Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 11261, 2024 05 17.
Article in English | MEDLINE | ID: mdl-38760405

ABSTRACT

Here, we focused on the role of Nucleobindin 2 (NUCB2), a multifunctional protein, in gastric carcinoma (GC) progression. NUCB2 expression was investigated in 150 GC cases (20 non-invasive (pT1) and 130 invasive (pT2/pT3/pT4) tumors) by immunohistochemistry (IHC), and in situ hybridization for detection of the mRNA in 21 cases. Using GC cell lines, we determined whether NUCB2 expression was associated with specific cellular phenotypes. In GC clinical samples, NUCB2 was transcriptionally upregulated when compared to normal tissues. High NUCB2 expression was associated with clinicopathological factors including deep tumor invasion, lymphovascular invasion, lymph node metastasis, and advanced clinical stages, and was a significant independent predictor of unfavorable progression-free survival in 150 non-invasive and invasive GC patients. Similar findings were also evident in 72 invasive GC cases in which patients received post-operative chemotherapy, but not in 58 invasive tumors from patients who did not receive the chemotherapy. In cell lines, NUCB2 knockout inhibited proliferation, susceptibility to apoptosis, and migration capability by inducting cellular senescence; this was consistent with higher proliferation and apoptotic indices in the NUCB2 IHC-high compared to NUCB2 IHC-low GC cases. NUCB2-dependent inhibition of senescence in GC engenders aggressive tumor behavior by modulating proliferation, apoptosis, and migration.


Subject(s)
Cellular Senescence , Nucleobindins , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Nucleobindins/metabolism , Female , Male , Cell Line, Tumor , Middle Aged , Aged , Cell Proliferation , Gene Expression Regulation, Neoplastic , Apoptosis , Cell Movement , Prognosis
2.
Am J Pathol ; 194(3): 459-470, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38096983

ABSTRACT

Notch signaling contributes to tissue development and homeostasis, but little is known about its role in morular differentiation of endometrial carcinoma (Em Ca) cells. The current study focused on crosstalk between Notch and ß-catenin signaling in Em Ca with morules. Promoters of hairy and enhancer of split 1 (Hes1) and mastermind-like 2 (MAML2) were activated by Notch intracellular domain 1 but not ß-catenin, and a positive feedback loop between Hes1 and MAML2 was observed. Immunoreactivities for nuclear ß-catenin, Hes1, and MAML2, as well as the interaction between ß-catenin and Hes1 or MAML2, were significantly higher in morular lesions compared with surrounding carcinoma in Em Ca. Inhibition of glycogen synthase kinase-3ß (GSK-3ß) increased expression of total nuclear and cytoplasmic GSK-3ß and its phosphorylated forms, as well as Notch intracellular domain 1, Hes1, and active ß-catenin. GSK-3ß inhibition also decreased proliferation and migration, consistent with the response of cells stably overexpressing Hes1. Finally, the nuclear/cytoplasmic GSK-3ß score was significantly higher in morules compared with surrounding carcinoma in Em Ca, and it was positively correlated with nuclear ß-catenin, Hes1, and MAML2 scores. This complex interplay between Notch effectors and ß-catenin signaling through GSK-3ß inhibition contributes to the establishment and maintenance of ß-catenin-mediated morular differentiation, which is, in turn, associated with reduced proliferation and inhibition of migration in Em Ca.


Subject(s)
Carcinoma , Endometrial Neoplasms , Female , Humans , beta Catenin/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Endometrial Neoplasms/pathology , Signal Transduction/physiology
3.
BMC Cancer ; 23(1): 765, 2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37592266

ABSTRACT

BACKGROUND: Although anaplastic lymphoma kinase (ALK) is overexpressed in several primary solid tumor types, its role in endometrial carcinoma (Em Ca) remains unclear. METHODS: We evaluated expression of ALK and its related molecules in clinical samples consisting of 168 Em Ca tissues. We also used Em Ca cell lines to evaluate the functional role of ALK. RESULTS: Cytoplasmic ALK immunoreactivity in the absence of chromosomal rearrangement was positively correlated with ALK mRNA expression, and was significantly higher in Grade (G) 3 Em Ca than in G1 or G2 tumors. ALK immunoreactivity was also significantly associated with expression of cancer stem cell (CSC)-related molecules (cytoplasmic CD133, ALDH1, Sox2) and neuroendocrine markers (CD56 and synaptophysin). Although the proliferative index was significantly higher in ALK-positive Em Ca when compared to ALK- negative malignancies, there was no association between ALK expression and other clinicopathological factors in this disease. In Em Ca cell lines, full-length ALK overexpression increased proliferation, decreased susceptibility to apoptosis, enhanced cancer stem cell features, and accelerated cell mobility, whereas these phenotypes were abrogated in ALK-knockdown cells. Finally, patients with tumors harboring either wild-type ALK or high ALK mRNA expression had a poorer prognosis than those with either mutant ALK or low ALK mRNA expression. CONCLUSION: Full-length ALK overexpression occurs in a subset of Em Ca, particularly in G3 tumors, and contributes to the establishment and maintenance of aggressive phenotypic characteristics through modulation of several biological processes.


Subject(s)
Anaplastic Lymphoma Kinase , Endometrial Neoplasms , Female , Humans , Anaplastic Lymphoma Kinase/genetics , Cytoplasm , Endometrial Neoplasms/genetics , Phenotype , RNA, Messenger
4.
Mol Oncol ; 17(10): 2168-2182, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37539980

ABSTRACT

Ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50) is a scaffold protein that is required for epithelial polarity. Knockout (KO) of membranous EBP50 (Me-EBP50) in ovarian clear cell carcinoma (OCCC) cells induced an epithelial-mesenchymal transition (EMT)-like phenotype, along with decreased proliferation, accelerated migration capability, and induction of cancer stem cell (CSC)-like properties. Shotgun proteomics analysis of proteins that co-immunoprecipitated with EBP50 revealed that Me-EBP50 strongly interacts with myosin 9 (MYH9). Specific inhibition of MYH9 with blebbistatin phenocopied Me-EBP50 KO, and blebbistatin treatment potentiated the effects of Me-EBP50 KO. In OCCC cells from clinical samples, Me-EBP50 and MYH9 were co-localized at the apical plasma membrane. Patients with a combination of Me-EBP50-high and MYH9-high scores had the best prognosis for overall and progression-free survival. Our data suggest that Me-EBP50 has tumor-suppressive effects through the establishment and maintenance of epithelial polarization. By contrast, loss of Me-EBP50 expression induces EMT-like phenotypes, probably due to MYH9 dysfunction; this results in increased cell mobility and enhanced CSC-like properties, which in turn promote OCCC progression.

5.
Am J Pathol ; 193(8): 1116-1128, 2023 08.
Article in English | MEDLINE | ID: mdl-37169340

ABSTRACT

Epithelial-mesenchymal transition is a hallmark of uterine carcinosarcoma (UCS). Here, shotgun proteomics analysis used to identify biomarkers associated with blebbistatin-mediated epithelial-mesenchymal transition in UCS indicated up-regulation of nucleobindin-2 (NUCB2) in endometrial carcinoma (Em Ca) cells. Expression of N-cadherin, Snail, Slug, and ZEB1 was reduced in NUCB2 knockout Em Ca cells, whereas ZEB1, Twist1, and vimentin were up-regulated in NUCB2-overexpressing Em Ca cells. NUCB2 knockout reduced cell proliferation and migration, whereas NUCB2 overexpression had the opposite effect. Treatment of Em Ca cells with transforming growth factor (TGF)-ß1 dramatically altered morphology toward a fibroblastic appearance; concomitantly, expression of NUCB2 and ZEB1 increased. The NUCB2 promoter was also activated by transfection of Smad2. In UCS tissues, NUCB2 expression was significantly higher in sarcomatous compared with carcinomatous components, which was consistent with increased TGF-ß1 mRNA expression in stromal and sarcomatous components compared with carcinomatous components. In addition, NUCB2 score correlated positively with ZEB1 and vimentin scores, whereas ZEB1 score correlated positively with Slug and vimentin scores and inversely with the E-cadherin score. Collectively, these data indicate that TGF-ß-dependent up-regulation of NUCB2 and ZEB1 contributes to the phenotypic characteristics of sarcomatous components in UCS.


Subject(s)
Carcinosarcoma , Uterine Neoplasms , Humans , Female , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Nucleobindins/genetics , Nucleobindins/metabolism , Transforming Growth Factor beta1/metabolism , Vimentin/metabolism , Genes, Homeobox , Cadherins/genetics , Cadherins/metabolism , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Phenotype , Carcinosarcoma/genetics , Carcinosarcoma/pathology , Zinc Fingers , Epithelial-Mesenchymal Transition/genetics , Cell Line, Tumor
6.
Cancers (Basel) ; 16(1)2023 Dec 29.
Article in English | MEDLINE | ID: mdl-38201610

ABSTRACT

Ezin-radixin-moesin-binding phosphoprotein 50 (EBP50) is a scaffold protein that interacts with several partner molecules including ß-catenin. Here, we examined the crosstalk between EBP50 and nuclear catenin during colorectal carcinoma (CRC) progression. In clinical samples, there were no correlations between the subcellular location of EBP50 and any clinicopathological factors. However, EBP50 expression was significantly lower specifically in the outer areas of tumor lesions, in regions where tumor budding (BD) was observed. Low EBP50 expression was also significantly associated with several unfavorable prognostic factors, suggesting that EBP50 depletion rather than its overexpression or subcellular distribution plays an important role in CRC progression. In CRC cell lines, knockout of EBP50 induced epithelial-mesenchymal transition (EMT)-like features, decreased proliferation, accelerated migration capability, and stabilized nuclear ß-catenin due to disruption of the interaction between EBP50 and ß-catenin at the plasma membrane. In addition, Slug expression was significantly higher in outer lesions, particularly in BD areas, and was positively correlated with nuclear ß-catenin status, consistent with ß-catenin-driven transactivation of the Slug promoter. Together, our data suggest that EBP50 depletion releases ß-catenin from the plasma membrane in outer tumor lesions, allowing ß-catenin to accumulate and translocate to the nucleus, where it transactivates the Slug gene to promote EMT. This in turn triggers tumor budding and contributes to the progression of CRC to a more aggressive phase.

7.
Cell Commun Signal ; 20(1): 181, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36411429

ABSTRACT

BACKGROUND: Although a lack of functional PTEN contributes to tumorigenesis in a wide spectrum of human malignancies, little is known about the functional role of its overexpression in the tumors. The current study focused on PTEN overexpression in endometrial carcinoma (Em Ca). METHODS: The functional impact of PTEN overexpression was assessed by Em Ca cell lines. Immunohistochemical analyses were also conducted using 38 Em Ca with morular lesions. RESULTS: Em Ca cell lines stably overexpressing PTEN (H6-PTEN) exhibited epithelial-mesenchymal transition (EMT)-like features, probably through ß-catenin/Slug-meditated suppression of E-cadherin. PTEN overexpression also inhibited cell proliferation, accelerated cellular senescence, increased apoptotic features, and enhanced migration capability. Moreover, H6-PTEN cells exhibited cancer stem cell (CSC)-like properties, along with high expression of aldehyde dehydrogenase 1 and CD44s, a large ALDH 1high population, enriched spheroid formation, and ß-catenin-mediated upregulation of cyclin D2, which is required for persistent CSC growth. In clinical samples, immunoreactivities for PTEN, as well as CSC-related molecules, were significantly higher in morular lesions as compared to the surrounding carcinomas. PTEN score was positively correlated with expression of nuclear ß-catenin, cytoplasmic CD133, and CD44v6, and negatively with cell proliferation. Finally, estrogen receptor-α (ERα)-dependent expression of Ezrin-radixin-moesin-binding phophoprotein-50 (EBP50), a multifunctional scaffolding protein, acts as a negative regulator of morular formation by Em Ca cells through interacting with PTEN and ß-catenin. CONCLUSION: In the abscess of ERα/EBP50 expression, PTEN overexpression and nuclear ß-catenin stabilization promote the establishment and maintenance of morular phenotype associated with EMT/CSC-like features in Em Ca cells. Video Abstract.


Subject(s)
Endometrial Neoplasms , PTEN Phosphohydrolase , Animals , Female , Humans , beta Catenin , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Epithelial-Mesenchymal Transition , Estrogen Receptor alpha , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
8.
J Pathol Clin Res ; 8(5): 458-469, 2022 09.
Article in English | MEDLINE | ID: mdl-35762092

ABSTRACT

Programmed cell death-1 (PD-1) and its ligand (PD-L1) are significant mediators of immune suppression in the tumor microenvironment. We focused on the immunological impact of PD-1/PD-L1 signaling during tumor progression in colorectal carcinoma (CRC) and its association with resistance to neoadjuvant chemoradiotherapy (NCRT) in locally advanced rectal carcinoma (LAd-RC). Histopathological and immunohistochemical analyses of 100 CRC cases (including 34 RC) without NCRT and 109 NCRT-treated LAd-RC cases were performed. Membranous tumoral PD-L1 expression was identified in 9 of 100 (9%) CRC cases, including 1 of 34 (2.9%) RC cases, but PD-L1 immunopositivity was not associated with any clinicopathological factors, with the exception of deficient mismatch repair (dMMR) status. In contrast, stromal PD-L1+ immune cells, which frequently exhibited coexpression of PD-1 and CD8 markers, were significantly correlated with tumor vessel invasion, nuclear ß-catenin+ tumor budding cancer stem cell (CSC)-like features, and unfavorable prognosis. In the LAd-RC cases, stromal CD8+ (but not PD-L1+) immune cell infiltration in pretreatment-biopsied samples was significantly and positively associated with therapeutic efficacy. After NCRT, tumoral PD-L1 expression was observed in only 2 of 83 (2.4%) tumors, independent of dMMR status, whereas high stromal PD-L1+ and tumoral nuclear ß-catenin positivity were significantly linked to a poor response to NCRT and high tumor budding features. In addition, high stromal PD-L1 immunoreactivity was significantly associated with poorer overall survival. In conclusion, a combination of stromal PD-L1+ immune cells and nuclear ß-catenin+ tumor budding may contribute to tumor progression in CRC and resistance to NCRT in LAd-RC, through formation of niche-like lesions that exhibit immune resistance and CSC properties.


Subject(s)
B7-H1 Antigen , Drug Resistance, Neoplasm , Programmed Cell Death 1 Receptor , Radiation Tolerance , Rectal Neoplasms , beta Catenin , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell Nucleus/genetics , Cell Nucleus/immunology , Chemoradiotherapy, Adjuvant , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Disease Progression , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Neoadjuvant Therapy , Prognosis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Radiation Tolerance/genetics , Radiation Tolerance/immunology , Rectal Neoplasms/genetics , Rectal Neoplasms/immunology , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , beta Catenin/genetics , beta Catenin/immunology
9.
Cell Commun Signal ; 20(1): 46, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35392912

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most aggressive form of brain tumor and has vascular-rich features. The S100A4/non-muscle myosin IIA (NMIIA) axis contributes to aggressive phenotypes in a variety of human malignancies, but little is known about its involvement in GBM tumorigenesis. Herein, we examined the role of the S100A4/NMIIA axis during tumor progression and vasculogenesis in GBM. METHODS: We performed immunohistochemistry for S100A4, NMIIA, and two hypoxic markers, hypoxia-inducible factor-1α (HIF-1α) and carbonic anhydrase 9 (CA9), in samples from 94 GBM cases. The functional impact of S100A4 knockdown and hypoxia were also assessed using a GBM cell line. RESULTS: In clinical GBM samples, overexpression of S100A4 and NMIIA was observed in both non-pseudopalisading (Ps) and Ps (-associated) perinecrotic lesions, consistent with stabilization of HIF-1α and CA9. CD34(+) microvascular densities (MVDs) and the interaction of S100A4 and NMIIA were significantly higher in non-Ps perinecrotic lesions compared to those in Ps perinecrotic areas. In non-Ps perinecrotic lesions, S100A4(+)/HIF-1α(-) GBM cells were recruited to the surface of preexisting host vessels in the vascular-rich areas. Elevated vascular endothelial growth factor A (VEGFA) mRNA expression was found in S100A4(+)/HIF-1α(+) GBM cells adjacent to the vascular-rich areas. In addition, GBM patients with high S100A4 protein expression had significantly worse OS and PFS than did patients with low S100A4 expression. Knockdown of S100A4 in the GBM cell line KS-1 decreased migration capability, concomitant with decreased Slug expression; the opposite effects were elicited by blebbistatin-dependent inhibition of NMIIA. CONCLUSION: S100A4(+)/HIF-1α(-) GBM cells are recruited to (and migrate along) preexisting vessels through inhibition of NMIIA activity. This is likely stimulated by extracellular VEGF that is released by S100A4(+)/HIF-1α(+) tumor cells in non-Ps perinecrotic lesions. In turn, these events engender tumor progression via acceleration of pro-tumorigenic vascular functions. Video abstract.


Subject(s)
Brain Neoplasms , Glioblastoma , Nonmuscle Myosin Type IIA , S100 Calcium-Binding Protein A4 , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carcinogenesis , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nonmuscle Myosin Type IIA/metabolism , S100 Calcium-Binding Protein A4/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
BMC Cancer ; 22(1): 184, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35177036

ABSTRACT

BACKGROUND: S100A1 expression is deregulated in a variety of human malignancies, but its role in normal and malignant endometrial cells is unclear. METHODS: We used endometrial carcinoma (Em Ca) cell lines to evaluate the physical and functional interaction of S100A1 with p53 and its negative regulator, mouse double minute 2 (MDM2). We also evaluated the expression of S100A1, p53, and MDM2 in clinical samples consisting of 89 normal endometrial and 189 Em Ca tissues. RESULTS: S100A1 interacted with MDM2 but not p53 in Em Ca cell lines. Treatment of cells stably overexpressing S100A1 with Nutlin-3A, an inhibitor of the p53/MDM2 interaction, increased expression of p53-target genes including p21waf1 and BAX. S100A1 overexpression enhanced cellular migration, but also sensitized cells to the antiproliferative and proapoptotic effects of Adriamycin, a genotoxic agent; these phenotypes were abrogated when S100A1 was knocked down using shRNA. In clinical samples from normal endometrium, S100A1 expression was significantly higher in endometrial glandular cells of the middle/late secretory and menstrual stages when compared to cells in the proliferative phases; high S100A1 was also positively correlated with expression of MDM2 and p21waf1 and apoptotic status, and inversely correlated with Ki-67 scores. However, such correlations were absent in Em Ca tissues. CONCLUSION: The interaction between S100A1 and MDM2 may modulate proliferation, susceptibility to apoptosis, and migration through alterations in p53 signaling in normal- but not malignant-endometrial cells.


Subject(s)
Endometrial Neoplasms/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , S100 Proteins/metabolism , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation/genetics , Endometrium/cytology , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice
11.
Am J Pathol ; 191(10): 1837-1850, 2021 10.
Article in English | MEDLINE | ID: mdl-34214505

ABSTRACT

Deregulated full-length anaplastic lymphoma kinase (ALK) overexpression has been found in some primary solid tumors, but little is known about its role in ovarian high-grade serous carcinoma (HGSC). The current study focused on the functional roles of ALK in HGSC. Cytoplasmic ALK immunoreactivity without chromosomal rearrangement and gene mutations was significantly higher in HGSC compared with non-HGSC-type ovarian carcinomas, and was significantly associated with several unfavorable clinicopathologic factors and poor prognosis. HGSC cell lines stably overexpressing ALK exhibited increased cell proliferation, enhanced cancer stem cell features, and accelerated cell mobility, whereas these phenotypes were abrogated in ALK-knockdown cells. Expression of the nervous system-associated gene, ELAVL3, and the corresponding protein (commonly known as HuC) was significantly increased in cells overexpressing ALK. Expression of SRY-box transcription factor (Sox)2 and Sox3 (genes associated with the neural progenitor population) increased in ALK-overexpressing but not ALK-knockdown cells. Furthermore, overexpression of Sox2 or Sox3 enhanced both ALK and ELAVL3 promoter activities, suggesting the existence of ALK/Sox/HuC signaling loops. Finally, ALK overexpression was attributed to increased expression of neuroendocrine markers, including synaptophysin, CD56, and B-cell lymphoma 2, in HGSC tissues. These findings suggest that overexpression of full-length ALK may influence the biological behavior of HGSC through cooperation with ELAVL3 and Sox factors, leading to the establishment and maintenance of the aggressive phenotypic characteristics of HGSC.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/pathology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Adult , Aged , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cytoplasm/enzymology , ELAV-Like Protein 3/metabolism , Female , Humans , Middle Aged , Models, Biological , Multivariate Analysis , Neoplasm Grading , Neoplastic Stem Cells/pathology , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Phenotype , Prognosis , Progression-Free Survival , SOX Transcription Factors/metabolism
12.
Mol Carcinog ; 59(12): 1409-1419, 2020 12.
Article in English | MEDLINE | ID: mdl-33111989

ABSTRACT

Glioblastomas (GBM) contain numerous hypoxic foci associated with a rare fraction of glioma stem cells (GSCs). Left-right determination factor (LEFTY) and Nodal, members of the transforming growth factor ß (TGF-ß) superfamily, have glycogen synthase kinase 3ß (GSK-3ß) phosphorylation motifs and are linked with stemness in human malignancies. Herein, we investigated the roles of LEFTY and Nodal in GBM hypoxic foci. In clinical samples, significantly higher expression of LEFTY, Nodal, phospho (p) GSK-3ß, pSmad2, and Nestin, as well as higher apoptotic and lower proliferation rates, were observed in nonpseudopalisading (non-Ps) perinecrotic lesions as compared to Ps and non-necrotic tumor lesions, with a positive correlation between LEFTY, Nodal, pGSK-3ß, or pSmad2 scores. In KS-1, a GBM cell line that lacks endogenous Nodal expression, treatment with the hypoxic mimetic CoCl2 increased LEFTY, pGSK-3ß, and pSmad2 levels, but decreased pAkt levels. Moreover, the promoter for LEFTY, but not Nodal, was activated by Smad2 or TGF-ß1, suggesting that overexpression of LEFTY and Nodal may be due to Akt-independent GSK-3ß inactivation, with or without cooperation of the TGF-ß1/Smad2 axis. LEFTY and Nodal overexpression increased proliferation rates and reduced susceptibility to CoCl2 -induced apoptosis, and increased the expression of epithelial-mesenchymal transition (EMT)/GSC-related markers. An increased ALDH1high population and more efficient spheroid formation was also observed in LEFTY-overexpressing cells. These findings suggest that LEFTY and Nodal may contribute to cell survival in non-Ps GBM perinecrotic lesions, leading to alterations in apoptosis, proliferation, or EMT/GCS features.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Left-Right Determination Factors/metabolism , Nodal Protein/metabolism , Up-Regulation , Adolescent , Adult , Aged , Brain Neoplasms/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Cobalt/adverse effects , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Left-Right Determination Factors/genetics , Male , Middle Aged , Nodal Protein/genetics , Phosphorylation , Signal Transduction , Young Adult
13.
Am J Pathol ; 190(11): 2304-2316, 2020 11.
Article in English | MEDLINE | ID: mdl-32805233

ABSTRACT

S100A4 is a small calcium-binding protein that exerts its biological functions by interacting with nonmuscle myosin IIA (NMIIA) and p53. Although S100A4 promotes metastasis in several tumors, little is known about its involvement in the progression of ovarian high-grade serous carcinomas (HGSCs). Herein, we focused on functional roles of the S100A4/NMIIA/p53 axis in these tumors. In HGSC cell lines harboring mutant p53, knockdown (KD) of S100A4 reduced the expression of several epithelial-mesenchymal transition/cancer stem cell markers and the ALDH1high population, consistent with an inhibition of stemness features. S100A4-KD also increased apoptosis, decreased cell proliferation, and accelerated cell mobility. This was accompanied by increased Snail expression, which, in turn, was likely due to loss of p53 function. In contrast, specific inhibition of NMIIA by blebbistatin induced phenotypes that-with the exception of cell proliferation and mobility-were opposite to those observed in S100A4-KD cells. In clinical samples, cytoplasmic and/or nuclear interactions between S100A4, NMIIA, and mutant p53 were observed. In addition, high expression of S100A4, but not NMIIA or p53, was a significant and independent unfavorable prognostic factor in HGSC patients. These findings suggest that, via its interaction with NMIIA and p53, overexpressed S100A4 may induce epithelial-mesenchymal transition/cancer stem cell properties in HGSC and elicit several other tumor-associated phenotypes.


Subject(s)
Cystadenocarcinoma, Serous/metabolism , Neoplastic Stem Cells/metabolism , Nonmuscle Myosin Type IIA/metabolism , Ovarian Neoplasms/metabolism , S100 Calcium-Binding Protein A4/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cystadenocarcinoma, Serous/pathology , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Snail Family Transcription Factors/biosynthesis
14.
Cell Commun Signal ; 18(1): 103, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32635925

ABSTRACT

BACKGROUND: We previously demonstrated that ovarian high grade serous carcinomas (OHGSeCa) and ovarian clear cell carcinomas (OCCCa) with an HNF-1ß+/p53+/ARID1A+ immunophenotype were associated with the worst unfavorable prognosis. To clarify the molecular mechanisms underlying this finding, we focused on alterations in the p53 signaling pathway in these tumors. METHODS: Changes in cell phenotype and function following knockdown of wild-type p53 (p53-KD) were assessed using OCCCa cells expressing endogenous HNF-1ß and ARID1A. The prognostic significance of molecules that were deregulated following p53-KD was also examined using 129 OCCCa/OHGSeCa cases. RESULTS: p53-KD cells had increased expression of Snail, phospho-Akt (pAkt), and pGSK3ß, and decreased E-cadherin expression, leading to epithelial-mesenchymal transition (EMT)/cancer stem cell (CSC) features. The cells also exhibited acceleration of cell motility and inhibition of cell proliferation and apoptosis. Next generation sequencing revealed that fibronectin (FN) expression was significantly increased in the p53 KD-cells, in line with our observation that wild-type p53 (but not mutant p53) repressed FN1 promoter activity. In addition, treatment of OCCCa cells with FN significantly increased cell migration capacity and decreased cell proliferation rate, independent of induction of EMT features. In clinical samples, FN/p53 scores were significantly higher in OCCCa/OHGSeCa with the HNF-1ß+/p53+/ARID1A+ immunophenotype when compared to others. Moreover, high FN/high p53 expression was associated with the worst overall survival and progression-free survival in OCCCa/OHGSeCa patients. CONCLUSION: These findings suggest that upregulation of FN following loss of p53 function may impact the biological behavior of OCCCa/OHGSeCa, particularly in tumors with an HNF-1ß+/p53+/ARID1A+ immunophenotype, through alterations in cell mobility and cell proliferation. The accompanying induction of EMT/CSC properties and inhibition of apoptosis due to p53 abnormalities also contribute to the establishment and maintenance of tumor phenotypic characteristics. Video Abstract.


