Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Cell Mol Med ; 27(9): 1250-1260, 2023 05.
Article in English | MEDLINE | ID: mdl-36967712

ABSTRACT

Oral squamous cell carcinoma (OSCC) has a high recurrence rate and poor prognosis. Hispolon, a polyphenolic compound with antiviral, antioxidant, and anticancer activities, is a potential chemotherapy agent. However, few studies have investigated the anti-cancer mechanism of hispolon in oral cancer. This present study used the cell viability assay, clonogenic assay, fluorescent nuclear staining, and flow cytometry assay to analyse the apoptosis-inducing effects of hispolon in OSCC cells. After hispolon treatment, the apoptotic initiators, cleaved caspase-3, -8, and - 9, were upregulated, whereas the cellular inhibitor of apoptosis protein-1 (cIAP1) was downregulated. Furthermore, a proteome profile analysis using a human apoptosis array revealed the overexpression of heme oxygenase-1 (HO-1) by hispolon, which was determined to be involved in caspase-dependent apoptosis. Moreover, cotreatment with hispolon and mitogen-activated protein kinase (MAPK) inhibitors revealed that hispolon induces apoptosis in OSCC cells through activation of the c-Jun N-terminal kinase (JNK) pathway and not the extracellular signal-regulated kinase (ERK) or p38 pathway. These findings indicate that hispolon may exert an anticancer effect on oral cancer cells by upregulating HO-1 and inducing caspase-dependent apoptosis by activating the JNK pathway.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , MAP Kinase Signaling System , Heme Oxygenase-1 , Carcinoma, Squamous Cell/drug therapy , Squamous Cell Carcinoma of Head and Neck , Mouth Neoplasms/drug therapy , Apoptosis , JNK Mitogen-Activated Protein Kinases , Cell Line, Tumor , p38 Mitogen-Activated Protein Kinases
2.
J Cell Mol Med ; 26(21): 5452-5462, 2022 11.
Article in English | MEDLINE | ID: mdl-36226563

ABSTRACT

Tumour necrosis family superfamily (TNFSF) member 15 (TNFSF15), encoded by TNFSF15, regulates immune responses and inflammation. However, the roles of TNFSF15 single-nucleotide variants (SNVs; formerly SNPs) in oral cavity squamous cell carcinoma (OCSCC) remain unclear. This case-control study included 2523 participants (1324 patients with OCSCC [52.5%] and 1199 healthy controls [47.5%]). The effects of TNFSF15 rs3810936, rs6478108 and rs6478109 on cancer development and prognosis were analysed by real-time PCR genotype assay. The Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) databases were used to validate our findings. The results demonstrated that the patients with altered TNFSF15 SNVs had poorer histological differentiation than did those with wild-type alleles. TNFSF15 SNVs were significantly associated with moderate-to-poor histological differentiation in univariate logistic regression. In the GTEx database, the expression of altered TNFSF15 SNVs in whole blood was lower than that of wild-type alleles. However, the expression of altered SNVs in the upper aerodigestive mucosa was higher than that of wild-type alleles. In the TCGA database, the patients with higher TNFSF15 expression had shorter overall survival than did those with lower TNFSF15 expression, especially for human papillomavirus-negative and advanced staging groups. In conclusion, although TNFSF15 SNVs did not affect OCSCC development, the patients with altered TNFSF15 SNVs exhibited poorer histological differentiation. The patients with higher TNFSF15 expression had poorer prognosis than did those with lower TNFSF15 expression.


Subject(s)
Genetic Predisposition to Disease , Mouth Neoplasms , Humans , Case-Control Studies , Genotype , Polymorphism, Single Nucleotide/genetics , Mouth Neoplasms/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
3.
J Oral Pathol Med ; 51(8): 730-737, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35880802

ABSTRACT

OBJECTIVE: To evaluate the associations between dipeptidyl peptidase IV (DPP4) single-nucleotide polymorphism (SNP) and clinicopathological characteristics of oral cancer. METHODS: Four loci of DPP4 SNPs (rs7608798 A/G, rs3788979 C/T, rs2268889 T/C, and rs6741949 G/C) were genotyped by using the TaqMan allelic discrimination in 1238 oral cancers patients and 1197 non-cancer individuals. RESULTS: The percentage of DPP4 SNP rs2268889 TC + CC was significantly higher in the oral cancer participants compared to the control group (odds ratio [OR]: 1.178, 95% confidence interval (CI): 1.004-1.382, p = 0.045). Among 1676 smokers, DPP4 polymorphisms carriers with betel quid chewing were found to have an 8.785- to 10.903-fold risk to have oral cancer compared to DPP4 wild-type carriers without betel quid chewing. Similar trend was found in individuals with alcohol consumption. Moreover, the oral cancer individuals without cigarette smoking history with at least one varied C allele of DPP4 rs2268889 had a significantly higher percentage of large tumor size with the wild-type TT homozygote (p = 0.011). CONCLUSIONS: The DPP4 SNP may correlate to the development of oral cancer in those with cigarette smoking and alcohol consumption. Besides, the DPP4 SNP rs2268889 could relate to worse clinical course of oral cancer in non-smokers.


Subject(s)
Dipeptidyl Peptidase 4 , Mouth Neoplasms , Alleles , Areca/adverse effects , Dipeptidyl Peptidase 4/genetics , Genotype , Humans , Mouth Neoplasms/genetics , Polymorphism, Single Nucleotide
4.
Int J Med Sci ; 19(7): 1198-1204, 2022.
Article in English | MEDLINE | ID: mdl-35919812

ABSTRACT

Oral squamous cell carcinoma (OSCC) is particularly prevalent in Taiwan. The goal of this study was to determine the clinicopathological role of insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) proteins as an indicator of clinical outcomes in OSCC patients. In this study, immunohistochemical (IHC) analysis was used to examine IGF2BP2 protein expression in 244 OSCC patients. We investigated the relationships among IGF2BP2 expression, clinicopathological variables, and patient survival. Our results showed that IGF2BP2 cytoplasmic protein expression was significantly correlated with lymph node metastasis, cancer stage, and patient survival. Kaplan-Meier survival curves revealed that elevated cytoplasmic IGF2BP2 expression levels in OSCC patients were associated with poor overall survival. Moreover, multivariate cox proportional hazard models revealed that cytoplasmic IGF2BP2 expression, T status, and lymph node metastasis were independent prognostic factors for survival. In conclusion, IGF2BP2 protein was found to be a helpful predictive marker for OSCC patients, as well as a possible therapeutic target for OSCC treatment.


Subject(s)
Head and Neck Neoplasms , RNA-Binding Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck , Biomarkers, Tumor/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/physiopathology , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , Prognosis , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/physiopathology
5.
Int J Mol Sci ; 23(13)2022 Jun 26.
Article in English | MEDLINE | ID: mdl-35806120

ABSTRACT

Deoxyshikonin (DSK), a phytochemical constituent, has been documented to elicit various oncostatic properties alone or in combination with established therapeutics. However, its role in restraining oral squamous cell carcinoma (OSCC) is mostly unclear. Here, we examined the tumor-suppressive effect of DSK and explored the molecular mechanisms underlying DSK's activities on controlling oral cancer. Our results showed that DSK dose-dependently lessened the cell viability of tongue cancer cell lines, involving induction of cell cycle arrest at the sub-G1 phase and apoptotic cell death. Moreover, a unique signature of apoptosis-related proteins, including augmented nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) expression and caspase activation, was observed in DSK-treated tongue cancer cell lines. Furthermore, DSK-mediated upregulation of HO-1 and cleavage of caspase-9 and -3 were significantly inhibited by pharmacological blockage of p38 kinase. Collectively, these data revealed that DSK halted cell cycle progression and elicited cell apoptosis in tongue cancer cell lines, reshaping a p38-dependent profile of apoptotic proteome. Our findings provided novel insights into the therapeutic implications of a natural compound on the management of OSCC.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Tongue Neoplasms , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Heme Oxygenase-1/metabolism , Humans , Mouth Neoplasms/pathology , NF-E2-Related Factor 2/metabolism , Naphthoquinones , Tongue Neoplasms/drug therapy , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Int J Mol Sci ; 22(21)2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34769290

ABSTRACT

Oral cancer is the most common oral malignant tumor in Taiwan. Although there exist several methods for treatment, oral cancer still has a poor prognosis and high recurrence. FLLL32, a synthetic analog of curcumin with antitumor activity, is currently known to induce melanoma apoptosis and inhibit tumor growth in various cancers. However, few studies have examined the mechanisms of FLLL32 in oral cancer. In this study, we explore whether FLLL32 induces apoptosis in oral cancer. We determined that FLLL32 can inhibit the cell viability of oral cancer. Next, we analyzed the effect of FLLL32 on the cell cycle of oral cancer cells and observed that the proportion of cells in the G2/M phase was increased. Additionally, annexin-V/PI double staining revealed that FLLL32 induced apoptosis in oral cancer cells. Data from the Human Apoptosis Array revealed that FLLL32 increases the expression of cleaved caspase-3 and heme oxygenase-1 (HO-1). FLLL32 activates proteins such as caspase-8, caspase-9, caspase-3, PARP, and mitogen-activated protein kinases (MAPKs) in apoptosis-related molecular mechanisms. Moreover, by using MAPK inhibitors, we suggest that FLLL32 induces the apoptosis of oral cancer cells through the p38 MAPK signaling pathway. In conclusion, our findings suggest that FLLL32 is a potential therapeutic agent for oral cancer by inducing caspase-dependent apoptosis and HO-1 activation through the p38 pathway. We believe that the activation of HO-1 and the p38 pathway by FLLL32 represent potential targets for further research in oral cancer.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Caspase 3/metabolism , Curcumin/analogs & derivatives , Heme Oxygenase-1/metabolism , Mouth Neoplasms/metabolism , Carcinoma, Squamous Cell/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Curcumin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Mouth Neoplasms/drug therapy
7.
Cancer Metastasis Rev ; 38(4): 829-837, 2019 12.
Article in English | MEDLINE | ID: mdl-31802358

ABSTRACT

Matrix metalloproteinases (MMPs) play a major role in extracellular matrix remodeling and are involved in tumor cell invasion. Cancers such as childhood leukemia are characterized by their capacity to infiltrate different organs. MMP production by leukemic cells may indicate a leukemic subtype or subpopulation with a more invasive phenotype. Therefore, clarifying the action mechanisms of MMPs as prognostic predictors or MMP targeting as a therapeutic strategy is necessary. MMP-targeting drugs have been developed for the treatment of hematological malignancies. In this review, we highlight current advances in understanding the molecular mechanisms and pathological characteristics of various MMPs, as well as recent therapeutic advances targeting MMPs in childhood leukemia. Several studies have been conducted on the therapeutic efficacy of MMP inhibitors in cancer, such as collagen peptidomimetics, nonpeptidomimetic inhibitors of MMP active sites, bisphosphonates, and tetracycline derivatives. Here, we conclude that more clinical trials are necessary to estimate the role of selective MMP inhibitors in the treatment and prevention of childhood leukemia.


Subject(s)
Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/therapy , Matrix Metalloproteinases/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Animals , Child , Humans
8.
Environ Toxicol ; 35(5): 619-627, 2020 May.
Article in English | MEDLINE | ID: mdl-31916385

ABSTRACT

Cantharidic acid (CA) is the hydrolysis product of the acid anhydride cantharidin, which is a natural toxin secreted by several species of blister beetles. Several studies have indicated that as an inhibitor of protein phosphatase 2 (PP2A), CA induces apoptosis in various human cancer cells. However, the effect of CA on human nasopharyngeal carcinoma (NPC) cells and the underlying pathways have not been addressed. In our current study, we tested the hypothesis that CA treatment reduces the viability of human NPC cells (HONE-1, NPC-39, and NPC-BM) by inducing apoptosis. Results indicated that CA markedly reduced cell viability, which was revealed by the upregulation of caspase activation in extrinsic and intrinsic apoptosis pathways as well as the upregulation of extracellular-signal-regulated kinase 1/2 (ERK1/2), p38, and c-Jun N-terminal kinase 1/2 (JNK1/2) pathways. Coadministration of a p38 inhibitor (SB203580) with CA abolished the activation of caspase proteins. These findings indicated that CA treatment leads to apoptosis in human NPC cells through the upregulation of caspase activation, mediated particularly by the p38 pathway. Hence, CA is a promising therapeutic agent for human NPC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cantharidin/analogs & derivatives , Caspases/metabolism , Transcriptional Activation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Cantharidin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , MAP Kinase Signaling System/drug effects , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Signal Transduction , Up-Regulation
9.
Exp Eye Res ; 175: 181-191, 2018 10.
Article in English | MEDLINE | ID: mdl-29935949

ABSTRACT

Matrix metalloproteinase (MMP)-8 is the most potent MMP for degrading collagen type-1 and plays an important role in inflammatory reactions and tissue remolding processes. MMP-8 is expressed mainly by polymorphonuclear leukocytes and is not expressed constitutively by most non-leukocytes. We studied the constitutive and TNF-α-induced expression of MMP-8 in cultured human uveal melanocytes (UM) and the relevant signal pathways involved. Conditioned media and cells were collected from UM and other cell types. MMP-8 proteins and mRNA were measured using ELISA kit, western blot and real time RT-PCR, respectively. Phosphorylated p38 MAPK, ERK1/2, and JNK1/2 were measured by ELISA kit and western blot. Very high levels of MMP-8 proteins and mRNA were detected in the conditioned media and cell lysates in 11 UM cell lines and three uveal melanoma cell lines cultured without serum, but not in media and cell lysates from other ocular resident cells or 12 malignant cell lines from other tissues, with exception of cutaneous melanoma cells. TNF-α moderately increased MMP-8 mRNA and protein levels in a dose- and time-dependent manner, accompanied by a significant increase of phosphorylated JNK1/2 and ERK1/2 in cell lysates. ERK1/2 (U0126) and JNK1/2 (SP600125) inhibitors significantly blocked TNF-α-induced and constitutive expression of MMP-8 in UM. This is the first report on the expression and secretion of MMP-8 by UM and uveal melanoma cells. The data suggest that UM may play a role in the remolding process and pathogenesis of inflammatory-related diseases in the eye via secretion of MMP-8.


Subject(s)
Gene Expression Regulation/physiology , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/metabolism , Melanocytes/drug effects , Mitogen-Activated Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Uvea/cytology , Adult , Aged , Blotting, Western , Cell Line , Enzyme-Linked Immunosorbent Assay , Female , Humans , MAP Kinase Signaling System/physiology , Male , Melanocytes/metabolism , Middle Aged , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Environ Toxicol ; 33(5): 527-534, 2018 May.
Article in English | MEDLINE | ID: mdl-29383865

ABSTRACT

Fisetin, a diatery flavonoid, been reported that possess anticancer effects in various cancers. The purpose of the study was to investigate the antitumor effects of fisetin in cultured uveal melanoma cell lines and compared with normal retinal pigment epithelial (RPE) cells. MTT assay was used for evaluating cytotoxic effects of fisetin. Flow cytometry study was used for the determination of apoptosis. JC-1 fluorescent reader was used to determine mitochondrial transmembrane potential changes. The results shown that fisetin dose-dependently decreased the cell viability of uveal melanoma cells but not influenced the cell viability of RPE cells. Apoptosis of uveal melanoma cells was induced by fisetin efficiently. Fisetin inhibited antiapoptotic Bcl-2 family proteins and damaged the mitochondrial transmembrane potential. The levels of proapoptotic Bcl-2 proteins, cytochrome c, and various caspase activities were increased by fisetin. In conclusion, fisetin induces apoptosis of uveal melanoma cells selectively and may be a promising agent to be explored for the treatment of uveal melanoma.


Subject(s)
Apoptosis/drug effects , Flavonoids/pharmacology , Melanoma/pathology , Mitochondria/drug effects , Uveal Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Flavonols , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
11.
J Pineal Res ; 62(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-27706852

ABSTRACT

Melatonin is a naturally occurring molecule secreted by the pineal gland and known as a gatekeeper of circadian clocks. Mounting evidence indicates that melatonin, employing multiple and interrelated mechanisms, exhibits a variety of oncostatic properties in a myriad of tumors during different stages of their progression. Tumor metastasis, which commonly occurs at the late stage, is responsible for the majority of cancer deaths; metastases lead to the development of secondary tumors distant from a primary site. In reference to melatonin, the vast majority of investigations have focused on tumor development and progression at the primary site. Recently, however, interest has shifted toward the role of melatonin on tumor metastases. In this review, we highlight current advances in understanding the molecular mechanisms by which melatonin counteracts tumor metastases, including experimental and clinical observations; emphasis is placed on the impact of both cancer and non-neoplastic cells within the tumor microenvironment. Due to the broad range of melatonin's actions, the mechanisms underlying its ability to interfere with metastases are numerous. These include modulation of cell-cell and cell-matrix interaction, extracellular matrix remodeling by matrix metalloproteinases, cytoskeleton reorganization, epithelial-mesenchymal transition, and angiogenesis. The evidence discussed herein will serve as a solid foundation for urging basic and clinical studies on the use of melatonin to understand and control metastatic diseases.


Subject(s)
Melatonin/metabolism , Neoplasm Metastasis/pathology , Animals , Humans
12.
Int J Med Sci ; 14(11): 1094-1100, 2017.
Article in English | MEDLINE | ID: mdl-29104463

ABSTRACT

In Taiwan, oral cancer is the fourth most common cancer and the most common malignancy with a poor prognosis. Endothelial cell-specific molecule-1 (ESM-1) is secreted by vascular endothelial cells in the liver, lungs, kidneys, and gastrointestinal tract. ESM-1 expression is associated with tumor prognosis, metastasis, and angiogenesis in many cancers. However, few studies have examined the association of plasma ESM-1 levels with oral squamous cell carcinoma (OSCC) progression. We measured the plasma ESM-1 levels of 438 male OSCC patients through a commercial enzyme-linked immunosorbent assay. The Cancer Genome Atlas (TCGA) dataset was also used to analyze the ESM-1 levels in 328 OSCC patients and 33 normal tissues. Our results revealed that the plasma levels of ESM-1 in OSCC patients were significantly associated with the tumor (T) status but not with the lymph node status, metastasis, and cell differentiation. TCGA bioinformatics database analysis revealed that ESM-1 expression was significantly higher in OSCC patients than in normal individuals (p < 0.05). In addition, the examination revealed similar results for the ESM-1 expression levels and pathological stage in OSCC. In conclusion, plasma ESM-1 is a novel biomarker for predicting the T status in OSCC patients.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/blood , Mouth Neoplasms/pathology , Neoplasm Proteins/blood , Proteoglycans/blood , Adult , Aged , Cell Differentiation/genetics , Cell Differentiation/physiology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged
13.
Int J Med Sci ; 14(5): 419-424, 2017.
Article in English | MEDLINE | ID: mdl-28539817

ABSTRACT

EGF-like domain 6 (EGFL6), a member of the epidermal growth factor (EGF) repeat protein superfamily, is a secreted protein that promotes endothelial cell migration and angiogenesis. The current study investigated the association between the clinicopathological characteristics and plasma level of EGFL6 in patients with oral squamous cell carcinoma (OSCC). We measured the plasma EGFL6 levels of 392 OSCC patients by using a commercial enzyme-linked immunosorbent assay. We also analyzed EGFL6 mRNA levels of 328 OSCC patients from The Cancer Genome Atlas (TCGA) dataset. The results showed that plasma EGFL6 levels were significantly higher in patients with OSCC than in healthy controls (p < 0.001). Similar results were observed for the TCGA bioinformatics database. Moreover, plasma EGFL6 levels were significantly higher in the patients with advanced T status (p = 0.002), distant metastasis (p = 0.001), and higher TNM stage (p=0.033). In conclusion, our results suggest that plasma level of EGFL6 may be useful to assess disease progression, and especially advanced T status and higher TNM stage in patients with OSCC.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/blood , Membrane Glycoproteins/blood , Mouth Neoplasms/blood , Adult , Aged , Calcium-Binding Proteins , Carcinoma, Squamous Cell/pathology , Cell Adhesion Molecules , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Mouth Neoplasms/pathology
14.
Carcinogenesis ; 37(7): 712-722, 2016 07.
Article in English | MEDLINE | ID: mdl-27207653

ABSTRACT

Lipocalin 2 (LCN2), a secreted glycoprotein, is up- or downregulated in different human cancers. At present, the functional role of LCN2 in the progression of oral squamous cell carcinoma (OSCC), which accounts for most head and neck cancers, remains poorly understood, particularly with respect to its involvement in invasion and metastasis. In this study, we observed that LCN2 expression decreased in patients with OSCC and lymph node metastasis compared with that in patients without metastasis. A higher LCN2 expression correlated with the survival of patients with OSCC. Furthermore, LCN2 overexpression in OSCC cells reduced in vitro migration and invasion and in vivo metastasis, whereas its silencing induced an increase in cell motility. Mechanistically, LCN2 inhibited the cell motility of OSCC cells through hypoxia-inducible factor (HIF)-1α-dependent transcriptional inhibition of the carbonic anhydrase IX (CAIX). CAIX overexpression relieved the migration inhibition imposed by LCN2 overexpression in OSCC cells. Moreover, a microRNA (miR) analysis revealed that LCN2 can suppress CAIX expression and cell migration through miR-4505 induction. Examination of tumour tissues from patients with OSCC and OSCC-transplanted mice revealed an inverse correlation between LCN2 and CAIX expression. Furthermore, patients with LCN2(strong)/CAIX(weak) revealed the lowest frequency of lymph node metastasis and the longest survival. Our findings suggest that LCN2 suppresses tumour metastasis by targeting the transcriptional and post-transcriptional regulation of CAIX in OSCC cells. LCN2 overexpression may be a novel OSCC treatment strategy and a useful biomarker for predicting OSCC progression.


Subject(s)
Antigens, Neoplasm/biosynthesis , Carbonic Anhydrase IX/biosynthesis , Carcinoma, Squamous Cell/genetics , Lipocalin-2/biosynthesis , Mouth Neoplasms/genetics , Prognosis , Adult , Animals , Antigens, Neoplasm/genetics , Carbonic Anhydrase IX/genetics , Cell Line, Tumor , Cell Movement/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lipocalin-2/genetics , Lymphatic Metastasis/pathology , Male , Mice , Middle Aged , Mouth Neoplasms/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
15.
Am J Chin Med ; 52(2): 565-581, 2024.
Article in English | MEDLINE | ID: mdl-38480502

ABSTRACT

L48H37 is a synthetic curcumin analog that has anticancer potentials. Here, we further explored the anticancer effect of L48H37 on oral cancer cells and its mechanistic acts. Cell cycle distribution was assessed using flow cytometric analysis. Apoptosis was elucidated by staining with PI/Annexin V and activation of the caspase cascade. Cellular signaling was explored using apoptotic protein profiling, Western blotting, and specific inhibitors. Our findings showed that L48H37 significantly reduced the cell viability of SCC-9 and HSC-3 cells, resulting in sub-G1 phase accumulation and increased apoptotic cells. Apoptotic protein profiling revealed that L48H37 increased cleaved caspase-3, and downregulated cellular inhibitor of apoptosis protein 1 (cIAP1) and X-linked inhibitor of apoptosis protein (XIAP) in SCC-9 cells, and the downregulated cIAP1 and XIAP in both oral cancer cells were also demonstrated by Western blotting. Meanwhile, L48H37 triggered the activation of caspases and mitogen-activated protein kinases (MAPKs). The involvement of c-Jun N-terminal kinase (JNK) and p38 MAPK (p38) in the L48H37-triggered apoptotic cascade in oral cancer cells was also elucidated by specific inhibitors. Collectively, these findings indicate that L48H37 has potent anticancer activity against oral cancer cells, which may be attributed to JNK/p38-mediated caspase activation and the resulting apoptosis. This suggests a potential benefit for L48H37 for the treatment of oral cancer.


Subject(s)
Curcumin , Mouth Neoplasms , Humans , Caspases/metabolism , Curcumin/pharmacology , Cell Line, Tumor , Apoptosis , p38 Mitogen-Activated Protein Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Caspase 3/metabolism , Mouth Neoplasms/drug therapy , Inhibitor of Apoptosis Proteins/pharmacology
16.
Int J Biol Sci ; 20(3): 818-830, 2024.
Article in English | MEDLINE | ID: mdl-38250159

ABSTRACT

Insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2), with high affinity to a myriad of RNA transcripts, has been shown to elicit promotive effects on tumorigenesis and metastasis. Yet, the functional involvement of IGF2BP2 in the progression of oral squamous cell carcinoma (OSCC) remains poorly understood. In this study, we showed that IGF2BP2 was upregulated in head and neck cancer, and high levels of IGF2BP2 were associated with poor survival. In in vitro experiments, IGF2BP2 promoted migration and invasion responses of OSCC cells. Moreover, we identified an IGF2BP2-regulated gene, EREG, which functioned as a modulator of OSCC invasion downstream of IGF2BP2. In addition, EREG expression triggered the epithelia-mesenchymal transition (EMT) in OSCC, as evidenced by the observation that knockdown of EREG weakened the induction of EMT mediated by IFG2BP2, and replenishment of EREG favored the EMT in IGF2BP2-depleted cells. Such IGF2BP2-regulated EREG expression, EMT, and cell invasion were dependent on the activation of FAK/Src signaling pathway. Collectively, these findings suggest that EREG, serving as a functional mediator of IGF2BP2-regulated EMT and cell invasion in oral cancer, may be implicated as a potential target for antimetastatic therapies.


Subject(s)
Mouth Neoplasms , RNA-Binding Proteins , Squamous Cell Carcinoma of Head and Neck , Humans , Epiregulin , Epithelial-Mesenchymal Transition/genetics , Mouth Neoplasms/genetics , RNA-Binding Proteins/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Up-Regulation/genetics
17.
Aging (Albany NY) ; 14(10): 4556-4571, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35613852

ABSTRACT

The gene encoding aldehyde dehydrogenase 7 family member A1 (ALDH7A1) has been associated with the development and prognosis in multiple cancers; however, the role of ALDH7A1 polymorphisms in oral cancer remains unknown. For this purpose, the influences of ALDH7A1 rs13182402 and rs12659017 on oral cancer development and prognosis were analyzed. Our resulted showed that ALDH7A1 rs13182402 genotype had less pathologic nodal metastasis among betel quid chewer. ALDH7A1 rs13182402 also corresponded to higher expressions in upper aerodigestive mucosa, whole blood, the musculoskeletal system and oral cancer tissues than did the ALDH7A1 wild type. Furthermore, ALDH7A1 overexpression in oral cancer cells increased in vitro migration, whereas its silencing reduced cell migration. Conversely, ALDH7A1 expression in tumor tissues and in patients with advanced disease was lower than that in normal tissues and in patients with early-stage disease. When the patients were classified into ALDH7A1-high and -low-expression groups, the high-ALDH7A1 group had superior outcomes in progression-free survival than the low-ALDH7A1 group (5-year survival of 58.7% vs. 48.0%, P = 0.048) did. In conclusion, patients with high ALDH7A1 expression might, however, have more favorable prognoses than those with low ALDH7A1 expression have.


Subject(s)
Aldehyde Dehydrogenase , Mouth Neoplasms , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Humans , Mouth Neoplasms/enzymology , Mouth Neoplasms/genetics , Polymorphism, Genetic , Prognosis
18.
Am J Chin Med ; 50(6): 1663-1679, 2022.
Article in English | MEDLINE | ID: mdl-35786173

ABSTRACT

Gambogic acid (GA), a natural and bioactive compound from the gamboge resin, has been reported to exhibit many oncostatic activities against several types of malignancies. However, its effects on the progression of oral squamous cell carcinoma (OSCC) remain largely unexplored. To fill this gap, we investigated the anticancer role of GA and molecular mechanisms underlying GA's actions in combating oral cancer. We found that GA negatively regulated the viability of OSCC cells, involving induction of the sub-G1 phase and cell apoptosis. In addition, a specific signature of apoptotic proteome, such as upregulation of heme oxygenase-1 (HO-1) and activation of caspase cascades, was identified in GA-treated OSCC. Moreover, such induction of HO-1 expression and caspase cleavage by GA was significantly diminished through the pharmacological inhibition of p38 kinase. In conclusion, these results demonstrate that GA promotes cell apoptosis in OSCC, accompanied with the activation of a p38-dependent apoptotic pathway. Our findings provide potential avenues for the use of GA with high safety and therapeutic implications in restraining oral cancer.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Apoptosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Squamous Cell Carcinoma of Head and Neck , Xanthones
19.
Curr Eye Res ; 46(11): 1681-1694, 2021 11.
Article in English | MEDLINE | ID: mdl-33979551

ABSTRACT

Purpose: Lipopolysaccharide (LPS) can activate Toll-like receptor 4 (TLR4) and increase the expression of CXCL1 and CXCL2, the potent neutrophils chemoattractants, in various cell types. These effects have not been previously reported in the uveal melanocytes. This study was designed to investigate the effects of LPS on the activation of TLR4 and expression of CXCL1/CXCL2 in cultured human uveal melanocytes and the relevant signal pathways.Methods: Effects of LPS on the expression of TLR4 were tested using real-time PCR, flow cytometry and fluorescence immunostaining. Effects of LPS-induced expression/secretion of CXCL1/CXCL2 were studied using real-time PCR in cell lysates and ELISA in conditioned media of cultured uveal melanocytes. Activated NF-κB and phosphorylated MAPK signals were tested in cells with and without LPS treatment using flow cytometry. Effects of various signal inhibitors on p38, ERK1/2, JNK1/2 and NF-κB on the secretion of CXCL1/CXCL2 were tested by ELISA. The effects of neutralized antibodies of CXCL1/CXCL2 on the severity of LPS-induced uveitis were tested in a mouse model.Results: LPS stimulation increased the expression of TLR4 mRNA and protein in culture uveal melanocytes. Constitutive secretion of CXCL1/CXCL2 was detected in uveal melanocytes and was significantly increased dose- and time-dependently by LPS stimulation. LPS mainly increased the activated NF-κB and phosphorylated JNK1/2. LPS-induced expression of CXCL1/CXCL2 was blocked by NF-κB and JNK1/2 inhibitors. The severity of LPS-induced uveitis was significantly inhibited by neutralizing antibody to CXCL1/CXCL2Conclusions: This is the first report on the LPS-induced expression of CXCL1 and CXCL2 by uveal melanocytes via the activation of TLR4. These results suggest that uveal melanocytes may play a role in the immune reaction that eliminates the invading pathogens. Conversely, an excessive LPS-induced inflammatory reaction may also lead to the development of inflammatory ocular disorders, such as non-infectious uveitis.


Subject(s)
Chemokine CXCL1/metabolism , Lipopolysaccharides/pharmacology , Melanocytes/drug effects , Toll-Like Receptor 4/metabolism , Uvea/cytology , Animals , Antibodies, Neutralizing/pharmacology , Cells, Cultured , Chemokine CXCL1/immunology , Chemokine CXCL2/immunology , Chemokine CXCL2/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , MAP Kinase Signaling System/physiology , Melanocytes/metabolism , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Protein Serine-Threonine Kinases/metabolism , Real-Time Polymerase Chain Reaction , Uveal Neoplasms/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , NF-kappaB-Inducing Kinase
20.
Biomedicines ; 9(10)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34680412

ABSTRACT

Magnolol is a natural compound extracted from Chinese herbal medicine and can induce apoptosis in numerous types of cancer cells. However, the molecular mechanisms of magnolol in oral cancer are still unclear. In this study, we investigated the anti-cancer effects and underlying mechanisms of magnolol in human oral cancer cell lines. Our results exhibited that magnolol inhibited the cell proliferation via inducing the sub-G1 phase and cell apoptosis of HSC-3 and SCC-9 cells. The human apoptosis array and Western blot assay showed that magnolol increased the expression of cleaved caspase-3 proteins and heme oxygenase-1 (HO-1). Moreover, we proved that magnolol induces apoptosis in oral cancer cell lines via the c-Jun N-terminal kinase (JNK)1/2 and p38 pathways. Overall, the current study supports the role for magnolol as a therapeutic approach for oral cancer through JNK1/2- and p38-mediated caspase activation.

SELECTION OF CITATIONS
SEARCH DETAIL