Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Anal Bioanal Chem ; 412(26): 7253-7262, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32879994

RESUMEN

Fourier transform infrared (FTIR) microspectroscopy provides a biochemical fingerprint of the cells. In this study, chemical changes in 143B osteosarcoma cells were investigated using FTIR analysis of cancer cells after their treatment with polymeric invertible micellar assemblies (IMAs) and curcumin-loaded IMAs and compared with untreated osteosarcoma cells. A comprehensive principal component analysis (PCA) was applied to analyze the FTIR results and confirm noticeable changes in cell surface chemical structures in the fingerprint regions of 1480-900 cm-1. The performed clustering shows visible differences for all investigated groups of cancer cells. It is demonstrated that a combination of FTIR microspectroscopy with PCA can be an efficient approach in determining interactions of osteosarcoma cells and drug-loaded polymer micellar assemblies. Graphical abstract.


Asunto(s)
Neoplasias Óseas/patología , Osteosarcoma/patología , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Línea Celular Tumoral , Humanos
2.
Chin J Cancer Res ; 31(1): 203-211, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30996578

RESUMEN

OBJECTIVE: To examine the role of store-operated calcium entry (SOCE) and stromal interaction molecule 1 (STIM1) in survival and migration of osteosarcoma cells and investigate what blockade of store-operated Ca2+ contributes to the regulation of osteosarcoma cells. METHODS: First, we examined the expression levels of STIM1 in osteosarcoma cell lines by Western analysis and in tissue specimens by immunohistochemistry. Second, we investigated the effect of SOCE and STIM1 on osteosarcoma cell viability using MTS assays and on cell proliferation using colony formation. Third, we investigated the role of SOCE and STIM1 in cell migration using wound healing assays and Boyden chamber assays. Finally, we studied the effect of SOCE on the nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) activity by luciferase assays. RESULTS: STIM1 was overexpressed in osteosarcoma cell lines and tissue specimens and was associated with poor survival of osteosarcoma patients. Also, inhibition of SOCE and STIM1 decreased the cell viability and migration of osteosarcoma cells. Furthermore, our results showed that blockade of store-operated Ca2+ channels involved down-regulation of NFATc1 in osteosarcoma cells. CONCLUSIONS: STIM1 is essential for osteosarcoma cell functions, and STIM1 and Ca2+ entry pathway could be further explored as molecular targets in the treatment of osteosarcoma.

3.
J Cell Biochem ; 118(6): 1497-1504, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27883247

RESUMEN

Osteosarcoma is a bone tumor that mainly affects children and adolescents. Although its pathogenesis is still not fully understood, activation of Wnt signaling has been implicated in the development and metastasis of osteosarcoma. In this report, we have investigated the effect of the anti-tumor compound, 2-methoxyestradiol (2-ME) on Wnt antagonist frizzled-related protein b (Frzb), also known as secreted frizzled-related protein (sFRP)3 in human osteosarcoma (MG63) cells. Our results show that 2-ME treatment induces Frzb gene promoter activity, and increases Frzb mRNA and protein levels in osteosarcoma cells. In addition, 2-ME treatment regulates downstream Wnt signaling, increasing the cytoplasmic levels of ß-catenin, and blocking ß-catenin-mediated Wnt activation in osteosarcoma cells. 2-ME-mediated induction of Frzb protein expression is specific to osteosarcoma cells, as it does not affect Frzb expression in normal primary human osteoblasts. Furthermore, 2-ME-induced apoptosis and autophagy are blocked in osteosarcoma cells transfected with Frzb siRNAs. Taken together, these studies demonstrate that Frzb protein plays an important role in 2-ME-mediated anti-tumor mechanisms in osteosarcoma cells. J. Cell. Biochem. 118: 1497-1504, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/genética , Estradiol/análogos & derivados , Glicoproteínas/genética , Osteosarcoma/genética , 2-Metoxiestradiol , Autofagia , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Estradiol/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glicoproteínas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
4.
Biochem Biophys Res Commun ; 486(2): 307-313, 2017 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-28300555

RESUMEN

Osteosarcoma survival rate has not improved over the past three decades, and the debilitating side effects of the surgical treatment suggest the need for alternative local control approaches. Radiotherapy is largely ineffective in osteosarcoma, indicating a potential role for radiosensitizers. Blocking DNA repair, particularly by inhibiting the catalytic subunit of DNA-dependent protein kinase (DNA-PKCS), is an attractive option for the radiosensitization of osteosarcoma. In this study, the expression of DNA-PKCS in osteosarcoma tissue specimens and cell lines was examined. Moreover, the small molecule DNA-PKCS inhibitor, KU60648, was investigated as a radiosensitizing strategy for osteosarcoma cells in vitro. DNA-PKCS was consistently expressed in the osteosarcoma tissue specimens and cell lines studied. Additionally, KU60648 effectively sensitized two of those osteosarcoma cell lines (143B cells by 1.5-fold and U2OS cells by 2.5-fold). KU60648 co-treatment also altered cell cycle distribution and enhanced DNA damage. Cell accumulation at the G2/M transition point increased by 55% and 45%, while the percentage of cells with >20 γH2AX foci were enhanced by 59% and 107% for 143B and U2OS cells, respectively. These results indicate that the DNA-PKCS inhibitor, KU60648, is a promising radiosensitizing agent for osteosarcoma.


Asunto(s)
Neoplasias Óseas/terapia , Cromonas/farmacología , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/antagonistas & inhibidores , Osteosarcoma/terapia , Inhibidores de Proteínas Quinasas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias Óseas/enzimología , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Cromonas/química , Cromonas/metabolismo , Daño del ADN , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Rayos gamma/uso terapéutico , Histonas/genética , Histonas/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteosarcoma/enzimología , Osteosarcoma/genética , Osteosarcoma/patología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/metabolismo , Análisis de Secuencia de ARN
5.
J Cell Biochem ; 117(3): 751-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26335692

RESUMEN

Chondrosarcoma is a cartilage tumor and is the second most common malignant bone cancer. Unlike many tumors, chondrosarcomas are resistant to conventional chemotherapy and radiotherapy. Autophagy is a homeostatic mechanism through which cellular proteins and organelles are subjected to lysosomal degradation and recycling. Autophagy could play a dual role in cancer by facilitating either cell death or cell survival. To determine whether autophagy plays a role in cell death in chondrosarcoma, we have studied the effect of the anti-tumor compound 2-methoxyestradiol (2-ME) in chondrosarcoma cells in culture. Transmission electron microscopy imaging indicates that 2-ME treatment leads to the accumulation of autophagosomes in human chondrosarcoma (SW1353 and Hs819T) cells. Also, 2-ME induces the conversion of microtubule-associated protein LC3-I to LC3-II, a protein marker that is correlated with the formation of autophagosomes. Our results show that siRNAs directed against ATG3 blocks 2-ME-induced autophagosome formation in chondrosarcoma cells. In addition, treatment with Bafilomycin A1 (Baf) and 3-methyladenine (3-MA), the inhibitors of autophagy, further increased the cell death in 2-ME-treated chondrosarcoma cells. Taken together, our studies demonstrate that autophagy causes resistance to cytotoxicity in chondrosarcoma cells, and the efficacy and anti-tumor effects of drugs in chondrosarcoma could be enhanced by modulating autophagy.


Asunto(s)
Antineoplásicos/farmacología , Autofagia , Estradiol/análogos & derivados , 2-Metoxiestradiol , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Condrosarcoma/tratamiento farmacológico , Ensayos de Selección de Medicamentos Antitumorales , Estradiol/farmacología , Humanos , Transducción de Señal
6.
J Biol Chem ; 288(29): 21307-21319, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23720736

RESUMEN

Osteosarcoma (OS) is a primary bone tumor that is most prevalent during adolescence. RUNX2, which stimulates differentiation and suppresses proliferation of osteoblasts, is deregulated in OS. Here, we define pathological roles of RUNX2 in the etiology of OS and mechanisms by which RUNX2 expression is stimulated. RUNX2 is often highly expressed in human OS biopsies and cell lines. Small interference RNA-mediated depletion of RUNX2 inhibits growth of U2OS OS cells. RUNX2 levels are inversely linked to loss of p53 (which predisposes to OS) in distinct OS cell lines and osteoblasts. RUNX2 protein levels decrease upon stabilization of p53 with the MDM2 inhibitor Nutlin-3. Elevated RUNX2 protein expression is post-transcriptionally regulated and directly linked to diminished expression of several validated RUNX2 targeting microRNAs in human OS cells compared with mesenchymal progenitor cells. The p53-dependent miR-34c is the most significantly down-regulated RUNX2 targeting microRNAs in OS. Exogenous supplementation of miR-34c markedly decreases RUNX2 protein levels, whereas 3'-UTR reporter assays establish RUNX2 as a direct target of miR-34c in OS cells. Importantly, Nutlin-3-mediated stabilization of p53 increases expression of miR-34c and decreases RUNX2. Thus, a novel p53-miR-34c-RUNX2 network controls cell growth of osseous cells and is compromised in OS.


Asunto(s)
Neoplasias Óseas/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , MicroARNs/metabolismo , Osteosarcoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Regulación hacia Abajo/genética , Regulación hacia Abajo/efectos de la radiación , Rayos gamma , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Ratones , Osteosarcoma/genética , Osteosarcoma/patología , Estabilidad Proteica/efectos de la radiación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/deficiencia
7.
BMC Cancer ; 12: 93, 2012 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-22429849

RESUMEN

BACKGROUND: Osteosarcoma is a bone tumor that often affects children and young adults. Although a combination of surgery and chemotherapy has improved the survival rate in the past decades, local recurrence and metastases still develop in 40% of patients. A definite therapy is yet to be determined for osteosarcoma. Anti- tumor compound and a metabolite of estrogen, 2-methoxyestradiol (2-ME) induces cell death in osteosarcoma cells. In this report, we have investigated whether interferon (IFN) pathway is involved in 2-ME-induced anti-tumor effects in osteosarcoma cells. METHODS: 2-ME effects on IFN mRNA levels were determined by Real time PCR analysis. Transient transfections followed by reporter assays were used for investigating 2-ME effects on IFN-pathway. Western blot analyses were used to measure protein and phosphorylation levels of IFN-regulated eukaryotic initiation factor-2 alpha (eIF-2α). RESULTS: 2-ME regulates IFN and IFN-mediated effects in osteosarcoma cells. 2 -ME induces IFN gene activity and expression in osteosarcoma cells. 2-ME treatment induced IFN-stimulated response element (ISRE) sequence-dependent transcription and gamma-activated sequence (GAS)-dependent transcription in several osteosarcoma cells. Whereas, 2-ME did not affect IFN gene and IFN pathways in normal primary human osteoblasts (HOB). 2-ME treatment increased the phosphorylation of eIF-2α in osteosarcoma cells. Furthermore, analysis of osteosarcoma tissues shows that the levels of phosphorylated form of eIF-2α are decreased in tumor compared to normal controls. CONCLUSIONS: 2-ME treatment triggers the induction and activity of IFN and IFN pathway genes in 2-ME-sensitive osteosarcoma tumor cells but not in 2-ME-resistant normal osteoblasts. In addition, IFN-signaling is inhibited in osteosarcoma patients. Thus, IFN pathways play a role in osteosarcoma and in 2-ME-mediated anti-proliferative effects, and therefore targeted induction of IFN signaling could lead to effective treatment strategies in the control of osteosarcoma.


Asunto(s)
Neoplasias Óseas/metabolismo , Estradiol/análogos & derivados , Interferones/metabolismo , Osteosarcoma/metabolismo , Transducción de Señal/efectos de los fármacos , 2-Metoxiestradiol , Neoplasias Óseas/genética , Línea Celular Tumoral , Estradiol/farmacología , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interferones/genética , Osteosarcoma/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
8.
Genes Chromosomes Cancer ; 50(11): 849-58, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21793095

RESUMEN

Well-differentiated liposarcoma (WDLS) is one of the most common malignant mesenchymal tumors and dedifferentiated liposarcoma (DDLS) is a malignant tumor consisting of both WDLS and a transformed nonlipogenic sarcomatous component. Cytogenetically, WDLS is characterized by the presence of ring or giant rod chromosomes containing several amplified genes, including MDM2, TSPAN31, CDK4, and others mainly derived from chromosome bands 12q13-15. However, the 12q13-15 amplicon is large and discontinuous. The focus of this study was to identify novel critical genes that are consistently amplified in primary (nonrecurrent) WDLS and with potential relevance for future targeted therapy. Using a high-resolution (5.0 kb) "single nucleotide polymorphism"/copy number variation microarray to screen the whole genome in a series of primary WDLS, two consistently amplified areas were found on chromosome 12: one region containing the MDM2 and CPM genes, and another region containing the FRS2 gene. Based on these findings, we further validated FRS2 amplification in both WDLS and DDLS. Fluorescence in situ hybridization confirmed FRS2 amplification in all WDLS and DDLS tested (n = 57). Real time PCR showed FRS2 mRNA transcriptional upregulation in WDLS (n = 19) and DDLS (n = 13) but not in lipoma (n = 5) and normal fat (n = 9). Immunoblotting revealed high expression levels of phospho-FRS2 at Y436 and slightly overexpression of total FRS2 protein in liposarcoma but not in normal fat or preadipocytes. Considering the critical role of FRS2 in mediating fibroblast growth factor receptor signaling, our findings indicate that FRS2 signaling should be further investigated as a potential therapeutic target for liposarcoma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Liposarcoma/genética , Liposarcoma/patología , Proteínas de la Membrana/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Western Blotting , Estudios de Casos y Controles , Diferenciación Celular/genética , Mapeo Cromosómico , Cromosomas Humanos Par 12 , Variaciones en el Número de Copia de ADN , Proteínas Ligadas a GPI/genética , Amplificación de Genes , Humanos , Hibridación Fluorescente in Situ , Liposarcoma/metabolismo , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-mdm2/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Reproducibilidad de los Resultados , Estudios Retrospectivos , Transducción de Señal
9.
Gene ; 802: 145865, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34352301

RESUMEN

Osteosarcoma is a bone tumor that mainly affects children and adolescents. Interferons (IFNs) have been shown to exert antitumor effects in osteosarcoma cells, although the molecular mechanisms have not been fully realized. We investigated IFN-γ actions on osteosarcoma cells. Our results show that IFN-γ induces the accumulation of autophagosomes in osteosarcoma cells. IFN-γ treatment leads to the conversion of autophagy marker light chain 3 (LC3)-I to LC3-II in osteosarcoma cells, and this conversion is accompanied by puncta formation. Also, IFN-γ-mediated induction of autophagosome formation and autophagic flux require RNA-dependent protein kinase (PKR) activity. In addition, our findings show that IFN-γ-mediated osteosarcoma cell death is not dependent on PKR. Our study suggests that IFN-γ has differential effects that lead to induction of cell death and autophagy in osteosarcoma cells. Further evaluation of the IFN-γ-mediated molecular mechanism could lead to improved understanding of and targeted treatment strategies for osteosarcoma.


Asunto(s)
Autofagia , Neoplasias Óseas/enzimología , Interferón gamma/metabolismo , Osteosarcoma/enzimología , eIF-2 Quinasa/metabolismo , Proteína 7 Relacionada con la Autofagia/metabolismo , Neoplasias Óseas/metabolismo , Osteosarcoma/metabolismo , Células Tumorales Cultivadas
10.
J Cell Biochem ; 109(5): 950-6, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20082321

RESUMEN

Osteosarcoma is a bone tumor that frequently develops during adolescence. 2-Methoxyestradiol (2-ME), a naturally occurring metabolite of 17beta-estradiol, induces cell cycle arrest and cell death in human osteosarcoma cells. To investigate whether the osteoprotegrin (OPG) protein plays a role in 2-ME actions, we studied the effect of 2-ME treatment on OPG gene expression in human osteosarcoma cells. 2-ME treatment induced OPG gene promoter activity and mRNA levels. Also, Western blot analysis showed that 2-ME treatment increased OPG protein levels in MG63, KHOS, 143B and LM7 osteosarcoma cells by 3-, 1.9-, 2.8-, and 2.5-fold, respectively, but did not affect OPG expression in normal bone cells. In addition, increases in OPG protein levels were observed in osteosarcoma cell culture media after 3 days of 2-ME treatment. The effect of 2-ME on osteosarcoma cells was ligand-specific as parent estrogen, 17beta-estradiol and a tumorigenic estrogen metabolite, 16alpha-hydroxyestradiol, which do not affect osteosarcoma cell cycle and cell death, had no effect on OPG protein expression. Furthermore, co-treating osteosarcoma cells with OPG protein did not further enhance 2-ME-mediated anti-tumor effects. OPG-released in 2-ME-treated cultures led to an increase in osteoblastic activity and a decrease in osteoclast number, respectively. These findings suggest that OPG is not directly involved in 2-ME-mediated anti-proliferative effects in osteosarcoma cells, but rather participates in anti-resorptive functions of 2-ME in bone tumor environment.


Asunto(s)
Estradiol/análogos & derivados , Osteoprotegerina/metabolismo , Osteosarcoma/metabolismo , 2-Metoxiestradiol , Fosfatasa Alcalina/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Estradiol/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/enzimología , Osteoprotegerina/genética , Osteosarcoma/genética , Osteosarcoma/patología , Péptidos/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Transcripción Genética/efectos de los fármacos
11.
Diagnostics (Basel) ; 10(2)2020 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-32023980

RESUMEN

Accurate clinical evaluation of renal osteodystrophy (ROD) is currently accomplished using invasive in vivo transiliac bone biopsy, followed by in vitro histomorphometry. In this study, we demonstrate that an alternative method for ROD assessment is through a fast, label-free Raman recording of multiple biomarkers combined with computational analysis for predicting the minimally required number of spectra for sample classification at defined accuracies. Four clinically relevant biomarkers: the mineral-to-matrix ratio, the carbonate-to-matrix ratio, phenylalanine, and calcium contents were experimentally determined and simultaneously considered as input to a linear discriminant analysis (LDA). Additionally, sample evaluation was performed with a linear support vector machine (LSVM) algorithm, with a 300 variable input. The computed probabilities based on a single spectrum were only marginally different (~80% from LDA and ~87% from LSVM), both providing an unacceptable classification power for a correct sample assignment. However, the Type I and Type II assignment errors confirm that a relatively small number of independent spectra (7 spectra for Type I and 5 spectra for Type II) is necessary for a p < 0.05 error probability. This low number of spectra supports the practicality of future in vivo Raman translation for a fast and accurate ROD detection in clinical settings.

12.
Cancer Invest ; 27(4): 435-42, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19212826

RESUMEN

There is no animal model that reflects the histological and radiographical heterogeneity of osteosarcoma. We assessed seven osteosarcoma cell lines for their potential to develop orthotopic tumors and lung metastasis in SCID mice. Whereas radiologically, 143B developed osteolytic tumors, SaOS-LM7 developed osteoblastic primary tumors. The mineralization status was confirmed by assessing the alkaline phosphatase activity and the microarray expression profile. We herein report a xenograft orthotopic osteosarcoma mouse model to assess osteoblastic and osteolytic lesions, which may contribute in the search for new diagnostic and therapeutic approaches.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias Pulmonares/secundario , Osteoblastos/patología , Osteólisis/patología , Osteosarcoma/patología , Tibia/patología , Fosfatasa Alcalina/metabolismo , Animales , Densidad Ósea , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/enzimología , Neoplasias Óseas/genética , Línea Celular Tumoral , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/diagnóstico por imagen , Osteoblastos/enzimología , Osteólisis/diagnóstico por imagen , Osteólisis/enzimología , Osteólisis/genética , Osteosarcoma/diagnóstico por imagen , Osteosarcoma/enzimología , Osteosarcoma/genética , Radiografía , Tibia/diagnóstico por imagen , Tibia/enzimología , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Endocrinology ; 149(11): 5735-46, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18635661

RESUMEN

Abnormal secretion of PTH by the parathyroid glands contributes to a variety of common skeletal disorders. Prior studies implicate platelet-derived growth factor-A (PDGF-A) as an important mediator of selective PTH actions on bone. The present studies used targeted gene profiling and small-molecule antagonists directed against candidate gene products to elucidate the roles of specific PTH-regulated genes and signaling pathways. A group of 29 genes in rats continuously infused with PTH and cotreated with the PDGF receptor antagonist trapidil were differentially expressed compared with PTH treatment alone. Several of the identified genes were functionally clustered as regulators of fibroblast differentiation and extracellular matrix modeling, including the matrix cross-linking enzyme lysyl oxidase (LOX). Treatment with beta-aminopropionitrile, an irreversible inhibitor of LOX activity, dramatically reduced diffuse mineralization but had no effect on PTH-induced fibrosis. In contrast, the receptor tyrosine kinase inhibitor Gleevec and the phosphoinositide 3-kinase inhibitor wortmannin each reduced bone marrow fibrosis. In summary, the present studies support the hypotheses that PTH-induced bone marrow fibrosis is mediated by PDGF-A via a phosphoinositide 3-kinase-dependent signaling pathway and that increased LOX gene expression plays a key role in abnormal mineralization, a hallmark of chronic hyperparathyroidism.


Asunto(s)
Hiperparatiroidismo/complicaciones , Osteítis Fibrosa Quística/etiología , Fosfatidilinositol 3-Quinasas/fisiología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Animales , Enfermedad Crónica , Análisis por Conglomerados , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Hiperparatiroidismo/genética , Hiperparatiroidismo/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteítis Fibrosa Quística/genética , Hormona Paratiroidea/administración & dosificación , Hormona Paratiroidea/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
14.
J Cell Biochem ; 104(5): 1937-45, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18384113

RESUMEN

2-Methoxyestradiol (2-ME), a naturally occurring mammalian metabolite of 17beta-Estradiol (E2), induces cell death in osteosarcoma cells. To further understand the molecular mechanisms of action, we have investigated cell cycle progression in 2-ME-treated human osteosarcoma (MG63, SaOS-2 and LM7 [corrected]) cells. At 5 microM, 2-ME induced growth arrest by inducing a block in cell cycle; 2-ME-treatment resulted in 2-fold increases in G1 phase cells and a decrease in S phase cells in MG63 and SaOS-2 osteosarcoma cell lines, compared to the appropriate vehicle controls. 2-ME-treatment induced a threefold increase in the G2 phase in LM7 [corrected] osteosarcoma cells. The results demonstrated steroid specificity, as the tumorigenic metabolite, 16alpha-hydroxyestradiol (16-OHE), did not have any effect on cell cycle progression in osteosarcoma cells. The cell cycle arrest coincided with an increase in expression of the cell cycle markers p21, p27 and p53 proteins in 2-ME-treated osteosarcoma cells. Also, MG63 cells, transiently transfected with cDNA for a 'loss of function mutant' RNA-dependent protein kinase (PKR) protein, were resistant to 2-ME-induced cell cycle arrest. These results suggest that 2-ME works in concert with factors regulating cell cycle progression, and cell cycle arrest precedes cell death in 2-ME-treated osteosarcoma cells.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Estradiol/análogos & derivados , Osteosarcoma/patología , 2-Metoxiestradiol , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Estradiol/farmacología , Citometría de Flujo , Genes Dominantes , Humanos , Ligandos , Proteínas Mutantes/metabolismo , Osteosarcoma/enzimología , eIF-2 Quinasa/metabolismo
15.
Biomaterials ; 29(22): 3245-52, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18472153

RESUMEN

In this study, we investigated the in vitro and in vivo biological activities of bone morphogenetic protein 2 (BMP-2) released from four sustained delivery vehicles for bone regeneration. BMP-2 was incorporated into (1) a gelatin hydrogel, (2) poly(lactic-co-glycolic acid) (PLGA) microspheres embedded in a gelatin hydrogel, (3) microspheres embedded in a poly(propylene fumarate) (PPF) scaffold and (4) microspheres embedded in a PPF scaffold surrounded by a gelatin hydrogel. A fraction of the incorporated BMP-2 was radiolabeled with (125)I to determine its in vitro and in vivo release profiles. The release and bioactivity of BMP-2 were tested weekly over a period of 12 weeks in preosteoblast W20-17 cell line culture and in a rat subcutaneous implantation model. Outcome parameters for in vitro and in vivo bioactivities of the released BMP-2 were alkaline phosphatase (AP) induction and bone formation, respectively. The four implant types showed different in vitro release profiles over the 12-week period, which changed significantly upon implantation. The AP induction by BMP-2 released from gelatin implants showed a loss in bioactivity after 6 weeks in culture, while the BMP-2 released from the other implants continued to show bioactivity over the full 12-week period. Micro-CT and histological analysis of the delivery vehicles after 6 weeks of implantation showed significantly more bone in the microsphere/PPF scaffold composites (Implant 3, p<0.02). After 12 weeks, the amount of newly formed bone in the microsphere/PPF scaffolds remained significantly higher than that in the gelatin and microsphere/gelatin hydrogels (p<0.001), however, there was no statistical difference compared to the microsphere/PPF/gelatin composite. Overall, the results from this study show that BMP-2 could be incorporated into various bone tissue engineering composites for sustained release over a prolonged period of time with retention of bioactivity.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacocinética , Microesferas , Ingeniería de Tejidos/métodos , Factor de Crecimiento Transformador beta/farmacocinética , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/administración & dosificación , Proteínas Morfogenéticas Óseas/farmacología , Huesos/citología , Huesos/efectos de los fármacos , Línea Celular , Fumaratos/química , Gelatina/química , Hidrogeles/química , Ácido Láctico/química , Masculino , Ratones , Osteogénesis/efectos de los fármacos , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polipropilenos/química , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta/administración & dosificación , Factor de Crecimiento Transformador beta/farmacología
16.
Biochem Pharmacol ; 150: 141-149, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29412166

RESUMEN

Osteosarcoma is the most common bone tumor that affects children and young adults. Despite advances in the use of combination chemotherapy regimens, response to neoadjuvant chemotherapy in osteosarcoma remains a key determinant of patient outcome. Recently, highly potent small molecule inhibitors of canonical Wnt signaling through the poly(ADP-ribose) polymerase (PARP)-family enzymes, tankyrases 1 & 2 (Tnks1/2), have been considered as possible chemotherapy sensitizing agents. The goal of this study was to determine the ability of the highly specific Tnks1/2 inhibitor IWR-1-endo to sensitize chemotherapy-resistant osteosarcoma to doxorubicin. We found that IWR-1-endo significantly inhibited cellular efflux, as measured by cellular retention of Calcein AM and doxorubicin. In a model of doxorubicin resistant osteosarcoma, pre-treatment with IWR-1-endo strongly sensitized to doxorubicin. This sensitization reduced the doxorubicin IC50 in doxorubicin-resistant cells, but not in chemotherapy naïve cells and caused doxorubicin-treated cells to accumulate at the G2/M checkpoint. Further, we found that sensitization with IWR-1-endo produced increased γH2AX foci formation, indicating increased DNA damage by doxorubicin. Taken together, our findings show that IWR-1-endo increases cellular responses to doxorubicin, by blocking efflux transport in a drug-resistant model of osteosarcoma.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Óseas/metabolismo , Doxorrubicina/administración & dosificación , Resistencia a Antineoplásicos/efectos de los fármacos , Imidas/administración & dosificación , Osteosarcoma/metabolismo , Quinolinas/administración & dosificación , Antibióticos Antineoplásicos/administración & dosificación , Neoplasias Óseas/tratamiento farmacológico , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/fisiología , Sinergismo Farmacológico , Humanos , Osteosarcoma/tratamiento farmacológico , Tanquirasas/antagonistas & inhibidores , Tanquirasas/metabolismo
17.
Biosensors (Basel) ; 8(2)2018 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-29642494

RESUMEN

Defining the pathogenesis of renal osteodystrophy (ROD) and its treatment efficacy are difficult, since many factors potentially affect bone quality. In this study, confocal Raman microscopy and parallel statistical analysis were used to identify differences in bone composition between healthy and ROD bone tissues through direct visualization of three main compositional parametric ratios, namely, calcium content, mineral-to-matrix, and carbonate-to-matrix. Besides the substantially lower values found in ROD specimens for these representative ratios, an obvious accumulation of phenylalanine is Raman spectroscopically observed for the first time in ROD samples and reported here. Thus, elevated phenylalanine could also be considered as an indicator of the disease. Since the image results are based on tens of thousands of spectra per sample, not only are the average ratios statistically significantly different for normal and ROD bone, but the method is clearly powerful in distinguishing between the two types of samples. Furthermore, the statistical outcomes demonstrate that only a relatively small number of spectra need to be recorded in order to classify the samples. This work thus opens the possibility of future development of in vivo Raman sensors for assessment of bone structure, remodeling, and mineralization, where different biomarkers are simultaneously detected with unprecedented accuracy.


Asunto(s)
Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/diagnóstico por imagen , Microscopía/métodos , Espectrometría Raman/métodos , Anciano , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/patología , Humanos
18.
J Exp Clin Cancer Res ; 37(1): 244, 2018 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-30286779

RESUMEN

BACKGROUND: Osteosarcoma is the most common bone cancer. Despite advances, molecular mechanisms associated with osteosarcoma have not been fully understood. Hence, an effective treatment for osteosarcoma has yet to be developed. Even though signal transducer and activator of transcription3 (STAT3) has been implicated, its role in pathogenesis of osteosarcoma is not fully determined. In this study, we investigated the antitumor effect of napabucasin (NP) (BBI608), an inhibitor of STAT3 on osteosarcoma in vitro and in vivo and studied the underlying molecular mechanism. METHODS: Cell viability, colony formation, apoptosis, tumor growth and metastasis assays were performed to examine the effect of NP on osteosarcoma in vitro and in vivo. Real-time RT-PCR, western analysis, immunofluorescence and reporter assays were used to monitor the expression and activity of proteins and underlying molecular pathways. Protein synthesis, co-immunoprecipitation and CAP binding assays were carried out to understand NP-mediated mechanism of actions in osteosarcoma cells. RESULTS: Our results show that NP treatment decreases cell viability and induces apoptosis in several osteosarcoma cell lines. NP treatment suppresses both expression and phosphorylation of STAT3 in addition to blocking STAT3-mediated transcription and downstream target proteins in osteosarcoma cells. Furthermore, NP inhibits protein synthesis through regulation of the eukaryotic initiation factor 4E (eIF4E) and eIF4E-binding protein 1 (4E-BP1). NP also inhibits the progression of osteosarcoma tumors and metastasis in vivo in an orthotopic tibial model of osteosarcoma. CONCLUSIONS: Taken together, our investigation reveals that NP acts through a novel mechanism and inhibits osteosarcoma growth and metastasis, and could be investigated clinically for treating osteosarcoma patients alone or in combination with other drugs.


Asunto(s)
Benzofuranos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Naftoquinonas/farmacología , Osteosarcoma/tratamiento farmacológico , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Osteosarcoma/metabolismo , Osteosarcoma/patología , Inhibidores de la Síntesis de la Proteína/farmacología , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Gene ; 674: 1-7, 2018 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-29933019

RESUMEN

Osteosarcoma is a malignant bone tumor that occurs mainly in children and adolescents. Because Wnt signaling has been implicated in the pathogenesis of osteosarcoma, we have investigated the circulating and local levels of the Wnt antagonist protein, Secreted Frizzled Related Protein (sFRP) 3, in osteosarcoma patients. Enzyme linked immunosorbent assay (ELISA) analysis of 67 osteosarcoma and age-matched non-diseased control sera showed that sFPR3 protein levels were significantly lower in osteosarcoma than in normal. Analysis of tumor and adjacent normal tissues (9 pairs) from osteosarcoma patients showed a decrease in sFRP3 expression in 5 out of 9 tumor samples compared to normal tissues. Furthermore, immunohistochemical analysis of tissue microarray revealed a significant decrease in sFRP3 levels in tumor compared to normal bone. RNA sequencing analysis in osteosarcoma cells shows suppression of sFRP3 and concomitant expression of multiple Wnt family members mediating canonical or non-canonical Wnt signaling. Taken together, our findings show that the systemic and local levels of sFRP3 protein are downregulated in osteosarcoma and sFRP3 levels could be explored further in the diagnosis and the care of osteosarcoma patients.


Asunto(s)
Neoplasias Óseas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Osteosarcoma/metabolismo , Adolescente , Adulto , Anciano , Neoplasias Óseas/sangre , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Niño , Regulación hacia Abajo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/sangre , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Persona de Mediana Edad , Osteosarcoma/sangre , Osteosarcoma/genética , Osteosarcoma/patología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Adulto Joven
20.
J Bone Miner Res ; 22(1): 29-36, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17014383

RESUMEN

UNLABELLED: We studied the involvement of interferon-regulated, PKR on 2-ME-mediated actions in human osteosarcoma cells. Our results show that PKR is activated by 2-ME treatment and is necessary for 2-ME-mediated induction of osteosarcoma cell death. INTRODUCTION: Osteosarcoma is the most common primary bone tumor and most frequently develops during adolescence. 2-Methoxyestradiol (2-ME), a metabolite of 17beta-estradiol, induces interferon gene expression and apoptosis in human osteosarcoma cells. In this report, we studied the role of interferon-regulated double-stranded (ds)RNA-dependent protein kinase (PKR) protein on 2-ME-mediated cell death in human osteosarcoma cells. MATERIALS AND METHODS: Western blot analyses were used to measure PKR protein and phosphorylation levels. Cell survival and apoptosis assays were measured using trypan blue exclusion and Hoechst dye methods, respectively. A transient transfection protocol was used to express the dominant negative PKR mutants. RESULTS AND CONCLUSIONS: PKR was increased in 2-ME-treated MG63 cells, whereas 17beta-estradiol, 4-hydroxyestradiol, and 16alpha-hydroxyestradiol, which do not induce cell death, had no effect on PKR protein levels. Also, 2-ME treatment induced PKR kinase activity as indicated by increased autophosphorylation and phosphorylation of the endogenous substrate, eukaryotic initiation factor (eIF)-2alpha. dsRNA poly (I).poly (C), an activator of PKR protein, increased cell death when osteosarcoma cells were treated with a submaximal concentration of 2-ME. In contrast, a serine-threonine kinase inhibitor SB203580 and a specific PKR inhibitor 2-aminopurine (2-AP) blocked the 2-ME-induced cell death in MG63 cells. A dominant negative PKR mutant protein conferred resistance to 2-ME-induced cell death to MG63 osteosarcoma and 2-ME-mediated PKR regulation did not require interferon gene expression. PKR protein is activated in cell free extracts by 2-ME treatment, resulting in autophosphorylation and in the phosphorylation of the substrate eIF-2alpha. We conclude from these results that PKR is regulated by 2-ME independently of interferon and is essential for 2-ME-mediated cell death in MG63 osteosarcoma cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Óseas , Muerte Celular/efectos de los fármacos , Estradiol/análogos & derivados , Osteosarcoma , Proteínas Quinasas/genética , ARN Bicatenario/genética , 2-Metoxiestradiol , División Celular , Línea Celular Tumoral , Estradiol/farmacología , Humanos , Inhibidores de Proteínas Quinasas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA