Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 68
1.
Exp Mol Pathol ; 116: 104485, 2020 10.
Article En | MEDLINE | ID: mdl-32574668

Septic arthritis is a condition of bone disorder caused predominantly by Staphylococcus aureus. Following the bacterial entry activated immune cells specially macrophages and dendritic cells release pro-inflammatory mediators such as IL-6, TNF-α, IL-1ß etc., which not only create an inflammatory microenvironment but also play crucial roles in the proliferation of different CD+ T cell subsets. Among them, Th17 and Tregs are of major concern in recent times because of their potential roles in regulating the ongoing inflammation in many diseases including experimental arthritis. But the downstream signalling mechanism of these cells in regulating the severity of inflammation in case of septic arthritis is not known yet. So, here we have established a murine model of S. aureus induced septic arthritis and kept the animal upto 15 days post-infection. To examine the signalling mechanism, Th17 and Treg cells were isolated from blood, spleen and synovial joints of control and infected mice and observed the expression of JNK, NFκB and RANKL in the lysate of isolated Th17 and Tregs. We have also estimated the levels of serum IL-21 and TGF-ß. NFκB, JNK and RANKL expression was found to be higher at 3 and 15 days post-infection along with serum IL-21 levels. On the other hand, maximum TGF-ß level was observed at 9 days post-infection along with increased Treg population. In conclusion it was hypothesized that bone resorption is related with downstream signalling pathways of Th17 cells, which stimulate osteoclast generation via NFκB/JNK-RANKL axis and helps in the persistence of the disease.


Arthritis, Infectious/immunology , Inflammation/immunology , Staphylococcal Infections/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/microbiology , Arthritis, Experimental/pathology , Arthritis, Infectious/genetics , Arthritis, Infectious/microbiology , Arthritis, Infectious/pathology , Gene Expression Regulation/genetics , Humans , Inflammation/genetics , Inflammation/microbiology , Inflammation/pathology , Interleukin-1beta/genetics , Joints/immunology , Joints/microbiology , Joints/pathology , MAP Kinase Kinase 4/genetics , Mice , Osteoclasts/immunology , Osteoclasts/microbiology , Osteoclasts/pathology , RANK Ligand/genetics , Signal Transduction/genetics , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity , T-Lymphocytes, Regulatory/microbiology , Th17 Cells/microbiology , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/genetics
2.
Sci Rep ; 10(1): 7823, 2020 05 08.
Article En | MEDLINE | ID: mdl-32385413

This study investigates the role of NLRP3 inflammasome and its main effector Caspase-1 in inflammation and alveolar bone resorption associated with periodontitis. Heat-killed Aggregatibacter actinomycetemcomitans (Aa) was injected 3x/week (4 weeks) into gingival tissues of wild-type (WT), Nlrp3-KO and Caspase1-KO mice. Bone resorption was measured by µCT and osteoclast number was determined by tartrate-resistant acid phosphatase (TRAP) staining. Inflammation was assessed histologically (H/E staining and immunofluorescence of CD45 and Ly6G). In vitro studies determined the influence of Nlrp3 and Caspase-1 in Rankl-induced osteoclast differentiation and activity and on LPS-induced expression of inflammation-associated genes. Bone resorption was significantly reduced in Casp1-KO but not in Nlrp3-KO mice. Casp1-KO mice had increased in osteoclast numbers, whereas the inflammatory infiltrate or on gene expression were similar to those of WT and Nlrp3-KO mice. Strikingly, osteoclasts differentiated from Nlrp3-deficient macrophages had increased resorbing activity in vitro. LPS-induced expression of Il-10, Il-12 and Tnf-α was significantly reduced in Nlrp3- and Casp1-deficient macrophages. As an inceptive study, these results suggest that Nlrp3 inflammasome does not play a significant role in inflammation and bone resorption in vivo and that Caspase-1 has a pro-resorptive role in experimental periodontal disease.


Alveolar Bone Loss/genetics , Caspase 1/genetics , Inflammation/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Periodontitis/genetics , Aggregatibacter actinomycetemcomitans , Alveolar Bone Loss/microbiology , Alveolar Bone Loss/pathology , Animals , Cell Differentiation/drug effects , Disease Models, Animal , Gingiva/growth & development , Gingiva/microbiology , Humans , Inflammation/microbiology , Inflammation/pathology , Interleukin-10/genetics , Interleukin-12/genetics , Mice , Mice, Knockout , Osteoclasts/microbiology , Osteoclasts/pathology , Periodontitis/microbiology , Periodontitis/pathology , RANK Ligand/genetics , Tumor Necrosis Factor-alpha/genetics
3.
Infect Immun ; 88(4)2020 03 23.
Article En | MEDLINE | ID: mdl-31932325

