Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 124(Pt B): 110948, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37774483

RESUMO

OBJECTIVE: We aimed to explore the effect and mechanism of the Src inhibitor PP2 on osteoarthritis (OA) progression. METHODS: The protein expressions of Src, p-Src (y418) and p-FAK in normal and OA human chondrocytes were detected by immunofluorescence (IF) analysis. Chondrocytes from the femur and tibial plateau of 3-day-old mice were extracted and inoculated into 6-well plates. The chondrocytes were co-cultured with IL-1ß and different doses of PP2, and then the degeneration of extracellular matrix was analyzed. A mouse OA model was induced by destabilizing medial meniscectomy of the right knee. Two weeks after the operation, different doses of PP2 were injected intraperitoneally. The drug was given three times a week for 6 weeks, and then the mice were sacrificed. Histopathological, IF and immunoblotting analyses were used to detect key OA catabolic markers and protein expression and related signaling. RESULTS: The levels of Src, p-Src (y418) and p-FAK in the knee cartilage tissue of patients with OA were abnormally increased. After chondrocytes were co-treated with IL-1ß and different doses of PP2, the results showed that PP2 reduced the abnormal increase of ß-catenin, p-ß-catenin and other proteins induced by IL-1ß, and reversed the decrease of p-Smad3, aggrecan and collagen Ⅱ protein levels. Meanwhile, intraperitoneal injection of PP2 in vivo significantly reduced the degeneration of articular cartilage in the OA mouse model. CONCLUSION: Our data indicate that targeting Src with PP2 protected against cartilage destruction in an OA mouse model by inhibiting Wnt/ß-catenin and activating TGF-ß/Smad signaling, suggesting that Src may be a potential therapeutic target for OA treatment.


Assuntos
Cartilagem Articular , Osteoartrite , Humanos , Camundongos , Animais , beta Catenina/metabolismo , Via de Sinalização Wnt , Osteoartrite/metabolismo , Condrócitos , Cartilagem Articular/patologia , Modelos Animais de Doenças , Fator de Crescimento Transformador beta/metabolismo , Células Cultivadas
2.
Int Immunopharmacol ; 112: 109225, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36095950

RESUMO

Osteoarthritis (OA) is the most prevalent degenerative whole-joint disease characterized by cartilage degeneration, synovial hyperplasia, osteophyte formation, and subchondral bone sclerosis. Currently there are no disease-modifying treatments available for OA because its etiology and pathogenesis are largely unknown. Here we report that a natural carboxylic polyether ionophore that is used as an anti-tumor drug, salinomycin (SAL), may be a promising therapeutic drug for OA in the future. We found that SAL showed no cytotoxicity on mouse chondrocytes and displayed a protective effect against interleukin-1ß (IL-1ß), in cultured mouse chondrocytes and cartilage explants. Treatment with low SAL concentrations directly upregulated the anabolism factors collagen II and aggrecan, while it inhibited the catabolic factors matrix metalloproteinase-13 (MMP13) and metalloproteinase with thrombospondin motifs-5 (ADAMTS5) to protect against extracellular matrix (ECM) degradation, and also suppressed inflammatory responses in mouse chondrocytes. Furthermore, SAL reduced the severity of OA-associated changes and delayed cartilage destruction, subchondral bone sclerosis, and osteophyte formation in a destabilized medial meniscus (DMM) surgery-induced mouse OA model. Mechanistically, a low SAL concentration induced anabolism and inhibited catabolism in chondrocytes via inhibiting Lrp6 phosphorylation and Wnt/ß-catenin signaling. Our results suggested that SAL may serve as a potential disease-modifying therapeutic against OA pathogenesis.


Assuntos
Osteoartrite , Osteófito , Via de Sinalização Wnt , Animais , Camundongos , Agrecanas/metabolismo , beta Catenina/metabolismo , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos , Modelos Animais de Doenças , Interleucina-1beta/metabolismo , Ionóforos/metabolismo , Ionóforos/farmacologia , Ionóforos/uso terapêutico , Metaloproteinase 13 da Matriz/metabolismo , Meniscos Tibiais/patologia , Osteoartrite/metabolismo , Osteoartrite/patologia , Osteófito/metabolismo , Osteófito/patologia , Esclerose/metabolismo , Esclerose/patologia , Trombospondinas/metabolismo , Trombospondinas/farmacologia , Trombospondinas/uso terapêutico
4.
Dis Markers ; 2022: 3556372, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35069930

RESUMO

Osteoarthritis (OA) is a degenerative disease characterized by articular cartilage and/or chondrocyte destruction, and although it has long been considered as a primary disease, the importance of meniscus endothelial cell modulation in the subchondral microenvironment has recently drawn attention. Previous studies have shown that apelin could potentially inhibit cellular apoptosis; however, it remains unclear whether apelin could play a protective role in protecting the endothelium in the OA meniscus. In this study, with the advantages of single-cell RNA sequencing (scRNA-seq) data, in combination with flow cytometry, we identified two endothelial subclusters in the meniscus, featured by high expression of Homeobox A13 (HOXA13) and Ras Protein-Specific Guanine Nucleotide Releasing Factor 2 (RASGRF2), respectively. Compared with control patients, both subclusters decreased in absolute cell numbers and exhibited downregulated APJ endogenous ligand (APLN, coding for apelin) and upregulated apelin receptor (APLNR, coding apelin receptor). Furthermore, we confirmed that in OA, decreased endothelial cell numbers, including both subclusters, were related to intrinsic apoptosis factors: one more relevant to caspase 3 (CASP3) and the other to BH3-Interacting Domain Death agonist (BID). In vitro culturing of meniscal endothelial cells purified from patients proved that apelin could significantly inhibit apoptosis by downregulating these two factors in endothelial cell subclusters, suggesting that apelin could potentially serve as a therapeutic target for patients with OA.


Assuntos
Menisco , Osteoartrite , Apelina/genética , Apelina/farmacologia , Apelina/uso terapêutico , Apoptose , Células Endoteliais/metabolismo , Humanos , Menisco/metabolismo , Osteoartrite/metabolismo
5.
Mol Cell Endocrinol ; 540: 111520, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34838695

RESUMO

Osteoporosis (OP) is characterized by decreased bone mineral density and impaired bone strength. Carfilzomib (CFZ) is a new-generation proteasome inhibitor and has been found to affect bone metabolism. However, the effect and mechanism of CFZ on OP has not been investigated systematically. In this study, we found that protein levels of proteasome activator subunit 1/2 (PSME1/2) increased in OP, and accumulated mostly in osteoblasts and osteoclasts. Treatment with PSME1/2 recombinant protein inhibited osteogenesis and promoted osteoclast formation in vitro. Also, PSME1/2 inhibited the expression of ß-catenin protein, resulting in limitation of Wnt/ß-catenin signaling. CFZ inhibited PSME1 and PSME2 proteasome activities and increased ß-catenin protein level, resulting in the translocation of ß-catenin to the nucleus and activation of canonical Wnt/ß-catenin signaling, further promoting osteogenesis and inhibiting osteoclastic differentiation. In vivo, we conducted ovariectomy (OVX) to create a model of OVX-induced postmenopausal OP in mice. When analyzed by micro-CT scanning, enhancement of bone mineral density, bone volume, trabecular number, and thickness was seen in the CFZ-treated mice. Also, we noticed increased osteogenesis and decreased osteoclastogenesis, diminished expression of PSME1 and PSME2 and activated Wnt/ß-catenin signaling in bone sections from OP mice treated with CFZ. Overall, our data indicated that PSME1/2 may serve as new targets for the treatment of OP, and targeting PSME1/2 with CFZ provides a candidate therapeutic molecule for postmenopausal OP.


Assuntos
Oligopeptídeos/uso terapêutico , Osteoporose/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/genética , Animais , Densidade Óssea/efeitos dos fármacos , Densidade Óssea/genética , Remodelação Óssea/efeitos dos fármacos , Remodelação Óssea/genética , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Osteoporose/patologia , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Células RAW 264.7 , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo
6.
Int J Bioprint ; 7(4): 405, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34805594

RESUMO

The rapid development of scaffold-based bone tissue engineering strongly relies on the fabrication of advanced scaffolds and the use of newly discovered functional drugs. As the creation of new drugs and their clinical approval often cost a long time and billions of U.S. dollars, producing scaffolds loaded with repositioned conventional drugs whose biosafety has been verified clinically to treat critical-sized bone defect has gained increasing attention. Carfilzomib (CFZ), an approved clinical proteasome inhibitor with a much fewer side effects, is used to replace bortezomib to treat multiple myeloma. It is also reported that CFZ could enhance the activity of alkaline phosphatase and increase the expression of osteogenic transcription factors. With the above consideration, in this study, a porous CFZ/ß-tricalcium phosphate/poly lactic-co-glycolic acid scaffold (designated as "cytidine triphosphate [CTP]") was produced through cryogenic three-dimensional (3D) printing. The hierarchically porous CTP scaffolds were mechanically similar to human cancellous bone and can provide a sustained CFZ release. The implantation of CTP scaffolds into critical-sized rabbit radius bone defects improved the growth of new blood vessels and significantly promoted new bone formation. To the best of our knowledge, this is the first work that shows that CFZ-loaded scaffolds could treat nonunion of bone defect by promoting osteogenesis and angiogenesis while inhibiting osteoclastogenesis, through the activation of the Wnt/ß-catenin signaling. Our results suggest that the loading of repositioned drugs with effective osteogenesis capability in advanced bone tissue engineering scaffold is a promising way to treat critical-sized defects of a long bone.

7.
Oncol Lett ; 22(5): 797, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34584572

RESUMO

Osteosarcoma (OS) is the most common malignant bone tumor in children and adolescents and is characterized by early metastasis and frequent recurrence, which greatly affects patient prognosis and survival rates. However, the treatment of OS, its recurrence and subsequent metastasis is now at a clinical bottleneck. To explore new OS chemotherapeutic targets, investigate new therapeutic strategies and improve patient prognosis and survival rates, the roles of paclitaxel (PTX) and monopolar spindle kinase 1 (Mps1) in OS were investigated using in vivo and in vitro models. Mps1 expression was upregulated in OS samples and associated with patient survival times. Moreover, spindle assembly checkpoint (SAC) activation and upregulation of Akt/mTOR signaling were both positively associated with OS progression. PTX treatment significantly inhibited Mps1 expression, as well as migration of OS cells both in vitro. In addition, the combination of Mps1 knockdown and PTX treatment inhibited OS progression in vivo. Mps1 overexpression inhibited the expression of SAC markers and upregulated Akt and mTOR expression, while Mps1 knockdown had the opposite effect. Cells subjected to combined Mps1 knockdown and PTX treatment exhibited activation of SAC and inhibition of Akt/mTOR signaling compared with Mps1 knockdown or PTX treatment alone. Based on these observations, Mps1 inhibition combined with PTX treatment may represent a potentially effective strategy for the treatment of OS.

8.
Exp Cell Res ; 406(2): 112781, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34400174

RESUMO

The imbalance between osteogenesis and adipogenesis in the bone marrow is the main characteristic of osteoporosis (OP). Thus, exploring regulation of the differentiation of bone marrow stromal cells (BMSCs) into osteoblasts and adipocytes is important to identify novel targets for the treatment of OP. In the present study, the master regulator of endoplasmic reticulum (ER) stress, heat shock protein family A (Hsp70) member 5 (HSPA5) was shown to significantly accumulate in osteoblasts and adipocytes, but not in osteoclasts in bone sections from aged and postmenopausal OP mice. In vitro study revealed that HSPA5 negatively modulated osteogenic differentiation and positively promoted adipogenic differentiation, and that targeting HSPA5 with its inhibitor HA15 enhanced osteogenic differentiation and inhibited adipogenic differentiation. Also, HA15 treatment induces ER stress and autophagy, and decreases apoptosis in cells. We constructed a postmenopausal OP model in mice with ovariectomy surgery, and treated the mice with HA15. The results showed that HA15 treatment induced appropriate ER stress, activated autophagy and decreased apoptosis in osteoblasts, thereby alleviating bone loss in vivo. Our results indicated that HSPA5 participated in OP pathogenesis by regulating the differentiation of BMSCs. HSPA5 may serve as a new target for the treatment of OP, and targeting HSPA5 with HA15 prevents the progression of OP and provides a candidate therapeutic molecule for postmenopausal OP.


Assuntos
Estresse do Retículo Endoplasmático , Proteínas de Choque Térmico/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteogênese , Osteoporose Pós-Menopausa/tratamento farmacológico , Sulfonamidas/farmacologia , Tiazóis/farmacologia , Animais , Apoptose , Autofagia , Diferenciação Celular , Chaperona BiP do Retículo Endoplasmático , Feminino , Proteínas de Choque Térmico/genética , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoporose Pós-Menopausa/etiologia , Osteoporose Pós-Menopausa/metabolismo , Osteoporose Pós-Menopausa/patologia , Ovariectomia/efeitos adversos
9.
Am J Transl Res ; 13(4): 2320-2330, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34017392

RESUMO

Osteonecrosis of the femoral head (ONFH) is a devastating bone disease characterized by avascular or aseptic necrosis of the femoral head, and alcohol consumption is reported one of the leading risks to this disease. Previous studies have linked Dickkopf-1 (DKK1) to the occurrence of ONFH, but the role of DKK1 in alcohol-induced ONFH (AONFH) has not been fully discussed. In this study, we found that the expression level of DKK1 was dramatically increased in serum and bone samples from AONFH patients, experimental AONFH rats, and cultured bone marrow mesenchymal stem cells (BMMSCs) with ethanol stimulation. Elevated DKK1 inhibited Wnt/ß-catenin signaling in vivo and in vitro, while knockdown of DKK1 enhanced the nuclear translocation of ß-catenin and promoted osteogenesis and inhibited adipogenesis in the BMMSC cell line C3H10T1/2. Local injection of DKK1 knockout lentivirus into the femoral head of rats alleviated the progression of AONFH, with activated Wnt/ß-catenin signaling, increased bone formation, reduced number of empty adipose lacunae and restored blood supply. In conclusion, our findings confirmed the important role of DKK1 and canonical Wnt/ß-catenin pathway in AONFH. We propose that DKK1 may be a prognostic marker of AONFH and targeting DKK1 to activate the canonical Wnt/ß-catenin pathway and restore osteogenic potential could be a promising therapeutic strategy to prevent AONFH.

10.
Front Bioeng Biotechnol ; 9: 641130, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33842445

RESUMO

Design an implant similar to the human bone is one of the critical problems in bone tissue engineering. Metal porous scaffolds have good prospects in bone tissue replacement due to their matching elastic modulus, better strength, and biocompatibility. However, traditional processing methods are challenging to fabricate scaffolds with a porous structure, limiting the development of porous scaffolds. With the advancement of additive manufacturing (AM) and computer-aided technologies, the development of porous metal scaffolds also ushers in unprecedented opportunities. In recent years, many new metal materials and innovative design methods are used to fabricate porous scaffolds with excellent mechanical properties and biocompatibility. This article reviews the research progress of porous metal scaffolds, and introduces the AM technologies used in porous metal scaffolds. Then the applications of different metal materials in bone scaffolds are summarized, and the advantages and limitations of various scaffold design methods are discussed. Finally, we look forward to the development prospects of AM in porous metal scaffolds.

11.
Front Bioeng Biotechnol ; 9: 748151, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35118053

RESUMO

Avascular necrosis of femoral head (ANFH) is a disease that is characterized by structural changes and collapse of the femoral head. The exact causes of ANFH are not yet clear, but small advances in etiopathogenesis, diagnosis and treatment are achieved. In this study, ß-tricalcium phosphate/poly lactic-co-glycolic acid composite scaffolds incorporated with bisperoxovanadium [bpV (pic)] (bPTCP) was fabricated through cryogenic 3D printing and were utilized to treat rat models with early ANFH, which were constructed by alcohol gavage for 6 months. The physical properties of bPTCP scaffolds and in vitro bpV (pic) release from the scaffolds were assessed. It was found that the sustained release of bpV (pic) promoted osteogenic differentiation and inhibited adipose differentiation of bone marrow-derived mesenchymal stem cells. Micro-computed tomography scanning and histological analysis confirmed that the progression of ANFH in rats was notably alleviated in bPTCP scaffolds. Moreover, it was noted that the bPTCP scaffolds inhibited phosphatase and tensin homolog and activated the mechanistic target of rapamycin signaling. The autophagy induced by bPTCP scaffolds could partially prevent apoptosis, promote osteogenesis and angiogenesis, and hence eventually prevent the progression of ANFH, suggesting that the bPTCP scaffold are promising candidate to treat ANFH.

12.
Invest New Drugs ; 39(2): 416-425, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33128383

RESUMO

Tumor microenvironment is a critical participant in the initiation, progression and drug resistance of carcinomas, including osteosarcoma. Notoginsenoside R1 (NGR1) is a proverbial active ingredient of the traditional Chinese medicine Panax notoginseng (PN) and possess undeniable roles in several cancers. Nevertheless, its function in osteosarcoma and tumor microenvironment remains elusive. In the current study, exposure to NGR1 dose-dependently inhibited osteosarcoma cell viability and migration, and induced apoptosis. Furthermore, osteosarcoma cells that were incubated with conditioned medium (CM) from bone marrow mesenchymal stem cells (BMSCs) exhibited greater proliferation, migration capacity and MMP-2 and MMP-9 expression relative to control cells, which was reversed when BMSCs were treated with NGR1. Notably, administration with NGR1 antagonized CM-evoked doxorubicin resistance in osteosarcoma cells by decreasing cell viability and increasing cell apoptosis and caspase-3/9 activity. Mechanically, NGR1 suppressed IL-6 secretion from BMSCs, as well as the subsequent activation of the JAK2/STAT3 signaling in osteosarcoma cells. In addition, blocking the JAK2 pathway by its antagonist AG490 reversed CM-induced osteosarcoma cell proliferation, migration and doxorubicin resistance. Moreover, exogenous supplementation with IL-6 engendered not only the reactivation of the JAK2/STAT3 signaling but also muted NGR1-mediated efficacy against osteosarcoma cell malignancy and doxorubicin resistance. Collectively, NGR1 may directly restrain osteosarcoma cell growth and migration, or indirectly antagonize MSC-evoked malignancy and drug resistance by interdicting IL-6 secretion-evoked activation of the JAK2/STAT3 pathway. Consequently, the current study may highlight a promising therapeutic strategy against osteosarcoma by regulating tumor cells and the tumor microenvironment.


Assuntos
Carcinogênese/efeitos dos fármacos , Ginsenosídeos/farmacologia , Janus Quinase 2/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Fator de Transcrição STAT3/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Interleucina-6 , Osteossarcoma/patologia , Transdução de Sinais/efeitos dos fármacos
13.
Front Bioeng Biotechnol ; 8: 576969, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33330415

RESUMO

Titanium and its alloys have superb biocompatibility, low elastic modulus, and favorable corrosion resistance. These exceptional properties lead to its wide use as a medical implant material. Titanium itself does not have antibacterial properties, so bacteria can gather and adhere to its surface resulting in infection issues. The infection is among the main reasons for implant failure in orthopedic surgeries. Nano-modification, as one of the good options, has the potential to induce different degrees of antibacterial effect on the surface of implant materials. At the same time, the nano-modification procedure and the produced nanostructures should not adversely affect the osteogenic activity, and it should simultaneously lead to favorable antibacterial properties on the surface of the implant. This article scrutinizes and deals with the surface nano-modification of titanium implant materials from three aspects: nanostructures formation procedures, nanomaterials loading, and nano-morphology. In this regard, the research progress on the antibacterial properties of various surface nano-modification of titanium implant materials and the related procedures are introduced, and the new trends will be discussed in order to improve the related materials and methods.

14.
J Immunol Res ; 2020: 8873261, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33294469

RESUMO

Stroke is a cerebrovascular disease that results in decreased blood flow. Although Panax notoginseng (PN), a Chinese herbal medicine, has been proven to promote stroke recovery, its molecular mechanism remains unclear. In this study, middle cerebral artery occlusion (MCAO) was induced in rats with thrombi generated by thread and subsequently treated with PN. After that, staining with 2,3,5-triphenyltetrazolium chloride was employed to evaluate the infarcted area, and electron microscopy was used to assess ultrastructural changes of the neurovascular unit. RNA-Seq was performed to determine the differential expressed genes (DEGs) which were then verified by qPCR. In total, 817 DEGs were identified to be related to the therapeutic effect of PN on stroke recovery. Further analysis by Gene Oncology analysis and Kyoto Encyclopedia of Genes and Genomes revealed that most of these genes were involved in the biological function of nerves and blood vessels through the regulation of neuroactive live receptor interactions of PI3K-Akt, Rap1, cAMP, and cGMP-PKG signaling, which included in the 18 pathways identified in our research, of which, 9 were reported firstly that related to PN's neuroprotective effect. This research sheds light on the potential molecular mechanisms underlying the effects of PN on stroke recovery.


Assuntos
Biomarcadores , Medicamentos de Ervas Chinesas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Panax notoginseng/química , Traumatismo por Reperfusão/etiologia , Animais , Biópsia , Biologia Computacional/métodos , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Ontologia Genética , Ratos , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/diagnóstico , Traumatismo por Reperfusão/tratamento farmacológico , Roedores , Transdução de Sinais , Transcriptoma
15.
J Food Sci ; 85(4): 1302-1306, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32144772

RESUMO

The effects and mechanisms of soybean isoflavone on osteoblast (OB) proliferation in vitro were investigated. Fifty female Wistar rats were randomly divided into five groups with 10 rats in each group. Rat OBs were separated and cultured. The first generation of OBs cultured for 48 hr at various concentrations of isoflavone were set as the experimental groups, the OBs exposed to estradiol (E2 ) culture were considered as positive control group. The biological characterization of OBs was investigated by phase contrast microscopy and alkaline phosphatase (ALP) histochemistry. The concentrations of interleukin (IL-1), osteoprotegerin (OPG), transforming growth factor (TGF), granulocyte-macrophage colony-stimulating factor (GM-CSF), and vascular endothelial growth factor (VEGF) in isoflavone culture solutions were determined. Proliferation rate of OBs was increased in experimental group comparing that in the blank group. ALP activity in experimental group was higher than that in blank group. No significant differences of ALP activity were observed between E2 culture group and isoflavone group at concentrations of 10-5 and 10-7 mM (P > 0.05). Furthermore, in the experimental groups at low isoflavone concentrations, the concentrations of OPG, TGF, and VEGF were increased and positively correlated with OB proliferation. However, the concentrations of IL-1, GM-CSF were decreased at higher concentration of isoflavone and were negatively correlated with OB proliferation. Soybean isoflavone could promote the growth and proliferation of rat OB, it might act as the stimulator of OPG, TGF, and VEGF pathway, and the inhibitor of IL-1, GM-CSF pathway as well.


Assuntos
Glycine max/química , Isoflavonas/farmacologia , Osteoblastos/efeitos dos fármacos , Fosfatase Alcalina , Animais , Células Cultivadas , Citocinas/metabolismo , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Isoflavonas/química , Osteoblastos/metabolismo , Ratos , Ratos Wistar , Glycine max/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Exp Ther Med ; 19(1): 696-702, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31885708

RESUMO

Role of microRNA-429 (miRNA-429) in osteogenic differentiation of hADMSCs was elucidated to explore the potential mechanism. Serum level of miRNA-429 in osteoporosis patients and controls was determined by quantitative real-time polymerase chain reaction (qRT-PCR). After H2O2 induction in hADMSCs, cell viability and reactive oxygen species (ROS) level were determined by cell-counting kit (CCK-8) assay and flow cytometry, respectively. Alkaline phosphatase (ALP) activity in H2O2-induced hADMSCs was also detected. The binding condition between miRNA-429 and SCD-1 was verified by dual-luciferase reporter gene assay. Relative levels of osteogenesis-related genes influenced by SCD-1 and miRNA-429 were detected by qRT-PCR. Furthermore, regulatory effects of SCD-1 and miRNA-429 on ALP activity and calcification ability of hADMSCs were evaluated. miRNA-429 was significantly upregulated in serum of osteoporosis patients. During the process of osteogenesis differentiation, H2O2 induction gradually upregulated miRNA-429 in hADMSCs. Overexpression of miRNA-429 markedly reduced ALP activity. Subsequent dual-luciferase reporter gene assay verified that miRNA-429 could bind to SCD-1 and negatively regulated its protein level in hADMSCs. SCD-1 was obviously downregulated in the osteogenesis differentiation of hADMSCs under oxidative stress. Moreover, silencing of SCD-1 suppressed expression of osteogenesis-related gene, ALP activity and calcification ability. Notably, SCD-1 knockdown partially reversed the regulatory effect of miRNA-429 on the osteogenic differentiation of hADMSCs. miRNA-429 suppresses the osteogenic differentiation of hADMSCs under oxidative stress via downregulating SCD-1.

17.
Mol Immunol ; 116: 56-62, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31605961

RESUMO

Macrophages can be polarized towards either a classically activated pro-inflammatory (M1) state, or alternatively towards an activated anti-inflammatory (M2) state. M1 cells are activated by ligands of toll-like receptor (TLR) or interferon (IFN)-γ and have a toxic effect, whereas M2 cells are activated by interleukin (IL)-4, IL-10, and IL-13 and have a regenerative effect in vitro and in vivo. Previously studies have shown that these cells play an important role in the inflammatory responses following spinal cord injury (SCI). Mechanistically, the role of PTEN in the regulation of macrophage polarization has yet to be fully elucidated. In the present study, we first evaluated the expression of PTEN in macrophages after SCI. We found that PTEN expression was accumulated in the macrophages after the SCI surgery. Knock-down of PTEN or inhibition of phospho-PTEN with bpV(pic) in RAW264.7 cells resulted in increased M2 polarization and decreased M1 polarization. In a rat model of SCI, grafts containing bpV(pic) reduced spinal tissue cavitation and promoted locomotor improvement, while combining grafts of bpV(pic) and acellular spinal cord (ASC) scaffolds showed a better effect. Moreover, grafts containing bpV(pic) enhanced M2 polarization and decreased M1 polarization in the macrophages during SCI. Thus, we have established that PTEN is critical for the polarization of macrophages and the functional recovery of SCI. Targeting PTEN enhances the macrophages towards to M2 polarization and promoting the functional recovery in SCI, and this suggest that PTEN may be a future therapeutic target for SCI treatment.


Assuntos
Macrófagos/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Compostos de Vanádio/farmacologia , Animais , Linhagem Celular , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , PTEN Fosfo-Hidrolase/metabolismo , Células RAW 264.7 , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/metabolismo , Receptores Toll-Like/metabolismo
18.
Drug Des Devel Ther ; 13: 513-521, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30774313

RESUMO

BACKGROUND: Spinal cord injury (SCI) is a disease of the central nervous system with few restorative treatments. Autophagy has been regarded as a promising therapeutic target for SCI. The inhibitor of phosphatase and tensin homolog deleted on chromosome ten (PTEN) bisperoxovanadium (bpV[pic]) had been claimed to provide a neuroprotective effect on SCI; but the underlying mechanism is still not fully understood. MATERIALS AND METHODS: Acute SCI model were generated with SD Rats and were treated with control, acellular spinal cord scaffolds (ASC) obtained from normal rats, bpV(pic), and combined material of ASC and bpV(pic). We used BBB score to assess the motor function of the rats and the motor neurons were stained with Nissl staining. The expressions of the main autophagy markers LC3B, Beclin1 and P62, expressions of apoptosis makers Bax, Bcl2, PARP and Caspase 3 were detected with IF or Western Blot analysis. RESULTS: The bpV(pic) showed significant improvement in functional recovery by activating autophagy and accompanied by decreased neuronal apoptosis; combined ASC with bpV(pic) enhanced these effects. In addition, after treatment with ERK1/2 inhibitor SCH772984, we revealed that bpV(pic) promotes autophagy and inhibits apoptosis through activating ERK1/2 signaling after SCI. CONCLUSION: These results illustrated that the bpV(pic) protects against SCI by regulating autophagy via activation of ERK1/2 signaling.


Assuntos
Autofagia/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Traumatismos da Medula Espinal/prevenção & controle , Compostos de Vanádio/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Masculino , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia
19.
Eur Spine J ; 28(3): 470-476, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30014254

RESUMO

PURPOSE: Disorders of the upper thoracic spine can lead to serious disability and morbidity. However, operating on the upper thoracic vertebrae T2-T5 remains challenging because of the anatomical features of the thoracic spine. We describe a novel anterolateral upper thoracic approach, which is safe and reproducible and allows direct access to the upper thoracic spine from T2 to T6 inclusive, obviating the risk of damaging complex anatomical structures inherent in the anterior trans-sternal approach. METHODS: Three patients with upper thoracic spinal-related spinal cord compression disease, presented with progressive thoracic myelopathy and upper back pain. Magnetic resonance imaging showed direct spinal cord compression due to upper thoracic vertebral destruction. In addition preoperative computed tomography also revealed vertebral erosion and collapse. The surgical management of the three patients involved decompression and reconstruction via the right infraaxillary thoracotomy approach, and fixation with a titanium mesh cage and an anterior plate in each. RESULTS: Clinical outcome measures including pre- and postoperative radiographic parameters were assessed. There were no complications associated with this technique. The back pain and neural function gradually improved, and plate placement was achieved in all patients. None of the patients experienced clinical symptoms or screw loosening or breakage in this study. CONCLUSIONS: The technique described is a safe and novel right infraaxillary thoracotomy approach to provide direct access from vertebral bodies T2-T6 and to provide adequate room for upper thoracic vertebral decompression and fusion surgery. However, a suitable fixation implant should be designed. These slides can be retrieved under Electronic Supplementary Material.


Assuntos
Descompressão Cirúrgica/métodos , Compressão da Medula Espinal/cirurgia , Vértebras Torácicas/cirurgia , Toracotomia/métodos , Placas Ósseas , Descompressão Cirúrgica/efeitos adversos , Humanos , Complicações Pós-Operatórias , Toracotomia/efeitos adversos , Tomografia Computadorizada por Raios X
20.
Eur Spine J ; 27(10): 2664, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30143897

RESUMO

Unfortunately, the HTML version of this article was published with incorrect copyright line. It has been corrected now.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...