Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Arch Oral Biol ; 162: 105940, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38479277

ABSTRACT

OBJECTIVE: Pseudolaric acid B (PAB) is a novel diterpenoid derived from the traditional Chinese medicinal herb Cortex pseudolaricis that exerts anticancer, anti-inflammatory, and immunomodulatory properties. While the anticancer potential of PAB has been studied, its effects on metastasis have not been well-studied. This study aims to determine the inhibitory effects of PAB on HSC-3 human tongue squamous cell carcinoma (TSCC) cell line. DESIGN: Cell viability and soft agar colony formation assays were conducted to assess cellular proliferation and in vitro tumorigenic capacity of TSCC cells, respectively. Additionally, wound healing, transwell migration, and invasion assays were conducted to monitor the aggressive behavior of TSCC cells. Furthermore, Western blotting analysis was conducted to reveal the signaling pathways involved in the modulation of epithelial-mesenchymal transition (EMT). RESULTS: The migratory and invasive capacities of HSC-3 cells were suppressed by PAB irrespective of their proliferation states. PAB's effects on EMT involved upregulation of E-cadherin expression and downregulation of Twist; these were concomitantly accompanied by downregulated phosphorylation of epidermal growth factor receptor (EGFR). CONCLUSIONS: PAB suppresses human TSCC in vitro by regulating Twist/E-cadherin through the EGFR signaling pathway. PAB may have potential as a candidate antimetastatic drug for TSCC treatment.


Subject(s)
Carcinoma, Squamous Cell , Diterpenes , Tongue Neoplasms , Humans , Tongue Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Diterpenes/pharmacology , Cell Proliferation , Tongue/pathology , ErbB Receptors/metabolism , Cadherins/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic
2.
Oncol Rep ; 51(2)2024 02.
Article in English | MEDLINE | ID: mdl-38099422

ABSTRACT

Hypopharyngeal squamous cell carcinoma (HSCC) is a relatively rare form of head and neck cancer that is notorious for its poor prognosis and low overall survival rate. This highlights the need for new therapeutic options for this malignancy. The objective of the present study was to examine the ability of caffeic acid phenethyl ester (CAPE), which is an active compound found in propolis, to combat HSCC tumor growth. CAPE exerted its tumor­suppressive activity in HSCC cell lines through the induction of apoptosis. Mechanistically, the CAPE­mediated apoptotic process was attributed to the perturbation of the mitochondrial membrane potential and the activation of caspase­9. CAPE also modulated survivin and X­linked inhibitor of apoptosis, which are potent members of the inhibitors of apoptosis protein family, either through transcriptional or post­translational regulation, leading to HSCC cell line death. Therefore, the findings of the present study suggested that CAPE is an effective treatment alternative for HSCC via the stimulation of mitochondria­dependent apoptosis.


Subject(s)
Head and Neck Neoplasms , Phenylethyl Alcohol , Humans , Squamous Cell Carcinoma of Head and Neck/drug therapy , Cell Line, Tumor , Phenylethyl Alcohol/pharmacology , Phenylethyl Alcohol/therapeutic use , Apoptosis , Caffeic Acids/pharmacology , Caffeic Acids/therapeutic use , Head and Neck Neoplasms/drug therapy
3.
Int J Mol Med ; 52(5)2023 Nov.
Article in English | MEDLINE | ID: mdl-37711052

ABSTRACT

Podophyllotoxin (PPT), which is derived from the podophyllum plant, exhibits marked cytotoxic effects against cancer cells; however, the precise molecular mechanism underlying its activity against human oral squamous cell carcinoma (OSCC) has not been elucidated. In the present study, the mechanism by which PPT induced cytotoxicity in two OSCC cell lines, HSC3 and HSC4, was determined. The effects of PPT on cytotoxicity in HSC3 and HSC4 cells were analyzed using Annexin V/PI double staining, Sub­G1 analysis, soft agar assays, western blotting, and quantitative PCR. The changes in the mitochondrial membrane potential were assessed using a JC­1 assay and cytosolic and mitochondrial fractionation. A myeloid cell leukemia­1 (Mcl­1) overexpression cell lines were also established to study the role of Mcl­1 on apoptosis. The results showed that PPT inhibited the growth of the two human OSCC cell lines and induced apoptosis, which was accompanied by mitochondrial membrane depolarization. Compared with the control, PPT reduced the expression of Mcl­1 in both cell lines through a proteasome­dependent protein degradation process. Overall, these results suggested that targeting of Mcl­1 protein by PPT induced apoptosis, providing a foundation for further pre­clinical and clinical study of its value in the management of OSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Leukemia , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/drug therapy , Squamous Cell Carcinoma of Head and Neck , Podophyllotoxin/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Mouth Neoplasms/drug therapy , Myeloid Cells
4.
Arch Oral Biol ; 146: 105611, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36577313

ABSTRACT

OBJECTIVE: Caffeic acid phenethyl ester (CAPE), one of the components of propolis that is produced by honeybees, reportedly suppresses multiple diseases, including bacterial infection, inflammation, and cancer. We aimed to investigate the inhibitory effects of CAPE on epithelial-mesenchymal transition (EMT) status and aggressive behaviors of human head and neck squamous cell carcinoma (HNSCC) in vitro and the underlying signaling pathway. DESIGN: To examine the cell growth and in vitro tumorigenic potential of HNSCC cells, cell viability and soft agar colony formation assays, respectively, were performed. Transwell migration and invasion assays were conducted to monitor HNSCC cells' aggressive behaviors. Western blotting and immunocytochemistry analyses were done to investigate the signaling pathway responsible for relieving EMT progression and HNSCC cell aggressiveness. RESULTS: CAPE inhibited the in vitro tumorigenic potential of SNU-1041 cells stimulated by epidermal growth factor and suppressed the migratory and invasive capacities of SNU-1041 cells, irrespective of their cell proliferation state. CAPE was, at least partially, capable of inhibiting EMT progression by upregulating E-cadherin expression, which was accompanied by the reduction of phosphorylated focal adhesion kinase (FAK) and Paxillin. The inhibition of the FAK/Paxillin axis by PF-562271 was sufficient to alleviate the EMT progression through the induction of E-cadherin and aggressive behaviors of SNU-1041 cells. CONCLUSIONS: CAPE has a therapeutic potential as an anti-metastatic drug candidate for HNSCC therapy targeting the FAK/Paxillin axis.


Subject(s)
Caffeic Acids , Head and Neck Neoplasms , Squamous Cell Carcinoma of Head and Neck , Humans , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/therapeutic use , Head and Neck Neoplasms/drug therapy , Paxillin/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Phenylethyl Alcohol , Caffeic Acids/pharmacology
5.
Mol Med Rep ; 17(4): 5258-5264, 2018 04.
Article in English | MEDLINE | ID: mdl-29363716

ABSTRACT

Potentilla discolor has been used in traditional Chinese medicine for the treatment of hyperglycemia. However, the potential role of Potentilla discolor against cancer and its mode of action remain to be fully elucidated. The present study explored the apoptotic effect of methanol extract of Potentilla discolor (MEPD) in human mucoepidermoid carcinoma (MEC) cell lines of salivary glands. MEPD markedly suppressed the growth and induced apoptotic cell death in MC3 and YD15 cells. MEPD treatment significantly upregulated the expression of PUMA and reduced STAT3 phosphorylation. Overexpression of STAT3 partially recovered the growth of MEC cells inhibited by MEPD. In addition, dephosphorylation of STAT3 by cryptotanshinone (a potent STAT3 inhibitor) was sufficient to inhibit the growth of MEC cells and induce apoptosis via affecting PUMA protein. These results suggest that MEPD has a potential anticancer property via the STAT3/PUMA signaling axis in human MEC cells of salivary gland.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Carcinoma, Mucoepidermoid/metabolism , Plant Extracts/pharmacology , Potentilla/chemistry , Proto-Oncogene Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans
6.
Oncotarget ; 8(53): 91306-91315, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-29207645

ABSTRACT

Nitidine chloride (NC) is a natural alkaloid compound derived from the plant Zanthoxylum nitidum and is known for its therapeutic anticancer potential. In this study, we investigated the effects of NC on growth and signaling pathways in human oral cancer cell lines and a tumor xenograft model. The apoptotic effects and related molecular targets of NC on human oral cancer were investigated using trypan blue exclusion assay, DAPI staining, Live/Dead assay, Western blotting, Immunohistochemistry/Immunofluorescence and a nude mouse tumor xenograft. NC decreased cell viability in both HSC3 and HSC4 cell lines; further analysis demonstrated that cell viability was reduced via apoptosis. STAT3 was hyper-phosphorylated in human oral squamous cell carcinoma (OSCC) compared with normal oral mucosa (NOM) and dephosphorylation of STAT3 by the potent STAT3 inhibitor, cryptotanshinone or NC decreased cell viability and induced apoptosis. NC also suppressed cell viability and induced apoptosis accompanied by dephosphorylating STAT3 in four other oral cancer cell lines. In a tumor xenograft model bearing HSC3 cell tumors, NC suppressed tumor growth and induced apoptosis by regulating STAT3 signaling without liver or kidney toxicity. Our findings suggest that NC is a promising chemotherapeutic candidate against human oral cancer.

7.
Arch Oral Biol ; 84: 94-99, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28965045

ABSTRACT

OBJECTIVE: Caffeic acid phenethyl ester (CAPE), a natural honeybee product exhibits a spectrum of biological activities including antimicrobial, anti-inflammatory, antioxidant and antitumor actions. The purpose of this research was to investigate the anticancer potential of CAPE and its molecular mechanism in human oral cancer cell lines (YD15, HSC-4 and HN22 cells). DESIGN: To determine the apoptotic activity of CAPE and identify its molecular targets, trypan blue exclusion assay, soft agar assay, Western blot analysis, DAPI staining, and live/dead assay were performed. RESULTS: CAPE significantly suppressed transformation of neoplastic cells induced by epidermal growth factor (EGF) and 12-O-tetradecanoylphorbol 13-acetate (TPA) without inhibiting growth. CAPE treatment inhibited cell growth, increased the cleavages of caspase-3 and poly (ADP-ribose) polymerase (PARP), and augmented the number of fragmented nuclei in human oral cancer cell lines. CAPE activated Bax protein causing it to undergo a conformational change, translocate to the mitochondrial outer membrane, and oligomere. CAPE also significantly increased Puma expression and interestingly Puma and Bax were co-localized. CONCLUSION: Overall, these results suggest that CAPE is a potent apoptosis-inducing agent in human oral cancer cell lines. Its action is accompanied by up-regulation of Bax and Puma proteins.


Subject(s)
Apoptosis/drug effects , Caffeic Acids/pharmacology , Mouth Neoplasms/drug therapy , Phenylethyl Alcohol/analogs & derivatives , Apoptosis Regulatory Proteins/metabolism , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Humans , Immunohistochemistry , Phenylethyl Alcohol/pharmacology , Proto-Oncogene Proteins/metabolism , Staining and Labeling , bcl-2-Associated X Protein/metabolism
8.
Cell Oncol (Dordr) ; 40(3): 235-246, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28401485

ABSTRACT

PURPOSE: Approximately 20% of all salivary gland cancer patients who are treated with current treatment modalities will ultimately develop metastases. Its most common form, mucoepidermoid carcinoma (MEC) is a highly aggressive tumor with an overall 5-year survival rate of ~30%. Until now, several chemotherapeutic drugs have been tested for the treatment of salivary gland tumors, but the results have been disappointing and the drugs often cause unwanted side effects. Therefore, several recent studies have focused on the potential of alternative and/or complementary therapeutic options, including the use of silymarin. METHODS: The effects of silymarin and its active component silibinin on salivary gland cancer-derived MC3 and HN22 cells and their underlying molecular mechanisms were examined using trypan blue exclusion, 4'-6-diamidino-2-phenylindole (DAPI) staining, Live/Dead, Annexin V/PI staining, mitochondrial membrane potential (ΔΨm) measurement, quantitative RT-PCR, soft agar colony formation and Western blotting analyses. RESULTS: We found that silymarin and silibinin dramatically increased the expression of the pro-apoptotic protein Bim in a concentration- and time-dependent manner and, concomitantly, induced apoptosis in MC3 and HN22 cells. We also found that ERK1/2 signaling inhibition successfully sensitized these cells to the apoptotic effects of silymarin and silibinin, which indicates that the ERK1/2 signaling pathway may act as an upstream regulator that modulates the silymarin/silibinin-induced Bim signaling pathway. CONCLUSIONS: Taken together, we conclude that ERK1/2 signaling pathway inhibition by silymarin and silibinin increases the expression of the pro-apoptotic Bcl-2 family member Bim which, subsequently, induces mitochondria-mediated apoptosis in salivary gland cancer-derived cells.


Subject(s)
Bcl-2-Like Protein 11/drug effects , MAP Kinase Signaling System/drug effects , Salivary Gland Neoplasms/pathology , Silymarin/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Silybin
9.
Phytother Res ; 29(8): 1145-51, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25903972

ABSTRACT

Evodiamine, a bioactive alkaloid, has been regarded as having antioxidant, antiinflammatory, and anticancer properties. In the present study, we explored the effects of evodiamine on cell growth and apoptosis in human oral cancer cell lines. Our data revealed that evodiamine significantly inhibited the proliferation of human oral cancer cells and resulted in the cleavages of PARP (poly (ADP-ribose) polymerase) and caspase-3, in addition to causing the typical characteristics of apoptosis. Evodiamine also increased Bax protein levels and caused translocation of Bax into mitochondria and Bax oligomerization. In addition, evodiamine decreased expression of myeloid cell leukemia (Mcl-1) at the transcriptional modification, and knockdown of Mcl-1 clearly resulted in an increase in expression of Bax and active Bax, resulting in induction of apoptosis. Evodiamine reduced expression of phosphorylated AKT, and LY294002 potentiated evodiamine-induced apoptosis by regulating Mcl-1 protein. Our results suggest that evodiamine induces apoptosis in human oral cancer cells through the AKT pathway. These findings provide a rationale for its clinical application in the treatment of oral cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Mouth Neoplasms/metabolism , Quinazolines/pharmacology , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Chromones , Humans , Mitochondria/drug effects , Morpholines , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Phosphorylation , Plant Extracts/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , bcl-2-Associated X Protein/metabolism
10.
Oral Oncol ; 51(6): 602-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25817195

ABSTRACT

OBJECTIVES: Parthenolide (PTL), a representative sesquiterpene lactone that is responsible for medicinal properties of the feverfew, is known to modulate diverse intracellular signaling pathways, thereby exerting the tumor growth-inhibitory effects. In this study, authors attempted to examine the pro-apoptotic effects and possible biochemical mechanisms of PTL in human oral cancer cell lines and tumor xenografts. MATERIAL AND METHODS: The apoptotic effects and related molecular mechanisms of PTL on oral cancer were evaluated using cell viability assay, MTS assay, DAPI staining, western blot analysis, reverse transcriptase-polymerase chain reaction, small interfering RNA transfection and nude mouse xenograft assay. RESULTS: PTL treatment increased the cleavage of caspase-3 and poly (ADP-ribose) polymerase (PARP), and the nuclear fragmentation in a concentration- or time-dependent manner. PTL treatment increased Bim protein expression by enhancing the Bim mRNA expression as well as stabilizing Bim protein level. PTL treatment also induced the translocation of cytosolic Bim into the mitochondria and, more importantly, PTL-induced apoptosis was significantly attenuated, when the Bim expression was knockdown by siRNA transfection. PTL treatment also induced death receptor 5 (DR5) protein expression and this event was closely correlated with an increase in the cleavage of caspase-8 and formation of truncation of Bid (t-Bid). Finally, PTL shrunk tumor size and volume resulting from apoptotic cell death by increasing Bim and DR5 whereas there were no abnormal histopathological findings in normal organs. CONCLUSION: This study proposes that PTL is a strong apoptotic inducer that deserves the further investigations for potential chemotherapeutic agent of human oral cancers.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Mouth Neoplasms/metabolism , Sesquiterpenes/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Bcl-2-Like Protein 11 , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Female , Humans , Membrane Proteins/metabolism , Mice , Mice, Nude , Mitochondria/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
11.
J Oral Pathol Med ; 44(10): 785-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25475012

ABSTRACT

BACKGROUND: YM155 is a small-molecule pro-apoptotic agent which has shown to inhibit survivin expression and induce apoptosis in various cancer cells. In this study, we investigated the function and molecular mechanism of YM155 in human oral cancer cells. METHODS: The apoptotic effects and related signaling pathways of YM155 were evaluated using trypan blue exclusion assay, 4'-6-diamidino-2-phenylindole staining, Western blotting, RT-PCR, and siRNA. RESULTS: YM155 inhibited the growth and caused caspase-dependent apoptosis in MC3 and HN22 cells. YM155 significantly suppressed the level of survivin protein expression through proteasome-dependent protein degradation to confirm its survivin-inhibiting function. YM155 reduced myeloid cell leukemia-1 (Mcl-1) protein, but it did not alter Mcl-1 mRNA. It was associated with the facilitation of lysosome-dependent protein degradation. The modifications of Mcl-1 and survivin by YM155 were caspase-independent manner. Treatment of MC-3 and HN22 cells with YM155 inhibited specificity protein 1 (Sp1) and the knockdown of Sp1 by siRNA demonstrated that Mcl-1 was regulated by Sp1 protein. CONCLUSIONS: We demonstrated the novel mechanism that YM155 causes apoptosis of human oral cancer cell lines through downregulation of Sp1 and Mcl-1. Therefore, it may be a potential anticancer drug candidate for the treatment of oral cancer.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Mouth Neoplasms/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Naphthoquinones/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Gene Knockdown Techniques , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Survivin
12.
Head Neck ; 37(9): 1326-35, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25043125

ABSTRACT

BACKGROUND: The purpose of our study was to investigate the anticancer effect of sorafenib on mucoepidermoid carcinoma (MEC) and find its new molecular mechanism. METHODS: The apoptotic effects of sorafenib were performed using MTS assay, diamidino-phenylindole (DAPI) staining, Western blotting, reverse transcription-polymerase chain reaction (RT-PCR), siRNA, and xenograft. RESULTS: Sorafenib had apoptotic effects on MC-3 and YD15 cells and decreased myeloid cell leukemia-1 (Mcl-1) through proteasome-dependent protein degradation and the inhibition of protein synthesis. Sorafenib significantly affected truncated bid (t-Bid) and siMcl-1 resulting in the upregulation of t-Bid to induce apoptosis. Signal transducer and activator of transcription 3 (STAT3) phosphorylation was also blocked by sorafenib and a potent STAT3 inhibitor, cryptotanshinone clearly induced poly ADP-ribose polymerase (PARP) cleavage by inhibiting Mcl-1 and increasing t-Bid. Finally, administration of sorafenib significantly suppressed tumor growth and induced apoptosis in tumor xenograft model in association with downregulation of Mcl-1 without any side effects. CONCLUSION: Taken together, these findings suggest that sorafenib can be a good anticancer drug candidate for the treatment of MEC.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Animals , Apoptosis/genetics , Blotting, Western , Carcinoma, Mucoepidermoid/drug therapy , Carcinoma, Mucoepidermoid/pathology , Disease Models, Animal , Down-Regulation , Female , Heterografts/drug effects , Heterografts/pathology , Humans , Mice , Mice, Nude , Niacinamide/pharmacology , RNA, Small Interfering/metabolism , Random Allocation , Real-Time Polymerase Chain Reaction , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/pathology , Sensitivity and Specificity , Signal Transduction , Sorafenib , Tumor Cells, Cultured
13.
Pharmacogn Mag ; 10(Suppl 3): S622-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25298683

ABSTRACT

BACKGROUND: Cryptotanshinone (CT) is a biologically active compound from the root of Salvia miltiorrhiza that has been reported to induce apoptosis in various cancer cell lines; but, it has not yet been fully explored in human mucoepidermoid carcinoma (MEC). OBJECTIVE: Here, we demonstrated the apoptotic effects and its related mechanism in MC-3 and YD-15 human MEC cell lines. MATERIALS AND METHODS: The effects of CT on apoptotic activity were evaluated by cell proliferation assay, Western blotting, 4'-6-diamidino-2-phenylindole staining, reverse transcription-polymerase chain reaction, and luciferase assay. RESULTS: Our data show that CT treatment of MC-3 cells results in anti-proliferative and apoptotic activities in MC-3 and it is accompanied by a decrease in phosphorylation and dimerization of signal transducer and activators of transcription 3 (STAT3). CT decreased the expression levels of myeloid cell leukemia-1 (Mcl-1) and surviving, whereas Bcl-xL expression was not changed. CT clearly regulates survivin protein at a transcriptional level and alters Mcl-1 through proteasome-dependent protein degradation. In addition, CT-induced apoptotic cell death in YD-15, another human MEC cell line, was associated with the inhibition of STAT3 phosphorylation. CONCLUSION: These data suggest that CT could be a good apoptotic inducer through modification of STAT3 signaling in human MEC cell lines.

14.
Mol Med Rep ; 9(2): 663-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24270523

ABSTRACT

Smilax china L., a well­known Chinese traditional medicine, has been used as an anti­inflammatory, anti­cancer and analgesic agent, but its role has not yet been fully elucidated in oral mucoepidermoid carcinoma (MEC). The present study focused on addressing the anticancer activity and molecular mechanism of methanol extract of Smilax china L. (MESC) in MC­3 human oral MEC cells. The results indicated that MESC inhibited cell growth and induced apoptosis in MC­3 cells. These observations were found to correlate with increases in truncated BH3 interacting­domain death agonist and B­cell lymphoma 2 (Bcl­2) interacting mediator of cell death, but not Bcl­2 homologous antagonist killer, Bcl­2­associated X protein, Bcl­2, B­cell lymphoma­extra large and induced myeloid leukemia cell differentiation protein levels. MESC also damaged the mitochondrial membrane potential, cleaved caspase­8 protein and increased death receptor 5 (DR5) protein levels by enhancing the stability of DR5 protein. Furthermore, MESC affected the phosphorylation of extracellular signal­regulated kinase (ERK) only, and did not affect c­Jun N­terminal kinase or p38 phosphorylation. Co­treatment with MESC and an ERK inhibitor (PD98059) significantly increased the expression of DR5 to induce apoptosis in MC­3 cells. Therefore, these results suggest that MESC may induce apoptosis via the ERK pathway and may be a potential anticancer drug candidate against human oral MEC.


Subject(s)
Carcinoma, Mucoepidermoid/drug therapy , Mouth Neoplasms/drug therapy , Plant Extracts/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/drug effects , Carcinoma, Mucoepidermoid/genetics , Carcinoma, Mucoepidermoid/pathology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/drug effects , Flavonoids/administration & dosage , Humans , MAP Kinase Signaling System/drug effects , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Plant Extracts/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Smilax/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...