Osteoarticular disease is a frequent complication of human brucellosis. Vaccination remains a critical component of brucellosis control, but there are currently no vaccines for use in humans, and no in vitro models for assessing the safety of candidate vaccines in reference to the development of bone lesions currently exist. While the effect of Brucella infection on osteoblasts has been extensively evaluated, little is known about the consequences of osteoclast infection. Murine bone marrow-derived macrophages were derived into mature osteoclasts and infected with B. abortus 2308, the vaccine strain S19, and attenuated mutants S19vjbR and B. abortusΔvirB2 While B. abortus 2308 and S19 replicated inside mature osteoclasts, the attenuated mutants were progressively killed, behavior that mimics infection kinetics in macrophages. Interestingly, B. abortus 2308 impaired the growth of osteoclasts without reducing resorptive activity, while osteoclasts infected with B. abortus S19 and S19vjbR were significantly larger and exhibited enhanced resorption. None of the Brucella strains induced apoptosis or stimulated nitric oxide or lactose dehydrogenase production in mature osteoclasts. Finally, infection of macrophages or osteoclast precursors with B. abortus 2308 resulted in generation of smaller osteoclasts with decreased resorptive activity. Overall, Brucella exhibits similar growth characteristics in mature osteoclasts compared to the primary target cell, the macrophage, but is able to impair the maturation and alter the resorptive capacity of these cells. These results suggest that osteoclasts play an important role in osteoarticular brucellosis and could serve as a useful in vitro model for both analyzing host-pathogen interactions and assessing vaccine safety.


Brucella Vaccine/adverse effects , Brucella abortus/growth & development , Host-Pathogen Interactions , Osteoarthritis/physiopathology , Osteoclasts/immunology , Osteoclasts/microbiology , Animals , Bone Resorption , Brucella Vaccine/administration & dosage , Cell Proliferation , Cells, Cultured , Macrophages/immunology , Macrophages/microbiology , Mice , Microbial Viability , Osteoclasts/physiology
4.
J Periodontal Res ; 55(3): 410-425, 2020 Jun.
Article En | MEDLINE | ID: mdl-31944305

BACKGROUND AND OBJECTIVE: Excessive osteoclast activity is a major characteristic of pathogenic bone loss in inflammatory bone diseases including periodontitis. However, beyond the knowledge that osteoclasts are differentiated from the monocyte/macrophage lineage and share common ancestry with macrophages and DC, the nature and function of osteoclast precursors are not completely understood. Furthermore, little is known about how osteoclast precursors respond to bacterial infection in vivo. We have previously demonstrated in vitro that the periodontal pathogen Porphyromonas gingivalis (Pg) plays a biphasic role on the receptor activator of nuclear factor kappa B ligand (RANKL)-induced osteoclast differentiation. In this study, we investigated the in vivo effect of Pg infection on the regulation of osteoclast precursors, using a mouse calvarial infection model. METHODS AND RESULTS: C57BL/6 wild-type and the myeloid differentiation factor 88 knockout (MyD88-/- ) mice were infected with Pg by calvarial injection. Local and systemic bone loss, and the number and function of CD11b+ c-fms+ cells from bone marrow and spleen were analyzed. Our results show that Pg infection induces localized inflammatory infiltration and osteoclastogenesis, as well as increased number and osteoclastogenic potential of CD11b+ c-fms+ osteoclast precursors in the bone marrow and periphery. We also show that CD11b+ c-fms+ RANK+ and CD11b+ c-fms+ RANK- are precursors with similar osteoclastogenic and pro-inflammatory potentials. In addition, CD11b+ c-fms+ cells exhibit an antigen-specific T-cell immune-suppressive activity, which are increased with Pg infection. Moreover, we demonstrate that MyD88 is involved in the regulation of osteoclast precursors upon Pg infection. CONCLUSIONS: In this study, we demonstrate an enhanced dual function of osteoclast precursors following calvarial Pg infection. Based on our findings, we propose the following model: Pg infection increases a pool of precursor cells that can be shunted toward osteoclast formation at the infection/inflammation sites, while at the same time dampening host immune responses, which is beneficial for the persistence of infection and maintenance of the characteristic chronic nature of periodontitis. Understanding the nature, function, and regulation of osteoclast precursors will be helpful for identifying therapeutic interventions to aid in the control and prevention of inflammatory bone loss diseases including periodontitis.


Bacteroidaceae Infections/pathology , Osteoclasts/cytology , Skull/microbiology , Animals , Cell Differentiation , Mice , Mice, Inbred C57BL , Osteoclasts/microbiology , Porphyromonas gingivalis , RANK Ligand
5.
Inflammation ; 43(1): 220-230, 2020 Feb.
Article En | MEDLINE | ID: mdl-31720989

Periodontitis is an inflammation characterized by alveolar bone resorption caused by imbalance in bone homeostasis. It is known that autophagy is related to inflammation and bone metabolism. However, whether autophagy inhibitors could be used for periodontitis in animal models remains unknown. We investigated the role of two classical autophagy inhibitors, 3-methyladenine (3-MA) and chloroquine (CQ), on the development of rat experimental periodontitis in terms of the bone loss (micro-CT), the number of inflammatory cells (hematoxylin and eosin staining), and the osteoclastic activity (tartrate-resistant acid phosphatase staining). Expression of autophagy-related genes and nuclear factor kappa B p65 (NF-κB p65) were assessed by immunohistochemistry. Expression of Beclin-1 and microtubule-associated proteins 1A/1B light chain 3 (LC3) were analyzed by Western blot. To further observe the effect of autophagy inhibitors on osteoclasts (OCs) in vitro, bone marrow-derived mononuclear macrophages were used. Together, these findings indicated that topical administration of 3-MA or CQ reduced the infiltration of inflammatory cells and alveolar bone resorption in experimental periodontitis. Furthermore, 3-MA and CQ may attenuate activation of OCs by autophagy. Therefore, 3MA and CQ may have prophylactic and therapeutic potential for inflammation and alveolar bone resorption in periodontitis in the future.


Adenine/analogs & derivatives , Alveolar Bone Loss/prevention & control , Alveolar Process/drug effects , Anti-Inflammatory Agents/pharmacology , Autophagy/drug effects , Chloroquine/pharmacology , Osteoclasts/drug effects , Periodontitis/prevention & control , Adenine/pharmacology , Alveolar Bone Loss/metabolism , Alveolar Bone Loss/microbiology , Alveolar Bone Loss/pathology , Alveolar Process/metabolism , Alveolar Process/microbiology , Alveolar Process/pathology , Animals , Autophagy-Related Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Male , Osteoclasts/metabolism , Osteoclasts/microbiology , Osteoclasts/pathology , Osteogenesis/drug effects , Periodontitis/metabolism , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis , Rats, Sprague-Dawley , Transcription Factor RelA/metabolism
6.
Curr Osteoporos Rep ; 17(6): 395-404, 2019 12.
Article En | MEDLINE | ID: mdl-31721069

PURPOSE OF REVIEW: Staphylococcus aureus is the primary pathogen responsible for osteomyelitis, which remains a major healthcare burden. To understand its dominance, here we review the unique pathogenic mechanisms utilized by S. aureus that enable it to cause incurable osteomyelitis. RECENT FINDINGS: Using an arsenal of toxins and virulence proteins, S. aureus kills and usurps immune cells during infection, to produce non-neutralizing pathogenic antibodies that thwart adaptive immunity. S. aureus also has specific mechanisms for distinct biofilm formation on implants, necrotic bone tissue, bone marrow, and within the osteocyte lacuno-canicular networks (OLCN) of live bone. In vitro studies have also demonstrated potential for intracellular colonization of osteocytes, osteoblasts, and osteoclasts. S. aureus has evolved a multitude of virulence mechanisms to achieve life-long infection of the bone, most notably colonization of OLCN. Targeting S. aureus proteins involved in these pathways could provide new targets for antibiotics and immunotherapies.


Adaptive Immunity/immunology , Bone and Bones/immunology , Immune Evasion , Osteomyelitis/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/pathogenicity , Abscess/immunology , B-Lymphocytes/immunology , Biofilms , Bone and Bones/microbiology , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Osteoblasts/microbiology , Osteoclasts/microbiology , Osteocytes/microbiology , Osteomyelitis/microbiology , Staphylococcal Infections/microbiology , Staphylococcal Protein A/immunology , Staphylococcus aureus/immunology
7.
mBio ; 10(5)2019 10 15.
Article En | MEDLINE | ID: mdl-31615966

Osteomyelitis (OM), or inflammation of bone tissue, occurs most frequently as a result of bacterial infection and severely perturbs bone structure. OM is predominantly caused by Staphylococcus aureus, and even with proper treatment, OM has a high rate of recurrence and chronicity. While S. aureus has been shown to infect osteoblasts, it remains unclear whether osteoclasts (OCs) are also a target of intracellular infection. Here, we demonstrate the ability of S. aureus to intracellularly infect and divide within OCs. OCs were differentiated from bone marrow macrophages (BMMs) by exposure to receptor activator of nuclear factor kappa-B ligand (RANKL). By utilizing an intracellular survival assay and flow cytometry, we found that at 18 h postinfection the intracellular burden of S. aureus increased dramatically in cells with at least 2 days of RANKL exposure, while the bacterial burden decreased in BMMs. To further explore the signals downstream of RANKL, we manipulated factors controlling OC differentiation, NFATc1 and alternative NF-κB, and found that intracellular bacterial growth correlates with NFATc1 levels in RANKL-treated cells. Confocal and time-lapse microscopy in mature OCs showed a range of intracellular infection that correlated inversely with S. aureus-phagolysosome colocalization. The propensity of OCs to become infected, paired with their diminished bactericidal capacity compared to BMMs, could promote OM progression by allowing S. aureus to evade initial immune regulation and proliferate at the periphery of lesions where OCs are most abundant.IMPORTANCE The inflammation of bone tissue is called osteomyelitis, and most cases are caused by an infection with the bacterium Staphylococcus aureus To date, the bone-building cells, osteoblasts, have been implicated in the progression of these infections, but not much is known about how the bone-resorbing cells, osteoclasts, participate. In this study, we show that S. aureus can infect osteoclasts and proliferate inside these cells, whereas bone-residing macrophages, immune cells related to osteoclasts, destroy the bacteria. These findings elucidate a unique role for osteoclasts to harbor bacteria during infection, providing a possible mechanism by which bacteria could evade destruction by the immune system.


Osteoclasts/microbiology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Differentiation , Cells, Cultured , Female , Macrophages/metabolism , Male , Mice , Osteoblasts/microbiology , Osteomyelitis/metabolism , Osteomyelitis/microbiology , Phagosomes/metabolism , RANK Ligand/metabolism , Staphylococcus aureus/drug effects
8.
Oral Dis ; 25(7): 1769-1779, 2019 Oct.
Article En | MEDLINE | ID: mdl-31365165

