Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Exp Mol Pathol ; 116: 104485, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32574668

RESUMEN

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.


Asunto(s)
Artritis Infecciosa/inmunología , Inflamación/inmunología , Infecciones Estafilocócicas/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/microbiología , Artritis Experimental/patología , Artritis Infecciosa/genética , Artritis Infecciosa/microbiología , Artritis Infecciosa/patología , Regulación de la Expresión Génica/genética , Humanos , Inflamación/genética , Inflamación/microbiología , Inflamación/patología , Interleucina-1beta/genética , Articulaciones/inmunología , Articulaciones/microbiología , Articulaciones/patología , MAP Quinasa Quinasa 4/genética , Ratones , Osteoclastos/inmunología , Osteoclastos/microbiología , Osteoclastos/patología , Ligando RANK/genética , Transducción de Señal/genética , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/patogenicidad , Linfocitos T Reguladores/microbiología , Células Th17/microbiología , Factor de Crecimiento Transformador beta/genética , Factor de Necrosis Tumoral alfa/genética
2.
Sci Rep ; 10(1): 7823, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32385413

RESUMEN

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.


Asunto(s)
Pérdida de Hueso Alveolar/genética , Caspasa 1/genética , Inflamación/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Periodontitis/genética , Aggregatibacter actinomycetemcomitans , Pérdida de Hueso Alveolar/microbiología , Pérdida de Hueso Alveolar/patología , Animales , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encía/crecimiento & desarrollo , Encía/microbiología , Humanos , Inflamación/microbiología , Inflamación/patología , Interleucina-10/genética , Interleucina-12/genética , Ratones , Ratones Noqueados , Osteoclastos/microbiología , Osteoclastos/patología , Periodontitis/microbiología , Periodontitis/patología , Ligando RANK/genética , Factor de Necrosis Tumoral alfa/genética
3.
Infect Immun ; 88(4)2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-31932325

RESUMEN

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.


Asunto(s)
Vacuna contra la Brucelosis/efectos adversos , Brucella abortus/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Osteoartritis/fisiopatología , Osteoclastos/inmunología , Osteoclastos/microbiología , Animales , Resorción Ósea , Vacuna contra la Brucelosis/administración & dosificación , Proliferación Celular , Células Cultivadas , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Viabilidad Microbiana , Osteoclastos/fisiología
4.
J Periodontal Res ; 55(3): 410-425, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31944305

RESUMEN

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.


Asunto(s)
Infecciones por Bacteroidaceae/patología , Osteoclastos/citología , Cráneo/microbiología , Animales , Diferenciación Celular , Ratones , Ratones Endogámicos C57BL , Osteoclastos/microbiología , Porphyromonas gingivalis , Ligando RANK
5.
Inflammation ; 43(1): 220-230, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31720989

RESUMEN

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.


Asunto(s)
Adenina/análogos & derivados , Pérdida de Hueso Alveolar/prevención & control , Proceso Alveolar/efectos de los fármacos , Antiinflamatorios/farmacología , Autofagia/efectos de los fármacos , Cloroquina/farmacología , Osteoclastos/efectos de los fármacos , Periodontitis/prevención & control , Adenina/farmacología , Pérdida de Hueso Alveolar/metabolismo , Pérdida de Hueso Alveolar/microbiología , Pérdida de Hueso Alveolar/patología , Proceso Alveolar/metabolismo , Proceso Alveolar/microbiología , Proceso Alveolar/patología , Animales , Proteínas Relacionadas con la Autofagia/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Masculino , Osteoclastos/metabolismo , Osteoclastos/microbiología , Osteoclastos/patología , Osteogénesis/efectos de los fármacos , Periodontitis/metabolismo , Periodontitis/microbiología , Periodontitis/patología , Porphyromonas gingivalis , Ratas Sprague-Dawley , Factor de Transcripción ReIA/metabolismo
6.
mBio ; 10(5)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31615966

RESUMEN

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.


Asunto(s)
Osteoclastos/microbiología , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Diferenciación Celular , Células Cultivadas , Femenino , Macrófagos/metabolismo , Masculino , Ratones , Osteoblastos/microbiología , Osteomielitis/metabolismo , Osteomielitis/microbiología , Fagosomas/metabolismo , Ligando RANK/metabolismo , Staphylococcus aureus/efectos de los fármacos
7.
Bone ; 127: 315-323, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31233933

RESUMEN

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.


Asunto(s)
Bacterias/metabolismo , Osteoclastos/microbiología , Osteoclastos/virología , Animales , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/patología , Endocitosis , Humanos , Osteoclastos/patología , Virosis/patología
8.
FASEB J ; 33(9): 10515-10527, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31251083

RESUMEN

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.


Asunto(s)
Inflamación/prevención & control , Histona Demetilasas con Dominio de Jumonji/fisiología , Enfermedades de la Boca/prevención & control , FN-kappa B/antagonistas & inhibidores , Osteogénesis , Porphyromonas gingivalis/patogenicidad , Animales , Infecciones por Bacteroidaceae/complicaciones , Infecciones por Bacteroidaceae/microbiología , Diferenciación Celular , Citocinas , Histonas , Humanos , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Enfermedades de la Boca/etiología , Enfermedades de la Boca/metabolismo , Enfermedades de la Boca/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Osteoclastos/metabolismo , Osteoclastos/microbiología , Osteoclastos/patología , Fosforilación , Transducción de Señal
9.
Molecules ; 23(7)2018 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-29937485

RESUMEN

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.


Asunto(s)
Remodelación Ósea/genética , Microbioma Gastrointestinal/fisiología , Vida Libre de Gérmenes , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis/genética , Simbiosis/fisiología , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Biomarcadores/metabolismo , Catepsina K/genética , Catepsina K/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Osteoblastos/citología , Osteoblastos/microbiología , Osteocalcina/genética , Osteocalcina/metabolismo , Osteoclastos/citología , Osteoclastos/microbiología , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo
10.
Exp Cell Res ; 362(1): 152-158, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29129564

RESUMEN

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.


Asunto(s)
Enterococcus faecalis/inmunología , Macrófagos/microbiología , Osteoclastos/microbiología , Osteogénesis/inmunología , Animales , Apoptosis/inmunología , Biomarcadores/metabolismo , Diferenciación Celular/inmunología , Línea Celular , Citocinas/metabolismo , Inflamación/metabolismo , Inflamación/microbiología , Macrófagos/metabolismo , Ratones , Osteoclastos/metabolismo , Fagocitosis/inmunología , Células RAW 264.7
11.
Med Sci Monit ; 23: 4579-4590, 2017 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-28942456

RESUMEN

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.


Asunto(s)
FN-kappa B/fisiología , Osteogénesis/efectos de los fármacos , Staphylococcus aureus/patogenicidad , Animales , Resorción Ósea/metabolismo , Resorción Ósea/microbiología , Diferenciación Celular/fisiología , Regulación de la Expresión Génica/genética , Macrófagos/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Subunidad p52 de NF-kappa B/fisiología , Factores de Transcripción NFATC , Osteoclastos/metabolismo , Osteoclastos/microbiología , Osteogénesis/inmunología , Osteogénesis/fisiología , Osteomielitis/microbiología , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Staphylococcus aureus/metabolismo , Factor de Transcripción ReIA/metabolismo
12.
J Cell Physiol ; 232(9): 2396-2406, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28185243

RESUMEN

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.


Asunto(s)
Remodelación Ósea , Huesos/enzimología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteoclastos/enzimología , Osteomielitis/enzimología , Infecciones Estafilocócicas/enzimología , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/metabolismo , Animales , Apoptosis , Resorción Ósea/enzimología , Resorción Ósea/microbiología , Resorción Ósea/patología , Huesos/microbiología , Huesos/patología , Estudios de Casos y Controles , Diferenciación Celular , Activación Enzimática , Interacciones Huésped-Patógeno , Humanos , Ratones , Factores de Transcripción NFATC/metabolismo , Osteoclastos/microbiología , Osteoclastos/patología , Osteomielitis/microbiología , Osteomielitis/patología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ligando RANK/metabolismo , Células RAW 264.7 , Transducción de Señal , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/patogenicidad
13.
Infect Immun ; 85(1)2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27795356

RESUMEN

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.


Asunto(s)
Aggregatibacter actinomycetemcomitans/patogenicidad , Pérdida de Hueso Alveolar/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/metabolismo , Macrófagos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Pérdida de Hueso Alveolar/microbiología , Pérdida de Hueso Alveolar/fisiopatología , Animales , Células Cultivadas , Quimiocinas/metabolismo , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/fisiopatología , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoclastos/metabolismo , Osteoclastos/microbiología , Infecciones por Pasteurellaceae/metabolismo , Infecciones por Pasteurellaceae/microbiología , ARN/metabolismo
14.
FASEB J ; 30(12): 4033-4041, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27535487

RESUMEN

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.


Asunto(s)
Pérdida de Hueso Alveolar/prevención & control , Melanocortinas/agonistas , Osteoclastos/microbiología , Infecciones por Pasteurellaceae/prevención & control , Enfermedades Periodontales/metabolismo , Aggregatibacter actinomycetemcomitans , Pérdida de Hueso Alveolar/etiología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Macrófagos/inmunología , Ratones Endogámicos C57BL , Periodontitis/tratamiento farmacológico , Periodontitis/metabolismo
15.
PLoS One ; 11(6): e0156708, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27311019

RESUMEN

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.


Asunto(s)
Resorción Ósea/inmunología , Osteogénesis/inmunología , Hueso Parietal/inmunología , Ligando RANK/genética , Infecciones Estafilocócicas/inmunología , Receptor Toll-Like 2/genética , Animales , Animales Recién Nacidos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/microbiología , Resorción Ósea/microbiología , Resorción Ósea/patología , Regulación del Desarrollo de la Expresión Génica , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/inmunología , Osteoblastos/microbiología , Osteoclastos/inmunología , Osteoclastos/microbiología , Osteogénesis/genética , Osteoprotegerina/genética , Osteoprotegerina/inmunología , Hueso Parietal/crecimiento & desarrollo , Hueso Parietal/microbiología , Cultivo Primario de Células , Prostaglandinas/biosíntesis , Ligando RANK/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/inmunología
16.
Free Radic Biol Med ; 97: 330-341, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27343691

RESUMEN

Osteoclastogenesis was induced by RANKL stimulation in mouse monocytes to examine the possible bactericidal function of osteoclast precursors (OCp) and mature osteoclasts (OCm) relative to their production of NO and ROS. Tartrate-resistant acid phosphatase (TRAP)-positive OCp, but few or no OCm, phagocytized and killed Escherichia coli in association with the production of reactive oxygen species (ROS) and nitric oxide (NO). Phagocytosis of E. coli and production of ROS and NO were significantly lower in TRAP+ OCp derived from Toll-like receptor (TLR)-4 KO mice than that derived from wild-type (WT) or TLR2-KO mice. Interestingly, after phagocytosis, TRAP+ OCp derived from wild-type and TLR2-KO mice did not differentiate into OCm, even with continuous exposure to RANKL. In contrast, E. coli-phagocytized TRAP+ OCp from TLR4-KO mice could differentiate into OCm. Importantly, neither NO nor ROS produced by TRAP+ OCp appeared to be engaged in phagocytosis-induced suppression of osteoclastogenesis. These results suggested that TLR4 signaling not only induces ROS and NO production to kill phagocytized bacteria, but also interrupts OCm differentiation. Thus, it can be concluded that TRAP+ OCp, but not OCm, can mediate bactericidal activity via phagocytosis accompanied by the production of ROS and NO via TLR4-associated reprograming toward phagocytic cell type.


Asunto(s)
Óxido Nítrico/fisiología , Osteoclastos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Fosfatasa Ácida Tartratorresistente/fisiología , Receptor Toll-Like 4/fisiología , Animales , Escherichia coli/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Viabilidad Microbiana , Osteoclastos/microbiología , Fagocitosis , Ligando RANK/fisiología , Células RAW 264.7
17.
Oncol Rep ; 36(1): 410-8, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27177272

RESUMEN

Bone is the most common distant metastatic site of lung cancer, and is particularly prone to osteolytic damage. Soluble factors secreted from bone marrow-derived cells and tumor cells contribute to the growth and metastasis of cancer cells, and enhance osteolytic damage. BMP9, as the most powerful osteogenetic factor of the bone morphogenetic protein (BMP) family, can regulate the development of various tumors. However, the effects and underlying mechanisms of BMP9 in regards to lung cancer and the bone metastatic microenvironment are poorly understood. Here, we determined the inhibitory effects of BMP9 on the proliferation and migration of lung adenocarcinoma A549 cells. When a co-culture system of A549 cells and bone marrow-derived cells (HS-5) was established, it was shown that HS-5 cells promoted the proliferation and migration of A549 cells, and metastasis and osteoclast-related factors IL-6 and IL-8 were increased in the A549 and HS-5 cells. However, BMP9 inhibited the proliferation and migration of the A549 cells in the bone microenvironment, and decreased the levels of IL-6 and IL-8. In addition, mitogen-activated protein kinase (MAPK/ERK) and nuclear factor-κB (NF-κB) signaling pathway may be involved in these effects.


Asunto(s)
Adenocarcinoma/genética , Movimiento Celular/genética , Microambiente Celular/genética , Factores de Diferenciación de Crecimiento/genética , Neoplasias Pulmonares/genética , Células Madre Mesenquimatosas/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , FN-kappa B/genética , Células A549 , Adenocarcinoma/metabolismo , Adenocarcinoma del Pulmón , Línea Celular Tumoral , Proliferación Celular/genética , Técnicas de Cocultivo/métodos , Factor 2 de Diferenciación de Crecimiento , Humanos , Interleucina-6/genética , Interleucina-8/genética , Neoplasias Pulmonares/metabolismo , Osteoclastos/microbiología , Transducción de Señal/genética
18.
Arch Oral Biol ; 66: 108-19, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26945169

RESUMEN

OBJECTIVES: The purposes of this study were to evaluate, in rats: i) the effects of Bacillus species on the development of experimental periodontitis (EP) via microtomographic, immunological and hematological assays (Experiment 1-E1); ii) the effects of Bacillus species as adjuncts to scaling and root planing (SRP) for the treatment of EP via histomorphometric and immunohistochemical analyses (Experiment 2-E2). METHODS: In E1, 24 rats were divided into groups C1 (control), PROB1, EP1 and EP-PROB1. In groups with EP, the mandibular first molar of each animal received a ligature for 14 days. In groups PROB1, animals received Bacillus species for 44 days, starting 30 days before EP induction in Group EP-PROB1. In E2, 24 rats were assigned to groups C2 (control), PROB2, EP-SRP2 and EP-SRP-PROB2. In groups with SRP, EP was induced as described in E1. The ligatures were removed after 14 days and SRP was performed. In groups PROB2, animals received Bacillus species for 15 days, starting after SRP in Group EP-SRP-PROB2. RESULTS: In E1, Group EP1 presented bone loss (BL) and eosinophil numbers greater than Group EP-PROB1 (P<0.05). In Group EP-PROB1, the receptor activator of nuclear factor-kB ligand (RANKL)/osteoprotegerin (OPG) ratio was similar to that of groups without EP. In E2, Group EP-SRP-PROB2 presented fewer TRAP-positive osteoclasts, lower immunolabeling pattern for a proinflammatory cytokine and decreased BL and attachment loss than Group EP-SRP2 (P<0.05). CONCLUSIONS: Bacillus species supplementation provided a protective effect against BL and enhanced the effects of SRP in the treatment of EP in rats.


Asunto(s)
Bacillus licheniformis/fisiología , Bacillus subtilis/fisiología , Periodontitis/microbiología , Periodontitis/terapia , Probióticos/farmacología , Pérdida de Hueso Alveolar/diagnóstico por imagen , Pérdida de Hueso Alveolar/microbiología , Pérdida de Hueso Alveolar/patología , Pérdida de Hueso Alveolar/terapia , Animales , Raspado Dental/métodos , Inmunohistoquímica , Masculino , Diente Molar/microbiología , Osteoclastos/metabolismo , Osteoclastos/microbiología , Osteoclastos/patología , Osteoprotegerina/metabolismo , Periodontitis/sangre , Periodontitis/patología , Distribución Aleatoria , Ratas , Ratas Wistar , Aplanamiento de la Raíz/métodos , Microtomografía por Rayos X/métodos
19.
Arch Oral Biol ; 66: 77-85, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26921718

RESUMEN

OBJECTIVE: P. gingivalis is a gram-negative anaerobic bacterium and a major periodontal pathogen. LPS produced by P. gingivalis promotes osteoclast formation. TECK is a CC chemokine whose expression is increased in gingival epithelial cells exposed to P. gingivalis LPS. In this study, we investigated the effect of TECK in osteoclastogenesis induced by P. gingivalis LPS. DESIGNS: Real time reverse transcriptase polymerase chain reaction (RTPCR) analysis and western blotting were performed to confirm TECK in MG63, human osteoblast cell line and primary murine osteoblasts and CCR9 in RAW 264.7 cells and murine bone marrow macrophages (BMMs) as osteoclast precursors. P. gingivalis LPS-treated BMMs and Raw 264.7 cells were cultured with or without TECK or TECK antibody to examine the effect of TECK on osteoclast formation. Cocultures with murine osteoblasts and bone marrow cells were also treated with or without TECK or TECK antibody. Luciferase assay and western blotting were used to determine whether TECK-CCR9 induced osteoclastogenesis was mediated through NFATc1 or NF-kB signaling. RESULTS: TECK was shown to be expressed by osteoblasts, and its receptor, CCR9, by osteoclast precursors. TECK increased P. gingivalis LPS-induced osteoclast numbers in an in vitro osteoclast formation assay using osteoclast precursors. The enhanced osteoclast formation by TECK was mediated by NFATc1, but not by NF-kB signaling. CONCLUSION: TECK may be a novel regulator of osteoclast formation induced by P. gingivalis LPS in periodontitis.


Asunto(s)
Quimiocinas CC/farmacología , Lipopolisacáridos/farmacología , Factores de Transcripción NFATC/metabolismo , Osteoclastos/efectos de los fármacos , Porphyromonas gingivalis/fisiología , Animales , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Quimiocinas CC/biosíntesis , Encía/citología , Encía/metabolismo , Humanos , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Osteoclastos/microbiología , Osteogénesis , Porphyromonas gingivalis/efectos de los fármacos , Células RAW 264.7 , Receptores CCR/biosíntesis , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos
20.
J Dent Res ; 95(2): 223-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26450512

RESUMEN

Obesity is associated with abnormal lipid metabolism and impaired bone homeostasis. The aim of our study was to investigate the impact of specific elevated fatty acid (FA) levels on alveolar bone loss in a Porphyromonas gingivalis-induced model of periodontal disease and to analyze underlying cellular mechanisms in bone-resorbing osteoclasts and bone-forming osteoblasts in mice. Four-week-old male C57BL/6 mice were randomly divided in groups and subjected to a palmitic acid (PA)- or oleic acid (OA)-enriched high-fat diet (HFD) (20% of calories from FA) or a normal caloric diet (C group) (10% of calories from FA) for 16 wk. Starting at week 10, mice were infected orally with P. gingivalis (W50) or placebo to induce alveolar bone loss. Animals were sacrificed, and percentage fat, serum inflammation (tumor necrosis factor [TNF]-α), and bone metabolism (osteocalcin [OC], carboxy-terminal collagen crosslinks [CTX], and N-terminal propeptides of type I procollagen [P1NP]) markers were measured. Osteoblasts and osteoclasts were cultured in the presence of elevated PA or OA levels and exposed to P. gingivalis. Animals on FA-enriched diets weighed significantly more compared with animals on a normal caloric diet (P < 0.05). Both obese groups had similar percentages of fat (P = nonsignificant); however, alveolar bone loss was significantly greater in animals that were on the PA-enriched HFD (P < 0.05). TNF-α levels were highest in the PA group (P < 0.001) and increased in all groups in response to P. gingivalis inoculation (P < 0.01), whereas bone remodeling markers OC, CTX, and P1NP were lowest in the PA group (P < 0.001) and highest in the C group. Bacterial challenge decreased bone metabolism markers in all groups (P < 0.01). Further, osteoclasts showed an augmented inflammatory response to P. gingivalis in the presence of hyperlipidemic PA levels as opposed to OA cultures, which responded similarly to controls. These findings indicate that the specific FA profile of diet rather than weight gain and obesity alone modulates bone metabolism and can therefore influence alveolar bone loss.


Asunto(s)
Pérdida de Hueso Alveolar/etiología , Dieta Alta en Grasa/efectos adversos , Obesidad/complicaciones , Pérdida de Hueso Alveolar/inmunología , Pérdida de Hueso Alveolar/microbiología , Animales , Peso Corporal , Remodelación Ósea/fisiología , Células Cultivadas , Colágeno Tipo I/sangre , Interleucina-6/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Obesidad/inmunología , Obesidad/microbiología , Ácido Oléico/sangre , Ácido Oléico/farmacología , Osteoblastos/inmunología , Osteoblastos/microbiología , Osteocalcina/sangre , Osteoclastos/inmunología , Osteoclastos/microbiología , Ácido Palmítico/sangre , Ácido Palmítico/farmacología , Fragmentos de Péptidos/sangre , Péptidos/sangre , Placebos , Porphyromonas gingivalis/fisiología , Procolágeno/sangre , Distribución Aleatoria , Receptor Toll-Like 2/análisis , Receptor Toll-Like 4/análisis , Factor de Necrosis Tumoral alfa/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA