Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
1.
Medicina (Kaunas) ; 60(6)2024 May 26.
Article in English | MEDLINE | ID: mdl-38929487

ABSTRACT

Background and Objectives: Lung adenocarcinoma is a leading cause of cancer-related mortality despite recent therapeutic advances. Cancer stem cells have gained increasing attention due to their ability to induce cancer cell proliferation through self-renewal and differentiation into multiple cell lineages. OCT4 and LIN28 (and their homologs A and B) have been identified as key regulators of pluripotency in mammalian embryonic (ES) and induced stem (IS) cells, and they are the crucial regulators of cancer progression. However, their exact role in lung adenocarcinoma has not yet been clarified. Materials and Methods: The aim of this study was to explore the role of the pluripotency factors OCT4 and LIN28 in a cohort of surgically resected human lung adenocarcinomas to reveal possible biomarkers for lung adenocarcinoma prognosis and potential therapeutic targets. The expressions of OCT4, LIN28A and LIN28B were analyzed in formalin-fixed, paraffin-embedded tissue samples from 96 patients with lung adenocarcinoma by immunohistochemistry. The results were analyzed with clinicopathologic parameters and were related to the prognosis of patients. Results: Higher OCT4 expression was related to an improved 5-year overall survival (OS) rate (p < 0.001). Nuclear LIN28B expression was lower in stage I and II tumors (p < 0.05) compared to advanced stage tumors. LIN28B cytoplasmic expression was associated with 5-year OS rates not only in univariate (p < 0.005), but also in multivariate analysis (where age, gender, histopathological subtype and stage were used as cofactors, p < 0.01 HR = 2.592). Patients with lower LIN28B expression showed improved 5-year OS rates compared to patients with increased LIN28B expression. Conclusions: Our findings indicate that OCT4 and LIN28B are implicated in lung adenocarcinoma progression and prognosis outcome; thus, they serve as promising prognostic biomarkers and putative therapeutic targets in lung adenocarcinomas.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Octamer Transcription Factor-3 , RNA-Binding Proteins , Humans , Octamer Transcription Factor-3/analysis , Octamer Transcription Factor-3/metabolism , Male , Female , RNA-Binding Proteins/analysis , Middle Aged , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/mortality , Aged , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Prognosis , Biomarkers, Tumor/analysis , Adult , Survival Analysis , Immunohistochemistry , Aged, 80 and over
2.
Pathol Res Pract ; 257: 155296, 2024 May.
Article in English | MEDLINE | ID: mdl-38615507

ABSTRACT

POU class 2 homeobox 3 (POU2F3)-positive small cell bladder carcinoma (SCBC) is an extremely rare entity, and its clinicopathologic features have not been fully described. Here, we investigated the clinicopathologic features of 4 cases of POU2F3-positive small cell bladder carcinoma (SCBC) and reviewed the literature. We collected 12 cases of SCBC from our departmental archives and detected the expression of POU2F3 by immunohistochemical (IHC) staining. Selected cases with or without POU2F3 expression were subjected to gene expression analysis between two different groups using DESeq2 software. We identified 4 POU2F3-positive SCBC patients, 2 males and 2 females, with a mean age of 77 years. Three patients had hematuria, and 1 patient had dysuria. Radiologic findings showed a bladder mass. Pathologic diagnosis showed that 3 cases were pure SCBC and 1 was mixed urothelial cancer (UC). Histopathologically, four POU2F3-positive SCBC tumors were composed of small round cells with sparse cytoplasm, the nuclei were salt-and-pepper-like or finely granular. Tumor cells showed characteristic cytoplasmic staining with punctate positive signals for cytokeratin. Syn and CD56 were diffusely positive in all the 4 patients. CgA was positive in only one patient. POU2F3-positive SCBC showed higher expression levels of POU2F3, HMGA2 and PLCG2 genes by RNA-Seq. Our data showed the specific clinicopathologic features of 4 rare POU2F3-positive SCBC cases, and the distinct molecular feature was observed between POU2F3-positive and negative SCBC in the limited number of cases.


Subject(s)
Biomarkers, Tumor , Carcinoma, Small Cell , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/genetics , Male , Female , Aged , Carcinoma, Small Cell/pathology , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/genetics , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Aged, 80 and over , Middle Aged , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/analysis
3.
Pathol Res Pract ; 227: 153637, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34619577

ABSTRACT

Lymphovascular invasion (LVI) is a relevant prognostic factor in germ cell tumors of the testis (GCTT) and it has been included in the AJCC staging system. Nevertheless, its histological assessment is challenging, with low/moderate interobserver agreement also among expert uropathologists. Few studies focused on the potential role of immunohistochemistry to solve this critical issue; as result, in current guidelines there is no indication for additional staining to detect this histological feature. In the present study, we investigated the detection of LVI invasion in a small cohort of GCTT with double staining for OCT4/CD34. Although our results need to be validated in larger case series with follow-up data, they suggest as OCT4/CD34 could be a useful tool for the histological assessment of these tumors, helping to identify some histological mimickers of LVI and modifying the pT/stage in a significant percentage of patients.


