Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
World Neurosurg ; 183: e963-e970, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38266990

RESUMO

OBJECTIVE: The objective of this study was to evaluate the surgical effectiveness of posterior procedure with long segment stabilization for treating thoracolumbar pseudarthrosis associated with ankylosing spinal disorders (ASDs) without anterior fusion or osteotomy. METHODS: Twelve patients with thoracolumbar pseudarthrosis in ASD were enrolled. All patients underwent posterior long-segment stabilization procedures. In some patients, the percutaneous technique or the aid of a robot or O-arm navigation was utilized for pedicle screw implantation. The clinical results were evaluated by means of the visual analog scale and Oswestry Disability Index. Radiological outcomes were evaluated for bone fusion, anterior column defect, local kyphotic correction, and position of the pedicle screws. RESULTS: All patients experienced effective bone fusion at the sites of pseudarthrosis. The mean operative time was 161.7 ± 57.1 minutes, and the average amount of blood loss was 305.8 ± 293.2 mL. For 6 patients who underwent surgery with the assistance of a robot or O-arm navigation, there was no statistically significant difference observed in terms of operative time and mean blood loss compared to those who used the freehand technique (P > 0.05). The visual analog scale score, Oswestry Disability Index value, and mean local kyphotic angle showed significant improvements at the final follow-up (P < 0.05). The accuracy of pedicle screw placement was 96%. CONCLUSIONS: Posterior surgery with long-segment fixation, without anterior fusion or osteotomy, can achieve satisfactory outcomes in ASD patients with thoracolumbar pseudarthrosis. The application of percutaneous techniques, as well as the assistance of robots or navigation technique may be a good choice for the treatment of pseudarthrosis in ASD patients.


Assuntos
Cifose , Parafusos Pediculares , Pseudoartrose , Fraturas da Coluna Vertebral , Fusão Vertebral , Cirurgia Assistida por Computador , Humanos , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/cirurgia , Vértebras Lombares/lesões , Pseudoartrose/diagnóstico por imagem , Pseudoartrose/cirurgia , Imageamento Tridimensional , Tomografia Computadorizada por Raios X , Cifose/diagnóstico por imagem , Cifose/etiologia , Cifose/cirurgia , Resultado do Tratamento , Fusão Vertebral/métodos , Estudos Retrospectivos , Vértebras Torácicas/diagnóstico por imagem , Vértebras Torácicas/cirurgia , Vértebras Torácicas/lesões , Fraturas da Coluna Vertebral/cirurgia
2.
JBMR Plus ; 7(12): e10811, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38130773

RESUMO

Bone homeostasis, the equilibrium between bone resorption and formation, is essential for maintaining healthy bone tissue in adult humans. Disruptions of this process can lead to pathological conditions such as osteoporosis. Dual-targeted agents, capable of inhibiting excessive bone resorption and stimulating bone formation, are being explored as a promising strategy for developing new treatments to address osteoporosis. In this study, we investigated the effects of P7C3 on bone remodeling and its potential therapeutic role in osteoporosis treatment in mice. Specifically, P7C3 can remarkably suppress receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclast differentiation in bone marrow macrophages via the Akt-NF-κB-NFATc1 signaling pathway. Additionally, RNA sequencing (RNAseq) analysis revealed that P7C3 promoted osteoblast differentiation and function through the Wnt/ß-catenin signaling pathway, thereby enhancing bone formation. Furthermore, µCT analysis and histological examination of bone tissues from P7C3-treated mice showed attenuation of both Ti-induced bone erosion and ovariectomy (OVX)-induced bone loss. These findings suggest that P7C3 may have a novel function in bone remodeling and may be a promising therapeutic agent for the treatment of osteoporosis. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

3.
Aging Dis ; 2023 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-37196128

RESUMO

Intervertebral disc degeneration is a leading cause of disability in the elderly population. Rigid extracellular matrix is a critical pathological feature of disc degeneration, leading to aberrant nucleus pulposus cells (NPCs) proliferation. However, the underlying mechanism is unclear. Here, we hypothesize that increased matrix stiffness induces proliferation and thus degenerative phenotypes of NPCs through YAP/TEAD1 signaling pathway. We established hydrogel substrates to mimic stiffness of degenerated human nucleus pulposus tissues. RNA-sequencing identified differentially expressed genes between primary rat NPCs cultured on rigid and soft hydrogels. Dual luciferase assay and gain- and loss-function experiments evaluated the correlation between YAP/TEAD1 and Cyclin B1. Furthermore, single-cell RNA-sequencing of human NPCs was performed to identify specific cell clusters with high YAP expression. Matrix stiffness increased in severely degenerated human nucleus pulposus tissues (p < 0.05). Rigid substrate enhanced rat NPCs proliferation mainly through Cyclin B1, which was directly targeted and positively regulated by YAP/TEAD1. Depletion of YAP or Cyclin B1 arrested G2/M phase progression of rat NPCs and reduced fibrotic phenotypes including MMP13 and CTGF (p < 0.05). Fibro NPCs with high YAP expression were identified in human tissues and responsible for fibrogenesis during degeneration. Furthermore, inhibition of YAP/TEAD interaction by verteporfin suppressed cell proliferation and alleviated degeneration in the disc needle puncture model (p < 0.05). Our results demonstrate that elevated matrix stiffness stimulates fibro NPCs proliferation through YAP/TEAD1-Cyclin B1 axis, indicating a therapeutic target for disc degeneration.

4.
Int J Biol Sci ; 19(6): 1910-1924, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37063428

RESUMO

The stem cell factor (SCF) binds to c-Kit in endothelial cells, thus activating downstream signaling and angiogenesis. Herein, we examined the role of G protein subunit alpha inhibitory (Gαi) proteins in this process. In MEFs and HUVECs, Gαi1/3 was associated with SCF-activated c-Kit, promoting c-Kit endocytosis, and binding of key adaptor proteins, subsequently transducing downstream signaling. SCF-induced Akt-mTOR and Erk activation was robustly attenuated by Gαi1/3 silencing or knockout (KO), or due to dominant negative mutations but was strengthened substantially following ectopic overexpression of Gαi1/3. SCF-induced HUVEC proliferation, migration, and capillary tube formation were suppressed after Gαi1/3 silencing or KO, or due to dominant negative mutations. In vivo, endothelial knockdown of Gαi1/3 by intravitreous injection of endothelial-specific shRNA adeno-associated virus (AAV) potently reduced SCF-induced signaling and retinal angiogenesis in mice. Moreover, mRNA and protein expressions of SCF increased significantly in the retinal tissues of streptozotocin-induced diabetic retinopathy (DR) mice. SCF silencing, through intravitreous injection of SCF shRNA AAV, inhibited pathological retinal angiogenesis and degeneration of retinal ganglion cells in DR mice. Finally, the expression of SCF and c-Kit increased in proliferative retinal tissues of human patients with proliferative DR. Taken together, Gαi1/3 mediate SCF/c-Kit-activated signaling and angiogenesis.


Assuntos
Células Endoteliais , Transdução de Sinais , Animais , Humanos , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Endoteliais/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/genética , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo
5.
Orthop Surg ; 15(1): 286-300, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36387061

RESUMO

OBJECTIVES: Osteosarcoma (OS) is a malignant tumor with frequent occurrence among teenagers. Long non-coding RNAs (lncRNAs) play pro-cancer roles in many tumors. The purpose of this study was to figure out the functional role of a novel lncRNA long intergenic non-protein coding RNA 665 (LINC00665) in OS by observing the OS cell behaviors. METHODS: Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to analyze LINC00665 expression in OS cells. Cell function assays assessed the impacts of LINC00665 on OS cell phenotype. Immunofluorescence and western blot analyzed the function of LINC00665 on epithelial-mesenchymal transition (EMT) in OS. Moreover, mechanistic assays analyzed the downstream mechanism of LINC00665 in OS cells. RESULTS: LINC00665 was significantly up-regulated in OS cells. LINC00665 silence facilitated OS cell proliferation, migration, invasion, and EMT while inhibiting cell apoptosis. Mechanically, LINC00665 acted as a competing endogenous RNA (ceRNA) to sponge miR-1249-5p and thereby modulated Wnt family member 2B (WNT2B) to activate Wnt pathway. Wnt pathway activated LINC00665 expression transcriptionally. CONCLUSIONS: Our study uncovered the cancer-promoting role of LINC00665 in OS, and the feedback loop of LINC00665/miR-1249-5p/WNT2B/Wnt might be a potential target for OS treatment.


Assuntos
Neoplasias Ósseas , MicroRNAs , Osteossarcoma , RNA Longo não Codificante , Humanos , MicroRNAs/metabolismo , Via de Sinalização Wnt , Transição Epitelial-Mesenquimal/genética , Retroalimentação , Osteossarcoma/metabolismo , Neoplasias Ósseas/patologia , Proliferação de Células , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética
6.
Commun Biol ; 5(1): 641, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35768581

RESUMO

Osteoarthritis (OA) is a highly prevalent and chronic disorder that is associated with a substantial social and economic burden. Itaconate, as an important regulator of cellular inflammation, is a metabolite synthesised by an enzyme encoded by immune-responsive gene 1. However, there are few studys regarding the effects of itaconate on OA. Here, we show the effect of the cell-permeable itaconate derivative 4-octyl itaconate (OI) on OA. OI attenuates the chondrocyte apoptosis induced by interleukin 1ß (IL-1ß) in vitro, indicating that OI protect chondrocytes against apoptosis. Moreover, OI ameliorates the chondrocyte autophagy inhibition induced by IL-1ß via the inhibition of PI3K/AKT/mTOR signalling pathway. Finally, OI enhances autophagy and reduces cartilage degradation in a rat model of OA established by destabilization of medial meniscus (DMM). In summary, our findings reveal that OI is involved in regulating the progression of OA. The above results shed light on the treatment of OA.


Assuntos
Condrócitos , Osteoartrite , Animais , Autofagia , Condrócitos/metabolismo , Humanos , Osteoartrite/tratamento farmacológico , Osteoartrite/genética , Osteoartrite/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Succinatos , Serina-Treonina Quinases TOR/metabolismo
7.
Biomaterials ; 284: 121482, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35358870

RESUMO

Fracture is one of the most common clinical diseases that reduce the quality of patients' lives significantly. In this study, we prepared gold nanorods modified by endogenous proteins which collected from the autologous blood of individual mice for enhanced photothermal therapy (PTT) to treat fracture. Due to the outermost layer being endogenous proteins, we find that GNRs neither activate the immune cells in vitro nor cause any rejection immune responses after entering the body as compared with PEG modification. In addition, the internal bleeding and edema of the fracture site result in a rapid enrichment of GNRs after intravenous injection. Under near infrared (NIR) light irradiation, the mild photothermal effect of the accumulated GNRs can effectively promote healing of fracture in mice. The molecular mechanism of osteogenic capability is revealed by transcriptome sequencing and subsequent confirmatory experiments, indicating enhanced two key osteogenic signal transduction (MAPK, PI3K-Akt) and multiple key osteogenesis related factors expression following the treatment. Our strategy offers an alternative way to promote bone regeneration following a fracture.


Assuntos
Ouro , Nanotubos , Animais , Linhagem Celular Tumoral , Ouro/uso terapêutico , Humanos , Camundongos , Osteogênese , Fosfatidilinositol 3-Quinases , Fototerapia , Transdução de Sinais
8.
Int J Biol Sci ; 18(4): 1508-1520, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35280670

RESUMO

Sustained activation of multiple receptor tyrosine kinases (RTKs) simultaneously is vital for tumorigenesis and progression of osteosarcoma (OS). Gαi proteins recruitment to various RTKs mediates downstream oncogenic signaling activation. The expression, functions and underlying mechanisms of Gαi3 in human OS were examined. Expression of Gαi3 is robustly elevated in human OS tissues and is correlated with a poor overall survival. In patient-derived primary OS cells and immortalized lines (MG63 and U2OS), Gαi3 depletion, by shRNA and CRISPR/Cas9 strategies, robustly suppressed cell viability, proliferation and migration, while provoking G1-S arrest and apoptosis activation. Conversely, Gαi3 overexpressing ectopically can accelerate proliferation and migration of OS cells. In OS cells, Gαi3 immunoprecipitated with VEGFR2, FGFR, PGDFR and EGFR, mediating downstream cascade transduction. Akt-mTOR activation in primary OS cells was potently inhibited by Gαi3 shRNA, knockout or dominant negative mutation, but augmented after Gαi3 overexpression. In vivo studies showed that Gαi3 shRNA AAV (adeno-associated viruses) intratumoral injection largely inhibited the growth of subcutaneous xenografts of primary OS cells. Moreover, the growth of the Gαi3-knockout primary OS xenografts was much slower than that of OS xenografts with empty vector. In Gαi3-depleted OS xenografts tissues, Gαi3 downregulation and Akt-mTOR inactivation were detected. Taken together, overexpressed Gαi3 mediates RTK-Akt signaling to drive OS progression.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Apoptose/genética , Neoplasias Ósseas/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Humanos , Osteossarcoma/genética , Osteossarcoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno , Receptores Proteína Tirosina Quinases , Serina-Treonina Quinases TOR/metabolismo
9.
Cell Death Discov ; 8(1): 48, 2022 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-35115496

RESUMO

Sphingosine kinase 1 (SphK1) expression and activity are elevated in human osteosarcoma (OS) and is a promising target of therapy. SKI-V is a non-competitive and highly-efficient non-lipid SphK1 inhibitor. The potential anti-OS cell activity by the SphK1 inhibitor was studied here. In primary OS cells and immortalized cell lines, SKI-V robustly suppressed cell survival, growth and proliferation as well as cell mobility, and inducing profound OS cell death and apoptosis. The SphK1 inhibitor was however non-cytotoxic nor pro-apoptotic in human osteoblasts. SKI-V robustly inhibited SphK1 activation and induced accumulation of ceramides, without affecting SphK1 expression in primary OS cells. The SphK1 activator K6PC-5 or sphingosine-1-phosphate partially inhibited SKI-V-induced OS cell death. We showed that SKI-V concurrently blocked Akt-mTOR activation in primary OS cells. A constitutively-active Akt1 (ca-Akt1, S473D) construct restored Akt-mTOR activation and mitigated SKI-V-mediated cytotoxicity in primary OS cells. In vivo, daily injection of SKI-V potently suppressed OS xenograft tumor growth in nude mice. In SKI-V-administrated OS xenograft tissues, SphK1 inhibition, ceramide increase and Akt-mTOR inhibition were detected. Together, SKI-V exerts significant anti-OS activity by inhibiting SphK1 and Akt-mTOR cascades in OS cells.

10.
Spine (Phila Pa 1976) ; 47(17): E570-E578, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34923548

RESUMO

STUDY DESIGN: The effect of amlodipine (AM) on spinal cord injury (SCI) and autophagy was researched by establishing ventral spinal cord cells (VSC4.1) oxygen and glucose deprivation model and SCI mice model. OBJECTIVE: To determine the neuroprotective effects of AM by upregulating autophagy during SCI repair. SUMMARY OF BACKGROUND DATA: AM, an antihypertensive medication, has been shown in several studies to inhibit neuronal apoptosis and exert neuroprotective effects in various central nervous system diseases. However, its effects on SCI are unexplored. Autophagy could inhibit cell apoptosis, which has been shown to promote SCI repair. However, the role of AM in autophagy remains unclear. METHODS: We examined the relationship between AM, apoptosis, and autophagy in ventral spinal cord cells and the injured spinal cords of C57BL/6 female mice respectively, following histological, behavioral, microscopic, immunofluorescence, and western blotting analyses. RESULTS: We found that AM could inhibit motor neuronal apoptosis in vitro. Furthermore, AM promoted locomotor recovery by upregulating autophagy and alleviating apoptosis, neuronal loss, and spinal cord damage after SCI. CONCLUSION: AM inhibited motoneuronal apoptosis by upregulating autophagy to improve SCI recovery.


Assuntos
Fármacos Neuroprotetores , Traumatismos da Medula Espinal , Anlodipino/farmacologia , Anlodipino/uso terapêutico , Animais , Apoptose , Autofagia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Medula Espinal , Regulação para Cima
11.
ACS Biomater Sci Eng ; 7(12): 5706-5716, 2021 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-34843223

RESUMO

Rheumatoid arthritis (RA) is a systemic autoimmune disease with clinical manifestations including joint cartilage, synovitis, and bone damage. Here we developed an injectable erythrocyte gel loaded with Bulleyaconitine A (BLA) for the treatment of RA and demonstrated its anti-inflammatory effects in vivo and in vitro. In vitro experiments showed that BLA could effectively down-regulate the expression of pro-inflammatory factor in activated macrophages through the nuclear factor-κB (NF-κB) pathway. In vivo experiments have shown that the injection of BLA@RBCs in the inflammatory joints of CIA mice increases the local concentration of BLA in a long time. Improved therapeutic outcomes and reduced toxicity of BLA are demonstrated in our work. Together, the developed BLA@RBCs drug delivery system provides an alternative strategy to treat RA joints and shows high potential in clinical RA treatment.


Assuntos
Artrite Experimental , Artrite Reumatoide , Aconitina/análogos & derivados , Animais , Artrite Reumatoide/tratamento farmacológico , Eritrócitos , Camundongos , NF-kappa B
12.
J Tissue Eng ; 12: 20417314211031378, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34345399

RESUMO

Skeletal muscle injury is a common disease accompanied by inflammation, and its treatment still faces many challenges. The local inflammatory microenvironment can be modulated by a novel ROS-scavenging hydrogel (Gel) we constructed. And MSCs could differentiate into myoblasts and contribute to muscle tissue homeostasis and regeneration. Here, Gel loaded with mesenchymal stem cells (MSCs) (Gel@MSCs) was developed for repairing the injured skeletal muscle. Results showed that the Gel improved the survivability and enhanced the proliferation of MSCs (≈two-fold), and the Gel@MSCs inhibited the local inflammatory responses as it promoted polarization of M2 macrophages (increased from 5% to 17%), the mediator of the production of anti-inflammatory factors. Western blotting and qPCR revealed the Gel promoted the expression of proteins (≈two-fold) and genes (≈two to six-fold) related to myogenesis in MSCs. Histological assessment indicated that the Gel or MSCs promoted regeneration of skeletal muscle, and the efficacy was more significant at Gel@MSCs than MSCs alone. Finally, behavioral experiments confirmed that Gel@MSCs improved the motor function of injured mice. In short, the Gel@MSCs system we constructed presented a positive effect on reducing skeletal muscle damage and promoted skeletal muscle regeneration, which might be a novel treatment for such injuries.

13.
Bioact Mater ; 6(11): 4014-4026, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33997490

RESUMO

The treatment of large-area bone defects still faces many difficulties and challenges. Here, we developed a blood clot delivery platform loaded with BMP-2 protein (BMP-2@BC) for enhanced bone regeneration. Blood clot gel platform as natural biomaterials can be engineered from autologous blood. Once implanted into the large bone defect site, it can be used for BMP-2 local delivery, as well as modulating osteoimmunology by recruiting a great number of macrophages and regulating their polarization at different stages. Moreover, due to the deep-red color of blood clot gel, mild localized hyperthermia under laser irradiation further accelerated bone repair and regeneration. We find that the immune niche within the bone defect microenvironment can be modulated in a controllable manner by the blood clots implantation and laser treatment. We further demonstrate that the newly formed bone covered almost 95% of the skull defect area by our strategy in both mice and rat disease models. Due to the great biocompatibility, photothermal potential, and osteoimmunomodulation capacity, such technology shows great promise to be used in further clinical translation.

14.
Mol Ther Nucleic Acids ; 24: 385-402, 2021 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-33868783

RESUMO

Osteosarcoma (OS) is the most common primary bone malignancy in the adolescent population. MAFG (v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog G) forms a heterodimer with Nrf2 (NF-E2-related factor 2), binding to antioxidant response element (ARE), which is required for Nrf2 signaling activation. We found that MAFG mRNA and protein expression is significantly elevated in human OS tissues as well as in established and primary human OS cells. In human OS cells, MAGF silencing or knockout (KO) largely inhibited OS cell growth, proliferation, and migration, simultaneously inducing oxidative injury and apoptosis activation. Conversely, ectopic overexpression of MAFG augmented OS cell progression in vitro. MicroRNA-4660 (miR-4660) directly binds the 3' untranslated region (UTR) of MAFG mRNA in the cytoplasm of OS cells. MAFG 3' UTR luciferase activity and expression as well as OS cell growth were largely inhibited with forced miR-4660 overexpression but augmented with miR-4660 inhibition. In vivo, MAGF short hairpin RNA (shRNA) or forced overexpression of miR-4660 inhibited subcutaneous OS xenograft growth in severe combined immunodeficient mice. Furthermore, MAFG silencing or miR-4660 overexpression inhibited OS xenograft in situ growth in proximal tibia of the nude mice. In summary, MAFG overexpression-driven OS cell progression is inhibited by miR-4660. The miR-4660-MAFG axis could be novel therapeutic target for human OS.

15.
J Orthop Translat ; 27: 9-16, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33344167

RESUMO

BACKGROUND: Hepatocyte growth factor (HGF) is a multifunctional growth factor that promotes various biological processes. However, the effect of HGF on bone metabolism in rheumatoid arthritis (RA) remains unknown. Here, we investigated the role of HGF in regulating osteoclastogenesis and bone resorption in RA. METHODS: The expression of HGF in RA patients and collagen-induced arthritis (CIA) mice was examined. The role of HGF on osteoclastogenesis was analysed by osteoclastogenesis and bone resorption assays. The effect of HGF inhibition was evaluated in a CIA mice model. The mechanism of HGF in regulating osteoclastogenesis and bone resorption was explored by a series of in vitro studies. RESULTS: HGF was overexpressed in CIA and RA. HGF stimulated osteoclastogenesis in vitro. SU11274, a selective small molecule blocker of c-Met, impeded the effect of HGF on osteoclastogenesis and bone resorption. HGF regulated osteoclastogenesis by JNK and AKT-GSK-3ß-NFATc1 signallings. SU11274 protected CIA mice from pathological bone loss. CONCLUSIONS: These data strongly suggest that the highly expressed HGF in the joint tissues contributes to bone loss in RA. Inhibition of HGF/c-Met could effectively alleviate pathological bone loss and inflammatory symptoms in CIA mice. HGF/c-Met may be used as a new target for the treatment of bone loss in RA.

16.
Cell Death Dis ; 11(9): 763, 2020 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-32938906

RESUMO

Monocyte-derived cells were shown to promote cartilage repair in osteoarthritis. The role of the long non-coding RNA (lncRNA) MM2P in this function of monocyte-derived cells remained unexplored. Treatment of RAW264.7 murine macrophages and mouse bone marrow-derived macrophages with IL-4 or IL-13 upregulated MM2P expression, upstream of STAT3 and STAT6 phosphorylation. Specifically, MM2P blocked SHP2-mediated dephosphorylation of STAT3 at Try705 and interacted with the RNA-binding protein FUS. In turn, p-STAT3 increased the Sox9 gene expression. These cells released Sox9 mRNA and protein-containing exosomes, as demonstrated by a transmission electron microscope, nanoparticle tracking analysis, and detection of typical surface markers. Their culture supernatant promoted the differentiation of mouse primary chondrocytes, i.e., upregulated the expression of Col1a2 and Acan genes and promoted the secretion of extracellular matrix components proteoglycan and type II collagen. These effects were mediated by Sox9 mRNA and protein delivered to chondrocytes by exosomes. Together, ex vivo treatment of monocyte-derived cells with IL-4 or IL-13 promoted chondrocyte differentiation and functions through exosome-mediated delivery of Sox9 mRNA and protein.


Assuntos
Condrócitos/metabolismo , Exossomos/metabolismo , Monócitos/metabolismo , RNA Longo não Codificante/genética , Fatores de Transcrição SOX9/metabolismo , Animais , Cartilagem Articular/metabolismo , Diferenciação Celular/genética , Condrogênese/genética , Condrogênese/fisiologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Osteoartrite/metabolismo , Proteoglicanas/metabolismo
17.
Adv Healthc Mater ; 9(3): e1901186, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31820852

RESUMO

The chronic inflammatory microenvironment is characterized by the elevated level of reactive oxygen species (ROS). Here, it is hypothesized that developing an ROS-scavenging scaffold loaded with rapamycin (Rapa@Gel) may offer a new strategy for modulating the local inflammatory microenvironment to improve intervertebral disk tissue regeneration. The therapeutic scaffold consisting of ROS-degradable hydrogel can be injected into the injured degeneration site of intervertebral disk (IVD) and can release therapeutics in a programmed manner. The ROS scavenged by scaffold reduces the inflammatory responses. It is found that when rats are treated with Rapa@Gel, this results in an increase in the percentage of M2-like macrophages and a decrease in M1-like macrophages in the inflammatory environment, respectively. Regeneration of IVD is achieved by Rapa@Gel local treatment, due to the increased M2 macrophages and reduced inflammation. This strategy may be extended to the treatment of many other inflammatory diseases.


Assuntos
Materiais Biocompatíveis/administração & dosagem , Materiais Biocompatíveis/química , Degeneração do Disco Intervertebral/tratamento farmacológico , Espécies Reativas de Oxigênio/metabolismo , Sirolimo/administração & dosagem , Animais , Materiais Biocompatíveis/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Hidrogéis , Degeneração do Disco Intervertebral/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Ratos Sprague-Dawley , Sirolimo/farmacocinética , Sirolimo/farmacologia
18.
Biochem Biophys Res Commun ; 503(4): 2255-2262, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-29959919

RESUMO

Dexamethasone (Dex) can induce injury to human osteoblasts. Long non-coding RNA (LncRNA) EPIC1 (Lnc-EPIC1) is a novel Myc-interacting LncRNA. Its effect on Dex-treated human osteoblasts is studied here. In OB-6 osteoblastic cells and primary human osteoblasts, treatment with Dex increased expression of Lnc-EPIC1. Its expression is also elevated in the necrotic femoral head tissues of Dex-taking patients. Ectopic overexpression of Lnc-EPIC1 inhibited Dex-induced apoptosis and programmed necrosis in OB-6 cells and primary human osteoblasts. Reversely, Lnc-EPIC1 silencing by targeted siRNA potentiated Dex-induced cytotoxicity. Myc is the target of Lnc-EPIC1 in osteoblasts. Exogenous overexpression of Myc protected OB-6 cells from Dex. Conversely, Myc knockout by CRISPR-Cas-9 method abolished Lnc-EPIC1-induced OB-6 cytoprotection against Dex. Together, Lnc-EPIC1 expression protects human osteoblasts from Dex possible via regulation of Myc.


Assuntos
Dexametasona/farmacologia , Osteoblastos/citologia , RNA Longo não Codificante/fisiologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Substâncias Protetoras/farmacologia , Proteínas Proto-Oncogênicas c-myc/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...