Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Biol Sci ; 20(7): 2555-2575, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38725861

RESUMEN

Staphylococcus aureus (S. aureus) persistence in macrophages, potentially a reservoir for recurrence of chronic osteomyelitis, contributes to resistance and failure in treatment. As the mechanisms underlying survival of S. aureus in macrophages remain largely unknown, there has been no treatment approved. Here, in a mouse model of S. aureus osteomyelitis, we identified significantly up-regulated expression of SLC7A11 in both transcriptomes and translatomes of CD11b+F4/80+ macrophages, and validated a predominant distribution of SLC7A11 in F4/80+ cells around the S. aureus abscess. Importantly, pharmacological inhibition or genetic knockout of SLC7A11 promoted the bactericidal function of macrophages, reduced bacterial burden in the bone and improved bone structure in mice with S. aureus osteomyelitis. Mechanistically, aberrantly expressed SLC7A11 down-regulated the level of intracellular ROS and reduced lipid peroxidation, contributing to the impaired bactericidal function of macrophages. Interestingly, blocking SLC7A11 further activated expression of PD-L1 via the ROS-NF-κB axis, and a combination therapy of targeting both SLC7A11 and PD-L1 significantly enhanced the efficacy of clearing S. aureus in vitro and in vivo. Our findings suggest that targeting both SLC7A11 and PD-L1 is a promising therapeutic approach to reprogram the bactericidal function of macrophages and promote bacterial clearance in S. aureus osteomyelitis.


Asunto(s)
Macrófagos , Osteomielitis , Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Osteomielitis/microbiología , Osteomielitis/metabolismo , Osteomielitis/genética , Ratones , Macrófagos/metabolismo , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
2.
Front Cell Infect Microbiol ; 13: 1187543, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37529351

RESUMEN

Introduction: Staphylococcus aureus (S. aureus) osteomyelitis causes a variety of metabolism disorders in microenvironment and cells. Defining the changes in cholesterol metabolism and identifying key factors involved in cholesterol metabolism disorders during S. aureus osteomyelitis is crucial to understanding the mechanisms of S. aureus osteomyelitis and is important in designing host-directed therapeutic strategies. Methods: In this study, we conducted in vitro and in vivo experiments to define the effects of S. aureus osteomyelitis on cholesterol metabolism, as well as the role of Apolipoprotein E (ApoE) in regulating cholesterol metabolism by macrophages during S. aureus osteomyelitis. Results: The data from GSE166522 showed that cholesterol metabolism disorder was induced by S. aureus osteomyelitis. Loss of cholesterol from macrophage obtained from mice with S. aureus osteomyelitis was detected by liquid chromatography-tandem mass spectrometry(LC-MS/MS), which is consistent with Filipin III staining results. Changes in intracellular cholesterol content influenced bactericidal capacity of macrophage. Subsequently, it was proven by gene set enrichment analysis and qPCR, that ApoE played a key role in developing cholesterol metabolism disorder in S. aureus osteomyelitis. ApoE deficiency in macrophages resulted in increased resistance to S. aureus. ApoE-deficient mice manifested abated bone destruction and decreased bacteria load. Moreover, the combination of transcriptional analysis, qPCR, and killing assay showed that ApoE deficiency led to enhanced cholesterol biosynthesis in macrophage, ameliorating anti-infection ability. Conclusion: We identified a previously unrecognized role of ApoE in S. aureus osteomyelitis from the perspective of metabolic reprogramming. Hence, during treating S. aureus osteomyelitis, considering cholesterol metabolism as a potential therapeutic target presents a new research direction.


Asunto(s)
Osteomielitis , Infecciones Estafilocócicas , Ratones , Animales , Staphylococcus aureus , Cromatografía Liquida , Espectrometría de Masas en Tándem , Macrófagos/metabolismo , Colesterol/metabolismo , Osteomielitis/microbiología , Infecciones Estafilocócicas/microbiología , Apolipoproteínas E/genética
3.
Mol Ther ; 31(1): 174-192, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36104974

RESUMEN

There is no effective therapy for implant-associated Staphylococcus aureus osteomyelitis, a devastating complication after orthopedic surgery. An immune-suppressive profile with up-regulated programmed cell death 1/programmed death ligand 1 (PD-1/PD-L1) was identified based on our transcriptional data (GEO: GSE166522) from a mouse model of S. aureus osteomyelitis. PD-1/PD-L1 expression was up-regulated mainly in F4/80+ macrophages surrounding the abscess in S. aureus-infected bone. Mechanistically, PD-1/PD-L1 activated mitophagy to suppress production of mitochondrial reactive oxygen species (ROS), suppressing the bactericidal function of macrophages. Using neutralizing antibodies for PD-L1 or PD-1, or knockout of PD-L1 adjuvant to gentamicin markedly reduced mitophagy in bone marrow F4/80+ cells, enhanced bacterial clearance in bone tissue and implants, and reduced bone destruction in mice. PD-1/PD-L1 expression was also increased in the bone marrow from individuals with S. aureus osteomyelitis. These findings uncover a so far unknown function of PD-1/PD-L1-mediated mitophagy in suppressing the bactericidal function of bone marrow macrophages.


Asunto(s)
Anticuerpos , Antígeno B7-H1 , Osteomielitis , Receptor de Muerte Celular Programada 1 , Animales , Ratones , Adyuvantes Inmunológicos , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Osteomielitis/metabolismo , Osteomielitis/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Staphylococcus aureus , Modelos Animales de Enfermedad , Anticuerpos/uso terapéutico
5.
Acta Biomater ; 152: 575-592, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36070834

RESUMEN

Implant-associated infections (IAI) and osseointegration disorders are the most common complications in orthopedics. Studies have shown that neutrophils surrounding implants play a vital role in regulating these complications. Although magnesium (Mg) and its alloys are considered promising biodegradable bone implants, their role in neutrophil-mediated antibacteria has not yet been examined. Considering the rapid corrosion of Mg, it is necessary to develop methods to inhibit its corrosion. To solve these issues, a zinc-doped ferric oxyhydroxide nano-layer modified plasma electrolytic oxidation (PEO)-coated Mg alloy (PEO-FeZn) was developed in this study, and its antibacterial, immune anti-infective, and osteogenic ability were systematically evaluated. The results showed that PEO-FeZn nano-layer enhanced the corrosion resistance, biocompatibility, bactericidal activity, and osteoblastic differentiation activity of the Mg alloy. Moreover, PEO-FeZn nano-layer inhibited immune evasion-related gene expression and contributed to the formation of neutrophil extracellular traps (NETs) by activating the extracellular release of DNA fibers and granule proteins, and thereby suppressing bacterial invasion and promoting osseointegration in vivo in Staphylococcus aureus (S. aureus)-infected rat femurs. Overall, the findings of this study could serve as a reference for the fabrication of highly biocompatible and corrosion resistant Mg alloys to address the challenges of IAI and osseointegration disorders. STATEMENT OF SIGNIFICANCE: The widely used metallic biomaterials usually come with the risk of IAI. As the first responder around the biomaterials, neutrophils could form NETs to defense against microorganism and promote tissue remodeling. Therefore, biomaterials addressing antibacterial and neutrophils-modulatory strategies are highly necessary in reducing IAI. To solve these issues, we grew PEO-FeZn nano-layers in situ on Mg alloy using a simple and green technique. The nano-layer not only enhanced the corrosion resistance and biocompatibility of Mg alloy, but also elevated the antibacterial and osteogenesis capability. Moreover, nano-layer contributed to NETs formation, thereby suppressing bacterial invasion and even promoting osseointegration in S.aureus-infected femurs. Accordingly, this functionalized multilayer coating with antibacterial immunity represents a novel therapeutic strategy for IAI and weak osseointegration.


Asunto(s)
Aleaciones , Trampas Extracelulares , Implantes Absorbibles , Aleaciones/farmacología , Animales , Antibacterianos/farmacología , Materiales Biocompatibles Revestidos/farmacología , Corrosión , Compuestos Férricos , Magnesio/farmacología , Oseointegración , Ratas , Staphylococcus aureus , Propiedades de Superficie , Zinc/farmacología
6.
Comput Intell Neurosci ; 2022: 1775496, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36072720

RESUMEN

The click-through rate (CTR) prediction task is used to estimate the probabilities of users clicking on recommended items, which are extremely important in recommender systems. Recently, the deep factorization machine (DeepFM) algorithm was proposed. The DeepFM algorithm incorporates a factorization machine (FM) to learn not only low-order features but also the interactions of higher-order features. However, DeepFM lacks user diversity representations and does not consider the text. In view of this, we propose a text-attention FM (TAFM) based on the DeepFM algorithm. First, the attention mechanism in the TAFM algorithm is used to address the diverse representations of users and goods and to mine the features that are most interesting to users. Second, the TAFM model can fully learn text features through its text component, text attention component, and N-gram text feature extraction component, which can fully explore potential user preferences and the diversity among user interests. In addition, the convolutional autoencoder in the TAFM can learn some higher-level features, and the higher-order feature mining process is more comprehensive. On the public dataset, the better performing models in the existing models are deep cross network (DCN), DeepFM, and product-based neural network (PNN), respectively, and the AUC score metrics of these models hover between 0.698 and 0.699. The AUC score of our design model is 0.730, which is at least 3% higher than that of the existing models. The accuracy metric of our model is at least 0.1 percentage points higher than that of existing models.


Asunto(s)
Algoritmos , Redes Neurales de la Computación , Aprendizaje
7.
FASEB J ; 35(10): e21851, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34547121

RESUMEN

It has been known that moderate mechanical loading, like that caused by exercise, promotes bone formation. However, its underlying mechanisms remain elusive. Here we showed that moderate running dramatically improved trabecular bone in mice tibias with an increase in bone volume fraction and trabecular number and a decrease in trabecular pattern factor. Results of immunohistochemical and histochemical staining revealed that moderate running mainly increased the number of osteoblasts but had no effect on osteoclasts. In addition, we observed a dramatic increase in the number of colony forming unit-fibroblast in endosteal bone marrow and the percentage of CD45- Leptin receptor+ (CD45- LepR+ ) endosteal mesenchymal progenitors. Bioinformatics analysis of the transcriptional data from gene expression omnibus (GEO) database identified chemokine c-c-motif ligands (CCL2) as a critical candidate induced by mechanical loading. Interestingly, we found that CCL2 was up-regulated mainly in osteoblastic cells in the tibia of mice after moderate running. Further, we found that mechanical loading up-regulated the expression of CCL2 by activating ERK1/2 pathway, thereby stimulating migration of endosteal progenitors. Finally, neutralizing CCL2 abolished the recruitment of endosteal progenitors and the increased bone formation in mice after 4 weeks running. These results therefore uncover an unknown connection between osteoblasts and endosteal progenitors recruited in the increased bone formation induced by mechanical loading.


Asunto(s)
Hueso Esponjoso/citología , Quimiocina CCL2/metabolismo , Células Madre Mesenquimatosas/citología , Osteoblastos/citología , Osteogénesis , Condicionamiento Físico Animal , Animales , Hueso Esponjoso/metabolismo , Movimiento Celular , Quimiocina CCL2/genética , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo
8.
Microb Cell Fact ; 20(1): 102, 2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001083

RESUMEN

As treatment of Staphylococcus aureus (S. aureus) osteomyelitis is often hindered by the development of antibiotic tolerance, novel antibacterial therapeutics are required. Here we found that the cell-free supernatant of Bacillus subtilis (B. subtilis CFS) killed planktonic and biofilm S. aureus, and increased S. aureus susceptibility to penicillin and gentamicin as well. Further study showed that B. subtilis CFS suppressed the expression of the genes involved in adhesive molecules (Cna and ClfA), virulence factor Hla, quorum sensing (argA, argB and RNAIII) and biofilm formation (Ica and sarA) in S. aureus. Additionally, our data showed that B. subtilis CFS changed the membrane components and increased membrane permeabilization of S. aureus. Finally, we demonstrated that B. subtilis CFS increased considerably the susceptibility of S. aureus to penicillin and effectively reduced S. aureus burdens in a mouse model of implant-associated osteomyelitis. These findings support that B. subtilis CFS may be a potential resistance-modifying agent for ß-lactam antibiotics against S. aureus.


Asunto(s)
Antibacterianos/farmacología , Bacillus subtilis/crecimiento & desarrollo , Medios de Cultivo/farmacología , Osteomielitis/microbiología , Staphylococcus aureus/efectos de los fármacos , Animales , Antibacterianos/administración & dosificación , Antibacterianos/química , Bacillus subtilis/química , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Biopelículas/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Medios de Cultivo/química , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Osteomielitis/tratamiento farmacológico , Percepción de Quorum , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética
9.
Infect Immun ; 89(5)2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33619031

RESUMEN

To investigate the molecular pathogenesis of bone with osteomyelitis, we developed implant-associated osteomyelitis (IAOM) models in mice. An orthopedic stainless pin was surgically placed in the right femoral midshaft of mice, followed by an inoculation of Staphylococcus aureus into the medullary cavity. Typical characteristics of IAOM, like periosteal reaction and intraosseous abscess, occurred by day 14 postinfection. By day 28 postinfection, necrotic abscess, sequestrum formation, and deformity of the whole femur were observed. Transcriptional analysis identified 101 and 1,702 differentially expressed genes (DEGs) between groups by days 3 and 14 postinfection, respectively. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed the enrichment of pathways in response to the bacterium, receptor-ligand activity, and chemokine signaling by day 3 postinfection. However, by day 14 postinfection, the enrichment switched to angiogenesis, positive regulation of cell motility and migration, skeletal system development, and cytokine-cytokine receptor interaction. Furthermore, protein-protein interaction network analysis identified 4 cytokines (interleukin 6 [IL-6], Cxcl10, gamma interferon [IFN-γ], and Cxcl9) associated with IAOM at an early stage of infection. Overall, as the pathological changes in this mouse model were consistent with those in human IAOM, our model may be used to investigate the mechanism and treatment of IAOM. Furthermore, the data for transcriptome sequencing and bioinformatic analysis will be an important resource for dissecting the molecular pathogenesis of bone with IAOM.


Asunto(s)
Osteomielitis/etiología , Infecciones Relacionadas con Prótesis/genética , Infecciones Relacionadas con Prótesis/microbiología , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus , Transcriptoma , Animales , Huesos/metabolismo , Huesos/patología , Biología Computacional/métodos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Ratones
11.
Artículo en Inglés | MEDLINE | ID: mdl-32793507

RESUMEN

Effective management of infectious osteomyelitis relies on timely microorganism identification and appropriate antibiotic therapy. Extracellular vesicles (EVs) carry protein and genetic information accumulated rapidly in the circulation upon infection. Rat osteomyelitis models infected by Staphylococcus aureus, Staphylococcus epidermidis, Pseudomonas aeruginosa, and Escherichia coli were established for the present study. Serum EVs were isolated 3 days after infection. The size and number of serum EVs from infected rats were significantly higher than those from controls. In addition, bacterial aggregation assay showed that the S. aureus and E. coli formed large aggregates in response to the stimulation of serum EVs from S. aureus-infected and E. coli-infected rats, respectively. Treatment of EVs-S. epidermidis led to large aggregates of S. epidermidis and E. coli, whereas stimulation of EVs-P. aeruginosa to large aggregates of S. aureus and P. aeruginosa. To evaluate the changes in EVs in osteomyelitis patients, 28 patients including 5 S. aureus ones and 21 controls were enrolled. Results showed that the size and number of serum EVs from S. aureus osteomyelitis patients were higher than those from controls. Further analysis using receiver operating characteristic curves revealed that only the particle size might be a potential diagnostic marker for osteomyelitis. Strikingly, serum EVs from S. aureus osteomyelitis patients induced significantly stronger aggregation of S. aureus and a cross-reaction with P. aeruginosa. Together, these findings indicate that the size and number of serum EVs may help in the diagnosis of potential infection and that EVs-bacteria aggregation assay may be a quick test to identify infectious microorganisms for osteomyelitis patients.


Asunto(s)
Vesículas Extracelulares , Osteomielitis , Infecciones Estafilocócicas , Animales , Biomarcadores , Escherichia coli , Humanos , Osteomielitis/diagnóstico , Ratas , Infecciones Estafilocócicas/diagnóstico , Staphylococcus aureus
12.
PeerJ ; 8: e9484, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32742785

RESUMEN

BACKGROUND: Bone marrow adipocyte (BMA), closely associated with bone degeneration, shares common progenitors with osteoblastic lineage. However, the intrinsic mechanism of cells fate commitment between BMA and osteogenic lineage remains unclear. METHODS: Gene Expression Omnibus (GEO) dataset GSE107789 publicly available was downloaded and analyzed. Differentially expressed genes (DEGs) were analyzed using GEO2R. Functional and pathway enrichment analyses of Gene Ontology and Kyoto Encyclopedia of Genes and Genomes were conducted by The Database for Annotation, Visualization and Integrated Discovery and Gene set enrichment analysis software. Protein-protein interactions (PPI) network was obtained using STRING database, visualized and clustered by Cytoscape software. Transcriptional levels of key genes were verified by real-time quantitative PCR in vitro in Bone marrow stromal cells (BMSCs) undergoing adipogenic differentiation at day 7 and in vivo in ovariectomized mice model. RESULTS: A total of 2,869 DEGs, including 1,357 up-regulated and 1,512 down-regulated ones, were screened out from transcriptional profile of human BMSCs undergoing adipogenic induction at day 7 vs. day 0. Functional and pathway enrichment analysis, combined with modules analysis of PPI network, highlighted ACSL1, sphingosine 1-phosphate receptors 3 (S1PR3), ZBTB16 and glypican 3 as key genes up-regulated at the early stage of BMSCs adipogenic differentiation. Furthermore, up-regulated mRNA expression levels of ACSL1, S1PR3 and ZBTB16 were confirmed both in vitro and in vivo. CONCLUSION: ACSL1, S1PR3 and ZBTB16 may play crucial roles in early regulation of BMSCs adipogenic differentiation.

13.
Food Chem Toxicol ; 145: 111689, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32810588

RESUMEN

α-hemolysin (Hla) is considered an essential virulent factor for Staphylococcus aureus (S. aureus) toxicity, the mechanism by which Hla affect bone metabolism is poorly understood. In this study, 2-month-old C57BL/6 mice were treated with Hla (40 µg/kg, i.p.) or S. aureus (1 × 106 CFU/ml, 100 µl, i.v.) with the presence or absence of methyl-ß-cyclodextrin (MßCD) (300 mg/kg, i.p.). MicroCT analysis showed progressive bone loss from week 2 to week 4 after Hla treatment, accompanied by a decreased osteoblasts and increased osteoclasts in femoral metaphysis in mice. Further, Hla stimulated the expression of Caveolin-1 in vivo and in vitro, activated lipid rafts accumulation in cell membrane of bone marrow stromal cells (BMSCs), and suppressed osteogenesis of BMSCs. Destruction of lipid rafts with MßCD or inhibition of Caveolin-1 with Daidzein blocked the detrimental effect of Hla on osteogenesis of BMSCs. Importantly, treating mice with MßCD rescued the loss of osteoblasts and increased osteoclastogenesis induced by Hla as well as the bone loss induced by S. aureus infection. Together, we demonstrate that Hla induces bone destruction directly by suppressing osteogenesis and indirectly by stimulating osteoclastogenesis, and that lipid rafts may mediate the detrimental effect of Hla and S. aureus on osteogenesis and bone formation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células de la Médula Ósea/citología , Proteínas Hemolisinas/metabolismo , Microdominios de Membrana/metabolismo , Osteogénesis , Infecciones Estafilocócicas/fisiopatología , Staphylococcus aureus/metabolismo , Animales , Proteínas Bacterianas/genética , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/microbiología , Regulación hacia Abajo , Proteínas Hemolisinas/genética , Interacciones Huésped-Patógeno , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoclastos/citología , Osteoclastos/metabolismo , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética
14.
Stem Cell Res Ther ; 11(1): 270, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32631432

RESUMEN

BACKGROUND: Prenatal dexamethasone exposure (PDE) induces low birth weight and retardation of fetal bone development which are associated with lower peak bone mass in adult offspring. Here we evaluated whether and how PDE affects postnatal long bone growth in mouse offspring. METHODS: Pregnant mice were injected subcutaneously with dexamethasone (1.2 mg/kg/day) every morning from gestational days (GD) 12-14. Femurs and tibias of 2-, 4-, 6-, and 12-week-old female offspring were harvested for histological, immunofluorescence, flow cytometric analysis, or microcomputed tomography (µCT) measurement. RESULTS: PDE leads to impaired bone remodeling as well as decreased bone mass in the long bone of female mouse offspring. During postnatal bone growth, significant decrease of CD45-CD29+CD105+Sca-1+ bone marrow mesenchymal stem cells (BMSCs) and CD45-Nestin+ cells, loss of type H vessels, and increment of cellular senescence were found in metaphysis of long bone in mouse offspring after PDE. We further show that eliminating the excessive senescent cells with dasatinib (5 mg/kg/day) and quercetin (50 mg/kg/day) during GD 12-14 rescues the above toxic effect of PDE on the postnatal long bone growth in female mouse offspring. CONCLUSION: Cellular senescence mediates the toxic effect of PDE on postnatal long bone growth in mouse offspring, and inhibition of cellular senescence may be proposed for treating the retardation of bone growth caused by PDE.


Asunto(s)
Dexametasona , Efectos Tardíos de la Exposición Prenatal , Animales , Desarrollo Óseo , Senescencia Celular , Dexametasona/toxicidad , Femenino , Ratones , Embarazo , Ratas , Ratas Wistar , Microtomografía por Rayos X
15.
Cell Microbiol ; 22(10): e13240, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32584493

RESUMEN

Internalisation of Staphylococcus aureus in osteoblasts plays a critical role in the persistence and recurrence of osteomyelitis, the mechanisms involved in this process remain largely unknown. In the present study, evidence of internalised S. aureus in osteoblasts was found in long bone of haematogenous osteomyelitis in mice after 2 weeks of infection. Meanwhile, eliminating extracellular S. aureus by gentamicin can partially rescue bone loss, whereas the remaining intracellular S. aureus in osteoblasts may be associated with continuous bone destruction. In osteoblastic MC3T3 cells, intracellular S. aureus was detectable as early as 15 min after infection, and the internalisation rates increased with the extension of infection time. Additionally, S. aureus invasion stimulated the expression of phosphor-focal adhesion kinase (FAK), phosphor-epidermal growth factor receptor (EGFR) and phosphor-c-Src in a time-dependent way, and blocking EGFR/FAK or c-Src signalling significantly reduced the internalisation rate of S. aureus in osteoblasts. Our findings provide new insights into the mechanism of S. aureus internalisation in osteoblast and raise the potential of targeting EGFR/FAK and c-Src as adjunctive therapeutics for treating chronic S. aureus osteomyelitis.


Asunto(s)
Receptores ErbB/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Osteoblastos/microbiología , Osteomielitis/microbiología , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Animales , Línea Celular , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Staphylococcus aureus/metabolismo
16.
J Orthop Translat ; 24: 12-22, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32518750

RESUMEN

BACKGROUND: Abnormal bone formation in subchondral bone resulting from uncoupled bone remodeling is considered a central feature in osteoarthritis (OA) pathogenesis. H-type vessels can couple angiogenesis and osteogenesis. We previously revealed that elevated H-type vessels in subchondral bone were correlated with OA and focal adhesion kinase (FAK) in MSCs is critical for H-type vessel formation in osteoporosis. The aim of this study was to explore the correlation between H-type vessels and MSCs in OA pathogenesis through regulation of H-type vessel formation using defactinib (an FAK inhibitor). METHODS: In vivo: 3-month-old male C57BL/6J (WT) mice were randomly divided into three groups: sham controls, vehicle-treated ACLT mice, and defactinib-treated ACLT mice (25 mg/kg, intraperitoneally weekly). In vitro: we explored the role of conditioned medium (CM) of MSCs from subchondral bone of different groups on the angiogenesis of endothelial cells (ECs). Flow cytometry, Western blotting, ELISA, real time (RT)-PCR, immunostaining, CT-based microangiography, and bone micro-CT (µCT) were used to detect changes in relative cells and tissues. RESULTS: This study demonstrated that inhibition of H-type vessels with defactinib alleviated OA by inhibiting H-type vessel-linked MSCs in subchondral bone. During OA pathogenesis, H-type vessels and MSCs formed a positive feedback loop contributing to abnormal bone formation in subchondral bone. Elevated H-type vessels provided indispensable MSCs for abnormal bone formation in subchondral bone. Flow cytometry and immunostaining results confirmed that the amount of MSCs in subchondral bone was obviously higher in vehicle-treated ACLT mice than that in sham controls and defactinib-treated ACLT mice. In vitro, p-FAK in MSCs from subchondral bone of vehicle-treated ALCT mice increased significantly relative to other groups. Further, the CM from MSCs of vehicle-treated ACLT mice enhanced angiogenesis of ECs through FAK-Grb2-MAPK-linked VEGF expression. CONCLUSIONS: Our results demonstrate that defactinib inhibits OA by suppressing the positive feedback loop between H-type vessels and MSCs in subchondral bone. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: Our results provide a mechanistic rationale for the use of defactinib as an effective candidate for OA treatment.

17.
Front Microbiol ; 11: 1301, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32595631

RESUMEN

Staphylococcus aureus (S. aureus) infection-induced osteomyelitis is a great challenge in clinic treatment. Identification of the essential genes and biological processes that are specifically changed in mononuclear cells at an early stage of S. aureus osteomyelitis is of great clinical significance. Based on transcriptional dataset GSE16129 available publicly, a bioinformatic analysis was performed to identify the differentially expressed genes of osteomyelitis caused by S. aureus infection. ERBB2, TWIST1, and NANOG were screened out as the most valuable osteomyelitis-related genes (OMRGs). A mice model of implant-associated S. aureus osteomyelitis was used to verify the above genes. We found significantly up-regulated expression of TWIST1 in macrophages and accumulation of macrophages around the infected implant. Meanwhile, S. aureus infection increased the expression of TWIST1, MMP9, and MMP13, and stimulated the migration and phagocytosis function of Raw 264.7 cells. Additionally, knock-down of the expression of TWIST1 by siRNA could significantly down-regulate MMP9 and MMP13 and suppress the migration and phagocytosis ability of macrophages in response to S. aureus infection. Furthermore, we found that NF-κB signaling was activated in Raw 264.7 cells by S. aureus and that inhibition of NF-κB signaling by Bay11-7082 blocked the expression of TWIST1, MMP9, and MMP13 as well as cell migration and phagocytosis evoked by S. aureus. Our findings demonstrate that NF-κB/TWIST1 is necessary for migration and phagocytosis of macrophages in response to S. aureus infection. Our study highlights the essential role of NF-κB/TWIST1 in early innate immune response to S. aureus infection in bone.

18.
J Biomater Appl ; 35(1): 97-107, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32233720

RESUMEN

Fabrication of osteoconductive scaffold with osteoinductive capability and appropriate resorption rate is of great significance for treating bone defects. To achieve this aim, strontium-substituted calcium sulfate hemihydrate (Sr-CSH) and hydroxyapatite (HA) were mixed to develop a novel composite. Sr-CSH containing 5% and 10% strontium was mixed with HA at the weight ratio of 6:4, respectively. Female Sprague-Dawley rats underwent bone defect surgery in left tibia were randomly assigned to three different treatment groups filled with CSH/HA, 5% and 10% Sr-CSH/HA. Micro-CT analysis showed increased new bone formation in 10% Sr-CSH/HA group compared to CSH/HA group. In addition, histological analysis showed large amounts of chondrocytes and osteoblasts within the pores of Sr-CSH/HA composites as a result of the CSH resorption. Further, CFU-F assay demonstrated the increased amount of bone marrow mesenchymal stromal cells (BMSCs) colonies in 10% Sr-CSH/HA group. In primary BMSCs, extraction from Sr-CSH/HA composite significantly increased the migration of cells, up-regulated the expression of osteoblastic marker genes, and increased the area of mineralized nodules. Together, Sr-CSH/HA may promote bone formation by recruiting and stimulating osteogenic differentiation of BMSCs. Therefore, this composite may be proposed as an ideal substitute to repair bone defects.


Asunto(s)
Regeneración Ósea , Sulfato de Calcio/química , Hidroxiapatitas/química , Células Madre Mesenquimatosas/citología , Estroncio/química , Andamios del Tejido/química , Animales , Regeneración Ósea/efectos de los fármacos , Sulfato de Calcio/farmacología , Proliferación Celular/efectos de los fármacos , Femenino , Hidroxiapatitas/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Ratas Sprague-Dawley , Estroncio/farmacología
19.
J Cell Physiol ; 235(11): 8653-8666, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32324278

RESUMEN

Osteoarthritis (OA), a disease of the entire joint, is characterized by abnormal bone remodeling and coalescent degradation of articular cartilage. We have previously found that elevated levels of H-type vessels in subchondral bone correlate with OA and that focal adhesion kinase (FAK) is critical for H-type vessel formation in osteoporosis. However, the potential role of FAK in OA remains unexplored. Here, we demonstrate that the p-FAK level was dramatically elevated in subchondral bone following anterior cruciate ligament transection (ACLT) in rats. Specific inhibition of FAK signaling with Y15 in subchondral bone resulted in the suppression of subchondral bone deterioration and this effect was mediated by H-type vessel-induced ectopic bone formation. Further, articular cartilage degeneration was also alleviated after Y15 treatment. In vitro, the p-FAK level was significantly elevated in mesenchymal stem cells (MSCs) from vehicle-treated ACLT rats as compared to that in MSCs from sham controls and Y15-treated ACLT rats. Elevated p-FAK level in MSCs promoted vascular endothelial growth factor (VEGF) expression, as demonstrated from the high VEGF level in the blood, subchondral bone, and conditioned medium (CM) of MSCs from vehicle-treated ACLT rats. The CM of MSCs from vehicle-treated ACLT rats might promote the angiogenesis of endothelial cells and the catabolic response of chondrocytes through the FAK-growth factor receptor-bound protein 2-mitogen-activated protein kinase-mediated expression of VEGF. The effect of the CM from MSCs of Y15-treated ACLT rats or that treated with a VEGF-neutralizing antibody on vessel formation and the catabolic response was lowered. Thus, the specific inhibition of FAK signaling may be a promising avenue for the prevention or early treatment of OA.


Asunto(s)
Cartílago Articular/metabolismo , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Osteoartritis/tratamiento farmacológico , Alendronato/farmacología , Animales , Ligamento Cruzado Anterior/patología , Remodelación Ósea/efectos de los fármacos , Remodelación Ósea/fisiología , Huesos/patología , Condrocitos/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Masculino , Osteoartritis/patología , Ratas Sprague-Dawley
20.
Stem Cell Res Ther ; 11(1): 131, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32197645

RESUMEN

BACKGROUND: Hypercholesterolemia increases the risk of tendon pain and tendon rupture. Tendon-derived stem cells (TDSCs) play a vital role in the development of tendinopathy. Our previous research found that high cholesterol inhibits tendon-related gene expression in TDSCs. Whether high cholesterol has other biological effects on TDSCs remains unknown. METHODS: TDSCs isolated from female SD rats were exposed to 10 mg/dL cholesterol for 24 h. Then, cell apoptosis was assessed using flow cytometry and fluorescence microscope. RFP-GFP-LC3 adenovirus transfection was used for measuring autophagy. Signaling transduction was measured by immunofluorescence and immunoblotting. In addition, Achilles tendons from ApoE -/- mice fed with a high-fat diet were histologically assessed using HE staining and immunohistochemistry. RESULTS: In this work, we verified that 10 mg/dL cholesterol suppressed cell proliferation and migration and induced G0/G1 phase arrest. Additionally, cholesterol induced apoptosis and autophagy simultaneously in TDSCs. Apoptosis induction was related to increased expression of cleaved caspase-3 and BAX and decreased expression of Bcl-xL. The occurrence of autophagic flux and accumulation of LC3-II demonstrated the induction of autophagy by cholesterol. Compared with the effects of cholesterol treatment alone, the autophagy inhibitor 3-methyladenine (3-MA) enhanced apoptosis, while the apoptosis inhibitor Z-VAD-FMK diminished cholesterol-induced autophagy. Moreover, cholesterol triggered reactive oxygen species (ROS) generation and activated the AKT/FOXO1 pathway, while the ROS scavenger NAC blocked cholesterol-induced activation of the AKT/FOXO1 pathway. NAC and the FOXO1 inhibitor AS1842856 rescued the apoptosis and autophagy induced by cholesterol. Finally, high cholesterol elevated the expression of cleaved caspase-3, Bax, LC3-II, and FOXO1 in vivo. CONCLUSION: The present study indicated that high cholesterol induced apoptosis and autophagy through ROS-activated AKT/FOXO1 signaling in TDSCs, providing new insights into the mechanism of hypercholesterolemia-induced tendinopathy. High cholesterol induces apoptosis and autophagy through the ROS-activated AKT/FOXO1 pathway in tendon-derived stem cells.


Asunto(s)
Hipercolesterolemia , Proteínas Proto-Oncogénicas c-akt , Animales , Apoptosis , Autofagia , Línea Celular Tumoral , Colesterol , Femenino , Proteína Forkhead Box O1 , Ratones , Proteínas del Tejido Nervioso , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Células Madre , Tendones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...