Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Comput Struct Biotechnol J ; 17: 537-560, 2019.
Article in English | MEDLINE | ID: mdl-31049165

ABSTRACT

The sphingolipid and lysophosphatidate regulatory networks impact diverse mechanisms attributed to cancer cells and the tumor immune microenvironment. Deciphering the complexity demands implementation of a holistic approach combined with higher-resolution techniques. We implemented a multi-modular integrative approach consolidating the latest accomplishments in gene expression profiling, prognostic/predictive modeling, next generation digital pathology, and systems biology for epithelial ovarian cancer. We assessed patient-specific transcriptional profiles using the sphingolipid/lysophosphatidate/immune-associated signature. This revealed novel sphingolipid/lysophosphatidate-immune gene-gene associations and distinguished tumor subtypes with immune high/low context. These were characterized by robust differences in sphingolipid-/lysophosphatidate-related checkpoints and the drug response. The analysis also nominates novel survival models for stratification of patients with CD68, LPAR3, SMPD1, PPAP2B, and SMPD2 emerging as the most prognostically important genes. Alignment of proprietary data with curated transcriptomic data from public databases across a variety of malignancies (over 600 categories; over 21,000 arrays) showed specificity for ovarian carcinoma. Our systems approach identified novel sphingolipid-lysophosphatidate-immune checkpoints and networks underlying tumor immune heterogeneity and disease outcomes. This holds great promise for delivering novel stratifying and targeting strategies.

2.
BMC Genomics ; 17(1): 643, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27527602

ABSTRACT

BACKGROUND: Building up of pathway-/disease-relevant signatures provides a persuasive tool for understanding the functional relevance of gene alterations and gene network associations in multifactorial human diseases. Ovarian cancer is a highly complex heterogeneous malignancy in respect of tumor anatomy, tumor microenvironment including pro-/antitumor immunity and inflammation; still, it is generally treated as single disease. Thus, further approaches to investigate novel aspects of ovarian cancer pathogenesis aiming to provide a personalized strategy to clinical decision making are of high priority. Herein we assessed the contribution of the AID/APOBEC family and their associated genes given the remarkable ability of AID and APOBECs to edit DNA/RNA, and as such, providing tools for genetic and epigenetic alterations potentially leading to reprogramming of tumor cells, stroma and immune cells. RESULTS: We structured the study by three consecutive analytical modules, which include the multigene-based expression profiling in a cohort of patients with primary serous ovarian cancer using a self-created AID/APOBEC-associated gene signature, building up of multivariable survival models with high predictive accuracy and nomination of top-ranked candidate/target genes according to their prognostic impact, and systems biology-based reconstruction of the AID/APOBEC-driven disease-relevant mechanisms using transcriptomics data from ovarian cancer samples. We demonstrated that inclusion of the AID/APOBEC signature-based variables significantly improves the clinicopathological variables-based survival prognostication allowing significant patient stratification. Furthermore, several of the profiling-derived variables such as ID3, PTPRC/CD45, AID, APOBEC3G, and ID2 exceed the prognostic impact of some clinicopathological variables. We next extended the signature-/modeling-based knowledge by extracting top genes co-regulated with target molecules in ovarian cancer tissues and dissected potential networks/pathways/regulators contributing to pathomechanisms. We thereby revealed that the AID/APOBEC-related network in ovarian cancer is particularly associated with remodeling/fibrotic pathways, altered immune response, and autoimmune disorders with inflammatory background. CONCLUSIONS: The herein study is, to our knowledge, the first one linking expression of entire AID/APOBECs and interacting genes with clinical outcome with respect to survival of cancer patients. Overall, data propose a novel AID/APOBEC-derived survival model for patient risk assessment and reconstitute mapping to molecular pathways. The established study algorithm can be applied further for any biologically relevant signature and any type of diseased tissue.


Subject(s)
APOBEC Deaminases/genetics , APOBEC Deaminases/metabolism , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Signal Transduction , Adult , Aged , Aged, 80 and over , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Combined Modality Therapy , Computational Biology/methods , Datasets as Topic , Female , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Middle Aged , Molecular Sequence Annotation , Multigene Family , Neoplasm Grading , Neoplasm Staging , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , Prognosis , Proportional Hazards Models
3.
Eur J Cancer ; 50(1): 99-110, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24176298

ABSTRACT

Cyclin E, coded by the genes CCNE1 and CCNE2, is the main regulator for transition from G1 to S phase determining cell division. CCNE1 and CCNE2 are known oncogenes in many cancer entities. Especially CCNE1 has frequently been associated with gene amplifications in various malignancies, emphasising its role as a putative oncogene. We determined gene expression and copy number of CCNE1 and CCNE2 by quantitative polymerase chain reaction (PCR) from 172 International Federation of Obstetrics and Gynecology (FIGO) II/III/IV stage serous epithelial ovarian cancer (EOC) tissues and analysed its impact on outcome. Furthermore, whole transcriptome gene expression changes correlating with CCNE1 expression were determined by microarray technology, interpreted by Signalling Pathway Impact Analysis (SPIA), Tool for Inferring Network of Genes (TINGe), and illustrated by hive plots. Protein-protein interaction (PPI) networks were also used for the interpretation. Interestingly, and contradictory to most reports and intuitive expectations, high CCNE1 expression correlated with better overall survival (p=0.005) if corrected for usual clinicopathologic parameters and a molecular subclassification. Using different grading systems or only high graded tumours had no impact on this correlation. Copy number of CCNE1 was increased in 25% of cases which correlated highly significantly with expression but showed no impact on outcome. CCNE2 had no impact on outcomes at all. Whole genome transcriptome analysis revealed 1872 differentially expressed genes correlated to CCNE1 expression, which were significantly enriched with genes from five pathways (e.g. cell cycle and viral carcinogenesis pathway were up-regulated and the Fanconi anaemia pathway was down-regulated). High CCNE1 gene expression is a significant and independent predictor for prolonged overall survival in FIGO III/IV EOC patients. This upside down impact of CCNE1 on survival probably reflects the special characteristic of EOC with tumour dissemination in the near anaerobic peritoneal cavity as the predominant cause of death, compared to other cancer entities where distant metastasis are predominantly lethal.


Subject(s)
Biomarkers, Tumor/genetics , Cyclin E/genetics , Neoplasms, Glandular and Epithelial/genetics , Oncogene Proteins/genetics , Ovarian Neoplasms/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial , Cell Proliferation , Cyclin E/metabolism , Cyclins/genetics , Cyclins/metabolism , Female , Gene Amplification , Gene Dosage , Gene Expression , Humans , Middle Aged , Neoplasms, Glandular and Epithelial/metabolism , Oncogene Proteins/metabolism , Ovarian Neoplasms/metabolism , Prognosis , Signal Transduction
4.
Cytotherapy ; 15(11): 1426-35, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24094492

ABSTRACT

BACKGROUND AIMS: Human endothelial progenitor cells (EPC) play an important role in regenerative medicine and contribute to neovascularization on vessel injury. They are usually enriched from peripheral blood, cord blood and bone marrow. In human fat tissue, EPC are rare and their isolation remains a challenge. METHODS: Fat tissue was prepared by collagenase digestion, and the expression of specific marker proteins was evaluated by flow cytometry in the stromal vascular fraction (SVF). For enrichment, magnetic cell sorting was performed with the use of CD133 microbeads and EPC were cultured until colonies appeared. A second purification was performed with CD34; additional isolation steps were performed with the use of a combination of CD34 and CD31 microbeads. Enriched cells were investigated by flow cytometry for the expression of endothelial specific markers, by Matrigel assay and by the uptake of acetylated low-density lipoprotein. RESULTS: The expression pattern confirmed the heterogeneous nature of the SVF, with rare numbers of CD133+ detectable. EPC gained from the SVF by magnetic enrichment showed cobblestone morphology of outgrowth endothelial cells and expressed the specific markers CD31, CD144, vascular endothelial growth factor (VEGF)R2, CD146, CD73 and CD105. Functional integrity was confirmed by uptake of acetylated low-density lipoprotein and the formation of tube-like structures on Matrigel. CONCLUSIONS: Rare EPC can be enriched from human fat tissue by magnetic cell sorting with the use of a combination of microbeads directed against CD133, an early EPC marker, CD34, a stem cell marker, and CD31, a typical marker for endothelial cells. In culture, they differentiate into EC and hence could have the potential to contribute to neovascularization in regenerative medicine.


Subject(s)
Antigens, CD34/immunology , Antigens, CD/immunology , Endothelial Cells/cytology , Glycoproteins/immunology , Peptides/immunology , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Stem Cells/cytology , 5'-Nucleotidase/biosynthesis , AC133 Antigen , Adipose Tissue/cytology , Antigens, CD/biosynthesis , CD146 Antigen/biosynthesis , Cadherins/biosynthesis , Cell Differentiation/immunology , Cells, Cultured , Endoglin , Endothelial Cells/immunology , GPI-Linked Proteins/biosynthesis , Humans , Lipectomy , Microspheres , Neovascularization, Physiologic/physiology , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Receptors, Cell Surface/biosynthesis , Stem Cells/immunology , Vascular Endothelial Growth Factor Receptor-2/biosynthesis
5.
BMC Cancer ; 13: 178, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23551967

ABSTRACT

BACKGROUND: The immune system is a key player in fighting cancer. Thus, we sought to identify a molecular 'immune response signature' indicating the presence of epithelial ovarian cancer (EOC) and to combine this with a serum protein biomarker panel to increase the specificity and sensitivity for earlier detection of EOC. METHODS: Comparing the expression of 32,000 genes in a leukocytes fraction from 44 EOC patients and 19 controls, three uncorrelated shrunken centroid models were selected, comprised of 7, 14, and 6 genes. A second selection step using RT-qPCR data and significance analysis of microarrays yielded 13 genes (AP2A1, B4GALT1, C1orf63, CCR2, CFP, DIS3, NEAT1, NOXA1, OSM, PAPOLG, PRIC285, ZNF419, and BC037918) which were finally used in 343 samples (90 healthy, six cystadenoma, eight low malignant potential tumor, 19 FIGO I/II, and 220 FIGO III/IV EOC patients). Using new 65 controls and 224 EOC patients (thereof 14 FIGO I/II) the abundances of six plasma proteins (MIF, prolactin, CA125, leptin, osteopondin, and IGF2) was determined and used in combination with the expression values from the 13 genes for diagnosis of EOC. RESULTS: Combined diagnostic models using either each five gene expression and plasma protein abundance values or 13 gene expression and six plasma protein abundance values can discriminate controls from patients with EOC with Receiver Operator Characteristics Area Under the Curve values of 0.998 and bootstrap .632+ validated classification errors of 3.1% and 2.8%, respectively. The sensitivities were 97.8% and 95.6%, respectively, at a set specificity of 99.6%. CONCLUSIONS: The combination of gene expression and plasma protein based blood derived biomarkers in one diagnostic model increases the sensitivity and the specificity significantly. Such a diagnostic test may allow earlier diagnosis of epithelial ovarian cancer.


Subject(s)
Biomarkers, Tumor/blood , Blood Proteins/genetics , Cystadenoma/blood , Cystadenoma/genetics , Gene Expression Profiling , Neoplasms, Glandular and Epithelial/blood , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/blood , Ovarian Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Analysis of Variance , Area Under Curve , CA-125 Antigen/blood , Carcinoma, Ovarian Epithelial , Case-Control Studies , Female , Humans , Insulin-Like Growth Factor II/metabolism , Intramolecular Oxidoreductases/blood , Leptin/blood , Macrophage Migration-Inhibitory Factors/blood , Membrane Proteins/blood , Middle Aged , Neoplasms, Glandular and Epithelial/pathology , Oligonucleotide Array Sequence Analysis , Osteopontin/blood , Ovarian Neoplasms/pathology , Prolactin/blood , ROC Curve , Retrospective Studies , Young Adult
6.
Cancer Sci ; 103(7): 1334-41, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22497737

ABSTRACT

Most patients with epithelial ovarian cancer (EOC) are diagnosed at advanced stage and have a poor prognosis. However, a small proportion of these patients will survive, whereas others will die very quickly. Clinicopathological factors do not allow precise identification of these subgroups. Thus, we have validated a molecular subclassification as new prognostic factor in EOC. One hundred and ninety-four patients with Stage II-IV EOC were characterized by whole-genome expression profiling of tumor tissues and were classified using a published 112 gene set, derived from an International Federation of Gynecology and Obstetrics (FIGO) stage-directed supervised classification approach. The 194 tumor samples were classified into two subclasses comprising 95 (Subclass 1) and 99 (Subclass 2) tumors. All nine FIGO II tumors were grouped in Subclass 1 (P = 0.001). Subclass 2 (54% of advanced-stage tumors) was significantly correlated with peritoneal carcinomatosis and non-optimal debulking. Patients with Subclass 2 tumors had a worse overall survival for both serous and non-serous histological subtypes, as revealed by univariate analysis (hazard ratios [HR] of 3.17 and 17.11, respectively; P ≤ 0.001) and in models corrected for relevant clinicopathologic parameters (HR 2.87 and 12.42, respectively; P ≤ 0.023). Significance analysis of microarrays revealed 2082 genes that were differentially expressed in advanced-grade serous tumors of both subclasses and the focal adhesion pathway as the most deregulated pathway. In the present validation study, we have shown that, in advanced-stage serous ovarian cancer, two approximately equally large molecular subtypes exist, independent of classical clinocopathological parameters and presenting with highly different whole-genome expression profiles and a markedly different overall survival. Similar results were obtained in a small cohort of patients with non-serous tumors.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Adult , European Union , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Multivariate Analysis , Neoplasm Grading , Neoplasm Staging , Neoplasms, Glandular and Epithelial/classification , Neoplasms, Glandular and Epithelial/pathology , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Ovarian Neoplasms/classification , Ovarian Neoplasms/pathology , Prognosis , Proportional Hazards Models
7.
Cancer Lett ; 293(1): 117-23, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20167419

ABSTRACT

Comprehensive in vitro and in vivo studies comparing EpCAM-based methods with other cytometric CTC enrichment technologies in metastatic colorectal cancer (mCRC) patients are lacking. We compare four manual cytometric methods to detect CTCs in vitro and in mCRC patients. The EpCAM-based technology, MACS HEA MicroBeads((R)), showed a significant better tumor cell recovery rate compared to other cytometric methods (p-value<0.0001). CTCs of 38 mCRC patients were enriched with MACS HEA MicroBeads(R). Progression-free survival did significantly differ between mCRC patients without detectable and with >or= 1 CTCs (p=0.007). CTC enrichment with EpCAM coupled antibodies is superior to other cytometric methods and is a feasible method for CTC detection in mCRC patients.


Subject(s)
Cell Separation/methods , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Centrifugation, Density Gradient/methods , Disease-Free Survival , Female , Humans , Immunomagnetic Separation/methods , Male , Middle Aged , Neoplasm Metastasis
8.
J Oral Pathol Med ; 35(8): 472-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16918598

ABSTRACT

BACKGROUND: 1Alpha,25-dihydroxyvitamin D(3) [1,25(OH)(2) Vitamin D(3)] induces growth inhibition in squamous cell carcinoma (SCC) cell lines of the head and neck by arresting the cells in the G0/G1 phase of the cell cycle, probably due to an enhanced expression of p21, which could be demonstrated in other cell lines (JPPA, SCC9) before. In SCC25, a SCC cell line isolated from tongue, growth inhibition but no overexpression of p21 was detected. The retinoblastoma gene, as a direct target of G1 cyclin-CDK complexes, showed an obvious shift from the hyperphosphorylated to the hypophosphorylated form under 1,25(OH)(2)Vitamin D(3), which indicates that the growth inhibition takes place in the G0/G1 phase. To explore the possible pathway of growth inhibition in SCC25 we investigated other cell cycle inhibitors (p18, p19, p27). METHODS: Synchronized cells were treated with 1,25(OH)(2)Vitamin D(3) over 96 h. The cell cycle status and expression of cell cycle-regulating proteins was determined by fluorescence-activated cell sorting (FACS) and Western blotting. An overexpression of p18 in 1,25(OH)(2)Vitamin D(3) vs. ethanol-treated cells was determined until 30 h in SCC25. No influence was detectable on the expression of p27 and p19. CONCLUSION: One mechanism by which 1,25(OH)(2)Vitamin D(3) controls cell growth might be the upregulation of p21. As p21 was unsusceptible to 1,25(OH)(2)Vitamin D(3) in SCC25, other inhibiting proteins were necessary to be tested. The proven upregulation of p18 seems to be the responsible step for growth inhibition of 1,25(OH)(2)Vitamin D(3) in SCC25.


Subject(s)
Calcitriol/pharmacology , Carcinoma, Squamous Cell/metabolism , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Head and Neck Neoplasms/metabolism , Neoplasm Proteins/metabolism , Animals , Carcinoma, Squamous Cell/pathology , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , G1 Phase/physiology , Head and Neck Neoplasms/pathology , Humans , Rabbits , Resting Phase, Cell Cycle/physiology , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology
9.
Gynecol Oncol ; 98(2): 211-6, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15967487

ABSTRACT

OBJECTIVE: Metastatic relapse due to early dissemination of tumor cells is associated with poor prognosis for epithelial cancer. The molecular characterization of these single cells or cell clusters that have evaded the tumor is indispensable in order to evaluate their biological behavior and metastatic potential. In this study, we established a sensitive immunomagnetic method to isolate rare cancer cells from peripheral blood based on their expression of epithelial- or tumor-cell-specific markers. METHODS: Low numbers of cells of breast cancer cell lines - ZR-75-1, MCF-7, HBL-100 - were spiked into peripheral blood specimens of healthy volunteers. Enrichment of tumor cells was performed using either pre-coupled HEA and/or ErbB2 microbeads or a mixture of three monoclonal antibodies against HEA, ErbB2 and EGFR. RESULTS: The recovery rate of spiked tumor cells correlated with the expression of the corresponding antigens. ZR-75-1 cells high expressing all three genes could be isolated to 60-71%. MCF-7 cells, which hardly express EGFR, showed a significant better recovery by using two specific antibodies in combination (50-68%) than one pre-coupled bead alone (31-42%). HBL-100 cells little expressing HEA could not be isolated with HEA microbeads and only to 27% in combination with ErbB2 beads -in contrast the use of an antibody cocktail achieved 38%. CONCLUSION: As tumor and epithelial specific cell marker antigens are expressed differently in disseminated tumor cells, the immunomagnetic enrichment from peripheral blood is most robust and reliable when using a combination of specific antibodies compared to single antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Breast Neoplasms/blood , Immunomagnetic Separation/methods , Neoplastic Cells, Circulating/pathology , Adult , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , ErbB Receptors/biosynthesis , ErbB Receptors/immunology , Humans , Microspheres , Neoplastic Cells, Circulating/immunology , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/immunology , Reproducibility of Results , Sensitivity and Specificity
10.
Acta Otolaryngol ; 124(1): 90-6, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14977084

ABSTRACT

OBJECTIVE: The biologically active 1,25(OH)2 vitamin D3 and its analogs have been shown to have antiproliferative and differentiating effects in a variety of malignant and non-malignant cells. For squamous carcinoma cell lines of the head and neck (SCCHN) we could show that this antiproliferative activity of 1,25(OH)2 vitamin D3 is due to induced expression of the cell-cycle inhibitory proteins p21 and p27, causing an arrest in the G0/G1 cell-cycle phase. MATERIAL AND METHODS: In this work we investigated the effects of three vitamin D3 analogs, EB1089, MC1288 and CB1093, on proliferation behavior and cell-cycle status in a laryngeal carcinoma cell line (JPPA) as well as in control human immortalized keratinocytes (HaCaT). To study the molecular mechanism the functional activity of the promoter region of p21, a potential target gene of vitamin D3 transcriptional regulation, was investigated. For this reason a 2.7-kb fragment of the p21 promoter was isolated by polymerase chain reaction from HaCaT, JPPA and SCC9 (tongue carcinoma) cells and directionally cloned into an enhanced green fluorescence protein (EGFP) reporter gene vector system. A construct was used to stably transfect HaCaT cells and to monitor the expression of the EGFP gene by confocal microscopy. RESULTS: Analysis of proliferation and cell-cycle status revealed decreased growth rates and G0/G1I cell-cycle arrest in cells treated with 1,25(OH)2 vitamin D3 and its analogs The EGFP reporter gene-transfected cells showed distinct fluorescence under the influence of 1,25(OH)2 vitamin D3 and its analogs compared to control cells. CONCLUSION: These results demonstrate that the cell-cycle inhibitor protein p21 is a direct target gene of biologically active 1,25(OH)2 vitamin D3, inducing G0/G1 cell-cycle arrest. The ability of vitamin D analogs to act via the same molecular mechanism as the natural hormone but with less hypercalcemic activity may have therapeutic implications for patients with SCCHN malignancy.


Subject(s)
Antineoplastic Agents/pharmacology , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Carcinoma, Squamous Cell/genetics , Cell Cycle/genetics , Cell Division/genetics , Laryngeal Neoplasms/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Cells, Cultured/drug effects , Calcitriol/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Cell Division/drug effects , Cell Survival/drug effects , G1 Phase/drug effects , G1 Phase/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter/genetics , Green Fluorescent Proteins , Humans , Laryngeal Neoplasms/pathology , Luminescent Proteins/genetics , Promoter Regions, Genetic/drug effects , Resting Phase, Cell Cycle/drug effects , Resting Phase, Cell Cycle/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transfection , Tumor Cells, Cultured/pathology
11.
Acta Otolaryngol ; 122(3): 338-42, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12030586

ABSTRACT

Epidemiologic studies have provided evidence of an alcohol-associated increased risk of upper aerodigestive tract cancers. Recently we reported ethanol-induced proliferation in a squamous cell carcinoma of the head and neck (SCCHN) cell line, but the underlying mechanisms are unknown. In order to further clarify these findings, major G0/G1-regulating proteins were investigated in the present study. Synchronized cells of a SCCHN line (JP-PA) and a human immortalized keratinocyte line (HaCaT)-used as a control-were cultured with or without 10(-3) M ethanol for up to 96 h. At distinct time intervals the expression of cyclin D1 and the inhibitors p16, p18, p19 and p21 were determined by Western blot analyses. In both lines ethanol had no influence on the protein expression of cyclin D1. In contrast, distinct downregulations of p21, p18 and p19 were detectable at the protein level. The p16 protein was not expressed in the SCCHN line and was unchanged in the control line after the addition of ethanol. In these in vitro experiments the marked downregulation of important cell-cycle inhibitors may accelerate progression from the G1 to the S phase of the cell cycle. The relevance of our findings to in vivo conditions remains speculative, but the observed mechanisms of significantly reduced expression of cell-cycle inhibitor proteins may be involved in the carcinogenesis of head and neck malignancies.


Subject(s)
Carcinoma, Squamous Cell/pathology , Ethanol/pharmacology , Head and Neck Neoplasms/pathology , Blotting, Western , Cell Cycle , Cell Line , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Down-Regulation , Enzyme Inhibitors/metabolism , Flow Cytometry , Humans , In Vitro Techniques , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...