OBJECTIVES: This study aimed to investigate the role of JAK2-STAT3 (Janus kinase 2/signal transducer and activator of transcription 3) in periapical disease caused by Enterococcus faecalis, as well as the correlation between lipoteichoic acid (LTA) in E. faecalis and the activity of the JAK2-STAT3 signaling pathway and osteoclast formation. MATERIALS AND METHODS: A rat model of periapical periodontitis induced by E. faecalis was established. Periapical bone resorption was confirmed by HE staining. The expression of JAK2, p-JAK2, STAT3, and p-STAT3 was assessed with immunohistochemical staining. Osteoclasts were observed through enzyme histochemical staining. LTA acted on mouse osteoclast precursor cells (RAW264.7 cells); a JAK2 inhibitor (AG490) was used to inhibit the JAK2-STAT3 pathway in RAW264.7 cells. The expression of proteins in the JAK2-STAT3 pathway and TRAP (tartrate resistant acid phosphatase) in RAW264.7 cells was also detected. RESULTS: Rat periapical periodontitis was successfully established and bone resorption peaked at day 21. The expression of critical components in the JAK2-STAT3 pathway increased with the progression of inflammation. LTA promoted the differentiation of RAW264.7 cells into osteoclasts. NFATc1 was highly expressed and was inhibited by AG490. CONCLUSIONS: JAK2-STAT3 signaling pathway plays an important role in the process of periapical bone resorption and osteoclastogenesis.


Bone Resorption , Enterococcus faecalis/physiology , Janus Kinase 2/metabolism , Osteoclasts/physiology , Osteogenesis , Periapical Periodontitis/physiopathology , STAT3 Transcription Factor/metabolism , Animals , Gene Expression Regulation , Janus Kinase 2/genetics , Mice , Osteoclasts/microbiology , Periapical Periodontitis/etiology , Rats , STAT3 Transcription Factor/genetics , Signal Transduction
9.
Bone ; 127: 315-323, 2019 10.
Article En | MEDLINE | ID: mdl-31233933

Bone is a highly adaptive tissue with regenerative properties that is subject to numerous diseases. Infection is one of the causes of altered bone homeostasis. Bone infection happens subsequently to bone surgery or to systemic spreading of microorganisms. In addition to osteoblasts, osteoclasts (OCs) also constitute cell targets for pathogens. OCs are multinucleated cells that have the exclusive ability to resorb bone mineral tissue. However, the OC is much more than a bone eater. Beyond its role in the control of bone turnover, the OC is an immune cell that produces and senses inflammatory cytokines, ingests microorganisms and presents antigens. Today, increasing evidence shows that several pathogens use OC as a host cell to grow, generating debilitating bone defects. In this review, we exhaustively inventory the bacteria and viruses that infect OC and report the present knowledge in this topic. We point out that most of the microorganisms enhance the bone resorption activity of OC. We notice that pathogen interactions with the OC require further investigation, in particular to validate the OC as a host cell in vivo and to identify the cellular mechanisms involved in altered bone resorption. Thus, we conclude that the OC is a new cell target for pathogens; this new research area paves the way for new therapeutic strategies in the infections causing bone defects.


Bacteria/metabolism , Osteoclasts/microbiology , Osteoclasts/virology , Animals , Bacterial Infections/microbiology , Bacterial Infections/pathology , Endocytosis , Humans , Osteoclasts/pathology , Virus Diseases/pathology
10.
FASEB J ; 33(9): 10515-10527, 2019 09.
Article En | MEDLINE | ID: mdl-31251083

Histone Lys-specific demethylases (KDMs) play a key role in many biological processes through epigenetic mechanisms. However, the role of KDMs in inflammatory responses to oral bacterial infection is poorly understood. Here, we show a novel regulatory role of KDM3C in inflammatory responses to oral bacterial infection. KDM3C expression is transiently suppressed in human and mouse macrophages exposed to LPS from Porphyromonas gingivalis (Pg LPS). Loss of KDM3C in both human and mouse macrophages led to notable induction of proinflammatory cytokines in response to Pg LPS stimulation. Also, KDM3C depletion led to strong induction of p65 phosphorylation and accelerated nuclear translocation in cells exposed to Pg LPS. Kdm3C knockout (KO) in mice led to increased alveolar bone destruction upon induction of experimental periodontitis or pulp exposure compared with those of the wild-type (WT) littermates. The Kdm3C KO mice also revealed an increased number of osteoclasts juxtaposed to the bony lesions. We also confirmed enhanced osteoclastogenesis by bone marrow-derived macrophages isolated from the Kdm3C KO compared with the WT controls. These findings suggest an anti-inflammatory function of KDM3C in regulating the inflammatory responses against oral bacterial infection through suppression of NF-κB signaling and osteoclastogenesis.-Lee, J. Y., Mehrazarin, S., Alshaikh, A., Kim, S., Chen, W., Lux, R., Gwack, Y., Kim, R. H., Kang, M. K. Histone Lys demethylase KDM3C demonstrates anti-inflammatory effects by suppressing NF-κB signaling and osteoclastogenesis.


Inflammation/prevention & control , Jumonji Domain-Containing Histone Demethylases/physiology , Mouth Diseases/prevention & control , NF-kappa B/antagonists & inhibitors , Osteogenesis , Porphyromonas gingivalis/pathogenicity , Animals , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/microbiology , Cell Differentiation , Cytokines , Histones , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/toxicity , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Mouth Diseases/etiology , Mouth Diseases/metabolism , Mouth Diseases/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoclasts/metabolism , Osteoclasts/microbiology , Osteoclasts/pathology , Phosphorylation , Signal Transduction
11.
PLoS Pathog ; 15(4): e1007744, 2019 04.
Article En | MEDLINE | ID: mdl-30978245

Staphylococcus aureus is able to infect virtually all organ systems and is a frequently isolated etiologic agent of osteomyelitis, a common and debilitating invasive infection of bone. Treatment of osteomyelitis requires invasive surgical procedures and prolonged antibiotic therapy, yet is frequently unsuccessful due to extensive pathogen-induced bone damage that can limit antibiotic penetration and immune cell influx to the infectious focus. We previously established that S. aureus triggers profound alterations in bone remodeling in a murine model of osteomyelitis, in part through the production of osteolytic toxins. However, staphylococcal strains lacking osteolytic toxins still incite significant bone destruction, suggesting that host immune responses are also major drivers of pathologic bone remodeling during osteomyelitis. The objective of this study was to identify host immune pathways that contribute to antibacterial immunity during S. aureus osteomyelitis, and to define how these immune responses alter bone homeostasis and contribute to bone destruction. We specifically focused on the interleukin-1 receptor (IL-1R) and downstream adapter protein MyD88 given the prominent role of this signaling pathway in both antibacterial immunity and osteo-immunologic crosstalk. We discovered that while IL-1R signaling is necessary for local control of bacterial replication during osteomyelitis, it also contributes to bone loss during infection. Mechanistically, we demonstrate that S. aureus enhances osteoclastogenesis of myeloid precursors in vitro, and increases the abundance of osteoclasts residing on bone surfaces in vivo. This enhanced osteoclast abundance translates to trabecular bone loss, and is dependent on intact IL-1R signaling. Collectively, these data define IL-1R signaling as a critical component of the host response to S. aureus osteomyelitis, but also demonstrate that IL-1R-dependent immune responses trigger collateral bone damage through activation of osteoclast-mediated bone resorption.


Bone Resorption/immunology , Myeloid Differentiation Factor 88/physiology , Osteoclasts/immunology , Osteomyelitis/immunology , Receptors, Interleukin-1 Type I/physiology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Bone Resorption/metabolism , Bone Resorption/microbiology , Cell Differentiation , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/metabolism , Osteoclasts/microbiology , Osteomyelitis/metabolism , Osteomyelitis/microbiology , Signal Transduction , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology
12.
Molecules ; 23(7)2018 06 23.
Article En | MEDLINE | ID: mdl-29937485

Recent studies suggest that the commensal microbiota affects not only host energy metabolism and development of immunity but also bone remodeling by positive regulation of osteoclast activity. However, the mechanism of regulation of bone cells by the commensal microbiota has not been elucidated. In this study, 8-week-old specific pathogen-free (SPF) and germ-free (GF) mice were compared in terms of alveolar bones and primary osteoblasts isolated from calvarias. Micro-CT analysis showed that SPF mice had larger body size associated with lower bone mineral density and bone volume fraction in alveolar bones compared with GF mice. Greater numbers of osteoclasts in alveolar bone and higher serum levels of tartrate-resistant acid phosphatase 5b were observed in SPF mice. Tissue extracts from SPF alveolar bone showed higher levels of cathepsin K, indicating higher osteoclast activity. SPF alveolar extracts also showed elevated levels of γ-carboxylated glutamic acid⁻osteocalcin as a marker of mature osteoblasts compared with GF mice. Polymerase chain reaction (PCR) array analysis of RNA directly isolated from alveolar bone showed that in SPF mice, expression of mRNA of osteocalcin, which also acts as an inhibitor of bone mineralization, was strongly enhanced compared with GF mice. Cultured calvarial osteoblasts from SPF mice showed reduced mineralization but significantly enhanced expression of mRNAs of osteocalcin, alkaline phosphatase, insulin-like growth factor-I/II, and decreased ratio of osteoprotegerin/receptor activator of nuclear factor-kappa B ligand compared with GF mice. Furthermore, PCR array analyses of transcription factors in cultured calvarial osteoblasts showed strongly upregulated expression of Forkhead box g1. In contrast, Gata-binding protein 3 was strongly downregulated in SPF osteoblasts. These results suggest that the commensal microbiota prevents excessive mineralization possibly by stimulating osteocalcin expression in osteoblasts, and enhances both osteoblast and osteoclast activity by regulating specific transcription factors.


Bone Remodeling/genetics , Gastrointestinal Microbiome/physiology , Germ-Free Life , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , Symbiosis/physiology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Biomarkers/metabolism , Cathepsin K/genetics , Cathepsin K/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/microbiology , Osteocalcin/genetics , Osteocalcin/metabolism , Osteoclasts/cytology , Osteoclasts/microbiology , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism
13.
Exp Cell Res ; 362(1): 152-158, 2018 01 01.
Article En | MEDLINE | ID: mdl-29129564

Persistent apical periodontitis (PAP) is characterized by refractory inflammation and progressive bone destruction. Enterococcus faecalis infection is considered an important etiological factor for the development of PAP, although the exact mechanisms remain unknown. This study aimed at investigating the role of E. faecalis in cell proliferation, inflammatory reactions and osteoclast differentiation of macrophages using an in vitro infection model of osteoclast precursor RAW264.7 cells. A cell viability assay of cultured RAW264.7 cells exposed to live E. faecalis at a multiplicity of infection of 100 for 2h, indicated that the infection exhibited no cytotoxic effect. Transmission electron microscopy images revealed no apoptotic changes but a rise of metabolic activity and phagocytic features in the infected RAW264.7 cells. Confocal laser scanning microscopic and flow cytometric analysis indicated that the phagocytosis of RAW264.7 cells was activated by E. faecalis infection. Furthermore, quantitative real-time PCR assays demonstrated that the expression of inflammatory cytokines was remarkably elevated in infected RAW264.7 cells. Differentiation of infected RAW264.7 cells into osteoclasts was remarkably attenuated, and expression of osteoclast marker genes as well as fusogenic genes significantly dropped. In summary, E. faecalis appears to attenuate osteoclastic differentiation of RAW264.7 precursor cells, rather stimulates them to function as macrophages.


Enterococcus faecalis/immunology , Macrophages/microbiology , Osteoclasts/microbiology , Osteogenesis/immunology , Animals , Apoptosis/immunology , Biomarkers/metabolism , Cell Differentiation/immunology , Cell Line , Cytokines/metabolism , Inflammation/metabolism , Inflammation/microbiology , Macrophages/metabolism , Mice , Osteoclasts/metabolism , Phagocytosis/immunology , RAW 264.7 Cells
14.
Med Sci Monit ; 23: 4579-4590, 2017 Sep 24.
Article En | MEDLINE | ID: mdl-28942456

BACKGROUND Osteomyelitis is one of the refractory diseases encountered in orthopedics, while Staphylococcus aureus (S. aureus) is the most common causative organism in osteomyelitis. However, the precise mechanisms underlying the bone loss caused by S. aureus infection have not been well defined. Here, we investigated the effect of S. aureus on osteoclast differentiation and the probable molecular mechanism. MATERIAL AND METHODS RAW 264.7 cells were treated for 5 days with live S. aureus, inactivated S. aureus, and S. aureus filtrate. Then, the formation of osteoclast-like cells and resorption pits was observed, and the expression of osteoclast-specific genes (TRAP, MMP-9, cathepsin K, CTR and Atp6v0d2) was detected by real-time PCR. Moreover, key proteins in the signaling pathway associated with osteoclast differentiation were detected with Western blot. RESULTS The data showed that live S. aureus, inactivated S. aureus, and S. aureus filtrate induced osteoclast formation, promoted bone resorption, and increased the expression of osteoclast-specific genes in a dose-dependent manner in the absence RANKL. In addition, we found that the S. aureus-induced osteoclastogenesis was related to the degradation of IκB-a, phosphorylation of NF-κB p65, and increased expression of NFATc1. Thus, we used JSH-23 to inhibit NF-κB transcriptional activity. The effect of the S. aureus-induced osteoclastogenesis and the expression of osteoclast-specific genes and NFATc1 were inhibited, which indicated that the NF-κB signaling pathway plays a role in S. aureus-induced osteoclastogenesis. CONCLUSIONS This study demonstrated that S. aureus induces osteoclastogenesis through its cell wall compound and secretion of small soluble molecules, and the NF-κB signaling pathway plays a role in this process.


NF-kappa B/physiology , Osteogenesis/drug effects , Staphylococcus aureus/pathogenicity , Animals , Bone Resorption/metabolism , Bone Resorption/microbiology , Cell Differentiation/physiology , Gene Expression Regulation/genetics , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , NF-kappa B p52 Subunit/physiology , NFATC Transcription Factors , Osteoclasts/metabolism , Osteoclasts/microbiology , Osteogenesis/immunology , Osteogenesis/physiology , Osteomyelitis/microbiology , RAW 264.7 Cells , Signal Transduction/drug effects , Staphylococcus aureus/metabolism , Transcription Factor RelA/metabolism
15.
J Cell Physiol ; 232(9): 2396-2406, 2017 Sep.
Article En | MEDLINE | ID: mdl-28185243

Bone infection is a common and serious complication in the orthopedics field, which often leads to excessive bone destruction and non-union. Osteoclast is the only type of cells which have the function of bone resorption. Its over activation is closely related to excessive bone loss. Staphylococcus aureus (S. aureus) is a major pathogen causing bone infection, which can produce a large number of strong pathogenic substances staphylococcal protein A (SPA). However, few studies were reported about the effects of SPA on osteoclastogenesis. In our study, we observed that S. aureus activated osteoclasts and promoted bone loss in bone infection specimens. Then, we investigated the effects of SPA on RANKL-induced osteoclastogenesis in vitro, the results revealed that SPA promoted osteoclastic differentiation and fusion, and enhanced osteoclastic bone resorption. In addition, we also showed that SPA upregulated the expression of NFATc1 and c-FOS through the activation of MAPK signaling to promote osteoclastogenesis. Our findings might help us better understand the pathogenic role of S. aureus in bone infection and develop new therapeutic strategies for infectious bone diseases.


Bone Remodeling , Bone and Bones/enzymology , Mitogen-Activated Protein Kinases/metabolism , Osteoclasts/enzymology , Osteomyelitis/enzymology , Staphylococcal Infections/enzymology , Staphylococcal Protein A/metabolism , Staphylococcus aureus/metabolism , Animals , Apoptosis , Bone Resorption/enzymology , Bone Resorption/microbiology , Bone Resorption/pathology , Bone and Bones/microbiology , Bone and Bones/pathology , Case-Control Studies , Cell Differentiation , Enzyme Activation , Host-Pathogen Interactions , Humans , Mice , NFATC Transcription Factors/metabolism , Osteoclasts/microbiology , Osteoclasts/pathology , Osteomyelitis/microbiology , Osteomyelitis/pathology , Proto-Oncogene Proteins c-fos/metabolism , RANK Ligand/metabolism , RAW 264.7 Cells , Signal Transduction , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity
16.
Infect Immun ; 85(1)2017 Jan.
Article En | MEDLINE | ID: mdl-27795356

Aggregatibacter actinomycetemcomitans is associated with aggressive periodontal disease, which is characterized by inflammation-driven alveolar bone loss. A. actinomycetemcomitans activates the p38 mitogen-activated protein kinase (MAPK) and MAPK-activated protein kinase 2 (MK2) stress pathways in macrophages that are involved in host responses. During the inflammatory process in periodontal disease, chemokines are upregulated to promote recruitment of inflammatory cells. The objective of this study was to determine the role of MK2 signaling in chemokine regulation during A. actinomycetemcomitans pathogenesis. Utilizing a murine calvarial model, Mk2+/+ and Mk2-/- mice were treated with live A. actinomycetemcomitans bacteria at the midsagittal suture. MK2 positively regulated the following macrophage RNA: Emr1 (F4/80), Itgam (CD11b), Csf1r (M-CSF Receptor), Itgal (CD11a), Tnf, and Nos2 Additionally, RNA analysis revealed that MK2 signaling regulated chemokines CCL3 and CCL4 in murine calvarial tissue. Utilizing the chimeric murine air pouch model, MK2 signaling differentially regulated CCL3 and CCL4 in the hematopoietic and nonhematopoietic compartments. Bone resorption pits in calvaria, observed by micro-computed tomography, and osteoclast formation were decreased in Mk2-/- mice compared to Mk2+/+ mice after A. actinomycetemcomitans treatment. In conclusion, these data suggest that MK2 in macrophages contributes to regulation of chemokine signaling during A. actinomycetemcomitans-induced inflammation and bone loss.


Aggregatibacter actinomycetemcomitans/pathogenicity , Alveolar Bone Loss/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Macrophages/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Alveolar Bone Loss/microbiology , Alveolar Bone Loss/physiopathology , Animals , Cells, Cultured , Chemokines/metabolism , Inflammation/metabolism , Inflammation/microbiology , Inflammation/physiopathology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Osteoclasts/metabolism , Osteoclasts/microbiology , Pasteurellaceae Infections/metabolism , Pasteurellaceae Infections/microbiology , RNA/metabolism
17.
FASEB J ; 30(12): 4033-4041, 2016 12.
Article En | MEDLINE | ID: mdl-27535487

Alveolar bone loss is a result of an aggressive form of periodontal disease (PD) associated with Aggregatibacter actinomycetemcomitans (Aa) infection. PD is often observed with other systemic inflammatory conditions, including arthritis. Melanocortin peptides activate specific receptors to exert antiarthritic properties, avoiding excessing inflammation and modulating macrophage function. Recent work has indicated that melanocortin can control osteoclast development and function, but whether such protection takes place in infection-induced alveolar bone loss has not been investigated. The purpose of this study was to evaluate the role of melanocortin in Aa-induced PD. Mice were orally infected with Aa and treated with the melanocortin analog DTrp8-γMSH or vehicle daily for 30 d. Then, periodontal tissue was collected and analyzed. Aa-infected mice treated with DTrp8-γMSH presented decreased alveolar bone loss and a lower degree of neutrophil infiltration in the periodontium than vehicle-treated animals; these actions were associated with reduced periodontal levels of TNF-α, IFN-γ, and IL-17A. In vitro experiments with cells differentiated into osteoclasts showed that osteoclast formation and resorptive activity were attenuated after treatment with DTrp8-γMSH. Thus, melanocortin agonism could represent an innovative way to tame overexuberant inflammation and, at the same time, preserve bone physiology, as seen after Aa infection.-Madeira, M. F. M., Queiroz-Junior, C. M., Montero-Melendez, T., Werneck, S. M. C., Corrêa, J. D., Soriani, F. M., Garlet, G. P., Souza, D. G., Teixeira, M. M., Silva, T. A., Perretti, M. Melanocortin agonism as a viable strategy to control alveolar bone loss induced by oral infection.


Alveolar Bone Loss/prevention & control , Melanocortins/agonists , Osteoclasts/microbiology , Pasteurellaceae Infections/prevention & control , Periodontal Diseases/metabolism , Aggregatibacter actinomycetemcomitans , Alveolar Bone Loss/etiology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Macrophages/immunology , Mice, Inbred C57BL , Periodontitis/drug therapy , Periodontitis/metabolism
18.
PLoS One ; 11(6): e0156708, 2016.
Article En | MEDLINE | ID: mdl-27311019

Severe Staphylococcus aureus (S. aureus) infections pose an immense threat to population health and constitute a great burden for the health care worldwide. Inter alia, S. aureus septic arthritis is a disease with high mortality and morbidity caused by destruction of the infected joints and systemic bone loss, osteoporosis. Toll-Like receptors (TLRs) are innate immune cell receptors recognizing a variety of microbial molecules and structures. S. aureus recognition via TLR2 initiates a signaling cascade resulting in production of various cytokines, but the mechanisms by which S. aureus causes rapid and excessive bone loss are still unclear. We, therefore, investigated how S. aureus regulates periosteal/endosteal osteoclast formation and bone resorption. S. aureus stimulation of neonatal mouse parietal bone induced ex vivo bone resorption and osteoclastic gene expression. This effect was associated with increased mRNA and protein expression of receptor activator of NF-kB ligand (RANKL) without significant change in osteoprotegerin (OPG) expression. Bone resorption induced by S. aureus was abolished by OPG. S. aureus increased the expression of osteoclastogenic cytokines and prostaglandins in the parietal bones but the stimulatory effect of S. aureus on bone resorption and Tnfsf11 mRNA expression was independent of these cytokines and prostaglandins. Stimulation of isolated periosteal osteoblasts with S. aureus also resulted in increased expression of Tnfsf11 mRNA, an effect lost in osteoblasts from Tlr2 knockout mice. S. aureus stimulated osteoclastogenesis in isolated periosteal cells without affecting RANKL-stimulated resorption. In contrast, S. aureus inhibited RANKL-induced osteoclast formation in bone marrow macrophages. These data show that S. aureus enhances bone resorption and periosteal osteoclast formation by increasing osteoblast RANKL production through TLR2. Our study indicates the importance of using different in vitro approaches for studies of how S. aureus regulates osteoclastogenesis to obtain better understanding of the complex mechanisms of S. aureus induced bone destruction in vivo.


Bone Resorption/immunology , Osteogenesis/immunology , Parietal Bone/immunology , RANK Ligand/genetics , Staphylococcal Infections/immunology , Toll-Like Receptor 2/genetics , Animals , Animals, Newborn , Bone Marrow Cells/immunology , Bone Marrow Cells/microbiology , Bone Resorption/microbiology , Bone Resorption/pathology , Gene Expression Regulation, Developmental , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/immunology , Osteoblasts/microbiology , Osteoclasts/immunology , Osteoclasts/microbiology , Osteogenesis/genetics , Osteoprotegerin/genetics , Osteoprotegerin/immunology , Parietal Bone/growth & development , Parietal Bone/microbiology , Primary Cell Culture , Prostaglandins/biosynthesis , RANK Ligand/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/immunology
19.
Biotechnol Lett ; 38(9): 1443-8, 2016 Sep.
Article En | MEDLINE | ID: mdl-27262294

OBJECTIVE: To investigate the effects of heat-killed Enterococcus faecalis ATCC 29212 and P25RC clinical strain (derived from an obturated root canal with apical periodontitis) on osteoclast differentiation within an osteoblast/osteoclast co-culture system. RESULTS: Heat-killed E. faecalis significantly increased the proportion of multinucleated osteoclastic cells (MNCs) within the co-culture system. The IL-6 level was significantly increased upon exposure to heat-killed E. faecalis. Gene expression levels of NFATc1 and cathepsin K were significantly up-regulated compared to the untreated control. EphrinB2 and EphB4 expressions at both the mRNA and protein levels were also significantly upregulated compared to the untreated control. CONCLUSIONS: Heat-killed E. faecalis can induce osteoclast differentiation within the osteoblast/osteoclast co-culture system in vitro, possibly through ephrinB2-EphB4 bidirectional signaling.


Enterococcus faecalis/pathogenicity , Osteoblasts/microbiology , Osteoclasts/microbiology , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Mice , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology
20.
Infect Immun ; 84(9): 2586-94, 2016 09.
Article En | MEDLINE | ID: mdl-27354444

We used a murine model of acute, posttraumatic osteomyelitis to evaluate the virulence of two divergent Staphylococcus aureus clinical isolates (the USA300 strain LAC and the USA200 strain UAMS-1) and their isogenic sarA mutants. The results confirmed that both strains caused comparable degrees of osteolysis and reactive new bone formation in the acute phase of osteomyelitis. Conditioned medium (CM) from stationary-phase cultures of both strains was cytotoxic to cells of established cell lines (MC3TC-E1 and RAW 264.7 cells), primary murine calvarial osteoblasts, and bone marrow-derived osteoclasts. Both the cytotoxicity of CM and the reactive changes in bone were significantly reduced in the isogenic sarA mutants. These results confirm that sarA is required for the production and/or accumulation of extracellular virulence factors that limit osteoblast and osteoclast viability and that thereby promote bone destruction and reactive bone formation during the acute phase of S. aureus osteomyelitis. Proteomic analysis confirmed the reduced accumulation of multiple extracellular proteins in the LAC and UAMS-1 sarA mutants. Included among these were the alpha class of phenol-soluble modulins (PSMs), which were previously implicated as important determinants of osteoblast cytotoxicity and bone destruction and repair processes in osteomyelitis. Mutation of the corresponding operon reduced the cytotoxicity of CM from both UAMS-1 and LAC cultures for osteoblasts and osteoclasts. It also significantly reduced both reactive bone formation and cortical bone destruction by CM from LAC cultures. However, this was not true for CM from cultures of a UAMS-1 psmα mutant, thereby suggesting the involvement of additional virulence factors in such strains that remain to be identified.


Bacterial Proteins/genetics , Osteomyelitis/microbiology , Osteomyelitis/pathology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Virulence Factors/genetics , Virulence/genetics , Animals , Gene Expression Regulation, Bacterial/genetics , Mice , Mice, Inbred C57BL , Mutation/genetics , Operon/genetics , Osteoblasts/microbiology , Osteoblasts/pathology , Osteoclasts/microbiology , Osteoclasts/pathology , Proteomics/methods , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology
...