Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Strahlenther Onkol ; 197(3): 231-245, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32588101

ABSTRACT

PURPOSE: Cancer stem cells (CSCs) are held accountable for the progress of head and neck squamous cell carcinoma (HNSCC). In the presented study, the authors evaluated the prognostic value of CSC markers in two particular HNSCC cohorts. METHODS: This two cohort study consisted of 85 patients with advanced stage HNSCC, treated with primary radio(chemo)therapy (pRCT), and 95 patients with HNSCC, treated with surgery and partially adjuvant radio(chemo)therapy. Overall survival (OS), disease-free survival (DFS), and disease-specific survival (DSS) were assessed. Samples were assessed for the expression of different molecular stem cell markers (ALDH1, BCL11B, BMI­1, and CD44). RESULTS: In the pRCT cohort, none of the baseline patient and tumor features exhibited a statistically significant relation with survival in either the cohort or the human papillomavirus (HPV)-stratified subcohorts. High expression of BMI­1 significantly decreased OS and DFS, while high expression of CD44 decreased all modes of survival. Multivariate analysis showed significant prognostic influence for all tested CSC markers, with high BMI­1 and CD44 decreasing survival (BMI-1: OS, DFS, DSS; CD44: OS, DFS) and high ALDH1 and BCL11B showing a beneficial effect on survival (ALDH1: OS, DFS; BCL11B: OS, DSS). In the surgical cohort, classical prognosticators such as HPV status, R1 resection, and nodal status in HPV-negative HNSCC played a significant role, but the tested CSC markers showed no significant effect on prognosis. CONCLUSION: Although validation in independent cohorts is still needed, testing for CSC markers in patients with advanced or late stage HNSCC might be beneficial, especially if many comorbidities exist or disease is irresectable. The findings might guide the development and earlier use of targeted therapies in the future.


Subject(s)
Aldehyde Dehydrogenase 1 Family/analysis , Head and Neck Neoplasms/diagnosis , Hyaluronan Receptors/analysis , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/analysis , Repressor Proteins/analysis , Squamous Cell Carcinoma of Head and Neck/diagnosis , Tumor Suppressor Proteins/analysis , Aged , Female , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/pathology , Survival Analysis
2.
J Appl Oral Sci ; 28: e20190532, 2020.
Article in English | MEDLINE | ID: mdl-32348447

ABSTRACT

Oral leukoplakia (OL) is a white lesion of an indeterminate risk not related to any excluded (other) known diseases or disorders that carry no increased risk for cancer. Many biological markers have been used in an attempt to predict malignant transformation; however, no reliable markers have been established so far. Objective To evaluate cell proliferation and immortalization in OL, comparing non-dysplastic (Non-dys OL) and dysplastic OL (Dys OL). Methodology This is a cross-sectional observational study. Paraffin-embedded tissue blocks of 28 specimens of Non-dys OL, 33 of Dys OL, 9 of normal oral mucosa (NOM), 17 of inflammatory hyperplasia (IH), and 19 of oral squamous cell carcinomas (OSCC) were stained for Ki-67 and BMI-1 using immunohistochemistry. Results A gradual increase in BMI-1 and K-i67 expression was found in oral carcinogenesis. The immunolabeling for those markers was higher in OSCC when compared with the other groups (Kruskal-Wallis, p<0.05). Ki-67 expression percentage was higher in OL and in IH when compared with NOM (Kruskal-Wallis/Dunn, p<0.05). Increased expression of BMI-1 was also observed in OL when compared with NOM (Kruskal-Wallis/Dunn, p<0.05). No differences were observed in expression of both markers when non-dysplastic and dysplastic leukoplakias were compared. A significant positive correlation between Ki-67 and BMI-1 was found (Spearman correlation coefficient, R=0.26, p=0.01). High-grade epithelial dysplasia was associated with malignant transformation (Chi-squared, p=0.03). Conclusions These findings indicate that BMI-1 expression increases in early oral carcinogenesis and is possibly associated with the occurrence of dysplastic changes. Furthermore, our findings indicate that both Ki-67 and BMI-1 are directly correlated and play a role in initiation and progression of OSCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Leukoplakia, Oral/pathology , Mouth Neoplasms/pathology , Polycomb Repressive Complex 1/analysis , Adult , Aged , Analysis of Variance , Carcinogenesis/pathology , Case-Control Studies , Cell Proliferation , Cross-Sectional Studies , Disease Progression , Female , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Male , Middle Aged , Mouth Mucosa/pathology , Reference Values , Risk Factors , Statistics, Nonparametric , Tumor Burden
3.
Medicina (Kaunas) ; 56(2)2020 Feb 12.
Article in English | MEDLINE | ID: mdl-32059385

ABSTRACT

Background and objectives: B-lymphoma Mo-MLV insertion region 1 (Bmi-1) is a stem cell factor that is overexpressed in various human cancer tissues. It has been implicated in cancer cell proliferation, cell invasion, distant metastasis, and chemosensitivity, and is associated with patient survival. Several reports have also identified Bmi-1 protein overexpression in endometrial carcinoma; however, the relationship between Bmi-1 expression and its significance as a clinicopathological parameter is still insufficiently understood. Accordingly, the present study aimed to clarify whether immunohistochemical staining for Bmi-1 in human endometrial carcinoma and normal endometrial tissues can be used as a prognostic and cell proliferation marker. Materials and Methods: Bmi-1 expression was assessed in endometrioid carcinoma (grade 1-3) and normal endometrial tissues (in the proliferative and secretory phases) by immunohistochemistry; protein expression was evaluated using the nuclear labeling index (%) in the hot spot. Furthermore, we examined other independent prognostic and proliferation markers, including the protein levels of Ki-67, p53, and cyclin A utilizing semi-serial sections of endometrial carcinoma tissues. Results: The expression of the Bmi-1 protein was significantly higher in all grades of endometrial carcinoma than in the secretory phase of normal tissues. Moreover, Bmi-1 levels tended to be higher in G2 and G3 tissues than in G1 tissue, without reaching significance. Bmi-1 expression showed no notable differences among International Federation of Gynecology and Obstetrics (FIGO) stages in endometrial carcinoma. Furthermore, we observed a significant positive relationship between Bmi-1 and Ki-67, cyclin A, or p53 by Spearman's rank correlation test, implying that high Bmi-1 expression can be an independent prognostic marker in endometrial carcinoma. Conclusions: Our study suggests that Bmi-1 levels in endometrial carcinoma tissues may be useful as a reliable proliferation and prognostic biomarker. Recently, the promise of anti-Bmi-1 strategies for the treatment of endometrial carcinoma has been detected. Our results provide fundamental data regarding this anti-Bmi-1 strategy.


Subject(s)
Endometrial Neoplasms/diagnosis , Immunohistochemistry/standards , Polycomb Repressive Complex 1/analysis , Predictive Value of Tests , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Biopsy/methods , Cyclin A/analysis , Early Detection of Cancer/methods , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry/methods , Immunohistochemistry/statistics & numerical data , Japan , Ki-67 Antigen/analysis , Middle Aged , Polycomb Repressive Complex 1/blood , Tumor Suppressor Protein p53/analysis
4.
J. appl. oral sci ; 28: e20190532, 2020. tab, graf
Article in English | BBO - Dentistry , LILACS | ID: biblio-1101257

ABSTRACT

Abstract Oral leukoplakia (OL) is a white lesion of an indeterminate risk not related to any excluded (other) known diseases or disorders that carry no increased risk for cancer. Many biological markers have been used in an attempt to predict malignant transformation; however, no reliable markers have been established so far. Objective To evaluate cell proliferation and immortalization in OL, comparing non-dysplastic (Non-dys OL) and dysplastic OL (Dys OL). Methodology This is a cross-sectional observational study. Paraffin-embedded tissue blocks of 28 specimens of Non-dys OL, 33 of Dys OL, 9 of normal oral mucosa (NOM), 17 of inflammatory hyperplasia (IH), and 19 of oral squamous cell carcinomas (OSCC) were stained for Ki-67 and BMI-1 using immunohistochemistry. Results A gradual increase in BMI-1 and K-i67 expression was found in oral carcinogenesis. The immunolabeling for those markers was higher in OSCC when compared with the other groups (Kruskal-Wallis, p<0.05). Ki-67 expression percentage was higher in OL and in IH when compared with NOM (Kruskal-Wallis/Dunn, p<0.05). Increased expression of BMI-1 was also observed in OL when compared with NOM (Kruskal-Wallis/Dunn, p<0.05). No differences were observed in expression of both markers when non-dysplastic and dysplastic leukoplakias were compared. A significant positive correlation between Ki-67 and BMI-1 was found (Spearman correlation coefficient, R=0.26, p=0.01). High-grade epithelial dysplasia was associated with malignant transformation (Chi-squared, p=0.03). Conclusions These findings indicate that BMI-1 expression increases in early oral carcinogenesis and is possibly associated with the occurrence of dysplastic changes. Furthermore, our findings indicate that both Ki-67 and BMI-1 are directly correlated and play a role in initiation and progression of OSCC.


Subject(s)
Humans , Animals , Male , Adult , Middle Aged , Aged , Aged, 80 and over , Leukoplakia, Oral/pathology , Mouth Neoplasms/pathology , Carcinoma, Squamous Cell/pathology , Ki-67 Antigen/analysis , Polycomb Repressive Complex 1/analysis , Mouth Mucosa/pathology , Immunohistochemistry , Cross-Sectional Studies , Risk Factors , Statistics, Nonparametric , Disease Progression , Cell Proliferation , Carcinogenesis/pathology
5.
Oncology ; 97(5): 311-318, 2019.
Article in English | MEDLINE | ID: mdl-31550723

ABSTRACT

INTRODUCTION: Human epidermal growth factor 2 (HER2) gene overexpression in breast carcinoma cell lines has been shown to drive mammary carcinogenesis and tumor growth and invasion through its effects on mammary stem cells. OBJECTIVE: Therefore, we investigated the mechanism by which HER2 regulates cancer stem cell (CSC) activity in gastric cancer cells. METHODS: HER2 was transfected into MKN28 gastric cancer cells, and its role in regulating CSC activity was determined by characterizing the HER2-overexpressing cells. RESULTS: The sphere formation assay revealed that the sphere sizes and frequency of sphere formation were significantly greater for the HER2-overexpressing cells than for the MKN28 control cells. The CSC markers Oct-4 and BMI1 were more highly expressed in the HER2-overexpressing cells, as were the EMT markers. This was accompanied by a significant enhancement in cellular invasion of the Matrigel and migration. The E-cadherin level was significantly downregulated, and the mesenchymal marker Snail upregulated, in the HER2-transfected cells. HER2 overexpression activated the well-characterized CSC-associated Wnt/ß-catenin signaling pathway, as shown by the luciferase assay. After treatment of these cells with the Wnt signal inhibitor PRI-724, the BMI1 and Oct-4 levels were decreased for 24 h and Snail was also downregulated. Immunofluorescence staining revealed the significant restoration of E-cadherin levels in the HER2-transfected cells after PRI-724 treatment. CONCLUSIONS: These results established a role for HER2 in regulating gastric CSC activity, with Wnt/ß-catenin signaling being mediated via a HER2-dependent pathway. In summary, HER2-overexpressing gastric cancer cells exhibited increased stemness and invasiveness and were regulated by Wnt/ß-catenin signaling.


Subject(s)
Neoplastic Stem Cells/physiology , Receptor, ErbB-2/physiology , Stomach Neoplasms/pathology , Wnt Signaling Pathway/physiology , Antigens, CD/analysis , Cadherins/analysis , Cell Line, Tumor , Humans , Neoplasm Invasiveness , Octamer Transcription Factor-3/analysis , Polycomb Repressive Complex 1/analysis , Receptor, ErbB-2/analysis , Stomach Neoplasms/chemistry , beta Catenin/analysis
7.
Tumour Biol ; 39(4): 1010428317695930, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28381191

ABSTRACT

Tumor necrosis factor receptor-associated factor 1, an adaptor protein of tumor necrosis factor 2, is involved in classical nuclear factor (NF)-κB activation and lymphocyte recruitment. However, less is known about the expression and association of tumor necrosis factor receptor-associated factor 1 with cancer stem cell markers in oral squamous cell carcinoma. This study aimed to investigate the expression of tumor necrosis factor receptor-associated factor 1 and stem cell characteristic markers (lin28 homolog B, B cell-specific Moloney murine leukemia virus integration site 1, and aldehyde dehydrogenase 1) in oral squamous cell carcinoma and analyze their relations. Paraffin-embedded tissues of 78 oral squamous cell carcinomas, 39 normal oral mucosa, and 12 oral dysplasia tissues were employed in tissue microarrays, and the expression of tumor necrosis factor receptor-associated factor 1, B cell-specific Moloney murine leukemia virus integration site 1, aldehyde dehydrogenase 1, and lin28 homolog B was measured by immunohistostaining and digital pathological analysis. The expression of tumor necrosis factor receptor-associated factor 1 was higher in the oral squamous cell carcinoma group as compared with the expression in the oral mucosa (p < 0.01) and oral dysplasia (p < 0.001) groups. In addition, the expression of tumor necrosis factor receptor-associated factor 1 was associated with those of B cell-specific Moloney murine leukemia virus integration site 1, aldehyde dehydrogenase 1, and lin28 homolog B (p = 0.032, r2 = 0.109; p < 0.0001, r2 = 0.64; and p < 0.001, r2 = 0.16) in oral squamous cell carcinoma. The patient survival rate was lower in the highly expressed tumor necrosis factor receptor-associated factor 1 group, although the difference was not significant. The clustering analysis showed that tumor necrosis factor receptor-associated factor 1 was most related to aldehyde dehydrogenase 1. These findings suggest that tumor necrosis factor receptor-associated factor 1 has potential direct/indirect regulations with the cancer stem cell markers in oral squamous cell carcinoma, which may help in further analysis of the cancer stem cell characteristics.


Subject(s)
Carcinoma, Squamous Cell/chemistry , Isoenzymes/analysis , Mouth Neoplasms/chemistry , Neoplastic Stem Cells/chemistry , Polycomb Repressive Complex 1/analysis , RNA-Binding Proteins/analysis , Retinal Dehydrogenase/analysis , TNF Receptor-Associated Factor 1/analysis , Aldehyde Dehydrogenase 1 Family , Biomarkers , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Humans , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology
8.
Oncotarget ; 7(52): 86186-86197, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27863389

ABSTRACT

Acute myeloid leukemia (AML) is an aggressive malignancy characterized by heterogeneous genetic and epigenetic changes in hematopoietic progenitors that lead to abnormal self-renewal and proliferation. Despite high initial remission rates, prognosis remains poor for most AML patients, especially for those harboring internal tandem duplication (ITD) mutations in the fms-related tyrosine kinase-3 (FLT3). Here, we report that a novel epidithiodiketopiperazine, NT1721, potently decreased the cell viability of FLT3-ITD+ AML cell lines, displaying IC50 values in the low nanomolar range, while leaving normal CD34+ bone marrow cells largely unaffected. The IC50 values for NT1721 were significantly lower than those for clinically used AML drugs (i.e. cytarabine, sorafenib) in all tested AML cell lines regardless of their FLT3 mutation status. Moreover, combinations of NT1721 with sorafenib or cytarabine showed better antileukemic effects than the single agents in vitro. Combining cytarabine with NT1721 also attenuated the cytarabine-induced FLT3 ligand surge that has been linked to resistance to tyrosine kinase inhibitors. Mechanistically, NT1721 depleted DNA methyltransferase 1 (DNMT1) protein levels, leading to the re-expression of silenced tumor suppressor genes and apoptosis induction. NT1721 concomitantly decreased the expression of EZH2 and BMI1, two genes that are associated with the maintenance of leukemic stem/progenitor cells. In a systemic FLT3-ITD+ AML mouse model, treatment with NT1721 reduced tumor burdens by > 95% compared to the control and significantly increased survival times. Taken together, our results suggest that NT1721 may represent a promising novel agent for the treatment of AML.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Piperazines/therapeutic use , Animals , Cell Line, Tumor , Cytarabine/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1/analysis , Humans , Membrane Proteins/analysis , Mice , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Piperazines/pharmacology , Polycomb Repressive Complex 1/analysis , Sorafenib
9.
Br J Nutr ; 116(7): 1188-1198, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27572423

ABSTRACT

Pro-inflammatory cytokines are critical in mechanisms of muscle atrophy. In addition, asparagine (Asn) is necessary for protein synthesis in mammalian cells. We hypothesised that Asn could attenuate lipopolysaccharide (LPS)-induced muscle atrophy in a piglet model. Piglets were allotted to four treatments (non-challenged control, LPS-challenged control, LPS+0·5 % Asn and LPS+1·0 % Asn). On day 21, the piglets were injected with LPS or saline. At 4 h post injection, piglet blood and muscle samples were collected. Asn increased protein and RNA content in muscles, and decreased mRNA expression of muscle atrophy F-box (MAFbx) and muscle RING finger 1 (MuRF1). However, Asn had no effect on the protein abundance of MAFbx and MuRF1. In addition, Asn decreased muscle AMP-activated protein kinase (AMPK) α phosphorylation, but increased muscle protein kinase B (Akt) and Forkhead Box O (FOXO) 1 phosphorylation. Moreover, Asn decreased the concentrations of TNF-α, cortisol and glucagon in plasma, and TNF-α mRNA expression in muscles. Finally, Asn decreased mRNA abundance of muscle toll-like receptor (TLR) 4 and nucleotide-binding oligomerisation domain protein (NOD) signalling-related genes, and regulated their negative regulators. The beneficial effects of Asn on muscle atrophy may be associated with the following: (1) inhibiting muscle protein degradation via activating Akt and inactivating AMPKα and FOXO1; and (2) decreasing the expression of muscle pro-inflammatory cytokines via inhibiting TLR4 and NOD signalling pathways by modulation of their negative regulators.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Asparagine/pharmacology , Gene Expression/drug effects , Muscular Atrophy/genetics , Proto-Oncogene Proteins c-akt/metabolism , Toll-Like Receptor 4/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Enzyme Activation/drug effects , F-Box Proteins/analysis , F-Box Proteins/genetics , Forkhead Box Protein O1/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Muscle Proteins/metabolism , Muscle, Skeletal/chemistry , Muscular Atrophy/chemically induced , Nod Signaling Adaptor Proteins/antagonists & inhibitors , Phosphorylation/drug effects , Polycomb Repressive Complex 1/analysis , Polycomb Repressive Complex 1/genetics , RNA, Messenger/analysis , Signal Transduction/drug effects , Sus scrofa , Toll-Like Receptor 4/genetics , Weaning
10.
Hum Pathol ; 57: 126-135, 2016 11.
Article in English | MEDLINE | ID: mdl-27476776

ABSTRACT

Minichromosomal maintenance (MCM) proteins are participants of DNA replication and may represent more accurate markers in determining the proliferative fraction within a tumor than proliferative marker Ki-67. Our study investigated the correlation between MCM4 and MCM7 expression and Ki-67, Bmi1, and cyclin E expression in esophageal adenocarcinoma, squamous cell carcinoma, and precancerous lesions. MCM4 and MCM7 expression had similar distribution as Ki-67 and Bmi1 expression in esophageal carcinoma and precancerous lesions. The mean percentage of MCM4, MCM7, and Ki-67 expression increased from squamous epithelium (5.5%, 7.3%, and 5.9%, respectively), to columnar cell metaplasia (11.2, 13.5%, and 3.4%), Barrett's esophagus (27.7%, 35.3%, and 8.3%), low-grade dysplasia (42.6%, 52.2%, and 12.9%), high-grade dysplasia (63.2%, 77.7%, and 29.6%), adenocarcinoma (61.3%, 75.5%, and 24.5%), and squamous cell carcinoma (74.1, 85.4%, and 36.3%). The percentages of MCM4 and MCM7 expression were significantly higher than Ki-67 expression. Using univariate analysis we found a high percentage of MCM4 expression (>70%) to be significantly associated with lymph node metastasis and shorter survival in the adenocarcinoma group. We also demonstrated the percentage of MCM4 and MCM7 expression to be significantly correlated with Ki-67, Bmi1, and cyclin E expression in esophageal carcinoma and precancerous lesions. MCM4 and MCM7 may serve as more sensitive proliferative markers for the evaluation of esophageal lesions.


Subject(s)
Adenocarcinoma/chemistry , Barrett Esophagus/metabolism , Carcinoma, Squamous Cell/chemistry , Cell Proliferation , Cyclin E/analysis , Esophageal Neoplasms/chemistry , Ki-67 Antigen/analysis , Minichromosome Maintenance Complex Component 4/analysis , Minichromosome Maintenance Complex Component 7/analysis , Polycomb Repressive Complex 1/analysis , Precancerous Conditions/chemistry , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Barrett Esophagus/mortality , Barrett Esophagus/pathology , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Chi-Square Distribution , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Multivariate Analysis , Neoplasm Grading , Precancerous Conditions/mortality , Precancerous Conditions/pathology , Predictive Value of Tests , Reproducibility of Results , Risk Factors , Tissue Array Analysis
11.
APMIS ; 124(7): 541-6, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27102362

ABSTRACT

The aim of this study was to investigate the prognostic value of B-cell-specific moloney murine leukemia virus insertion site 1 (BMI1) protein expression in primary tumors of stage II colon cancer patients. BMI1 protein expression was assessed by immunohistochemistry in a retrospective patient cohort consisting of 144 stage II colon cancer patients. BMI1 expression at the invasive front of the primary tumors correlated with mismatch repair status of the tumors. Furthermore, BMI1 expression at the luminal surface correlated with T-stage, tumor location, and the histological subtypes of the tumors. In a univariate Cox proportional hazard analysis, no statistical significant association between risk of relapse and BMI1 protein expression at the invasive front (HR: 1.12; 95% CI 0.78-1.60; p = 0.53) or at the luminal surface of the tumor (HR: 1.06; 95% CI 0.75-1.48; p = 0.70) was found. Likewise, there was no association between 5-year overall survival and BMI1 expression at the invasive front (HR: 1.12; 95% CI 0.80-1.56; p = 0.46) or at the luminal surface of the tumor (HR: 1.16; 95% CI 0.86-1.60; p = 0.33). In conclusion, BMI1 expression in primary tumors of stage II colon cancer patients could not predict relapse or overall survival of the patients, thus having a limited prognostic value in stage II colon cancer patients.


Subject(s)
Biomarkers, Tumor/analysis , Colonic Neoplasms/diagnosis , Colonic Neoplasms/pathology , Polycomb Repressive Complex 1/analysis , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Recurrence , Retrospective Studies , Survival Analysis
12.
Int J Mol Med ; 36(3): 783-91, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26202447

ABSTRACT

Aggressive tumor cells can mimic embryonic vasculogenic networks and form vasculogenic mimicry (VM). Preliminary studies demonstrated that hypoxia can promote VM formation; however, the underlying mechanism remains unclear. The present study aimed to investigate the role of the Twist1­Bmi1 connection in hypoxia­induced VM formation and the underlying mechanism. In the in vitro experiments, western blot analysis demonstrated that hypoxia upregulated the expression of Twist1, Bmi1, epithelial­mesenchymal transition (EMT) markers, stem cell markers and VM­associated markers. The 3D culture assay showed that hypoxia promoted VM formation in hepatocellular carcinoma (HCC) cell lines. Using transfection and in vitro cell experiments, the Twist1­Bmi1 connection was confirmed to have an important role in inducing EMT, cell stemness and VM formation. In the in vivo experiments, the murine hypoxia models were established via incomplete femoral artery ligation and the mechanism by which hypoxia promoted Twist1 and Bmi1 expression and led to VM formation was demonstrated by immunohistochemistry staining and endomucin/periodic acid Schiff double­staining. In conclusion, hypoxia upregulate the expression of Twist1 and Bmi1, and these two proteins have an important role in inducing EMT and cancer cell stemness, which contributed to VM formation.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Hypoxia/metabolism , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Twist-Related Protein 1/metabolism , Animals , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/pathology , Cell Hypoxia , Epithelial-Mesenchymal Transition , Hep G2 Cells , Humans , Hypoxia/complications , Hypoxia/pathology , Liver/blood supply , Liver/metabolism , Liver/pathology , Liver Neoplasms/blood supply , Liver Neoplasms/complications , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Nuclear Proteins/analysis , Polycomb Repressive Complex 1/analysis , Proto-Oncogene Proteins/analysis , Twist-Related Protein 1/analysis
13.
Biochem Biophys Res Commun ; 458(3): 470-475, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25660450

ABSTRACT

Epigenetic modifications such as DNA methylation and histone H3 lysine 27 methylation (H3K27me) are repressive marks that silence gene expression. The M phase phosphoprotein (MPP8) associates with proteins involved in both DNA methylation and histone modifications, and therefore, is a potential candidate to mediate crosstalk between repressive epigenetic pathways. Here, by performing immunohistochemical analyses we demonstrate that MPP8 is expressed in the rodent testis, especially in spermatocytes, suggesting a role in spermatogenesis. Interestingly, we found that MPP8 physically interacts with PRC1 (Polycomb Repressive Complex 1) components which are known to possess essential function in testis development by modulating monoubiquitination of Histone H2A (uH2A) and trimethylation of Histone H3 Lysine 27 (H3K27me3) residues. Knockdown analysis of MPP8 in HeLa cells resulted in derepression of a set of genes that are normally expressed in spermatogonia, spermatids and mature sperm, thereby indicating a role for this molecule in silencing testis-related genes in somatic cells. In addition, depletion of MPP8 in murine ES cells specifically induced expression of genes involved in mesoderm differentiation, such as Cdx2 and Brachyury even in the presence of LIF, which implicated that MPP8 might be required to repress differentiation associated genes during early development. Taken together, our results indicate that MPP8 could have a role for silencing genes that are associated with differentiation of the testis and the mesoderm by interacting with epigenetic repressors modules such as the PRC1 complex.


Subject(s)
Phosphoproteins/genetics , Phosphoproteins/metabolism , Polycomb Repressive Complex 1/metabolism , Spermatogenesis , Animals , Cell Line , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Knockdown Techniques , HeLa Cells , Histones/metabolism , Humans , Male , Mice , Phosphoproteins/analysis , Polycomb Repressive Complex 1/analysis , Protein Interaction Maps , Rats, Inbred F344 , Spermatocytes/cytology , Spermatocytes/metabolism , Transcriptional Activation
14.
Int J Oral Sci ; 6(4): 212-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25012868

ABSTRACT

Previous studies have demonstrated that spheroid type cells grown under suspension culture conditions have cancer stem cell (CSC) traits in a number of cancers, but this phenomenon has not yet been reported in the VX2 rabbit oral cancer model. Hence, this study aimed to study the spheroid cells from VX2 rabbit buccal squamous cell carcinomas (SCCs) and assess their CSC characteristics. Five adult male New Zealand white outbred rabbits were used to generate VX2 rabbit buccal SCC. Sphere-forming cell culture was performed for the VX2 rabbit buccal SCC specimens. The self-renewal capability; cluster of designation (CD) 44, CD133, acetaldehyde dehydrogenase 1 (ALDH1), B cell-specific Moloney murine leukemia virus integration site 1 (Bmi-1), Nestin, octamer-binding transcription factor 4 (Oct4) and reduced expression protein-1 (Rex-1) expression with reverse transcription-polymerase chain reaction (RT-PCR); chemoresistance to cisplatin and 5-fluorouracil; and in vivo tumorigenicity of spheroid cell transplantation in nude mice were evaluated to determine the CSC characteristics of the resulting spheroid cells. We successfully obtained spheroid cells from the VX2 rabbit OSCC tissues. The spheroid cells exhibited CSC traits, including the expression of CSC and stem cell markers (CD44, Bmi-1, Nestin, Oct4 and Rex-1), capacity to generate new spheroid colonies within 1 week of reseeding from single-dissociated spheroid cells, chemoresistance capacity and generation of tumour xenografts (with histological features resembling those of the original VX2 rabbit buccal SCC) from the transplantation of 10(3) undifferentiated spheroid cells into nude mice. In summary, we demonstrated that spheroid cells with CSC cell traits can be derived from VX2 rabbit buccal SCCs, indicating that this animal cancer model is applicable for studying CSCs in human oral cancers.


Subject(s)
Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/pathology , Neoplastic Stem Cells/classification , Spheroids, Cellular/classification , AC133 Antigen , Aldehyde Dehydrogenase 1 Family , Animals , Antigens, CD/analysis , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Cisplatin/pharmacology , DNA-Binding Proteins/analysis , Disease Models, Animal , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Glycoproteins/analysis , Heterografts/transplantation , Hyaluronan Receptors/analysis , Isoenzymes/analysis , Male , Mice , Mice, Nude , Neoplasm Transplantation , Nestin/analysis , Octamer Transcription Factor-3/analysis , Peptides/analysis , Polycomb Repressive Complex 1/analysis , Proto-Oncogene Proteins/analysis , Rabbits , Retinal Dehydrogenase/analysis
15.
Tumour Biol ; 35(10): 9619-25, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24964959

ABSTRACT

Mel-18 is a member of the polycomb group (PcG) of proteins, which are chromatin regulatory factors that play an important role in oncogenesis. This study was designed to investigate the clinical and prognostic significance of Mel-18 in colorectal cancer (CRC) patients. For this purpose, expression of Mel-18 mRNA was evaluated in 82 primary CRC and paired noncancerous mucosa samples by qRT-PCR and Western blotting. We found that overall Mel-18 mRNA expression in the CRC tissue was significantly lower than in the noncancerous mucosal tissue (p = 0.007, Wilcoxon matched-pairs signed-ranks test). Mel-18 was conversely correlated with the pathological classifications (p = 0.003 for T, p < 0.001 for N, and p = 0.015 for M classifications, respectively) and clinical AJCC stage (p < 0.001). Furthermore, CRC patients with a higher level of Mel-18 showed prolonged disease-free survivals (DFS) (p < 0.001). In multivariate analysis, the diminished Mel-18 expression may be a risk factor for the patients' 3-year DFS (HR = 1.895; 95 % CI 1.032, 3.477; p = 0.039). It was therefore concluded that the lower Mel-18 expression might contribute to the CRC development/progression.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/metabolism , Polycomb Repressive Complex 1/biosynthesis , Blotting, Western , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Polycomb Repressive Complex 1/analysis , Prognosis , Proportional Hazards Models , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
Ann Surg Oncol ; 21 Suppl 4: S657-64, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24833101

ABSTRACT

BACKGROUND: It has been suggested that markers associated with cancer stem cells (CSC) may play a role in esophageal cancer. Our aim was to investigate the expression pattern of proposed CSC markers ALDH1, Axin2, BMI1, CD44, and SOX2 in esophageal adenocarcinoma (EAC) and to relate their expression to survival. METHODS: In this study we included 94 EAC patients and examined the expression of the above-mentioned markers by using immunohistochemistry on tissue microarrays. Expression was scored as positive or negative or categorized as low or high in terms of an immunoreactivity score (IRS). Expression rates were related to clinicopathologic characteristics and overall and disease-free survival (DFS). RESULTS: In a multivariate analysis, negative expression of CD44 and of SOX2 were both significant prognostic factors for DFS [hazard ratio (HR), 1.73; 95 % confidence interval (CI), 1.00-2.96; P = 0.046 and HR, 2.06; 95 % CI 1.14-3.70 P = 0.016). When CD44 and SOX2 expression were analyzed together, negative SOX2 expression was an independent prognostic factor for DFS (HR, 1.91; 95 % CI 1.05-3.46; P = 0.034). Low IRS scores for ALDH1 or Axin2 were associated with a reduced median survival (12.8 vs. 28.7 and 12.1 vs. 25.5 months, respectively). However, these markers and BMI1 were not prognostic factors for survival. CONCLUSIONS: Loss of CD44 expression and loss of SOX2 expression are prognostic factors of poor survival in EAC patients. This suggests a role of these proteins in EAC that requires further investigation.


Subject(s)
Adenocarcinoma/chemistry , Biomarkers, Tumor/analysis , Esophageal Neoplasms/chemistry , Hyaluronan Receptors/analysis , SOXB1 Transcription Factors/analysis , Adenocarcinoma/surgery , Aged , Aldehyde Dehydrogenase 1 Family , Axin Protein/analysis , Disease-Free Survival , Esophageal Neoplasms/surgery , Esophagectomy , Female , Humans , Isoenzymes/analysis , Kaplan-Meier Estimate , Male , Middle Aged , Polycomb Repressive Complex 1/analysis , Retinal Dehydrogenase/analysis , Retrospective Studies , Survival Rate
17.
World J Gastroenterol ; 20(18): 5533-9, 2014 May 14.
Article in English | MEDLINE | ID: mdl-24833884

ABSTRACT

AIM: To determine the correlation between invasiveness, migration and prognosis in esophageal squamous cell carcinoma (ESCC) and expression of the B-cell-specific Moloney leukemia virus insert site 1 (Bmi-1) and plasminogen activator inhibitor-1 (PAI-1). METHODS: Eighty previously untreated patients who underwent surgical excision of ESCC were included. The expression of Bmi-1 and PAI-1 was examined immunohistochemically in formalin-fixed paraffin-embedded primary tissue specimens. The relationships between the expression of Bmi-1 and PAI-1, the clinicopathologic features of ESCC, and the survival rate of ESCC patients were also discussed. The correlation between Bmi-1 and PAI-1 protein expression in ESCC was analyzed. The relationship between Bmi-1 and PAI-1 expression and ESCC prognosis was evaluated using a Cox regression model and Kaplan-Meier survival curve analysis. RESULTS: The rates of positive Bmi-1 and PAI-1 expression in ESCC were higher than those in normal esophageal tissue (P < 0.05). The expression of Bmi-1 and PAI-1 was correlated with depth of invasion and lymph node metastasis (P < 0.05), but not with patient age, tumor size or nationality (P > 0.05). The expression of Bmi-1 was positively correlated with that of PAI-1 (P < 0.05). The 10-year overall survival rate for all patients was 20% (16/80). Univariate Kaplan-Meier survival analysis showed that patients with high expression of esophageal PAI-1 and Bmi-1 had lower survival, however, the difference was not statistically significant. Cox multivariate analysis showed that PAI-1 and Bmi-1 were not independent factors for survival rate, while the depth of tumor invasion and metastasis were independent factors affecting patient survival. CONCLUSION: The expression of Bmi-1 and PAI-1 plays a role in ESCC progression, and may be used as a prognostic marker in ESCC.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/chemistry , Esophageal Neoplasms/chemistry , Plasminogen Activator Inhibitor 1/analysis , Polycomb Repressive Complex 1/analysis , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/surgery , Esophageal Squamous Cell Carcinoma , Esophagectomy , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Invasiveness , Predictive Value of Tests , Proportional Hazards Models , Risk Factors
18.
Biol Cell ; 106(5): 151-65, 2014 May.
Article in English | MEDLINE | ID: mdl-24611931

ABSTRACT

BACKGROUND INFORMATION: The optimal repair of DNA lesions is fundamental for physiological processes. We asked whether the recruitment of HP1ß, 53BP1 and BMI1 proteins to ultraviolet (UVA)-induced DNA lesions requires functional A-type lamins. RESULTS: We found that UVA irradiation of nuclear lamina abolished the fluorescence of mCherry-tagged A-type lamins and destroyed the nuclear lamina as also observed by electron microscopy studies. Similarly, an absence of endogenous A- and B-type lamins was found in irradiated regions by UVA. However, irradiation did not affect the recruitment of HP1ß, 53BP1 and BMI1 to DNA lesions. The UVA-induced shrinkage of the nuclear lamina, which anchors chromatin, explains why UVA-micro-irradiated chromatin is relaxed. Conversely, additional experiments with γ-irradiation showed that the nuclear lamina remained intact and the genome-wide level of HP1ß was stable. Fluorescence intensity of HP1ß and BMI1 in UVA-induced DNA lesions and level of HP1ß after γ-irradiation were unaffected by deficiency in A-type lamins, whereas those parameters of 53BP1 were changed. CONCLUSIONS: We conclude that only the 53BP1 status in DNA lesions, induced by UVA or γ-rays, is affected by A-type lamin deficiency, which was not observed for heterochromatin-related proteins HP1ß and BMI1.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Damage/radiation effects , Lamin Type A/metabolism , 3T3 Cells , Animals , Chromosomal Proteins, Non-Histone/analysis , DNA/genetics , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Lamin Type A/analysis , Mice , Polycomb Repressive Complex 1/analysis , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1 , Ultraviolet Rays
19.
Free Radic Biol Med ; 65: 1464-1472, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23917144

ABSTRACT

Nitrative and oxidative DNA damage plays an important role in inflammation-related carcinogenesis. Chronic inflammation such as parasite infection and primary sclerosing cholangitis can be an etiological factor of cholangiocarcinoma. Using a proteomic approach and double-fluorescent staining, we identified high expression and colocalization of albumin and cytokeratin-19 in liver fluke-associated cholangiocarcinoma tissues, compared with normal livers from cholangiocarcinoma patients and cadaveric donors, respectively. Albumin was detected not only in cells of hyperplastic bile ducts and cholangiocarcinoma, but also in liver stem/progenitor cell origin, such as canal of Hering, ductules, and ductular reactions, suggesting the involvement of stem/progenitor cells in cholangiocarcinoma development. To clarify the involvement of liver stem/progenitor cells in cholangiocarcinoma, we examined several stem/progenitor cell markers (CD133, CD44, OV6, and Oct3/4) in cholangiocarcinoma tissues analyzed by immunohistochemical staining, and measured 8-oxodG levels by using HPLC-ECD as an inflammation-related DNA lesion. In addition, a stem/progenitor cell factor Bmi1, 8-nitroguanine (formed during nitrative DNA damage), DNA damage response (DDR) proteins (phosphorylated ATM and γ-H2AX), and manganese-SOD (Mn-SOD) were analyzed by immunohistochemistry. Stem/progenitor cell markers (CD133, OV6, CD44, and Oct3/4) were positively stained in 56, 38, 47, and 56% of 34 cholangiocarcinoma cases, respectively. Quantitative analysis of 8-oxodG revealed significantly increased levels in CD133- and/or Oct3/4-positive tumor tissues compared to negative tumor tissues, as well as 8-nitroguanine formation detected by immunohistochemistry. In the cases of CD44- and/or OV6-positive tissue, no significant difference was observed. Cholangiocarcinoma patients with CD133- and/or Oct3/4-positive tumor tissues showed significantly lower expression of Mn-SOD and higher DDR protein, γ-H2AX. Moreover, CD133- and/or Oct3/4-positive cholangiocarcinoma patients had significant associations with tumor histology types, tumor stage, and poor prognoses. Our results suggest that CD133 and Oct3/4 in cholangiocarcinoma are associated with increased formation of DNA lesions and the DDR protein, which may be involved in genetic instability and lead to cholangiocarcinoma development with aggressive clinical features.


Subject(s)
Antigens, CD/genetics , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , DNA Damage/genetics , Genomic Instability/genetics , Glycoproteins/genetics , Octamer Transcription Factor-3/genetics , Peptides/genetics , 8-Hydroxy-2'-Deoxyguanosine , AC133 Antigen , Albumins/metabolism , Antigens, CD/biosynthesis , Antigens, Differentiation/biosynthesis , Bile Ducts, Intrahepatic/pathology , Cell Transformation, Neoplastic/immunology , Cholangitis, Sclerosing , DNA Damage/immunology , DNA Repair , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/analysis , Female , Glycoproteins/biosynthesis , Guanine/analogs & derivatives , Guanine/analysis , Guanine/biosynthesis , Histones/biosynthesis , Humans , Hyaluronan Receptors/biosynthesis , Inflammation/immunology , Keratin-19/metabolism , Liver/cytology , Male , Middle Aged , Octamer Transcription Factor-3/biosynthesis , Oxidation-Reduction , Polycomb Repressive Complex 1/analysis , Prognosis , Stem Cells/cytology , Superoxide Dismutase
20.
PLoS One ; 8(1): e52993, 2013.
Article in English | MEDLINE | ID: mdl-23308129

ABSTRACT

BACKGROUND: Lack of reliable predictive biomarkers is a stumbling block in the management of prostate cancer (CaP). Prostate-specific antigen (PSA) widely used in clinics has several caveats as a CaP biomarker. African-American CaP patients have poor prognosis than Caucasians, and notably the serum-PSA does not perform well in this group. Further, some men with low serum-PSA remain unnoticed for CaP until they develop disease. Thus, there is a need to identify a reliable diagnostic and predictive biomarker of CaP. Here, we show that BMI1 stem-cell protein is secretory and could be explored for biomarker use in CaP patients. METHODOLOGY/PRINCIPAL FINDINGS: Semi-quantitative analysis of BMI1 was performed in prostatic tissues of TRAMP (autochthonous transgenic mouse model), human CaP patients, and in cell-based models representing normal and different CaP phenotypes in African-American and Caucasian men, by employing immunohistochemistry, immunoblotting and Slot-blotting. Quantitative analysis of BMI1 and PSA were performed in blood and culture-media of siRNA-transfected and non-transfected cells by employing ELISA. BMI1 protein is (i) secreted by CaP cells, (ii) increased in the apical region of epithelial cells and stromal region in prostatic tumors, and (iii) detected in human blood. BMI1 is detectable in blood of CaP patients in an order of increasing tumor stage, exhibit a positive correlation with serum-PSA and importantly is detectable in patients which exhibit low serum-PSA. The clinical significance of BMI1 as a biomarker could be ascertained from observation that CaP cells secrete this protein in higher levels than cells representative of benign prostatic hyperplasia (BPH). CONCLUSIONS/SIGNIFICANCE: BMI1 could be developed as a dual bio-marker (serum and biopsy) for the diagnosis and prognosis of CaP in Caucasian and African-American men. Though compelling these data warrant further investigation in a cohort of African-American patients.


Subject(s)
Polycomb Repressive Complex 1/blood , Prostate/pathology , Prostatic Neoplasms/blood , Prostatic Neoplasms/diagnosis , Black or African American , Animals , Biopsy , Cell Line , Cell Line, Tumor , Humans , Male , Mice , Mice, Transgenic , Polycomb Repressive Complex 1/analysis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/analysis , White People
SELECTION OF CITATIONS
SEARCH DETAIL