Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Altern Ther Health Med ; 29(8): 594-600, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37678860

RESUMO

Background: Since most patients with oral cancer do not benefit from current treatments, new therapeutic strategies or drugs must be developed to improve patient prognosis. Qing Yan Li Ge Tang (QYLGT), a Chinese herbal medicine, is known for its anticancer activity. This study aimed to investigate whether QYLGT has anticancer effects on human OEC-M1 oral cancer cells. Methods: To evaluate whether QYLGT affects viability, morphology, and colony formation ability of the OEC-M1 cells, the 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, morphology study, and colony formation assay were performed, respectively. Each assay was carried out in triplicate, and the whole set of experiments was performed three times independently. To investigate whether QYLGT induces apoptotic effects in OEC-M1 cells, the enzyme-linked immunosorbent (ELISA) was carried out to quantify cytokeratin 18 fragment (an apoptosis marker). Each assay was carried out in triplicate, and the whole set of experiments was performed three times independently. The immunoblotting assay was performed to detect the protein expression after QYLGT treatment. The whole set of experiments was performed two times independently. Results: The results from the MTT and colony formation assays indicate that QYLGT inhibited the cell viability and clonogenic growth capacity of OEC-M1 cells. The morphology study shows that QYLGT increased plasma membrane blebbing in OEC-M1 clles. The results of ELISA and an immunoblotting assay show that QYLGT increased cytokeratin 18 fragment release and poly ADP-ribose polymerase cleavage (another apoptosis marker) in OEC-M1 cells. In addition, the results from immunoblotting assay show that QYLGT also activated apoptotic executor proteins, including caspase-8, caspase-9, and caspase-3, and the results of ELISA indicate that treatment with the pan-caspase inhibitor, Z-VAD-FMK, inhibited QYLGT-induced cytokeratin 18 fragment release. These results indicate that QYLGT inhibited cell viability in OEC-M1 cells and induced OEC-M1 apoptosis through caspase activation. Additionally, QYLGT-activated c-Jun N-terminal kinase, extracellular signal-regulated kinase, p38 mitogen-activated protein kinase, and nuclear factor-kappa B (NF-κB), and the related inhibitors, including SP600125, PD184352, SB202190, and Bay11-7082, were used to confirm which signaling was involved in QYLGT-induced apoptosis. Moreover, only Bay11-7082, the NF-κB inhibitor, could suppress QYLGT-induced the release of cytokeratin 18 fragments from OEC-M1 cells. Conclusions: QYLGT induced apoptosis in OEC-M1 cells via the NF-κB pathway.


Assuntos
Neoplasias Bucais , NF-kappa B , Humanos , NF-kappa B/metabolismo , NF-kappa B/farmacologia , Queratina-18/farmacologia , Apoptose , Neoplasias Bucais/tratamento farmacológico , Linhagem Celular Tumoral
2.
Langmuir ; 38(48): 14712-14724, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36420594

RESUMO

A chitosan layer was covalently bonded to a polyetheretherketone (PEEK) surface using a simple facile self-assembly method to address inadequate biological activity and infection around the implant. The surface characterization, layer degradation, biological activity, and antibacterial adhesion properties of chitosan-modified PEEK (PEEK-CS) were studied. Through chitosan grafting, the surface morphology changed, the surface roughness increased, and the contact angle decreased significantly. PEEK-CS boosted cell adhesion, proliferation, increased alkaline phosphate activity, extracellular matrix mineralization, and expression of osteogenic genes. PEEK-CS demonstrated less adhesion to Porphyromonas gingivalis as well as less bacterial adhesion to P. gingivalis and Streptococcus mutans. According to our findings, chitosan modification significantly improved the osteogenic ability and antibacterial adhesion of PEEK in vitro.


Assuntos
Quitosana , Quitosana/farmacologia , Polímeros/farmacologia , Cetonas/farmacologia , Polietilenoglicóis/farmacologia , Osteogênese , Antibacterianos/farmacologia , Propriedades de Superfície
3.
Biochem Biophys Res Commun ; 503(3): 2160-2166, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30082032

RESUMO

Clinical studies suggest a positive association between malignant progression of nasopharyngeal carcinoma (NPC) and Rta, a transcription factor of Epstein-Barr virus (EBV). However, Rta induces cellular senescence in vitro. To provide an underlying mechanism integrating these clues, we adapted a concept of senescence-associated secretory phenotype (SASP), based on which senescent cells facilitate tumor progression through paracrine. First, Rta-expressing NPC cells themselves show reduced invasiveness but promote invasion of Rta-negative tumor cells through secreted factors. Secretion of matrix metalloproteinase 9 (MMP9), an SASP protein, is increased by Rta, which requires the C-terminus of Rta and Rta-induced activation of E2F. Furthermore, the Rta-induced, paracrine-mediated pro-invasive effect is blocked upon knockdown of MMP9 expression or treatment with an MMP9 inhibitor. This study not only indicates that Rta can contribute to NPC progression through paracrine but also supports that MMP9 is a potential therapeutic target to prevent NPC metastasis.


Assuntos
Efeito Espectador , Proteínas Imediatamente Precoces/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Comunicação Parácrina , Transativadores/metabolismo , Células Cultivadas , Humanos , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/patologia
4.
Neurochem Res ; 43(5): 1058-1066, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29671236

RESUMO

Reactive astrocyte-mediated neuroinflammatory responses in the spinal dorsal horn have been reported to play a pivotal role in pathological pain. Chronic constriction injury (CCI) enhances the activation of nuclear factor kappa B (NF-κB), which is involved in neuropathic pain (NP). Picroside II (PII), a major active component of Picrorhiza scrophulariiflora, has been investigated for its anti-oxidative, anti-inflammatory, and anti-apoptotic activities. Here, we explored the analgesic effects of PII on a model of CCI-induced NP and investigated the levels of the GFAP protein and the mRNA and protein levels of pro-inflammatory cytokines in the spinal cord, including interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). CCI significantly induced mechanical allodynia and thermal hyperalgesia. Intraperitoneal administration of PII remarkably reversed the CCI-induced mechanical allodynia and thermal hyperalgesia and reduced the mRNA and protein levels of IL-1ß, IL-6, and TNF-α in the spinal cord. Additionally, according to the in vitro data, the PII treatment inhibited LPS-induced increases in the mRNA and protein levels of IL-1ß, IL-6, and TNF-α and suppressed the NF-κB pathway by inhibiting the phosphorylation of NF-κB/p65 and the degradation of inhibitor of NF-κB (IκB) in astrocytes without toxicity to astrocytes. Overall, the analgesic effect of PII correlated with the inhibition of spinal reactive astrocyte-mediated neuroinflammation through the NF-κB pathway in rats with NP.


Assuntos
Analgésicos/uso terapêutico , Astrócitos/efeitos dos fármacos , Cinamatos/uso terapêutico , Glucosídeos Iridoides/uso terapêutico , NF-kappa B/efeitos dos fármacos , Neuralgia/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Animais , Astrócitos/patologia , Células Cultivadas , Constrição Patológica/complicações , Citocinas/metabolismo , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Glial Fibrilar Ácida/genética , Hiperalgesia/tratamento farmacológico , Inflamação/patologia , Inflamação/prevenção & controle , Masculino , Neuralgia/etiologia , Neuralgia/patologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/patologia
5.
Int Immunopharmacol ; 130: 111796, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38452412

RESUMO

There is a reciprocal comorbid relationship between periodontitis and type 2 diabetes mellitus (T2DM). Recent studies have suggested that mitochondrial dysfunction (MD) could be the key driver underlying this comorbidity. The aim of this study is to provide novel understandings into the potential molecular mechanisms between MD and the comorbidity, and identify potential therapeutic targets for personalized clinical management. MD-related differentially expressed genes (MDDEGs) were identified. Enrichment analyses and PPI network analysis were then conducted. Six algorithms were used to explore the hub MDDEGs, and these were validated by ROC analysis and qRT-PCR. Co-expression and potential drug targeting analyses were then performed. Potential biomarkers were identified using LASSO regression. The immunocyte infiltration levels in periodontitis and T2DM were evaluated via CIBERSORTx and validated in mouse models. Subsequently, MD-related immune-related genes (MDIRGs) were screened by WGCNA. The in vitro experiment verified that MD was closely associated with this comorbidity. GO and KEGG analyses demonstrated that the connection between periodontitis and T2DM was mainly enriched in immuno-inflammatory pathways. In total, 116 MDDEGs, eight hub MDDEGs, and two biomarkers were identified. qRT-PCR revealed a distinct hub MDDEG expression pattern in the comorbidity group. Altered immunocytes in disease samples were identified, and their correlations were explored. The in vivo examination revealed higher infiltration levels of inflammatory immunocytes. The findings of this study provide insight into the mechanism underlying the gene-mitochondria-immunocyte network and provide a novel reference for future research into the function of mitochondria in periodontitis and T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Doenças Mitocondriais , Periodontite , Animais , Camundongos , Algoritmos , Biomarcadores , Biologia Computacional
6.
Biochem Biophys Res Commun ; 436(4): 672-6, 2013 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-23770367

RESUMO

WW domain-containing oxidoreductase (WOX1) participates in tumor suppression and many other biologic functions, but its molecular and functional interactions with viral proteins remain largely unknown. This study reveals that WOX1 is physically associated with latent membrane protein 2A (LMP2A), an oncoprotein of Epstein-Barr virus. The molecular interaction involves the tyrosine residue 33 of WOX1 and the proline-rich motifs of LMP2A. Interestingly, endogenous WOX1 is required for some LMP2A-triggered, cancer-promoting effects, including activation of extracellular signal-regulated kinase-1/2, upregulation of matrix metalloproteinase 9 (MMP9) and promotion of cell invasion. Upon knockdown of endogenous WOX1, LMP2A-triggered MMP9 induction is restored by exogenous wild-type WOX1, but not by a WOX1 mutant defective in LMP2A binding. These results indicate that, through interaction with LMP2A, WOX1 is involved in MMP9 induction, suggesting a novel role of WOX1 in Epstein-Barr virus-associated cancer progression.


Assuntos
Metaloproteinase 9 da Matriz/metabolismo , Oxirredutases/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Regulação para Cima , Proteínas da Matriz Viral/metabolismo , Sequência de Bases , Humanos , Invasividade Neoplásica , Oxirredutases/genética , RNA Interferente Pequeno , Proteínas Supressoras de Tumor/genética , Oxidorredutase com Domínios WW
7.
J Virol ; 86(12): 6656-67, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22514348

RESUMO

Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC) is highly metastatic, and this malignant feature may be promoted by an EBV oncoprotein, latent membrane protein 2A (LMP2A). Acting as a signal regulator, LMP2A can enhance invasiveness and motility of epithelial cells. Downstream from the LMP2A-triggered signaling events, it is largely unknown what key effector proteins are induced and essentially promote cell invasion. In the present study, we found that in NPC cells, LMP2A upregulated matrix metalloproteinase 9 (MMP9), a metastasis-associated protease. LMP2A increased MMP9 expression at both the mRNA and protein levels. It also activated the MMP9 promoter, in which two AP-1 elements were required for the promoter activation. Among AP-1 transcription factors, Fra-1 was induced by LMP2A and is essential for LMP2A-triggered MMP9 expression. Induction of Fra-1 was dependent on the LMP2A-activated ERK1/2 pathway, and induction of the ERK1/2-Fra-1-MMP9 axis required PY motifs in the amino-terminal domain of LMP2A. Notably, LMP2A-promoted invasion of NPC cells was blocked when MMP9 expression, Fra-1 induction, or ERK1/2 activation was inhibited. In addition, we found an association of LMP2A with MMP9 expression in NPC tumor biopsy specimens, where Fra-1 was a major mediation factor. This study reveals an underlying mechanism of LMP2A-induced cell invasion, from signal transduction to upregulation of a critical protease. Considering that MMP9 can also be upregulated by another EBV oncoprotein, LMP1, this protease may be a pivotal effector at which the EBV-induced, invasion-promoting mechanisms converge, serving as an attractive therapeutic target for NPC treatment.


Assuntos
Infecções por Vírus Epstein-Barr/enzimologia , Herpesvirus Humano 4/metabolismo , Sistema de Sinalização das MAP Quinases , Metaloproteinase 9 da Matriz/metabolismo , Neoplasias Nasofaríngeas/enzimologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas da Matriz Viral/metabolismo , Carcinoma , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/genética , Humanos , Metaloproteinase 9 da Matriz/genética , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/virologia , Proteínas Proto-Oncogênicas c-fos/genética , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional , Regulação para Cima , Proteínas da Matriz Viral/genética
9.
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
10.
Anticancer Res ; 43(9): 4015-4022, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37648299

RESUMO

BACKGROUND/AIM: Cisplatin is a drug for treating oral cancer. However, several previous studies indicate that oral cancer cells can develop resistance to cisplatin, which may result in a poor prognosis for patients with oral cancer. Fucoidan, a natural health product extracted from brown seaweed, has anticancer abilities against various types of cancer cell. This study evaluated whether fucoidan can enhance the sensitivity of oral cancer cells to cisplatin and explored the underlying mechanism. MATERIALS AND METHODS: SCC-25 cells were used in the present study and treated with 0.3125 mg/ml fucoidan, 12.5 µg/ml cisplatin, or 0.3125 mg/ml fucoidan plus 12.5 µg/ml cisplatin for 48 h, 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, enzyme-linked immunosorbent, and immunoblotting assays were performed to evaluate cell survival, cytokeratin-18 fragment release, and expression of markers of apoptosis and autophagy, respectively. RESULTS: Cotreatment with fucoidan enhanced cisplatin-induced reduction of SCC-25 cell survival compared to cisplatin alone. In addition, cotreatment also increased the expression of apoptosis markers, including activated caspase-8, activated caspase-9, activated caspase-3, and cleaved poly(ADP-ribose) polymerase (PARP), but did not increase the expression of the two autophagy markers studied, beclin and autophagy-related 12-autophagy-related 5 conjugate. Fucoidan significantly inhibited cisplatin-induced AKT serine/threonine kinase 1 activation, which promoted PARP cleavage, caspase-3 activation, and cytokeratin-18 fragment expression in SCC-25 cells. CONCLUSION: Fucoidan promoted cisplatin-induced effects by inhibiting phosphatidylinositol 4,5 bisphosphate 3 kinase/AKT serine/threonine kinase 1 activation induced by cisplatin. The results of this study may provide a basis for the possible application of the combination of fucoidan and cisplatin in the clinical treatment of oral cancer in the future to improve the prognosis of patients with oral cancer.


Assuntos
Cisplatino , Neoplasias Bucais , Humanos , Caspase 3 , Cisplatino/farmacologia , Queratina-18 , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Inibidores de Poli(ADP-Ribose) Polimerases , Neoplasias Bucais/tratamento farmacológico , Serina
11.
Anticancer Res ; 43(6): 2583-2591, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37247927

RESUMO

BACKGROUND/AIM: Although clinical medicine has significantly progressed in treating nasopharyngeal carcinoma (NPC) in recent years, many patients still have poor prognoses due to distant metastasis. It is still relatively unclear why NPC has a highly metastatic ability. Especially whether the tumor microenvironment affects the invasion and metastasis of NPC still needs to be cleared. In this study, serum starvation was used to simulate nutrient deficiency in the tumor microenvironment to explore whether nutrient deficiency affects the malignancy of NPC cells. MATERIALS AND METHODS: Semiquantitative reverse transcription-polymerase chain reaction, ELISA, immunoblotting assay, reporter gene assay, and Matrigel invasion assay were carried out. RESULTS: Under serum starvation, NPC cells could induce the mRNA expression and protein secretion of matrix metalloproteinase 9 (MMP9). The ERK-AP1 pathway was activated under serum starvation in NPC cells, resulting in the expression of MMP9. In contrast, treatment with an MMP9 inhibitor or an MMP9 siRNA inhibited serum starvation-induced invasion. CONCLUSION: Serum starvation could up-regulate MMP9 expression in NPC cells, contributing to NPC invasion. Therefore, serum starvation may promote malignancy of NPC cells but also support MMP9 as a potential therapeutic target to prevent NPC cell invasion and metastasis.


Assuntos
Metaloproteinase 9 da Matriz , Neoplasias Nasofaríngeas , Humanos , Carcinoma Nasofaríngeo/genética , Metaloproteinase 9 da Matriz/metabolismo , Neoplasias Nasofaríngeas/patologia , Sistema de Sinalização das MAP Quinases , RNA Interferente Pequeno/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Invasividade Neoplásica/patologia , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral
12.
Anticancer Res ; 43(11): 4879-4885, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37910001

RESUMO

BACKGROUND/AIM: Current NPC treatment methods have improved the 5-year survival rates of patients; however, some patients do not benefit from the treatments. Therefore, the existing treatment methods or new drugs must be developed to improve the patient's prognosis. NAD (P)H:quinone oxidoreductase 1 (NQO1), an electron reductase highly expressed in various cancers, can convert aziridinyl-substituted quinone-derived compound into an alkylating agent, resulting in cell apoptosis. Therefore, a di-aziridinyl-substituted quinone-derived compound, AZ-1, was designed previously. The present study investigated whether AZ-1 has anticancer activities in NPC cells and explored the underlying mechanism. MATERIALS AND METHODS: NPC-TW01 cells were used in the study, and 3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide, colony formation, terminal deoxynucleotidyl transferase dUTP nick end labeling, and immunoblotting assays were performed to assess the cell viability, cell survival, DNA fragmentation, and protein expression, respectively. RESULTS: The results show that AZ-1 significantly inhibited the viability and survival of NPC-TW01 cells. AZ-1 also induced the expression of cleaved PARP, cleaved caspase-8, cleaved caspase-9, and cleaved caspase-3, and triggered DNA fragmentation in NPC-TW01 cells. In addition, AZ-1 induced γH2AX expression, a DNA damage marker, in NPC-TW01 cells. Treatment with dicoumarol, an NQO1 activity inhibitor, not only reversed AZ-1-induced cell viability inhibition but also decreased AZ-1-induced expression of γH2AX, cleaved caspase-8, cleaved caspase-9, and cleaved caspase-3. CONCLUSION: NQO1 reverses AZ-1-triggered cell viability inhibition, DNA damage, and apoptosis. The findings of this study may provide a basis for the possible clinical application of AZ-1 in the treatment of NPC to improve the prognosis of patients with NPC.


Assuntos
NAD(P)H Desidrogenase (Quinona) , NAD , Neoplasias Nasofaríngeas , Humanos , Caspase 3 , Caspase 8 , Caspase 9 , Carcinoma Nasofaríngeo/tratamento farmacológico , Neoplasias Nasofaríngeas/tratamento farmacológico , Quinonas , NAD(P)H Desidrogenase (Quinona)/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/metabolismo
13.
Int J Oncol ; 60(2)2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35029282

RESUMO

For a number of years, oral cancer has remained in the top ten most common types of cancer, with an incidence rate that is steadily increasing. In total, ~75% oral cancer cases are associated with lifestyle factors, including uncontrolled alcohol consumption, betel and tobacco chewing, and the excessive use of tobacco. Notably, betel chewing is highly associated with oral cancer in Southeast Asia. Arsenic is a key environmental toxicant; however, arsenic trioxide has been used as a medicine for the treatment of acute promyelocytic leukemia, highlighting its anticancer properties. The present study aimed to investigate the role of arsenic compounds in the treatment of cancer, using FaDu oral squamous carcinoma cells treated with sodium arsenite (NaAsO2) and dimethyl arsenic acid (DMA). The results demonstrated that FaDu cells exhibited membrane blebbing phenomena and high levels of apoptosis following treatment with 10 µM NaAsO2 and 1 mM DMA for 24 h. The results of cell viability assay demonstrated that the rate of FaDu cell survival was markedly reduced as the concentration of arsenic compounds increased from 10 to 100 µM NaAsO2, and 1 to 100 mM DMA. Moreover, flow cytometry was carried out to further examine the effects of arsenic compounds on FaDu cell cycle regulation; the results revealed that treatment with NaAsO2 and DMA led to a significant increase in the percentage of FaDu cells in the sub­G1 and G2/M phases of the cell cycle. An Annexin V/PI double staining assay was subsequently performed to verify the levels of FaDu cell apoptosis following treatment with arsenic compounds. Furthermore, the results of the western blot analyses revealed that the expression levels of caspase­8, ­9 and ­3, and poly ADP­ribose polymerase, as well the levels of phosphorylated JNK and ERK1/2 were increased following treatment with NaAsO2 and DMA in the FaDu cells. On the whole, the results of the present study revealed that treatment with NaAsO2 and DMA promoted the apoptosis of FaDu oral cancer cells, by activating MAPK pathways, as well as the extrinsic and intrinsic apoptotic pathways.


Assuntos
Apoptose/efeitos dos fármacos , Arsênio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Arsênio/metabolismo , Caspases/metabolismo , Caspases/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/farmacologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/fisiopatologia
14.
Artigo em Inglês | MEDLINE | ID: mdl-38023774

RESUMO

Cordycepin, a bioactive compound extracted from Cordyceps sinensis, can induce apoptosis in human OEC-M1 oral cancer cells. However, the exact mechanism is still unclear. The present study aimed to investigate the underlying mechanism of cordycepin-induced apoptosis in OEC-M1 cells. Following treatment with cordycepin, apoptosis was examined and quantified using a DNA laddering assay and a cytokeratin 18 fragment enzyme-linked immunosorbent assay, respectively. Expressions of mitogen-activated protein kinases (MAPKs) and apoptosis-related proteins were detected by the western blot analysis. Our results show that a pan-caspase inhibitor, Z-VAD-FMK, could significantly inhibit cordycepin-induced apoptosis in OEC-M1 cells. In addition, treatment with cordycepin not only activated caspase-8, caspase-9, and caspase-3 but also induced Bid and poly ADP-ribose polymerase cleavages. Furthermore, cordycepin also induced the activation of c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase, and p38 MAPKs. Among MAPKs, activation of JNK solely contributed to cordycepin-induced apoptosis with the activation of caspase-8, caspase-9, and caspase-3 and cleavage of PARP. Taken together, the present study demonstrated that cordycepin activated JNK and caspase pathways to induce apoptosis in OEC-M1 cells.

15.
Oncol Lett ; 22(4): 705, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34457060

RESUMO

It has been reported that paclitaxel activates cell cycle arrest and increases caspase protein expression to induce apoptosis in head and neck squamous cell carcinoma (HNSCC) cell lines. However, the potential signaling pathway regulating this apoptotic phenomenon remains unclear. The present study used OEC-M1 cells to investigate the underlying molecular mechanism of paclitaxel-induced apoptosis. Following treatment with paclitaxel, cell viability was assessed via the MTT assay. Necrosis, apoptosis, cell cycle and mitochondrial membrane potential (∆Ψm) were analyzed via flow cytometric analyses, respectively. Western blot analysis was performed to detect the expression levels of proteins associated with the MAPK and caspase signaling pathways. The results demonstrated that low-dose paclitaxel (50 nM) induced apoptosis but not necrosis in HNSCC cells. In addition, paclitaxel activated the c-Jun N-terminal kinase (JNK), but not extracellular signal-regulated kinase or p38 mitogen-activated protein kinase. The paclitaxel-activated JNK contributed to paclitaxel-induced apoptosis, activation of caspase-3, -6, -7, -8 and -9, and reduction of ∆Ψm. In addition, caspase-8 and -9 inhibitors, respectively, significantly decreased paclitaxel-induced apoptosis. Notably, Bid was truncated following treatment with paclitaxel. Taken together, the results of the present study suggest that paclitaxel-activated JNK is required for caspase activation and loss of ∆Ψm, which results in apoptosis of HNSCC cells. These results may provide mechanistic basis for designing more effective paclitaxel-combining regimens to treat HNSCC.

16.
Artigo em Inglês | MEDLINE | ID: mdl-34765012

RESUMO

Since a portion of patients with nasopharyngeal carcinoma (NPC) do not benefit much from current standard treatments, it is still needed to discover new therapeutic drugs to improve the prognosis of the patients. Considering that Chinese traditional medicine plays a role in inhibiting tumor progression, in this study, we aimed to investigate whether a Chinese herbal formula, Qing Yan Li Ge Tang (QYLGT), has the anticancer activity in NPC cells and explore the underlying mechanism as well. MTT assay, colony formation assay, immunoblotting assay, and DNA laddering assay were performed to assess cell viability, cell colony formation, protein expression, and DNA fragmentation, respectively. Results show that QYLGT was able to inhibit the cell viability and decrease colony formation ability in NPC cells. QYLGT could also increase the formation of intracellular vacuoles and induce the autophagy-related protein expressions, including Atg3, Atg6, and Atg12-Atg5 conjugate in NPC cells. Treatment with an autophagy inhibitor, 3-methyladenine, could significantly recover QYLGT-inhibited cell viability of NPC cells. In addition, QYLGT did not significantly induce apoptosis in NPC cells. We also found that QYLGT had the ability to activate phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of the rapamycin (mTOR) pathway. Treatment with PI3K inhibitors, LY294002 and wortmannin, or mTOR inhibitors, rapamycin and Torin 1, could not only recover QYLGT-inhibited cell viability of NPC cells but also inhibit Atg3 expression. Taken together, our results demonstrated that QYLGT could induce autophagic cell death in NPC cells through the PI3K/Akt/mTOR pathway.

17.
Microsc Res Tech ; 83(12): 1558-1565, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33220004

RESUMO

This vitro study aimed to evaluate the effects of 45S5 bioactive glass (BAG) and Er:YAG laser as desensitization treatments on the microtensile bond strength (MTBS) of fluorosed teeth. The 120 noncarious fluorosis were to obtain superficial dentin, being classified into four groups according to the Thylstrup and Fejerskov Index (TFI). Specimens from each group were randomly divided into five subgroups. After fluorosed teeth hypersensitivity models were established, the following pretreatments were applied on dentine surface: Subgroup 1: deionized water (Control); Subgroup 2: BAG; Subgroup 3: Er:YAG laser; Subgroup 4: BAG + Er:YAG laser, and Subgroup 5: Er:YAG laser + BAG. One sample was randomly selected from each subgroup for scanning electron microscope (SEM). The remaining samples were bonded with composite resin by Adper Single Bond 2 adhesive. Then water bath at 37°C for 24 hr. After 5,000 thermocycling, MTBS was tested and fracture mode was analyzed. The difference of MTBS between BAG group and Control group was found statistically significant (p < .05) in fluorosis. The Er:YAG laser + BAG group showed lowest MTBS values in fluorosis. In conclusion, the pretreatment of BAG might be beneficial to the adhesive of fluorosed teeth. Er:YAG laser desensitization alone or using BAG first and then Er:YAG laser desensitization might not affect the adhesive of fluorosed teeth, while Er:YAG laser desensitization followed by the pretreatment of BAG would be not conducive to the adhesive of fluorosed teeth.


Assuntos
Colagem Dentária , Lasers de Estado Sólido , Resinas Compostas , Dentina , Humanos , Microscopia Eletrônica de Varredura , Cimentos de Resina , Resistência à Tração
18.
Artigo em Zh | MEDLINE | ID: mdl-32791594

RESUMO

Objective:To explore the application value of anteromedial thigh flap(AMT) as alternative flap in repairing maxillofacial soft tissue defects. Method:Sixty patients were scheduled to underwent anterolateral thigh flap(ALT) reconstruction. Preoperative CT angiography were conducted. Imaging workstations were used to locate perforator vessels in the anterolateral and anteromedial areas respectively. Four patients had no suitable perforator during the preparation of AMT flaps. In the same operation area, ALT flaps were prepared to reconstruct the defect according to the location of the perforator vessels in the anteromedial areas. Result:All four AMT flaps survived uneventfully. Flap sizes ranged from 9 cm×6 cm to 7 cm×4 cm. The follow-up period ranged from 6 to 12 months, the functions of recipient and donor sites recovered well. Conclusion:Preoperative CT angiography can improve the accuracy of the preparation of skin flap effectively. When no sizable perforator is available during harvest of the ALT flap, successful reconstruction can be achieved using the ipsilateral AMT flap.


Assuntos
Retalho Perfurante , Procedimentos de Cirurgia Plástica , Lesões dos Tecidos Moles/cirurgia , Humanos , Transplante de Pele , Coxa da Perna/cirurgia
19.
Artigo em Zh | MEDLINE | ID: mdl-32791639

RESUMO

Objective:To investigate the application of free fibular flap based on digital technology in mandibular defects. Method:Eight cases of mandibular defects underwent virtual surgery and guide plate design before operation. The mandibular osteotomy guide plate, fibula plastic guide plate and mandibular reconstruction model were prepared by rapid prototyping technology. The individualized reconstruction titanium plates were prefabricated on the mandibular reconstruction model. Based on the guide plates and the individualized reconstruction titanium plates, the mandibular defects were repaired accurately. At the same time, CT angiography was used to observe the variation of peroneal artery. For patients with soft tissue defects, the superficial position of the point going out muscle of perforator vessels was located, and the skin flaps were designed to repair the soft tissue defect. Result:The free fibular flaps survived in all patients. The guide plates were successfully implanted, the position of the individualized reconstruction titanium plates were accurate, and the occlussions were well recovered. Preoperative CT angiography was carried out without complication in all patients, the desired anatomy was adequately demonstrated in all patients. The superficial position of the point going out muscle of perforator vessels during operation were basically in accordance with those detected by CT angiography. Conclusion:The free fibular flaps based on digital technology can successfully repair mandibular defects with good aesthetic and functional results.


Assuntos
Retalhos de Tecido Biológico , Reconstrução Mandibular , Procedimentos de Cirurgia Plástica , Placas Ósseas , Fíbula/cirurgia , Humanos , Mandíbula/cirurgia
20.
Anticancer Res ; 40(6): 3255-3264, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32487620

RESUMO

BACKGROUND/AIM: Rta, a transactivator of Epstein-Barr virus, is associated with progression of nasopharyngel carcinoma (NPC); however, its mechanism of contribution to the pathogenesis of NPC remains unclear. Interleukin-6 (IL-6), a tumor promoter, is detected in NPC. This in vitro study examined whether and how Rta promotes NPC progression by up-regulating IL-6. MATERIALS AND METHODS: Semiquantitative reverse transcription-polymerase chain reaction (RT-PCR), quantitative real-time PCR, ELISA, immunoblotting assays, reporter gene assays, and transwell migration assays were performed. RESULTS: In NPC cells, Rta up-regulated IL-6 expression at the mRNA and protein levels, and the Rta's C-terminus was essential for promoter activation and expression of IL-6. The induction of IL-6 by Rta also required activation of extracellular signal-regulated kinase 1/2 and activator protein-1. Furthermore, IL-6 secreted from Rta-expressing NPC cells promoted migration of Rta-negative NPC cells by activating IL-6 receptor/Janus kinase/signal transducer and activator of transcription 3 pathway. CONCLUSION: Rta contributes to progression of NPC cells through induction of IL-6 in vitro.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Interleucina-6/metabolismo , Janus Quinases/metabolismo , Neoplasias/metabolismo , Receptores de Interleucina-6/metabolismo , Fator de Transcrição STAT3/metabolismo , Transativadores/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/virologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Humanos , Proteínas Imediatamente Precoces/genética , Interleucina-6/biossíntese , Interleucina-6/genética , Sistema de Sinalização das MAP Quinases , Células MCF-7 , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patologia , Carcinoma Nasofaríngeo/virologia , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/virologia , Neoplasias/genética , Neoplasias/patologia , Neoplasias/virologia , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Neoplasias Gástricas/virologia , Transativadores/genética , Fator de Transcrição AP-1/metabolismo , Transfecção , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA