Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Dent Sci ; 17(4): 1722-1730, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36299346

RESUMO

Background/purpose: Photodynamic therapy (PDT) is a therapeutic alternative for malignant tumors that uses a photosensitizer. This study examined whether synthesized Pheophorbide a (Pa) -PDT induced apoptosis and autophagy involving endoplasmic reticulum (ER) stress in oral squamous cell carcinoma (OSCC) cells. Materials and methods: Human OSCC cells were treated with Pa-PDT, and cell proliferation was examined by MTT assay. Apoptosis and autophagy were measured using Western blot analysis. ER stress was examined using RT-PCR and Western blot analysis. In vivo murine OSCC animal model were treated with intratumoral (IT) Pa-PDT, and investigated the therapeutic effect. Results: Pa-PDT significantly inhibited the proliferation of human OSCC cells in a dose-dependent manner. Pa-PDT induced intrinsic apoptotic cell death and also induced autophagy. Pa-PDT induced ER stress which was observed as demonstrated by the up-regulation of the ER stress marker. Inhibition of the ER stress pathway using 4-phenylbutyric acid (PBA) decreased CHOP and induced inhibition of cell deaths. In addition, the inhibition of ER stress enhanced Pa-PDT mediated autophagy. IT Pa-PDT significantly inhibited the tumor growth and induced apoptosis, autophagy and ER stress in vivo OSCC cells transplanted model. Conclusion: This study showed that synthesized Pa-PDT induced ER stress trigger apoptosis and apoptotic cell death pathways in OSCC cells. The inhibition of ER stress declined Pa-PDT mediated cytotoxicity with an increase of autophagy. These results may provide Pa-PDT exerts anti-tumor effects through ER stress pathway in OSCC cells and may provide a basis for developing Pa-PDT targeting ER stress as a therapy for OSCC.

2.
J Dent Sci ; 15(1): 1-8, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32256993

RESUMO

BACKGROUND/PURPOSE: Oral lichen planus (OLP) is a chronic inflammatory disease of oral mucosa. The present study investigated the expression of nucleotide-binding oligomerization domain (NOD), a pivotal sensor protein of the innate immune system, in OLP. MATERIALS AND METHODS: Oral mucosal biopsies were collected from 20 patients with OLP and 6 individuals with normal oral mucosa (NOM). The expression of NOD1 and NOD2 was determined using RT-PCR and immunohistochemistry in OLP and NOM samples. RESULTS: The mRNA expression of NOD1 and NOD2 was significantly higher in the OLP group than in the NOM group. The protein expression of NOD1 was marginally upregulated in all mucosal layers in the OLP group compared with that of the NOM group; however, the differences were not significant. The expression of NOD2 was elevated in infiltrating lymphocytes of the submucosal layer in the OLP group compared with the NOM group, but was undetected in other inflammatory disease, inflammatory fibrous hyperplasia (IFH). This study revealed the upregulation of NOD2 mRNA and protein in the OLP group, but not in the NOM group. CONCLUSION: These findings suggest that NOD2 may play an important role in the pathogenesis of OLP and represents a new diagnostic and treatment target.

3.
Biomol Ther (Seoul) ; 28(2): 184-194, 2020 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-31476841

RESUMO

Histone deacetylase (HDAC) inhibitors represent a novel class of anticancer agents, which can be used to inhibit cell proliferation and induce apoptosis in several types of cancer cells. In this study, we investigated the anticancer activity of MHY4381, a newly synthesized HDAC inhibitor, against human prostate cancer cell lines and compared its efficacy with that of suberoylanilide hydroxamic acid (SAHA), a well-known HDAC inhibitor. We assessed cell viability, apoptosis, cell cycle regulation, and other biological effects in the prostate cancer cells. We also evaluated a possible mechanism of MHY4381 on the apoptotic cell death pathway. The IC50 value of MHY4381 was lower in DU145 cells (IC50=0.31 µM) than in LNCaP (IC50=0.85 µM) and PC-3 cells (IC50=5.23 µM). In addition, the IC50 values of MHY4381 measured in this assay were significantly lower than those of SAHA against prostate cancer cell lines. MHY4381 increased the levels of acetylated histones H3 and H4 and reduced the expression of HDAC proteins in the prostate cancer cell lines. MHY4381 increased G2/M phase arrest in DU145 cells, and G1 arrest in LNCaP cells. It also activated reactive oxygen species (ROS) generation, which induced apoptosis in the DU145 and LNCaP cells by increasing the ratio of Bax/Bcl-2 and releasing cytochrome c into the cytoplasm. Our results indicated that MHY4381 preferentially results in antitumor effects in DU145 and LNCaP cells via mitochondria-mediated apoptosis and ROS-facilitated cell death pathway, and therefore can be used as a promising prostate cancer therapeutic.

4.
Data Brief ; 26: 104484, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31667249

RESUMO

There is technology available for anti-thrombus with earthworms, but the procedure is complex and extracts protein with inferior purity. In order to develop a simplified process with a stronger purity of protease, we investigated the Lumbricus rubellus earthworm and Perinereis linea lugworm. We purified water extracts cut off at 10 kDa of molecular weight using ultrafiltration because proteins are large biomolecules, or macromolecules, consisting of one or more long chains of amino acid residues. We purified EW1 (raw earthworm extract), EW2 (molecular weight (m.w) > 10 kDa of earthworm extract), and EW3 (m.w < 10 kDa) from the Lumbricus rubellus earthworm. Likewise, we purified LW1 (wild lugworm extract), LW2 (m.w > 10 kDa), and LW3 (m.w < 10 kDa) from the Perinereis linea lugworm. Using a fibrin assay, we found that fibrinolytic activity of the specimens had a rank order of clear zone diameter: EW2 > EW1 > EW3 > LW2 > LW1 > LW3. In particular, EW2 and LW2 showed a potent fibrinolytic effect in two different worm specimens. The protein content of each sample was detected as 2.34 (EW1), 3.03 (EW2), 2.80 (LW1), 3.71 (LW2) mg/ml respectively, and their molecular weights were measured using SDS-PAGE. The samples contained the following amounts of total fatty acids: EW1, 3.61%; EW2, 0.48%; LW1, 4.96%; and LW2, 0.23%. We developed a process to increase the thrombolytic effect with a higher purity protein. The study results demonstrate this procedure and provide basic data for developing an anti-thrombolytic agent.

5.
Data Brief ; 25: 104088, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31294054

RESUMO

This study investigated the anti-inflammatory effects of mixed extracts of Achyranthes japonica Nakai (AJ) and Aralia continentalis Kitagawa (AC) (ratios of 1:2, 1:3, 1:5, 2:1, 3:1 and 5:1) on RAW264.7 macrophages and evaluated the anti-inflammatory effects of the mixed extracts of AJ and AC by measuring IL-1ß, IL-6, and TNFα using the ELISA kit assay. In particular, the formation of nitric oxide (NO) was found to decrease in the group treated with the combined extracts of AJ and AC at all ratios. In particular, extracts of ratio of 2:1 (AJ:AC) deceased the formation of NO level that is approximately 60% of the group treated with only lipopolysaccharide (LPS). Also, extracts of ratio of 2:1 (AJ:AC) reduced the production of IL-1ß, IL-6, TNFα and PGE2 with statistical significance. Volunteers over the age of 50 who complain of discomfort in knee joints were selected as the experimental subjects. The subjects took daily administration of 2000 mg of the combined extracts of ratio of 2:1 (AJ:AC) for 12 weeks. A survey (VAS (Visual Analog Scale), WOMAC (Western Ontario and McMaster Universities Osteoarthritis Index)) was conducted after the 12 weeks of oral administration. The experimental group showed the change between each visit and baseline time compared with the control group. In the intention-to-treat (ITT) analysis, VAS score and WOMAC stiffness score decreased significantly. And the WOMAC total score and function score tended to decrease. In the per-protocol (PP) analysis, the WOMAC stiffness score was significantly decreased and the VAS and WOMAC total and function scores were decreased. There was no significant difference in all parameters of ITT and PP in radiological examinations.

6.
Cell Physiol Biochem ; 52(6): 1535-1552, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31135122

RESUMO

BACKGROUND/AIMS: Pyruvate kinase M2 (PKM2) is essential for aerobic glycolysis. Although high PKM2 expression is observed in various cancer tissues, its functional role in cancer metabolism is unclear. Here, we investigated the role of PKM2 in regulating autophagy and its associated pathways in prostate cancer cells. METHODS: Immunohistochemistry was performed to compare the expression level of PKM2 in prostate cancer patients and normal human, whereas expression of PKM2 in several cell lines was also examined by using western blot. PKM2 expression was silenced using various small interfering RNAs (siRNAs). Cell viability was examined using IncuCyte ZOOM™ live cell imaging system. Western blotting and immunofluorescence were performed to investigate the PKM2 knockdown on other cellular signaling molecules. Acridine orange and Monodansylcadaverine staining was performed to check effect of PKM2 knockdown on autophagy induction. High performance thin layer chromatography was carried out to quantify the level of different cellular metabolites (pyruvate and lactate). Colony formation assay was performed to determine the ability of a cells to form large colonies. RESULTS: PKM2 was highly expressed in prostate cancer patients as compared to normal human. PKM2 siRNA-transfected prostate cancer cells showed significantly reduced viability. Acridine orange, Monodansylcadaverine staining and western blotting analysis showed that PKM2 downregulation markedly increased autophagic cell death. Results of western blotting analysis showed that PKM2 knockdown affected protein kinase B/mechanistic target of rapamycin 1 pathway, which consequently downregulated the expression of glycolytic enzymes lactate dehydrogenase A and glucose transporter 1. Knockdown of PKM2 also reduced the colony formation ability of human prostate cancer cell DU145. CONCLUSION: To the best of our knowledge, this is the first study to show that PKM2 inhibition alters prostate cancer cell metabolism and induces autophagy, thus providing new perspectives for developing PKM2-targeting anticancer therapies for treating prostate cancer.


Assuntos
Autofagia , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piruvato Quinase/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Linhagem Celular Tumoral , Transportador de Glucose Tipo 1/metabolismo , Humanos , Isoenzimas/metabolismo , L-Lactato Desidrogenase/metabolismo , Lactato Desidrogenase 5 , Masculino , Neoplasias da Próstata/metabolismo , Piruvato Quinase/antagonistas & inibidores , Piruvato Quinase/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
7.
Oncol Lett ; 16(5): 6552-6560, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30405794

RESUMO

Apicidin, a cyclic peptide histone deacetylase (HDAC) inhibitor, has been demonstrated to exhibit antitumor activity in a number of human cancer types. The present study examined the antitumor activity of apicidin in murine oral squamous cell carcinoma (OSCC) cells. Inhibition of cell proliferation and the expression of selective HDACs were determined in apicidin-treated AT-84 murine OSCC cells. A C3H mouse model with subcutaneous injection of AT-84 cells was used to assess the in vivo effect of apicidin on tumor growth. Apicidin-induced cell growth inhibition and selectively reduced HDAC8 expression in AT-84 cells. Induction of apoptosis and autophagy was observed in apicidin-treated AT-84 cells. Apicidin notably inhibited tumor growth by up to 46% relative to the control group at the end of a 14-day period in a murine tumor model. The immunohistochemistry results in tumor tissues indicated that apicidin inhibited cell proliferation and induced apoptosis and autophagy in AT-84 cell-derived tumor tissues. Overexpression of HDAC8 was observed in the nucleus and cytoplasm in tumor tissues and apicidin significantly inhibited the level of HDAC8 expression, compared with the vehicle group. These results indicated that apicidin inhibited cell proliferation through HDAC8 inhibition in murine OSCC cells in vitro and in vivo. The present study indicated that apicidin may be an effective therapeutic agent for OSCC.

8.
J Toxicol Environ Health A ; 81(11): 397-407, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29557720

RESUMO

The objective of this study was to elucidate the effect of hepatic damage on cisplatin (CDDP)-induced acute kidney injury (AKI). Thioacetamide (TAA, 150 mg/kg), a hepatotoxicant, was intraperitoneally (i.p.) injected to male Sprague-Dawley rats for 3 d prior to CDDP (5 mg/kg, i.p.) injection. All animals were sacrificed 5 d after CDDP treatment, and urine or blood was obtained to measure various parameters. No significant changes in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activity were observed after CDDP treatment. However, pretreatment with TAA significantly elevated ALT and AST activity. Serum blood urea nitrogen and creatinine levels significantly increased in CDDP-treated group compared to control. In addition, urinary excretion of novel protein-based biomarkers such as neutrophil gelatinase-associated lipocalin, vascular endothelial growth factor, kidney injury molecule-1, and tissue inhibitor of metalloproteinase-1 rose markedly in the CDDP-treated group. In particular, pretreatment with TAA markedly elevated CDDP-induced urinary excretion of protein-based nephrotoxic biomarkers compared with CDDP alone. Hematoxylin and eosin staining demonstrated that pretreatment with TAA following CDDP injection led to more severe tubular damage and apoptosis in rats compared with CDDP alone. Antioxidant status was significantly reduced in kidneys following pretreatment with TAA prior to CDDP. These findings indicate that liver injury enhanced the vulnerability of kidney to CDDP-induced AKI and this phenomenon may be associated with severe apoptotic damage.


Assuntos
Injúria Renal Aguda/fisiopatologia , Antineoplásicos/toxicidade , Cisplatino/toxicidade , Fígado/fisiopatologia , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/urina , Carcinógenos/toxicidade , Fígado/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Tioacetamida/toxicidade
9.
J Toxicol Environ Health A ; 81(11): 421-431, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29557728

RESUMO

Triclosan (TCS), a common antimicrobial ingredient, is present in many consumer products, including soaps, shampoos, and toothpaste. Owing to its widespread use, potential adverse effects on animals and humans may arise from lifetime exposure, but data on chronic prepubertal exposure of TCS are still lacking. The aim of the present study was to investigate the influence of subchronic TCS exposure (0.25, 25, 250, or 750 mg/kg) on target organ toxicity in prepubertal male rats. After daily administration of TCS to rats by oral gavage for 60 d, a significant reduction in body weight and relative weights of liver, kidneys, testes, and adrenal glands was observed in the 750-mg/kg (high dose) group. Serum alanine aminotransferase and aspartate aminotransferase activities as well as levels of blood urea nitrogen, and creatinine were significantly increased at 750 mg/kg TCS. Further, TCS (750 mg/kg) elevated the protein expressions of hepatic CYP2B1, RXR/PPAR, and levels of malondialdehyde. High-dose TCS exposure induced histological changes as evidenced by reduction of Bowman's space, occlusion of the tubular lumen, and degeneration of tubular epithelial cells in the kidney. Tubular necrosis was confirmed as evidenced by a rise in expression of high mobility group box 1 renal protein. Daily sperm production was significantly diminished by high doses of TCS with marked inhibition of androgen receptor protein expression. Our results indicated that subchronic exposure to excessively high concentrations of 750 mg/kg TCS induced hepatorenal and reproductive toxicities in prepubertal male rats; however, the biological relevance of these findings is questionable as these drug levels are not encountered in the environment.


Assuntos
Glândulas Suprarrenais/efeitos dos fármacos , Anti-Infecciosos Locais/toxicidade , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Triclosan/toxicidade , Animais , Relação Dose-Resposta a Droga , Masculino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Testículo/efeitos dos fármacos , Testes de Toxicidade Subcrônica
10.
J Toxicol Environ Health A ; 80(9): 453-464, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28665768

RESUMO

Acute kidney injury (AKI) is associated with increased mortality rate in patients but clinically available biomarkers for disease detection are currently not available. Recently, a new biomarker, selenium-binding protein 1 (SBP1), was identified for detection of nephrotoxicity using proteomic analysis. The aim of this study was to assess the sensitivity of urinary SBP1 levels as an early detection of AKI using animal models such as cisplatin or ischemia/reperfusion (I/R). Sprague-Dawley rats were injected with cisplatin (6 mg/kg, once i.p.) and sacrificed at 1, 3, or 5 days after treatment. Ischemia was achieved by bilaterally occluding both kidneys with a microvascular clamp for 45 min and verified visually by a change in tissue color. After post-reperfusion, urine samples were collected at 9, 24, and 48 hr intervals. Urinary excretion of protein-based biomarkers was measured by Western blot analysis. In cisplatin-treated rats, mild histopathologic alterations were noted at day 1 which became severe at day 3. Blood urea nitrogen (BUN) and serum creatinine (SCr) levels were significantly increased at day 3. Levels of urinary excretion of SBP1, neutrophil gelatinase-associated lipocalin (NGAL), and a tissue inhibitor of metalloproteinase-1 (TIMP-1) were markedly elevated at day 3 and 5 following drug treatment. In the vehicle-treated I/R group, serum levels of BUN and SCr and AST activity were significantly increased compared to sham. Urinary excretion of SBP1 and NGAL rose markedly following I/R. The urinary levels of SBP1, NGAL, TIMP-1, and KIM-1 proteins excreted by AKI patients and normal subjects were compared. Among these proteins, a marked rise in SBP1 was observed in urine of patients with AKI compared to normal subjects. Based upon receiver-operator curves (ROC), SBP1 displayed a higher area under the curve (AUC) scores than levels of SCr, BUN, total protein, and glucose. In particular, SBP1 protein was readily detected in small amounts of urine without purification. Data thus indicate that urinary excretion of SBP1 may be useful as a reliable biomarker for early diagnosis of AKI in patients.


Assuntos
Injúria Renal Aguda/diagnóstico , Injúria Renal Aguda/urina , Biomarcadores/urina , Diagnóstico Precoce , Proteínas de Ligação a Selênio/urina , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Animais , Valor Preditivo dos Testes , Proteômica , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
11.
J Oral Pathol Med ; 46(4): 276-283, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28178760

RESUMO

BACKGROUND: The overexpression of histone deacetylases (HDACs) has been observed in many cancers, and inhibition of specific HDACs has emerged as a new target for cancer therapy. We found that HDAC7 expression was selectively reduced by HDAC inhibitor apicidin in salivary mucoepidermoid carcinoma (MEC) cells. Here, we show that HDAC7 suppression has a potent antitumor effect in MEC cells. METHODS: Histone deacetylases7 was knocked down using HDAC7 siRNAs, and cell proliferation was quantified. Cell cycle progression, apoptosis, and autophagy were measured by flow cytometry and immunoblotting. RESULTS: Histone deacetylases 7 siRNAs inhibited cell proliferation and c-Myc expression, increased p27 expression, and caused G2/M phase cell cycle arrest in both YD-15 and Mc3 cells. HDAC7 silencing increased the sub-G1 population, Annexin V positive apoptotic cells and cleaved caspase3 levels. HDAC7 silencing induced an increase in autophagic markers, number of acidic vesicular organelles, and LC3B II levels, and decrease in p62 levels. HDAC7 siRNAs reduced the activation of ERK. HDAC7 knockdown resulted in growth inhibition through G2/M phase cell cycle arrest and induced both apoptosis and autophagy in MEC cells. CONCLUSIONS: This study indicates that inhibition of HDAC7 might become a novel and effective therapeutic approach for treating to MEC.


Assuntos
Apoptose , Autofagia , Carcinoma Mucoepidermoide/metabolismo , Histona Desacetilases/metabolismo , Neoplasias das Glândulas Salivares/metabolismo , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos
12.
Oncol Rep ; 37(1): 540-546, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28004115

RESUMO

The overexpression of histone deacetylases (HDACs) has been observed in many cancers and inhibition of specific HDAC has emerged as a new target for cancer therapy. The present study examined the expression of HDAC8 and the inhibitory effect of HDAC8 in oral squamous cell carcinoma (OSCC). The expression of HDAC8 was measured in human OSCC tissues and OSCC cell lines using immunohistochemistry and immunoblotting. HDAC8 was knocked down in OSCC cells by transfection with HDAC8 siRNAs and cell proliferation was quantified. Apoptosis and autophagy were measured using flow cytometry and immunoblotting. HDAC8 were overexpressed in OSCC tissues and OSCC cells, mainly localized in the cytoplasm. HDAC8 siRNAs effectively reduced the level of HDAC8 expression and HDAC8 silencing significantly inhibited the proliferation of OSCC cells. HDAC8 knockdown induced apoptotic cell death through caspases activation and pro-survival autophagy in OSCC cells. Combination with HDAC silencing and autophagy inhibition enhanced cell death by increasing apoptosis in OSCC cells. This study suggests that inhibition of HDAC8 might become a novel therapeutic strategy for OSCC.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Histona Desacetilases/metabolismo , Neoplasias Bucais/enzimologia , Proteínas Repressoras/metabolismo , Apoptose/genética , Autofagia , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Citoplasma/enzimologia , Feminino , Técnicas de Silenciamento de Genes , Histona Desacetilases/genética , Humanos , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular/métodos , Neoplasias Bucais/patologia , RNA Interferente Pequeno , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética
13.
Oncol Res ; 25(2): 295-304, 2017 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-27629775

RESUMO

Photodynamic therapy (PDT) is a therapeutic alternative for malignant tumors that uses a photosensitizer. Our group recently synthesized photosensitizer pheophorbide a (Pa) from chlorophyll-a. The present study investigated the therapeutic effect of PDT using intratumoral administration of the synthetic photosensitizer Pa in an in vivo murine oral squamous cell carcinoma (OSCC) animal model. Pa accumulation was measured using the fluorescence spectrum and imaging in living C3H mice. Intratumoral treatment of Pa-PDT (IT Pa-PDT) significantly inhibited the growth of transplanted OSCC cells. Histopathological examination of tumor tissues showed that PCNA expression was significantly decreased, while TUNEL-stained cells were markedly increased in the IT Pa-PDT group compared to controls. IT Pa-PDT-induced apoptosis was confirmed by immunoblot. Reduction of Bcl-2 and cleavage of caspase 3 and PARP were observed in IT Pa-PDT. These data demonstrate that IT Pa-PDT inhibited tumor cell proliferation and induced apoptosis, which is correlated with the anticancer activity of IT Pa-PDT. These potent antitumor activities of IT Pa-PDT were observed in both the immunohistochemistry and Western blot experiments. Our findings suggest the intratumoral therapeutic potential of Pa-PDT on OSCC. Additionally, demonstrated detection of Pa using a fluorescence spectroscopy system or molecular imaging system provides a means for simultaneous diagnosis and treatment of OSCC.


Assuntos
Clorofila/análogos & derivados , Neoplasias Bucais/tratamento farmacológico , Fotoquimioterapia/métodos , Radiossensibilizantes/síntese química , Radiossensibilizantes/uso terapêutico , Animais , Linhagem Celular Tumoral , Clorofila/síntese química , Clorofila/uso terapêutico , Relação Dose-Resposta a Droga , Camundongos , Camundongos Endogâmicos C3H , Neoplasias Bucais/patologia
14.
J Oral Pathol Med ; 45(4): 262-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26332444

RESUMO

OBJECTIVES: Microbial Pattern-recognition receptors (PRRs), such as nucleotide-binding oligomerization domains (NODs), are essential for mammalian innate immune response. This study was designed to determine the effect of NOD1 and NOD2 agonist on innate immune responses and antitumor activity in oral squamous cell carcinoma (OSCC) cells. MATERIALS AND METHODS: NODs expression was examined by RT-PCR, and IL-8 production by NODs agonist was examined by ELISA. Western blot analysis was performed to determine the MAPK activation in response to their agonist. Cell proliferation was determined by MTT assay. Flow cytometry and Western blot analysis were performed to determine the MDP-induced cell death. RESULTS: The levels of NODs were apparently expressed in OSCC cells. NODs agonist, Tri-DAP and MDP, led to the production of IL-8 and MAPK activation. NOD2 agonist, MDP, inhibited the proliferation of YD-10B cells in a dose-dependent manner. Also, the ratio of Annexin V-positive cells and cleaved PARP was increased by MDP treatment in YD-10B cells, suggesting that MDP-induced cell death in YD-10B cells may be owing to apoptosis. CONCLUSIONS: Our results indicate that NODs are functionally expressed in OSCC cells and can trigger innate immune responses. In addition, NOD2 agonist inhibited cell proliferation and induced apoptosis. These findings provide the potential value of MDP as novel candidates for antitumor agents of OSCC.


Assuntos
Acetilmuramil-Alanil-Isoglutamina/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/tratamento farmacológico , Ácido Diaminopimélico/análogos & derivados , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias Bucais/tratamento farmacológico , Proteína Adaptadora de Sinalização NOD2/agonistas , Oligopeptídeos/farmacologia , Antineoplásicos/farmacologia , Apoptose/fisiologia , Western Blotting , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ácido Diaminopimélico/farmacologia , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Imunidade Inata/efeitos dos fármacos , Interleucina-8/biossíntese , Interleucina-8/metabolismo , Proteínas Quinases Ativadas por Mitógeno/biossíntese , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias Bucais/imunologia , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Proteína Adaptadora de Sinalização NOD1/agonistas , Proteína Adaptadora de Sinalização NOD1/biossíntese , Proteína Adaptadora de Sinalização NOD1/genética , Proteína Adaptadora de Sinalização NOD2/biossíntese , Proteína Adaptadora de Sinalização NOD2/genética , RNA Mensageiro/biossíntese , Carcinoma de Células Escamosas de Cabeça e Pescoço
15.
Biomol Ther (Seoul) ; 23(5): 434-41, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26336583

RESUMO

Histone deacetylase (HDAC) inhibitors are considered novel agents for cancer chemotherapy. We previously investigated MHY219, a new HDAC inhibitor, and its potent anticancer activity in human prostate cancer cells. In the present study, we evaluated MHY219 molecular mechanisms involved in the regulation of prostate cancer cell migration. Similar to suberanilohydroxamic acid (SAHA), MHY219 inhibited HDAC1 enzyme activity in a dose-dependent manner. MHY219 cytotoxicity was higher in LNCaP (IC50=0.67 µM) than in DU145 cells (IC50=1.10 µM) and PC3 cells (IC50=5.60 µM) after 48 h of treatment. MHY219 significantly inhibited the HDAC1 protein levels in LNCaP and DU145 cells at high concentrations. However, inhibitory effects of MHY219 on HDAC proteins levels varied based on the cell type. MHY219 significantly inhibited LNCaP and DU145 cells migration by down-regulation of matrix metalloprotease-1 (MMP-1) and MMP-2 and induction of tissue inhibitor of metalloproteinases-1 (TIMP-1). These results suggest that MHY219 may potentially be used as an anticancer agent to block cancer cell migration through the repression of MMP-1 and MMP-2, which is related to the reduction of HDAC1.

16.
Eur J Pharm Sci ; 79: 122-31, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26342773

RESUMO

This study examined the anti-tumor effects of AGM130, a novel indirubin-3'-oxime derivative in A549 human non-small cell lung cancer cells. AGM130 significantly inhibited the proliferation and arrested the cell cycle of G2/M phase. Induction of apoptosis was detected in AGM130-treated A549 cells. The protein levels of Cytochrome c release, Bax, cleaved caspases and PARP were increased in AGM130 treated cells, whereas Bcl-2 levels were decreased. AGM130 inhibited Insulin-like growth factor 1 receptor (IGF1R), AKT/mTOR signaling and inactivated mitogen-activated protein kinases (MAPK). AGM130 also induced slight autophagy as pro-survival function and autophagy inhibition by chloroquine (CQ) induced necrosis. In vivo tumor xenograft model, AGM130 dose-dependently suppressed transplanted A549 cell tumor growth and induced the expression of proliferative cell nuclear antigen (PCNA). AGM130 also increased TUNEL positive apoptotic cell populations and the induction of glandular differentiation with mucin pool compared with vehicle-treated control in tumor tissue. These results suggest that AGM130 is an effective novel indirubin-3'-oxime derivative of anti-cancer drug and may be an attractive candidate for non-small cell lung cancer therapy.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Indóis/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Oximas/uso terapêutico , Animais , Apoptose , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Indóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteína Oncogênica v-akt/antagonistas & inibidores , Oximas/farmacologia , Receptor IGF Tipo 1 , Receptores de Somatomedina/antagonistas & inibidores
17.
Oncol Rep ; 33(4): 1899-907, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25647264

RESUMO

Inhibition of histone deacetylases (HDACs) has emerged as a new target for cancer therapies. The present study examined the antitumor effect and molecular mechanism of the HDAC inhibitor apicidin in YD-15 human salivary mucoepidermoid carcinoma (MEC) cells. The cells were treated with apicidin and cell death was quantified using an MTT assay. Apoptosis and autophagy were measured using flow cytometry, immunoblot analysis and cell staining. Regulation of the signaling pathways was monitored using immunoblot analysis and co-treatment with specific inhibitors. Insulin-like growth factor 1 receptor (IGF-1R) was knocked down using specific siRNA. Apicidin significantly inhibited the proliferation of MEC cells. Apicidin also induced apoptosis through the inactivation of extracellular signal-regulated kinase (ERK) and AKT/mTOR signaling and activation of c-Jun NH2-terminal kinase (JNK), whereas apicidin promoted autophagy through inactivation of the AKT/mTOR signaling. These effects may be mediated by the inhibition of IGF-1R, an upstream regulator of MAPK and AKT/mTOR pathways. These results suggested that apicidin is an attractive chemotherapeutic agent against salivary MEC and may be a good candidate for targeting IGF-1R for cancer therapies.


Assuntos
Carcinoma Mucoepidermoide/patologia , Inibidores de Histona Desacetilases/farmacologia , Proteínas de Neoplasias/biossíntese , Peptídeos Cíclicos/farmacologia , Receptores de Somatomedina/biossíntese , Neoplasias das Glândulas Salivares/patologia , Transdução de Sinais/efeitos dos fármacos , Clorometilcetonas de Aminoácidos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Carcinoma Mucoepidermoide/genética , Carcinoma Mucoepidermoide/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Terapia de Alvo Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , Receptor IGF Tipo 1 , Receptores de Somatomedina/genética , Receptores de Somatomedina/fisiologia , Neoplasias das Glândulas Salivares/genética , Neoplasias das Glândulas Salivares/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/fisiologia
18.
Obstet Gynecol Sci ; 56(2): 102-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24327988

RESUMO

OBJECTIVE: This study was performed to compare the expression of CD44 in endometrial stromal cells (ESCs) of women with and without endometriosis and to evaluate the role of CD44 in the adherence of ESCs to peritoneal mesothelial cells (PMCs). METHODS: A PMC adherence assay was performed to evaluate the adherence of ESCs to PMCs in women with and without endometriosis. The expression of CD44 mRNA was measured by reverse transcription-polymerase chain reaction. CD44 protein was evaluated by Western blot analysis. RESULTS: There were no significant differences in the expression of CD44 mRNA and protein in ESCs or in the binding of ESCs to PMCs between patients with endometriosis and controls. Although the expression of CD44 protein was decreased in both women with endometriosis and controls after anti-CD44 antibody treatment, there was no effect on binding of ESCs to PMCs. Treatment of ESCs with peritoneal fluid from endometriosis patients resulted in a significant increase in binding of ESCs to PMCs compared to untreated ESCs in the endometriosis group. CONCLUSION: This study demonstrates that the expression of CD44 protein in ESCs from women with endometriosis might not be directly associated with adherence to PMCs.

19.
Biomed Pharmacother ; 67(5): 407-15, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23583193

RESUMO

Histone deacetylase (HDAC) inhibitors are a new class of anticancer agents that act by inhibiting cancer cell proliferation and inducing apoptosis in various cancer cell lines. To investigate the anticancer effect of a novel histone deacetylase (HDAC) inhibitor MHY219, its efficacy was compared to that of suberoylanilide hydroxamic acid (SAHA) in human prostate cancer cells. The anticancer effects of MHY219 on cell viability, HDAC enzyme activity, cell cycle regulation, apoptosis and other biological assays were performed. MHY219 was shown to enhance the cytotoxicity on DU145 cells (IC50, 0.36 µM) when compared with LNCaP (IC50, 0.97 µM) and PC3 cells (IC50, 5.12 µM). MHY219 showed a potent inhibition of total HDAC activity when compared with SAHA. MHY219 increased histone H3 hyperacetylation and reduced the expression of class I HDACs (1, 2 and 3) in prostate cancer cells. MHY219 effectively increased the sub-G1 fraction of cells through p21 and p27 dependent pathways in DU145 cells. MHY219 significantly induced a G2/M phase arrest in DU145 and PC3 cells and arrested the cell cycle at G0/G1 phase in LNCaP cells. Furthermore, MHY219 effectively increased apoptosis in DU145 and LNCaP cells, but not PC3 cells, according to Annexin V/PI staining and Western blot analysis. These results indicate that MHY219 is a potent HDAC inhibitor that targets regulating multiple aspects of cancer cell death and might have preclinical value in human prostate cancer chemotherapy, warranting further investigation.


Assuntos
Antineoplásicos/farmacologia , Difenilamina/análogos & derivados , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Difenilamina/administração & dosagem , Difenilamina/farmacologia , Inibidores de Histona Desacetilases/administração & dosagem , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Concentração Inibidora 50 , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptores Androgênicos/genética , Regulação para Cima/efeitos dos fármacos , Vorinostat
20.
J Oral Pathol Med ; 42(1): 17-25, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22742535

RESUMO

BACKGROUND: Pheophorbide a (Pa) is a chlorine-based photosensitizer derived from an ethnopharmacological herb, and our group recently synthesized Pa by the removal of a magnesium ion and a phytyl group from chlorophyll-a. In this study, the effect of photodynamic therapy (PDT) with synthesized Pa was examined in a human oral squamous cell carcinoma (OSCC) cells. METHODS: Cells were treated with PDT with Pa, and reactive oxygen species (ROS) and mitochondrial membrane potential [ΔΨ (m)] were examined. Apoptosis was measured using annexin V staining and immunoblot. Autophagy was characterized by the increase in LC3B-II and the formation of autophagosome and acidic vesicular organelles (AVOs). RESULTS: Pa-PDT inhibited the proliferation of OSCC cells in a dose-dependent manner. Pa-PDT increased the number of apoptotic cells by inactivating ERK pathway. Pa-PDT also induced autophagy in OSCC cells evidenced by the increased levels of LC3 type II expression and the accumulation of AVOs. The inhibition of autophagy enhanced Pa-PDT-mediated cytotoxicity through an increase in necrosis. CONCLUSIONS: These results suggest that synthesized Pa-PDT exerts anti-tumor effects by inducing apoptosis and autophagy and provide novel evidence that Pa-PDT induces autophagy, and autophagy inhibition enhances Pa-PDT-mediated necrosis in OSCC cells.


Assuntos
Apoptose , Autofagia , Carcinoma de Células Escamosas/tratamento farmacológico , Clorofila/análogos & derivados , Neoplasias Bucais/tratamento farmacológico , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Clorofila/síntese química , Clorofila/farmacologia , Clorofila/uso terapêutico , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/biossíntese , Necrose , Fagossomos , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA