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










Database
Language
Publication year range
1.
J Orthop Translat ; 44: 47-59, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38229660

ABSTRACT

Background: Osteoarthritis (OA) is a chronic degenerative joint disease that primarily affects middle-aged and elderly individuals. The decline in chondrocyte function plays a crucial role in the development of OA. Inflammasome-mediated chondrocyte pyroptosis is implicated in matrix degradation and cartilage degeneration in OA patients. Guanylate binding protein 5 (GBP5), a member of the GTPase family induced by Interferon-γ (IFN-γ), significantly influences cellular inflammatory responses, including intracellular inflammasome activation and cytokine release. However, the role of GBP5 in chondrocyte pyroptosis and OA progression remains unclear. Methods: In this study, we used tumor necrosis factor-α (TNF-α) to induce inflammation and created an OA mouse model with surgically-induced destabilization of the medial meniscus (DMM). We isolated and cultured primary chondrocytes from the knee joints of suckling C57 mice. TNF-α-stimulated primary chondrocytes served as an in vitro model for OA and underwent RNA sequencing. Chondrocytes were transfected with GBP5-overexpression plasmids and small interfering RNA and were subsequently treated with TNF-α. We assessed the expression of cartilage matrix components (COL2A1 and aggrecan), catabolic factors (MMP9 and MMP13), and NLRP3 inflammasome pathway genes (NLRP3, Caspase1, GSDMD, Pro-IL-1ß, and Pro-Caspase1) using RT-qPCR and Western blotting. We analyzed the expression of GBP5, NLRP3, and Caspase1 in the cartilage of DMM-induced post-traumatic OA mice and human OA patients. Immunohistochemistry (IHC) was used to detect the expression of GBP5, NLRP3 and GSDMD in cartilage specimens from OA patients and mouse DMM models. Chondrocyte pyroptosis was assessed using flow cytometry, and the levels of interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) were measured with ELISA. We conducted double luciferase reporter gene and chromatin immunoprecipitation (ChIP) assays to confirm the relationship between IRF1 and GBP5. Results: GBP5 expression increased in TNF-α-induced chondrocytes, as revealed by RNA sequencing. GBP5 inhibited COL2A1 and aggrecan expression while promoting the expression of MMP9, MMP13, NLRP3, Caspase1, GSDMD, Pro-IL-1ß, and Pro-Caspase1. GBP5 expression also increased in the cartilage of DMM-induced post-traumatic OA mice and human OA patients. Knockout of GBP5 reduced chondrocyte injury in OA mice. GBP5 promoted chondrocyte pyroptosis and the production of IL-1ß and IL-18. Additionally, we found that IRF1 bound to the promoter region of GBP5, enhancing its expression. After co-transfected with ad-IRF1 and siGBP5, the expression of pyroptosis-related genes was significantly decreased compared with ad-IRF1 group. Conclusions: The IRF1/GBP5 axis enhances extracellular matrix (ECM) degradation and promotes pyroptosis during OA development, through the NLRP3 inflammasome signaling pathway. The translational potential of this article: This study underscores the significance of the IRF1/GBP5 axis in NLRP3 inflammasome-mediated chondrocyte pyroptosis and osteoarthritic chondrocyte injury. Modulating IRF1 and GBP5 expression could serve as a novel therapeutic target for OA.

2.
Arch Biochem Biophys ; 751: 109827, 2024 01.
Article in English | MEDLINE | ID: mdl-38000494

ABSTRACT

Osteoarthritis (OA) is a common aging-related disease affecting entire joint structures, encompassing articular cartilage and subchondral bone. Although senescence and dysfunction of chondrocytes are considered crucial factors in the occurrence of OA, the exact pathogenesis remains to be investigated. In our study, chondrocytes were incubated with a conditioned medium obtained from osteoclasts at different differentiation stages, suggesting that osteoclasts and osteoclast precursors suppressed anabolism and promoted the catabolism of chondrocytes in vitro. In contrast, the function of osteoclasts was more significant than osteoclast precursors. Further blocking of osteoclast exosome secretion by using GW4869 abolished the effect of osteoclasts on chondrocytes. Functionally, exosomal transfer of osteoclast-derived miR-212-3p inhibited Smad2 to mediate chondrocyte dysfunction, thus accelerating cartilage matrix degradation in OA via TGF-ß1/Smad2 signaling. The mechanism was also confirmed within the articular cartilage in OA patients and surgery-induced OA mice. Our study provides new information on intercellular interactions in the bone microenvironment within articular cartilage and subchondral bone during OA progression. The miR-212-3p/Smad2 axis is a potential target for the prevention and therapy of OA.


Subject(s)
Cartilage, Articular , MicroRNAs , Osteoarthritis , Animals , Humans , Mice , Cartilage, Articular/metabolism , Chondrocytes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoarthritis/metabolism , Osteoclasts/metabolism , Transforming Growth Factor beta1/metabolism
3.
Neurospine ; 20(4): 1306-1318, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38171298

ABSTRACT

OBJECTIVE: To illustrate a simultaneous single-position oblique lateral interbody fusion (SPOLIF) combined with unilateral percutaneous pedicle screw fixation in treating single-level lumbar tuberculosis, compared with posterior-only approach in clinical and radiographic evaluations. METHODS: Consecutive patients who had undergone surgeries for single-level lumbar tuberculosis from January 2018 to December 2020 were retrospectively reviewed. The patients included were divided into SP-OLIF and posterior-only groups according to surgical methods applied, with follow-up for at least 36 months. Outcomes included estimated blood loss, operative time, and complications for safety evaluation; visual analogue scale (VAS), Oswestry Disability Index (ODI) for efficacy evaluation; erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) for evaluating tuberculosis activity; x-ray and computed tomography scan were used for radiographic evaluation. RESULTS: A total of 136 patients had been enrolled in the study (60 for SP-OLIF and 76 for Posterior-only). The median operative time, blood loss, and hospital stay in SP-OLIF group were significantly less, with a lower complication rate. Meanwhile, the SP-OLIF group showed substantially lower VAS in 1 and 7 days and decreased ODI in the first month postoperatively, without significant difference afterward. Similarly, the median CRP and ESR in SP-OLIF group were significantly lower in 3 and 7 days postoperatively. All indicators had reduced to normal after 3 months. No recurrence had been reported throughout the whole follow-up. CONCLUSION: SP-OLIF was an efficient minimally invasive protocol for single-level lumbar tuberculosis, facilitating earlier clinical improvement, with decreased blood loss, operative time and hospital stay compared with posterior-only approach.

4.
FASEB J ; 36(2): e22115, 2022 02.
Article in English | MEDLINE | ID: mdl-35032415

ABSTRACT

Bone loss is a hallmark of inflammatory bone diseases caused by aberrantly activated osteoclasts (OCLs). Studies have shown that OCLs exhibit various phenotypes and functions due to variations in the source(s) of precursor cells, cytokine expressions, and microenvironment-dependent factors. During these conditions, inflammatory osteoclasts (iOCLs) lose their immune-suppressive effect relative to OCLs under physiological conditions. This induces TNF α-producing CD4+ T cells in an antigen-dependent manner and finally leads to cascade amplification of iOCLs. OCL-derived exosomes have been reported to regulate OCL formation and inhibit the osteoblast activity. However, the specific function and mechanism of iOCL-derived exosomes on osteoblast have not been studied yet. In the present study, we compare the osteoblast promoting activities of iOCL-derived exosomes and OCL-derived exosomes. We found that iOCLs exosomes specifically target osteoblasts through ephrinA2/EphA2. Mechanistically, the lncRNA LIOCE is enriched in iOCL exosomes and promotes the osteoblast activity after being incorporated into osteoblasts. Furthermore, our results revealed that exosomal lncRNA LIOCE stabilizes osteogenic transcription factor Osterix by interacting and reducing the ubiquitination level of Osterix. This study demonstrated that the bone loss is alleviated in the inflammatory osteolysis mice model after injection of iOCL exosomes encapsulating lncRNA LIOCE. The role of exosomes encapsulating lncRNA LIOCE in promoting bone formation was well established in the rat bone repair model. Our results indicate that iOCL-derived exosomal lncRNA LIOCE promotes bone formation by upregulating Osx expression, and thus, the exosomes encapsulating lncRNA LIOCE may be an effective strategy to increase bone formation in osteoporosis and other bone metabolic disorders.


Subject(s)
Exosomes/genetics , Inflammation/genetics , Osteoblasts/physiology , Osteoclasts/physiology , Osteogenesis/genetics , RNA, Long Noncoding/genetics , Sp7 Transcription Factor/genetics , 3T3 Cells , Animals , Cell Differentiation/genetics , Cell Line , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Osteolysis/genetics , Osteoporosis/genetics , Rats , Transcription Factors/genetics , Ubiquitination/genetics , Up-Regulation/genetics
5.
Front Cell Dev Biol ; 9: 730095, 2021.
Article in English | MEDLINE | ID: mdl-34595178

ABSTRACT

The periosteum is critical for bone healing. Studies have shown that the periosteum contains periosteal stem cells (PSCs) with multidirectional differentiation potential and self-renewal ability. PSCs are activated in early fracture healing and are committed to the chondrocyte lineage, which is the basis of callus formation. However, the mechanism by which PSCs are activated and committed to chondrocytes in bone regeneration remains unclear. Here, we show that tartrate acid phosphatase (TRAP)-positive monocytes secrete CTGF to activate PSCs during bone regeneration. The loss function of TRAP-positive monocytes identifies their specific role during bone healing. Then, the secreted CTGF promotes endochondral ossification and activates PSCs in mouse bone fracture models. The secreted CTGF enhances PSC renewal by upregulating the expression of multiple pluripotent genes. CTGF upregulates c-Jun expression through αVß5 integrin. Then, c-Jun transcription activates the transcription of the pluripotent genes Sox2, Oct4, and Nanog. Simultaneously, CTGF also activates the transcription and phosphorylation of Smad3 through αVß5 integrin, which is the central gene in chondrogenesis. Our study indicates that TRAP-positive monocyte-derived CTGF promotes bone healing by activating PSCs and directing lineage commitment and that targeting PSCs may be an effective strategy for preventing bone non-union.

6.
FASEB J ; 35(6): e21543, 2021 06.
Article in English | MEDLINE | ID: mdl-34046950

ABSTRACT

Clinically, bone destruction caused by Mycobacterium tuberculosis was serious especially in patients with vitamin D (VD) deficiency. However, the role of VD in M. tuberculosis-induced bone destruction remains clear. In this context, we investigate the role of VD and vitamin D receptor (VDR) in the M. tuberculosis-induced bone destruction. First, we infected RAW264.7 and bone marrow-derived macrophages (BMMs) with Mycobacterium bovis Bacillus Calmette-Guérin (M. bovis BCG) in vitro. Then, we activated VDR through VD administration. TRAP and FAK staining, bone resorption assays, immunofluorescence staining, qPCR, and western blot were carried out. In vivo, the M. tuberculosis-induced osteolytic model on the murine skull was established and the µCT and histological analyses were performed. We found that VDR and TRAP were upregulated in bone tuberculosis tissue and proved that M. tuberculosis infection promoted osteoclastogenesis in RAW264.7 and BMMs. VD could inhibit osteoclasts differentiation, fusion, and bone resorption dose-dependently. However, when VDR was knocked down, the inhibitory effect of VD on osteoclasts disappeared. In mechanism, activation of VDR inhibits the phosphorylation of IκB α, thereby inhibiting NFκB signaling pathway and alleviating osteoclastogenesis. Furthermore, in the skull osteolysis model, VD administration reduced osteolysis, but not in VDR-/- mice. Our study, for the first time, demonstrates that activation of VDR by VD administration inhibits M. tuberculosis-induced bone destruction. Our results reveal that VD and VDR are potential therapeutic targets for M. tuberculosis-induced bone destruction, and are of great clinical significance for the development of new therapeutic strategies.


Subject(s)
Macrophages/drug effects , Mycobacterium tuberculosis/pathogenicity , NF-kappa B/antagonists & inhibitors , Osteolysis/prevention & control , Receptors, Calcitriol/metabolism , Tuberculosis/complications , Vitamin D/administration & dosage , Animals , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Osteolysis/etiology , Osteolysis/metabolism , Osteolysis/pathology , Receptors, Calcitriol/genetics , Tuberculosis/microbiology , Vitamins/administration & dosage
7.
Orthop Surg ; 12(6): 1579-1588, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31568641

ABSTRACT

OBJECTIVE: To evaluate the efficacy of three surgical approaches for the treatment of cervicothoracic tuberculosis. METHODS: This is a multicenter retrospective study. We analyzed 74 patients with cervicothoracic tuberculosis who were treated in six institutions between January 2000 and January 2015. There were 37 male and 37 female patients, with an average age of 24 years (range, 5-62 years). The operative method was selected according to the indications. A total of 33 patients underwent one-stage anterior surgery (group A); 16 underwent a combined anterior and posterior surgery (group B) and 25 underwent one-stage posterior surgery (group C). Clinical outcomes, laboratory indexes, and radiological results were analyzed. RESULTS: All cases were followed up for approximately 36-96 months post-surgery (average, 39 months). At the last follow-up, patients in all three groups had achieved bone fusion, with pain relief and neurological recovery. No major vessel and nerve injuries were found during the operation. There were significant differences before and after treatment for visual analogue scale (VAS), neck disability index (NDI), and Japanese Orthopedic Association (JOA) score (P < 0.001). Three surgical strategies significantly improved kyphosis (P < 0.001). CONCLUSION: The choice of operation for cervicothoracic tuberculosis should be selected based on the pathological changes, scope, and general physical condition of the patient. The indication for a posterior approach is narrow and it should be used selectively. The combined anterior and posterior approach involved a longer operating time, larger blood loss, and greater trauma, and also required a higher level of surgical skill. Therefore, the indications for this approach should be strictly controlled. Anterior approach surgery for the treatment of cervicothoracic tuberculosis showed excellent efficacy and fewer complications.


Subject(s)
Cervical Vertebrae/microbiology , Cervical Vertebrae/surgery , Spinal Fusion/methods , Thoracic Vertebrae/microbiology , Thoracic Vertebrae/surgery , Tuberculosis, Spinal/surgery , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome , Young Adult
8.
Cell Physiol Biochem ; 49(4): 1512-1522, 2018.
Article in English | MEDLINE | ID: mdl-30205409

ABSTRACT

BACKGROUND/AIMS: Tuberculosis induces bone loss and activates Th1 cells that play an important role in the host defense of Bacille Calmette-Guérin tuberculosis vaccine. However, the role of tuberculosis-activated Th1 cells in differentiation of osteoclast precursors to osteoclasts is unclear. As secretion of IFN-γ in Th1 cells is induced by tuberculosis, we aimed to investigate the role of anti-IFN-γ antibody on the differentiation and activation of osteoclasts in bone marrow monocyte-derived macrophages (BMMs). METHODS: BMMs were isolated and co-cultured with CD4+T helper 1 cells (Th1 cells), pretreated with anti-IFN-γ antibody. Then, cell proliferation, expression and release of cytokines, formation of actin ring, differentiation of osteoclasts and bone resorption function were measured by CCK8 assay, qRT-PCR/Western blot/flow cytometry, ELISA, immunofluorescence, tartrate-resistant acidic phosphatase (TRAP) staining and bone absorbance assay, respectively. RESULTS: Anti-IFN-γ antibody inhibited the cell viability of BMMs, and induced the expressions of RANKL, TNF-α, NF-κB and TRAF6 in BMMs. In addition, it led to increased expression levels of RANK on cell surfaces, and increased production of RANKL, TNF-α, MCP-1 and SDF-1. Anti-IFN-γ antibody also induced the expression of osteoclast differentiation factor and actin ring formation, but inhibited the expression of osteoprotegerin. TRAP staining and bone resorption assays showed that anti-IFN-γ antibody induced an increase in osteoclast formation and bone resorption. CONCLUSION: The anti-IFN-γ antibody induced osteoclast formation, and is probably mediated by RANKL-induced activation of NF-κB, that induces TRAF6 in the RANKL-RANK signaling pathway. Our data suggest an inhibitory role for IFN-γ in osteoclast formation induced by tuberculosis.


Subject(s)
Antibodies/pharmacology , Interferon-gamma/immunology , Osteogenesis/drug effects , Tuberculosis/pathology , Animals , Cell Differentiation , Chemokine CCL2/metabolism , Chemokine CXCL12/metabolism , Coculture Techniques , Humans , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Osteoclasts/cytology , Osteoclasts/metabolism , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction/drug effects , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Tuberculosis/immunology , Tuberculosis/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
9.
Microb Cell Fact ; 16(1): 154, 2017 Sep 18.
Article in English | MEDLINE | ID: mdl-28923077

ABSTRACT

BACKGROUND: Lactococcus lactis is a food grade probiotics and widely used to express heterologous proteins. Generally, target genes are knocked into the L. lactis genome through double-crossover recombination to express heterologous proteins stably. However, creating marker-less heterologous genes knocked-in clones is laborious. In this study, an efficient heterologous gene knock-in reporter system was developed in L. lactis NZ9000. RESULTS: Our knock-in reporter system consists of a temperature-sensitive plasmid pJW and a recombinant L. lactis strain named NZB. The pJW contains homologous arms, and was constructed to knock-in heterologous genes at a fixed locus of NZ9000 genome. lacZ (ß-galactosidase) gene was knocked into the chromosome of NZ9000 as a counter-selective marker through the plasmid pJW to generate NZB. The engineered NZB strain formed blue colonies on X-Gal plate. The desired double-crossover mutants formed white colonies distinctive from the predominantly blue colonies (parental and plasmid-integrated clones) when the embedded lacZ was replaced with the target heterologous genes carried by pJW in NZB. CONCLUSIONS: By using the system, the heterologous gene knocked-in clones are screened by colony phenotype change rather than by checking colonies individually. Our new knock-in reporter system provides an efficient method to create heterologous genes knocked-in clones.


Subject(s)
Gene Knock-In Techniques/methods , Genes, Reporter , Lactococcus lactis/genetics , Chromosomes, Bacterial , Genetic Vectors , Lac Operon , Mutation , Phenotype , Probiotics , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...