Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Arch Oral Biol ; 116: 104774, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32470830

RESUMO

OBJECTIVES: The aim of the present study was to identify the anticancer effects and the mechanisms of action of shikonin and its analogue isobutyrylshikonin in oral squamous carcinoma cells. DESIGNS: The cytotoxic effects of isobutyrylshikonin and shikonin in Ca9-22 and SCC-25 cells were analyzed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, flow cytometry analysis of Annexin V/Propidium Iodide (PI) staining, western blot analysis and immunohistochemistry. RESULTS: Treatment with both isobutyrylshikonin and shikonin induced dose- and time-dependent apoptotic cell death in Ca9-22 cells, although the IC50 of isobutyrylshikonin was less than that of shikonin. The induction of apoptosis by both isobutyrylshikonin and shikonin was accompanied by activation of caspase-8, -9, -3, and PARP, loss of mitochondrial trans-membrane potential, and release of cytochrome c from the mitochondria. ROS mediated the apoptosis induced by isobutyrylshikonin and shikonin, indicating that ROS may play a critical role in the distinctive cytotoxic effects of isobutyrylshikonin and shikonin in Ca9-22. Isobutyrylshikonin showed a similar cytotoxic effect in SCC-25 cells at concentrations showing the effects in Ca9 cells, but not in human normal keratinocyte cells. Although there is no biological difference between isobutyrylshikonin and shikonin, isobutyrylshikonin exerts the same cytotoxic effect at a concentration 6 times lower than shikonin. CONCLUSIONS: The present study suggest that isobutyrylshikonin may be a more potent chemotherapeutic agent against oral cancer cells than shikonin. In addition, our data exhibit that both isobutyrylshikonin and shikonin induce caspase-dependent apoptosis via the mitochondrial pathway through accumulation of ROS in oral squamous carcinoma cells.


Assuntos
Neoplasias Bucais , Naftoquinonas , Apoptose , Linhagem Celular Tumoral , Humanos , Potencial da Membrana Mitocondrial , Neoplasias Bucais/tratamento farmacológico , Naftoquinonas/farmacologia , Naftoquinonas/toxicidade , Espécies Reativas de Oxigênio , Células Tumorais Cultivadas
2.
Molecules ; 25(3)2020 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-32050534

RESUMO

This study aimed to investigate whether quercetin exerts anticancer effects on oral squamous cell carcinoma (OSCC) cell lines and to elucidate its mechanism of action. These anticancer effects in OSCC cells were assessed using an MTT assay, flow cytometry (to assess the cell cycle), wound-healing assay, invasion assay, Western blot analysis, gelatin zymography, and immunofluorescence. To investigate whether quercetin also inhibits transforming growth factor ß1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) in human keratinocyte cells, HaCaT cells were treated with TGF-ß1. Overall, our results strongly suggest that quercetin suppressed the viability of OSCC cells by inducing cell cycle arrest at the G2/M phase. However, quercetin did not affect cell viability of human keratinocytes such as HaCaT (immortal keratinocyte) and nHOK (primary normal human oral keratinocyte) cells. Additionally, quercetin suppresses cell migration through EMT and matrix metalloproteinase (MMP) in OSCC cells and decreases TGF-ß1-induced EMT in HaCaT cells. In conclusion, this study is the first, to our knowledge, to demonstrate that quercetin can inhibit the survival and metastatic ability of OSCC cells via the EMT-mediated pathway, specifically Slug. Quercetin may thus provide a novel pharmacological approach for the treatment of OSCCs.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Bucais/tratamento farmacológico , Quercetina/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/fisiologia , Humanos , Queratinócitos/efeitos dos fármacos , Metaloproteinases da Matriz/metabolismo , Neoplasias Bucais/patologia , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
3.
J Agric Food Chem ; 66(48): 12719-12729, 2018 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-30395462

RESUMO

Inhibition of matrix metalloproteinases (MMPs), which degrade collagen and elastin in the dermis of normal skin, is a key strategy for anti-skin aging. In this study, we identified five low-molecular-weight (LMW, <1 kDa) MMP-1-suppressive peptides in feather keratin hydrolysate (FKH) obtained by anaerobic digestion with an extremophilic bacterium. FKH was first subjected to ultrafiltration, followed by size-exclusion chromatography and liquid chromatography/electrospray ionization tandem mass spectrometry analysis. Chemically synthesized peptides identical to the sequences identified suppressed MMP expression in human dermal fibroblasts (HDFs). To investigate the impact of the MMP-1-suppressive peptides on the signaling pathway, we performed antibody array phosphorylation profiling of HDFs. The results suggested that the peptide GGFDL regulates ultraviolet-B-induced MMP-1 expression by inhibiting mitogen-activated protein kinases and nuclear factor κB signaling pathways as well as histone modification. Thus, LMW feather keratin peptides could serve as novel bioactive compounds to protect the skin against intrinsic and extrinsic factors.


Assuntos
Proteínas Aviárias/química , Plumas/química , Queratinas/química , Inibidores de Metaloproteinases de Matriz/química , Peptídeos/química , Envelhecimento da Pele/efeitos dos fármacos , Animais , Galinhas , Regulação para Baixo/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/efeitos da radiação , Humanos , Metaloproteinase 1 da Matriz/química , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/isolamento & purificação , Mapeamento de Peptídeos , Peptídeos/isolamento & purificação , Fosforilação , Hidrolisados de Proteína/química , Pele/efeitos dos fármacos , Pele/enzimologia , Pele/efeitos da radiação , Envelhecimento da Pele/genética , Envelhecimento da Pele/efeitos da radiação , Raios Ultravioleta
5.
Mol Cells ; 41(4): 290-300, 2018 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-29562732

RESUMO

Using an in vitro model of intestinal organoids derived from intestinal crypts, we examined effects of indole-3-carbinol (I3C), a phytochemical that has anticancer and aryl hydrocarbon receptor (AhR)-activating abilities and thus is sold as a dietary supplement, on the development of intestinal organoids and investigated the underlying mechanisms. I3C inhibited the in vitro development of mouse intestinal organoids. Addition of α-naphthoflavone, an AhR antagonist or AhR siRNA transfection, suppressed I3C function, suggesting that I3C-mediated interference with organoid development is AhR-dependent. I3C increased the expression of Muc2 and lysozyme, lineage-specific genes for goblet cells and Paneth cells, respectively, but inhibits the expression of IAP, a marker gene for enterocytes. In the intestines of mice treated with I3C, the number of goblet cells was reduced, but the number of Paneth cells and the depth and length of crypts and villi were not changed. I3C increased the level of active nonphosphorylated ß-catenin, but suppressed the Notch signal. As a result, expression of Hes1, a Notch target gene and a transcriptional repressor that plays a key role in enterocyte differentiation, was reduced, whereas expression of Math1, involved in the differentiation of secretory lineages, was increased. These results provide direct evidence for the role of AhR in the regulation of the development of intestinal stem cells and indicate that such regulation is likely mediated by regulation of Wnt and Notch signals.


Assuntos
Indóis/metabolismo , Receptores Notch/genética , Via de Sinalização Wnt/genética , Animais , Diferenciação Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
6.
Oncol Rep ; 39(4): 1930-1938, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29436668

RESUMO

Various amino acids regulate cell growth and differentiation. In the present study, we examined the ability of HT-29 cells to differentiate into goblet cells in RPMI and DMEM which are largely different in the amounts of numerous amino acids. Most of the HT-29 cells differentiated into goblet cells downregulating the stem cell marker Lgr5 when cultured in DMEM, but remained undifferentiated in RPMI. The goblet cell differentiation in DMEM was inhibited by 1-methyl-tryptophan (1-MT), an inhibitor of indoleamine 2,3 dioxygenase-1 which is the initial enzyme in tryptophan metabolism along the kynurenine (KN) pathway, whereas tryptophan and KN induced goblet cell differentiation in RPMI. The levels of Notch1 and its activation product Notch intracytoplasmic domain in HT-29 cells were lower in DMEM than those in RPMI and were increased by 1-MT in both media. HT-29 cells grown in both media expressed ß-catenin at the same level on day 2 when goblet cell differentiation was not observed. ß-catenin expression, which was increased by 1-MT in both media, was decreased by KN. DMEM reduced Hes1 expression while enhancing Hath1 expression. Finally, aryl hydrocarbon receptor (AhR) activation moderately induced goblet cell differentiation. Our results suggest that KN promotes goblet cell differentiation by regulating Wnt, Notch, and AhR signals and expression of Hes1 and Hath1.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Cinurenina/administração & dosagem , Fatores de Transcrição HES-1/genética , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Caliciformes/efeitos dos fármacos , Células Caliciformes/patologia , Células HT29 , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Receptor Notch1/genética , Receptores de Hidrocarboneto Arílico/genética , Triptofano/análogos & derivados , Triptofano/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/genética
7.
Mol Med Rep ; 17(2): 2327-2334, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29207110

RESUMO

The development of pharmaceutical agents possessing anti­invasive and anti­metastatic abilities, as well as apoptotic activity, is important in decreasing the incidence and recurrence of oral cancer. Cancer cells are known to acquire invasiveness not only through epigenetic changes, but also from inflammatory stimuli within the tumor microenvironment. Accordingly, the identification of agents that can suppress the inflammation­promoted invasiveness of cancer cells may be important in treating cancer and improving the prognosis of patients with cancer. Acetylshikonin, a flavonoid with anti­inflammatory activity, inhibits proliferation and induces apoptosis of oral cancer cells. In the present study, the anti­invasive effect of acetylshikonin on YD10B oral cancer cells infected with Porphyromonas gingivalis, a major pathogen of chronic periodontitis, and the mechanisms involved were investigated. Firstly, we examined whether P. gingivalis infection increased the invasiveness of YD10B cells. Results suggested that YD10B oral cancer cells become more aggressive when they are infected with P. gingivalis. Secondly, acetylshikonin significantly inhibited the invasion of P. gingivalis­infected YD10B cells by suppressing IL­8 release and IL­8­dependent MMP release. These data suggest that acetylshikonin may be a useful preventive and therapeutic candidate for oral cancer that is chronically infected with periodontal pathogens.


Assuntos
Antraquinonas/farmacologia , Interleucina-8/metabolismo , Metaloproteinases da Matriz/metabolismo , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/fisiologia , Infecções por Bacteroidaceae/metabolismo , Infecções por Bacteroidaceae/microbiologia , Linhagem Celular Tumoral , Células Cultivadas , Transição Epitelial-Mesenquimal , Expressão Gênica , Humanos , Interleucina-8/genética , Metaloproteinases da Matriz/genética , Neoplasias Bucais/metabolismo , Neoplasias Bucais/microbiologia , Neoplasias Bucais/patologia
8.
Oncotarget ; 8(29): 46981-46992, 2017 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-28388583

RESUMO

Major obstacles to improving the prognosis of patients with oral squamous cell carcinoma (OSCC) are the acquisition of resistance to chemotherapeutic agents and development of metastases. Recently, inflammatory signals are suggested to be one of the most important factors in modulating chemoresistance and establishing metastatic lesions. In addition, epidemiological studies have demonstrated that periodontitis, the most common chronic inflammatory condition of the oral cavity, is closely associated with oral cancer. However, a correlation between chronic periodontitis and chemoresistance/metastasis has not been well established. Herein, we will present our study on whether sustained infection with Porphyromonas gingivalis, a major pathogen of chronic periodontitis, could modify the response of OSCC cells to chemotherapeutic agents and their metastatic capability in vivo. Tumor xenografts composed of P. gingivalis-infected OSCC cells demonstrated a higher resistance to Taxol through Notch1 activation, as compared with uninfected cells. Furthermore, P. gingivalis-infected OSCC cells formed more metastatic foci in the lung than uninfected cells.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Infecções por Bacteroidaceae/complicações , Infecções por Bacteroidaceae/microbiologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Bucais/etiologia , Neoplasias Bucais/patologia , Paclitaxel/farmacologia , Porphyromonas gingivalis , Animais , Infecções por Bacteroidaceae/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Expressão Gênica , Humanos , Masculino , Camundongos , Neoplasias Bucais/metabolismo , Metástase Neoplásica , Receptor Notch1/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cytokine ; 86: 64-72, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27468958

RESUMO

Recent studies indicate that chronic inflammation promotes the aggressiveness of cancers. However, the direct molecular mechanisms underlying a functional link between chronic periodontitis, the most common form of oral inflammatory diseases, and the malignancy of oral cancer remain unknown. To elucidate the role of chronic periodontitis in progression of oral cancer, we examined the effect of Porphyromonas gingivalis (P. gingivalis), a major pathogen that causes chronic periodontitis, on the invasiveness of oral squamous cell carcinoma (OSCC) cells, including SCC-25, OSC-20 and SAS cells. Exposures to P. gingivalis promoted the invasive ability of OSC-20 and SAS cells via the upregulation of matrix metalloproteinases (MMPs), specifically MMP-1 and MMP-2. However, P. gingivalis-infected SCC-25 cells did not exhibit changes in their invasive properties or the low expression levels of MMPs. In an effort to delineate the molecular players that control the invasiveness, we first assessed the level of interleukin-8 (IL-8), a well-known inflammatory cytokine, in P. gingivalis-infected OSCC cells. IL-8 secretion was substantially increased in the OSC-20 and SAS cells, but not in the SCC-25 cells, following P. gingivalis infection. When IL-8 was directly applied to SCC-25 cells, their invasive ability and MMP level were significantly increased. Furthermore, the downregulation of IL-8 in P. gingivalis-infected OSC-20 and SAS cells attenuated their invasive potentials and MMP levels. Taken together, our findings strongly suggest that P. gingivalis infection plays an important role in the promotion of the invasive potential of OSCC cells via the upregulation of IL-8 and MMPs.


Assuntos
Carcinoma de Células Escamosas/patologia , Interleucina-8/genética , Metaloproteinases da Matriz/genética , Neoplasias Bucais/patologia , Invasividade Neoplásica , Porphyromonas gingivalis/fisiologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/microbiologia , Linhagem Celular Tumoral , Humanos , Interleucina-8/imunologia , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 2 da Matriz/genética , Neoplasias Bucais/metabolismo , Neoplasias Bucais/microbiologia , Reação em Cadeia da Polimerase em Tempo Real , Regulação para Cima
10.
Arch Oral Biol ; 70: 149-157, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27371806

RESUMO

OBJECTIVES: Recently, shikonin derivatives from Lithospermum erythrorhizon have been suggested as potential chemotherapeutic agents against numerous types of cancers in addition to their traditional uses, e.g., as anti-inflammatory agents. Acetylshikonin, one of shikonin derivatives, has also been reported to possess anticancer activity. However, few studies of the effectiveness of acetylshikonin against cancer cells have been conducted, and there are no studies of oral cancers. In this study, we investigated the usefulness of acetylshikonin as a treatment regimen for oral cancers by observing the growth inhibitory function of acetylshikonin and the involved mechanisms. DESIGNS: The viability, cell cycle, and ratio of apoptotic cells of oral squamous cell carcinoma (OSCC) cells were observed after treatment with acetylshikonin using MTT assay, flow cytometric analysis, and Annexin V/PI staining, respectively. In addition, molecular changes of apoptosis-related pathways and the role of reactive oxygen species (ROS) were analyzed in acetylshikonin-treated cells. RESULTS: We observed that acetylshikonin significantly suppressed the growth of OSCC cells by inducing apoptotic cell death, and acetylshikonin affected the viability of a normal keratinocyte cell line HaCaT to a lesser degree, suggesting that acetylshikonin may be a good chemotherapeutic reagent with less toxicity to normal tissues. In addition, we found that acetylshikonin-induced apoptosis of OSCC cells is mediated by ROS as well as G2 cell cycle arrest. ROS production in response to acetylshikonin treatment enhanced the phosphorylation of JNK and p38 MAPK, which are in the major pathways of apoptotic cell death mechanisms. CONCLUSIONS: In summary, our data suggest that acetylshikonin is a strong candidate for use as a selective chemotherapeutic agent for the treatment of OSCC.


Assuntos
Antraquinonas/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias Bucais/tratamento farmacológico , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , MAP Quinase Quinase 4/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Neoplasias Bucais/patologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Tumour Biol ; 36(12): 9947-60, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26178482

RESUMO

Periodontitis is the most common chronic inflammatory condition occurring in the human oral cavity, but our knowledge on its contribution to oral cancer is rather limited. To define crosstalk between chronic periodontitis and oral cancer, we investigated whether Porphyromonas gingivalis, a major pathogen of chronic periodontitis, plays a role in oral cancer progression. To mimic chronic irritation by P. gingivalis in the oral cavity, oral squamous cell carcinoma (OSCC) cells were infected with P. gingivalis twice a week for 5 weeks. Repeated infection of oral cancer cells by P. gingivalis resulted in morphological changes of host cancer cells into an elongated shape, along with the decreased expression of epithelial cell markers, suggesting acquisition of an epithelial-to-mesenchymal transition (EMT) phenotype. The prolonged exposure to P. gingivalis also promoted migratory and invasive properties of OSCC cells and provided resistance against a chemotherapeutic agent, all of which are described as cellular characteristics undergoing EMT. Importantly, long-term infection by P. gingivalis induced an increase in the expression level of CD44 and CD133, well-known cancer stem cell markers, and promoted the tumorigenic properties of infected cancer cells compared to non-infected controls. Furthermore, increased invasiveness of P. gingivalis-infected OSCC cells was correlated with enhanced production of matrix metalloproteinase (MMP)-1 and MMP-10 that was stimulated by interleukin-8 (IL-8) release. This is the first report demonstrating that P. gingivalis can increase the aggressiveness of oral cancer cells via epithelial-mesenchymal transition-like changes and the acquisition of stemness, implicating P. gingivalis as a potential bacterial risk modifier.


Assuntos
Neoplasias Bucais/patologia , Células-Tronco Neoplásicas/patologia , Periodontite/patologia , Porphyromonas gingivalis/patogenicidade , Carcinoma de Células Escamosas , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-8/metabolismo , Metaloproteinase 1 da Matriz/biossíntese , Neoplasias Bucais/complicações , Neoplasias Bucais/microbiologia , Células-Tronco Neoplásicas/microbiologia , Periodontite/complicações , Periodontite/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA