Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Tissue Bank ; 24(3): 515-522, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36508120

RESUMEN

To determine the expression of signal transducer and activator of transcription 3 (STAT3) in patients with fragility fractures (FFs) and its effect on the biological function of osteoblasts. The study included 32 patients with FFs who were diagnosed and treated in the research group and 30 concurrent healthy individuals in the control group. We observed STAT3 mRNA expression in the patients with FFs and controls and altered STAT3 mRNA to detect changes in the proliferation, invasion, and apoptosis of osteoblasts. The patients with FFs presented higher serum STAT3 mRNA expression than the controls (P < 0.05). We plotted receiver operating characteristic curves based on the STAT3 mRNA expression and found that the area under the curve for STAT3 mRNA was 0.856 (P < 0.05). Transfection of STAT3 mRNA mimics resulted in increased STAT3 mRNA expression, inhibited cell proliferation as detected by an MTT assay, and increased apoptosis rate, which was determined using flow cytometry with human fetal osteoblastic cell line 1.19 cells. STAT3 mRNA expression was elevated in the serum of patients with FFs and can be used as a biomarker for the diagnosis of the disease. Regulating STAT3 mRNA can inhibit the proliferation and induce the osteoblasts apoptosis.


Asunto(s)
Apoptosis , Factor de Transcripción STAT3 , Humanos , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Osteoblastos/metabolismo , ARN Mensajero/genética , Proliferación Celular , Línea Celular Tumoral
2.
Front Chem ; 10: 1088823, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36531327

RESUMEN

Bone metastatic breast cancer has severely threatened the survival and life quality of patients. Due to the suboptimal efficacy of anti-metastatic chemotherapeutic drugs and the complicated bone marrow microenvironments, effective treatment of metastatic breast cancer remains challenging for traditional clinical approaches. In this work, we developed a mesoporous nanoplatform (m-CuS-PEG) with the co-loading of CuS nanodots and a chemotherapeutic drug cisplatin for the combined photothermal-chemotherapy of bone-metastasized breast cancer. The CuS nanodots were decorated onto mesoporous silica (m-SiO2) surface with dendritic mesoporous channels, into which the cisplatin was accommodated. The carboxyl-terminated poly (ethylene glycol) (PEG) was further functionalized onto the surface to obtain the functional nanoplatform m-CuS-PEG. The drug release of the loaded cisplatin exhibited pH- and thermal-dual responsive manner. The attached CuS nanodots rendered the mesoporous nanoplatform with high photothermal conversion ability. Upon irradiation with a near-infrared laser in the second near-infrared (NIR-II) window, m-CuS-PEG dispersions exhibited rapid temperature elevation and high photostability. The results revealed that m-CuS-PEG had excellent biocompatibility. The cisplatin-loaded m-CuS-PEG not only showed superior cancer cell-killing effects, but also significantly inhibit the growth of metastatic tumors. The tumor-induced bone destruction was also dramatically attenuated by the mesoporous nanoplatform-mediated combined therapy. Overall, the developed functional nanoplatform integrates photothermal therapy and efficient chemotherapeutic drug delivery to offer an alternative approach for combating breast cancer bone metastasis.

3.
Orthop Surg ; 11(5): 770-776, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31464084

RESUMEN

OBJECTIVE: Wear-induced aseptic loosening has been accepted as one of the main reasons for failure of total hip arthroplasty. Ceramic wear debris is generated following prosthesis implantation and plays an important part in the upregulation of inflammatory factors in total hip arthroplasty. The present study investigates the influence of ceramic debris on osteoblasts and inflammatory factors. METHODS: Ceramic debris was prepared by mechanical grinding of an aluminum femoral head and added to cultures of MC3T3-E subclone 14 cells at different concentrations (i.e. 0, 5, 10, and 15 µg/mL). Cell proliferation was evaluated using a Cell Counting Kit (CCK-8), and cell differentiation was assessed by mRNA expression of alkaline phosphatase (ALP), osteocalcin (OCN), and osteopontin (OPN). In addition, cell bio-mineralization was evaluated through alizarin red S staining, and release of tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1ß), and interleukin-6 (IL-6) was measured through enzyme-linked immunosorbent assays (ELISA). Furthermore, mRNA expression of Smad1, Smad4, and Smad5 and protein expression of phosphorylated Smad1, Smad4, and Smad5 were measured by reverse transcriptase polymerase chain reaction (RT-PCR) and western blotting. RESULTS: The ceramic debris had irregular shapes and sizes, and analysis of the size distribution using a particle size analyzer indicated that approximately 90% of the ceramic debris was smaller than 3.2 µm (2.0 ± 0.4 µm), which is considered clinically relevant. The results for mRNA expression of ALP, OCN, and OPN and alizarin red S staining indicated that cell differentiation and bio-mineralization were significantly inhibited by the presence of ceramic debris at all tested concentrations (P < 0.05, and the values decreased gradually with the increase of ceramic debris concentration), but the results of the CCK-8 assay showed that cell proliferation was not significantly affected (P > 0.05; there was no significant difference between the groups at 1, 3, and 5 days). In addition, the results of ELISA, RT-PCR, and western blotting demonstrated that ceramic debris significantly promoted the release of inflammatory factors, including TNF-α, IL-ß, and IL-6 (P < 0.05, and the values increased gradually with the increase of ceramic debris concentration), and also greatly reduced the mRNA expression of Smad1, Smad4, and Smad5 (the values decreased gradually with the increase of ceramic debris concentration) as well as protein expression of phosphorylated Smad1, Smad4, and Smad5. CONCLUSION: Ceramic debris may affect differentiation and bio-mineralization of MC3T3-E subclone 14 cells through the bone morphogenetic protein/Smad signaling pathway.


Asunto(s)
Cerámica/efectos adversos , Cuerpos Extraños/complicaciones , Prótesis de Cadera/efectos adversos , Osteoblastos/citología , Células 3T3 , Fosfatasa Alcalina/metabolismo , Animales , Artroplastia de Reemplazo de Cadera , Biomarcadores/metabolismo , Western Blotting , Diferenciación Celular , Proliferación Celular , Citocinas/metabolismo , Ratones , Osteocalcina/metabolismo , Osteopontina/metabolismo , Proteínas Smad/metabolismo
4.
Immunol Invest ; 47(5): 492-503, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29671649

RESUMEN

The lymphocyte activation gene 3 (LAG-3) is a CD4 homolog with binding affinity to MHC class II molecules. It is thought that LAG-3 exerts a bimodal function, such that co-ligation of LAG-3 and CD3 could deliver an inhibitory signal in conventional T cells, whereas, on regulatory T cells, LAG-3 expression could promote their inhibitory function. In this study, we investigated the role of LAG-3 expression on CD4+ T cells in patients with long bone fracture. We found that LAG-3+ cells represented approximately 13% of peripheral blood CD4+ T cells on average. Compared to LAG-3- CD4+ T cells, LAG-3+ CD4+ T cells presented significantly higher Foxp3 and CTLA-4 expression. Directly ex vivo or with TCR stimulation, LAG-3+ CD4+ T cells expressed significantly higher levels of IL-10 and TGF-ß than LAG-3- CD4+ T cells. Interestingly, blocking the LAG-3-MHC class II interaction actually increased the IL-10 expression by LAG-3+ CD4+ T cells. The frequency of LAG-3+ CD4+ T cell was positively correlated with restoration of healthy bone function in long bone fracture patients. These results together suggested that LAG-3 is a marker of CD4+ T cells with regulatory function; at the same time, LAG-3 might have limited the full suppressive potential of Treg cells.


Asunto(s)
Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Fracturas Óseas/inmunología , Fracturas Óseas/metabolismo , Inmunomodulación , Adulto , Anciano , Antígenos CD/genética , Antígenos de Superficie/metabolismo , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Femenino , Fracturas Óseas/diagnóstico , Fracturas Óseas/genética , Expresión Génica , Humanos , Inmunohistoquímica , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Proteína del Gen 3 de Activación de Linfocitos
5.
Clin Exp Pharmacol Physiol ; 45(5): 430-436, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29215756

RESUMEN

Bone fracture healing is a multistage regenerative process that requires the collaboration of various cell types, with approximately 5%-10% of fractures not healing properly. Accumulating evidence suggests that dysregulations in the immune system are associated with defective healing. In a cohort of 30 bone fracture patients between 50 and 62 years of age, 8 patients displayed delayed healing. Compared to the 22 normal healing patients, these 8 delayed healing patients presented significantly lower frequencies of CD4+ CD25hi Foxp3+ canonical regulatory T cells immediately following bone fracture and early on during the healing process. The CD4+ CD25+/hi T cells from delayed healing patients also presented reduced capacity to express transforming growth factor beta (TGF-ß), and presented reduced surface expression levels of inhibitory molecules, including CTLA-4 and Lag-3, compared to CD4+ CD25+/hi T cells from normal healing patients. Moreover, CD4+ CD25+/hi T cells from delayed healing patients were less potent in the suppression of CD4+ CD25- autologous conventional T cell proliferation, and presented reduced expansion capacity in response to interleukin (IL)-2 stimulation. Overall, our results demonstrated multiple reductions in regulatory T cell function in delayed healing patients that could produce long-lasting consequences in the bone fracture healing process.


Asunto(s)
Regulación hacia Abajo , Curación de Fractura , Linfocitos T Reguladores/citología , Antígenos CD/metabolismo , Antígeno CTLA-4/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Curación de Fractura/efectos de los fármacos , Humanos , Interleucina-2/farmacología , Masculino , Persona de Mediana Edad , Linfocitos T Reguladores/efectos de los fármacos , Proteína del Gen 3 de Activación de Linfocitos
6.
APMIS ; 125(8): 752-760, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28493494

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is a key signaling protein in the skeletal system as well as in the immune system. Accumulating evidence demonstrates that the inflammatory response is deeply involved in the healing process of bone fractures, but how the immune system is regulated during this process is unclear. In this study, we examined STAT3-mediated regulation of immunity in adult patients with closed tibia fracture. In all patients, the expression and activation of STAT3 peaked at around day 7 to day 14 after surgery, and gradually decreased during the rest of the healing period. At day 7 (peak STAT3 expression and phosphorylation), the CD4+ CD25+ T cells from bone fracture patients presented the highest level of STAT3 activation among lymphocyte subsets. Therefore, we investigated the role of STAT3 in CD4+ CD25+ T cells. The level of FOXP3 expression by CD4+ CD25+ T cells was directly correlated with the level of STAT3 phosphorylation in these cells. The level of STAT3 phosphorylation in CD4+ CD25+ T cells was also inversely correlated with the level of IFN-γ and TNF-α secretion in peripheral blood mononuclear cells. Inhibition of STAT3 significantly suppressed FOXP3 and IL-10 expression by CD4+ CD25+ T cells, as well as the ability of CD4+ CD25+ T cells to suppress T-cell IFN-γ and TNF-α secretion. Furthermore, early healers patients presented significantly higher STAT3 expression and phosphorylation than late healers, possibly due to the higher IL-6 and IL-10 levels in the serum of early healing patients. Together, these data demonstrated that STAT3 was beneficial to bone fracture healing, possibly by enhancing Treg-mediated suppression of counteracting inflammations, and suggested that STAT3 could be used as a prognostic marker to identify otherwise undistinguishable patients at risk of developing delayed union or nonunion.


Asunto(s)
Factores de Transcripción Forkhead/biosíntesis , Curación de Fractura , Fracturas Óseas/patología , Regulación de la Expresión Génica , Factor de Transcripción STAT3/metabolismo , Linfocitos T Reguladores/inmunología , Adolescente , Adulto , Anciano , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/inmunología , Femenino , Humanos , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-2/análisis , Masculino , Persona de Mediana Edad , Subgrupos de Linfocitos T/química , Subgrupos de Linfocitos T/inmunología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
7.
Exp Biol Med (Maywood) ; 242(4): 422-428, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-26764266

RESUMEN

The bradykinin B2 receptor (BDKRB2) plays a key role in the inflammation process of osteoarthritis. Nitric oxide has also long been considered to be a catabolic factor that contributes to inflammatory response and the osteoarthritis disease pathology. Several studies have reported that the BDKRB2 +9/-9 bp polymorphisms are associated with transcription of the receptor. However, the roles of BDKRB2 polymorphisms in inflammation in osteoarthritis remain unclear. This study enrolled 156 subjects with primary knee osteoarthritis and 58 healthy volunteers. BDKRB2 polymorphisms were genotyped, and the mRNA and protein levels of BDKRB2 in synovial tissues from osteoarthritis patients were measured by quantitative real-time polymerase chain reaction and western blot analysis, respectively. Nitric oxide production in serum from patients with osteoarthritis was measured using a nitric oxide assay kit. We found that the mean BDKRB2 mRNA levels were significantly higher in Kallgren-Lawrence grade-4 osteoarthritis patients than patients with lower grade osteoarthritis. The +9/-9 bp polymorphisms significantly affected the BDKRB2 mRNA and protein expression levels in synovial tissues from osteoarthritis subjects. Osteoarthritis patients with +9/-9 and -9/-9 genotypes had higher BDKRB2 expression levels in synovial tissue and nitric oxide production in serum. Moreover, positive correlation was found between the BDKRB2 levels in synovial tissue and nitric oxide production. Compared with health controls, significant increases of nitric oxide production in osteoarthritis were detected which were associated with increasing severity of osteoarthritis. Multiple linear regression analysis (adjusted for gender and age) showed serum nitric oxide level was positively associated with BDKRB2 polymorphism and Kallgren-Lawrence grade and was inversely associated with obesity. Our findings showed that the BDKRB2 +9/-9 bp polymorphisms affected the gene expression and nitric oxide production, which were associated with radiographic severity of osteoarthritis, suggesting that the BDKRB2 +9/ -9 bp polymorphisms may act as a genetic modulator of osteoarthritis, and play an essential role in inflammatory process in osteoarthritis.


Asunto(s)
Óxido Nítrico/sangre , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/patología , Receptor de Bradiquinina B2/genética , Membrana Sinovial/citología , Femenino , Humanos , Inflamación/patología , Masculino , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Polimorfismo Genético/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Bradiquinina B2/metabolismo
8.
Clin Exp Pharmacol Physiol ; 44(4): 455-462, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28008635

RESUMEN

Bone fractures may result in delayed union (DU) or non-union (NU) in some patients. Evidence suggests that the skewing of the immune system toward the proinflammatory type is a contributing factor. Because B cells were previously found to infiltrate the fracture healing site at abundant levels, we examined the regulatory B cells (Bregs) in DU/NU patients. In bone fracture patients with normal healing, the frequency of interleukin (IL)-10-expressing B cells was significantly upregulated in the early healing process (6 weeks post-surgery) and was downregulated later on (18 weeks post-surgery), whereas in DU/NU patients, the early upregulation of IL-10-expressing B cells was missing. The majority of IL-10-expressing B cells were concentrated in the IgM+ CD27+ fraction in both controls and patients. IgM+ CD27+ B cells effectively suppressed interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), and IL-2 expression from CD4+ T cells, as well as IFN-γ and TNF-α expression from CD8+ T cells. The IgM+ CD27+ B cell-mediated suppression was restricted to the sample from the early healing time point in controls, as the IgM+ CD27+ B cells from normal healing patients later on or from DU/NU patients did not present significant regulatory function. In addition, culturing of CD4+ CD25+ Tregs with IgM+ CD27+ B cells from controls at early healing time point resulted in higher Foxp3 expression, a function absent in controls at later time point, or in DU/NU patients. In conclusion, our results support a role of B cell-mediated regulation early during the bone healing process.


Asunto(s)
Linfocitos B Reguladores/metabolismo , Citocinas/metabolismo , Factores de Transcripción Forkhead/metabolismo , Curación de Fractura , Fracturas Óseas/inmunología , Fracturas Óseas/fisiopatología , Linfocitos T Reguladores/metabolismo , Linfocitos B Reguladores/inmunología , Femenino , Regulación de la Expresión Génica , Humanos , Interleucina-10/metabolismo , Masculino , Factor de Crecimiento Transformador beta/metabolismo
9.
Cell Physiol Biochem ; 38(3): 1245-56, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26982743

RESUMEN

BACKGROUND/AIMS: The bradykinin B2 receptor (BDKRB2) +9/-9 gene polymorphisms have been shown to be associated with the susceptibility and severity of osteoarthritis (OA); however, the underlying mechanisms are unclear. In this study, we investigated the correlation between the BDKRB2 +9/-9 polymorphisms and pro-inflammatory cytokine levels in OA and the molecular mechanisms involved. METHODS: A total of 156 patients with primary knee OA and 121 healthy controls were enrolled. The BDKRB2 +9/-9 polymorphisms were genotyped. The tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, and IL-8 levels were determined using Enzyme-linked immunosorbent assay (ELISA). The toll-like receptor (TLR)-2 and TLR-4 mRNA levels were determined by quantitative real-time PCR. The basal and bradykinin-stimulated pro-inflammatory cytokine secretion in human OA synoviocytes and the involvement of TLR-2 and mitogen-activated protein kinases (MAPKs) were investigated. RESULTS: The presence of -9 bp genotype is associated with higher TNF-α, IL-6, and IL-8 levels and higher TLR-2 expression in OA patients. The basal and bradykinin-induced TLR-2 expressions in human OA synoviocytes were significantly reduced by specific inhibitors of p38, JNK1/2, and ERK1/2. Both the B2 receptor antagonist MEN16132 and TLR-2 silencing inhibited IL-6 and IL-8 secretion in human OA synoviocytes. CONCLUSION: The data suggested that the BDKRB2 +9/-9 polymorphisms influence pro-inflammatory cytokine levels in knee osteoarthritis by altering TLR-2 expression.


Asunto(s)
Bradiquinina/farmacología , Citocinas/metabolismo , Osteoartritis de la Rodilla/genética , Receptor de Bradiquinina B2/genética , Sinoviocitos/efectos de los fármacos , Receptor Toll-Like 2/genética , Anciano , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Sinoviocitos/citología , Sinoviocitos/metabolismo
10.
Cell Tissue Res ; 363(2): 427-47, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26004143

RESUMEN

Aseptic loosening secondary to periprosthetic inflammatory osteolysis results from the biological response to wear particles and is a leading cause of arthroplasty failure. The origin of this inflammatory response remains unclear. We aim to validate the definite link between endoplasmic reticulum (ER) stress and particle-induced inflammatory signaling pathways in periprosthetic osteolysis. We examine the histopathologic changes of osteolysis and the expression of specific biomarkers for ER-stress-mediated inflammatory signaling pathways (IRE1α, GRP78/Bip, c-Fos, NF-κB, ROS and Ca(2+)). Moreover, pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6) and osteoclastogenic molecules (VEGF, OPG, RANKL and M-CSF) were assessed in clinical interface membranes and murine periosteum tissues. We found wear particles to be capable of inducing ER stress in macrophages within clinical osteolytic interface membranes and murine osteolytic periosteum tissues and to be associated with the inflammatory response and osteoclastogenesis. Blocking ER stress with sodium 4-phenylbutyrate (4-PBA) results in a dramatic amelioration of particle-induced osteolysis and a significant reduction of ER-stress intensity. Simultaneously, this ER-stress blocker also lessens inflammatory cell infiltration, diminishes the capability of osteoclastogenesis and reduces the inflammatory response by lowering IRE1α, GRP78/Bip, c-Fos, NF-κB, ROS and Ca(2+) levels. Thus, ER stress plays an important role in particle-induced inflammatory osteolysis and osteoclastogenic reactions. The pharmacological targeting of ER-stress-mediated inflammatory signaling pathways might be an appealing approach for alleviating or preventing particle-induced osteolysis in at-risk patients.


Asunto(s)
Estrés del Retículo Endoplásmico , Inflamación/patología , Nanopartículas/efectos adversos , Osteólisis/etiología , Osteólisis/patología , Periostio/patología , Transducción de Señal , Adolescente , Anciano , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Chaperón BiP del Retículo Endoplásmico , Femenino , Humanos , Inflamación/complicaciones , Mediadores de Inflamación/metabolismo , Masculino , Membranas , Ratones , Persona de Mediana Edad , FN-kappa B/metabolismo , Osteoclastos/patología , Osteólisis/complicaciones , Osteólisis/diagnóstico por imagen , Osteoprotegerina/metabolismo , Periostio/diagnóstico por imagen , Prótesis e Implantes/efectos adversos , Falla de Prótesis , Ligando RANK/metabolismo , Radiografía , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
J Mol Histol ; 46(6): 467-73, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26514989

RESUMEN

Mesenchymal stem cells (MSCs) have several features that make them an attractive option for potentiating cartilage repair. Synovium-derived (SMSCs) have been recently recognized as an excellent source. SRY-related HMG-box (Sox) family plays an important role in the proliferation and differentiation of SMSCs. However, the role of Sox4 in human SMSCs remains elusive. In the present study, we investigated the role of Sox4 in SMSCs through gain-of-function studies and found that Sox4 promoted cell proliferation and chondrogenesis. Furthermore, Sox4 could directly bind to the promoter of long noncoding RNA DANCR and increased its expression. Finally, knockdown of DANCR could reverse the stimulative effect of Sox4 on the proliferation and chondrogenesis of SMSCs. Taken together, our data highlights the pivotal role of Sox4 in the proliferation and differentiation of SMSCs.


Asunto(s)
Diferenciación Celular/genética , Condrogénesis/genética , ARN Largo no Codificante/genética , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo , Células Madre/citología , Células Madre/metabolismo , Membrana Sinovial/citología , Proliferación Celular , Células Cultivadas , Expresión Génica , Regulación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Interferencia de ARN , Activación Transcripcional
12.
Cell Biochem Biophys ; 71(2): 741-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25257130

RESUMEN

Osteoclast is the only cell that can degrade bone tissue in vivo. Recent studies have shown the important role of cytoskeleton dynamics in osteolysis and the formation of podosome belt in osteoclasts. This process is regulated by the dynamic microtubule (MT) network. We treated osteoclast precursor cells Raw264.7 with low concentration of nocodazole (10 nM) and antineoplastic drug taxol (10 nM) to block MT turnover, and used end binding protein 1 fused to GFP to track the movement of microtubules in induced osteoclasts. We show that low concentrations of nocodazole and taxol interfere with the formation of podosome belt, and reduce TRAP activity of induced osteoclasts. These results suggest that the effect of taxol on MT dynamics may be used clinically to reduce osteoclast activity and potentially prevent development of osteoporosis and other metabolic bone diseases.


Asunto(s)
Microtúbulos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Fosfatasa Ácida/metabolismo , Animales , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Relación Dosis-Respuesta a Droga , Isoenzimas/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/efectos de los fármacos , Nocodazol/farmacología , Osteoclastos/efectos de los fármacos , Paclitaxel/farmacología , Ligando RANK/farmacología , Fosfatasa Ácida Tartratorresistente
13.
Zhongguo Gu Shang ; 27(6): 529-32, 2014 Jun.
Artículo en Chino | MEDLINE | ID: mdl-25241478

RESUMEN

Osteoclast, a huge coenocytes,originates from mononuclear macrophages or monocytic series hematopoietic precursor cell, plays an important role in the progree of bone resorption. Formation and abnormal activity of osteoclast may cause osteoprosis, rheumatoid arthritis and aseptic loosening after arthroplasty. Therefore, osteoclast is the target for treating these disease. At present, a lot of study on formation of osteoclast were reported, but the study on how to identify and degradation of bone tissue is not yet reported. Bone mineral are seen as important component of identifing osteoclast, and the research suggested that bone matrix is not the essential ingredients of activiting osteoclast, petri dish covered by vitronectin also can make osteoclast occure certain form of bone resorption, vitronectin plays an significant role in activiting osteoclast. Otherwise, the research found that swallowing and secretion of bone matrix degradation products is benefit for differentiation of osteoclast and maintain of function, and this may be therapeutic target for treatment of musculoskeletal disorders.


Asunto(s)
Resorción Ósea , Osteoclastos/fisiología , Animales , Matriz Ósea/metabolismo , Humanos
14.
Cell Biochem Biophys ; 70(3): 1671-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24972690

RESUMEN

To investigate the effects of the HDMX polymorphism on sarcoma risk. Relevant studies were identified by searching the PubMed, Embase, and Web of Science databases. Data were extracted by two independent investigators. Odds ratios (ORs) and 95 % confidence intervals (CIs) were calculated using a fixed-effects model to assess the association between the HDMX polymorphism and sarcoma risk. We also conducted heterogeneity test, sensitivity analysis, and publication bias test. A meta-analysis of four published case-control studies involving 1,115 subjects (379 cases and 736 controls) showed no statistical association between the HDMX polymorphism and sarcoma risk (ORTT vs. GG 0.88, 95 % CI 0.68-1.14, P heterogeneity 0.819; ORTT + TG vs. GG 0.95, 95 % CI 0.79-1.15, P heterogeneity 0.937; ORTT vs. TG + GG 0.82, 95 % CI 0.65-1.04, P heterogeneity 0.589; ORT allele vs. G allele 0.91, 95 % CI 0.79-1.05, P heterogeneity 0.727; ORTG vs. GG 0.95, 95 % CI 0.74-1.22, P heterogeneity = 0.869). This null result did not alter when data were stratified according to ethnicity. Our meta-analysis indicates that the HDMX polymorphism is unlikely to contribute to individual susceptibility to sarcoma.


Asunto(s)
Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/genética , Sarcoma/genética , Alelos , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Estudios de Casos y Controles , Proteínas de Ciclo Celular , Bases de Datos Factuales , Femenino , Genotipo , Humanos , Oportunidad Relativa , Polimorfismo de Nucleótido Simple , Sarcoma/patología , Neoplasias Uterinas/genética , Neoplasias Uterinas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...