Subject(s)
Antigens, CD34/analysis , Biomarkers, Tumor/analysis , Carcinoma, Embryonal/chemistry , Immunohistochemistry , Lymphatic Vessels/chemistry , Neoplasms, Complex and Mixed/chemistry , Neoplasms, Germ Cell and Embryonal/chemistry , Octamer Transcription Factor-3/analysis , Seminoma/chemistry , Testicular Neoplasms/chemistry , Adult , Carcinoma, Embryonal/pathology , Humans , Lymphatic Vessels/pathology , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Neoplasms, Complex and Mixed/pathology , Neoplasms, Germ Cell and Embryonal/pathology , Predictive Value of Tests , Retrospective Studies , Seminoma/pathology , Testicular Neoplasms/pathology , Young Adult
4.
Anal Bioanal Chem ; 413(8): 2081-2089, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33655347

ABSTRACT

Human embryonic stem cells (hESCs) can be self-propagated indefinitely in culture while holding the capacity to generate almost all cell types. Although this powerful differentiation ability of hESCs has become a potential source of cell replacement therapies, application of stem cells in clinical practice relies heavily on the exquisite control of their developmental fate. In general, an essential first step in differentiation is to exit the pluripotent state, which is precariously balanced and depends on a variety of factors, mainly centering on the core transcriptional mechanism. To date, much evidence has indicated that transcription factors such as Sox2, Oct4, and Nanog control the self-renewal and pluripotency of hESCs. Their expression displays a restricted spatial-temporal pattern and their small changes in level can significantly affect directed differentiation and the cell type derived. So far, few assays have been developed to monitor this process. Herein, we provided a mass spectrometry (MS)-based approach for simultaneous and quantitative monitoring of these transcription factors, in an attempt to provide insight into their contributions in hESC differentiation.


Subject(s)
Human Embryonic Stem Cells/cytology , Transcription Factors/analysis , Cell Differentiation , Cell Line , Humans , Mass Spectrometry , Octamer Transcription Factor-3/analysis , Proteomics , SOXB1 Transcription Factors/analysis
5.
BMC Cancer ; 20(1): 521, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32503462

ABSTRACT

BACKGROUND: Octamer-binding transcription factor 4A (OCT4A) is essential for cell pluripotency and reprogramming both in humans and mice. To date, however, the function of human OCT4 in somatic and/or tumour tissues is largely unknown. METHODS: RT-PCR was used to identify full-length splice forms of OCT4 transcripts in normal and cancer cells. A FLAG-tagged OCT4 genomic transgene was used to identify OCT4-positive cancer cells. A potential role for OCT4 in somatic cancer cells was examined by cell ablation of OCT4-positive cells using promoter-driven diphtheria toxin A. OCT4 and secreted phosphoprotein 1 (SPP1) transcripts in early-stage lung adenocarcinoma tumours were analysed and compared with pathohistological features. RESULTS: The results show that, unlike in murine cells, OCT4A and OCT4B variants are transcribed in both human cancer cells and in adult tissues such as lung, kidney, uterus, breast, and eye. We found that OCT4A and SPP1C are co-expressed in highly aggressive human breast, endometrial, and lung adenocarcinoma cell lines, but not in mesothelial tumour cell lines. Ablation of OCT4-positive cells in lung adenocarcinoma cells significantly decreased cell migration and SPP1C mRNA levels. The OCT4A/SPP1C axis was found in primary, early-stage, lung adenocarcinoma tumours. CONCLUSIONS: Co-expression of OCT4 and SPP1 may correlate with cancer aggressiveness, and the OCT4A/SPP1C axis may help identify early-stage high-risk patients with lung adenocarcinoma. Contrary to the case in mice, our data strongly suggest a critical role for OCT4A and SPP1C in the development and progression of human epithelial cancers.


Subject(s)
Adenocarcinoma of Lung/pathology , Biomarkers, Tumor/metabolism , Lung Neoplasms/pathology , Octamer Transcription Factor-3/metabolism , Osteopontin/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/mortality , Adolescent , Adult , Aged , Animals , Biomarkers, Tumor/analysis , Cell Differentiation , Cell Line, Tumor , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Infant , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Mice , Middle Aged , Neoplasm Staging , Octamer Transcription Factor-3/analysis , Osteopontin/analysis , Prognosis , Protein Isoforms/analysis , Protein Isoforms/metabolism , Young Adult
6.
PLoS One ; 15(5): e0232934, 2020.
Article in English | MEDLINE | ID: mdl-32428045

ABSTRACT

AIMS: Much work has been done to find markers of cancer stem cells (CSCs) that distinguish them from the tumor bulk cells and normal cells. Recent CSC research has applied the induced pluripotent stem cell (iPSC) concept. In this study, we investigated the expression of a panel of iPSC markers in primary colon adenocarcinoma (CA)-derived cell lines. MATERIALS AND METHODS: Expression of iPSC markers by CA-derived primary cell lines was interrogated using immunocytochemistry, western blotting and RT-qPCR. The stem cell function of these cells was then assessed in vitro using differentiation and tumorsphere assays. RESULTS: Expression of iPSC markers OCT4, SOX2, NANOG, KLF4 and c-MYC was more widespread in high-grade CA (HGCA) cell lines than low-grade CA (LGCA) cell lines, as demonstrated by western blotting and RT-qPCR. These cells could be induced to differentiate down the three embryonic lineages. Cells derived from HGCA were more capable of forming tumorspheres than those derived from LGCA. EpCAM sorting revealed that a population enriched for EpCAMHigh cells formed larger tumorspheres than EpCAMLow cells. Pluripotency markers, SSEA4 and TRA-1-60, were co-expressed by a small subpopulation of cells that also co-expressed SOX2 in 75% and OCT4 in 50% of the cell lines. CONCLUSIONS: CA-derived primary cell lines contain tumorsphere-forming cells which express key pluripotency genes and can differentiate down 3 embryonic lineages, suggesting a pluripotent CSC-like phenotype. There appear to be two iPSC-like subpopulations, one with high EpCAM expression which forms larger tumorspheres than another with low EpCAM expression. Furthermore, these cells can be characterized based on iPSC marker expression, as we have previously demonstrated in the original CA tumor tissues.


Subject(s)
Adenocarcinoma/metabolism , Induced Pluripotent Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Biomarkers, Tumor/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cellular Reprogramming/genetics , Colon/cytology , Colon/metabolism , Colonic Neoplasms/metabolism , DNA-Binding Proteins/analysis , Genes, Homeobox , Genes, myc , Humans , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/analysis , Nanog Homeobox Protein/analysis , Octamer Transcription Factor-3/analysis , Primary Cell Culture , SOXB1 Transcription Factors/analysis , Transcription Factors/analysis
7.
Head Neck Pathol ; 14(3): 749-757, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31902091

ABSTRACT

The cancer stem cells deliver uncontrolled proliferative capacity within the tumor imparting to increasing size while epithelial mesenchymal transition adds to the invasive potential. Studies using specific markers elucidating the role of these phenomena may bring advancement in the targeted therapy of tumor. SOX2 and OCT4 are two among few stem cell markers indicative of proliferative potential and WNT5A is an epithelial mesenchymal transition marker indicative of invasive potential. We aimed to determine the association between expression of SOX2, OCT4 and WNT5A in oral epithelial dysplasia, oral squamous cell carcinoma and normal oral mucosa. 20 cases of oral squamous cell carcinoma, 20 cases of oral epithelial dysplasia (leukoplakia with dysplasia) and 25 normal oral mucosa tissues specimens were immunohistochemically stained to assess SOX2, OCT4 and WNT5A expression. SOX2 expression was higher in oral squamous cell carcinoma than in oral epithelial dysplasia and very low in normal oral mucosa. OCT4 was very low in oral squamous cell carcinoma and oral epithelial dysplasia when compared to SOX2, while negative in normal tissues. Co-expression of SOX2 and OCT4 showed statistically non-significant difference for tumor proliferation. WNT5A expression was found to be increasing from normal oral mucosa to oral epithelial dysplasia and oral squamous cell carcinoma. In conformity with present study, SOX2 itself can act as a potential marker for proliferation in tumor cells while OCT4 has non-significant role in regulation of tumor behavior in oral squamous cell carcinoma as well as in oral epithelial dysplasia. WNT5A can be a putative marker in studying invasive potential of oral squamous cell carcinoma.


Subject(s)
Biomarkers, Tumor/analysis , Mouth Mucosa/pathology , Mouth Neoplasms/pathology , Precancerous Conditions/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Adult , Aged , Female , Humans , Immunohistochemistry , Male , Middle Aged , Octamer Transcription Factor-3/analysis , Octamer Transcription Factor-3/biosynthesis , SOXB1 Transcription Factors/analysis , SOXB1 Transcription Factors/biosynthesis , Wnt-5a Protein/analysis , Wnt-5a Protein/biosynthesis
8.
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
9.
Int J Radiat Oncol Biol Phys ; 103(1): 161-168, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30213750

ABSTRACT

PURPOSE: Although cancer stem cells (CSCs) have been reported across solid tumors, there is a dearth of data regarding CSC and its impact on outcomes of cervical cancer. METHODS AND MATERIALS: From October 2013 to December 2015, patients with squamous cancer of the cervix (stage IB2-IVA) were included. Pretreatment and posttreatment biopsy was obtained and immunohistochemistry was performed for SOX-2, OCT-4, Nanog, CD44, and Podoplanin. All patients received concurrent radiation and brachytherapy to an equivalent dose of 80 to 84 Gy to point A with concurrent weekly cisplatin. Correlation of CSC expression was performed with known prognostic factors. The effect of stem cell expression on disease outcomes was tested within multivariate analysis. RESULTS: One hundred fifty patients were included. The median dose to point A was 83 Gy (46-89 Gy) and a median of 4 cycles (range, 0-6 cycles) of chemotherapy was administered. At baseline, moderate to strong immunohistochemical expression of SOX-2, OCT-4, Nanog, CD44, and Podoplanin was observed in 12.8%, 4.8%, 24.4%, 15.5%, and 1.3% of patients, respectively. At median follow-up of 30 months (range, 3-51 months), locoregional and distant relapse was observed in 12.2% and 23.1% of patients, of whom 4.7% had both local and distant relapse. The 3-year disease-free survival rate was 87%. On multivariate analysis, moderate to high CSC expression and CD44 low status (hazard ratio [HR] = 8.8; 95% confidence interval [CI], 1.0-77.2; P < .04) independently predicted for locoregional relapse-free survival. International Federation of Gynecology and Obstetrics stage (HR = 2.6; 95% CI, 1.3-5.4; P = .004) and presence of residual tumor after external radiation (HR = 3.5; 95% CI, 1.8-6.5; P = .0001) predicted for a detriment in disease-free survival. CONCLUSIONS: The presence of stem cell proteins and loss of CD44 independently predicts for reduced locoregional control in locally advanced cervical cancer. Further investigation into the interaction of stem cell and CD44 biology is warranted.


Subject(s)
Chemoradiotherapy , Hyaluronan Receptors/analysis , Neoplastic Stem Cells/chemistry , Uterine Cervical Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Octamer Transcription Factor-3/analysis , Prospective Studies , SOXB1 Transcription Factors/analysis , Translational Research, Biomedical , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/pathology
10.
Hum Pathol ; 82: 68-75, 2018 12.
Article in English | MEDLINE | ID: mdl-30067948

ABSTRACT

In the present study, we investigated protein expression of the transcription factors mammalian doublesex and mab-3 related transcription factor 1 (DMRT1), basic helix-loop-helix transcription factor-like 5 (TCLF5), and octamer-binding transcription factor 4 (OCT4) in normal human spermatogenesis, testicular mixed germ cell-sex cord stromal tumor (MGC-SCST), spermatocytic tumor, and seminoma. In normal human spermatogenesis, DMRT1 is expressed in the nuclei of spermatogonia but not in those of more mature germ cells. By way of contrast, TCLF5 is expressed in the nuclei of some clusters of primary spermatocytes that have entered meiosis 1, in secondary spermatocytes, and in round (early) spermatids in the seminiferous tubules of adults during the reproductive years. OCT4 is expressed in primordial germ cells but not in the seminiferous tubules of the normal adult testis during the reproductive years. DMRT1 is expressed in the germ cells of both testicular MGC-SCST and spermatocytic tumor, whereas TCLF5 is not expressed in either neoplasm. These low-grade neoplasms, however, differ histologically in that all the germ cell nuclei of testicular MGC-SCST resemble spermatogonia, whereas in spermatocytic tumor, the nuclei of the medium-sized and large cells resemble those of primary spermatocytes. Both neoplasms lack expression of OCT4. By way of contrast, in seminoma, a fully malignant testicular germ cell tumor, the germ cell nuclei express OCT4 but do not express either DMRT1 or TCLF5.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/analysis , Biomarkers, Tumor/analysis , Neoplasms, Germ Cell and Embryonal/chemistry , Octamer Transcription Factor-3/analysis , Seminiferous Tubules/chemistry , Seminoma/chemistry , Spermatocytes/chemistry , Testicular Neoplasms/chemistry , Transcription Factors/analysis , Biopsy , Cell Nucleus/chemistry , Cell Nucleus/pathology , Humans , Immunohistochemistry , Male , Neoplasms, Germ Cell and Embryonal/pathology , Seminiferous Tubules/pathology , Seminoma/pathology , Spermatocytes/pathology , Spermatogenesis , Testicular Neoplasms/pathology
11.
Histol Histopathol ; 33(8): 859-870, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29569698

ABSTRACT

Double primary endometrioid endometrial and ovarian carcinomas (DPEEOCs) are the most common multiple gynecological carcinomas. In recent years, gene sequential comparison analysis has strongly supported the opinion that sporadic double endometrioid endometrial and ovarian cancers (DEEOCs) are clonally related in both primary and metastatic tumors. In order to find more clonal evidence for DPEEOC, we investigated cancer stem cells (CSCs). SOX2 and OCT4 are two common factors in CSCs. MicroRNA (miRNA)-145, a small non-coding RNA, has effects in regulating gene expression and tumorigenesis in CSCs. The aim of this study was to assess the involvements of SOX2, OCT4, and miRNA-145 in the tumorigenesis of DPEEOCs. In our study, twenty DPEEOC patients were chosen. Metastatic DEEOCs and normal endometrial and ovarian tissues were also included. The expression of miRNA-145 was detected by real-time quantitative PCR. Immunohistochemical staining was used to measure the expression of OCT4 and SOX2. The results showed that miRNA-145 expression was lower in DPEEOC endometrial tissues and higher in DPEEOC ovarian tissues compared to the corresponding normal tissues. Both SOX2 and OCT4 were over-expressed in cancer tissues compared with that in normal tissues. MiRNA-145, SOX2, and OCT4 were expressed at similar levels in two cancer sites of a given DPEEOC or metastatic DEEOC sample. Besides, metastatic DEEOC sections expressed a higher level of SOX2 and OCT4 compared to the corresponding DPEEOC tissues. Together, these results support the clonality of DPEEOCs. Moreover, SOX2 and OCT4 may have some implication in DPEEOC and metastatic DEEOC diagnosis.


Subject(s)
Biomarkers, Tumor , Carcinoma, Endometrioid , Endometrial Neoplasms , MicroRNAs/genetics , Neoplasms, Multiple Primary , Octamer Transcription Factor-3/analysis , Ovarian Neoplasms , SOXB1 Transcription Factors/analysis , Adult , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Carcinoma, Endometrioid/chemistry , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/secondary , Endometrial Neoplasms/chemistry , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Neoplasm Staging , Neoplasms, Multiple Primary/chemistry , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/pathology , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
12.
Clin Transl Oncol ; 19(9): 1147-1153, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28374320

ABSTRACT

PURPOSE: Circulating tumor cells (CTCs) with epithelial-to-mesenchymal transition (EMT) phenotypes might be related to tumor progression while OCT4 expression is involved in tumor metastasis and poor prognosis. But the possible clinical significance of EMT phenotypes of CTCs from non-small-cell lung cancer (NSCLC) patients has still to be demonstrated. Furthermore, none has been investigated the expression of OCT4 in CTCs. We therefore identified the EMT phenotype-based subsets of CTCs and determined the OCT4 expression status of CTCs in NSCLC patients, to explore their possible clinical relevance. METHODS: 37 NSCLC patients and ten healthy volunteers were enrolled, respectively. The Canpatrol™ CTC enrichment technique was used to isolate and identify the EMT phenotype-based subsets of CTCs. OCT4 expression in each CTC was also determined. Results were correlated with patients' clinico-pathological features. RESULTS: CTCs were detected in 33 of 37 (89.2%) NSCLC patients, and no CTCs were identified in ten healthy volunteers. Three CTCs phenotypes, including epithelial, biophenotypic, and mesenchymal CTCs were identified based on the expression of EMT markers. Mesenchymal CTCs were more commonly found in patients with distant metastasis. Patients with distant metastasis tended to have a higher median CTCs number. OCT4-positive was observed in 21 of 28 (75.0%) patients. High expression of OCT4 tended to occur in advanced patients as well as in distant metastatic patients. CONCLUSIONS: The findings suggest that identification of CTCs by EMT markers as well as evaluation of OCT4 expression status by assessment of OCT4 expression in CTCs could serve as potential adjuncts for evaluating metastasis and prognosis in NSCLC patients.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Octamer Transcription Factor-3/biosynthesis , Adult , Aged , Carcinoma, Non-Small-Cell Lung/mortality , Epithelial-Mesenchymal Transition/physiology , Female , Humans , Lung Neoplasms/mortality , Male , Middle Aged , Octamer Transcription Factor-3/analysis , Prognosis
13.
J Gastroenterol Hepatol ; 32(3): 567-576, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28320060

ABSTRACT

BACKGROUND AND AIM: The role of octamer-binding transcription factor 4 (Oct4) has been implicated in the clinical prognosis of various kinds of digestive system cancers, but the results remain controversial. The purpose of this meta-analysis is to assess the potential role of Oct4 as a prognostic marker in digestive system tumors. METHODS: Relevant articles were retrieved from Pubmed, Web of Science, and Cochrane Library up to July 2016. The software Stata 12.0 was used to analyze the outcomes, including overall survival (OS), disease-free survival, recurrence-free survival, and clinicopathological characteristics. RESULTS: A total of 13 eligible studies with 1538 patients were included. Elevated Oct4 expression was significantly associated with poor OS (pooled hazard ratio [HR] = 2.183, 95% confidence interval [CI]: 1.824-2.612), disease-free survival (pooled HR = 1.973, 95% CI: 1.538-2.532), and recurrence-free survival (pooled HR = 2.209, 95% CI: 1.461-3.338) of digestive system malignancies. Subgroup analyses showed that cancer type, sample size, study quality, and laboratory detection method did not alter the significant prognostic value of Oct4. Additionally, Oct4 expression was found to be an independent predictive factor for OS (HR = 2.068, 95% CI: 1.633-2.619). No significant association was found between Oct4 and clinicopathological features of digestive system malignancies. CONCLUSION: This study provided evidence of Oct4 and/or its closely related homolog protein as a predictive factor for patients with digestive system cancers. More large-scale clinical studies on the prognostic value of Oct4 are warranted.


Subject(s)
Biomarkers, Tumor/analysis , Digestive System Neoplasms/diagnosis , Octamer Transcription Factor-3/analysis , Databases, Bibliographic , Digestive System Neoplasms/mortality , Gene Expression , Humans , Octamer Transcription Factor-3/genetics , Predictive Value of Tests , Prognosis , Survival Rate
14.
Stem Cell Reports ; 8(4): 1005-1017, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28285878

ABSTRACT

Malignant mesothelioma (MM) is an aggressive neoplasm characterized by a poor patient survival rate, because of rapid tumor recurrence following first-line therapy. Cancer stem cells (CSCs) are assumed to be responsible for initiating tumorigenesis and driving relapse after therapeutic interventions. CSC-enriched MM cell subpopulations were identified by an OCT4/SOX2 reporter approach and were characterized by (1) increased resistance to cisplatin, (2) increased sensitivity toward the FAK inhibitor VS-6063 in vitro, and (3) a higher tumor-initiating capacity in vivo in orthotopic xenograft and allograft mouse models. Overexpression of NF2 (neurofibromatosis 2, merlin), a tumor suppressor often mutated or lost in MM, did not affect proliferation and viability of CSC-enriched MM populations but robustly decreased the viability of reporter-negative cells. In contrast, downregulation of calretinin strongly decreased proliferation and viability of both populations. In summary, we have enriched and characterized a small MM cell subpopulation that bears the expected CSC characteristics.


Subject(s)
Lung Neoplasms/pathology , Lung/pathology , Mesothelioma/pathology , Neoplastic Stem Cells/pathology , Stem Cell Factor/analysis , Animals , Antineoplastic Agents/pharmacology , Calbindin 2/analysis , Cell Proliferation , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Humans , Lung/drug effects , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Mesothelioma, Malignant , Mice , Neoplastic Stem Cells/drug effects , Neurofibromin 2/analysis , Octamer Transcription Factor-3/analysis , SOXB1 Transcription Factors/analysis
15.
Braz Oral Res ; 31: e17, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28146221

ABSTRACT

Periodontitis develops as a result of a continuous interaction between host cells and subgingival pathogenic bacteria. The periodontium has a limited capacity for regeneration, probably due to changes in periodontal ligament stem cells (PDLSCs) phenotype. The aim of this study was to evaluate the effects of lipopolysaccharides from Porphyromonas gingivalis (PgLPS) on mesenchymal phenotype and osteoblast/cementoblast (O/C) potential of PDLSCs. PDLSCs were assessed for Toll-like receptor 2 (TLR2) expression by immunostaining technique. After, cells were exposed to PgLPS, and the following assays were carried out: (i) cell metabolic activity using MTS; (ii) gene expression for IL-1ß, TNF-α and OCT-4 by real-time polymerase chain reaction (RT-qPCR); (iii) flow cytometry for STRO-1 and CD105, and (iv) osteogenic differentiation. PDLSCs were positive for TLR2. PgLPS promoted cell proliferation, produced IL-1ß and TNF-α, and did not affect the expression of stem cell markers, STRO-1, CD105 and OCT-4. Under osteogenic condition, PDLSCs exposed to PgLPS showed a similar potential to differentiate toward osteoblast/cementoblast phenotype compared to control group as revealed by mineralized matrix deposition and levels of transcripts for RUNX2, ALP and OCN. These results provide evidence that PgLPS induces pro-inflammatory cytokines, but does not change the mesenchymal phenotype and osteoblast/cementoblast differentiation potential of PDLSCs.


Subject(s)
Lipopolysaccharides/toxicity , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Periodontal Ligament/cytology , Porphyromonas gingivalis , Alkaline Phosphatase/analysis , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/analysis , Flow Cytometry , Gene Expression , Humans , Interleukin-1beta/analysis , Mesenchymal Stem Cells/metabolism , Octamer Transcription Factor-3/analysis , Osteocalcin/analysis , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric , Time Factors , Toll-Like Receptors/analysis , Tumor Necrosis Factor-alpha/analysis
16.
Anticancer Agents Med Chem ; 17(7): 961-965, 2017.
Article in English | MEDLINE | ID: mdl-27671309

ABSTRACT

BACKGROUND: The high rates of women&#039;s death from breast cancer occur due to acquired resistance by patients to certain treatments, enabling the recurrence and/or tumor growth, invasion and metastasis. It has been demonstrated that the presence of cancer stem cells in human tumors, as responsible for recurrence and resistance to therapy. Studies have identified OCT4 as responsible for self-renewal and maintenance of pluripotency of stem cells. Thus, it is interesting to study potential drugs that target this specific population in breast cancer. Melatonin, appears to have oncostatic effects on cancer cells, however, little is known about its therapeutic effect on cancer stem cells. OBJECTIVE: Evaluate the viability and the expression of OCT4 in breast cancer stem cells, MCF-7 and MDA-MB- 231, after melatonin treatment. METHOD: The cells were grown in a 3-dimensional model of mammospheres, representing the breast cancer stem cell population and treated or not with melatonin. The cell viability of mammospheres were evaluated by MTT assay and the OCT4 expression, a cancer stem cells marker, was verified by immunocitochemistry. RESULTS: Our results demonstrated that the melatonin treatment decreased the cell viability of MCF-7 and MDAMB- 231 mammospheres. Furthermore, it was observed that in both cell lines, the expression of OCT4 was decreased in melatonin-treated cells compared to the control group. CONCLUSIONS: This fact suggests that melatonin is effective against breast cancer stem cells inhibiting the cell viability via OCT 4. Based on that, we believe that melatonin has a high potential to be used as an alternative treatment for breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast/drug effects , Cell Survival/drug effects , Melatonin/pharmacology , Neoplastic Stem Cells/drug effects , Breast/cytology , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/analysis
17.
Braz. oral res. (Online) ; 31: e17, 2017. tab, graf
Article in English | LILACS | ID: biblio-839523

ABSTRACT

Abstract Periodontitis develops as a result of a continuous interaction between host cells and subgingival pathogenic bacteria. The periodontium has a limited capacity for regeneration, probably due to changes in periodontal ligament stem cells (PDLSCs) phenotype. The aim of this study was to evaluate the effects of lipopolysaccharides from Porphyromonas gingivalis (PgLPS) on mesenchymal phenotype and osteoblast/cementoblast (O/C) potential of PDLSCs. PDLSCs were assessed for Toll-like receptor 2 (TLR2) expression by immunostaining technique. After, cells were exposed to PgLPS, and the following assays were carried out: (i) cell metabolic activity using MTS; (ii) gene expression for IL-1β, TNF-α and OCT-4 by real-time polymerase chain reaction (RT-qPCR); (iii) flow cytometry for STRO-1 and CD105, and (iv) osteogenic differentiation. PDLSCs were positive for TLR2. PgLPS promoted cell proliferation, produced IL-1β and TNF-α, and did not affect the expression of stem cell markers, STRO-1, CD105 and OCT-4. Under osteogenic condition, PDLSCs exposed to PgLPS showed a similar potential to differentiate toward osteoblast/cementoblast phenotype compared to control group as revealed by mineralized matrix deposition and levels of transcripts for RUNX2, ALP and OCN. These results provide evidence that PgLPS induces pro-inflammatory cytokines, but does not change the mesenchymal phenotype and osteoblast/cementoblast differentiation potential of PDLSCs.


Subject(s)
Humans , Osteogenesis/drug effects , Periodontal Ligament/cytology , Lipopolysaccharides/toxicity , Porphyromonas gingivalis , Mesenchymal Stem Cells/drug effects , Time Factors , Gene Expression , Osteocalcin/analysis , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Tumor Necrosis Factor-alpha/analysis , Statistics, Nonparametric , Cell Proliferation/drug effects , Alkaline Phosphatase/analysis , Octamer Transcription Factor-3/analysis , Toll-Like Receptors/analysis , Core Binding Factor Alpha 1 Subunit/analysis , Interleukin-1beta/analysis , Mesenchymal Stem Cells/metabolism , Real-Time Polymerase Chain Reaction , Flow Cytometry
18.
Elife ; 52016 09 29.
Article in English | MEDLINE | ID: mdl-27684073

ABSTRACT

Formation of the three embryonic germ layers is a fundamental developmental process that initiates differentiation. How the zebrafish pluripotency factor Pou5f3 (homologous to mammalian Oct4) drives lineage commitment is unclear. Here, we introduce fluorescence lifetime imaging microscopy and fluorescence correlation spectroscopy to assess the formation of Pou5f3 complexes with other transcription factors in real-time in gastrulating zebrafish embryos. We show, at single-cell resolution in vivo, that Pou5f3 complexes with Nanog to pattern mesendoderm differentiation at the blastula stage. Later, during gastrulation, Sox32 restricts Pou5f3-Nanog complexes to the ventrolateral mesendoderm by binding Pou5f3 or Nanog in prospective dorsal endoderm. In the ventrolateral endoderm, the Elabela / Aplnr pathway limits Sox32 levels, allowing the formation of Pou5f3-Nanog complexes and the activation of downstream BMP signaling. This quantitative model shows that a balance in the spatiotemporal distribution of Pou5f3-Nanog complexes, modulated by Sox32, regulates mesendoderm specification along the dorsoventral axis.


Subject(s)
Mesoderm/embryology , Nanog Homeobox Protein/analysis , Octamer Transcription Factor-3/analysis , Zebrafish Proteins/analysis , Zebrafish/embryology , Animals , Intravital Microscopy , Mesoderm/chemistry , Microscopy, Fluorescence , Protein Binding , Spatio-Temporal Analysis , Spectrometry, Fluorescence
19.
Proteomics ; 16(20): 2605-2614, 2016 10.
Article in English | MEDLINE | ID: mdl-27392809

ABSTRACT

We present a fully defined culture system (adapted Essential8TM [E8TM ] medium in combination with vitronectin) for human embryonic stem cells that can be used for SILAC purposes. Although a complete incorporation of the labels was observed after 4 days in culture, over 90% of precursors showed at least 10% conversion. To reduce this arginine conversion, E8TM medium was modified by adding (1) l-proline, (2) l-ornithine, (3) Nω -hydroxy-nor-l-arginine acetate, or by (4) lowering the arginine concentration. Reduction of arginine conversion was best obtained by adding 5 mM l-ornithine, followed by 3.5 mM l-proline and by lowering the arginine concentration in the medium to 99.5 µM. No major changes in pluripotency and cell amount could be observed for the adapted E8TM media with ornithine and proline. However, our subsequent ion mobility assisted data-independent acquisition (high-definition MS) proteome analysis cautions for ongoing changes in the proteome when aiming at longer term suppression of arginine conversion.


Subject(s)
Arginine/metabolism , Culture Media/metabolism , Human Embryonic Stem Cells/metabolism , Proteome/analysis , Proteomics/methods , Arginine/analysis , Cell Count , Cell Culture Techniques/methods , Cell Line , Culture Media/chemistry , Human Embryonic Stem Cells/chemistry , Human Embryonic Stem Cells/cytology , Humans , Isotope Labeling/methods , Octamer Transcription Factor-3/analysis , Octamer Transcription Factor-3/metabolism , Proteome/metabolism , Tandem Mass Spectrometry/methods
20.
Am J Surg Pathol ; 40(7): 950-7, 2016 07.
Article in English | MEDLINE | ID: mdl-27035612

ABSTRACT

OCT4 and SALL4 are transcription factors within a complex network that functions to maintain pluripotency in primitive stem cells and germ cells. Nuclear expression of OCT4 is widely cited as sensitive and specific for primary and metastatic germ cell tumors and is commonly used in the diagnosis of central nervous system (CNS) germinomas. Studies have failed to systematically examine the expression of OCT4 or SALL4 in diffuse large B-cell lymphoma (DLBCL), although this entity enters the morphologic differential diagnosis of some germ cell tumors. A retrospective review was conducted on 145 consecutive cases of DLBCL and testicular lymphoma to evaluate the prevalence of OCT4 and SALL4 expression. Nuclear OCT4 expression was present in 2/11 (18%) testicular DLBCLs and 6/134 (4.5%) nontesticular DLBCLs. Most OCT4 cases demonstrated moderate to strong expression in >50% of neoplastic cells. Rare, weak nuclear SALL4 expression was detected in only 3 nontesticular DLBCLs. Within the extratesticular DLBCL group, 2/6 (33%) primary CNS DLBCLs expressed nuclear OCT4. In addition, OCT4 DLBCL showed an overall predilection toward non-germinal center B-cell phenotype (7/8; 88%) and had a higher than expected rate of CD5 coexpression (4/8, 50%). These results are cautionary against using OCT4 as a sole marker of germ cell differentiation in testicular and extratesticular sites, especially in the CNS. The apparent associations of OCT4 expression with primary CNS DLBCL, non-germinal center B-cell phenotype, and CD5 coexpression raise the question of whether OCT4 expression in DLBCL may reflect more aggressive biology.


Subject(s)
Biomarkers, Tumor/analysis , Lymphoma, Large B-Cell, Diffuse/diagnosis , Neoplasms, Germ Cell and Embryonal/diagnosis , Octamer Transcription Factor-3/biosynthesis , Testicular Neoplasms/diagnosis , Transcription Factors/analysis , Transcription Factors/biosynthesis , Adult , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry , Male , Middle Aged , Octamer Transcription Factor-3/analysis , Retrospective Studies , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL