Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Cells ; 12(18)2023 09 07.
Article in English | MEDLINE | ID: mdl-37759448

ABSTRACT

Emerging evidence indicates that intracellular calcium (Ca2+) levels and their regulatory proteins play essential roles in normal stem cell proliferation and differentiation. Cancer stem-like cells (CSCs) are subpopulations of cancer cells that retain characteristics similar to stem cells and play an essential role in cancer progression. Recent studies have reported that the Orai3 calcium channel plays an oncogenic role in human cancer. However, its role in CSCs remains underexplored. In this study, we explored the effects of Orai3 in the progression and stemness of oral/oropharyngeal squamous cell carcinoma (OSCC). During the course of OSCC progression, the expression of Orai3 exhibited a stepwise augmentation. Notably, Orai3 was highly enriched in CSC populations of OSCC. Ectopic Orai3 expression in non-tumorigenic immortalized oral epithelial cells increased the intracellular Ca2+ levels, acquiring malignant growth and CSC properties. Conversely, silencing of the endogenous Orai3 in OSCC cells suppressed the CSC phenotype, indicating a pivotal role of Orai3 in CSC regulation. Moreover, Orai3 markedly increased the expression of inhibitor of DNA binding 1 (ID1), a stemness transcription factor. Orai3 and ID1 exhibited elevated expression within CSCs compared to their non-CSC counterparts, implying the functional importance of the Orai3/ID1 axis in CSC regulation. Furthermore, suppression of ID1 abrogated the CSC phenotype in the cell with ectopic Orai3 overexpression and OSCC. Our study reveals that Orai3 is a novel functional CSC regulator in OSCC and further suggests that Orai3 plays an oncogenic role in OSCC by promoting cancer stemness via ID1 upregulation.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Oropharyngeal Neoplasms , Humans , Mouth Neoplasms/genetics , Squamous Cell Carcinoma of Head and Neck , Calcium Channels , Hyperplasia , Inhibitor of Differentiation Protein 1
2.
Exp Cell Res ; 405(1): 112656, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34033760

ABSTRACT

DYRK1A, one of the dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs), plays an important role in various biological processes by regulating downstream targets via kinase-dependent and independent mechanisms. Here, we report a novel role of DYRK1A in maintaining tumor growth and stemness of oral/oropharyngeal squamous cell carcinoma (OSCC) cells. Deletion of DYRK1A from OSCC cells abrogated their in vivo tumorigenicity and self-renewal capacity, the key features of cancer stem-like cells (CSCs; also referred to as tumor-initiating cells). The DYRK1A deletion also induced the suppression of CSC populations and properties, such as migration ability and chemoresistance. Conversely, ectopic expression of DYRK1A in OSCC cells augmented their CSC phenotype. Among five DYRK members (DYRK1A, 1B, 2, 3, and 4), DYRK1A is the most dominantly expressed kinase, and its expression is upregulated in OSCC compared to normal oral epithelial cells. More importantly, DYRK1A was highly enriched in various CSC-enriched OSCC populations compared to their corresponding non-CSC populations, indicating its pivotal role in cancer progression and stemness. Further, our study revealed that fibroblast growth factor 2 (FGF2) is a key regulator in the DYRK1A-mediated CSC regulation. Functional studies demonstrated that the loss of DYRK1A inhibits CSC phenotype via reduction of FGF2. Overexpression of DYRK1A promotes CSC phenotype via upregulation of FGF2. Our study delineates a novel mechanism of cancer stemness regulation by DYRK1A-FGF2 axis in OSCC. Thus, inhibition of DYRK1A would lead to a potential novel therapeutic option for targeting CSCs in OSCC.


Subject(s)
Carcinogenesis/pathology , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/pathology , Neoplastic Stem Cells/pathology , Oropharyngeal Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Proliferation , Humans , Mice , Mice, Nude , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Dyrk Kinases
3.
Oncol Rep ; 45(1): 291-298, 2021 01.
Article in English | MEDLINE | ID: mdl-33155662

ABSTRACT

Cancer stem­like cells (CSCs; also referred to as tumor­initiating cells) play crucial roles in tumor progression and aggressiveness. Recent studies have demonstrated the antitumor activity of zoledronic acid (ZA), a third­generation bisphosphonate, in various types of human cancer. However, its effect on oral CSCs and the underlying mechanism remain obscure. The present study demonstrated that ZA suppresses the growth and stemness properties of oral/oropharyngeal squamous cell carcinoma (OSCC) cells. ZA inhibited the malignant characteristics of OSCC cells, such as anchorage­independent growth and epithelial thickening in organotypic raft cultures. Moreover, ZA treatment resulted in suppression of self­renewal capacity, a key feature of CSCs. ZA also inhibited important CSC properties, such as migration and chemo­radioresistance. Mechanistically, ZA exposure significantly decreased chemokine (C­C motif) ligand 3 (CCL3) expression in OSCC cells. It was further demonstrated that CCL3 signaling via its receptor is crucial for supporting the CSC phenotype in OSCC cells. Moreover, an antagonist of the CCL3 receptor reversed the effect of CCL3 on CSC properties, and exogenous CCL3 rescued the suppressaed CSC phenotype in ZA­treated OSCC cells. These results demonstrated that ZA suppresses the CSC phenotype in OSCC cells by reducing CCL3 expression, suggesting that ZA may be an effective therapeutic agent for oral cancer by targeting CSCs.


Subject(s)
Chemokine CCL3/physiology , Mouth Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Squamous Cell Carcinoma of Head and Neck/drug therapy , Zoledronic Acid/pharmacology , Cell Line, Tumor , Chemokine CCL3/analysis , Humans , Mouth Neoplasms/immunology , Mouth Neoplasms/pathology , Neoplastic Stem Cells/chemistry , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology
4.
Oncotarget ; 10(23): 2306-2319, 2019 Mar 19.
Article in English | MEDLINE | ID: mdl-31040921

ABSTRACT

Nuclear factor of activated T cells (NFATc1-c4), a family of transcription factors, is involved in many biological processes by regulating various downstream target genes. However, their role in cancer progression remains controversial. We here report that NFATc3 is the dominant isoform of NFAT in human oral epithelial cells, and its expression was increased in a stepwise manner during the progression of oral/oropharyngeal squamous cell carcinoma (OSCC). More importantly, NFATc3 was highly enriched in self-renewing cancer stem-like cells (CSCs) of OSCC. Increased expression of NFATc3 was required for the maintenance of CSC self-renewal, as NFATc3 inhibition suppressed tumor sphere formation in OSCC cells. Conversely, ectopic NFATc3 expression in non-tumorigenic immortalized oral epithelial cells resulted in the acquisition of self-renewal and increase in CSC phenotype, such as enhanced ALDH1HIGH cell population, mobility and drug resistance, indicating the functional role of NFATc3 in the maintenance of CSC phenotype. NFATc3 expression also converted the non-tumorigenic oral epithelial cells to malignant phenotypes. Mechanistic investigations further reveal that NFATc3 binds to the promoter of OCT4, a stemness transcription factor, for its activation, thereby promoting CSC phenotype. Moreover, suppression of OCT4 abrogated CSC phenotype in the cell with ectopic NFATc3 overexpression and OSCC, and ectopic OCT4 expression sufficiently induced CSC phenotype. Our study indicates that NFATc3 plays an important role in the maintenance of cancer stemness and OSCC progression via novel NFATc3-OCT4 axis, suggesting that this axis may be a potential therapeutic target for OSCC CSCs.

5.
J Endod ; 43(8): 1302-1308, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28606672

ABSTRACT

INTRODUCTION: Mesenchymal stem cells (MSCs) are typically cultured as adherent monolayer using a conventional tissue culture technique. However, this technique incompletely reproduces an in vivo microenvironment of stem cells and results in the loss of stemness properties. Three-dimensional (3D) sphere culture is one of the most widely used 3D culture techniques that have been developed to recapitulate the in vivo microenvironment. However, the stemness and multilineage differentiation capacity of spheres derived from dental pulp stem cells (DPSCs) have not been well investigated. METHODS: DPSCs were cultured and examined for the sphere-forming ability in serum-free, nonadherent conditions. The expression of pluripotency transcription factors was assayed by reverse transcription quantitative polymerase chain reaction and Western blot analysis. The expression of MSC-associated markers was determined by flow cytometry. Multilineage differentiation capacity was examined by alkaline phosphatase, alizarin red S, and oil red O assays. Subcutaneous transplantation in nude mice was used to examine the in vivo mineralized tissue-forming ability of sphere and adherent monolayer cells derived from DPSCs. RESULTS: We showed that DPSCs form spheres. DPSC spheres exhibited a distinct stem cell phenotype characterized by robust expression of pluripotency transcription factors and decreased expression of MSC-associated markers compared with their corresponding adherent monolayer cells. Functionally, DPSC spheres exhibited enhanced in vitro multilineage differentiation capacity. The expression of multilineage differentiation-related genes was also highly increased in DPSC spheres. Furthermore, DPSC sphere cells possessed higher in vivo mineralized tissue-forming ability than adherent monolayer cells. CONCLUSIONS: Our findings indicate that sphere-forming cells are unique multipotent cell populations in DPSCs. Our study further suggests that DPSC spheres may provide a unique opportunity for pulp tissue regeneration.


Subject(s)
Dental Pulp/cytology , Multipotent Stem Cells/cytology , Animals , Blotting, Western , Cell Differentiation/physiology , Cells, Cultured , Dental Pulp/metabolism , Humans , Mice , Mice, Nude , Multipotent Stem Cells/metabolism , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL