 DOI: 10.3390/ijms150915778] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 08/20/2014] [Accepted: 09/03/2014] [Indexed: 11/16/2022] Open
Abstract
Inflammatory response and articular destruction are common symptoms of osteoarthritis (OA) and rheumatoid arthritis (RA). Leptin, an adipocyte-secreted hormone that centrally regulates weight control, may exert proinflammatory effects in the joint, depending on the immune response. Yet, the mechanism of leptin interacting with the arthritic inflammatory response is unclear. This study finds that leptin increased expression of oncostatin M (OSM) in human osteoblasts in a concentration- and time-dependent manner. In addition, OBRl, but not OBRs receptor antisense oligonucleotide, abolished the leptin-mediated increase of OSM expression. On the other hand, leptin inhibited miR-93 expression; an miR-93 mimic reversed leptin-increased OSM expression. Stimulation of osteoblasts with leptin promoted Akt phosphorylation, while pretreatment of cells with Akt inhibitor or siRNA reversed leptin-inhibited miR-93 expression. Our results showed that leptin heightened OSM expression by downregulating miR-93 through the Akt signaling pathway in osteoblasts, suggesting leptin as a novel target in arthritis treatment.
Collapse
Affiliation(s)
- Wei-Hung Yang
- Department of Orthopedic Surgery, Taichung Hospital, Department of Health Executive Yuan, Taichung 403, Taiwan.
| | - Chun-Hao Tsai
- Department of Medicine and Graduate Institute of Clinical Medical Science, China Medical University, Taichung 404, Taiwan.
| | - Yi-Chin Fong
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
| | - Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung 500, Taiwan.
| | - Shoou-Jyi Wang
- Department of Orthopedic Surgery, Chang-Hua Hospital, Department of Health Executive Yuan, Changhua Country 500, Taiwan.
| | - Yung-Sen Chang
- Department of Orthopedic Surgery, Taichung Hospital, Department of Health Executive Yuan, Taichung 403, Taiwan.
| | - Chih-Hsin Tang
- Department of Biotechnology, College of Health Science, Asia University, Taichung 500, Taiwan.
| |
Collapse
|
19
|
Chen CY, Su CM, Huang YL, Tsai CH, Fuh LJ, Tang CH. CCN1 induces oncostatin M production in osteoblasts via integrin-dependent signal pathways. PLoS One 2014; 9:e106632. [PMID: 25187949 PMCID: PMC4154729 DOI: 10.1371/journal.pone.0106632] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 08/11/2014] [Indexed: 11/19/2022] Open
Abstract
Inflammatory response and articular destruction are common symptoms of osteoarthritis. Cysteine-rich 61 (CCN1 or Cyr61), a secreted protein from the CCN family, is associated with the extracellular matrix involved in many cellular activities like growth and differentiation. Yet the mechanism of CCN1 interacting with arthritic inflammatory response is unclear. This study finds CCN1 increasing expression of oncostatin m (OSM) in human osteoblastic cells. Pretreatment of αvβ3 monoclonal antibody and inhibitors of focal adhesion kinase (FAK), c-Src, phosphatidylinositol 3-kinase (PI3K), and NF-κB inhibited CCN1-induced OSM expression in osteoblastic cells. Stimulation of cells with CCN1 increased phosphorylation of FAK, c-Src, PI3K, and NF-κB via αvβ3 receptor; CCN1 treatment of osteoblasts increased NF-κB-luciferase activity and p65 binding to NF-κB element on OSM promoter. Results indicate CCN1 heightening OSM expression via αvβ3 receptor, FAK, c-Src, PI3K, and NF-κB signal pathway in osteoblastic cells, suggesting CCN1 as a novel target in arthritis treatment.
Collapse
Affiliation(s)
- Cheng-Yu Chen
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chen-Ming Su
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Medicine and Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Lih-Jyh Fuh
- Department of Prosthodontics, China Medical University Hospital, Taichung, Taiwan
- * E-mail: (CHT); (LJF)
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- * E-mail: (CHT); (LJF)
| |
Collapse
|
20
|
Sims NA, Quinn JMW. Osteoimmunology: oncostatin M as a pleiotropic regulator of bone formation and resorption in health and disease. BONEKEY REPORTS 2014; 3:527. [PMID: 24876928 DOI: 10.1038/bonekey.2014.22] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/07/2014] [Indexed: 01/14/2023]
Abstract
Bone remodeling in health and disease is carried out by osteoblasts and osteoclasts, which respectively produce bone matrix and resorb it. Endocrine and paracrine control of these cells can be direct, but they are also exerted indirectly, either by influencing progenitor cell differentiation or by stimulating paracrine signals from local accessory cells including osteocytes (which form a critical communication and regulation network within the bone matrix), macrophages and T lymphocytes. Here we review the osteotropic actions of the interleukin-6 family member cytokine oncostatin M (OSM), which is of particular interest because of its ability to stimulate bone accrual. OSM is produced within the bone microenvironment by cells of both mesenchymal and hematopoietic origin, including osteocytes, osteoblasts, macrophages and T lymphocytes, and can act via two receptor complexes: OSM receptor:gp130 and leukemia inhibitory factor receptor (LIFR):gp130. Although OSM can directly stimulate osteoblast mineralization activity and differentiation, it can also stimulate mesenchymal stem cell osteoblastic commitment at the expense of adipogenesis. In osteocytes, OSM can suppress the production of the bone formation inhibitor sclerostin, an action that is mediated by LIFR:gp130. OSM also stimulates the production of receptor activator of nuclear factor κB ligand by osteoblasts and thereby drives the formation of osteoclasts particularly in pathological conditions. Thus, cellular effects of OSM on bone metabolism include direct and indirect actions mediated by two related receptor/ligand complexes. OSM therefore provides an example of paracrine and endocrine control mechanisms that regulate bone mass by controlling both bone formation and resorption.
Collapse
Affiliation(s)
- Natalie A Sims
- Bone Cell Biology and Disease Unit, St Vincent's Institute of Medical Research , Melbourne, Victoria, Australia ; Department of Medicine at St Vincent's Hospital Melbourne, The University of Melbourne , Melbourne, Victoria, Australia
| | - Julian M W Quinn
- Prince Henry's Institute, Monash Medical Centre , Melbourne, Victoria, Australia ; Department of Biochemistry and Molecular Biology, Monash University , Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Sanguineti R, Puddu A, Mach F, Montecucco F, Viviani GL. Advanced glycation end products play adverse proinflammatory activities in osteoporosis. Mediators Inflamm 2014; 2014:975872. [PMID: 24771986 PMCID: PMC3977495 DOI: 10.1155/2014/975872] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/19/2014] [Indexed: 12/18/2022] Open
Abstract
Osteoporosis is a major public health burden that is expected to further increase as the global population ages. In the last twenty years, advanced glycation end products (AGEs) have been shown to be critical mediators both in the pathogenesis and development of osteoporosis and other chronic degenerative diseases related to aging. The accumulation of AGEs within the bone induces the formation of covalent cross-links with collagen and other bone proteins which affects the mechanical properties of tissue and disturbs bone remodelling and deterioration, underlying osteoporosis. On the other hand, the gradual deterioration of the immune system during aging (defined as immunosenescence) is also characterized by the generation of a high level of oxidants and AGEs. The synthesis and accumulation of AGEs (both localized within the bone or in the systemic circulation) might trigger a vicious circle (in which inflammation and aging merged in the word "Inflammaging") which can establish and sustain the development of osteoporosis. This narrative review will update the molecular mechanisms/pathways by which AGEs induce the functional and structural bone impairment typical of osteoporosis.
Collapse
Affiliation(s)
- Roberta Sanguineti
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Alessandra Puddu
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Faculty of Medicine, Geneva University Hospitals, 64 Avenue de la Roseraie, 1211 Geneva, Switzerland
| | - Fabrizio Montecucco
- Division of Cardiology, Foundation for Medical Researches, Faculty of Medicine, Geneva University Hospitals, 64 Avenue de la Roseraie, 1211 Geneva, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, 6 Viale Benedetto XV, 16132 Genoa, Italy
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 Gabrielle-Perret-Gentil, 1205 Geneva, Switzerland
| | - Giorgio Luciano Viviani
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| |
Collapse
|
22
|
Güven EP, Taşlı PN, Yalvac ME, Sofiev N, Kayahan MB, Sahin F. In vitrocomparison of induction capacity and biomineralization ability of mineral trioxide aggregate and a bioceramic root canal sealer. Int Endod J 2013; 46:1173-82. [DOI: 10.1111/iej.12115] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 04/02/2013] [Indexed: 12/29/2022]
Affiliation(s)
- E. P. Güven
- Department of Endodontics; Faculty of Dentistry; Yeditepe University; Istanbul Turkey
| | - P. N. Taşlı
- Department of Genetics and Bioengineering; Faculty of Engineering and Architecture; Yeditepe University; Istanbul Turkey
| | - M. E. Yalvac
- Center for Gene Therapy; Department of Pediatrics; The Research Institute at Nationwide Children's Hospital; Ohio State University; Columbus OH USA
| | - N. Sofiev
- Department of Oral and Maxillofacial Surgery; Faculty of Dentistry; Istanbul University; Capa; Istanbul Turkey
| | - M. B. Kayahan
- Department of Endodontics; Faculty of Dentistry; Yeditepe University; Istanbul Turkey
| | - F. Sahin
- Department of Genetics and Bioengineering; Faculty of Engineering and Architecture; Yeditepe University; Istanbul Turkey
| |
Collapse
|
23
|
Williams EL, Edwards CJ, Cooper C, Oreffo ROC. Impact of inflammation on the osteoarthritic niche: implications for regenerative medicine. Regen Med 2012; 7:551-70. [DOI: 10.2217/rme.12.34] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis worldwide and is the sixth leading cause of disability. It costs the UK economy approximately 1% of gross national product per annum. With an aging population, the cost of chronic conditions such as OA continues to rise. Historically, treatments for OA have been limited to painkillers, physiotherapy and joint injections. When these fail, patients are referred for joint replacement surgery. With the advent of tissue engineering strategies aimed at generating new bone and cartilage for repair of osteochondral defects, there has been considerable interest in exploiting these techniques to devise new treatments for OA. To date, little consideration has been given to the OA niche and attendant inflammatory milieu for any regenerative skeletal strategy. This review highlights the importance of understanding the osteoarthritic niche in order to modify existing tissue engineering and regenerative medicine strategies for the future treatment of OA.
Collapse
Affiliation(s)
- Emma L Williams
- Bone & Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Human Development & Health, University of Southampton Medical School, Southampton, UK
| | - Christopher J Edwards
- Bone & Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Human Development & Health, University of Southampton Medical School, Southampton, UK
- Rheumatology Department, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Cyrus Cooper
- Rheumatology Department, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- MRC Lifecourse Epidemiology Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Richard OC Oreffo
- Bone & Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Human Development & Health, University of Southampton Medical School, Southampton, UK
| |
Collapse
|
24
|
Clérigues V, Guillén MI, Castejón MA, Gomar F, Mirabet V, Alcaraz MJ. Heme oxygenase-1 mediates protective effects on inflammatory, catabolic and senescence responses induced by interleukin-1β in osteoarthritic osteoblasts. Biochem Pharmacol 2011; 83:395-405. [PMID: 22155307 DOI: 10.1016/j.bcp.2011.11.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 11/22/2011] [Accepted: 11/23/2011] [Indexed: 11/30/2022]
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease showing altered bone metabolism. Osteoblasts contribute to the regulation of cartilage metabolism and bone remodeling. We have shown previously that induction of heme oxygenase-1 (HO-1) protects OA cartilage against inflammatory and degradative responses. In this study, we investigated the effects of HO-1 induction on OA osteoblast metabolism. HO-1 was induced with cobalt protoporphyrin IX (CoPP) and by transduction with LV-HO-1. In osteoblasts stimulated with interleukin (IL)-1β, CoPP enhanced mineralization, the expression of a number of markers of osteoblast differentiation such as Runx2, bone morphogenetic protein-2, osteocalcin, and collagen 1A1 and 1A2, as well as the ratio osteoprotegerin/receptor activator of nuclear factor-κB ligand. HO-1 induction significantly reduced the expression of matrix metalloproteinase (MMP)-1, MMP-2 and MMP-3, and the production of pro-inflammatory cytokines such as tumor necrosis factor-α and IL-6 whereas IL-10 levels increased. HO-1 also exerted inhibitory effects on prostaglandin (PG)E(2) production which could be dependent on cyclooxygenase-2 and microsomal PGE synthase-1 down-regulation. The activity of senescence-associated β-galactosidase and the expression of the senescence marker caveolin-1 were significantly decreased after HO-1 induction. The inhibition of nuclear factor-κB activation induced by IL-1β in OA osteoblasts may contribute to some HO-1 effects. Our results have shown that HO-1 decreases the production of relevant inflammatory and catabolic mediators that participate in OA pathophysiology thus eliciting protective effects in OA osteoblasts.
Collapse
Affiliation(s)
- Victoria Clérigues
- Department of Pharmacology, University of Valencia, Burjasot, 46100 Valencia, Spain
| | | | | | | | | | | |
Collapse
|
25
|
Bone osteoblastic and mesenchymal stromal cells lack primarily tumoral features in multiple myeloma patients. Leukemia 2010; 24:1368-70. [PMID: 20485369 DOI: 10.1038/leu.2010.96] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
|
27
|
Todoerti K, Lisignoli G, Storti P, Agnelli L, Novara F, Manferdini C, Codeluppi K, Colla S, Crugnola M, Abeltino M, Bolzoni M, Sgobba V, Facchini A, Lambertenghi-Deliliers G, Zuffardi O, Rizzoli V, Neri A, Giuliani N. Distinct transcriptional profiles characterize bone microenvironment mesenchymal cells rather than osteoblasts in relationship with multiple myeloma bone disease. Exp Hematol 2009; 38:141-53. [PMID: 19963035 DOI: 10.1016/j.exphem.2009.11.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 11/05/2009] [Accepted: 11/24/2009] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Multiple myeloma (MM) is characterized by a high incidence of osteolytic bone lesions, which have been previously correlated with the gene expression profiles of MM cells. The aim of this study was to investigate the transcriptional patterns of cells in the bone microenvironment and their relationships with the presence of osteolysis in MM patients. MATERIALS AND METHODS Both mesenchymal (MSC) and osteoblastic (OB) cells were isolated directly from bone biopsies of MM patients and controls to perform gene expression profiling by microarrays and real-time polymerase chain reaction on selected bone-related genes. RESULTS We identified a series of upregulated and downregulated genes that were differentially expressed in the MSC cells of osteolytic and nonosteolytic patients. Comparison of the osteolytic and nonosteolytic samples also showed that the MSC cells and OB had distinct transcriptional patterns. No significantly modulated genes were found in the OBs of the osteolytic and nonosteolytic patients. CONCLUSIONS Our data suggest that the gene expression profiles of cells of the bone microenvironment are different in MM patients and controls, and that MSC cells, but not OBs, have a distinct transcriptional pattern associated with the occurrence of bone lesions in MM patients. These data support the idea that alterations in MSC cells may be involved in MM bone disease.
Collapse
Affiliation(s)
- Katia Todoerti
- Dipartimento di Scienze Mediche, Università di Milano e U.O. Ematologia 1, Fondazione IRCCS Policlinico, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Lisignoli G, Manferdini C, Codeluppi K, Piacentini A, Grassi F, Cattini L, Filardo G, Facchini A. CCL20/CCR6 chemokine/receptor expression in bone tissue from osteoarthritis and rheumatoid arthritis patients: Different response of osteoblasts in the two groups. J Cell Physiol 2009; 221:154-60. [DOI: 10.1002/jcp.21839] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
29
|
Tonnarelli B, Manferdini C, Piacentini A, Codeluppi K, Zini N, Ghisu S, Facchini A, Lisignoli G. Surface-dependent modulation of proliferation, bone matrix molecules, and inflammatory factors in human osteoblasts. J Biomed Mater Res A 2009; 89:687-96. [DOI: 10.1002/jbm.a.32019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
30
|
Botter SM, Glasson SS, Hopkins B, Clockaerts S, Weinans H, van Leeuwen JPTM, van Osch GJVM. ADAMTS5-/- mice have less subchondral bone changes after induction of osteoarthritis through surgical instability: implications for a link between cartilage and subchondral bone changes. Osteoarthritis Cartilage 2009; 17:636-45. [PMID: 19010693 DOI: 10.1016/j.joca.2008.09.018] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Accepted: 09/30/2008] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is characterized by damaged articular cartilage and changes in subchondral bone. Previous work demonstrated aggrecanase-2 deficient (ADAMTS5-/-) mice to be protected from cartilage damage induced by joint instability. This study analyzed whether this protective effect on cartilage is also reflected in the subchondral bone structure. METHODS Right knee joints from 10-week old male wild type (WT) and ADAMTS5-/- mice received transection of the medial meniscotibial ligament to induce OA, whereas left knees were left unoperated. After 8 weeks knee joints were scanned by micro-CT. The proximal tibia was selected for further analysis. Histology was performed to evaluate cartilage damage and osteoclast presence. RESULTS ADAMTS5-/- joints had a significantly thinner subchondral plate and less epiphyseal trabecular bone compared to WT joints. Histology confirmed previous findings that ADAMTS5-/- mice have significantly less cartilage damage than WT in the instability-induced OA model. Although the subchondral bone plate became significantly thicker at the medial tibial plateau in operated joints of both genotypes, the percentage increase was significantly smaller in ADAMTS5-/- mice (WT: 20.7+/-4.7%, ADAMTS5-/-: 8.3+/-1.2% compared to the left unoperated control joint). In ADAMTS5-/- animals a significant decrease was found in both Oc.N./BS and Oc.S./BS. Finally, in WT but not in ADAMTS5-/- mice a significant correlation was found between medial subchondral bone plate thickness and cartilage damage at the medial tibial plateau. CONCLUSION ADAMTS5-/- joints that were protected from cartilage damage showed minor changes in the subchondral bone structure, in contrast to WT mice where substantial changes were found. This finding suggests links between the process of cartilage damage and subchondral bone changes in instability-induced OA.
Collapse
Affiliation(s)
- S M Botter
- Erasmus MC, University Medical Center Rotterdam, Department of Orthopedics, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
31
|
Giuliani N, Lisignoli G, Colla S, Lazzaretti M, Storti P, Mancini C, Bonomini S, Manferdini C, Codeluppi K, Facchini A, Rizzoli V. CC-chemokine ligand 20/macrophage inflammatory protein-3α and CC-chemokine receptor 6 are overexpressed in myeloma microenvironment related to osteolytic bone lesions. Cancer Res 2008; 68:6840-50. [PMID: 18703490 DOI: 10.1158/0008-5472.can-08-0402] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The expression of the chemokine CC-chemokine ligand 20 (CCL20)/macrophage inflammatory protein (MIP)-3alpha and its receptor CC-chemokine receptor 6 (CCR6) by multiple myeloma (MM) and microenvironment cells and their potential relationship with osteoclast (OC) formation and osteolytic bone lesions in MM patients was investigated in this study. First, we found that MM cells rarely produce CCL20/MIP-3alpha but up-regulate its production by bone marrow (BM) osteoprogenitor cells and osteoblasts in coculture with the involvement of soluble factors as interleukin-1beta and tumor necrosis factor alpha. MM cells also stimulate both CCL20/MIP-3alpha and CCR6 expression by OCs in coculture. Thereafter, we showed that CCL20/MIP-3alpha significantly increases both the number of multinucleated tartrate-resistant acid phosphatase-positive OCs and receptor activator of nuclear factor-kappaB-positive OC progenitor cells similar to CCL3/MIP-1alpha. Finally, we found that blocking anti-CCL20/MIP-3alpha and anti-CCR6 antibodies significantly inhibits MM-induced OC formation. In vitro data were further expanded in vivo analyzing a total number of 64 MM patients. Significantly higher CCL20/MIP-3alpha levels were detected in MM patients versus monoclonal gammopathy of uncertain significance (MGUS) subjects and in MM osteolytic patients versus nonosteolytic ones. Moreover, a significant increase of CCL20/MIP-3alpha-positive osteoblasts in osteolytic MM patients compared with nonosteolytic ones was observed. Interestingly, no significant difference in BM CCL20/MIP-3alpha expression and level was observed between MGUS and nonosteolytic MM patients. Our data indicate that CCL20/MIP-3alpha and its receptor CCR6 are up-regulated in the bone microenvironment by MM cells and contribute to OC formation and osteolytic bone lesions in MM patients.
Collapse
Affiliation(s)
- Nicola Giuliani
- Hematology and Bone Marrow Transplantation Center, University of Parma, Parma, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kastl SP, Speidl WS, Kaun C, Katsaros KM, Rega G, Afonyushkin T, Bochkov VN, Valent P, Assadian A, Hagmueller GW, Hoeth M, de Martin R, Ma Y, Maurer G, Huber K, Wojta J. In human macrophages the complement component C5a induces the expression of oncostatin M via AP-1 activation. Arterioscler Thromb Vasc Biol 2008; 28:498-503. [PMID: 18187666 DOI: 10.1161/atvbaha.107.160580] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Macrophages produce the cytokine oncostatin M (OSM), which beside other functions is also involved in inflammation. The complement component C5a mobilizes and activates these cells at inflammatory sites. We examined the effect of C5a on OSM production in human monocytes and in human monocyte-derived macrophages. METHODS AND RESULTS For macrophage transformation peripheral blood monocytes were cultivated for 8 to 10 days in the presence of human serum. C5a significantly increased in these cells OSM antigen as determined by specific ELISA and mRNA as quantitated by real-time polymerase chain reaction in these cells as well as in plaque macrophages. This effect was blocked by antibodies against the receptor C5aR/CD88 and by pertussis toxin. The C5a-induced phosphorylation of p38 and JNK and the C5a-induced increase in OSM production in macrophages was abolished by 2 p38 inhibitors and by a JNK inhibitor. Furthermore C5a increased the nuclear translocation of c-fos and c-jun. Using different OSM promoter deletion mutant constructs we show that the putative AP-1 element is responsible for activation of OSM promoter activity by C5a. CONCLUSIONS Our data establish a link between the complement system and the gp130 receptor cytokine family with possible implications for the pathology of inflammatory diseases.
Collapse
Affiliation(s)
- Stefan P Kastl
- Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lisignoli G, Cristino S, Piacentini A, Zini N, Noël D, Jorgensen C, Facchini A. Chondrogenic differentiation of murine and human mesenchymal stromal cells in a hyaluronic acid scaffold: differences in gene expression and cell morphology. J Biomed Mater Res A 2007; 77:497-506. [PMID: 16482549 DOI: 10.1002/jbm.a.30632] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chondrogenesis is a complex process characterized by a sequence of different steps that start with the condensation of the cells, followed by the expression of specific components, such as collagens and proteoglycans. We evaluated in vitro chondrogenic differentiation of C3H10T1/2 murine mesenchymal cells and compared them with human mesenchymal stromal cells (h-MSCs) in a hyaluronic acid scaffold. We analyzed (from day 0 to day 28) cellular morphology, proliferation, and chondrogenic/osteogenic gene expression at different time points. Our data demonstrate that, during chondrogenic differentiation, murine cells proliferate both in the absence and presence of TGFbeta, while h-MSCs require the presence of this activating factor. Murine cells, even if viable, differentiate on hyaluronan scaffold, maintain a fibroblastic morphology, and form a capsule outside the scaffold. At the mRNA level, murine cells showed a decrease in collagen type I combined with a significant increase in collagen type II (from day 0), and aggrecan (on day 28), as found for h-MSCs. Immunohistochemical data confirmed that chondrogenic differentiation of murine cells, induced by TGFbeta, occurred only in some restricted areas inside the scaffold that were positive to collagen type II, but did not show a cartilage-like tissue structure, as we had found using h-MSCs. These data demonstrate that C3H10T1/2 murine cells, widely used as a chondrogenic model, show a different sequence of chondrogenic events in hyaluronic acid scaffold, compared with primary h-MSCs.
Collapse
Affiliation(s)
- Gina Lisignoli
- Laboratorio di Immunologia e Genetica, Istituti Ortopedici Rizzoli, 40136 Bologna, Italy.
| | | | | | | | | | | | | |
Collapse
|
34
|
Hwang SH, Kim MH, Yang IH, Bahk JY, Han H. Analysis of cytokines in umbilical cord blood-derived multipotent stem cell. BIOTECHNOL BIOPROC E 2007. [DOI: 10.1007/bf02931800] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
35
|
Lisignoli G, Toneguzzi S, Piacentini A, Cristino S, Grassi F, Cavallo C, Facchini A. CXCL12 (SDF-1) and CXCL13 (BCA-1) chemokines significantly induce proliferation and collagen type I expression in osteoblasts from osteoarthritis patients. J Cell Physiol 2006; 206:78-85. [PMID: 15965952 DOI: 10.1002/jcp.20435] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
To evaluate the role of CXC chemokines CXCL8 (IL8), CXCL10 (IP-10), CXCL12 (SDF-1), and CXCL13 (BCA-1) in bone remodeling, we analyzed their effects on osteoblasts (OBs) obtained from subchondral trabecular bone tissue of osteoarthritis (OA) and post-traumatic (PT) patients. The expression of CXC receptors/ligands (CXCR1/CXCL8, CXCR2/CXCL8, CXCR3/CXCL10, CXCR4/CXCL12, and CXCR5/CXCL13) was analyzed in cultured OBs by flow cytometry and immunocytochemistry. Functional assays on CXC chemokine-treated-OBs in the presence or absence of their specific inhibitors were performed to analyze cellular proliferation and the enzymatic response to chemokine activation. The expression of chemokine ligands/receptors was also confirmed in bone tissue samples by immunohistochemical analysis. Collagen type I and alkaline phosphatase mRNA expression were analyzed on CXCL12- and CXCL13-treated OBs by real-time PCR. OBs from both OA and PT patients expressed high levels of CXCR3 and CXCR5 and lower amounts of CXCR1 and CXCR4. CXCL12 and CXCL13, only in OBs from OA patients, induced a significant proliferation that was also confirmed by specific blocking experiments. Moreover, OBs from OA patients released a higher amount of CXCL13 than those of PT patients while no differences were found for CXCL12. In the remodeling area of bone tissue samples, immunohistochemical analysis confirmed that OBs expressed CXCL12/CXCR4 and CXCL13/CXCR5 both in OA and PT samples. CXCL12 and CXCL13 upregulated collagen type I mRNA expression in OBs from OA patients. These data suggest that CXCL12 and CXCL13 may directly modulate cellular proliferation and collagen type I in OA patients, so contributing to the remodeling process that occurs in the evolution of this disease.
Collapse
Affiliation(s)
- Gina Lisignoli
- Laboratorio di Immunologia e Genetica, Istituti Ortopedici Rizzoli, Bologna, Italy.
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Rheumatoid arthritis, juvenile idiopathic arthritis, the seronegative spondyloarthropathies including psoriatic arthritis, and systemic lupus erythematosus are all examples of rheumatic diseases in which inflammation is associated with skeletal pathology. Although some of the mechanisms of skeletal remodeling are shared among these diseases, each disease has a unique impact on articular bone or on the axial or appendicular skeleton. Studies in human disease and in animal models of arthritis have identified the osteoclast as the predominant cell type mediating bone loss in arthritis. Many of the cytokines and growth factors implicated in the inflammatory processes in rheumatic diseases have also been demonstrated to impact osteoclast differentiation and function either directly, by acting on cells of the osteoclast-lineage, or indirectly, by acting on other cell types to modulate expression of the key osteoclastogenic factor receptor activator of nuclear factor (NF) kappaB ligand (RANKL) and/or its inhibitor osteoprotegerin (OPG). Further elucidation of the mechanisms responsible for inflammation-induced bone loss will potentially lead to the identification of novel therapeutic strategies for the prevention of bone loss in these diseases. In this review, we provide an overview of the cell types, inflammatory mediators, and mechanisms that are implicated in bone loss and new bone formation in inflammatory joint diseases.
Collapse
Affiliation(s)
- Nicole C Walsh
- Beth Israel Deaconess Medical Center, New England Baptist Bone and Joint Institute, Harvard Institutes of Medicine, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
37
|
Chen SH, Benveniste EN. Oncostatin M: a pleiotropic cytokine in the central nervous system. Cytokine Growth Factor Rev 2005; 15:379-91. [PMID: 15450253 DOI: 10.1016/j.cytogfr.2004.06.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Oncostatin M (OSM), a member of the interleukin-6 (IL-6) cytokine family, has yet to be well studied, especially in the context of the central nervous system (CNS). The biological functions of OSM are complex and variable, depending on the cellular microenvironment. Inflammatory responses and tumor development are among two of the major events that OSM is involved in. Although OSM levels remain low in the normal CNS, elevated expression occurs in pathological conditions. Therefore, it is crucial to understand the regulation of OSM to control its expression and/or its effects. Accumulating data demonstrate that OSM binds to specific receptor complexes, then activates two major signaling pathways: Janus Kinase-Signal Transducers and Activators of Transcription (JAK-STAT) and Mitogen-Activated Protein Kinase (MAPK), to regulate downstream events. In this review, we focus on the biological functions of OSM, the signaling pathways of OSM in the CNS, and OSM involvement in CNS diseases.
Collapse
Affiliation(s)
- Shao-Hua Chen
- Department of Cell Biology, MCLM 386, University of Alabama at Birmingham, 1918 University Boulevard, Birmingham, AL 35294-0005, USA.
| | | |
Collapse
|
38
|
Abstract
Besides the well-known activities of the prototypical inflammatory cytokines (IL-1beta, TNFalpha), a role for chemokines and their receptors in cartilage degradation in osteoarthritis has recently been reported. Human chondrocytes can produce CC and CXC chemokines and express chemokine receptors for both chemokine subfamilies. Engagement of these receptors can induce the release of matrix degrading enzymes such as matrix metalloproteinases 1, 3, and 13, and N-acetyl-beta-D-glucosaminidase. Furthermore GROalpha, a CXC chemokine acting on CXCR2, can activate an apoptotic pathway in chondrocytes that leads to chondrocyte cell death. These findings suggest that chemokines can act as an autocrine or paracrine loop on chondrocytes and can contribute to many pathophysiological patterns present in osteoarthritis. Chemokines and their downstream signaling pathways can be considered novel therapeutic targets in osteoarthritis.
Collapse
Affiliation(s)
- Rosa Maria Borzì
- Laboratorio di Immunologia e Genetica, Istituti Ortopedici Rizzoli, Bologna, Italy
| | | | | | | |
Collapse
|
39
|
Grassi F, Cristino S, Toneguzzi S, Piacentini A, Facchini A, Lisignoli G. CXCL12 chemokine up-regulates bone resorption and MMP-9 release by human osteoclasts: CXCL12 levels are increased in synovial and bone tissue of rheumatoid arthritis patients. J Cell Physiol 2004; 199:244-51. [PMID: 15040007 DOI: 10.1002/jcp.10445] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chemokines are involved in a number of inflammatory pathologies and some of them show a pivotal role in the modulation of osteoclast development. Therefore, we evaluated the role of CXCL12 chemokine on osteoclast differentiation and function and we analyzed its expression on synovial and bone tissue biopsies from rheumatoid arthritis (RA) patients. Osteoclasts were obtained by 7 days in vitro differentiation with RANKL and M-CSF of CD11b positive cells in the presence or absence of CXCL12. The total number of osteoclast was analyzed by Tartrate-resistant acid phosphatase (TRAP)-staining and bone-resorbing activity was assessed by pit assay. MMP-9 and TIMP-1 release was evaluated by ELISA assay. CXCL12 expression on biopsies from RA patients was analyzed by immunohistochemistry. Osteoclasts obtained in the presence of CXCL12 at 10 nM concentration displayed a highly significant increase in bone-resorbing activity as measured by pit resorption assay, while the total number of mature osteoclasts was not affected. The increased resorption is associated with overexpression of MMP-9. Immunostaining for CXCL12 on synovial and bone tissue biopsies from both rheumatoid arthritis (RA) and osteoarthritis (OA) samples revealed a strong increase in the expression levels under inflammatory conditions. CXCL12 chemokine showed a clear activating role on mature osteoclast by inducing bone-resorbing activity and specific MMP-9 enzymatic release. Moreover, since bone and synovial biopsies from RA patients showed an elevated CXCL12 expression, these findings may provide useful tools for achieving a full elucidation of the complex network that regulates osteoclast function in course of inflammatory diseases.
Collapse
Affiliation(s)
- Francesco Grassi
- Laboratorio di Immunologia e Genetica, Istituti Ortopedici Rizzoli, Bologna, Italy
| | | | | | | | | | | |
Collapse
|
40
|
Lisignoli G, Toneguzzi S, Piacentini A, Cristino S, Cattini L, Grassi F, Facchini A. Recruitment and proliferation of T lymphocytes is supported by IFN?- and TNF?-activated human osteoblasts: Involvement of CD54 (ICAM-1) and CD106 (VCAM-1) adhesion molecules and CXCR3 chemokine receptor. J Cell Physiol 2004; 198:388-98. [PMID: 14755544 DOI: 10.1002/jcp.10427] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The mechanism by which osteoblasts (OB) interact and modulate the phenotype and proliferation of T lymphocytes during inflammation is not well known. The effects of two regulatory cytokines, TNFalpha and IFNgamma, on the expression of CD54 (ICAM-1) and CD106 (VCAM-1) adhesion molecules and the CXCR3 ligands (CXCL9, CXCL10, CXCL11), were assessed in a primary culture of human OB by real-time PCR, flow cytometry, and immunohistochemistry. In addition, we functionally evaluated the recruitment and proliferation of T lymphocytes grown with resting or stimulated OB. According to the present data IFNgamma, either alone or in combination with TNFalpha, significantly up-regulates the expression of CD54 and CD106 and induces the expression and release of CXCL9, CXCL10, CXCL11 in OB. The supernatant of TNFalpha- and IFNgamma-activated OB induces the recruitment of T lymphocytes more significantly than stimulation by CXCR3 ligands. T lymphocyte proliferation is significantly enhanced by direct contact with TNFalpha- and IFNgamma-activated OB or by incubation with the supernatant of TNFalpha- and IFNgamma-activated OB. Blocking experiments with anti-CD11a, anti-CD49d, anti-CXCR3, and Bordetella pertussis toxin demonstrate that adhesion molecules and the CXCR3 chemokine receptor play a key role in the proliferation of T lymphocytes. The present study demonstrates the involvement of adhesion molecules (CD11a and CD49d) and chemokine receptor (CXCR3) in the mechanism by which OB recruit, interact, and modulate T lymphocyte proliferation under inflammatory conditions.
Collapse
Affiliation(s)
- Gina Lisignoli
- Laboratorio di Immunologia e Genetica, Istituti Ortopedici Rizzoli, Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
41
|
Grassi F, Piacentini A, Cristino S, Toneguzzi S, Cavallo C, Facchini A, Lisignoli G. Human osteoclasts express different CXC chemokines depending on cell culture substrate: molecular and immunocytochemical evidence of high levels of CXCL10 and CXCL12. Histochem Cell Biol 2003; 120:391-400. [PMID: 14600836 DOI: 10.1007/s00418-003-0587-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2003] [Indexed: 10/26/2022]
Abstract
Chemokines are important mediators of chemotaxis, cell adherence, and proliferation and exert specific functions in bone remodeling. Despite the potential intriguing role of chemokines in the regulation of osteoclast (OC) functions, little is known about the expression of chemokines and their receptors in human OCs at different stages of differentiation. Therefore, we analyzed the expression of CXC chemokine receptors (CXCR1, CXCR2, CXCR3, CXCR4 and CXCR5) and ligands (CXCL8, CXCL10, CXCL12 and CXCL13) both at molecular and protein levels, in human OCs grown on plastic or calcium phosphate-coated slides at different stages of differentiation. Real-time PCR showed that CXCR1, CXCR2, CXCR3, CXCR4, CXCR5 and CXCL8 were expressed in undifferentiated cells and significantly decreased during OC differentiation. By contrast, CXCL10 and CXCL12 were strongly upregulated from day 0 to day 8 in cells grown on calcium phosphate-coated slides. Immunocytochemistry showed that OCs grown on plastic expressed CXCR3, CXCR4, CXCR5, CXCL8 and CXCL12, while they were negative for CXCR1, CXCR2 and CXCL10. Interestingly, both at molecular and protein levels CXCL10 and CXCL12 significantly increased only when cells were differentiated on calcium phosphate-coated slides. These data suggest that the selection of a substrate that better mimics the tridimensional structure of bone tissue, thus favoring OC maturation and differentiation, may be necessary when studying osteoclastogenesis in vitro.
Collapse
Affiliation(s)
- Francesco Grassi
- Laboratorio di Immunologia e Genetica, Istituti Ortopedici Rizzoli, Via di Barbiano 1/10, 40136, Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
42
|
Dvornyk V, Recker RR, Deng HW. Gene expression studies of osteoporosis: implications for microarray research. Osteoporos Int 2003; 14:451-61. [PMID: 12730742 DOI: 10.1007/s00198-002-1373-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2002] [Accepted: 11/27/2002] [Indexed: 10/26/2022]
Abstract
Osteoporosis is one of the most common bone-related disorders in the elderly. It is a complex disease, and largely determined genetically. Traditional gene expression studies have shown that osteoporosis has complex regulating mechanisms which are controlled by multiple inherent factors, such as hormones, cytokines, various receptors, etc. The complex nature of osteoporosis, and the large number of genes involved in its onset and development, suggest the use of the state of the art microarray technique as a powerful tool to unravel mechanisms underlying etiology of osteoporosis.
Collapse
Affiliation(s)
- V Dvornyk
- Osteoporosis Research Center, Creighton University, 601 N. 30th St., Suite 6730, Omaha, NE 68131, USA.
| | | | | |
Collapse
|
43
|
Ertenli I, Kiraz S, Oztürk MA, Haznedaroğlu IC, Celik I, Calgüneri M. Pathologic thrombopoiesis of rheumatoid arthritis. Rheumatol Int 2003; 23:49-60. [PMID: 12634936 DOI: 10.1007/s00296-003-0289-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2002] [Accepted: 12/16/2002] [Indexed: 10/27/2022]
Abstract
Rheumatoid arthritis (RA) is frequently complicated by thrombocytosis correlated with disease activity. The exact pathogenetic mechanism(s) that cause increased platelet counts in RA are still unknown. Recent investigations indicate that proinflammatory pleiotropic cytokines of RA also have megakaryocytopoietic/thrombopoietic properties. Moreover, several lineage-dominant hematopoietic cytokines can also act as acute phase responders and contribute to the inflammation. This review focuses on the current literature and our experience regarding the dual relationships of the pathologic thrombopoiesis of RA. Growth factors contributing to it, namely interleukin (IL)-6, IL-11, stem cell factor, leukemia inhibitory factor, granulocyte colony stimulating factor, thrombopoietin (TPO), and the regulation of megakaryocytopoiesis during the inflammatory cascade are reviewed. Some data indicate that thrombopoietin could contribute to the reactive thrombocytosis of RA. In the non-lineage-specific gp130 cytokine family, IL-6 appears to predominate for the induction of megakaryopoiesis. However, other cytokines and growth factors may also contribute to the pathologic megakaryocytopoiesis of RA. Those pleiotropic mediators seem to act in concert to regulate this enigmatic process. Clarification of the pathobiologic basis of thrombopoiesis in RA may improve understanding of the disease pathogenesis and management of the inflammatory thrombocytosis.
Collapse
Affiliation(s)
- Ihsan Ertenli
- Department of Rheumatology, Hacettepe University School of Medicine, Ankara, Turkey
| | | | | | | | | | | |
Collapse
|
44
|
Lisignoli G, Toneguzzi S, Piacentini A, Cattini L, Lenti A, Tschon M, Cristino S, Grassi F, Facchini A. Human osteoblasts express functional CXC chemokine receptors 3 and 5: activation by their ligands, CXCL10 and CXCL13, significantly induces alkaline phosphatase and beta-N-acetylhexosaminidase release. J Cell Physiol 2003; 194:71-9. [PMID: 12447991 DOI: 10.1002/jcp.10188] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Osteoblasts (OBs) contribute to the maintenance of bone homeostasis and their activity can be influenced by immune cells localized in bone lacunae. We investigated the expression of the chemokine receptors in isolated human OBs by reverse transcriptase-polymerase chain reaction (RT-PCR) and flow cytometry, and report a novel finding, namely, that OBs express high levels of CXC chemokine receptor 3 (CXCR3) and 5 (CXCR5). Functional assays to evaluate CXCR3 and CXCR5 demonstrated that their ligands-CXCL10 and CXCL13, respectively-significantly induce the release of beta-N-acetylhexosaminidase, an enzyme involved in endochondral ossification and bone remodeling able to degrade important extracellular matrix components. Alkaline phosphatase activity, a useful index of matrix formation was also up-regulated by CXCL10 and CXCL13. However, OB activation by these ligands does not affect OB proliferation. Both Bordetella pertussis toxin and neutralizing anti-CXCR3/anti-CXCR5 monoclonal antibodies block CXCL10 and CXCL13 induction, respectively. We also demonstrated the expression of CXCL10 and CXCL13 in human bone tissue biopsies. These results indicate that both CXCR3/CXCL10 and CXCR5/CXCL13 receptor-ligand pairs may play an important role in OB activity through the specific up-regulation of two enzymes, which are involved in the bone remodeling process. Moreover, our data suggest that OBs may play a role in the modulation of bone formation through the combined action of these two enzymes.
Collapse
MESH Headings
- Alkaline Phosphatase/metabolism
- Cell Division/drug effects
- Cell Division/immunology
- Cells, Cultured
- Chemokine CXCL10
- Chemokine CXCL13
- Chemokines, CXC/metabolism
- Chemokines, CXC/pharmacology
- Exocytosis/immunology
- Extracellular Matrix/drug effects
- Extracellular Matrix/metabolism
- Humans
- Immunohistochemistry
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Middle Aged
- Osteoblasts/drug effects
- Osteoblasts/enzymology
- Osteoblasts/immunology
- Osteogenesis/drug effects
- Osteogenesis/immunology
- Receptors, CXCR3
- Receptors, CXCR5
- Receptors, Chemokine/drug effects
- Receptors, Chemokine/metabolism
- Receptors, Cytokine/drug effects
- Receptors, Cytokine/metabolism
- beta-N-Acetylhexosaminidases/metabolism
Collapse
Affiliation(s)
- Gina Lisignoli
- Laboratorio di Immunologia e Genetica, Istituti Ortopedici Rizzoli, Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Oncostatin M (OM) is a pleiotropic cytokine of the interleukin 6 family, whose in vivo properties and physiological function remain in dispute and poorly defined. These in vivo studies strongly suggest that OM is anabolic, promoting wound healing and bone formation, and anti-inflammatory. In models of inflammation OM is produced late in the cytokine response and protects from lipopolysaccharide (LPS)-induced toxicities, promoting the re-establishment of homoeostasis by cooperating with proinflammatory cytokines and acute phase molecules to alter and attenuate the inflammatory response. Administration of OM inhibited bacterial LPS-induced production of tumour necrosis factor alpha and septic lethality in a dose dependent manner. Consistent with these findings, in animal models of chronic inflammatory disease OM potently suppressed inflammation and tissue destruction in murine models of rheumatoid arthritis and multiple sclerosis. T cell function and antibody production were not impaired by OM treatment. Taken together, these data indicate that the activities of this cytokine in vivo are anti-inflammatory without concordant immunosuppression.
Collapse
Affiliation(s)
- A F Wahl
- Seattle Genetics, Inc, Bothell, WA 98021, USA.
| | | |
Collapse
|
46
|
Chenoufi HL, Diamant M, Rieneck K, Lund B, Stein GS, Lian JB. Increased mRNA expression and protein secretion of interleukin-6 in primary human osteoblasts differentiated in vitro from rheumatoid and osteoarthritic bone. J Cell Biochem 2001; 81:666-78. [PMID: 11329621 DOI: 10.1002/jcb.1104] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We have investigated the expression and synthesis of potential bone-resorbing cytokines, interleukin-6 (IL-6), interleukin-1 (IL-1), and tumor necrosis factor (TNF) in rheumatoid arthritic (RA) and osteoarthritic (OA) bone, two common diseases which are associated with bone loss. Primary human osteoblast (hOB) cultures were established to determine the temporal mRNA expression of IL-6, IL-1 (alpha and beta), and TNF (alpha and beta) in relation to osteoblast growth and phenotypic genes. IL-6 mRNA levels were found to be significantly higher (P < 0.04) in both OA hOB (17 patients) and RA hOB (10 patients) compared to normal (NO) hOB (9 patients) and reached five-fold increases in OA hOB and 13-fold increases in RA hOB. Maximal levels of IL-6 are expressed at Day 21 which corresponds to the mineralization stage reflected by decreasing collagen I (alpha(1)), osteopontin, bone sialoprotein, alkaline phosphatase mRNA levels, while osteocalcin (OC) mRNA levels increased. IL-6 protein levels also were significantly higher (P < 0.05) in OA hOB and RA hOB compared to NO hOB. These increases were not attributable to sex or age of the donor bone. Neither the mRNA encoding IL-1(alpha and beta) and TNF(alpha and beta) nor the related proteins were detectable. These results indicate that differentiated OA hOB and RA hOB within a bone tissue-like matrix constitutively express and secrete high levels of IL-6. This inherent property suggests that these osteoblasts, independent of local inflammatory parameters, can contribute to enhanced recruitment of osteoclast progenitors and thereby bone resorption.
Collapse
Affiliation(s)
- H L Chenoufi
- Department of Orthopaedic Surgery U-2161, Rigshospitalet, DK-2100 Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Bone tissue is continually being remodelled according to physiological circumstances. Two main cell populations (osteoblasts and osteoclasts) are involved in this process, and cellular activities (including cell differentiation) are modulated by hormones, cytokines and growth factors. Within the last 20 years, many factors involved in bone tissue metabolism have been found to be closely related to the inflammatory process. More recently, a cytokine family sharing a common signal transducer (gp130) had been identified, which appears to be a key factor in bone remodelling. This family includes interleukin 6, interleukin 11, oncostatin M, leukaemia inhibitory factor, ciliary neurotrophic factor and cardiotrophin-1. This paper provides an exhaustive review of recent knowledge on the involvement of gp130 cytokine family in bone cell (osteoblast, osteoclast, etc.) differentiation/activation and in osteoarticular pathologies.
Collapse
Affiliation(s)
- D Heymann
- Faculté de Médecine, Laboratoire de Physiopathologie de la Résorption Osseuse, 1 rue Gaston Veil, Nantes cedex 1, 44035, France.
| | | |
Collapse
|