Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Drug Dev Res ; 85(3): e22195, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38704831

RESUMO

We investigated the angiogenesis-modulating ability of noscapine in vitro using osteosarcoma cell line (MG-63) and in vivo using a zebrafish model. MTT assay and the scratch wound healing assay were performed on the osteosarcoma cell line (MG-63) to analyze the cytotoxic effect and antimigrative ability of noscapine, respectively. We also observed the antiangiogenic ability of noscapine on zebrafish embryos by analyzing the blood vessels namely the dorsal aorta, and intersegmental vessels development at 24, 48, and 72 h postfertilization. Real-time polymerase chain reaction was used to analyze the hypoxia signaling molecules' gene expression in MG-63 cells and zebrafish embryos. The findings from the scratch wound healing demonstrated that noscapine stopped MG-63 cancer cells from migrating under both hypoxia and normoxia. Blood vessel development and the heart rate in zebrafish embryos were significantly reduced by noscapine under both hypoxia and normoxia which showed the hemodynamics impact of noscapine. Noscapine also downregulated the cobalt chloride (CoCl2) induced hypoxic signaling molecules' gene expression in MG-63 cells and zebrafish embryos. Therefore, noscapine may prevent MG-63 cancer cells from proliferating and migrating, as well as decrease the formation of new vessels and the production of growth factors linked to angiogenesis in vivo under both normoxic and hypoxic conditions.


Assuntos
Hemodinâmica , Neovascularização Patológica , Noscapina , Peixe-Zebra , Animais , Humanos , Noscapina/farmacologia , Linhagem Celular Tumoral , Hemodinâmica/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Hipóxia , Movimento Celular/efeitos dos fármacos , Embrião não Mamífero/efeitos dos fármacos , Osteossarcoma/tratamento farmacológico , Angiogênese
2.
Biochem Biophys Res Commun ; 646: 56-62, 2023 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-36706706

RESUMO

G protein coupled free fatty acid receptors (FFARs) are involved in the pathogenesis of several human diseases. FFAR2 and FFAR3 are activated by the binding of short-chain fatty acids (SCFAs). This study aimed to evaluate the roles of FFAR2 in the regulation of cellular functions in osteosarcoma HOS cells, using acetic acid and propanoic acid as FFAR2 and FFAR3 agonists. FFAR2 and FFAR3 genes were expressed in HOS cells. The cell motile activity of HOS cells was significantly stimulated by acetic acid and propanoic acid. In contrast, acetic acid and propanoic acid had no impact on the activation of matrix metalloproteinase-2 (MMP-2) and MMP-9. In cell survival assay, the cell survival rate to cisplatin (CDDP) of HOS cells was elevated by acetic acid and propanoic acid. To assess the effects of FFAR2 on cellular functions, FFAR2 knockdown (HOS-FFAR2) cells were generated from HOS cells. The cell motile activity of HOS-FFAR2 cells was enhanced by acetic acid and propanoic acid. In the presence of acetic acid and propanoic acid, MMP-2 and MMP-9 activities were reduced in HOS-FFAR2 cells, compared with control cells. When cells were treated with acetic acid and propanoic acid, the cell survival rate to CDDP of HOS-FFAR2 cells was significantly lower than that of control cells. These results suggest that activation of FFAR2-mediated signaling is involved in the modulation of cellular functions in HOS cells.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Humanos , Propionatos/farmacologia , Metaloproteinase 2 da Matriz/genética , Receptores Acoplados a Proteínas G/metabolismo , Ácidos Graxos não Esterificados , Metaloproteinase 9 da Matriz/genética , Ácidos Graxos Voláteis/farmacologia , Osteossarcoma/genética , Cisplatino/farmacologia , Ácido Acético , Neoplasias Ósseas/genética
3.
Exp Cell Res ; 417(2): 113223, 2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35643180

RESUMO

Many studies indicated that static magnetic fields (SMFs) have anti-cancer effects. However, effect of SMFs on cancer cells with strength exceeding 12 T are rarely reported. The intracellular iron could participate in the reactive oxygen species (ROS) production and affect cell proliferation. This study aimed to investigate the effect of 12 T high static magnetic field (HiSMF) on osteosarcoma cells and the relationship with intracellular iron. The 12 T HiSMF was generated by a superconducting magnet. The proliferation was evaluated by CCK-8 assays and cell counting. The apoptosis, cell cycle distribution, and ROS were evaluated by flow cytometry. Intracellular iron status was evaluated by atomic absorption spectroscopy and Calcein-AM/2,2'-bipyridyl. The expression of cell cycle and iron metabolism-related genes were analyzed by Western Blot. The result showed that 12 T HiSMF exposure suppressed the proliferation of osteosarcoma cell lines MNNG/HOS, U-2 OS, and MG63 via cell cycle arrest in S and G2/M. Meanwhile, 12 T HiSMF increasing intracellular ROS, and its antitumor effect was reduced by antioxidant. Furthermore, the intracellular total and free iron levels, the expression of FTH1 and DMT1 were increased by 12 HiSMF. The iron chelator (DFO) could reduce the cytotoxicity of 12 T HiSMF on osteosarcoma cells. Moreover, 12 T HiSMF could enhance the cytotoxicity of cisplatin and sorafenib in osteosarcoma cells. In Conclusion, 12 T HiSMF could suppress osteosarcoma cells proliferation via intracellular iron and ROS related cell cycle arrest, and have application potential in osteosarcoma therapy combined with sorafenib and cisplatin.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Apoptose , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Cisplatino/farmacologia , Humanos , Ferro , Campos Magnéticos , Osteossarcoma/genética , Espécies Reativas de Oxigênio/metabolismo , Sorafenibe/farmacologia
4.
Int J Mol Sci ; 24(24)2023 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-38139388

RESUMO

This work investigated the mechanisms of action of conventional drugs, cisplatin and oxaliplatin, and the potentially less deleterious drug Pd2Spermine (Spm) and its Pt(II) analog, against osteosarcoma MG-63 cells, using nuclear-magnetic-resonance metabolomics of the cellular lipidome. The Pt(II) chelates induced different responses, namely regarding polyunsaturated-fatty-acids (increased upon cisplatin), suggesting that cisplatin-treated cells have higher membrane fluidity/permeability, thus facilitating cell entry and justifying higher cytotoxicity. Both conventional drugs significantly increased triglyceride levels, while Pt2Spm maintained control levels; this may reflect enhanced apoptotic behavior for conventional drugs, but not for Pt2Spm. Compared to Pt2Spm, the more cytotoxic Pd2Spm (IC50 comparable to cisplatin) induced a distinct phospholipids profile, possibly reflecting enhanced de novo biosynthesis to modulate membrane fluidity and drug-accessibility to cells, similarly to cisplatin. However, Pd2Spm differed from cisplatin in that cells had equivalent (low) levels of triglycerides as Pt2Spm, suggesting the absence/low extent of apoptosis. Our results suggest that Pd2Spm acts on MG-63 cells mainly through adaptation of cell membrane fluidity, whereas cisplatin seems to couple a similar effect with typical signs of apoptosis. These results were discussed in articulation with reported polar metabolome adaptations, building on the insight of these drugs' mechanisms, and particularly of Pd2Spm as a possible cisplatin substitute.


Assuntos
Antineoplásicos , Neoplasias Ósseas , Osteossarcoma , Humanos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Metabolismo dos Lipídeos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Osteossarcoma/tratamento farmacológico , Osteossarcoma/metabolismo , Espermina/metabolismo , Apoptose , Neoplasias Ósseas/tratamento farmacológico , Linhagem Celular Tumoral
5.
Int J Mol Sci ; 24(19)2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37834071

RESUMO

Cancer cells overexpress IF1, the endogenous protein that inhibits the hydrolytic activity of ATP synthase when mitochondrial membrane potential (ΔµH+) falls, as in ischemia. Other roles have been ascribed to IF1, but the associated molecular mechanisms are still under debate. We investigated the ability of IF1 to promote survival and proliferation in osteosarcoma and colon carcinoma cells exposed to conditions mimicking ischemia and reperfusion, as occurs in vivo, particularly in solid tumors. IF1-silenced and parental cells were exposed to the FCCP uncoupler to collapse ΔµH+ and the bioenergetics of cell models were validated. All the uncoupled cells preserved mitochondrial mass, but the implemented mechanisms differed in IF1-expressing and IF1-silenced cells. Indeed, the membrane potential collapse and the energy charge preservation allowed an increase in both mitophagy and mitochondrial biogenesis in IF1-expressing cells only. Interestingly, the presence of IF1 also conferred a proliferative advantage to cells highly dependent on oxidative phosphorylation when the uncoupler was washed out, mimicking cell re-oxygenation. Overall, our results indicate that IF1, by allowing energy preservation and promoting mitochondrial renewal, can favor proliferation of anoxic cells and tumor growth. Therefore, hindering the action of IF1 may be promising for the therapy of tumors that rely on oxidative phosphorylation for energy production.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Humanos , Mitocôndrias/metabolismo , Hipóxia/metabolismo , Osteossarcoma/metabolismo , Neoplasias Ósseas/metabolismo , Isquemia/metabolismo , Proliferação de Células , Trifosfato de Adenosina/metabolismo
6.
J Bioenerg Biomembr ; 54(2): 109-117, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35260987

RESUMO

Lysophosphatidic acid (LPA) signaling via LPA receptors (LPA1 to LPA6) exhibits a variety of malignant properties in cancer cells. Intracellular ATP depletion leads to the development of necrosis and apoptosis. The present study aimed to evaluate the effects of LPA receptor-mediated signaling on the regulation of cancer cell functions associated with ATP reduction. Long-term ethidium bromide (EtBr) treated (MG63-EtBr) cells were established from osteosarcoma MG-63 cells. The intracellular ATP levels of MG63-EtBr cells were significantly lower than that of MG-63 cells. LPAR2, LPAR3, LPAR4 and LPAR6 gene expressions were elevated in MG63-EtBr cells. The cell motile and invasive activities of MG63-EtBr cells were markedly higher than those of MG-63 cells. The cell motile activity of MG-63 cells was increased by LPA4 and LPA6 knockdowns. In cell survival assay, cells were treated with cisplatin (CDDP) every 24 h for 3 days. The cell survival to CDDP of MG63-EtBr cells was lower than that of MG-63 cells. LPA2 knockdown decreased the cell survival to CDDP of MG-63 cells. The cell survival to CDDP of MG-63 cells was inhibited by (2 S)-OMPT (LPA3 agonist). Moreover, the cell survival to CDDP of MG-63 cells was enhanced by LPA4 and LPA6 knockdowns. These results indicate that LPA signaling via LPA receptors is involved in the regulation of cellular functions associated with ATP reduction in MG-63 cells treated with EtBr.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Trifosfato de Adenosina/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/genética , Movimento Celular , Etídio/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Lisofosfolipídeos/metabolismo , Osteossarcoma/tratamento farmacológico , Osteossarcoma/genética , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/metabolismo
7.
Luminescence ; 37(2): 323-331, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34871472

RESUMO

There have been strong demands for nanofibrous scaffolds fabricated by electrospinning for various fields due to their various advantages. Electrospun poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) fibre mats were prepared. The effects of processing variables as well as the inclusion of poly(ethylene glycol) (PEG) on the morphologies of generated fibres were investigated using Fourier-transform infrared spectroscopy and scanning electron microscopy. The average fibrous diameter was monitored in the range 400-3000 nm relying on the total content of PEG. The fluorescence cell imaging of electrospun mats was also explored. The results of cell viability demonstrated that skin fibroblast BJ-1 cells showed different adhesions and growth rates for the three kinds of PHBV fibres. Electrospun PHBV mats with low amount of PEG offer a high-quality medium for cell growth. Therefore, those mats exhibited high potential for soft tissue engineering, in particular wound healing.


Assuntos
Nanofibras , Engenharia Tecidual , Ácido 3-Hidroxibutírico , Proliferação de Células , Hidroxibutiratos , Poliésteres , Polietilenoglicóis
8.
Int J Mol Sci ; 23(19)2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36232858

RESUMO

Several semisynthetic, low-cardiotoxicity doxorubicin (DOXO) conjugated have been extensively described, considering the risk of cytotoxicity loss against resistant tumor cells, which mainly present drug efflux capacity. Doxorubicin 14-[4-(4-phenyl-5-thioxo-5H-[1,2]dithiol-3-yl)]-benzoate (H2S-DOXO) was synthetized and tested for its ability to overcome drug resistance with good intracellular accumulation. In this paper, we present a formulation study aimed to develop naked and decorated H2S-DOXO-loaded lipid nanoparticles (NPs). NPs prepared by the "cold dilution of microemulsion" method were decorated with hyaluronic acid (HA) to obtain active targeting and characterized for their physicochemical properties, drug entrapment efficiency, long-term stability, and in vitro drug release. Best formulations were tested in vitro on human-sensitive (MCF7) and human/mouse DOXO-resistant (MDA-MDB -231 and JC) breast cancer cells, on human (U-2OS) osteosarcoma cells and DOXO-resistant human/mouse osteosarcoma cells (U-2OS/DX580/K7M2). HA-decoration by HA-cetyltrimethyl ammonium bromide electrostatic interaction on NPs surface was confirmed by Zeta potential and elemental analysis at TEM. NPs had mean diameters lower than 300 nm, 70% H2S-DOXO entrapment efficiency, and were stable for almost 28 days. HA-decorated NPs accumulated H2S-DOXO in Pgp-expressing cells reducing cell viability. HA-decorated NPs result in the best formulation to increase the inter-cellular H2S-DOXO delivery and kill resistant cells, and therefore, as a future perspective, they will be taken into account for further in vivo experiments on tumor animal model.


Assuntos
Neoplasias Ósseas , Nanopartículas , Osteossarcoma , Animais , Benzoatos , Linhagem Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacologia , Humanos , Ácido Hialurônico/química , Lipossomos , Camundongos , Nanopartículas/química , Osteossarcoma/patologia
9.
Int J Mol Sci ; 23(18)2022 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-36142445

RESUMO

The hEag1 (Kv10.1) K+ channel is normally found in the brain, but it is ectopically expressed in tumor cells, including osteosarcoma. Based on the pivotal role of ion channels in osteogenesis, we tested whether pharmacological modulation of hEag1 may affect osteogenic differentiation of osteosarcoma cell lines. Using molecular biology (RT-PCR), electrophysiology (patch-clamp) and pharmacology (astemizole sensitivity, IC50 = 0.135 µM) we demonstrated that SaOS-2 osteosarcoma cells also express hEag1 channels. SaOS-2 cells also express to KCa1.1 K+ channels as shown by mRNA expression and paxilline sensitivity of the current. The inhibition of hEag1 (2 µM astemizole) or KCa1.1 (1 mM TEA) alone did not induce Ca2+ deposition in SaOS-2 cultures, however, these inhibitors, at identical concentrations, increased Ca2+ deposition evoked by the classical or pathological (inorganic phosphate, Pi) induction pathway without causing cytotoxicity, as reported by three completer assays (LDH release, MTT assay and SRB protein assay). We observed a similar effect of astemizole on Ca2+ deposition in MG-63 osteosarcoma cultures as well. We propose that the increase in the osteogenic stimuli-induced mineral matrix formation of osteosarcoma cell lines by inhibiting hEag1 may be a useful tool to drive terminal differentiation of osteosarcoma.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Astemizol/farmacologia , Linhagem Celular Tumoral , Canais de Potássio Éter-A-Go-Go , Humanos , Osteogênese , Osteossarcoma/tratamento farmacológico , Fosfatos/metabolismo , RNA Mensageiro/genética
10.
Angew Chem Int Ed Engl ; 61(44): e202210568, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36102872

RESUMO

Herein, we show intranuclear nanoribbons formed upon dephosphorylation of leucine-rich L- or D-phosphopeptide catalyzed by alkaline phosphatase (ALP) to selectively kill osteosarcoma cells. Being dephosphorylated by ALP, the peptides are first transformed into micelles and then converted into nanoribbons. The peptides/assemblies first aggregate on cell membranes, then enter cells via endocytosis, and finally accumulate in nuclei (mainly in nucleoli). Proteomics analysis suggests that the assemblies interact with histone proteins. The peptides kill osteosarcoma cells rapidly and are nontoxic to normal cells. Moreover, the repeated stimulation of the osteosarcoma cells by the peptides sensitizes the cancer cells rather than inducing resistance. This work not only illustrates a novel mechanism for nucleus targeting, but may also pave a new way for selectively killing osteosarcoma cells and minimizing drug resistance.


Assuntos
Neoplasias Ósseas , Nanotubos de Carbono , Osteossarcoma , Humanos , Fosfatase Alcalina/metabolismo , Micelas , Fosfopeptídeos/metabolismo , Histonas , Leucina , Osteossarcoma/tratamento farmacológico , Linhagem Celular Tumoral , Neoplasias Ósseas/tratamento farmacológico
11.
Exp Mol Pathol ; 118: 104596, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33347862

RESUMO

Lysophosphatidic acid (LPA) signaling via LPA receptors (LPA1 to LPA6) exhibits a variety of biological responses. In tumor microenvironment, endothelial cells promote cancer cell functions. In this study, we investigated the roles of endothelial cells in the regulation of cell motile activity via LPA2 and LPA3 in human osteosarcoma MG-63 cells. In cell motility assay, the cell motile activity of MG-63 cells was markedly increased by the supernatants of endothelial F2 cells. MG-63 cell motility elevated by the supernatants was enhanced by GRI-977143 (LPA2 agonist) and reduced by (2S)-OMPT (LPA3 agonist). LPAR2 and LPAR3 expressions were increased in highly migratory MG63-CR7(F2) cells, which were generated from MG-63 cells by co-culture with F2 cell supernatants. MG63-CR7(F2) cell motility was stimulated by LPA treatment. In the presence of F2 cell supernatants, MG63-CR7(F2) cell motility was markedly enhanced by GRI-977143 and suppressed by (2S)-OMPT. Autotaxin (ATX) enzymatically converts lysophosphatidylcholine (LPC) to LPA. ATX expression was higher in MG63-CR(F2) cells than in MG-63 cells. MG63-CR7(F2) cell motility was markedly increased by LPC in comparison with MG-63 cells. In addition, MG63-CR(F2) cell motility was significantly stimulated by the supernatants of LPC treated F2 cells. The present results suggest that the activation of LPA signaling via LPA2 and LPA3 by endothelial cells is involved in the modulation of cell motile activity of MG-63 cells.


Assuntos
Neoplasias Ósseas/patologia , Movimento Celular , Células Endoteliais/patologia , Regulação Neoplásica da Expressão Gênica , Osteossarcoma/patologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Apoptose , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Proliferação de Células , Células Endoteliais/metabolismo , Humanos , Lisofosfolipídeos/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Transdução de Sinais , Células Tumorais Cultivadas
12.
Exp Cell Res ; 388(1): 111813, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31904382

RESUMO

Lysophosphatidic acid (LPA) signaling via LPA receptors (LPA1 to LPA6) regulates a variety of malignant properties of cancer cells. It is known that endothelial cells promote tumor progression and chemoresistance. The present study aimed to investigate the roles of LPA5 in cellular functions modulated by endothelial cells and anticancer drug in osteosarcoma cells. Human osteosarcoma MG-63 cells were maintained in endothelial F2 cell supernatants. After culturing for 3 months, MG63-F2 cells were established. LPAR5 expression level in MG63-F2 cells was significantly elevated, compared with MG-63 cells. The cell motile activity of MG63-F2 cells was markedly higher than that of MG-63 cells. To validate the effects of LPA5 on cell motile activity, LPA5 knockdown cells were generated from MG-63 cells. The cell motile activity of MG-63 cells was inhibited by LPA5 knockdown. The cell survival to cisplatin (CDDP) was reduced in MG-63 cells treated with LPA. In the presence of LPA, the cell survival rate was significantly lower in MG63-F2 cells than MG-63 cells, correlating with LPAR5 expression. LPA5 knockdown cells indicated the high cell survival rate to CDDP. Moreover, LPAR5 expression level was increased in the long-term CDDP treated MG63-C cells. The cell survival to CDDP of MG63-C cells was enhanced by LPA5 knockdown. These results suggest that cellular functions are regulated through LPA5-mediatd signaling induced by endothelial cells and CDDP in MG-63 cells.


Assuntos
Células Endoteliais/metabolismo , Osteossarcoma/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Antineoplásicos/farmacologia , Morte Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Cisplatino/análogos & derivados , Cisplatino/farmacologia , Meios de Cultivo Condicionados/farmacologia , Humanos , Lisofosfolipídeos/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Transdução de Sinais , Células Tumorais Cultivadas
13.
Int J Mol Sci ; 22(8)2021 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-33924370

RESUMO

The mineralization process is initiated by osteoblasts and chondrocytes during intramembranous and endochondral ossifications, respectively. Both types of cells release matrix vesicles (MVs), which accumulate Pi and Ca2+ and form apatites in their lumen. Tissue non-specific alkaline phosphatase (TNAP), a mineralization marker, is highly enriched in MVs, in which it removes inorganic pyrophosphate (PPi), an inhibitor of apatite formation. MVs then bud from the microvilli of mature osteoblasts or hypertrophic chondrocytes and, thanks to the action of the acto-myosin cortex, become released to the extracellular matrix (ECM), where they bind to collagen fibers and propagate mineral growth. In this report, we compared the mineralization ability of human fetal osteoblastic cell line (hFOB 1.19 cells) with that of osteosarcoma cell line (Saos-2 cells). Both types of cells were able to mineralize in an osteogenic medium containing ascorbic acid and beta glycerophosphate. The composition of calcium and phosphate compounds in cytoplasmic vesicles was distinct from that in extracellular vesicles (mostly MVs) released after collagenase-digestion. Apatites were identified only in MVs derived from Saos-2 cells, while MVs from hFOB 1.19 cells contained amorphous calcium phosphate complexes. In addition, AnxA6 and AnxA2 (nucleators of mineralization) increased mineralization in the sub-membrane region in strongly mineralizing Saos-2 osteosarcoma, where they co-localized with TNAP, whereas in less mineralizing hFOB 1.19 osteoblasts, AnxA6, and AnxA2 co-localizations with TNAP were less visible in the membrane. We also observed a reduction in the level of fetuin-A (FetuA), an inhibitor of mineralization in ECM, following treatment with TNAP and Ca channels inhibitors, especially in osteosarcoma cells. Moreover, a fraction of FetuA was translocated from the cytoplasm towards the plasma membrane during the stimulation of Saos-2 cells, while this displacement was less pronounced in stimulated hFOB 19 cells. In summary, osteosarcoma Saos-2 cells had a better ability to mineralize than osteoblastic hFOB 1.19 cells. The formation of apatites was observed in Saos-2 cells, while only complexes of calcium and phosphate were identified in hFOB 1.19 cells. This was also evidenced by a more pronounced accumulation of AnxA2, AnxA6, FetuA in the plasma membrane, where they were partly co-localized with TNAP in Saos-2 cells, in comparison to hFOB 1.19 cells. This suggests that both activators (AnxA2, AnxA6) and inhibitors (FetuA) of mineralization were recruited to the membrane and co-localized with TNAP to take part in the process of mineralization.


Assuntos
Anexina A2/metabolismo , Anexina A6/metabolismo , Calcificação Fisiológica , Osteoblastos/metabolismo , Osteossarcoma/metabolismo , alfa-2-Glicoproteína-HS/metabolismo , Fosfatase Alcalina/metabolismo , Cálcio/metabolismo , Linhagem Celular Tumoral , Forma Celular , Humanos , Fósforo/metabolismo
14.
Molecules ; 26(16)2021 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-34443394

RESUMO

This paper reports the first metabolomics study of the impact of new chelates Pt2Spm and Pd2Spm (Spm = Spermine) on human osteosarcoma cellular metabolism, compared to the conventional platinum drugs cisplatin and oxaliplatin, in order to investigate the effects of different metal centers and ligands. Nuclear Magnetic Resonance metabolomics was used to identify meaningful metabolite variations in polar cell extracts collected during exposure to each of the four chelates. Cisplatin and oxaliplatin induced similar metabolic fingerprints of changing metabolite levels (affecting many amino acids, organic acids, nucleotides, choline compounds and other compounds), thus suggesting similar mechanisms of action. For these platinum drugs, a consistent uptake of amino acids is noted, along with an increase in nucleotides and derivatives, namely involved in glycosylation pathways. The Spm chelates elicit a markedly distinct metabolic signature, where inverse features are observed particularly for amino acids and nucleotides. Furthermore, Pd2Spm prompts a weaker response from osteosarcoma cells as compared to its platinum analogue, which is interesting as the palladium chelate exhibits higher cytotoxicity. Putative suggestions are discussed as to the affected cellular pathways and the origins of the distinct responses. This work demonstrates the value of untargeted metabolomics in measuring the response of cancer cells to either conventional or potential new drugs, seeking further understanding (or possible markers) of drug performance at the molecular level.


Assuntos
Antineoplásicos/farmacologia , Quelantes/farmacologia , Desenho de Fármacos , Osteossarcoma/patologia , Paládio/química , Platina/química , Antineoplásicos/química , Linhagem Celular Tumoral , Quelantes/química , Humanos
15.
J Cell Mol Med ; 24(21): 12789-12798, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32985079

RESUMO

In this study, a new water and alkaline-soluble polysaccharide (ALP), with an average molecular weight of 6.63 × 104  Da, was successfully purified from the rhizomes of Atractylodes lancea. GC analysis demonstrated that ALP was a kind of glucan. The effect of the ALP on the interaction between E-selectin and sialyl Lewis X (sLex ) was examined in human osteosarcoma U-2 OS cells. It was obvious that the expression of sLex antigen on the surface of U-2 OS cells was visible under fluorescence microscopy. The addition of ALP (0.5, 1 and 2 mg/mL) resulted in a marked inhibition on the adhesion, migration and invasion of U-2 OS cells to human umbilical vein endothelial cells (HUVECs), which was achieved by the decreased sLex expression on U-2 OS cells. Additionally, the induction of apoptosis can be observed in U-2 OS cells following ALP treatment using TUNEL staining and Annexin V-FITC/PI double-staining analysis on flow cytometry. In conclusion, these results indicated that ALP exerted anti-metastatic activity towards osteosarcoma cells via inhibition of sLex /E-selectin binding, which suggested that ALP could be a potent agent for human osteosarcoma intervention.


Assuntos
Atractylodes/química , Selectina E/metabolismo , Osteossarcoma/patologia , Polissacarídeos/farmacologia , Antígeno Sialil Lewis X/metabolismo , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Monossacarídeos/análise , Metástase Neoplásica , Polissacarídeos/isolamento & purificação , Ligação Proteica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
16.
J Recept Signal Transduct Res ; 40(2): 181-186, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32026734

RESUMO

Free fatty acid receptor 1 (FFA1) and FFA4 belong to a family of free fatty acid (FFA) receptors. FFA1- and FFA4-mediated signaling regulates a variety of malignant properties in cancer cells. It is known that stromal cells in the tumor microenvironment promote tumor progression. In the present study, to assess the roles of FFA1 and FFA4 in cellular functions modulated by endothelial cells, highly migratory MG63-CR7(F2) cells were generated from osteosarcoma MG-63 cells, using endothelial F2 cell supernatants. Expression levels of FFAR1 and FFAR4 genes in MG63-CR7(F2) cells were significantly higher than those of MG-63 cells. In cell survival assay, cells were treated with cisplatin (CDDP) every 24 h for 2 days. The cell survival rate of MG-63 cells was significantly elevated by an FFA1 agonist TUG-770 as well as an FFA4 agonist TUG-891. Moreover, the cell survival rate of MG63-CR7(F2) cells was higher than that of MG-63 cells in the presence of TUG-770 or TUG-891, correlating with FFAR1 and FFAR4 expression levels. To validate the effects of FFA1 and FFA4 on cell survival to CDDP, FFA1 and FFA4 knockdown cell were generated from MG-63 cells. The cell survival rate of MG-63 cells was markedly inhibited by FFA1 or FFA4 knockdown. These results suggest that FFA1 and FFA4 may play an important role in the modulation of cellular functions by endothelial cells in osteosarcoma cells.


Assuntos
Carcinogênese/genética , Osteossarcoma/tratamento farmacológico , Receptores Acoplados a Proteínas G/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Células Endoteliais/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Osteossarcoma/genética , Osteossarcoma/patologia , Microambiente Tumoral/efeitos dos fármacos
17.
Int J Med Sci ; 17(5): 577-590, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32210707

RESUMO

Confocal Raman microspectral imaging was adopted to elucidate the cellular drug responses of osteosarcoma cells (OC) to N-[N-(3, 5-difluorophenyl acetyl)-L-alanyl]-sphenylglycine butyl ester (DAPT), a γ-secretase inhibitor, by identifying the drug induced subcellular compositional and structural changes. Methods: Spectral information were acquired from cultured osteosarcoma cells treated with 0 (Untreated Group, UT), 10 (10 µM DAPT treated, 10T), 20 µM (20 µM DAPT treated, 20T) DAPT for 24 hours. A one-way ANOVA and Tukey's honest significant difference (HSD) post hoc multiple test were sequentially applied to address spectral features among three groups. Multivariate algorithms such as K-means clustering analysis (KCA) and Principal component analysis (PCA) were used to highlight the structural and compositional differences, while, univariate imaging was applied to illustrate the distribution pattern of certain cellular components after drug treatment. Results: Major biochemical changes in DAPT-induced apoptosis came from changes in the content and structure of proteins, lipids, and nucleic acids. By adopted multivariate algorithms, the drug induced cellular changes was identified by the morphology and spectral characteristics between untreated cells and treated cells, testified that DAPT mainly acted in the nuclear region. With the increase of the drug concentration, the content of main subcellular compositions, such nucleic acid, protein, and lipid decreased. In an addition, DAPT-induced nuclear fragmentation and apoptosis was depicted by the univariate Raman image of major cellular components (nucleic acids, proteins and lipids). Conclusions: The achieved Raman spectral and imaging results illustrated detailed DAPT-induced subcellular compositional and structural variations as a function of drug dose. Such observations can not only explain drug therapeutic mechanisms of OC DAPT treatment, and also provide new insights for accessing the medicine curative efficacy and predicting prognosis.


Assuntos
Estruturas Celulares/efeitos dos fármacos , Dipeptídeos/farmacologia , Osteossarcoma/tratamento farmacológico , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Dipeptídeos/uso terapêutico , Ensaios de Seleção de Medicamentos Antitumorais , Camundongos , Osteossarcoma/química , Análise de Componente Principal , Análise Espectral Raman
18.
Clin Exp Pharmacol Physiol ; 47(3): 517-519, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31705676

RESUMO

Our results showed that the expression of calcium-sensing receptor (CaSR) and the concentration of intracellular calcium were enhanced in the osteosarcoma cells MG-63 compared with in the normal osteoblasts hFOB1.19. GdCl3 (CaSR agonist) significantly increased the proliferation rate of MG-63 cells, the expression of PCNA and Cyclin D1 and decreased the expression of p21Cip/WAF-1 . The effect of NPS2390 (CaSR antagonist) on the above indicators was opposite to GdCl3 . In addition, GdCl3 up-regulated the phosphorylation of ERK1/2, PI3K and Akt and the proliferation rate of MG-63 cells. However, these effects of GdCl3 were cancelled by LY294002 (a PI3K inhibitor) or PD98059 (an ERK1/2 inhibitor). Our results demonstrate that activation of CaSR promotes osteosarcoma cell multiplication by up-regulating ERK1/2 and PI3K-Akt pathways.


Assuntos
Neoplasias Ósseas/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Osteossarcoma/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Neoplasias Ósseas/patologia , Humanos , Osteossarcoma/patologia
19.
Molecules ; 25(1)2020 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-31935900

RESUMO

Currently, significant attention is attracted to the problem of the development of the specific architecture and composition of the surface layer in order to control the biocompatibility of implants made of titanium and its alloys. The titanium surface properties can be tuned both by creating an inorganic sublayer with the desired morphology and by organic top coating contributing to bioactivity. In this work, we developed a composite biologically active coatings based on hybrid molecules obtained by chemical cross-linking of amino acid bisphosphonates with a linear tripeptide RGD, in combination with inorganic porous sublayer created on titanium by plasma electrolytic oxidation (PEO). After the addition of organic molecules, the PEO coated surface gets nobler, but corrosion currents increase. In vitro studies on proliferation and viability of fibroblasts, mesenchymal stem cells and osteoblast-like cells showed the significant dependence of the molecule bioactivity on the structure of bisphosphonate anchor and the linker. Several RGD-modified bisphosphonates of ß-alanine, γ-aminobutyric and ε-aminocaproic acids with BMPS or SMCC linkers can be recommended as promising candidates for further in vivo research.


Assuntos
Materiais Revestidos Biocompatíveis , Oligopeptídeos , Ácidos Fosforosos , Próteses e Implantes , Titânio , Linhagem Celular , Materiais Revestidos Biocompatíveis/química , Eletroquímica , Humanos , Teste de Materiais , Células-Tronco Mesenquimais/metabolismo , Oligopeptídeos/química , Osteoblastos/metabolismo , Ácidos Fosforosos/química , Análise Espectral , Propriedades de Superfície , Titânio/química
20.
J Cell Physiol ; 234(4): 4179-4190, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30146726

RESUMO

Sendai virus strain Tianjin, a novel genotype of Sendai virus, has been proven to possess potent antitumor effect on certain cancer cell types although inactivated by ultraviolet (UV). This study was carried out to investigate the in vitro anticancer properties of UV-inactivated Sendai virus strain Tianjin (UV-Tianjin) on human osteosarcoma cells and the underlying molecular mechanism. Our studies demonstrated UV-Tianjin significantly inhibited the viability of human osteosarcoma cell lines and triggered apoptosis through activation of both extrinsic and intrinsic pathways in MG-63 cells. Meanwhile, autophagy occurred in UV-Tianjin-treated cells. Blockade of autophagy with 3-methyladenine remarkably attenuated the inhibition of cell proliferation by UV-Tianjin, suggesting that UV-Tianjin-induced autophagy may be contributing to cell death. Furthermore, UV-Tianjin induced reactive oxygen species (ROS) production, which was involved in the execution of MG-63 cell apoptosis and autophagy, as evidenced by the result that treatment of N-acetyl-L-cysteine, a ROS scavenger, attenuated both apoptosis and autophagy. In addition, inhibition of apoptosis promoted autophagy, whereas suppression of autophagy attenuated apoptosis. Our results suggest that UV-Tianjin triggers apoptosis and autophagic cell death via generation of the ROS in MG-63 cells, which might provide important insights into the effectiveness of novel strategies for osteosarcoma therapy.


Assuntos
Apoptose , Autofagia , Neoplasias Ósseas/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Osteossarcoma/terapia , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Vírus Sendai , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Neoplasias Ósseas/virologia , Linhagem Celular Tumoral , Proliferação de Células , Interações Hospedeiro-Patógeno , Humanos , Vírus Oncolíticos/efeitos da radiação , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Osteossarcoma/virologia , Vírus Sendai/efeitos da radiação , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA