Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Environ Toxicol ; 39(1): 299-313, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37705323

RESUMO

Increased neddylation benefits the survival of several types of cancer cells. The inhibition of neddylation has the potential to exert anticancer effects but is rarely assessed in oral cancer cells. This study aimed to investigate the antiproliferation potential of a neddylation inhibitor MLN4924 (pevonedistat) for oral cancer cells. MLN4924 inhibited the cell viability of oral cancer cells more than that of normal oral cells (HGF-1) with 100% viability, that is, IC50 values of oral cancer cells (CAL 27, OC-2, and Ca9-22) are 1.8, 1.4, and 1.9 µM. MLN4924 caused apoptotic changes such as the subG1 accumulation, activation of annexin V, pancaspase, and caspases 3/8/9 of oral cancer cells at a greater rate than in normal oral cells. MLN4924 induced greater oxidative stress in oral cancer cells compared to normal cells by upregulating reactive oxygen species and mitochondrial superoxide and depleting the mitochondrial membrane potential and glutathione. In oral cancer cells, preferential inductions also occurred for DNA damage (γH2AX and 8-oxo-2'-deoxyguanosine). Therefore, this investigation demonstrates that MLN4924 is a potential anti-oral-cancer agent showing preferential inhibition of apoptosis and promotion of DNA damage with fewer cytotoxic effects on normal cells.


Assuntos
Apoptose , Neoplasias Bucais , Humanos , Proliferação de Células , Linhagem Celular Tumoral , Neoplasias Bucais/metabolismo
2.
Clin Exp Pharmacol Physiol ; 47(5): 857-866, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31943292

RESUMO

Asparaginase is fundamental to the treatment of haematological malignancies. However, little has been studied on the effects that asparaginase could exert on solid tumours. Thus, this study aimed to evaluate the effects of asparaginase on an oral carcinoma cell line. The cytotoxicity of asparaginase in SCC-9 (tongue squamous cell carcinoma) and HaCaT (human keratinocyte) cell lines was evaluated with MTT cell viability assay. The cells were treated with asparaginase at 0.04, 0.16, 0.63, 1.0, 1.5, 2.5, and 5.0 IU/mL. Dose-response curves and IC50 values were obtained and the Tumour Selectivity Index (TSI) was calculated. The effect of asparaginase on procaspase-3 and nuclear factor κB (NFκB) expression was evaluated with western blot because it was reported that the overexpression of NFκB has been shown to contribute to tumour cell survival, proliferation, and migration. Caspase 3/7 staining was performed to identify cell death using flow cytometry. Effective asparaginase concentrations were lower for SCC-9 cells when compared to HaCaT cells. The cytotoxicity results at 48 and 72 hours were significantly different for SCC-9 cells. The TSI indicated that asparaginase was selective for the tumour cells. A decrease in procaspase-3 and NFκB protein levels was observed in SCC-9 cells. Furthermore, asparaginase resulted in significant apoptosis after 48 and 72 hours. Based on these results, asparaginase was cytotoxic in a dose- and time-dependent manner, induces apoptosis, and reduces NFκB expression in oral cancer cells. These results encourage further studies on the effectiveness of this enzyme as a treatment for solid tumours, especially head and neck cancer.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Asparaginase/farmacologia , NF-kappa B/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Neoplasias da Língua/tratamento farmacológico , Caspase 3/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Regulação para Baixo , Células HaCaT , Humanos , Transdução de Sinais , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Fatores de Tempo , Neoplasias da Língua/metabolismo , Neoplasias da Língua/patologia
3.
Toxicol Appl Pharmacol ; 382: 114734, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31470033

RESUMO

Curcumin (Cur), a natural product, has been shown to have anti-tumor activities in many human cancer cells. Gefitinib (Gef) is a clinical drug for cancer patients. However, there is no available information to show whether Gef/Cur nanoparticles (NPs) increased cell apoptosis and anti-tumor effects on xenograft mice model in vivo. In this study, γ-polyglutamic acid-coated nanoparticles loaded with Gef and Cur (γ-PGA-Gef/Cur NPs) were developed and its physicochemical properties and antitumor effects were investigated in vitro and in vivo. The γ-PGA-Gef/Cur NPs showed 548.5 ±â€¯93.7 nm in diameter and -40.3 ±â€¯3.87 mV on surface charge. The loading efficiencies of Gef and Cur were 89.5 and 100%, respectively. γ-PGA-Gef/Cur NPs could be internalized into SAS cells and significantly decreased total cell viability of SAS cells. Western blotting results indicated that both free Gef/Cur and γ-PGA-Gef/Cur NPs induced apoptotic cell death via caspase- and mitochondria-dependent pathways. In vivo studies indicated that treatments of PLGA NPs, free Gef/Cur, and γ-PGA-Gef/Cur NPs did not significantly affect appearances and bodyweights of mice. But the γ-PGA-Gef/Cur NPs significantly suppressed tumor size when comparing to free Gef/Cur-treated group. The nanoparticles developed in this study may be used as a potential therapy for oral cancer.


Assuntos
Apoptose/efeitos dos fármacos , Curcumina/administração & dosagem , Gefitinibe/administração & dosagem , Neoplasias Bucais/tratamento farmacológico , Nanopartículas/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Antineoplásicos/administração & dosagem , Apoptose/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Bucais/patologia , Inibidores de Proteínas Quinases/administração & dosagem
4.
Chin J Physiol ; 62(3): 123-130, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31249266

RESUMO

Chlorzoxazone is a skeletal muscle relaxant. However, the effect of chlorzoxazone on intracellular Ca2+ concentrations ([Ca2+]i) in oral cancer cells is unclear. This study examined whether chlorzoxazone altered Ca2+ signaling and cell viability in OC2 human oral cancer cells. [Ca2+]iin suspended cells was measured using the fluorescent Ca2+-sensitive dye fura-2. Cell viability was examined by water-soluble tetrazolium-1 assay. Chlorzoxazone (250-1000 µM) induced [Ca2+]irises in a concentration-dependent manner. Ca2+ removal reduced the signal by approximately 50%. Mn2+ has been shown to enter cells through similar mechanisms as Ca2+ but quenches fura-2 fluorescence at all excitation wavelengths. Chlorzoxazone (1000 µM) induced Mn2+ influx, suggesting that Ca2+ entry occurred. Chlorzoxazone-induced Ca2+ entry was inhibited by 20% by inhibitors of store-operated Ca2+ channels and protein kinase C (PKC) modulators. In Ca2+-free medium, treatment with the endoplasmic reticulum Ca2+ pump inhibitor thapsigargin (TG) inhibited chlorzoxazone-evoked [Ca2+]irises by 88%. Conversely, treatment with chlorzoxazone-suppressed TG-evoked [Ca2+]irises 75%. Chlorzoxazone induced [Ca2+]irises by exclusively releasing Ca2+ from the endoplasmic reticulum. Inhibition of phospholipase C (PLC) with U73122 did not alter chlorzoxazone-induced [Ca2+]irises. PLC activity was not involved in chlorzoxazone-evoked [Ca2+]irises. Chlorzoxazone at 200-700 µM decreased cell viability, which was not reversed by pretreatment with Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid/acetoxy methyl. In sum, in OC2 cells, chlorzoxazone induced [Ca2+]irises by evoking PLC-independent Ca2+ release from the endoplasmic reticulum and Ca2+ entry via PKC-sensitive store-operated Ca2+ entry. Chlorzoxazone also caused Ca2+-independent cell death. Since [Ca2+]irises play a triggering or modulatory role in numerous cellular phenomena, the effect of chlorzoxazone on [Ca2+]iand cell viability should be taken into account in other in vitro studies.


Assuntos
Sinalização do Cálcio , Neoplasias Bucais , Apoptose , Cálcio , Linhagem Celular Tumoral , Sobrevivência Celular , Clorzoxazona , Humanos , Fosfolipases Tipo C
5.
Biochem Biophys Res Commun ; 495(3): 2118-2124, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29253571

RESUMO

How to effectively delivering therapeutic agents, including γ-secretase inhibitors (GSIs), into live cells, remains a significant challenge. This study assessed the effect of Notch signaling inhibition by examining levels of the Notch1 intracellular domain (N1ICD) in cultured oral cancer cells analyzed with random stitched images (2D) and 3D visualizations using confocal microscopy and quantitative gene analysis. Substantially, we have developed a novel method to assist the delivery of γ-secretase inhibitor, DAPT, into live cells in the presence of an effective minimum concentration of Triton-X100 (0.001%) without damaging cell activity and membrane integrity assessed with cell proliferation assays. The images obtained in this study showed that DAPT alone could not block the γ-secretase inhibitor despite inhibiting cell growth. Further analysis of quantitative gene expressions of Notch signaling canonical pathway to verify the effectiveness of the novel method for delivering inhibitor into live cells, displayed deregulation of Notch1, Delta-like ligand 1 (DLL1) and hairy and enhancer of split 1 (Hes1). Our data suggest that Notch1/Hes1 signaling pathway is deactivated using DAPT with a low dose of Triton-X100 in this cancer cells. And the finding also suggests that Notch1 could be engaged by DLL1 to promote differentiation in oral cancer cells. Using this approach, we demonstrate that Triton-X100 is a promising and effective permeabilization agent to deliver γ-secretase inhibitor DAPT into live oral epithelial cells. This strategy has the potential to implicate in the treatment of cancer diseases.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Dipeptídeos/administração & dosagem , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/metabolismo , Octoxinol/administração & dosagem , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dipeptídeos/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Emulsões/química , Humanos , Neoplasias Bucais/patologia , Octoxinol/química , Octoxinol/farmacologia , Resultado do Tratamento
6.
Exp Cell Res ; 360(2): 404-412, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28943462

RESUMO

DNA polymeric films (DNA-PFs) are a promising drug delivery system (DDS) in modern medicine. In this study, we evaluated the growth behavior of oral squamous cell carcinoma (OSCC) cells on DNA-PFs. The morphological, biochemical, and cytometric features of OSCC cell adhesion on DNA-PFs were also assessed. An initial, temporary alteration in cell morphology was observed at early time points owing to the inhibition of cell attachment to the film, which then returned to a normal morphological state at later time points. MTT and resazurin assays showed a moderate reduction in cell viability related to increased DNA concentration in the DNA-PFs. Flow cytometry studies showed low cytotoxicity of DNA-PFs, with cell viabilities higher than 90% in all the DNA-PFs tested. Flow cytometric cell cycle analysis also showed average cell cycle phase distributions at later time points, indicating that OSCC cell growth is maintained in the presence of DNA-PFs. These results show high biocompatibility of DNA-PFs and suggest their use in designing "dressing material," where the DNA film acts as a support for cell growth, or with incorporation of active or photoactive compounds, which can induce tissue regeneration and are useful to treat many diseases, especially oral cancer.


Assuntos
Proliferação de Células , DNA/química , Membranas Artificiais , Polímeros/química , Medicina Regenerativa , Técnicas de Cultura de Tecidos/instrumentação , Alicerces Teciduais/química , Materiais Biocompatíveis/análise , Materiais Biocompatíveis/farmacologia , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , DNA/farmacologia , Humanos , Teste de Materiais , Neoplasias Bucais/patologia , Polímeros/farmacologia , Medicina Regenerativa/instrumentação , Medicina Regenerativa/métodos , Técnicas de Cultura de Tecidos/métodos
7.
Environ Toxicol ; 33(2): 127-141, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29098808

RESUMO

Casticin, a polymethoxyflavone, present in natural plants, has been shown to have biological activities including anti-cancer activities. Herein, we investigated the anti-oral cancer activity of casticin on SCC-4 cells in vitro. Viable cells, cell cycle distribution, apoptotic cell death, reactive oxygen species (ROS) production, and Ca2+ production, levels of ΔΨm and caspase activity were measured by flow cytometric assay. Cell apoptosis associated protein expressions were examined by Western blotting and confocal laser microscopy. Results indicated that casticin induced cell morphological changes, DNA condensation and damage, decreased the total viable cells, induced G2 /M phase arrest in SCC-4 cells. Casticin promoted ROS and Ca2+ productions, decreases the levels of ΔΨm , promoted caspase-3, -8, and -9 activities in SCC-4 cells. Western blotting assay demonstrated that casticin affect protein level associated with G2/M phase arrest and apoptosis. Confocal laser microscopy also confirmed that casticin increased the translocation of AIF and cytochrome c in SCC-4 cells. In conclusion, casticin decreased cell number through G2 /M phase arrest and the induction of cell apoptosis through caspase- and mitochondria-dependent pathways in SCC-4 cells.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Flavonoides/toxicidade , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Cálcio/metabolismo , Caspase 3/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Dano ao DNA/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Microscopia Confocal , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Espécies Reativas de Oxigênio/metabolismo
8.
Toxicol Appl Pharmacol ; 332: 81-91, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28782507

RESUMO

Antioxidant enzymes are related to oral diseases. We investigated the roles of heme oxygenase-1 (HO-1) and catalase in cadmium (Cd)-induced oxidative stress and the underlying molecular mechanism in oral cancer cells. Exposing YD8 cells to Cd reduced the expression levels of catalase and superoxide dismutase 1/2 and induced the expression of HO-1 as well as autophagy and apoptosis, which were reversed by N-acetyl-l-cysteine (NAC). Cd-exposed YD10B cells exhibited milder effects than YD8 cells, indicating that Cd sensitivity is associated with antioxidant enzymes and autophagy. Autophagy inhibition via pharmacologic and genetic modulations enhanced Cd-induced HO-1 expression, caspase-3 cleavage, and the production of reactive oxygen species (ROS). Ho-1 knockdown increased autophagy and apoptosis. Hemin treatment partially suppressed Cd-induced ROS production and apoptosis, but enhanced autophagy and CHOP expression, indicating that autophagy induction is associated with cellular stress. Catalase inhibition by pharmacological and genetic modulations increased Cd-induced ROS production, autophagy, and apoptosis, but suppressed HO-1, indicating that catalase is required for HO-1 induction. p38 inhibition upregulated Cd-induced phospho-JNK and catalase, but suppressed HO-1, autophagy, apoptosis. JNK suppression exhibited contrary results, enhancing the expression of phospho-p38. Co-suppression of p38 and JNK1 failed to upregulate catalase and procaspase-3, which were upregulated by JNK1 overexpression. Overall, the balance between the responses of p38 and JNK activation to Cd appears to have an important role in maintaining cellular homeostasis via the regulation of antioxidant enzymes and autophagy induction. In addition, the upregulation of catalase by JNK1 activation can play a critical role in cell protection against Cd-induced oxidative stress.


Assuntos
Autofagia/efeitos dos fármacos , Cádmio/toxicidade , Catalase/metabolismo , Heme Oxigenase-1/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Acetilcisteína/farmacologia , Antioxidantes/farmacologia , Caspase 3/genética , Caspase 3/metabolismo , Catalase/genética , Linhagem Celular Tumoral , Heme Oxigenase-1/genética , Humanos , Proteína Quinase 8 Ativada por Mitógeno/genética , Neoplasias Bucais/induzido quimicamente , Neoplasias Bucais/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/genética
9.
Cell Mol Biol (Noisy-le-grand) ; 63(10): 131-136, 2017 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-29096753

RESUMO

Oral cancer represents the sixth most common cancer type worldwide. Patients with oral cancer express high levels of IL-6 which is associated with very poor prognosis. Previous studies illustrated that IL-6 cytokine induces angiogenesis. It has also been reported that the presence of Cancer- Associated Fibroblasts (CAFs) is essential for angiogenesis. In this study, we examined the correlation between IL-6 and CAF and the role of this correlation on VEGF production. In this study, quantitative expression level of IL-6 and VEGF in CAF and Oral Cancer Cells (OCCs) examined through Real Time PCR and ELISA and western blot analysis. In addition, maintenance and retention of IL-6 and VEGF checked out in co-culture experiment of CAF and OCC cells. These experiments demonstrated that in oral cancer, CAF cell line secretes significantly more IL-6 than OCC. Also IL-6 is a factor that causes VEGF secretion in CAF cell line. CAF is the basic and the most essential source for producing IL-6 in patients with oral cancer. Secreted IL-6 is able to induce VEGF production in both CAF and OCCs. Correlation between CAF, IL-6 and VEGF could be considered as an approach for cancer therapy.


Assuntos
Interleucina-6/análise , Fator A de Crescimento do Endotélio Vascular/análise , Actinas/metabolismo , Técnicas de Cocultura , Ensaio de Imunoadsorção Enzimática , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolissacarídeos/toxicidade , Masculino , Microscopia de Fluorescência , Pessoa de Meia-Idade , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Reação em Cadeia da Polimerase em Tempo Real , Células Estromais/citologia , Células Estromais/metabolismo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vimentina/metabolismo
10.
Microb Pathog ; 99: 135-141, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27554277

RESUMO

The knowhow followed for synthesis, characterization and application of nanomaterials has become an important branch of nanoscience. The use of marine sponges for the synthesis of metal nanoparticles is still in the budding level of current nanobiotechnology. This paper reports a single step one pot biosynthesis utilizing marine sponge (Haliclona exigua) extract as a reducing agent by means of a conventional ultrasonic bath on the formation and growth of flower like silver nanocolloids. These silver nanocolloids were characterized through UV visible spectroscopy, High Resolution Transmission Electron Microscope, Fourier Transform Infrared spectroscopy and X-ray Diffractometer. Further, antibacterial activity and antiproliferative activity were done against oral biofilm bacteria and oral cancer cell lines for the biosynthesized flower like silver nanocolloids. Water soluble organic amines were responsible for the syntheses of nanomaterials which have a size range from 100 to 120 nm. An average size of 9.1 mm zone of inhibition was recorded with 10.0 µg of silver nanocolloids against oral biofilm bacteria. The estimated half maximal inhibitory concentration value for flower like silver nanocolloids was 0.6 µg/ml for oral cancer cell lines.


Assuntos
Anti-Infecciosos/farmacologia , Antineoplásicos/farmacologia , Biofilmes/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Nanoestruturas/química , Poríferos/química , Prata/metabolismo , Animais , Anti-Infecciosos/metabolismo , Antineoplásicos/metabolismo , Organismos Aquáticos/química , Bactérias/efeitos dos fármacos , Extratos Celulares/isolamento & purificação , Linhagem Celular Tumoral , Microscopia Eletrônica de Transmissão , Nanoestruturas/ultraestrutura , Prata/farmacologia , Espectroscopia de Infravermelho com Transformada de Fourier , Análise Espectral , Ondas Ultrassônicas , Difração de Raios X
11.
Environ Toxicol ; 31(12): 2045-2056, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26464283

RESUMO

Helioxanthin, an active compound from Taiwania cryptomerioides Hayata, has been shown to have various biological activities. However, their anticancer effect in oral squamous cell carcinoma has not been well established yet. Helioxanthin inhibited the proliferation of oral squamous cell carcinoma cells in a dose-dependent manner by inducing G2/M phase arrest. Similarly, helioxanthin inhibited cyclooxygenase-2, (COX-2), phosphorylated EGFR, and extracellular-signal-regulated kinases (ERK) protein level and further reduced the nuclear accumulation of phosphorylated epidermal growth factor receptor (pEGFR) and activator protein-1(AP-1) family protein, c-fos. Moreover, helioxanthin at the dose of 20 and 30 mg kg-1 for 15 days reduced the tumor growth in animal model. This study demonstrated that Helioxanthin exerts its anticancer activity against oral cancer cells by downregulating EGFR/ERK/c-fos signaling pathway to inhibit COX-2 level and by activating cyclin-dependent kinase inhibitor (p27) to further induce G2/M cell cycle arrest. This helioxanthin may serve as a novel candidate for oral cancer prevention. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 2045-2056, 2016.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Lignanas/farmacologia , Neoplasias Bucais/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Arecolina , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Xenoenxertos , Lignanas/uso terapêutico , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos Nus , Neoplasias Bucais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo
12.
Elife ; 122024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38567911

RESUMO

The antibiotic heliomycin (resistomycin), which is generated from Streptomyces resistomycificus, has multiple activities, including anticancer effects. Heliomycin was first described in the 1960s, but its clinical applications have been hindered by extremely low solubility. A series of 4-aminomethyl derivatives of heliomycin were synthesized to increase water solubility; studies showed that they had anti-proliferative effects, but the drug targets remained unknown. In this study, we conducted cellular thermal shift assays (CETSA) and molecular docking simulations to identify and validate that heliomycin and its water-soluble derivative, 4-(dimethylaminomethyl)heliomycin (designated compound 4-dmH) engaged and targeted with sirtuin-1 (SIRT1) in p53-functional SAS and p53-mutated HSC-3 oral cancer cells. We further addressed the cellular outcome of SIRT1 inhibition by these compounds and found that, in addition to SIRT1, the water-soluble 4-dmH preferentially targeted a tumor-associated NADH oxidase (tNOX, ENOX2). The direct binding of 4-dmH to tNOX decreased the oxidation of NADH to NAD+ which diminished NAD+-dependent SIRT1 deacetylase activity, ultimately inducing apoptosis and significant cytotoxicity in both cell types, as opposed to the parental heliomycin-induced autophagy. We also observed that tNOX and SIRT1 were both upregulated in tumor tissues of oral cancer patients compared to adjacent normal tissues, suggesting their clinical relevance. Finally, the better therapeutic efficacy of 4-dmH was confirmed in tumor-bearing mice, which showed greater tNOX and SIRT1 downregulation and tumor volume reduction when treated with 4-dmH compared to heliomycin. Taken together, our in vitro and in vivo findings suggest that the multifaceted properties of water-soluble 4-dmH enable it to offer superior antitumor value compared to parental heliomycin, and indicated that it functions through targeting the tNOX-NAD+-SIRT1 axis to induce apoptosis in oral cancer cells.


Assuntos
Neoplasias Bucais , Compostos Policíclicos , Sirtuína 1 , Humanos , Animais , Camundongos , Sirtuína 1/metabolismo , Linhagem Celular Tumoral , NAD/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Simulação de Acoplamento Molecular , Apoptose , Neoplasias Bucais/tratamento farmacológico
13.
Oncol Rep ; 50(4)2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37615224

RESUMO

Despite significant advancements in therapeutic approaches, oral neoplasms remain formidable and life­threatening conditions that affect a substantial number of individuals worldwide. Within oral malignancies, a subset of cancer stem cells (CSCs) represent a crucial population responsible for tumor initiation and progression. The identification of reliable markers for the detection and characterization of CSCs in solid tumors, particularly in the context of oral cancers, remains an ongoing challenge. Stage­specific embryonic antigen 3 (SSEA3), previously associated with mesenchymal stem cells and linked to the progression of breast neoplasms and poor prognosis, has yet to be comprehensively elucidated in the context of oral malignancies. The present study aimed to investigate the expression and properties of SSEA3 in 16 distinct subsets of human oral neoplastic cell lines, classified as either CD44 positive (+) or CD44 negative (­). For the first time, SSEA3 was examined as an indicator of tumorigenicity and resistance to taxane­derived chemotherapeutic agents. In the majority of oral neoplastic cell lines analyzed, SSEA3 was expressed in a small population of CD44(+) cells. Significantly, SSEA3(+) cells exhibited heightened proliferative activity and upregulated expression of genes associated with stem cells compared with SSEA3(­) cells. The aforementioned findings suggested that SSEA3 may contribute to the evolution and progression of oral malignancies by fostering tumor growth. Furthermore, SSEA3(+) cells displayed increased sensitivity to taxane­based pharmaceuticals, indicating the potential for SSEA3 to be a viable target in the treatment schema for oral cavity neoplasms. In conclusion, the present study provides novel insight into the role of SSEA3 in the progression and management of oral neoplasms, potentially paving the way for more effective therapeutic approaches.


Assuntos
Neoplasias Bucais , Humanos , Antígenos Embrionários Estágio-Específicos , Neoplasias Bucais/tratamento farmacológico , Transformação Celular Neoplásica , Linhagem Celular Tumoral , Células-Tronco Neoplásicas
14.
Anticancer Res ; 43(9): 3905-3911, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37648334

RESUMO

BACKGROUND/AIM: Cervical lymph node metastasis worsens oral cancer prognosis. Cancer cells with high metastatic ability can delay or resist apoptosis and survive in the floating condition during circulation. The involved genes and pathways in this process remain largely unknown. This study aimed to establish an oral cancer cell line adapted to suspension culture by in vitro selection and perform gene expression analysis. MATERIALS AND METHODS: The oral cancer cell subline adapted to suspension culture was isolated by in vitro selection from the oral cancer cell line, HSC-3. The transcriptome profiles of HSC-3 and its subline were compared using gene expression microarrays. Gene Ontology (GO) enrichment analysis, Gene Set Enrichment Analysis (GSEA), and Ingenuity Pathway Analysis (IPA) were performed to predict the involved pathways and molecules in cancer progression. RESULTS: The subline was designated as HSC-3S5 The cellular viability of HSC-3S5 cells at the suspension culture was higher than that of HSC-3 cells. A total of 961 genes were differentially expressed between HSC-3 and HSC-3S5 cells under the threshold cut-off (FDR-adjusted p-value of <0.05 and absolute fold change of >1.5). GO terms, such as growth regulation, were enriched in the DEGs. GSEA revealed the association between the DEGs and significant gene sets, including metastasis and stemness. IPA predicted that the proliferation-related pathways were enhanced while the apoptotic pathway was inhibited in HSC-3S5 cells compared to HSC-3 cells. CONCLUSION: Our transcriptome analysis revealed several potentially activated pathways and molecules in the floating-adapted oral cancer cells and indicated molecular implications for cancer progression.


Assuntos
Neoplasias Bucais , Transcriptoma , Humanos , Neoplasias Bucais/genética , Perfilação da Expressão Gênica , Testes Imunológicos , Apoptose/genética
15.
Mol Med Rep ; 27(2)2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36524366

RESUMO

Although arsenic is an environmental toxicant, arsenic trioxide (ATO) is used to treat acute promyelocytic leukemia (APL) with anticancer effects. Studies have demonstrated oral cancer is in the top 10 cancers in Taiwan. High rate of oral cancers is linked to various behaviors, such as excessive alcohol consumption and tobacco use. Similarly, betel chewing is a strong risk factor in oral cancer. In the present study, oral squamous carcinoma OC3 cells were investigated with the treatments of sodium arsenite (NaAsO2) and dimethylarsenic acid (DMA), respectively, to examine if arsenic compounds have anti­cancer efforts. It was found that 1 µM NaAsO2 and 1 mM DMA for 24 h induced rounded contours with membrane blebbing phenomena in OC3 cells, revealing cell apoptotic characteristics. In addition, NaAsO2 (10­100 µM) and DMA (1­100 mM) significantly decreased OC3 cell survival. In cell cycle regulation detected by flow cytometry, NaAsO2 and DMA significantly augmented percentage of subG1 and G2/M phases in OC3 cells, respectively. Annexin V/PI double staining assay was further used to confirm NaAsO2 and DMA did induce OC3 cell apoptosis. In mechanism investigation, western blotting assay was applied and the results showed that NaAsO2 and DMA significantly induced phosphorylation of JNK, ERK1/2 and p38 and then the cleavages of caspase­8, ­9, ­3 and poly ADP­ribose polymerase (PARP) in OC3 cells, dynamically. In conclusion, NaAsO2 and DMA activated MAPK pathways and then apoptotic pathways to induce OC3 oral cancer cell apoptosis.


Assuntos
Arsenicais , Neoplasias Bucais , Humanos , Ácido Cacodílico/farmacologia , Linhagem Celular Tumoral , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/patologia , Apoptose , Arsenicais/farmacologia
16.
J Dent Sci ; 18(3): 1280-1287, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37404653

RESUMO

Background/purpose: Econazole is an antifungal drug. Antifungal activity of econazole against non-dermatophyte molds was reported. Econazole inhibited Ca2+ channels and stimulated cytotoxicity in lymphoma and leukemia cells. Ca2+ cations are crucial second envoy that triggers various processes. This research was aimed to investigate action of econazole on Ca2+ levels and cytotoxicity in OC2 human oral cancer cells. Materials and methods: Cytosolic Ca2+ levels ([Ca2+]i) were detected employing fura-2 as a probe in a RF-5301PC spectrofluorophotometer (Shimadzu). Cytotoxicity was determined using 4-[3-[4-lodophenyl]-2-4(4-nitrophenyl)-2H-5-tetrazolio-1,3-benzene disulfonate] (WST-1) to detect fluorescence changes. Results: Econazole at 10-50 µmol/L provoked [Ca2+]i raises. Forty % of 50 µml//L econazole-induced signal was diminished when external Ca2+ was eliminated. The Ca2+ influx provoked by econazole was suppressed by different degrees by store-induced Ca2+ influx suppressors SKF96365 and nifedipine; GF109203X (a protein C [PKC] inhibitor); an extracellular signaling pathway (ERK) 1/2 blocker PD98059, and phospholipase A2 suppressor aristolochic acid, but was enhanced by phorbol 12-myristate 13 acetate (PMA; a PKC activator) by 18%. Without external Ca2+, econazole-caused [Ca2+]i raises were abolished by thapsigargin. In contrast, econazole partially suppressed the [Ca2+]i raises caused by thapsigargin. U73122 fell short to change econazole-caused [Ca2+]i responses. Econazole (10-70 µmol/L) elicited cytotoxicity in a dose-dependent fashion. Blockade of 50 µmol/L econazole-induced [Ca2+] rises with BAPTA/AM enhanced econazole-induced cytotoxicity by 72%. Conclusion: Econazole evoked [Ca2+]i raises and provoked cytotoxicity in a concentration-dependent manner in OC2 human oral cancer cells. In Ca2+-containing solution, BAPTA/AM enhanced 50 µmol/L econozole-induced cytotoxicity.

17.
Artif Cells Nanomed Biotechnol ; 51(1): 83-94, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36752159

RESUMO

The present research was carried out to look into therapeutic insight of biosynthesized silver nanoparticles (AgNPs) by leaf extract of Byttneria herbacea Roxb (BH). The analysis of biosynthesized BH-AgNPs by UV-visible spectroscopy shows an intense surface plasmon resonance (SPR) peak at 422 nm initially and 437 nm after 30 min which certainly reveals the formation of BH-AgNPs. Fourier Infra-red Spectroscopy (FT-IR) reveals that BH-AgNPs are biosynthesized by using different bioactive compounds like O-H stretch of free hydroxyl alcohol and phenols, N-H bond of primary amines present in the leaf extract. Transmission Electron Microscope (TEM) analysis revealed that BH-AgNPs are almost spherical in nature with an average size range from of 2 nm to 12 nm. The particle size analysis by Dynamic Light Scattering (DLS) reveals that the BH-AgNPs are poly-dispersed in nature with an average size of 8 nm ± 2 nm, with a negative zeta potential value of -21 mV which reveals the biosynthesized BH-AgNPs are very stable. The BH-AgNPs (Byttneria herbacea -AgNPs) revealed excellent free radical scavenging activity and exceptional antimicrobial activity. The anti-proliferative and cytotoxic studies in KB oral cancer cells revealed biosynthesized BH-AgNPs can employ as future novel therapeutic agents in cancer treatment and other biomedical applications.


Assuntos
Nanopartículas Metálicas , Neoplasias Bucais , Humanos , Nanopartículas Metálicas/química , Prata/química , Espectroscopia de Infravermelho com Transformada de Fourier , Extratos Vegetais/química , Antibacterianos/química
18.
Front Oncol ; 12: 824043, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35494068

RESUMO

Z-ligustilide (or ligustilide) is found in Angelica sinensis (Oliv.) Diels and may exert potential benefits in cancer treatment. Previous research has reported that ligustilide has anti-cancer effects on several types of cancer cells. However, studies of ligustilide on oral cancer cells have not been reported, especially under hypoxic conditions. This study focuses on the molecular mechanism of ligustilide-induced apoptosis in hypoxic oral cancer cells. We found that in hypoxic TW2.6 cells, ligustilide inhibited cell migration and induced caspase-dependent apoptosis. Accumulation of c-Myc accompanied by BH3-only members suggests that ligustilide may induce c-Myc-dependent apoptosis. In addition, we reported that ligustilide has an effect on ER-stress signaling. By using inhibitors of c-Myc, IRE1α, and ER-stress inhibitors, we found that cell morphologies or cell viability were rescued to some degree. Moreover, ligustilide is able to increase the expression of γ-H2AX and enhance the occurrence of DNA damage in oral cancer cells after radiation treatment. This result suggests that ligustilide has potential as a radiation sensitizer. Altogether, we propose that ligustilide may induce c-Myc-dependent apoptosis via ER-stress signaling in hypoxic oral cancer cells.

19.
Antioxidants (Basel) ; 11(10)2022 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-36290795

RESUMO

A novel nitrated [6,6,6]tricycles-derived compound containing nitro, methoxy, and ispropyloxy groups, namely SK1, was developed in our previous report. However, the anticancer effects of SK1 were not assessed. Moreover, SK1 contains two nitro groups (NO2) and one nitrogen-oxygen (N-O) bond exhibiting the potential for oxidative stress generation, but this was not examined. The present study aimed to evaluate the antiproliferation effects and oxidative stress and its associated responses between oral cancer and normal cells. Based on the MTS assay, SK1 demonstrated more antiproliferation ability in oral cancer cells than normal cells, reversed by N-acetylcysteine. This suggests that SK1 causes antiproliferation effects preferentially in an oxidative stress-dependent manner. The oxidative stress-associated responses were further validated, showing higher ROS/MitoSOX burst, MMP, and GSH depletion in oral cancer cells than in normal cells. Meanwhile, SK1 caused oxidative stress-causing apoptosis, such as caspases 3/8/9, and DNA damages, such as γH2AX and 8-OHdG, to a greater extent in oral cancer cells than in normal cells. Siilar to cell viability, these oxidative stress responses were partially diminished by NAC, indicating that SK1 promoted oxidative stress-dependent responses. In conclusion, SK1 exerts oxidative stress, apoptosis, and DNA damage to a greater extent to oral cancer cells than in normal cells.

20.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 40(1): 22-31, 2022 Jan 25.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-38596989

RESUMO

OBJECTIVES: In this study, we aimed to investigate whether oral cancer cells affect pancreatic ß-cells function through transmissible endoplasmic reticulum stress (TERS). METHODS: Tunicamycin (TM) was selected as the endoplasmic reticulum stress (ERS) inducer. The human oral cancer cell lines CAl-27 and SCC-25 were selected as the donor cells, and mouse insulinoma 6 (MIN6) cell lines were chosen as the recipient cells. Quantitative real-time polymerase chain reaction (qPCR) and Western blot (WB) analysis were used to detect ERS markers and insulin expression. The TdT-mediated dUTP nick-end labeling (TUNEL) method was applied to detect apoptosis levels. The clone formation method was utilized to detect cell proliferation capability. The secretory function of pancreatic ß-cells was detected with an enzyme linked immunosorbent assay (ELISA) kit and a bicinchoninic acid (BCA) kit. RESULTS: The MIN6 cells were subjected to TM stimulation. qPCR and WB analysis revealed that ERS markers were upregulated. This result implied that the MIN6 cells can induce ERS. The supernatant of oral cancer cells under ERS was added to the MIN6 cells. qPCR and WB analysis showed that the oral cancer cells that had been subjected to ERS could induce ERS in the MIN6 cells, that is, the phenomenon of TERS occurred. The TUNEL assay indicated that the apoptosis of the MIN6 cells increased under TERS. The clone formation assay demonstrated that the proliferation capability of the MIN6 cells decreased under TERS. qPCR and WB analysis revealed that under TERS, insulin synthesis by the MIN6 cells decreased and insulin synthesis was inhibited at the translation level. The ELISA and BCA kits demonstrated that insulin secretion by the MIN6 cells was reduced under TERS. CONCLUSIONS: Oral cancer cells can affect pancreatic ß-cells through TERS, resulting in increased apoptosis, decreased viability, and reduced insulin secretion and synthesis capability.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa