Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Int J Oral Sci ; 12(1): 3, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31911577

ABSTRACT

High-risk human papillomaviruses (HPVs) are involved in the development of several human cancers, including oropharyngeal squamous cell carcinomas. However, many studies have demonstrated that HPV alone is not sufficient for the oncogenic transformation of normal human epithelial cells, indicating that additional cofactors are required for the oncogenic conversion of HPV-infected cells. Inasmuch as chronic inflammation is also closely associated with carcinogenesis, we investigated the effect of chronic exposure to tumor necrosis factor α (TNFα), the major proinflammatory cytokine, on oncogenesis in two immortalized oral keratinocyte cell lines, namely, HPV16-immortalized and human telomerase reverse transcriptase (hTERT)-immortalized cells. TNFα treatment led to the acquisition of malignant growth properties in HPV16-immortalized cells, such as (1) calcium resistance, (2) anchorage independence, and (3) increased cell proliferation in vivo. Moreover, TNFα increased the cancer stem cell-like population and stemness phenotype in HPV16-immortalized cells. However, such transforming effects were not observed in hTERT-immortalized cells, suggesting an HPV-specific role in TNFα-promoted oncogenesis. We also generated hTERT-immortalized cells that express HPV16 E6 and E7. Chronic TNFα exposure successfully induced the malignant growth and stemness phenotype in the E6-expressing cells but not in the control and E7-expressing cells. We further demonstrated that HPV16 E6 played a key role in TNFα-induced cancer stemness via suppression of the stemness-inhibiting microRNAs miR-203 and miR-200c. Overexpression of miR-203 and miR-200c suppressed cancer stemness in TNFα-treated HPV16-immortalized cells. Overall, our study suggests that chronic inflammation promotes cancer stemness in HPV-infected cells, thereby promoting HPV-associated oral carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/genetics , Human papillomavirus 16/metabolism , MicroRNAs/metabolism , Mouth Neoplasms/genetics , Mouth/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/virology , Telomerase/genetics , Tumor Necrosis Factor-alpha/metabolism , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinoma, Squamous Cell/pathology , Cell Transformation, Viral/genetics , Gene Expression Regulation , Genes, Viral , Human papillomavirus 16/genetics , Humans , MicroRNAs/genetics , Mouth/virology , Mouth Neoplasms/pathology , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Telomerase/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
2.
Biochem Biophys Res Commun ; 424(1): 58-64, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22728043

ABSTRACT

Cancer stem-like cell (CSC; also known as tumor initiating cell) is defined as a small subpopulation of cancer cells within a tumor and isolated from various primary tumors and cancer cell lines. CSCs are highly tumorigenic and resistant to anticancer treatments. In this study, we found that prolonged exposure to tumor necrosis factor alpha (TNFα), a major proinflammatory cytokine, enhances CSC phenotype of oral squamous cell carcinoma (OSCC) cells, such as an increase in tumor sphere-forming ability, stem cell-associated genes expression, chemo-radioresistance, and tumorigenicity. Moreover, activation of Notch1 signaling was detected in the TNFα-exposed cells, and suppression of Notch1 signaling inhibited CSC phenotype. Furthermore, we demonstrated that inhibition of a Notch downstream target, Hes1, led to suppression of CSC phenotype in the TNFα-exposed cells. We also found that Hes1 expression is commonly upregulated in OSCC lesions compared to precancerous dysplastic lesions, suggesting the possible involvement of Hes1 in OSCC progression and CSC in vivo. In conclusion, inflammatory cytokine exposure may enhance CSC phenotype of OSCC, in part by activating the Notch-Hes1 pathway.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Squamous Cell/pathology , Homeodomain Proteins/metabolism , Mouth Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Receptor, Notch1/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/chemically induced , Homeodomain Proteins/genetics , Humans , Mouth Neoplasms/metabolism , Neoplastic Stem Cells/pathology , Receptor, Notch1/genetics , Transcription Factor HES-1
3.
Biochem Biophys Res Commun ; 404(4): 896-902, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21167132

ABSTRACT

MicroRNAs (miRNAs) are epigenetic regulators of gene expression, and their deregulation plays an important role in human cancer, including oral squamous cell carcinoma (OSCC). Recently, we found that miRNA-181a (miR-181a) was upregulated during replicative senescence of normal human oral keratinocytes. Since senescence is considered as a tumor suppressive mechanism, we thus investigated the expression and biological role of miR-181a in OSCC. We found that miR-181a was frequently downregulated in OSCC. Ectopic expression of miR-181a suppressed proliferation and anchorage independent growth ability of OSCC. Moreover, miR-181a dramatically reduces the growth of OSCC on three dimensional organotypic raft culture. We also identified K-ras as a novel target of miR-181a. miR-181a decreased K-ras protein level as well as the luciferase activity of reporter vectors containing the 3'-untranslated region of K-ras gene. Finally, we defined a minimal regulatory region of miR-181a and found a positive correlation between its promoter activity and the level of miR-181a expression. In conclusion, miR-181a may function as an OSCC suppressor by targeting on K-ras oncogene. Thus, miR-181a should be considered for therapeutic application for OSCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Genes, ras , MicroRNAs/physiology , Mouth Neoplasms/pathology , Base Sequence , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Humans , MicroRNAs/genetics , Mouth Neoplasms/genetics , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...