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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
FASEB J ; 33(10): 11137-11147, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31298955

RESUMEN

So far, there has been no effective cure for osteoporotic cortical bone, the most significant change in long bone structure during aging and the main cause of bone fragility fractures, because its underlying molecular and cellular mechanisms remain largely unknown. We used 3- and 15-mo-old mice as well as 15-mo-old mice treated with vehicle and gefitinib to evaluate structural, cellular, and molecular changes in cortical bone. We found that the senescence of osteoprogenitors was increased, whereas the expression of phosphorylated epidermal growth factor receptor (EGFR) on the endosteal surface of cortical bone down-regulated in middle-aged 15-mo-old mice compared with young 3-mo-old mice. Further decreasing EGFR signaling by gefitinib treatment in middle-aged mice resulted in promoted senescence of osteoprogenitors and accelerated cortical bone degeneration. Moreover, inhibiting EGFR signaling suppressed the expression of enhancer of zeste homolog 2 (Ezh2), the repressor of cell senescence-inducer genes, through ERK1/2 pathway, thereby promoting senescence in osteoprogenitors. Down-regulated EGFR signaling plays a physiologically significant role during aging by reducing Ezh2 expression, leading to the senescence of osteoprogenitors and the decline in bone formation on the endosteal surface of cortical bone.-Liu, G., Xie, Y., Su, J., Qin, H., Wu, H., Li, K., Yu, B., Zhang, X. The role of EGFR signaling in age-related osteoporosis in mouse cortical bone.


Asunto(s)
Hueso Cortical/metabolismo , Receptores ErbB/metabolismo , Osteoporosis/metabolismo , Transducción de Señal/fisiología , Envejecimiento/metabolismo , Animales , Senescencia Celular/fisiología , Regulación hacia Abajo/fisiología , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteogénesis/fisiología
8.
J Cell Physiol ; 234(10): 17946-17958, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30834523

RESUMEN

To identify the effects of running on articular cartilage and subchondral bone remodeling, C57BL/6 mice were randomly divided into three groups: control, moderate-, and strenuous running. Magnetic resonance imaging showed bone marrow lesions in the knee subchondral bone in the strenuous-running group in contrast with the other two groups. The microcomputed tomography analysis showed promoted bone formation in the subchondral bone in mice subjected to strenuous running. Histological and immunohistochemistry results indicated that terminal differentiation of chondrocytes and degeneration of articular cartilage were enhanced but, synthesis of platelet-derived growth factor-AA (PDGF-AA) in the subchondral bone was suppressed after strenuous running. In vitro, excessive mechanical treatments suppressed the expression of PDGF-AA in osteoblasts, and the condition medium from mechanical-treated osteoblasts stimulated maturation and terminal differentiation of chondrocytes. These results indicate that strenuous running suppresses the synthesis of PDGF-AA in subchondral bone, leading to downregulated PDGF/Akt signal in articular cartilage and thus cartilage degeneration.


Asunto(s)
Cartílago Articular/metabolismo , Condrocitos/metabolismo , Fémur/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Esfuerzo Físico , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Carrera , Tibia/metabolismo , Animales , Cartílago Articular/patología , Diferenciación Celular , Células Cultivadas , Condrocitos/patología , Regulación hacia Abajo , Femenino , Fémur/diagnóstico por imagen , Ratones Endogámicos C57BL , Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Tibia/diagnóstico por imagen
9.
J Cell Physiol ; 234(10): 18017-18028, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30825206

RESUMEN

Clinical studies have indicated that increased serum cholesterol levels raised the risk of tendinopathy in hypercholesterolemia, but the effect of cholesterol on tendon-derived stem cells (TDSCs) and its underlying mechanism have not been studied. The purpose of this study is to investigate the association between cholesterol and tendinopathy in vitro and in vivo, and its underlying molecular mechanism as well. In TDSCs, the effect of cholesterol was assessed by quantitative polymerase chain reaction, western blot analysis, and immunofluorescence staining. Intracellular levels of reactive oxygen species (ROS) was detected, using flow cytometry. The link between nuclear factor (NF)-κB signaling and the effect of cholesterol was evaluated using a representative IκB kinase (IKK) inhibitor, BAY 11-7082. In addition, Achilles tendons from apolipoprotein E mice fed with a high-fat diet were histologically assessed using hematoxylin and eosin staining and immunohistochemistry. We found that high cholesterol apparently lowered the expression of tendon cell markers (collagen 1, scleraxis, tenomodulin), and elevated ROS levels via the NF-κB pathway both in vitro and in vivo. The ROS scavenger N-acetylcysteine (NAC) and BAY 11-7082 reversed the inhibiting effect of cholesterol on the tendon-related gene expressions of TDSCs. Moreover, NAC blocked cholesterol-induced phosphorylation of IκBα and p65. Significant histological alternation in vivo was shown in Achilles tendon in the hypercholesterolemic group. These results indicated that high cholesterol may inhibit the tendon-related gene expressions in TDSCs via ROS-activated NF-кB signaling, implying pathogenesis of tendinopathy in hypercholesterolemia and suggesting a new mechanism underlying hypercholesterolemia-induced tendinopathy.


Asunto(s)
Tendón Calcáneo/metabolismo , Colesterol/metabolismo , Hipercolesterolemia/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Células Madre/metabolismo , Tendinopatía/metabolismo , Tendón Calcáneo/efectos de los fármacos , Tendón Calcáneo/patología , Animales , Antioxidantes/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Masculino , Ratones Noqueados para ApoE , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal , Células Madre/efectos de los fármacos , Células Madre/patología , Tendinopatía/genética , Tendinopatía/patología , Tendinopatía/prevención & control
10.
Clin Sci (Lond) ; 133(12): 1297-1308, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31175224

RESUMEN

Bone loss in Staphylococcus aureus (S. aureus) osteomyelitis poses a serious challenge to orthopedic treatment. The present study aimed to elucidate how S. aureus infection in bone might induce bone loss. The C57BL/6 mice were injected with S. aureus (106 CFU/ml, 100 µl) or with the same amount of vehicle (control) via the tail vein. Microcomputed tomography (microCT) analysis showed bone loss progressing from week 1 to week 5 after infection, accompanied by a decreased number of osteocalcin-positive stained osteoblasts and the suppressed mRNA expression of Runx2 and osteocalcin. Transcriptome profiles of GSE30119 were downloaded and analyzed to determine the differences in expression of inflammatory factors between patients with S. aureus infected osteomyelitis and healthy controls, the data showed significantly higher mRNA expression of granulocyte colony-stimulating factor (G-CSF) in the whole blood from patients with S. aureus infection. Enzyme-linked immunosorbent assay (ELISA) analysis confirmed an increased level of G-CSF in the bone marrow and serum from S. aureus infected mice, which might have been due to the increased amount of F4/80+ macrophages. Interestingly, G-CSF neutralizing antibody treatment significantly rescued the bone loss after S. aureus infection, as evidenced by its roles in improving BV/TV and preserving osteocalcin- and osterix-positive stained cells. Importantly, we found that G-CSF level was significantly up-regulated in the serum from osteomyelitis patients infected by S. aureus Together, S. aureus infection might suppress the function of osteoblastic cells and induce progressive bone loss by up-regulating the level G-CSF, suggesting a therapeutic potential for G-CSF neutralization in combating bone loss in S. aureus osteomyelitis.


Asunto(s)
Remodelación Ósea , Factor Estimulante de Colonias de Granulocitos/metabolismo , Osteoblastos/metabolismo , Osteomielitis/metabolismo , Infecciones Estafilocócicas/metabolismo , Tibia/metabolismo , Adulto , Animales , Anticuerpos Neutralizantes/farmacología , Remodelación Ósea/efectos de los fármacos , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos/inmunología , Humanos , Masculino , Ratones Endogámicos C57BL , Osteoblastos/efectos de los fármacos , Osteoblastos/microbiología , Osteomielitis/diagnóstico por imagen , Osteomielitis/tratamiento farmacológico , Osteomielitis/microbiología , Transducción de Señal , Infecciones Estafilocócicas/diagnóstico por imagen , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Tibia/diagnóstico por imagen , Tibia/efectos de los fármacos , Tibia/microbiología , Factores de Tiempo , Microtomografía por Rayos X
11.
Proc Natl Acad Sci U S A ; 113(50): 14360-14365, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27911782

RESUMEN

Osteoarthritis (OA) is the most common joint disease, characterized by progressive destruction of the articular cartilage. The surface of joint cartilage is the first defensive and affected site of OA, but our knowledge of genesis and homeostasis of this superficial zone is scarce. EGFR signaling is important for tissue homeostasis. Immunostaining revealed that its activity is mostly dominant in the superficial layer of healthy cartilage but greatly diminished when OA initiates. To evaluate the role of EGFR signaling in the articular cartilage, we studied a cartilage-specific Egfr-deficient (CKO) mouse model (Col2-Cre EgfrWa5/flox). These mice developed early cartilage degeneration at 6 mo of age. By 2 mo of age, although their gross cartilage morphology appears normal, CKO mice had a drastically reduced number of superficial chondrocytes and decreased lubricant secretion at the surface. Using superficial chondrocyte and cartilage explant cultures, we demonstrated that EGFR signaling is critical for maintaining the number and properties of superficial chondrocytes, promoting chondrogenic proteoglycan 4 (Prg4) expression, and stimulating the lubrication function of the cartilage surface. In addition, EGFR deficiency greatly disorganized collagen fibrils in articular cartilage and strikingly reduced cartilage surface modulus. After surgical induction of OA at 3 mo of age, CKO mice quickly developed the most severe OA phenotype, including a complete loss of cartilage, extremely high surface modulus, subchondral bone plate thickening, and elevated joint pain. Taken together, our studies establish EGFR signaling as an important regulator of the superficial layer during articular cartilage development and OA initiation.


Asunto(s)
Artritis Experimental/metabolismo , Cartílago Articular/metabolismo , Receptores ErbB/metabolismo , Osteoartritis/metabolismo , Animales , Artritis Experimental/patología , Artritis Experimental/prevención & control , Células Cultivadas , Condrocitos/metabolismo , Condrocitos/patología , Condrogénesis , Receptores ErbB/deficiencia , Receptores ErbB/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Osteoartritis/patología , Osteoartritis/prevención & control , Proteoglicanos/metabolismo , Transducción de Señal
12.
Biochem Biophys Res Commun ; 506(1): 194-203, 2018 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-30342851

RESUMEN

Osteoporosis is widely viewed as a major public health concern, but the exact magnitude of the problem is uncertain. MicroRNAs play a key role in maintaining bone development and metabolism. This study aims to investigate the effects that microRNA-874 (miR-874) has on osteoblast proliferation and differentiation in osteoporosis rats by targeting SUFU through the Hedgehog signaling pathway. Twenty Wistar female rats were selected for following experiment, and another 20 rats were served as the normal group. Their osteogenic tissues were obtained and the positive expression of SUFU in tissues was determined. Rat osteoblasts were isolated and. The targeting relationship between SUFU and miR-874 was verified and the expression of miR-874, SUFU, Sonic Hedgehog (Shh), Ptch, Smoothened (Smo), bone morphogenetic protein (BMP2), Runx2, proliferating cell nuclear antigen (PCNA) and Bcl-2 associated X protein (Bax) were identified. Besides, cell viability apoptosis, and differentiation were confirmed respectively. Moreover, calcium nodules were observed. Overexpression of SUFU and Bax but lower expression of miR-874, Shh, Ptch, Smo, BMP2, Runx2, and PCNA were found in osteoporosis mice. Besides, elevated expression of miR-874, Shh, Ptch, Smo, BMP2, Runx2 and PCNA, as well as increased cell viability, ALP activity and calcium nodules but decreased expression of SUFU and Bax, and reduced cell apoptosis were confirmed when treated with miR-874 mimic. And it is reciprocal when miR-874 was inhibited. Our study demonstrated that through targeted inhibition of SUFU and activation of Hedgehog signaling pathway, miR-874 could promote the proliferation and differentiation of osteoblasts in osteoporosis rats.


Asunto(s)
Proteínas Hedgehog/metabolismo , MicroARNs/fisiología , Osteoblastos/citología , Osteoporosis/patología , Proteínas Represoras/antagonistas & inhibidores , Animales , Apoptosis , Diferenciación Celular , Proliferación Celular , Femenino , Osteoporosis/metabolismo , Ratas , Ratas Wistar , Proteínas Represoras/análisis , Transducción de Señal
13.
Toxicol Appl Pharmacol ; 351: 12-20, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29753006

RESUMEN

Dexamethasone is routinely used for treating those mothers at risk for preterm delivery. However, overexposure to exogenous glucocorticoids induces bone loss in offspring, and the "critical window" and safe dose of this treatment are largely unknown. In this study, we found that femoral length, and the length of the primary ossification center were significantly reduced in fetal mice after repeated prenatal dexamethasone exposure (PDE). Compared with single-course exposure on gestational day (GD)15, newborn mice with repeated PDE (3 times, from GD15 to 17) showed a significant decrease in femoral trabecular bone mass with decreased trabecular number and thickness. For those newborn mice treated after repeated PDE at different doses (0, 0.2, 0.8, and 1.2 mg/kg/d), the toxic effect of dexamethasone on bone development was observed at 0.8 and 1.2 mg/kg/d. More severe retardation in bone development was observed in the fetal mice after PDE at 0.8 mg/kg/d during GD12-14, compared with that during GD15-17. Interestingly, stronger toxic effects were observed in male newborn mice after PDE than were observed in female newborn mice. In conclusion, PDE with multiple course, higher dose, or exposure at an early stage of pregnancy have stronger toxic effects on bone development of fetal mice.


Asunto(s)
Desarrollo Óseo/efectos de los fármacos , Dexametasona/toxicidad , Fémur/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Glucocorticoides/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Desarrollo Óseo/fisiología , Dexametasona/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Fémur/embriología , Fémur/patología , Desarrollo Fetal/fisiología , Glucocorticoides/administración & dosificación , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología
15.
FASEB J ; 30(2): 785-97, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26499267

RESUMEN

This study aimed to evaluate whether female adult offspring born with intrauterine growth retardation induced by prenatal nicotine exposure (PNE) are susceptible to osteoarthritis (OA) and to explore the underlying programming mechanisms. Pregnant rats were treated with nicotine or saline at 2.0 mg/kg/d from gestational d 11 to 20. The female adult offspring with or without PNE were forced with a strenuous treadmill running for 6 wk to induce OA. Nicotine's effects on fetal articular chondrocytes were studied by exposing chondrocytes to nicotine for 10 d, and dihydro-ß-erythroidine, a selective α4ß2-nicotinic acetylcholine receptor (nAChR) inhibitor, was used to identify the change of nicotine's effect. For adult offspring, increased cartilage destruction and accelerated OA progression were observed in the PNE group with running; the expression of α1 chain of type II collagen (Col2A1), aggrecan, SRY-type high mobility group box 9 (Sox9), and IGF1 signaling molecules in the cartilage of PNE offspring were decreased. For fetuses, elevated serum corticosteroid and nicotine levels and suppressed IGF1 levels were observed; expression of Col2A1, aggrecan, Sox9, and IGF1 were reduced. The result of chondrocytes revealed that nicotine impeded the expression of Col2A1, aggrecan, and IGF1; blocking α4ß2-nAChR rescued nicotine's suppression. In conclusion, PNE increases the susceptibility of adult offspring to OA; the potential mechanism involves IGF1 low-functional programming in articular cartilage caused directly by the action of nicotine on α4ß2-nAChR.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Exposición Materna/efectos adversos , Nicotina/efectos adversos , Osteoartritis/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Cartílago Articular/metabolismo , Cartílago Articular/patología , Susceptibilidad a Enfermedades , Femenino , Factor I del Crecimiento Similar a la Insulina/genética , Nicotina/farmacología , Osteoartritis/inducido químicamente , Osteoartritis/genética , Osteoartritis/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Wistar
16.
Toxicol Appl Pharmacol ; 305: 234-241, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27338645

RESUMEN

Prenatal ethanol exposure (PEE) inhibits longitudinal growth of fetal bones, but the underlying mechanisms remain unknown. In this study, we aimed to investigate how PEE induces the retardation of long bone development in fetal rats. Pregnant Wistar rats were treated with ethanol or distilled water (control group) by gavage from gestational day (GD) 9 to 20. Fetuses were delivered by cesarean section on GD20. Fetal sera were collected for assessing corticosterone (CORT) level. Fetal long bones were harvested for histochemical, immunohistochemical and gene expression analysis. Primary chondrocytes were treated with ethanol or CORT for analyzing genes expression. PEE fetuses showed a significant reduction in birth weight and body length. The serum CORT concentration in PEE group was significantly increased, while the body weight, body length and femur length all were significantly decreased in the PEE group. The length of the epiphyseal hypertrophy zone was enlarged, whereas the length of the primary ossification center was significantly reduced in PEE fetuses. TUNEL assay showed reduced apoptosis in the PEE group. Further, the gene expression of osteoprotegerin (OPG) was markedly up-regulated. In vitro experiments showed that CORT (but not ethanol) treatment significantly activated the expression of OPG, while the application of glucocorticoid receptor inhibitor, mifepristone, attenuated these change induced by CORT. These results indicated that PEE-induced glucocorticoid over-exposure enhanced the expression of OPG in fetal epiphyseal cartilage and further lead to the suppressed osteoclast differentiation in the chondro-osseous junction and consequently inhibited the endochondral ossification in long bones of fetal rats.


Asunto(s)
Etanol/toxicidad , Fémur/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/fisiología , Femenino , Fémur/crecimiento & desarrollo , Feto , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Embarazo , Ratas Wistar
17.
J Biol Chem ; 288(45): 32229-32240, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24047892

RESUMEN

The epidermal growth factor receptor (EGFR) is an essential player in the development of multiple organs during embryonic and postnatal stages. To understand its role in epiphyseal cartilage development, we generated transgenic mice with conditionally inactivated EGFR in chondrocytes. Postnatally, these mice exhibited a normal initiation of cartilage canals at the perichondrium, but the excavation of these canals into the cartilage was strongly suppressed, resulting in a delay in the formation of the secondary ossification center (SOC). This delay was accompanied by normal chondrocyte hypertrophy but decreased mineralization and apoptosis of hypertrophic chondrocytes and reduced osteoclast number at the border of marrow space. Immunohistochemical analyses demonstrated that inactivation of chondrocyte-specific EGFR signaling reduced the amounts of matrix metalloproteinases (MMP9, -13, and -14) and RANKL (receptor activator of NF-κB ligand) in the hypertrophic chondrocytes close to the marrow space and decreased the cartilage matrix degradation in the SOC. Analyses of EGFR downstream signaling pathways in primary epiphyseal chondrocytes revealed that up-regulation of MMP9 and RANKL by EGFR signaling was partially mediated by the canonical Wnt/ß-catenin pathway, whereas EGFR-enhanced MMP13 expression was not. Further biochemical studies suggested that EGFR signaling stimulates the phosphorylation of LRP6, increases active ß-catenin level, and induces its nuclear translocation. In line with these in vitro studies, deficiency in chondrocyte-specific EGFR activity reduced ß-catenin amount in hypertrophic chondrocytes in vivo. In conclusion, our work demonstrates that chondrocyte-specific EGFR signaling is an important regulator of cartilage matrix degradation during SOC formation and epiphyseal cartilage development and that its actions are partially mediated by activating the ß-catenin pathway.


Asunto(s)
Condrocitos/metabolismo , Receptores ErbB/metabolismo , Placa de Crecimiento/embriología , Vía de Señalización Wnt/fisiología , Animales , Colagenasas/biosíntesis , Colagenasas/genética , Receptores ErbB/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Placa de Crecimiento/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Ratones Noqueados , Fosforilación/fisiología , Ligando RANK/genética , Ligando RANK/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
18.
Am J Pathol ; 182(3): 917-27, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23274133

RESUMEN

Osteochondromas and enchondromas are the most common tumors affecting the skeleton. Osteochondromas can occur as multiple lesions, such as those in patients with hereditary multiple exostoses. Unexpectedly, while studying the role of ß-catenin in cartilage development, we found that its conditional deletion induces ectopic chondroma-like cartilage formation in mice. Postnatal ablation of ß-catenin in cartilage induced lateral outgrowth of the growth plate within 2 weeks after ablation. The chondroma-like masses were present in the flanking periosteum by 5 weeks and persisted for more than 6 months after ß-catenin ablation. These long-lasting ectopic masses rarely contained apoptotic cells. In good correlation, transplants of ß-catenin-deficient chondrocytes into athymic mice persisted for a longer period of time and resisted replacement by bone compared to control wild-type chondrocytes. In contrast, a ß-catenin signaling stimulator increased cell death in control chondrocytes. Immunohistochemical analysis revealed that the amount of detectable ß-catenin in cartilage cells of osteochondromas obtained from hereditary multiple exostoses patients was much lower than that in hypertrophic chondrocytes in normal human growth plates. The findings in our study indicate that loss of ß-catenin expression in chondrocytes induces periosteal chondroma-like masses and may be linked to, and cause, the persistence of cartilage caps in osteochondromas.


Asunto(s)
Neoplasias Óseas/patología , Condroma/patología , Periostio/patología , beta Catenina/deficiencia , Fosfatasa Ácida/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/metabolismo , Cartílago/diagnóstico por imagen , Cartílago/patología , Proliferación Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/patología , Condrocitos/trasplante , Condroma/diagnóstico por imagen , Condroma/metabolismo , Coristoma/diagnóstico por imagen , Coristoma/patología , Colágeno Tipo II/metabolismo , Placa de Crecimiento/efectos de los fármacos , Placa de Crecimiento/metabolismo , Placa de Crecimiento/patología , Humanos , Etiquetado Corte-Fin in Situ , Indoles/farmacología , Integrasas/metabolismo , Isoenzimas/metabolismo , Ratones , Osteocondroma/metabolismo , Osteocondroma/patología , Oximas/farmacología , Periostio/diagnóstico por imagen , Periostio/efectos de los fármacos , Periostio/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Radiografía , Costillas/patología , Tamoxifeno/farmacología , Fosfatasa Ácida Tartratorresistente , beta Catenina/metabolismo
19.
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)
Sistema de Transporte de Aminoácidos y+ , Macrófagos , Osteomielitis , Infecciones Estafilocócicas , Animales , Ratones , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Osteomielitis/metabolismo , Osteomielitis/microbiología , Especies Reactivas de Oxígeno/metabolismo , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA