Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Publication year range
1.
Exp Ther Med ; 22(5): 1244, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34539840

ABSTRACT

Long non-coding (lnc) RNAs have been associated with osteoarthritis (OA) progression. The aim of the present study was to investigate the regulatory mechanism of lncRNA LINC01385 in OA in vitro. The mRNA expression level of LINC01385, microRNA(miR)-140-3p, and Toll-like receptor 4 (TLR4) was detected using reverse transcription-quantitative PCR, while ELISA was used to determine the concentration of different inflammatory factors [tumor necrosis factor-α (TNF-α), IL-6, and prostaglandin E2 (PGE2)]. The viability of human articular chondrocytes (HC-a) was measured using a MTT assay and western blot analysis was performed to quantify the protein expression level of TLR4. The associations between miR-140-3p and LINC01385/TLR4 were confirmed using a dual-luciferase reporter assay. LINC01385 mRNA expression level was increased in OA tissues and IL-1ß-induced HC-a. LINC01385 knockdown and miR-140-3p mimics reduced the concentration of inflammatory factors in IL-1ß-induced HC-a and promoted cell survival. In addition, it was confirmed that LINC01385 targeted miR-140-3p, while TLR4 was a target gene of miR-140-3p. Negative correlations between LINC01385 and miR-140-3p, and between miR-140-3p and TLR4 were observed in OA tissues. Low mRNA expression level of miR-140-3p and high protein expression level of TLR4 reversed the inhibitory effect of LINC01385 knockdown on the inflammatory responses of IL-1ß-induced HC-a and exhibited a stimulating effect on cell viability. LINC01385 knockdown reduced the progression of OA by modulating the miR-140-3p/TLR4 axis in vitro; thus, LINC01385 may be a therapeutic target for OA.

2.
Pathol Res Pract ; 216(2): 152766, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31796334

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the effect of Toll like receptor 4 (TLR4) on fracture healing. METHODS: The open tibial fracture models in TLR4 knockout (TLR4-/-) and wild type (WT) C57BL-6 J mice were established. The radiological examination, tartrate-resistant acid phosphatase (TRAP) staining, Micro-CT scan and biological torsion test were performed on 7, 14 and 21 days after operation. Enzyme Linked Immunosorbent Assay (ELISA) kit was used to detect the expression levels of tumor necrosis factor-α (TNF-α), interleukin-1 beta (IL-1ß) and interleukin 6 (IL-6). Western blotting was used to detect the expression of ß-catenin, Wingless-type MMTV integration site family, member 4 and 5B (Wnt4 and Wnt5B), proliferating cell nuclear antigen (PCNA) and bone morphogenetic protein-2 (BMP-2) of the callus tissue obtained from mice. RESULTS: TLR4 knockout promoted fracture healing, reduced the number of osteoclasts, increased bone callus volume (BV) and callus mineralized volume fraction (BV/TV%) (P < 0.05), increased the maximum torque and torsional stiffness of callus (P < 0.05), reduced TNF-α, IL-1ß and IL-6 expression (P < 0.01), and increased the expression levels of ß-catenin, Wnt4, Wnt5B, PCNA and BMP-2 (P < 0.01). CONCLUSION: TLR4 knockout reduced inflammatory and promoted fracture healing by activating Wnt/ß-catenin signaling pathway.


Subject(s)
Fracture Healing/genetics , Toll-Like Receptor 4/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Bony Callus/diagnostic imaging , Disease Models, Animal , Fracture Healing/physiology , Inflammation/diagnostic imaging , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/physiology , Specific Pathogen-Free Organisms , Tibia/diagnostic imaging , Tibial Fractures/diagnostic imaging , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , X-Ray Microtomography , beta Catenin/genetics
3.
Braz J Med Biol Res ; 52(12): e8735, 2019.
Article in English | MEDLINE | ID: mdl-31826179

ABSTRACT

Exosomes, a kind of extracellular vesicle, are promising therapeutic agents for spinal cord injury (SCI). This article aimed to investigate effects of exosomes secreted from miRNA-29b-modified bone marrow mesenchymal stem cells (BMSCs) on SCI. Exosomes were extracted from BMSCs transfected with miRNA-29b or negative control (miR NC). SCI rats were injected intravenously with exosomes (control exosomes, miRNA-29b exosomes) and BMSCs (miR NC, miRNA-29b) through the tail vein. The expression of miRNA-29b in spinal cord tissues of SCI rats was detected by qRT-PCR. The hind limb motor function was evaluated by Basso Beattie Bresnahan (BBB) score. The histopathological damage and neuronal regeneration in spinal cord tissues was observed by HE staining and immunohistochemistry, respectively. The injection of miRNA-29b exosomes and miRNA-29b BMSCs both significantly increased the expression of miRNA-29b in spinal cord tissues of SCI rats (P<0.05). Compared with SCI rats, rats in the miRNA-29b exosomes and the miRNA-29b groups exhibited improved SCI, including increased BBB score, NF200 and GAP-43 positive neurons, as well as decreased contractile nerve cell numbers and GFAP positive neurons (all P<0.05). The relieving degree of SCI was significantly higher in the miRNA-29b exosomes group than in the miRNA-29b BMSCs group (P<0.05). Exosomes secreted from miRNA-29b-modified BMSCs were effective in the repair of SCI in rats.


Subject(s)
Exosomes/metabolism , Mesenchymal Stem Cell Transplantation , MicroRNAs/metabolism , Recovery of Function , Spinal Cord Injuries/therapy , Transfection , Animals , Disease Models, Animal , Female , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
4.
Braz. j. med. biol. res ; 52(12): e8735, 2019. graf
Article in English | LILACS | ID: biblio-1055473

ABSTRACT

Exosomes, a kind of extracellular vesicle, are promising therapeutic agents for spinal cord injury (SCI). This article aimed to investigate effects of exosomes secreted from miRNA-29b-modified bone marrow mesenchymal stem cells (BMSCs) on SCI. Exosomes were extracted from BMSCs transfected with miRNA-29b or negative control (miR NC). SCI rats were injected intravenously with exosomes (control exosomes, miRNA-29b exosomes) and BMSCs (miR NC, miRNA-29b) through the tail vein. The expression of miRNA-29b in spinal cord tissues of SCI rats was detected by qRT-PCR. The hind limb motor function was evaluated by Basso Beattie Bresnahan (BBB) score. The histopathological damage and neuronal regeneration in spinal cord tissues was observed by HE staining and immunohistochemistry, respectively. The injection of miRNA-29b exosomes and miRNA-29b BMSCs both significantly increased the expression of miRNA-29b in spinal cord tissues of SCI rats (P<0.05). Compared with SCI rats, rats in the miRNA-29b exosomes and the miRNA-29b groups exhibited improved SCI, including increased BBB score, NF200 and GAP-43 positive neurons, as well as decreased contractile nerve cell numbers and GFAP positive neurons (all P<0.05). The relieving degree of SCI was significantly higher in the miRNA-29b exosomes group than in the miRNA-29b BMSCs group (P<0.05). Exosomes secreted from miRNA-29b-modified BMSCs were effective in the repair of SCI in rats.


Subject(s)
Animals , Male , Female , Rats , Spinal Cord Injuries/therapy , Transfection , Recovery of Function , MicroRNAs/metabolism , Mesenchymal Stem Cell Transplantation , Exosomes/metabolism , Immunohistochemistry , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Disease Models, Animal
5.
PLoS One ; 9(1): e85472, 2014.
Article in English | MEDLINE | ID: mdl-24427311

ABSTRACT

BACKGROUND: A novel injectable cement composed of chitosan-bonded borate bioactive glass (BG) particles was evaluated as a carrier for local delivery of vancomycin in the treatment of osteomyelitis in a rabbit tibial model. MATERIALS AND METHODS: The setting time, injectability, and compressive strength of the borate BG cement, and the release profile of vancomycin from the cement were measured in vitro. The capacity of the vancomycin-loaded BG cement to eradicate methicillin-resistant Staphylococcus aureus (MRSA)-induced osteomyelitis in rabbit tibiae in vivo was evaluated and compared with that for a vancomycin-loaded calcium sulfate (CS) cement and for intravenous injection of vancomycin. RESULTS: The BG cement had an injectability of >90% during the first 3 minutes after mixing, hardened within 30 minutes and, after hardening, had a compressive strength of 18 ± 2 MPa. Vancomycin was released from the BG cement into phosphate-buffered saline for up to 36 days, and the cumulative amount of vancomycin released was 86% of the amount initially loaded into the cement. In comparison, vancomycin was released from the CS cement for up 28 days and the cumulative amount released was 89%. Two months post-surgery, radiography and microbiological tests showed that the BG and CS cements had a better ability to eradicate osteomyelitis when compared to intravenous injection of vancomycin, but there was no significant difference between the BG and CS cements in eradicating the infection. Histological examination showed that the BG cement was biocompatible and had a good capacity for regenerating bone in the tibial defects. CONCLUSIONS: These results indicate that borate BG cement is a promising material both as an injectable carrier for vancomycin in the eradication of osteomyelitis and as an osteoconductive matrix to regenerate bone after the infection is cured.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Biocompatible Materials , Borates , Drug Carriers , Glass , Osteomyelitis/drug therapy , Animals , Anti-Bacterial Agents/pharmacokinetics , Compressive Strength , Disease Models, Animal , Kinetics , Materials Testing , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Osteomyelitis/microbiology , Osteomyelitis/pathology , Rabbits , Staphylococcal Infections/drug therapy , Staphylococcal Infections/pathology , Vancomycin/administration & dosage
6.
J Mater Sci Mater Med ; 25(3): 733-45, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24477872

ABSTRACT

Osteomyelitis (bone infection) is often difficult to cure. The commonly-used treatment of surgical debridement to remove the infected bone combined with prolonged systemic and local antibiotic treatment has limitations. In the present study, an injectable borate bioactive glass cement was developed as a carrier for the antibiotic vancomycin, characterized in vitro, and evaluated for its capacity to cure osteomyelitis in a rabbit tibial model. The cement (initial setting time = 5.8 ± 0.6 min; compressive strength = 25.6 ± 0.3 MPa) released vancomycin over ~25 days in phosphate-buffered saline, during which time the borate glass converted to hydroxyapatite (HA). When implanted in rabbit tibial defects infected with methicillin-resistant Staphylococcus aureus (MRSA)-induced osteomyelitis, the vancomycin-loaded cement converted to HA and supported new bone formation in the defects within 8 weeks. Osteomyelitis was cured in 87 % of the defects implanted with the vancomycin-loaded borate glass cement, compared to 71 % for the defects implanted with vancomycin-loaded calcium sulfate cement. The injectable borate bioactive glass cement developed in this study is a promising treatment for curing osteomyelitis and for regenerating bone in the defects following cure of the infection.


Subject(s)
Bone Cements/therapeutic use , Bone Regeneration/drug effects , Drug Carriers/administration & dosage , Glass/chemistry , Osteomyelitis/therapy , Vancomycin/administration & dosage , Vancomycin/chemistry , Animals , Bone Cements/chemistry , Borates/chemistry , Compressive Strength , Drug Carriers/chemistry , Female , Injections, Intralesional , Materials Testing , Rabbits , Tibia
7.
Antimicrob Agents Chemother ; 57(7): 3293-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23629702

ABSTRACT

The treatment of osteomyelitis induced by Gram-negative bacilli is rarely reported in the literature. This study established a rabbit tibia model of osteomyelitis induced by the Gram-negative bacillus Escherichia coli. Using this model, pellets composed of a chitosan-bonded mixture of borate bioactive glass and gentamicin were evaluated in vitro and in vivo for the treatment of osteomyelitis induced by Escherichia coli. Our results showed that the pellets in phosphate-buffered saline released gentamicin continuously over 26 days. Without the simultaneous use of a systemic antibiotic, the implantation of the gentamicin-loaded pellets into the osteomyelitis region of the tibia resulted in the eradication of 81.82% of infections, as determined by microbiological, histological and radiographic evaluation, and supported the ingrowth of new bone into the tibia defects after 6 weeks of implantation. The results indicate that the gentamicin-loaded borate bioactive glass implant, combining sustained drug release with the ability to support new bone formation, could provide a method for treating osteomyelitis induced by Gram-negative bacilli.


Subject(s)
Escherichia coli Infections/drug therapy , Gentamicins/administration & dosage , Osteomyelitis/drug therapy , Animals , Anti-Bacterial Agents/therapeutic use , Biocompatible Materials/therapeutic use , Borates/therapeutic use , Ceramics , Delayed-Action Preparations/therapeutic use , Disease Models, Animal , Drug Carriers , Drug Implants , Escherichia coli/drug effects , Escherichia coli Infections/microbiology , Gentamicins/therapeutic use , Glass , Male , Osteomyelitis/microbiology , Rabbits
8.
Biomaterials ; 33(29): 7008-18, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22818985

ABSTRACT

The aims of this study were to (1) determine whether platelet-rich plasma (PRP) could be prepared as a bioactive scaffold capable of endogenous growth factor release for cartilage repair; (2) compare the chondrogenic differentiation ability of mesenchymal stem cells (MSCs) from bone marrow (BMSC) and from adipose (ADSC) seeded within the PRP scaffold; and (3) test the efficacy of ADSC-PRP construct in cartilage regeneration in vivo. In vitro evaluation showed that a 3-dimensional scaffold with a mesh-like microstructure was formed from PRP, with the capability of endogenous growth factor release and ready cell incorporation. Upon seeding in the PRP scaffold, BMSC showed higher proliferation rate, and higher expression of cartilage-specific genes and proteins than ADSC. In an osteochondral defect model in rabbits, implanted BMSC seeded within PRP scaffold also exhibited better gross appearance and histological and immunohistochemical characteristics, higher cartilage-specific gene and protein expression as well as subchondral bone regeneration. ADSC seeded constructs developed into functional chondrocytes secreting cartilaginous matrix in rabbits at 9 weeks post-implantation. Our findings suggest that PRP is a candidate bioactive scaffold capable of releasing endogenous growth factors and that BMSC and ADSC seeded within the PRP scaffold differentiate into chondrocytes and may be suitable for cell-based cartilage repair.


Subject(s)
Adipose Tissue/cytology , Cartilage/cytology , Mesenchymal Stem Cells/cytology , Regeneration/physiology , Animals , Blood Platelets/cytology , Bone Marrow Cells/cytology , Cell Differentiation , Cell Proliferation , Chondrogenesis , Male , Microscopy, Electron, Scanning/methods , Platelet-Rich Plasma/metabolism , Rabbits , Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry
9.
Article in Chinese | MEDLINE | ID: mdl-22792754

ABSTRACT

OBJECTIVE: To evaluate the characterization, biocompatibility in vitro and in vivo, and antimicrobial activity of an injectable vancomycin-loaded borate glass/chitosan composite (VBC) so as to lay the foundation for its further clinical application. METHODS: The solid phase of VBC was constituted by borate glass and vancomycin, liquid phase was a mixture of chitosan, citric acid, and glucose with the proportion of 1 : 10 : 20. Solid phase and liquid phase was mixed with the ratio of 2 : 1. Vancomycin-loaded calcium sulfate (VCS) was produced by the same method using calcium sulfate instead of borate glass and saline instead of chitosan, as control. High performance liquid chromatography was applied to detect the release rate of antibiotics from VBC and VCS, and minimum inhibitory concentration (MIC) was tested by using an antibiotic tube dilution method. The structure of the VBC and VCS specimens before and 2, 4, 8, 16, and 40 days after immersion in D-Hank's was examined by scanning electron microscopy, and the phase composition of VBC was analysed by X-ray diffraction after soaked for 40 days. Thirty-three healthy adult New Zealand white rabbits (weighing, 2.25-3.10 kg; male or female) were used to establish the osteomyelitis models according to Norden method. After 4 weeks, the models of osteomyelitis were successfully established in 28 rabbits, and they were randomly divided into 4 groups (groups A, B, C, and D). In group A (n=8), simple debridement was performed; in groups B and C (n=8), defect was treated by injecting VCS or VBC after debridement; and in group D (n=4), no treatment was given. The effectiveness of treatment was assessed using radiological and histological techniques after 2 months. RESULTS: The releases of vancomycin from VBC lasted for 30 days; the release rate of vancomycin reached 75% at the first 8 days, then could reached more than 90%. The releases of vancomycin from VCS lasted only for 16 days. The MIC of VBC and VCS were both 2 microg/mL. The VCS had a smooth glass crystal surface before immersion, however, it was almost degradated after 4 days. The fairly smooth surface of the VBC pellet became more porous and rougher with time, X-ray diffraction analysis of VBC soaked for 40 days indicated that the borate glass had gradually converted to hydroxyapatite. After 2 months, the best result of treatment was observed in group C, osteomyelitis symptoms disappeared. The X-ray scores of groups A, B, C, and D were 3.50 +/- 0.63, 2.29 +/- 0.39, 2.00 +/- 0.41, and 4.25 +/- 0.64, respectively; Smeltzer scores were 6.00 +/- 0.89, 4.00 +/- 0.82, 3.57 +/- 0.98, and 7.25 +/- 0.50, respectively. The scores were significantly higher in group D than in groups A, B, and C (P < 0.05), and in group A than in groups B and C (P < 0.05). The scores were higher in group B than in group C, but no significant difference was found (P > 0.05). CONCLUSION: The VBC is effective in treating chronic osteomyelitis of rabbit by providing a sustained release of vancomycin, in addition to stimulating bone regeneration, so it may be a promising biomaterial for treating chronic osteomyelitis.


Subject(s)
Borates/chemistry , Chitosan/chemistry , Drug Carriers/therapeutic use , Osteomyelitis/drug therapy , Vancomycin/administration & dosage , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/therapeutic use , Biocompatible Materials/chemistry , Calcium Sulfate/chemistry , Calcium Sulfate/therapeutic use , Delayed-Action Preparations , Disease Models, Animal , Drug Carriers/chemistry , Female , Glass/chemistry , Injections , Male , Materials Testing , Osteomyelitis/microbiology , Rabbits , Staphylococcal Infections/drug therapy , Vancomycin/chemistry , Vancomycin/metabolism , Vancomycin/therapeutic use , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL