Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Methods Cell Biol ; 171: 23-32, 2022.
Article in English | MEDLINE | ID: mdl-35953204

ABSTRACT

Quantitative real-time polymerase chain reaction (qRT-PCR) flexibility, robustness and reproducibility have rapidly extended the scope of the method. Cancer stem cells are gaining increasing importance since their role in cancer initiation, treatment resistance and recurrence give rise to a wide range of potential diagnostic and therapeutic applications. The expression of several characteristic markers is proven a reliable method to assess stem-like-phenotype of cancer cells. Here, we provided a thorough protocol for the study of cancer stem cells in hepatocellular carcinoma mouse models and cell cultures using qRT-PCR.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , AC133 Antigen/genetics , AC133 Antigen/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Neoplastic Stem Cells/pathology , Real-Time Polymerase Chain Reaction , Reproducibility of Results
2.
Clin Transl Oncol ; 23(9): 1743-1751, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33721187

ABSTRACT

OBJECTIVES: The promoting roles of cyclin dependent kinase 1 (CDK1) have been revealed in various tumors, however, its effects in the progression of cancer stem cells are still confusing. This work aims to explore the roles of CDK1 in regulating the stemness of lung cancer cells. METHODS: Online dataset analysis was performed to evaluate the correlation between CDK1 exression and the survival of lung cancer patients. RT-qPCR, western blot, cell viability, sphere-formation analysis and ALDH activity detection were used to investigate the roles of CDK1 on lung cancer cell stemness, viability and chemotherapeutic sensitivity. Immunocoprecipitation (Co-IP) analysis and rescuing experiments were performed to reveal the underlying mechanisms contributing to CDK1-mediated effects on lung cancer cell stemness. RESULTS: CDK1 mRNA expression was negatively correlated with the overall survival of lung cancer patients and remarkably increased in tumor spheres formed by lung cancer cells compared to the parental cells. Additionally, CDK1 positively regulated the stemness of lung cancer cells. Mechanistically, CDK1 could interact with Sox2 protein, but not other stemness markers (Oct4, Nanog and CD133). Furthermore, CDK1 increased the phosphorylation, cytoplasm-nuclear translocation and transcriptional activity of Sox2 protein in lung cancer cells. Moreover, CDK1 positively regulated the stemness of lung cancer cells in a Sox2-dependent manner. Finally, we revealed that inhibition of CDK1 enhanced the chemotherapeutic sensitivity, which was also rescued by Sox2 overexpression. CONCLUSIONS: This work reveals a novel CDK1/Sox2 axis responsible for maintaining the stemness of lung cancer cells.


Subject(s)
CDC2 Protein Kinase/metabolism , Lung Neoplasms/pathology , Neoplastic Stem Cells/pathology , SOXB1 Transcription Factors/metabolism , A549 Cells , AC133 Antigen/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Aldehyde Dehydrogenase/metabolism , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Survival , Disease Progression , Humans , Immunoprecipitation/methods , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Nanog Homeobox Protein/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/drug effects , Octamer Transcription Factor-3/metabolism , Phosphorylation , RNA, Messenger/metabolism , Spheroids, Cellular/pathology
3.
Asian Pac J Cancer Prev ; 21(9): 2501-2506, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32986345

ABSTRACT

OBJECTIVE: to investigate CD133 immunoexpression, cancer stem cells marker, in oral epithelial dysplasias (OEDs) and oral squamous cells carcinomas (OSCCs) and understandits possible involvement in the malignant transformation process of these lesions and to better elucidate their biological behavior. MATERIAL AND METHODS: Tissue samples of 15 cases of OSCCs and 15 OEDs were subjected to CD133 antibody immunohistochemistry reactions. The analysis used quantitative parameters (number of immunostained cells regardless of immunostaining sublocations). RESULTS: All samples of OSCCs and OEDs showed positive immunostaining, with no significant difference between these groups (p = 0.283). We did not observe statistical difference between the degree of dysplasia and the amount of CD133+ cells (p = 0.899). CD133 immunoexpression showed no association with the OEDs and OSCCs sites. It was observed that nuclear and cytoplasmic immunostaining was more evident with the progression of the malignant process. CONCLUSION: It is suggested that the CD133 cellular localization together with the histopathological criteria of OEDs classification can contribute to provide more concrete indications about the oral carcinogenesis process.


Subject(s)
AC133 Antigen/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Focal Epithelial Hyperplasia/pathology , Mouth Neoplasms/pathology , Precancerous Conditions/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/metabolism , Female , Focal Epithelial Hyperplasia/metabolism , Follow-Up Studies , Humans , Male , Middle Aged , Mouth Neoplasms/metabolism , Precancerous Conditions/metabolism , Prognosis
4.
J Mol Neurosci ; 70(6): 981-992, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32056089

ABSTRACT

Medulloblastoma (MB), which originates from embryonic neural stem cells (NSCs) or neural precursors in the developing cerebellum, is the most common malignant brain tumor of childhood. Recurrent and metastatic disease is the principal cause of death and may be related to resistance within cancer stem cells (CSCs). Chromatin state is involved in maintaining signaling pathways related to stemness, and inhibition of histone deacetylase enzymes (HDAC) has emerged as an experimental therapeutic strategy to target this cell population. Here, we observed antitumor actions and changes in stemness induced by HDAC inhibition in MB. Analyses of tumor samples from patients with MB showed that the stemness markers BMI1 and CD133 are expressed in all molecular subgroups of MB. The HDAC inhibitor (HDACi) NaB reduced cell viability and expression of BMI1 and CD133 and increased acetylation in human MB cells. Enrichment analysis of genes associated with CD133 or BMI1 expression showed mitogen-activated protein kinase (MAPK)/ERK signaling as the most enriched processes in MB tumors. MAPK/ERK inhibition reduced expression of the stemness markers, hindered MB neurosphere formation, and its antiproliferative effect was enhanced by combination with NaB. These results suggest that combining HDAC and MAPK/ERK inhibitors may be a novel and more effective approach in reducing MB proliferation when compared to single-drug treatments, through modulation of the stemness phenotype of MB cells.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Medulloblastoma/metabolism , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , AC133 Antigen/genetics , AC133 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Tumor Cells, Cultured
5.
Clin Transl Oncol ; 22(7): 1040-1048, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31630355

ABSTRACT

OBJECTIVE: To investigate the gene expression profile of CSCs and to explore the key pathways and specific molecular signatures involved in the characteristic of CSCs. MATERIALS AND METHODS: CD133+ /CD44+ CSCs and bulk population (non-CSCs) were isolated from DU-145 cells using fluorescence-activated cell sorting (FACS). We used Illumina HumanHT-12 v4 Expression to investigate gene expression profiling of CSCs and non-CSCs. Protein-protein interaction (PPI) network analysis was performed using the STRING database. Biomarkers selected based on gene expression profiling were visually analyzed using immunofluorescence staining method. An image analysis program, ImageJ®, was used for the analysis of fluorescence intensity. RESULTS: In microarray analysis, we found that many ribosomal proteins and translation initiation factors that constitute the mTOR complex were highly expressed. PPI analysis using the 33 genes demonstrated that there was a close interaction between ribosome biogenesis, translation, and mTOR signaling. The fluorescence amount of mTOR and MLST8 were higher in CSCs compared to non-CSCs. CONCLUSIONS: The increase in a number of genes associated with ribosome biogenesis, translation, and mTOR signaling may be important to evaluate prognosis and determine treatment approach for prostate cancer (PCa). A better understanding of the molecular pathways associated with CSCs may be promising to develop targeted therapies to prolong survival in PCa.


Subject(s)
Eukaryotic Initiation Factors/genetics , Neoplastic Stem Cells/metabolism , Organelle Biogenesis , Prostatic Neoplasms/genetics , Ribosomes/genetics , TOR Serine-Threonine Kinases/genetics , Transcriptome , mTOR Associated Protein, LST8 Homolog/genetics , AC133 Antigen/metabolism , Cell Line, Tumor , Eukaryotic Initiation Factors/metabolism , Flow Cytometry , Humans , Hyaluronan Receptors/metabolism , Male , Prostatic Neoplasms/metabolism , Protein Interaction Maps , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Spheroids, Cellular , TOR Serine-Threonine Kinases/metabolism , mTOR Associated Protein, LST8 Homolog/metabolism
6.
Clin Transl Oncol ; 21(10): 1364-1373, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30798512

ABSTRACT

PURPOSE: Patients with recurrent glioblastoma (rGBM) have a poor prognosis, with survival ranging from 25 to 40 weeks. Antiangiogenic agents are widely used, showing a variable response. In this study, we explored the efficacy of carmustine plus bevacizumab (BCNU/Bev) for treating rGBM. METHODS/PATIENTS: In this study, we assessed 59 adult patients with histologically confirmed rGBM who were treated with BCNU/Bev as second-line regimen. The response rate (RR), progression-free survival (PFS) and overall survival (OS) were evaluated according to their molecular expression profile, including CD133 mRNA expression, MGMT methylation (pMGMT), PDGFR amplification, YKL40 mRNA expression, IDH1/2 condition, p53 and EGFRvIII mutation status. RESULTS: Median follow-up was 18.6 months, overall RR to the combination was 56.3%, and median PFS was 9.0 months (95% CI 8.0-9.9). OS from time of diagnosis was 21.0 months (95% CI 13.2-28.7) and from starting BCNU/Bev it was 10.7 months (95% CI 9.5-11.8). IDH1/2 mutations were found in 30.5% of the patients, pMGMT in 55.9% and high CD133 mRNA expression in 57.6%. Factors which positively affected PFS included performance status (p = 0.015), IDH+ (p = 0.05), CD133 mRNA expression (p = 0.009) and pMGMT+ (p = 0.007). OS was positively affected by pMGMT+ (p = 0.05). Meanwhile, YKL40 negatively affected PFS (p = 0.01) and OS (p = 0.0001). Grade ≥ 3 toxicities included hypertension (22%) and fatigue (12%). CONCLUSIONS: BCNU/Bev is a safe and tolerable treatment for rGBM. Patients with MGMT+/IDH+ derive the greatest benefit from the treatment combination in the second-line setting. Nonetheless, high YKL40 expression discourages the use of antiangiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Brain Neoplasms/drug therapy , Carmustine/therapeutic use , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , AC133 Antigen/genetics , AC133 Antigen/metabolism , Adult , Aged , Angiogenesis Inhibitors/adverse effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Bevacizumab/adverse effects , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Carmustine/adverse effects , Chitinase-3-Like Protein 1/genetics , Colombia , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Drug Administration Schedule , Female , Genes, erbB-1 , Genes, p53 , Glioblastoma/blood supply , Glioblastoma/genetics , Glioblastoma/mortality , Humans , Isocitrate Dehydrogenase/genetics , Male , Methylation , Middle Aged , Mutation , Neoplasm Recurrence, Local/blood supply , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Progression-Free Survival , RNA, Messenger/metabolism , Receptors, Platelet-Derived Growth Factor/genetics , Survival Analysis , Tumor Suppressor Proteins/metabolism , Young Adult
7.
Stem Cell Res Ther ; 9(1): 310, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30413179

ABSTRACT

BACKGROUND: Previous studies have demonstrated remarkable tropism of mesenchymal stem cells (MSCs) toward malignant gliomas, making these cells a potential vehicle for delivery of therapeutic agents to disseminated glioblastoma (GBM) cells. However, the potential contribution of MSCs to tumor progression is a matter of concern. It has been suggested that CD133+ GBM stem cells secrete a variety of chemokines, including monocytes chemoattractant protein-1 (MCP-1/CCL2) and stromal cell-derived factor-1(SDF-1/CXCL12), which could act in this tropism. However, the role in the modulation of this tropism of the subpopulation of CD133+ cells, which initiate GBM and the mechanisms underlying the tropism of MSCs to CD133+ GBM cells and their effects on tumor development, remains poorly defined. METHODS/RESULTS: We found that isolated and cultured MSCs (human umbilical cord blood MSCs) express CCR2 and CXCR4, the respective receptors for MCP-1/CCL2 and SDF-1/CXCL12, and demonstrated, in vitro, that MCP-1/CCL2 and SDF-1/CXC12, secreted by CD133+ GBM cells from primary cell cultures, induce the migration of MSCs. In addition, we confirmed that after in vivo GBM tumor establishment, by stereotaxic implantation of the CD133+ GBM cells labeled with Qdots (705 nm), MSCs labeled with multimodal iron oxide nanoparticles (MION) conjugated to rhodamine-B (Rh-B) (MION-Rh), infused by caudal vein, were able to cross the blood-brain barrier of the animal and migrate to the tumor region. Evaluation GBM tumors histology showed that groups that received MSC demonstrated tumor development, glial invasiveness, and detection of a high number of cycling cells. CONCLUSIONS: Therefore, in this study, we validated the chemotactic effect of MCP-1/CCL2 and SDF-1/CXCL12 in mediating the migration of MSCs toward CD133+ GBM cells. However, we observed that, after infiltrating the tumor, MSCs promote tumor growth in vivo probably by release of exosomes. Thus, the use of these cells as a therapeutic carrier strategy to target GBM cells must be approached with caution.


Subject(s)
AC133 Antigen/metabolism , Brain Neoplasms/pathology , Glioblastoma/pathology , Mesenchymal Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tropism , Animals , Brain Neoplasms/ultrastructure , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Migration Assays , Cell Proliferation , Cell Separation , Chemokines/metabolism , Glioblastoma/ultrastructure , Humans , Immunophenotyping , Male , Mesenchymal Stem Cells/ultrastructure , Models, Biological , Neoplastic Stem Cells/ultrastructure , Quantum Dots/metabolism , Rats, Wistar , Receptors, Chemokine/metabolism , Spheroids, Cellular/pathology , Tumor Cells, Cultured
8.
Appl Radiat Isot ; 141: 156-161, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29452949

ABSTRACT

Glioblastoma contains self-renewing, tumorigenic cancer stem-like cells that contribute to tumor initiation and therapeutic resistance. The aim of this research was to estimate and compare the effectiveness ratio (α/ß) of stem-like cells and differentiated glioma cells derived from the U87MG glioblastoma cell line. Cell survival experiments were obtained in a dose range of 0-20 Gy (13.52 ± 0.09 Gy/h) as a hyperfractionationated accelerated radiotherapy scheme. Biochemical characterization of the post-irradiated cells was performed by flow cytometry analysis and the percentage of stem-like cells that resisted irradiation was determined by the CD133 expression. Results showed that U87MG stem-like cells are highly proliferative and more radioresistant than the U87MG adherent group (with a lesser stem-like character), this in association with the calculated α/ß ratio of 17 and 14.1, respectively.


Subject(s)
Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , AC133 Antigen/metabolism , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Radiation Tolerance , Spheroids, Cellular/pathology , Spheroids, Cellular/radiation effects , Tumor Microenvironment/radiation effects
9.
Neurobiol Dis ; 108: 13-28, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28743634

ABSTRACT

Diabetes mellitus (DM) is reaching epidemic conditions worldwide and increases the risk for cognition impairment and dementia. Here, we postulated that progenitors in adult neurogenic niches might be particularly vulnerable. Therefore, we evaluated the different components of the mouse subventricular zone (SVZ) during the first week after chemical induction of type 1 and type 2 diabetes-like (T1DM and T2DM) conditions. Surprisingly, only T2DM mice showed SVZ damage. The initial lesions were localized to ependymal cilia, which appeared disorientated and clumped together. In addition, they showed delocalization of the ciliary membrane protein prominin-1. Impairment of neuroprogenitor proliferation, neurogenic marker abnormalities and ectopic migration of neuroblasts were found at a later stage. To our knowledge, our data describe for the first time such an early impact of T2DM on the SVZ. This is consistent with clinical data indicating that brain damage in T2DM patients differs from that in T1DM patients.


Subject(s)
AC133 Antigen/metabolism , Cilia/physiology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Neurogenesis/physiology , Stem Cell Niche/physiology , AC133 Antigen/genetics , Animals , Cells, Cultured , Cerebral Ventricles , Cilia/pathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/pathology , Disease Progression , Ependyma/pathology , Ependyma/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Random Allocation
10.
Clin Transl Oncol ; 19(11): 1350-1357, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28600675

ABSTRACT

PURPOSE: Androgen receptor (AR) splice variant 7 (AR-V7) has been related with both a higher risk of prostate cancer (PC) progression and differential responsiveness to hormonal agents versus chemotherapy. The objective of this study was to investigate the feasibility of a novel capillary nano-immunoassay in assessing AR-V7 in plasma from PC patients. METHODS: Patients with either localized or advanced PC were included in the study. Assessment of AR-V7 in plasma was performed through a capillary nano-immunoassay platform. Correlation with clinical data, stem cell biomarkers (such as CD133+), AR amplification and PTEN status was identified. RESULTS: The study included 72 PC patients. AR-V7 signal was detected in 21 (29%) patients: 17 (81%) had a Gleason score ≥7, 15 (71%) castration-resistant prostate cancer (CRPC), 18 (86%) metastatic disease and PSA (median) high than AR-V7 negative (p < 0.05). CD133 was expressed in 69 (96%) patients. The median CD133+ expression in circulating tumor cells CTCs was higher among the 21 AR-V7 positive cases versus AR-V7 negative (7 vs. 3). Androgen Receptor and PTEN fluorescence in situ hybridization (FISH) on CD133+ captured cells were performed: 37 cases showed ≥four CD133+ CTCs, of which 81% showed an increased AR copy number. This percentage was similar in both AR-V7-positive and AR-V7-negative patients. A total of 68% of the cases showed deletion of PTEN: 70% were ARV-7 positive vs. 67%, which were AR-V7 negative. CONCLUSIONS: Assessing the presence of AR-V7 in plasma from PC patients is feasible by a novel capillary nano-immunoassay. AR-V7 was observed in 29% of the tumors and is more frequent in aggressive tumors.


Subject(s)
AC133 Antigen/metabolism , Alternative Splicing , Biomarkers, Tumor/blood , Neoplastic Cells, Circulating/metabolism , Prostatic Neoplasms/blood , Receptors, Androgen/genetics , Biomarkers, Tumor/genetics , Follow-Up Studies , Humans , Immunoassay , Male , Nanomedicine , Neoplastic Cells, Circulating/pathology , Pilot Projects , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
11.
Genet Mol Res ; 16(1)2017 Mar 30.
Article in English | MEDLINE | ID: mdl-28362995

ABSTRACT

MiR-200b, a member of the microRNA-200 family, has been identified to be capable of suppressing glioma cell growth through targeting CREB1 or CD133. However, whether miR-200b affects the biological behavior (proliferation, invasion, and migration) of glioma cells is poorly understood. The aim of this study was to evaluate the effect of miR-200b on the biological behavior of glioma cells in vitro. MiRNA-200b mimics, miRNA-200b inhibitor, and mimic control were transfected into conventionally cultured glioma U251 cells, followed by measuring the expression of miR-200b and CD133 in transfected cells by RT-PCR; effect of miR-200b on CD133 mRNA 3'-UTR luciferase activity by luciferase reporter assay; proliferation activity of transfected U251 cells by MTT method; and changes in U251 cell invasion and migration by Transwell method after transfection. Compared to that in the miRNA-200b inhibitor, mimic control, and blank control groups, miRNA-200b expression was significantly increased and CD133 mRNA expression was significantly decreased in the mimic miRNA-200b group in a time-dependent manner (P < 0.05). Meanwhile, dual luciferase reporter assay showed that miR-200b could inhibit CD133 activity through binding to the 3'-UTR of CD133 mRNA (P < 0.05). Furthermore, the proliferation activity and invasion and migration abilities of U251 cells transfected with miRNA-200b mimic were significantly decreased (P < 0.05). In conclusion, overexpression of miR-200b inhibited the proliferation, invasion, and migration of glioma cells possibly through targeting CD133.


Subject(s)
AC133 Antigen/genetics , Glioma/genetics , MicroRNAs/genetics , 3' Untranslated Regions , AC133 Antigen/metabolism , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Glioma/pathology , Humans , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Nervous System Neoplasms/genetics , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
12.
Article in English | MEDLINE | ID: mdl-27866975

ABSTRACT

OBJECTIVES: The aim of this study was to analyze the expression of CD24, CD44, CD133, ALDH1, CD29 (integrin-ß1), and Ki-67 in squamous cell carcinoma of the oral cavity and oropharynx. STUDY DESIGN: Fifty-two tumors and 21 metastatic lymph nodes were evaluated by using immunohistochemistry. RESULTS: Seven of 52 cases (13.5%) showed positive cytoplasmic staining of aldehyde dehydrogenase 1; integrin-ß1 was expressed in 45 of 50 cases (90%); 30 of 52 cases (57.7%) had positive membranous staining of CD44; CD24 was expressed in 44 of 50 cases (88%); and three of 52 cases (5.8%) stained positively for membranous CD133. Median proliferation rate, measured by Ki-67, was 37.1% for tumors. Five-year cancer-specific survival rates for the CD44-negative and CD44-positive groups were 74% and 38%, respectively, although this difference did not reach statistical significance (P = .052). CONCLUSIONS: Our study demonstrated the expression of putative stem cell markers in squamous cell carcinoma of the oral cavity and oropharynx, with participation of CD44-positive cells in association with poor survival outcome.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Mouth Neoplasms/metabolism , Oropharyngeal Neoplasms/metabolism , Stem Cells/metabolism , AC133 Antigen/metabolism , Aldehyde Dehydrogenase 1 Family , CD24 Antigen/metabolism , Carcinoma, Squamous Cell/pathology , Female , Humans , Hyaluronan Receptors/metabolism , Immunohistochemistry , Integrin beta1/metabolism , Isoenzymes/metabolism , Ki-67 Antigen/metabolism , Male , Middle Aged , Mouth Neoplasms/pathology , Neoplasm Staging , Oropharyngeal Neoplasms/pathology , Retinal Dehydrogenase/metabolism , Survival Rate
13.
Oncotarget ; 7(26): 40546-40557, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27244897

ABSTRACT

Glioblastoma is composed of dividing tumor cells, stromal cells and tumor initiating CD133+ cells. Recent reports have discussed the origin of the glioblastoma CD133+ cells and their function in the tumor microenvironment. The present work sought to investigate the multipotent and mesenchymal properties of primary highly purified human CD133+ glioblastoma-initiating cells. To accomplish this aim, we used the following approaches: i) generation of tumor subspheres of CD133+ selected cells from primary cell cultures of glioblastoma; ii) analysis of the expression of pluripotency stem cell markers and mesenchymal stem cell (MSC) markers in the CD133+ glioblastoma-initiating cells; iii) side-by-side ultrastructural characterization of the CD133+ glioblastoma cells, MSC and CD133+ hematopoietic stem cells isolated from human umbilical cord blood (UCB); iv) assessment of adipogenic differentiation of CD133+ glioblastoma cells to test their MSC-like in vitro differentiation ability; and v) use of an orthotopic glioblastoma xenograft model in the absence of immune suppression. We found that the CD133+ glioblastoma cells expressed both the pluripotency stem cell markers (Nanog, Mush-1 and SSEA-3) and MSC markers. In addition, the CD133+ cells were able to differentiate into adipocyte-like cells. Transmission electron microscopy (TEM) demonstrated that the CD133+ glioblastoma-initiating cells had ultrastructural features similar to those of undifferentiated MSCs. In addition, when administered in vivo to non-immunocompromised animals, the CD133+ cells were also able to mimic the phenotype of the original patient's tumor. In summary, we showed that the CD133+ glioblastoma cells express molecular signatures of MSCs, neural stem cells and pluripotent stem cells, thus possibly enabling differentiation into both neural and mesodermal cell types.


Subject(s)
AC133 Antigen/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/cytology , Adipocytes/cytology , Animals , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Line, Tumor , Fetal Blood/cytology , Humans , Immunophenotyping , Male , Mesenchymal Stem Cells/cytology , Microspheres , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL