101
|
Yu Y, Yang D, Qiu L, Okamura H, Guo J, Haneji T. Tumor necrosis factor-α induces interleukin-34 expression through nuclear factor‑κB activation in MC3T3-E1 osteoblastic cells. Mol Med Rep 2014; 10:1371-6. [PMID: 24970360 PMCID: PMC4121411 DOI: 10.3892/mmr.2014.2353] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 04/24/2014] [Indexed: 01/27/2023] Open
Abstract
Osteoblasts produce various types of cytokines under pathological conditions and control osteoclast differentiation. Tumor necrosis factor-α (TNF-α) has been demonstrated to exert complex effects in osteoblasts under local inflammatory conditions, including in periodontal and periapical diseases. Interleukin-34 (IL-34) has been recently identified as a novel regulatory factor for the differentiation and function of osteoclasts. The present study provides the first evidence, to the best of our knowledge, that the expression of IL-34 is induced by TNF-α through nuclear factor-κB (NF-κB) activation in MC3T3-E1 osteoblastic cells. TNF-α induced IL-34 expression in a dose- and time-dependent manner. Immunocytochemistry with an NF-κB antibody demonstrated that NF-κB was mainly localized in the cytoplasm of the untreated MC3T3-E1 cells. Rapid translocation of NF-κB from the cytoplasm to the nucleus was observed in the cells treated with TNF-α for 15 min. Translocation and transcriptional activity of NF-κB were also determined by western blotting and a luciferase reporter assay, respectively. Pretreatment with 100 μM CAPE, an inhibitor of NF-κB, significantly inhibited TNF-α-induced IL-34 expression. These results indicate that TNF-α induces IL-34 expression via NF-κB in osteoblasts.
Collapse
Affiliation(s)
- Yaqiong Yu
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Di Yang
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Lihong Qiu
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Hirohiko Okamura
- Department of Histology and Oral Histology, Institute of Health Biosciences, The University of Tokushima Graduate School, Kuramoto, Tokushima 770-8504, Japan
| | - Jiajie Guo
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Tatsuji Haneji
- Department of Histology and Oral Histology, Institute of Health Biosciences, The University of Tokushima Graduate School, Kuramoto, Tokushima 770-8504, Japan
| |
Collapse
|
102
|
Jenner F, IJpma A, Cleary M, Heijsman D, Narcisi R, van der Spek PJ, Kremer A, van Weeren R, Brama P, van Osch GJVM. Differential gene expression of the intermediate and outer interzone layers of developing articular cartilage in murine embryos. Stem Cells Dev 2014; 23:1883-98. [PMID: 24738827 DOI: 10.1089/scd.2013.0235] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Nascent embryonic joints, interzones, contain a distinct cohort of progenitor cells responsible for the formation of the majority of articular tissues. However, to date the interzone has largely been studied using in situ analysis for candidate genes in the context of the embryo rather than using an unbiased genome-wide expression analysis on isolated interzone cells, leaving significant controversy regarding the exact role of the intermediate and outer interzone layers in joint formation. Therefore, in this study, using laser capture microdissection (three biological replicates), we selectively harvested the intermediate and outer interzones of mouse embryos at gestational age 15.5 days, just prior to cavitation, when the differences between the layers should be most profound. Microarray analysis (Agilent Whole Mouse Genome Oligo Microarrays) was performed and the differential gene expression between the intermediate interzone cells and outer interzone cells was examined by performing a two-sided paired Student's t-test and pathway analysis. One hundred ninety-seven genes were differentially expressed (≥ 2-fold) between the intermediate interzone and the outer interzone with a P-value ≤ 0.01. Of these, 91 genes showed higher expression levels in the intermediate interzone and 106 were expressed higher in the outer interzone. Pathway analysis of differentially expressed genes suggests an important role for inflammatory processes in the interzone layers, especially in the intermediate interzone, and hence in joint and articular cartilage development. The high representation of genes relevant to chondrocyte hypertrophy and endochondral ossification in the outer interzone suggests that it undergoes endochondral ossification.
Collapse
Affiliation(s)
- Florien Jenner
- 1 Equine University Hospital, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna , Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Zhang H, Hilton MJ, Anolik JH, Welle SL, Zhao C, Yao Z, Li X, Wang Z, Boyce BF, Xing L. NOTCH inhibits osteoblast formation in inflammatory arthritis via noncanonical NF-κB. J Clin Invest 2014; 124:3200-14. [PMID: 24892805 DOI: 10.1172/jci68901] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/10/2014] [Indexed: 12/29/2022] Open
Abstract
NOTCH-dependent signaling pathways are critical for normal bone remodeling; however, it is unclear if dysfunctional NOTCH activation contributes to inflammation-mediated bone loss, as observed in rheumatoid arthritis (RA) patients. We performed RNA sequencing and pathway analyses in mesenchymal stem cells (MSCs) isolated from transgenic TNF-expressing mice, a model of RA, to identify pathways responsible for decreased osteoblast differentiation. 53 pathways were dysregulated in MSCs from RA mice, among which expression of genes encoding NOTCH pathway members and members of the noncanonical NF-κB pathway were markedly elevated. Administration of NOTCH inhibitors to RA mice prevented bone loss and osteoblast inhibition, and CFU-fibroblasts from RA mice treated with NOTCH inhibitors formed more new bone in recipient mice with tibial defects. Overexpression of the noncanonical NF-κB subunit p52 and RELB in a murine pluripotent stem cell line increased NOTCH intracellular domain-dependent (NICD-dependent) activation of an RBPjκ reporter and levels of the transcription factor HES1. TNF promoted p52/RELB binding to NICD, which enhanced binding at the RBPjκ site within the Hes1 promoter. Furthermore, MSC-enriched cells from RA patients exhibited elevated levels of HES1, p52, and RELB. Together, these data indicate that persistent NOTCH activation in MSCs contributes to decreased osteoblast differentiation associated with RA and suggest that NOTCH inhibitors could prevent inflammation-mediated bone loss.
Collapse
MESH Headings
- Animals
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Bone Resorption/metabolism
- Bone Resorption/pathology
- Bone Resorption/prevention & control
- Cell Differentiation
- Dipeptides/pharmacology
- Disease Models, Animal
- Gene Expression
- Homeodomain Proteins/genetics
- Humans
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mesenchymal Stem Cells/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- NF-kappa B/genetics
- NF-kappa B/metabolism
- NF-kappa B p52 Subunit/genetics
- NF-kappa B p52 Subunit/metabolism
- Osteoblasts/drug effects
- Osteoblasts/metabolism
- Osteoblasts/pathology
- Promoter Regions, Genetic
- Receptors, Notch/antagonists & inhibitors
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction
- Transcription Factor HES-1
- Transcription Factor RelB/genetics
- Transcription Factor RelB/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
|
104
|
Marie PJ. Bone cell senescence: mechanisms and perspectives. J Bone Miner Res 2014; 29:1311-21. [PMID: 24496911 DOI: 10.1002/jbmr.2190] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 12/15/2022]
Abstract
Age-related bone loss is in large part the consequence of senescence mechanisms that impact bone cell number and function. In recent years, progress has been made in the understanding of the molecular mechanisms underlying bone cell senescence that contributes to the alteration of skeletal integrity during aging. These mechanisms can be classified as intrinsic senescence processes, alterations in endogenous anabolic factors, and changes in local support. Intrinsic senescence mechanisms cause cellular dysfunctions that are not tissue specific and include telomere shortening, accumulation of oxidative damage, impaired DNA repair, and altered epigenetic mechanisms regulating gene transcription. Aging mechanisms that are more relevant to the bone microenvironment include alterations in the expression and signaling of local growth factors and altered intercellular communications. This review provides an integrated overview of the current concepts and interacting mechanisms underlying bone cell senescence during aging and how they could be targeted to reduce the negative impact of senescence in the aging skeleton.
Collapse
Affiliation(s)
- Pierre J Marie
- Inserm UMR-1132, Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
105
|
Tao K, Zeng H, Xiao DM, Xiong A, Weng J, Kang B. Influences of IL-6R antibody on PMMA bone cement-mediated expression of OPG and RANKL in synovial fibroblasts. ACTA ACUST UNITED AC 2014; 34:241-246. [PMID: 24710939 DOI: 10.1007/s11596-014-1265-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 12/02/2013] [Indexed: 11/24/2022]
Abstract
Effect of interleukin-6 receptor (IL-6R) antibody on polymethyl methacrylate (PMMA) bone cement-mediated expression of osteoprotegerin (OPG) and receptor activator of nuclear factor-kappaB ligand (RANKL) in synovial fibroblasts was investigated. Synovial tissue obtained from total knee arthroplasty was digested and cultured. Inverted microscope was employed to observe the synovial cells and immunocytochemistry (SABC method) staining was used to identify synovial fibroblasts. This experiment was divided into three groups according to different culture media: PMMA group (75 μg/mL PMMA bone cement particles), IL-6R antibody group (10 ng/mL IL-6R antibody+75 μg/mL PMMA bone cement particles), and control group (no IL-6R antibody or PMMA bone cement particles). Influence of IL-6R antibody and PMMA on proliferation of synovial fibroblasts was measured by cell counting kit-8 (CCK-8). ELISA method was used to measure OPG and RANKL levels in culture solution. Fluorescence quantitative real-time PCR (FQ-PCR) was used to detect the expression of OPG and RANKL mRNA. After three consecutive passages, more than 95% of the primary synovial cells became long spindle fibroblast-like cells. SABC staining results showed that the fibroblast-like cells were negative for anti-CD68 antibody and positive for anti-vimentin antibody, with brown madder stained. CCK-8 test demonstrated that the absorbance (A) value at 450 nm was significantly lower in IL-6R antibody group than in PMMA group and control group (P<0.01), but there was no statistically significant difference in A value at 450 nm between the control group and PMMA group (P>0.05). Results of ELISA indicated that the expression of OPG was significantly higher in IL-6R antibody group than in PMMA group and control group (P<0.01). The expression of RANKL was inhibited (P<0.05), and the ratio of OPG/RANKL was significantly increased in IL-6R antibody group as compared with PMMA group and control group. There was no significant difference in the expression of OPG between control group and PMMA group (P>0.05), but the expression of RANKL was higher in PMMA group than in control group (P<0.05), and there was a significant difference in the ratio of OPG/RANKL between them (P<0.05). Results of FQ-PCR revealed the expression of RANKL mRNA was significantly inhibited (P<0.01) and the expression of OPG mRNA was significantly increased (P<0.01) in IL-6R antibody group as compared with PMMA group and control group. The expression of RANKL mRNA was higher in PMMA group than in control group (P<0.05), but the expression of OPG mRNA had no significant difference between them (P>0.05). IL-6R antibody could significantly increase the expression of OPG, but inhibit the expression of RANKL, which might provide a theoretical basis of molecular biology for the prevention and treatment of aseptic loosening of prosthesis.
Collapse
Affiliation(s)
- Ke Tao
- Department of Orthopedics, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Hui Zeng
- Department of Orthopedics, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - De-Ming Xiao
- Department of Orthopedics, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Ao Xiong
- Department of Orthopedics, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Jian Weng
- Department of Orthopedics, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Bin Kang
- Department of Orthopedics, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| |
Collapse
|
106
|
Constitutive activation of IKK2/NF-κB impairs osteogenesis and skeletal development. PLoS One 2014; 9:e91421. [PMID: 24618907 PMCID: PMC3949987 DOI: 10.1371/journal.pone.0091421] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/11/2014] [Indexed: 12/17/2022] Open
Abstract
Pathologic conditions impair bone homeostasis. The transcription factor NF-κB regulates bone homeostasis and is central to bone pathologies. Whereas contribution of NF-κB to heightened osteoclast activity is well-documented, the mechanisms underlying NF-κB impact on chondrocytes and osteoblasts are scarce. In this study, we examined the effect of constitutively active IKK2 (IKK2ca) on chondrogenic and osteogenic differentiation. We show that retroviral IKK2ca but not GFP, IKK2WT, or the inactive IKK2 forms IKK2KM and IKK2SSAA, strongly suppressed osteogenesis and chondrogenesis, in vitro. In order to explore the effect of constitutive NF-κB activation on bone formation in vivo, we activated this pathway in a conditional fashion. Specifically, we crossed the R26StopIKK2ca mice with mice carrying the Col2-cre in order to express IKK2ca in osteoblasts and chondrocytes. Both chondrocytes and osteoblasts derived from Col2Cre/IKK2ca expressed IKK2ca. Mice were born alive yet died shortly thereafter. Histologically, newborn Col2Cre+/RosaIKK2ca heterozygotes (Cre+IKK2ca_w/f (het)) and homozygotes (Cre+IKK2ca_f/f (KI)) showed smaller skeleton, deformed vertebrate and reduced or missing digit ossification. The width of neural arches, as well as ossification in vertebral bodies of Cre+IKK2ca_w/f and Cre+IKK2ca_f/f, was reduced or diminished. H&E staining of proximal tibia from new born pups revealed that Cre+IKK2ca_f/f displayed disorganized hypertrophic zones within the smaller epiphysis. Micro-CT analysis indicated that 4-wk old Cre+IKK2ca_w/f has abnormal trabecular bone in proximal tibia compared to WT littermates. Mechanistically, ex-vivo experiments showed that expression of differentiation markers in calvarial osteoblasts derived from newborn IKK2ca knock-in mice was diminished compared to WT-derived cells. In situ hybridization studies demonstrated that the hypertrophic chondrocyte marker type-X collagen, the pre-hypertrophic chondrocyte markers Indian hedgehog and alkaline phosphatase, and the early markers Aggrecan and type-II collagen were reduced in Cre+IKK2ca_w/f and Cre+IKK2ca_f/f mice. Altogether, the in-vitro, in vivo and ex-vivo evidence suggest that IKK2ca perturbs osteoblast and chondrocyte maturation and impairs skeletal development.
Collapse
|
107
|
Wu CM, Chen PC, Li TM, Fong YC, Tang CH. Si-Wu-tang extract stimulates bone formation through PI3K/Akt/NF-κB signaling pathways in osteoblasts. Altern Ther Health Med 2013; 13:277. [PMID: 24156308 PMCID: PMC4015792 DOI: 10.1186/1472-6882-13-277] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 10/17/2013] [Indexed: 01/21/2023]
Abstract
BACKGROUND Si-Wu-Tang (SWT), a Traditional Chinese Medicine (TCM) formula, is widely used for the treatment of gynopathies diseases such as menstrual discomfort, climacteric syndrome, dysmenorrhea, and other estrogen-related diseases. Recent studies have shown that SWT can treat primary dysmenorrhea, have anti-pruritic anti-inflammatory effects, and protect against radiation-induced bone marrow damage in an animal model. It has been reported that anti-inflammatory and anti-oxidant agents have the potential to treat osteoporosis by increasing bone formation and/or suppressing bone resorption. However, the effect of SWT on bone cell function has not yet been reported. METHODS Alkaline phosphatase (ALP), bone morphogenetic proteins (BMP)-2, and osteopontin (OPN) mRNA expression was analyzed by qPCR. The mechanism of action of SWT extract was investigated using western blotting. The in vivo anti-osteoporotic effect of SWT extract was assessed in ovariectomized mice. RESULTS Here, we report that SWT increases ALP, BMP-2, and OPN expression as well as bone mineralization. In addition, we show that the PI3K, Akt, and NF-κB signaling pathways may be involved in the SWT-mediated increase in gene expression and bone mineralization. Notably, treatment of mice with SWT extract prevented bone loss induced by ovariectomy in vivo. CONCLUSION SWT may be used to stimulate bone formation for the treatment of osteoporosis.
Collapse
|
108
|
Wang M, Chen G, Lu C, Xiao C, Li L, Niu X, He X, Jiang M, Lu A. Rheumatoid arthritis with deficiency pattern in traditional chinese medicine shows correlation with cold and hot patterns in gene expression profiles. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:248650. [PMID: 24174973 PMCID: PMC3794642 DOI: 10.1155/2013/248650] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 07/15/2013] [Accepted: 07/17/2013] [Indexed: 12/14/2022]
Abstract
In our precious study, the correlation between cold and hot patterns in traditional Chinese medicine (TCM) and gene expression profiles in rheumatoid arthritis (RA) has been explored. Based on TCM theory, deficiency pattern is another key pattern diagnosis among RA patients, which leads to a specific treatment principle in clinical management. Therefore, a further analysis was performed aiming at exploring the characteristic gene expression profile of deficiency pattern and its correlation with cold and hot patterns in RA patients by bioinformatics analysis approach based on gene expression profiles data detected with microarray technology. The TCM deficiency pattern-related genes network comprises 7 significantly, highly connected regions which are mainly involved in protein transcription processes, protein ubiquitination, toll-like receptor activated NF- κ B regulated gene transcription and apoptosis, RNA clipping, NF- κ B signal, nucleotide metabolism-related apoptosis, and immune response processes. Toll-like receptor activated NF- κ B regulated gene transcription and apoptosis pathways are potential specific pathways related to TCM deficiency patterns in RA patients; TCM deficiency pattern is probably related to immune response. Network analysis can be used as a powerful tool for detecting the characteristic mechanism related to specific TCM pattern and the correlations between different patterns.
Collapse
Affiliation(s)
- Minzhi Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing 100700, China
| | - Gao Chen
- School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing 100700, China
| | - Cheng Xiao
- Sino-Japan Friendship Hospital, Beijing 100029, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing 100700, China
| | - Xuyan Niu
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing 100700, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing 100700, China
| | - Miao Jiang
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing 100700, China
| | - Aiping Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing 100700, China
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
- E-Institute of Shanghai Municipal Education Commission, Shanghai TCM University, Shanghai 201203, China
| |
Collapse
|
109
|
Sinomenine suppresses osteoclast formation and Mycobacterium tuberculosis H37Ra-induced bone loss by modulating RANKL signaling pathways. PLoS One 2013; 8:e74274. [PMID: 24066131 PMCID: PMC3774760 DOI: 10.1371/journal.pone.0074274] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 07/30/2013] [Indexed: 01/01/2023] Open
Abstract
Receptor activator of NF-κB ligand (RANKL) is essential for osteoclastogenesis. Targeting RANKL signaling pathways has been an encouraging strategy for treating lytic bone diseases such as osteoporosis and rheumatoid arthritis (RA). Sinomenine (SIN), derived from Chinese medicinal plant Sinomenioumacutum, is an active compound to treat RA, but its effect on osteoclasts has been hitherto unknown. In the present study, SIN was found to ameliorate M. tuberculosis H37Ra (Mt)-induced bone loss in rats with a decreased serum level of TRACP5b and RANKL, and an increased level of osteoprotegerin (OPG). In vitro study also showed that SIN could inhibit RANKL-induced osteoclast formation and bone resorption. The osteoclastic specific marker genes induced by RANKL including c-Src, MMP-9, TRACP were inhibited by SIN in a dose dependent manner. Signal transduction studies showed that SIN could obviously reduce the expression of RANK adaptor molecule TRAF6 and down-regulate RANKL-induced NF-κB activation. It decreased the RANKL-induced p38, JNK posphorylation but not ERK1/2 posphorylation. SIN could also reduce RANKL-mediated calcium influx which is associated with TRAF6/c-Src complex. Finally, SIN suppressed RANKL induced AP-1 and NFAT transcription, as well as the gene expression of NFATc1 and AP-1 components (Fra-1, Fra-2, c-Fos). The protein expression of c-Fos and TRAF6 were also inhibited by SIN after RANKL stimulation. Taken together, SIN could attenuate osteoclast formation and Mt-induced bone loss by mediating RANKL signaling pathways.
Collapse
|
110
|
|
111
|
Tokunaga F. Linear ubiquitination-mediated NF-κB regulation and its related disorders. J Biochem 2013; 154:313-23. [PMID: 23969028 DOI: 10.1093/jb/mvt079] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Ubiquitination is a post-translational modification involved in the regulation of a broad variety of cellular functions, such as protein degradation and signal transduction, including nuclear factor-κB (NF-κB) signalling. NF-κB is crucial for inflammatory and immune responses, and aberrant NF-κB signalling is implicated in multiple disorders. We found that linear ubiquitin chain assembly complex (LUBAC), composed of HOIL-1L, HOIP and SHARPIN, generates a novel type of Met1 (M1)-linked linear polyubiquitin chain and specifically regulates the canonical NF-κB pathway. Moreover, specific deubiquitinases, such as CYLD, A20 (TNFAIP3) and OTULIN/gumby, inhibit LUBAC-induced NF-κB activation by different molecular mechanisms, and several M1-linked ubiquitin-specific binding domains have been structurally defined. LUBAC and these linear ubiquitination-regulating factors contribute to immune and inflammatory processes and apoptosis. Functional impairments of these factors are correlated with multiple disorders, including autoinflammation, immunodeficiencies, dermatitis, B-cell lymphomas and Parkinson's disease. This review summarizes the molecular basis and the pathophysiological implications of the linear ubiquitination-mediated NF-κB activation pathway regulation by LUBAC.
Collapse
Affiliation(s)
- Fuminori Tokunaga
- Laboratory of Molecular Cell Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma 371-8912, Japan
| |
Collapse
|
112
|
Zhang Y, Otero JE, Abu-Amer Y. Ubiquitin-like domain of IKKβ regulates osteoclastogenesis and osteolysis. Calcif Tissue Int 2013; 93:78-85. [PMID: 23686246 PMCID: PMC3706195 DOI: 10.1007/s00223-013-9735-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 04/19/2013] [Indexed: 01/01/2023]
Abstract
The transcription factor NF-κB family is central for osteoclastogenesis and inflammatory osteolysis. Activation of NF-κB dimers is regulated by a kinase complex predominantly containing IKKα (IKK1), IKKβ (IKK2), and a regulatory subunit, IKKγ/NEMO. IKKα and IKKβ catalyze the cytoplasmic liberation and nuclear translocation of various NF-κB subunits. The requirement of IKKα and IKKβ for normal bone homeostasis has been established. Congruently, mice devoid of IKKα or IKKβ exhibit in vitro and in vivo defects in osteoclastogenesis, and IKKβ-null mice are refractory to inflammatory arthritis and osteolysis. To better understand the molecular mechanism underlying IKKβ function in bone homeostasis and bone pathologies, we conducted structure-function analysis to determine IKKβ functional domains in osteoclasts. IKKβ encompasses several domains, of which the ubiquitination-like domain (ULD) has been shown essential for IKKβ activation. In this study, we examined the role of ULD in IKKβ-mediated NF-κB activation in osteoclast precursors and its contribution to osteoclastogenesis and osteolysis. We generated and virally introduced IKKβ in which the ULD domain has been deleted (IKKβ∆ULD) into osteoclast progenitors. The results show that deletion of ULD diminishes IKKβ activity and that IKKβ∆ULD strongly inhibits osteoclastogenesis. In addition, unlike wild type (WT)-IKKβ, IKKβ∆ULD fail to restore RANKL-induced osteoclastogenesis by IKKβ-null precursors. Finally, we provide evidence that IKKβ∆ULD blocks inflammatory osteolysis in a model of murine calvarial osteolysis. Thus, we identified the ULD as crucial for IKKβ activity and osteoclastogenesis and found that ULD-deficient IKKβ is a potent inhibitor of osteoclastogenesis and osteolysis.
Collapse
Affiliation(s)
- Yanhong Zhang
- Department of Orthopaedic Surgery and Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110
- Department of Medicine, Raritan Bay Medical Center, 530 New Brunswick Ave, Perth Amboy, NJ 08861
| | - Jesse E. Otero
- University of Iowa Hospitals and Clinics, Department of Orthopaedic Surgery, Iowa City, Iowa 52242
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery and Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
113
|
Zhang H, Wu C, Matesic LE, Li X, Wang Z, Boyce BF, Xing L. Ubiquitin E3 ligase Itch negatively regulates osteoclast formation by promoting deubiquitination of tumor necrosis factor (TNF) receptor-associated factor 6. J Biol Chem 2013; 288:22359-68. [PMID: 23782702 DOI: 10.1074/jbc.m112.442459] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Itch is a ubiquitin E3 ligase that regulates protein stability. Itch(-/-) mice develop an autoimmune disease phenotype characterized by itchy skin and multiorgan inflammation. The role of Itch in the regulation of osteoclast function has not been examined. We report that Itch(-/-) bone marrow and spleen cells formed more osteoclasts than cells from WT littermates in response to receptor activator of NF-κB ligand (RANKL) and was associated with increased expression of the osteoclastogenic transcription factors c-fos and Nfatc1. Overexpression of Itch in Itch(-/-) cells rescued increased osteoclastogenesis. RANKL increased Itch expression, which can be blocked by a NF-κB inhibitor. The murine Itch promoter contains NF-κB binding sites. Overexpression of NF-κB p65 increased Itch expression, and RANKL promoted the binding of p65 onto the NF-κB binding sites in the Itch promoter. Itch(-/-) osteoclast precursors had prolonged RANKL-induced NF-κB activation and delayed TNF receptor-associated factor 6 (TRAF6) deubiquitination. In WT osteoclast precursors, Itch bound to TRAF6 and the deubiquitinating enzyme cylindromatosis. Adult Itch(-/-) mice had normal bone volume, but they had significantly increased LPS-induced osteoclastogenesis and bone resorption. Thus, Itch is a new RANKL target gene that is induced during osteoclastogenesis. Itch interacts with the deubiquitinating enzyme and is required for deubiquitination of TRAF6, thus limiting RANKL-induced osteoclast formation.
Collapse
Affiliation(s)
- Hengwei Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
114
|
Boyce BF. Advances in osteoclast biology reveal potential new drug targets and new roles for osteoclasts. J Bone Miner Res 2013; 28:711-22. [PMID: 23436579 PMCID: PMC3613781 DOI: 10.1002/jbmr.1885] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 01/22/2013] [Accepted: 01/28/2013] [Indexed: 01/06/2023]
Abstract
Osteoclasts are multinucleated myeloid lineage cells formed in response to macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL) by fusion of bone marrow-derived precursors that circulate in the blood and are attracted to sites of bone resorption in response to factors, such as sphingosine-1 phosphate signaling. Major advances in understanding of the molecular mechanisms regulating osteoclast functions have been made in the past 20 years, mainly from mouse and human genetic studies. These have revealed that osteoclasts express and respond to proinflammatory and anti-inflammatory cytokines. Some of these cytokines activate NF-κB and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) signaling to induce osteoclast formation and activity and also regulate communication with neighboring cells through signaling proteins, including ephrins and semaphorins. Osteoclasts also positively and negatively regulate immune responses and osteoblastic bone formation. These advances have led to development of new inhibitors of bone resorption that are in clinical use or in clinical trials; and more should follow, based on these advances. This article reviews current understanding of how bone resorption is regulated both positively and negatively in normal and pathologic states.
Collapse
Affiliation(s)
- Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
115
|
Marzaro G, Guiotto A, Borgatti M, Finotti A, Gambari R, Breveglieri G, Chilin A. Psoralen derivatives as inhibitors of NF-κB/DNA interaction: synthesis, molecular modeling, 3D-QSAR, and biological evaluation. J Med Chem 2013; 56:1830-42. [PMID: 23414143 DOI: 10.1021/jm3009647] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Some new psoralen derivatives were synthesized and evaluated as inhibitors of NF-κB/DNA interaction, with the aim to investigate the structural determinants required to inhibit this interaction. Starting from molecular docking studies, several possible protein binding sites were proposed and several three-dimensional quantitative structure-activity relationship (3D-QSAR) models were built using the docked poses of 29 (the most active psoralen in the series) as templates for alignment of the inhibitors. The selected best model was validated through the prediction of the activity of 17 novel compounds. All the experimental data agreed with the computational experiments, supporting the reliability of the computational approach. The hypothesis about the interaction with NF-κB was also supported by surface plasmon resonance based assays using compound 29. All the collected data allowed the identification of compound 29 as a potential candidate for the development of pharmaceutical strategies against the inflammatory phenotype of cystic fibrosis.
Collapse
Affiliation(s)
- Giovanni Marzaro
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Via Marzolo 5, 35131 Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
116
|
Lerner UH. Osteoblasts, Osteoclasts, and Osteocytes: Unveiling Their Intimate-Associated Responses to Applied Orthodontic Forces. Semin Orthod 2012. [DOI: 10.1053/j.sodo.2012.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
117
|
Kim HJ, Yoon KA, Lee MK, Kim SH, Lee IK, Kim SY. A novel small molecule, NecroX-7, inhibits osteoclast differentiation by suppressing NF-κB activity and c-Fos expression. Life Sci 2012; 91:928-934. [PMID: 23000100 DOI: 10.1016/j.lfs.2012.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 08/30/2012] [Accepted: 09/07/2012] [Indexed: 12/25/2022]
Abstract
AIMS Osteoclasts, the unique bone-resorbing polykaryons, are responsible for many bone-destructive diseases, such as osteoporosis and rheumatoid arthritis. Hence, the regulation of osteoclast formation is considered a potential therapeutic approach for these diseases. In this study, we investigated the effect of a novel small compound, C(25)H(32)N(4)O(4)S(2) (NecroX-7) on osteoclast formation. MAIN METHODS We analyzed the effects of NecoX-7 on receptor activator of nuclear factor kappa B ligand (RANKL)-induced osteoclast differentiation in vitro and LPS-induced bone loss in vivo. KEY FINDINGS We observed that NecroX-7 suppressed osteoclast formation from primary bone marrow macrophages (BMMs) in a dose-dependent manner. NecroX-7 significantly inhibited the NF-κB signaling pathway without affecting the activation of the mitogen-activated protein kinases (MAPKs) JNK, p38, and ERK in response to RANKL. In addition, NecroX-7 strongly attenuated the induction of c-Fos and nuclear factor of activated T cells c1 (NFATc1), which are crucial transcription factors for osteoclast differentiation. Mirroring the down-regulation of c-Fos and NFATc1, the expression of osteoclastogenic markers, such as tartrate-resistant acid phosphatase (TRAP) and cathepsin K, was also reduced by the addition of NecroX-7. Furthermore, confirming the in vitro anti-osteoclastogenic effect, NecroX-7 inhibited lipopolysaccharide (LPS)-induced bone loss in vivo. SIGNIFICANCE Our data imply that NecroX-7 is useful as a therapeutic drug for the treatment of bone resorption-associated diseases.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Skeletal Diseases Genome Research Center, Kyungpook National University and Hospital, Daegu 700-412, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
118
|
Zhao C, Xiu Y, Ashton J, Xing L, Morita Y, Jordan CT, Boyce BF. Noncanonical NF-κB signaling regulates hematopoietic stem cell self-renewal and microenvironment interactions. Stem Cells 2012; 30:709-18. [PMID: 22290873 DOI: 10.1002/stem.1050] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RelB and nuclear factor κB (NF-κB2) are the main effectors of NF-κB noncanonical signaling and play critical roles in many physiological processes. However, their role in hematopoietic stem/progenitor cell (HSPC) maintenance has not been characterized. To investigate this, we generated RelB/NF-κB2 double-knockout (dKO) mice and found that dKO HSPCs have profoundly impaired engraftment and self-renewal activity after transplantation into wild-type recipients. Transplantation of wild-type bone marrow cells into dKO mice to assess the role of the dKO microenvironment showed that wild-type HSPCs cycled more rapidly, were more abundant, and had developmental aberrancies: increased myeloid and decreased lymphoid lineages, similar to dKO HSPCs. Notably, when these wild-type cells were returned to normal hosts, these phenotypic changes were reversed, indicating a potent but transient phenotype conferred by the dKO microenvironment. However, dKO bone marrow stromal cell numbers were reduced, and bone-lining niche cells supported less HSPC expansion than controls. Furthermore, increased dKO HSPC proliferation was associated with impaired expression of niche adhesion molecules by bone-lining cells and increased inflammatory cytokine expression by bone marrow cells. Thus, RelB/NF-κB2 signaling positively and intrinsically regulates HSPC self-renewal and maintains stromal/osteoblastic niches and negatively and extrinsically regulates HSPC expansion and lineage commitment through the marrow microenvironment.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
119
|
Tokunaga F, Iwai K. LUBAC, a novel ubiquitin ligase for linear ubiquitination, is crucial for inflammation and immune responses. Microbes Infect 2012; 14:563-72. [DOI: 10.1016/j.micinf.2012.01.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 01/19/2012] [Indexed: 10/14/2022]
|
120
|
Abstract
Both embryonic and adult neurogenesis involves the self-renewal/proliferation, survival, migration and lineage differentiation of neural stem/progenitor cells. Such dynamic process is tightly regulated by intrinsic and extrinsic factors and complex signaling pathways. Misregulated neurogenesis contributes much to a large range of neurodevelopmental defects and neurodegenerative diseases. The signaling of NFκB regulates many genes important in inflammation, immunity, cell survival and neural plasticity. During neurogenesis, NFκB signaling mediates the effect of numerous niche factors such as cytokines, chemokines, growth factors, extracellular matrix molecules, but also crosstalks with other signaling pathways such as Notch, Shh, Wnt/β-catenin. This review summarizes current progress on the NFκB signaling in all aspects of neurogenesis, focusing on the novel role of NFκB signaling in initiating early neural differentiation of neural stem cells and embryonic stem cells.
Collapse
Affiliation(s)
- Yonggang Zhang
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | |
Collapse
|
121
|
Otero JE, Chen T, Zhang K, Abu-Amer Y. Constitutively active canonical NF-κB pathway induces severe bone loss in mice. PLoS One 2012; 7:e38694. [PMID: 22685599 PMCID: PMC3369901 DOI: 10.1371/journal.pone.0038694] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/14/2012] [Indexed: 12/23/2022] Open
Abstract
Physiologic osteoclastogenesis entails activation of multiple signal transduction pathways distal to the cell membrane receptor RANK. However, atypical osteoclastogenesis driven by pro-inflammatory stimuli has been described. We have reported recently a novel mechanism whereby endogenous mutational activation of the classical NF-κB pathway is sufficient to induce RANKL/RANK-independent osteoclastogenesis. Here we investigate the physiologic relevance of this phenomenon in vivo. Using a knock-in approach, the active form of IKK2, namely IKK2SSEE, was introduced into the myeloid lineage with the aid of CD11b-cre mice. Phenotypic assessment revealed that expression of IKK2SSEE in the myeloid compartment induced significant bone loss in vivo. This observation was supported by a dramatic increase in the number and size of osteoclasts in trabecular regions, elevated levels of circulating TRACP-5b, and reduced bone volume. Mechanistically, we observed that IKK2SSEE induced high expression of not only p65 but also p52 and RelB; the latter two molecules are considered exclusive members of the alternative NF-κB pathway. Intriguingly, RelB and P52 were both required to mediate the osteoclastogenic effect of IKK2SSEE and co-expression of these two proteins was sufficient to recapitulate osteoclastogenesis in the absence of RANKL or IKK2SSEE. Furthermore, we found that NF-κB2/p100 is a potent inhibitor of IKK2SSEE-induced osteoclastogenesis. Deletion of p52 enabled more robust osteoclast formation by the active kinase. In summary, molecular activation of IKK2 may play a role in conditions of pathologic bone destruction, which may be refractory to therapeutic interventions targeting the proximal RANKL/RANK signal.
Collapse
Affiliation(s)
- Jesse E. Otero
- Department of Orthopedic Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa, United States of America
| | - Tim Chen
- Department of Orthopedic Surgery and Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Kaihua Zhang
- Department of Orthopedic Surgery and Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Yousef Abu-Amer
- Department of Orthopedic Surgery and Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
122
|
Corilagin is a potent inhibitor of NF-kappaB activity and downregulates TNF-alpha induced expression of IL-8 gene in cystic fibrosis IB3-1 cells. Int Immunopharmacol 2012; 13:308-15. [PMID: 22561123 DOI: 10.1016/j.intimp.2012.04.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 04/04/2012] [Accepted: 04/18/2012] [Indexed: 01/21/2023]
Abstract
Corilagin (beta-1-O-galloyl-3,6-(R)-hexahydroxydiphenoyl-d-glucose), a gallotannin identified in several plants, including Phyllanthus urinaria, has been shown to exhibit versatile medicinal activities. As far as possible anti-inflammatory effects of corilagin, only few reports are available, and the potential use of corilagin as possible therapeutic molecule for cystic fibrosis has not been evaluated. In the present paper we report experiments aimed at determining the activity of corilagin on nuclear factor kappaB (NF-kappaB) binding to DNA target and on the expression of the major pro-inflammatory gene involved in cystic fibrosis, interleukin-8 (IL-8). Both IL-8 mRNA content and IL-8 protein secretion were analyzed in cystic fibrosis bronchial IB3-1 cells stimulated by tumor necrosis factor-alpha (TNF-alpha), one of the most potent pro-inflammatory agents. The data obtained demonstrate that corilagin binds to NF-kappaB, inhibits NF-kappaB/DNA interactions and affects IL-8 gene expression in TNF-alpha treated IB3-1 cells. In addition, corilagin inhibits TNF-alpha induced secretion of MCP-1 and RANTES, exhibiting low or no effect on the release of G-CSF, IL-6 and VEGF. Therefore, corilagin might be of interest for experimental anti-inflammatory therapy of cystic fibrosis.
Collapse
|
123
|
Li TM, Huang HC, Su CM, Ho TY, Wu CM, Chen WC, Fong YC, Tang CH. Cistanche deserticola extract increases bone formation in osteoblasts. ACTA ACUST UNITED AC 2012; 64:897-907. [PMID: 22571269 DOI: 10.1111/j.2042-7158.2012.01483.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES We investigated the effect of Cistanche deserticola Ma. (CD) on bone formation by cultured osteoblasts. METHODS The mineralized nodule formation assay was used to examine the in-vitro effects of CD on bone formation. Alkaline phosphatase (ALP), bone morphogenetic proteins (BMP)-2 and osteopontin (OPN) mRNA expression was analysed by quantitative real-time polymerase chain reaction. The mechanism of action of CD extract was investigated using Western blotting. The in-vivo anti-osteoporotic effect of CD extract was assessed in ovariectomized mice. KEY FINDINGS CD extract had no effect on the proliferation, migration or wound healing of cultured osteoblasts, but increased ALP, BMP-2 and OPN mRNA and bone mineralization. Mitogen-activated protein kinase (MAPK) or nuclear factor (NF)-κB inhibitors reduced CD extract-induced bone formation and ALP, BMP-2 and OPN expression. However, CD extract did not affect osteoclastogenesis. In addition, CD extract prevented the bone loss induced by ovariectomy in vivo. CONCLUSIONS CD may be a novel bone formation agent for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Te-Mao Li
- School of Chinese Medicine Chi-Mei Medical Center, Tainan, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
124
|
Li J, Zhao Q, Wang E, Zhang C, Wang G, Yuan Q. Transplantation of Cbfa1-overexpressing adipose stem cells together with vascularized periosteal flaps repair segmental bone defects. J Surg Res 2012; 176:e13-20. [PMID: 22341352 DOI: 10.1016/j.jss.2011.12.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 11/03/2011] [Accepted: 12/06/2011] [Indexed: 10/14/2022]
Abstract
BACKGROUND Segmental bone defect is still a challenge to orthopedic surgeons. Currently available therapies for segmental bone defects have some drawbacks. Tissue engineering using pluripotent stem cells is a new, promising method for bone repair. The present study aims to promote the effect of bone defect repair using the tissue engineered bone in combination with vascularized periosteal flaps. METHODS The adenoviral vector carrying Cbfa1 transduced rabbit adipose-derived mesenchymal stem cells and gene modified tissue engineering bone (GMB) were constructed. Rabbits with radial defects were implanted with the GMB together with vascularized periosteum (group A); or GMB with free periosteum (group B); or GMB (group C), and scaffold (group D). The bone repair effect was evaluated at 4, 8, or 12 wk, respectively, after the operations. RESULTS Cbfa1 proteins were strongly expressed in adipose stem cells (ADSCs) that formed a stratified network on the inner surface of the polylactic acid/ polycaprolacton (PLA/PCL) pores. Bone repair was well achieved in the rabbits treated with the Cbfa1-expressing ADSCs and vascularized flap that was markedly better than those treated with either Cbfa1-expressing ADSCs alone or with vascularized flap alone. CONCLUSIONS Combination with implanting the Cbfa1 gene-modified tissue-engineered bone and vascularized periosteum can better repair the segmental bone defects by stimulating osteogenesis, osteoinduction, and osteoconduction than using either one of the approaches.
Collapse
Affiliation(s)
- Jianjun Li
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, PR China
| | | | | | | | | | | |
Collapse
|
125
|
Tokunaga F, Iwai K. Linear ubiquitination: a novel NF-κB regulatory mechanism for inflammatory and immune responses by the LUBAC ubiquitin ligase complex. Endocr J 2012; 59:641-52. [PMID: 22673407 DOI: 10.1507/endocrj.ej12-0148] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The NF-κB pathway is a central signaling pathway for inflammatory and immune responses, and aberrant NF-κB signaling is implicated multiple disorders, such as cancer and autoimmune, chronic inflammatory and metabolic diseases. NF-κB is regulated by various post-translational modifications, including phosphorylation and multiple ubiquitinations. We determined that LUBAC (linear ubiquitin chain assembly complex), composed of SHARPIN, HOIL-IL and HOIP, generates a novel type of Met1-linked linear polyubiquitin chain and specifically regulates the canonical NF-κB pathway via the linear ubiquitination of NEMO and RIP1. In the absence of LUBAC components, NF-κB signaling was attenuated and induced apoptosis and inflammation. Many studies on the pathophysiological functions of LUBAC, such as in B cell development, innate immune response, carcinogenesis, and osteogenesis, have been performed recently. This review summarizes these new findings on LUBAC- and linear ubiquitination-mediated NF-κB regulation and their implications in disorders.
Collapse
Affiliation(s)
- Fuminori Tokunaga
- Laboratory of Molecular Cell Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan.
| | | |
Collapse
|
126
|
Beck GR, Ha SW, Camalier CE, Yamaguchi M, Li Y, Lee JK, Weitzmann MN. Bioactive silica-based nanoparticles stimulate bone-forming osteoblasts, suppress bone-resorbing osteoclasts, and enhance bone mineral density in vivo. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 8:793-803. [PMID: 22100753 DOI: 10.1016/j.nano.2011.11.003] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 08/29/2011] [Accepted: 11/03/2011] [Indexed: 12/25/2022]
Abstract
Bone is a dynamic tissue that undergoes renewal throughout life in a process whereby osteoclasts resorb worn bone and osteoblasts synthesize new bone. Imbalances in bone turnover lead to bone loss and development of osteoporosis and ultimately fracture, a debilitating condition with high morbidity and mortality. Silica is a ubiquitous biocontaminant that is considered to have high biocompatibility. The authors report that silica nanoparticles (NPs) mediate potent inhibitory effects on osteoclasts and stimulatory effects on osteoblasts in vitro. The mechanism of bioactivity is a consequence of an intrinsic capacity to antagonize activation of NF-κB, a signal transduction pathway required for osteoclastic bone resorption but inhibitory to osteoblastic bone formation. We further demonstrate that silica NPs promote a significant enhancement of bone mineral density (BMD) in mice in vivo, providing a proof of principle for the potential application of silica NPs as a pharmacological agent to enhance BMD and protect against bone fracture.
Collapse
Affiliation(s)
- George R Beck
- The Division of Endocrinology, Metabolism and Lipids, Emory University School of Medicine, Atlanta, Georgia 30322-0001, USA
| | | | | | | | | | | | | |
Collapse
|
127
|
Borgatti M, Chilin A, Piccagli L, Lampronti I, Bianchi N, Mancini I, Marzaro G, Francesco dall’Acqua, Guiotto A, Gambari R. Development of a novel furocoumarin derivative inhibiting NF-κB dependent biological functions: Design, synthesis and biological effects. Eur J Med Chem 2011; 46:4870-7. [DOI: 10.1016/j.ejmech.2011.07.032] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 07/13/2011] [Accepted: 07/18/2011] [Indexed: 12/15/2022]
|
128
|
Yamaguchi M, Neale Weitzmann M. The intact strontium ranelate complex stimulates osteoblastogenesis and suppresses osteoclastogenesis by antagonizing NF-κB activation. Mol Cell Biochem 2011; 359:399-407. [DOI: 10.1007/s11010-011-1034-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 08/05/2011] [Indexed: 10/17/2022]
|
129
|
Platt MP, Soler Z, Metson R, Stankovic KM. Pathways analysis of molecular markers in chronic sinusitis with polyps. Otolaryngol Head Neck Surg 2011; 144:802-8. [PMID: 21493366 DOI: 10.1177/0194599810395091] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To perform a comprehensive molecular pathways analysis of genes identified through genome-wide expression profiling and the published literature for chronic sinusitis with polyps. STUDY DESIGN Molecular pathways analysis. SETTING Academic medical center. METHODS A molecular pathways analysis of gene biomarkers discovered through hypothesis-driven and high-throughput molecular studies was performed. Genes identified with a PubMed literature search were analyzed with Ingenuity Pathways Analysis software to identify central molecules implicated in the pathogenesis of chronic sinusitis with polyps. The central pathways were then compared with those identified through genome-wide expression profiling of ethmoid polyps. RESULTS A total of 97 molecules were investigated with Ingenuity Pathways Analysis based on 55 studies that evaluated differences in gene expression (39), genetic variation (12), or proteomics (4). The analysis revealed 9 statistically significant molecular networks containing central nodes that included transcription factors, protein kinases, cytokines, and growth factors/receptors. The highest scoring networks implicated nuclear factor kappa-B, tumor necrosis factor, and mitogen-activated protein kinases. The majority of pathways in the literature review analysis overlapped with those identified through a single genome-wide expression study. CONCLUSIONS Chronic sinusitis with polyps is a complex disease with suspected contribution of multiple genetic and environmental factors. The search for causative genes has led to the discovery of numerous candidates. Pathways analysis applied to these candidate genes identified common central molecules that are likely to be key mediators of the disease process. Novel therapies targeting these molecules may be applicable for the treatment of chronic sinusitis with polyps.
Collapse
Affiliation(s)
- Michael P Platt
- Department of Otolaryngology-Head and Neck Surgery, Boston University, Boston, Massachusetts 02118, USA.
| | | | | | | |
Collapse
|
130
|
Yamaguchi M, Weitzmann MN. Zinc stimulates osteoblastogenesis and suppresses osteoclastogenesis by antagonizing NF-κB activation. Mol Cell Biochem 2011; 355:179-86. [DOI: 10.1007/s11010-011-0852-z] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 04/20/2011] [Indexed: 12/21/2022]
|
131
|
Diverse effects of Porphyromonas gingivalis on human osteoclast formation. Microb Pathog 2011; 51:149-55. [PMID: 21539907 DOI: 10.1016/j.micpath.2011.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 04/12/2011] [Accepted: 04/20/2011] [Indexed: 11/21/2022]
Abstract
Porphyromonas gingivalis is associated with periodontitis, a chronic inflammatory disease of the tooth-supporting tissues. A major clinical symptom is alveolar bone loss due to excessive resorption by osteoclasts. P. gingivalis may influence osteoclast formation in diverse ways; by interacting directly with osteoclast precursors that likely originate from peripheral blood, or indirectly by activating gingival fibroblasts, cells that can support osteoclast formation. In the present study we investigated these possibilities. Conditioned medium from viable or dead P. gingivalis, or from gingival fibroblasts challenged with viable or dead P. gingivalis were added to human mononuclear osteoclast precursors. After 21 days of culture the number of multinucleated (≥3 nuclei) tartrate resistant acid phosphatase (TRACP)-positive cells was determined as a measure for osteoclast formation. Conditioned medium from viable P. gingivalis, and from fibroblasts with viable P. gingivalis stimulated osteoclast formation (1.6-fold increase p < 0.05). Conditioned medium from dead bacteria had no effect on osteoclast formation, whereas conditioned medium from fibroblasts with dead bacteria stimulated formation (1.4-fold increase, p < 0.05). Inhibition of P. gingivalis LPS activity by Polymyxin B reduced the stimulatory effect of conditioned medium. Interestingly, when RANKL and M-CSF were added to cultures, conditioned media inhibited osteoclast formation (0.6-0.7-fold decrease, p < 0.05). Our results indicate that P. gingivalis influences osteoclast formation in vitro in different ways. Directly, by bacterial factors, likely LPS, or indirectly, by cytokines produced by gingival fibroblasts in response to P. gingivalis. Depending on the presence of RANKL and M-CSF, the effect of P. gingivalis is either stimulatory or inhibitory.
Collapse
|
132
|
Huang L, Mackenzie G, Ouyang N, Sun Y, Xie G, Johnson F, Komninou D, Rigas B. The novel phospho-non-steroidal anti-inflammatory drugs, OXT-328, MDC-22 and MDC-917, inhibit adjuvant-induced arthritis in rats. Br J Pharmacol 2011; 162:1521-33. [PMID: 21175575 PMCID: PMC3057290 DOI: 10.1111/j.1476-5381.2010.01162.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2010] [Revised: 11/01/2010] [Accepted: 11/03/2010] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE The use of non-steroidal anti-inflammatory drugs (NSAIDs) in the treatment of rheumatoid arthritis (RA) is limited by their toxicity. We evaluated the anti-inflammatory efficacy and safety of three novel modified NSAIDs, phospho-aspirin, phospho-ibuprofen and phospho-sulindac. EXPERIMENTAL APPROACH We determined the anti-inflammatory effects and gastrointestinal safety of the phospho-NSAIDs in the rat adjuvant arthritis model and studied their mechanism of action in cultured cells, Cytokines were measured with elisa and activation of nuclear factor-κB (NF-κB) by immunohistochemistry. KEY RESULTS All three phospho-NSAIDs showed less gastrointestinal toxicity than their parent compounds and demonstrated strong anti-inflammatory effects, essentially reversing joint inflammation and oedema. They have a broad but not uniform effect on the expression of relevant cytokines, in general decreasing IL-6 and IL-1β and increasing IL-10 levels in rat plasma and cultured cells. Phospho-sulindac and phospho-ibuprofen but not phospho-aspirin suppressed PGE(2) production in vitro, whereas phospho-aspirin (in contrast to aspirin) showed the same effect in vivo. In joint tissues, phospho-aspirin inhibited NF-κB activation, and suppressed inflammation and bone resorption. Phospho-aspirin also inhibited Jurkat T cell proliferation. In general, phospho-aspirin had greater efficacy but different effects upon inflammatory mediators compared with aspirin. The chemical modification of the parent NSAIDs seems crucial for their safety and efficacy. CONCLUSIONS AND IMPLICATIONS Phospho-aspirin, phospho-ibuprofen and phospho-sulindac were safer than their parent NSAIDs, were highly effective in rat adjuvant arthritis and inhibited many key mediators in the pathophysiology of RA. These novel compounds are promising candidate drugs for the treatment of RA and merit further evaluation.
Collapse
Affiliation(s)
- L Huang
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
133
|
Mahoney DJ, Swales C, Athanasou NA, Bombardieri M, Pitzalis C, Kliskey K, Sharif M, Day AJ, Milner CM, Sabokbar A. TSG-6 inhibits osteoclast activity via an autocrine mechanism and is functionally synergistic with osteoprotegerin. ARTHRITIS AND RHEUMATISM 2011; 63:1034-43. [PMID: 21162099 DOI: 10.1002/art.30201] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE TSG-6 (the product of tumor necrosis factor [TNF]-stimulated gene 6) has a potent inhibitory effect on RANKL-mediated bone erosion. The aim of this study was to compare the activity of TSG-6 with that of osteoprotegerin (OPG) and to investigate its role as an autocrine modulator of cytokine-mediated osteoclast formation/activation. We also determined TSG-6 expression in inflammatory joint disease. METHODS The effects of TSG-6, OPG, and the inflammation mediators TNFα, interleukin-1 (IL-1), and IL-6 on the formation of osteoclasts from peripheral blood mononuclear cells and synovial fluid (SF) macrophages were determined by tartrate-resistant acid phosphatase staining. Lacunar resorption and filamentous actin ring formation were measured as indicators of osteoclast activity. The amount of TSG-6 in culture media or SF was quantified by enzyme-linked immunosorbent assay, and expression of TSG-6 in synovial tissue was assessed by immunohistochemistry. RESULTS TSG-6 acted in synergy with OPG to inhibit RANKL-mediated bone resorption and was produced by osteoclast precursors and mature osteoclasts in response to TNFα, IL-1, and IL-6. Expression of TSG-6 correlated with inhibition of lacunar resorption; this effect was ameliorated by an anti-TSG-6 antibody. The level of TSG-6 protein was determined in SF from patients with various arthritides; it was highest in patients with inflammatory conditions such as rheumatoid arthritis, in which it correlated with the amount of TSG-6 immunostaining in the synovium. TSG-6 inhibited the activation but not the formation of osteoclasts from SF macrophages. CONCLUSION In the presence of inflammatory cytokines, osteoclasts produced TSG-6 at concentrations that are sufficient to inhibit lacunar resorption. This may represent an autocrine mechanism to limit the degree of bone erosion during joint inflammation.
Collapse
|
134
|
Inflammation as death or life signal in diabetic fracture healing. Inflamm Res 2010; 60:3-10. [PMID: 20845059 DOI: 10.1007/s00011-010-0246-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 08/03/2010] [Accepted: 08/25/2010] [Indexed: 12/11/2022] Open
Abstract
Increased apoptosis of chondrocytes and osteoblasts and prolonged survival of osteoclasts lead to early destruction of callus tissue and impair bone remodeling in fracture healing of diabetic patients. Diabetes is accompanied by an increased inflammatory state, reactive oxygen species (ROS) generation and accumulation of advanced glycation end products (AGEs), a heterogenous group of toxic metabolites that can induce inflammation. Prolonged hyperglycemia and insulin resistance correlate with increased apoptosis rate and, accordingly, the proapoptotic role of several inflammatory mediators, ROS and AGEs has been also documented. In this review we summarize the most recent reports supporting the idea that inflammatory signaling increases chondrocyte and osteoblast death and prolongs osteoclast survival, resulting in impaired bone regeneration in diabetes. Antagonising inflammatory signal pathways and solution of inflammation may deserve greater attention in the management of diabetic fracture healing.
Collapse
|