Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biofactors ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38777369

ABSTRACT

Drug-induced gingival enlargement (DIGE) is a side effect of ciclosporin, calcium channel blockers, and phenytoin. DIGE is a serious disease that leads to masticatory and esthetic disorders, severe caries, and periodontitis but currently has no standard treatment. We recently reported that nuclear receptor 4A1 (NR4A1) is a potential therapeutic target for DIGE. This study aimed to evaluate the therapeutic effects of n-butylidenephthalide (BP), which increases the expression of NR4A1, on DIGE. In this study, NR4A1 mRNA expression was analyzed in the patients with periodontal disease (PD) and DIGE. We evaluated the effect of BP on NR4A1 expression in gingival fibroblasts and in a DIGE mouse model. RNA sequencing (RNA-seq) was conducted to identify the mechanisms by which BP increases NR4A1 expression. The results showed that NR4A1 mRNA expression in the patients with DIGE was significantly lower than the patients with PD. BP suppressed the upregulation of COL1A1 expression, which was upregulated by TGF-ß. BP also ameliorated gingival overgrowth in DIGE mice and reduced Col1a1 and Pai1 expression. BP also decreased Il1ß mRNA expression in gingival tissue in DIGE. RNA-seq results showed an increase in the expression of several genes related to mitogen-activated protein kinase including DUSP genes in gingival fibroblasts stimulated by BP. Treatment with ERK and JNK inhibitors suppressed the BP-induced increase in NR4A1 expression. In addition, BP promoted the phosphorylation of ERK in gingival fibroblasts. In conclusion, BP increases NR4A1 expression in gingival fibroblasts through ERK and JNK signaling, demonstrating its potential as a preventive and therapeutic agent against DIGE.

2.
JBMR Plus ; 8(6): ziae050, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38699440

ABSTRACT

Cherubism (OMIM 118400) is a rare craniofacial disorder in children characterized by destructive jawbone expansion due to the growth of inflammatory fibrous lesions. Our previous studies have shown that gain-of-function mutations in SH3 domain-binding protein 2 (SH3BP2) are responsible for cherubism and that a knock-in mouse model for cherubism recapitulates the features of cherubism, such as increased osteoclast formation and jawbone destruction. To date, SH3BP2 is the only gene identified to be responsible for cherubism. Since not all patients clinically diagnosed with cherubism had mutations in SH3BP2, we hypothesized that there may be novel cherubism genes and that these genes may play a role in jawbone homeostasis. Here, using whole exome sequencing, we identified homozygous loss-of-function variants in the opioid growth factor receptor like 1 (OGFRL1) gene in 2 independent autosomal recessive cherubism families from Syria and India. The newly identified pathogenic homozygous variants were not reported in any variant databases, suggesting that OGFRL1 is a novel gene responsible for cherubism. Single cell analysis of mouse jawbone tissue revealed that Ogfrl1 is highly expressed in myeloid lineage cells. We generated OGFRL1 knockout mice and mice carrying the Syrian frameshift mutation to understand the in vivo role of OGFRL1. However, neither mouse model recapitulated human cherubism or the phenotypes exhibited by SH3BP2 cherubism mice under physiological and periodontitis conditions. Unlike bone marrow-derived M-CSF-dependent macrophages (BMMs) carrying the SH3BP2 cherubism mutation, BMMs lacking OGFRL1 or carrying the Syrian mutation showed no difference in TNF-ɑ mRNA induction by LPS or TNF-ɑ compared to WT BMMs. Osteoclast formation induced by RANKL was also comparable. These results suggest that the loss-of-function effects of OGFRL1 in humans differ from those in mice and highlight the fact that mice are not always an ideal model for studying rare craniofacial bone disorders.

3.
Stem Cell Rev Rep ; 20(1): 347-361, 2024 01.
Article in English | MEDLINE | ID: mdl-37917410

ABSTRACT

Mesenchymal stem cells (MSCs) have gained significant attention in cell therapies due to their multipotency and immunomodulatory capacities. The transcriptional co-activators YAP/TAZ, central to the mechanotransduction system in MSCs, dominantly direct MSCs lineage commitment. However, their role in immunomodulation remains elusive. Accordingly, this present study aimed to investigate the role of mechanotransducer YAP/TAZ and their binding target transcriptional factor, TEAD, in the immunomodulatory capacities of human bone marrow-derived MSCs. Reducing YAP/TAZ activity by altering the matrix stiffness, disrupting the F-actin integrity with chemical inhibitors, or using siRNAs increased the expression of immunomodulatory genes, such as TSG-6 and IDO, upon TNF-α stimulation. Similarly, transfection of TEAD siRNA also increased the immunomodulatory capacities in MSCs. RNA-seq analysis and inhibition assays demonstrated that the immunomodulatory capacities caused by YAP/TAZ-TEAD axis disruption were due to the NF-κB signaling pathway activation. Then, we also evaluated the in vivo anti-inflammatory efficacy of MSCs in a dextran sulfate sodium (DSS)-induced mice colitis model. The administration of human MSCs transfected with TEAD siRNA, which exhibited enhanced immunomodulatory properties in vitro, significantly ameliorated inflammatory bowel disease symptoms, such as body weight loss and acute colon inflammation, in the DSS-induced mice colitis model. Our findings underscore the mechanosignaling YAP/TAZ-TEAD axis as a regulator of MSCs immunomodulation. Targeting these signaling pathways could herald promising MSCs-based therapies for immune disorders.


Subject(s)
Colitis , Mesenchymal Stem Cells , YAP-Signaling Proteins , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Colitis/metabolism , Immunomodulation , Mechanotransduction, Cellular , RNA, Small Interfering/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , TEA Domain Transcription Factors/metabolism
4.
J Bone Miner Res ; 38(10): 1521-1540, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37551879

ABSTRACT

Mouse ligature-induced periodontitis (LIP) has been used to study bone loss in periodontitis. However, the role of osteocytes in LIP remains unclear. Furthermore, there is no consensus on the choice of alveolar bone parameters and time points to evaluate LIP. Here, we investigated the dynamics of changes in osteoclastogenesis and bone volume (BV) loss in LIP over 14 days. Time-course analysis revealed that osteoclast induction peaked on days 3 and 5, followed by the peak of BV loss on day 7. Notably, BV was restored by day 14. The bone formation phase after the bone resorption phase was suggested to be responsible for the recovery of bone loss. Electron microscopy identified bacteria in the osteocyte lacunar space beyond the periodontal ligament (PDL) tissue. We investigated how osteocytes affect bone resorption of LIP and found that mice lacking receptor activator of NF-κB ligand (RANKL), predominantly in osteocytes, protected against bone loss in LIP, whereas recombination activating 1 (RAG1)-deficient mice failed to resist it. These results indicate that T/B cells are dispensable for osteoclast induction in LIP and that RANKL from osteocytes and mature osteoblasts regulates bone resorption by LIP. Remarkably, mice lacking the myeloid differentiation primary response gene 88 (MYD88) did not show protection against LIP-induced bone loss. Instead, osteocytic cells expressed nucleotide-binding oligomerization domain containing 1 (NOD1), and primary osteocytes induced significantly higher Rankl than primary osteoblasts when stimulated with a NOD1 agonist. Taken together, LIP induced both bone resorption and bone formation in a stage-dependent manner, suggesting that the selection of time points is critical for quantifying bone loss in mouse LIP. Pathogenetically, the current study suggests that bacterial activation of osteocytes via NOD1 is involved in the mechanism of osteoclastogenesis in LIP. The NOD1-RANKL axis in osteocytes may be a therapeutic target for bone resorption in periodontitis. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).

5.
Stem Cell Rev Rep ; 19(6): 1812-1827, 2023 08.
Article in English | MEDLINE | ID: mdl-37166558

ABSTRACT

Three-dimensional clumps of mesenchymal stem cells (MSCs)/extracellular matrix (ECM) complexes (C-MSCs) can be implanted into tissue defects with no artificial scaffolds. In addition, the cellular properties and characteristics of the ECM in C-MSCs can be regulated in vitro. Most bone formation in the developmental and healing process is due to endochondral ossification, which occurs after bone collar formation surrounding cartilage derived from MSCs. Thus, to develop a rapid and reliable bone-regenerative cell therapy, the present study aimed to generate cartilaginous tissue covered with a mineralized bone collar-like structure from human C-MSCs by combining chondrogenic and osteogenic induction. Human bone marrow-derived MSCs were cultured in xeno-free/serum-free (XF) growth medium. Confluent cells that formed cellular sheets were detached from the culture plate using a micropipette tip. The floating cellular sheet contracted to round clumps of cells (C-MSCs). C-MSCs were maintained in XF-chondro-inductive medium (CIM) and XF-osteo-inductive medium (OIM). The biological and bone-regenerative properties of the generated cellular constructs were assessed in vitro and in vivo. C-MSCs cultured in CIM/OIM formed cartilaginous tissue covered with a mineralized matrix layer, whereas CIM treatment alone induced cartilage with no mineralization. Transplantation of the cartilaginous tissue covered with a mineralized matrix induced more rapid bone reconstruction via endochondral ossification in the severe combined immunodeficiency mouse calvarial defect model than that of cartilage generated using only CIM. These results highlight the potential of C-MSC culture in combination with CIM/OIM to generate cartilage covered with a bone collar-like structure, which can be applied for novel bone-regenerative cell therapy.


Subject(s)
Bone Regeneration , Osteogenesis , Mice , Animals , Humans , Bone and Bones , Cartilage , Extracellular Matrix , Disease Models, Animal
6.
Nat Commun ; 13(1): 6648, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333322

ABSTRACT

The impact of bone cell activation on bacterially-induced osteolysis remains elusive. Here, we show that matrix-embedded osteocytes stimulated with bacterial pathogen-associated molecular patterns (PAMPs) directly drive bone resorption through an MYD88-regulated signaling pathway. Mice lacking MYD88, primarily in osteocytes, protect against osteolysis caused by calvarial injections of bacterial PAMPs and resist alveolar bone resorption induced by oral Porphyromonas gingivalis (Pg) infection. In contrast, mice with targeted MYD88 restoration in osteocytes exhibit osteolysis with inflammatory cell infiltration. In vitro, bacterial PAMPs induce significantly higher expression of the cytokine RANKL in osteocytes than osteoblasts. Mechanistically, activation of the osteocyte MYD88 pathway up-regulates RANKL by increasing binding of the transcription factors CREB and STAT3 to Rankl enhancers and by suppressing K48-ubiquitination of CREB/CREB binding protein and STAT3. Systemic administration of an MYD88 inhibitor prevents jawbone loss in Pg-driven periodontitis. These findings reveal that osteocytes directly regulate inflammatory osteolysis in bone infection, suggesting that MYD88 and downstream RANKL regulators in osteocytes are therapeutic targets for osteolysis in periodontitis and osteomyelitis.


Subject(s)
Alveolar Bone Loss , Osteolysis , Osteomyelitis , Periodontitis , Mice , Animals , Osteocytes/metabolism , Osteolysis/chemically induced , Osteolysis/complications , Osteolysis/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , RANK Ligand/metabolism , Porphyromonas gingivalis/metabolism , Periodontitis/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Osteoclasts/metabolism
7.
JBMR Plus ; 4(6): e10352, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32537546

ABSTRACT

Cherubism (OMIM#118400) is a craniofacial disorder characterized by destructive jaw expansion. Gain-of-function mutations in SH3-domain binding protein 2 (SH3BP2) are responsible for this rare disorder. We have previously shown that homozygous knock-in (KI) mice (Sh3bp2 KI/KI ) recapitulate human cherubism by developing inflammatory lesions in the jaw. However, it remains unknown why heterozygous KI mice (Sh3bp2 KI/+ ) do not recapitulate the excessive jawbone destruction in human cherubism, even though all mutations are heterozygous in humans. We hypothesized that Sh3bp2 KI/+ mice need to be challenged for developing exacerbated jawbone destruction and that bacterial stimulation in the oral cavity may be involved in the mechanism. In this study, we applied a ligature-induced periodontitis model to Sh3bp2 KI/+ mice to induce inflammatory alveolar bone destruction. Ligature placement induced alveolar bone resorption with gingival inflammation. Quantification of alveolar bone volume revealed that Sh3bp2 KI/+ mice developed more severe bone loss (male: 43.0% ± 10.6%, female: 42.6% ± 10.4%) compared with Sh3bp2 +/+ mice (male: 25.8% ± 4.0%, female: 30.9% ± 6.5%). Measurement of bone loss by the cement-enamel junction-alveolar bone crest distance showed no difference between Sh3bp2 KI/+ and Sh3bp2 +/+ mice. The number of osteoclasts on the alveolar bone surface was higher in male Sh3bp2 KI/+ mice, but not in females, compared with Sh3bp2 +/+ mice. In contrast, inflammatory cytokine levels in gingiva were comparable between Sh3bp2 KI/+ and Sh3bp2 +/+ mice with ligatures. Genetic deletion of the spleen tyrosine kinase in myeloid cells and antibiotic treatment suppressed alveolar bone loss in Sh3bp2 KI/+ mice, suggesting that increased osteoclast differentiation and function mediated by SYK and accumulation of oral bacteria are responsible for the increased alveolar bone loss in Sh3bp2 KI/+ mice with ligature-induced periodontitis. High amounts of oral bacterial load caused by insufficient oral hygiene could be a trigger for the initiation of jawbone destruction in human cherubism. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.

8.
Bone Rep ; 12: 100258, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32258251

ABSTRACT

Even though the receptor activator of the nuclear factor-κB ligand (RANKL) and its receptor RANK have an exclusive role in osteoclastogenesis, the possibility of RANKL/RANK-independent osteoclastogenesis has been the subject of a long-standing debate in bone biology. In contrast, it has been reported that calvarial injection of TNF-ɑ elicits significant osteoclastogenesis in the absence of RANKL/RANK in NF-κB2- and RBP-J-deficient mice, suggesting that inflammatory challenges and secondary gene manipulation are the prerequisites for RANKL/RANK-deficient mice to develop osteoclasts in vivo. Here we report that, even in the absence of RANKL (Rankl -/- ), cherubism mice (Sh3bp2 KI/KI ) harboring the homozygous gain-of-function mutation in SH3-domain binding protein 2 (SH3BP2) develop tartrate-resistant acid phosphatase (TRAP)-positive multinucleated osteoclasts spontaneously. The Sh3bp2 KI/KI Rankl -/- mice exhibit an increase in tooth exposure and a decrease in bone volume/total volume compared to Sh3bp2 +/+ Rankl -/- mice. The multinucleated cells were stained positively for cathepsin K. Osteoclastic marker gene expression in bone and serum TRAP5b levels were elevated in Sh3bp2 KI/KI Rankl -/- mice. Elevation of the serum TNF-ɑ levels suggested that TNF-ɑ is a driver for the RANKL-independent osteoclast formation in Sh3bp2 KI/KI mice. Our results provide a novel mutant model that develops osteoclasts independent of RANKL and establish that the gain-of-function of SH3BP2 promotes osteoclastogenesis not only in the presence of RANKL but also in the absence of RANKL.

9.
Biochem Biophys Res Commun ; 525(4): 889-894, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32171527

ABSTRACT

A sophisticated and delicate balance between bone resorption by osteoclasts and bone formation by osteoblasts regulates bone metabolism. Optineurin (OPTN) is a gene involved in primary open-angle glaucoma and amyotrophic lateral sclerosis. Although its function has been widely studied in ophthalmology and neurology, recent reports have shown its possible involvement in bone metabolism through negative regulation of osteoclast differentiation. However, little is known about the role of OPTN in osteoblast function. Here, we demonstrated that OPTN controls not only osteoclast but also osteoblast differentiation. Different parameters involved in osteoblastogenesis and osteoclastogenesis were assessed in Optn-/- mice. The results showed that osteoblasts from Optn-/- mice had impaired alkaline phosphatase activity, defective mineralized nodules, and inability to support osteoclast differentiation. Moreover, OPTN could bind to signal transducer and activator of transcription 1 (STAT1) and regulate runt-related transcription factor 2 (RUNX2) nuclear localization by modulating STAT1 levels in osteoblasts. These data suggest that OPTN is involved in bone metabolism not only by regulating osteoclast function but also by regulating osteoblast function by mediating RUNX2 nuclear translocation via STAT1.


Subject(s)
Cell Cycle Proteins/metabolism , Membrane Transport Proteins/metabolism , Osteoblasts/cytology , Osteogenesis/physiology , STAT1 Transcription Factor/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Differentiation/physiology , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Membrane Transport Proteins/genetics , Mice, Inbred C57BL , Mice, Mutant Strains , Osteoclasts/cytology , Osteoclasts/metabolism
10.
J Bone Miner Res ; 35(2): 382-395, 2020 02.
Article in English | MEDLINE | ID: mdl-31613396

ABSTRACT

Periodontitis is a bacterially induced chronic inflammatory condition of the oral cavity where tooth-supporting tissues including alveolar bone are destructed. Previously, we have shown that the adaptor protein SH3-domain binding protein 2 (SH3BP2) plays a critical role in inflammatory response and osteoclastogenesis of myeloid lineage cells through spleen tyrosine kinase (SYK). In this study, we show that SH3BP2 is a novel regulator for alveolar bone resorption in periodontitis. Micro-CT analysis of SH3BP2-deficient (Sh3bp2 -/- ) mice challenged with ligature-induced periodontitis revealed that Sh3bp2 -/- mice develop decreased alveolar bone loss (male 14.9% ± 10.2%; female 19.0% ± 6.0%) compared with wild-type control mice (male 25.3% ± 5.8%; female 30.8% ± 5.8%). Lack of SH3BP2 did not change the inflammatory cytokine expression and osteoclast induction. Conditional knockout of SH3BP2 and SYK in myeloid lineage cells with LysM-Cre mice recapitulated the reduced bone loss without affecting both inflammatory cytokine expression and osteoclast induction, suggesting that the SH3BP2-SYK axis plays a key role in regulating alveolar bone loss by mechanisms that regulate the bone-resorbing function of osteoclasts rather than differentiation. Administration of a new SYK inhibitor GS-9973 before or after periodontitis induction reduced bone resorption without affecting inflammatory reaction in gingival tissues. In vitro, GS-9973 treatment of bone marrow-derived M-CSF-dependent macrophages suppressed tartrate-resistant acid phosphatase (TRAP)-positive osteoclast formation with decreased mineral resorption capacity even when GS-9973 was added after RANKL stimulation. Thus, the data suggest that SH3BP2-SYK is a novel signaling axis for regulating alveolar bone loss in periodontitis and that SYK can be a potential therapeutic target to suppress alveolar bone resorption in periodontal diseases. © 2019 American Society for Bone and Mineral Research. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Alveolar Bone Loss , Osteoclasts , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone and Bones/metabolism , Cell Differentiation , Female , Male , Mice , Mice, Inbred C57BL , Osteoclasts/metabolism , RANK Ligand/metabolism , Tartrate-Resistant Acid Phosphatase
11.
Prog Neurol Surg ; 32: 1-13, 2018.
Article in English | MEDLINE | ID: mdl-29990969

ABSTRACT

Recently, the clinical applications of photodynamic therapy (PDT) in the management of malignant brain tumors have attracted significant attention. Meta-analysis of the observational studies on this treatment in high-grade gliomas (Eljamel, 2010) included more than 1,000 patients and reported median survival in cases of newly diagnosed and recurrent glioblastoma multiforme (GBM) of 16.1 and 10.3 months, respectively. In some series, increase in the long-term survival rates was also observed. Few controlled trials demonstrated statistically significant impact of PDT on prolongation of survival in patients with GBM in comparison to conventional management. The main treatment-related adverse event is short-lasting excessive photosensitivity of the skin and retina after photosensitizer administration, but its negative consequences can be easily avoided with appropriate protective measures. Overall, PDT may be considered to be a safe and effective adjuvant therapeutic option for patients with newly diagnosed and recurrent malignant gliomas. Aggressive tumor resection seems to be an important prerequisite to maximize treatment efficacy.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Neoplasm Recurrence, Local/therapy , Outcome Assessment, Health Care , Photochemotherapy/methods , Photosensitivity Disorders/etiology , Photosensitizing Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Humans , Neoplasm Recurrence, Local/drug therapy , Photochemotherapy/adverse effects , Photosensitizing Agents/adverse effects
12.
Jpn Dent Sci Rev ; 54(2): 66-75, 2018 May.
Article in English | MEDLINE | ID: mdl-29755617

ABSTRACT

Periodontal disease is a bacterial biofilm-associated inflammatory disease that has been implicated in many systemic diseases. A new preventive method for periodontal disease needs to be developed in order to promote the health of the elderly in a super-aged society. The gingival epithelium plays an important role as a mechanical barrier against bacterial invasion and a part of the innate immune response to infectious inflammation in periodontal tissue. The disorganization of cell-cell interactions and subsequent inflammation contribute to the initiation of periodontal disease. These make us consider that regulation of host defensive functions, epithelial barrier and neutrophil activity, may become novel preventive methods for periodontal inflammation. Based on this concept, we have found that several agents regulate the barrier function of gingival epithelial cells and suppress the accumulation of neutrophils in the gingival epithelium. We herein introduce the actions of irsogladine maleate, azithromycin, amphotericin B, and Houttuynia cordata (dokudami in Japanese), which is commonly used in traditional medicine, on the epithelial barrier and neutrophil migration in gingival epithelial cells in vivo and in vitro, in order to provide support for the clinical application of these agents to the prevention of periodontal inflammation.

13.
J Bone Miner Res ; 33(8): 1513-1519, 2018 08.
Article in English | MEDLINE | ID: mdl-29669173

ABSTRACT

Cherubism is a craniofacial disorder characterized by maxillary and mandibular bone destruction. Gain-of-function mutations in the SH3-domain binding protein 2 (SH3BP2) are responsible for the excessive bone resorption caused by fibrous inflammatory lesions. A homozygous knock-in (KI) mouse model for cherubism (Sh3bp2KI/KI ) develops autoinflammation resulting in systemic bone destruction. Although administration of the TNF-α blocker etanercept to neonatal Sh3bp2KI/KI mice prevented the disease onset, this therapy was not effective for adult Sh3bp2KI/KI mice or human cherubism patients who already had lesions. Because genetic ablation of spleen tyrosine kinase (SYK) in myeloid cells rescues Sh3bp2KI/KI mice from inflammation, we examined whether SYK inhibitor administration can improve fully developed cherubism symptoms in adult Sh3bp2KI/KI mice. Entospletinib (GS-9973) was intraperitoneally injected into 10-week-old Sh3bp2KI/KI mice every day for 6 weeks. Treatment with GS-9973 improved facial swelling and histomorphometric analysis of lung and liver tissue showed that GS-9973 administration significantly reduced inflammatory infiltrates associated with decreased levels of serum TNF-α. Micro-computed tomography (µCT) analysis showed that GS-9973 treatment reduced bone erosion in mandibles, calvariae, and ankle and elbow joints of Sh3bp2KI/KI mice compared to Sh3bp2KI/KI mice treated with dimethyl sulfoxide (DMSO). Taken together, the results demonstrate that administration of the SYK inhibitor ameliorates an already established cherubism phenotype in mice, suggesting that pharmacological inhibition of SYK may be a treatment option for cherubism patients with active disease progression. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Bone and Bones/pathology , Cherubism/drug therapy , Indazoles/therapeutic use , Inflammation/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazines/therapeutic use , Syk Kinase/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone and Bones/drug effects , Cherubism/complications , Disease Models, Animal , Indazoles/administration & dosage , Indazoles/pharmacology , Inflammation/complications , Inflammation/pathology , Mice , Protein Kinase Inhibitors/pharmacology , Pyrazines/administration & dosage , Pyrazines/pharmacology , Syk Kinase/metabolism
14.
J Bone Miner Res ; 33(1): 167-181, 2018 01.
Article in English | MEDLINE | ID: mdl-28914985

ABSTRACT

Currently, it is believed that osteoclasts positive for tartrate-resistant acid phosphatase (TRAP+) are the exclusive bone-resorbing cells responsible for focal bone destruction in inflammatory arthritis. Recently, a mouse model of cherubism (Sh3bp2KI/KI ) with a homozygous gain-of-function mutation in the SH3-domain binding protein 2 (SH3BP2) was shown to develop auto-inflammatory joint destruction. Here, we demonstrate that Sh3bp2KI/KI mice also deficient in the FBJ osteosarcoma oncogene (c-Fos) still exhibit noticeable bone erosion at the distal tibia even in the absence of osteoclasts at 12 weeks old. Levels of serum collagen I C-terminal telopeptide (ICTP), a marker of bone resorption generated by matrix metalloproteinases (MMPs), were elevated, whereas levels of serum cross-linked C-telopeptide (CTX), another resorption marker produced by cathepsin K, were not increased. Collagenolytic MMP levels were increased in the inflamed joints of the Sh3bp2KI/KI mice deficient in c-Fos. Resorption pits contained a large number of F4/80+ macrophages and genetic depletion of macrophages rescued these erosive changes. Importantly, administration of NSC405020, an MMP14 inhibitor targeted to the hemopexin (PEX) domain, suppressed bone erosion in c-Fos-deficient Sh3bp2KI/KI mice. After activation of the NF-κB pathway, macrophage colony-stimulating factor (M-CSF)-dependent macrophages from c-Fos-deficient Sh3bp2KI/KI mice expressed increased amounts of MMP14 compared with wild-type macrophages. Interestingly, receptor activator of NF-κB ligand (RANKL)-deficient Sh3bp2KI/KI mice failed to show notable bone erosion, whereas c-Fos deletion did restore bone erosion to the RANKL-deficient Sh3bp2KI/KI mice, suggesting that osteolytic transformation of macrophages requires both loss-of-function of c-Fos and gain-of-function of SH3BP2 in this model. These data provide the first genetic evidence that cells other than osteoclasts can cause focal bone destruction in inflammatory bone disease and suggest that MMP14 is a key mediator conferring pathological bone-resorbing capacity on c-Fos-deficient Sh3bp2KI/KI macrophages. In summary, the paradigm that osteoclasts are the exclusive cells executing inflammatory bone destruction may need to be reevaluated based on our findings with c-Fos-deficient cherubism mice lacking osteoclasts. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Bone and Bones/pathology , Cherubism/pathology , Inflammation/pathology , Macrophages/pathology , Matrix Metalloproteinase 14/metabolism , Osteoclasts/metabolism , Proto-Oncogene Proteins c-fos/deficiency , Tartrate-Resistant Acid Phosphatase/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Joints/drug effects , Joints/pathology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Matrix Metalloproteinase 2/metabolism , Mice , NF-kappa B/metabolism , Osteoclasts/drug effects , Proto-Oncogene Proteins c-fos/metabolism , RANK Ligand/deficiency , RANK Ligand/metabolism , Signal Transduction/drug effects
15.
Cell Physiol Biochem ; 39(5): 1777-1786, 2016.
Article in English | MEDLINE | ID: mdl-27744428

ABSTRACT

BACKGROUND: An investigation of the mechanisms underlying the production of inflammatory cytokines through the stimulation of microorganisms on gingival epithelial cells may provide insights into the pathogenesis of the initiation of periodontitis. Lipid rafts, microdomains in the cell membrane, include a large number of receptors, and are centrally involved in signal transduction. We herein examined the involvement of lipid rafts in the expression of interleukin (IL-6) and IL-8 in gingival epithelial cells stimulated by periodontal pathogens. METHODS: OBA9, a human gingival cell line, was stimulated by Aggregatibacter actinomycetemcomitans or tumor necrosis factor (TNF)-α in the presence of methyl-ß-cyclodextrin (MßCD). RESULTS: A. actinomycetemcomitans or TNF-α increased IL-8 and IL-6 mRNA levels, and promoted the phosphorylation of ERK and p38 MAP kinase in OBA9. The pretreatment with MßCD abolished increases in IL-6 and IL-8 mRNA levels and the phosphorylation induced by A. actinomycetemcomitans, but did not suppress the response induced by TNF-α. The transfection of TLR4 inhibited A. actinomycetemcomitans-induced increases in IL-8 and IL-6 mRNA levels. Confocal microscopy revealed that MßCD inhibited the mobilization of TLR4 into lipid rafts. CONCLUSION: The mobilization of TLR4 into lipid rafts is involved in the expression of inflammatory cytokines and phosphorylation of MAP kinase in human gingival epithelial cells stimulated by A. actinomycetemcomitans.


Subject(s)
Aggregatibacter actinomycetemcomitans/growth & development , Epithelial Cells/immunology , Host-Pathogen Interactions , Membrane Microdomains/immunology , Toll-Like Receptor 4/genetics , p38 Mitogen-Activated Protein Kinases/immunology , Aggregatibacter actinomycetemcomitans/metabolism , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Gene Expression Regulation , Gingiva/immunology , Gingiva/microbiology , Gingiva/pathology , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-8/genetics , Interleukin-8/immunology , Membrane Microdomains/drug effects , Membrane Microdomains/microbiology , Membrane Microdomains/pathology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Phosphorylation , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/pharmacology , beta-Cyclodextrins/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics
16.
Cell Microbiol ; 18(12): 1723-1738, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27121139

ABSTRACT

Gingival junctional epithelial cell apoptosis caused by periodontopathic bacteria exacerbates periodontitis. This pathological apoptosis is involved in the activation of transforming growth factor ß (TGF-ß). However, the molecular mechanisms by which microbes induce the activation of TGF-ß remain unclear. We previously reported that Aggregatibacter actinomycetemcomitans (Aa) activated TGF-ß receptor (TGF-ßR)/smad2 signalling to induce epithelial cell apoptosis, even though Aa cannot bind to TGF-ßR. Additionally, outer membrane protein 29 kDa (Omp29), a member of the Aa Omps family, can induce actin rearrangements via focal adhesion kinase (FAK) signalling, which also plays a role in the activation of TGF-ß by cooperating with integrin. Accordingly, we hypothesized that Omp29-induced actin rearrangements via FAK activity would enhance the activation of TGF-ß, leading to gingival epithelial cell apoptosis in vitro. By using human gingival epithelial cell line OBA9, we found that Omp29 activated TGF-ßR/smad2 signalling and decreased active TGF-ß protein levels in the extracellular matrix (ECM) of cell culture, suggesting the transactivation of TGF-ßR. Inhibition of actin rearrangements by cytochalasin D or blebbistatin and knockdown of FAK or integrinß1 expression by siRNA transfection attenuated TGF-ßR/smad2 signalling activity and reduction of TGF-ß levels in the ECM caused by Omp29. Furthermore, Omp29 bound to fibronectin (Fn) to induce its aggregation on integrinß1, which is associated with TGF-ß signalling activity. All the chemical inhibitors and siRNAs tested blocked Omp29-induced OBA9 cells apoptosis. These results suggest that Omp29 binds to Fn in order to facilitate Fn/integrinß1/FAK signalling-dependent TGF-ß release from the ECM, thereby inducing gingival epithelial cell apoptosis via TGF-ßR/smad2 pathway.


Subject(s)
Aggregatibacter actinomycetemcomitans/genetics , Bacterial Outer Membrane Proteins/genetics , Epithelial Cells/microbiology , Fibronectins/genetics , Focal Adhesion Kinase 1/genetics , Integrin beta1/genetics , Transforming Growth Factor beta/genetics , Aggregatibacter actinomycetemcomitans/metabolism , Apoptosis/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/pharmacology , Cell Line, Transformed , Cytochalasin D/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibronectins/metabolism , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation , Gingiva/metabolism , Gingiva/microbiology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Host-Pathogen Interactions , Humans , Integrin beta1/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , Smad2 Protein/antagonists & inhibitors , Smad2 Protein/genetics , Smad2 Protein/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
17.
Arch Oral Biol ; 62: 64-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26655749

ABSTRACT

OBJECTIVE: The gingival epithelium plays an important role in protecting against the invasion of periodontal pathogens, and the permeability of gingival epithelial cells has been implicated in the initiation of periodontitis. Azithromycin (AZM) has been used in the treatment of chronic inflammatory airway diseases because it regulates cell-cell contact in airway epithelial cells. Therefore, AZM may also regulate barrier function in gingival epithelial cells. In the present study, we examined the effects of AZM on the permeability of human gingival epithelial cells (HGEC) under inflammatory conditions in vitro. MATERIALS AND METHODS: HGEC were stimulated by tumor necrosis factor-α (TNF-α) in the presence of AZM or p38 MAP kinase and ERK inhibitors. Permeability was assessed based on transepithelial electrical resistance (TER). The expression of E-cadherin, phosphorylated p38 MAP kinase, and ERK was analyzed by Western blotting. RESULTS: TNF-α decreased TER in HGEC, and AZM and the p38 MAP kinase and ERK inhibitors recovered this decrease. AZM inhibited the phosphorylation of ERK and p38 MAP kinase in TNF-α-stimulated HGEC. Furthermore, AZM recovered the decrease in E-cadherin expression in HGEC stimulated with TNF-α. CONCLUSIONS: These results suggested that AZM regulated gingival epithelial permeability through p38 MAP kinase and ERK signaling, and may contribute to suppress the inflammation in gingival tissue.


Subject(s)
Azithromycin/pharmacology , Epithelial Cells/drug effects , Gingiva/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Cadherins/metabolism , Cell Line , Cell Membrane Permeability/drug effects , Cells, Cultured , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Gingiva/cytology , Gingiva/metabolism , Gingivitis/metabolism , Humans , Interleukin-8/biosynthesis , Interleukin-8/metabolism , Phosphorylation , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Cytokine ; 75(1): 165-73, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25882870

ABSTRACT

Periodontitis is the most prevalent infectious disease caused by periodontopathic bacteria and is also a chronic inflammatory disease. Gingival crevicular fluid (GCF) is an inflammatory exudate that seeps into the gingival crevices or periodontal pockets around teeth with inflamed gingiva, and contains various materials including leukocytes and cytokines. Since gingival epithelial cells, which form a barrier against bacterial challenges, are affected by GCF, cytokines or other materials contained within GCF are engaged in the maintenance and disruption of the epithelial barrier. Accordingly, its compositional pattern has been employed as a reliable objective index of local inflammation. Transforming growth factor ß1 (TGF-ß1) levels in GCF were previously shown to be markedly higher in patients with periodontitis than in healthy subject. However, it currently remains unclear how TGF-ß1 affects gingival epithelial cell growth or apoptosis; therefore, elucidating the mechanism responsible may lead to a deeper understanding of pathogenic periodontitis. In the present study, the human gingival epithelial cell line, OBA9 cells were stimulated with recombinant TGF-ß1. Apoptosis-related protein levels were determined by Western blotting. Caspase-3/7 activity was measured with a Caspase-Glo assay kit. Surviving and apoptotic cells were detected using an MTS assay and TUNEL staining, respectively. TGF-ßRI siRNA and smad2 siRNA were transfected into cells using the lipofectamine RNAiMAX reagent. TGF-ß1 elevated caspase-3 activity and the number of TUNEL-positive apoptotic cells in OBA9 cells. Furthermore, while the levels of the pro-apoptotic proteins Bax, Bak, Bim, and Bad were increased in OBA9 cells stimulated with TGF-ß1, the TGF-ß1 treatment also decreased the levels of anti-apoptotic proteins such as Bcl-2 and Bcl-xL in a time-dependent manner. Additionally, TGF-ß1 up-regulated the protein levels of cleaved caspase-9. These results indicated that TGF-ß1-induced apoptosis was involved in a mitochondria-related intrinsic pathway. TGF-ß1 phosphorylated smad2 in OBA9 cells and this phosphorylation was clearly reduced by SB431542 (a TGF-ß type I receptor inhibitor). Consistent with this result, SB431542 or smad2 siRNA-induced reductions in smad2 protein expression levels attenuated TGF-ß1-induced apoptosis. On the other hand, the ligation of TGF-ß1 on its receptor also stimulated the phosphorylation of Erk and Akt, which are smad2-independent pathways. However, the inhibition of Erk/Akt signaling pathways by U0126, a MEK-Erk inhibitor and LY294002, a PI3Kinase-Akt inhibitor, augmented TGF-ß1-induced apoptosis in OBA9 cells. Taken together, the results of present study demonstrated that TGF-ß1 activated both the smad2 and Erk/Akt cascades via its receptor on gingival epithelial cells, even though these two pathways have opposite roles in cell death and survival, and the culmination of these signaling events induced mitochondria-dependent apoptosis in gingival epithelial cells. Based on the results of the present study, we herein proposed for the first time, that TGF-ß1 is a novel target cytokine for monitoring the progression of periodontal disease.


Subject(s)
Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gingiva/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta1/pharmacology , Acetylcysteine/metabolism , Apoptosis , Benzamides/chemistry , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Survival , Dioxoles/chemistry , Humans , Inflammation/metabolism , MAP Kinase Signaling System , Periodontitis/metabolism , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
19.
Int Immunopharmacol ; 15(2): 340-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23306101

ABSTRACT

Periodontitis is an infectious inflammatory disease. Our previous studies have revealed that irsogladine maleate (IM) regulates intercellular junctional function and chemokine secretion in gingival epithelium, resulting in the suppression of the onset of periodontal disease in a rat model. Therefore, it is plausible that IM is a promising preventive remedy for periodontal disease. In this study, to gain a better understanding of IM in gingival epithelial cells, we employed a DNA microarray analysis. More specifically, human gingival epithelial cells (HGEC) were exposed to Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) in the presence or absence of IM. Then, a human genome focus array was used. A. actinomycetemcomitans facilitated the expression of several inflammatory-related genes, including these for matrix metalloproteinase (MMP)-3, interleukin (IL)-6, and intercellular adhesion molecule-1 (ICAM-1) in HGEC, while these mRNA levels were attenuated by IM treatment. Importantly, consistent with mRNA levels, immunoblotting, immunofluorescence staining and ELISA analysis indicated that IM also abrogated the A. actinomycetemcomitans-induced increase in MMP-3, IL-6, and ICAM-1 at the protein level. In addition, inhibition of the ERK or p38 MAP kinase signaling cascade, previously reported to be disturbed by IM treatment in HGEC, clearly blocked A. actinomycetemcomitans-induced MMP-3, IL-6, or ICAM-1 protein expression. Moreover, animal study revealed that IM-pretreatment inhibited the A. actinomycetemcomitans-induced increase of ICAM-1 in gingival junctional epithelium. Taken together, these results suggested that IM can regulate inflammatory responses in HGEC by inhibiting the ERK or p38 MAP kinase signaling cascade, which may result in suppression of inflammation in gingival tissue, thereby contributing to the prevention of periodontitis.


Subject(s)
Actinobacillus Infections/immunology , Aggregatibacter actinomycetemcomitans/immunology , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Gingiva/pathology , Periodontitis/prevention & control , Triazines/pharmacology , Animals , Antigens, Bacterial/immunology , Cells, Cultured , Epithelial Cells/pathology , Humans , Oligonucleotide Array Sequence Analysis , Periodontitis/genetics , Periodontitis/microbiology , Rats , Signal Transduction/drug effects , Triazines/therapeutic use , p38 Mitogen-Activated Protein Kinases/metabolism
20.
J Mol Neurosci ; 39(1-2): 175-84, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19277492

ABSTRACT

The physiological function of interleukin-6 within the central nervous system (CNS) is complex; interleukin-6 exerts neurotrophic and neuroprotective effects and yet can also function as a mediator of inflammation, demyelination, and astrogliosis depending on the cellular context. However, the roles of interleukin-6 in astrocytes are poorly understood. In the present study, we investigated the effect of the pro-inflammatory cytokine interleukin-6 on the production of the inflammatory mediator prostaglandin E(2) in mouse astrocytes. Interleukin-6 stimulated prostaglandin E(2) production in a time-dependent fashion via a rapid and transient induction of cyclooxygenase-2 messenger RNA, followed by cyclooxygenase-2 protein synthesis. Interleukin-6 may act on the nervous system by interacting with its specific soluble interleukin-6 receptor and the signal transducer 130-kDa glycoprotein. Simultaneous treatment of astrocytes with interleukin-6 and soluble interleukin-6 receptor caused marked induction of prostaglandin E(2) synthesis, and this effect was suppressed by adding a neutralizing antibody against soluble interleukin-6 receptor. Furthermore, the mouse 130-kDa glycoprotein antibody suppressed prostaglandin E(2) formation induced by interleukin-6, as well as interleukin-6/soluble interleukin-6 receptor complexes, in a dose-dependent manner. These results indicate that interleukin-6/soluble interleukin-6 receptor complexes and the signal transducer 130-kDa glycoprotein play an important role in the regulation of cyclooxygenase-2 expression and subsequent prostaglandin E(2) formation in mouse astrocytes and that interleukin-6 is an important regulator of immune and inflammatory processes in the CNS.


Subject(s)
Astrocytes/metabolism , Dinoprostone/metabolism , Interleukin-6/metabolism , Animals , Astrocytes/cytology , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokine Receptor gp130/metabolism , Humans , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...