Subject(s)
Fibronectins/genetics , Immunophenotyping , Ovarian Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation/genetics , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 1-beta/metabolism , Humans , Kinetics , Middle Aged , Models, Biological , Multivariate Analysis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Phenotype , Prognosis , Progression-Free Survival , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism
15.
Lab Invest ; 100(5): 682-695, 2020 05.
Article in English | MEDLINE | ID: mdl-31857700

ABSTRACT

Uterine carcinosarcoma (UCS) represents a true example of cancer associated with epithelial-mesenchymal transition (EMT), which exhibits cancer stem cell (CSC)-like traits. Although S100A4 is an inducer of EMT, little is known about its involvement in UCS tumorigenesis. Herein, we focused on the functional role of S100A4 during development of UCS. Expression of S100A4 and molecules associated with its function were also examined in 35 UCS cases. In endometrial carcinoma cell lines, S100A4 promoter activity and mRNA levels were significantly increased by the transfection of NF-κB/p65, independent of a putative κB-binding site in the promoter. Cells stably overexpressing S100A4 showed enhancement of CSC properties, along with decreased cell proliferation and acceleration of cell migration. These phenotypes were abrogated in S100A4-knockdown cells. A combination of S100A4 antibody-mediated co-immunoprecipitation and shotgun proteomics analysis revealed that S100A4 strongly interacted with non-muscle myosin II (NMII) heavy chains, including myosin 9 and myosin 14. Specific inhibition of NMII by blebbistatin phenocopied S100A4 overexpression and induced a fibroblast-like morphology. In clinical samples, S100A4 score was significantly higher in sarcomatous as compared with carcinomatous components of UCS, and was positively correlated with ALDH1, Slug, and vimentin scores, and inversely with Ki-67 labeling indices. These findings suggest that an S100A4/NMII-related signaling cascade may contribute to the establishment and maintenance of EMT/CSC properties, along with changes in cell proliferation and migration capability. These events may be initiated in carcinomatous components in UCS and lead to divergent sarcomatous differentiation.


Subject(s)
Carcinosarcoma/pathology , Epithelial-Mesenchymal Transition/physiology , S100 Calcium-Binding Protein A4 , Signal Transduction/physiology , Uterine Neoplasms/pathology , Carcinosarcoma/chemistry , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism , Uterine Neoplasms/chemistry , Uterus/chemistry , Uterus/pathology
16.
BMC Cancer ; 19(1): 308, 2019 Apr 03.
Article in English | MEDLINE | ID: mdl-30943930

ABSTRACT

BACKGROUND: Expression of Nodal, a member of the TGF-ß superfamily, is commonly absent in differentiated tissues, while its re-expression occurs in a variety of human malignancy. However, little is known about its involvement in ovarian tumorigenesis. Herein, we focused on the functional roles of Nodal in ovarian endometriosis-carcinoma lesions. METHODS: Regulation and function of Nodal and its associated molecules, including Smad2, GSK-3ß, and several cell kinetics-related molecules, were assessed using clinical samples consisting of 108 ovarian carcinomas and 33 endometriotic lesions, as well as ES-2 (ovarian clear cell carcinoma; OCCCa) and Ishikawa (endometrial carcinoma) cell lines. RESULTS: Nodal expression was significantly higher in endometriosis and OCCCa lesions as compared to that of non-OCCCas, with positive correlations to phosphorylated forms of both Smad2 (pSmad2) and GSK-3ß. When compared to endometriotic lesions, the expression of Nodal and pSmad2 was significantly decreased in OCCCa. Treatment of Ishikawa cells with TGF-ß1 resulted in transcriptional upregulation of Nodal, along with increased pSmad2 expression, while inhibition of GSK-3ß also induced an increase in Nodal expression at the posttranslational level. Both ES-2 and Ishikawa cells stably overexpressing Nodal had increased susceptibility to apoptosis in response to treatment with cisplatin and doxorubicin, respectively, together with higher cleaved caspase-3 expression and decreased Bcl2/Bax ratio. Moreover, the stable Nodal-overexpressing cells showed reduced cell proliferation, along with increased expression of p27kip1 and p21waf1. In clinical samples, a significantly higher number of apoptotic cells and lower Ki-67 labeling indices were observed in Nodal-positive as compared to Nodal-negative OCCCa. CONCLUSIONS: These findings suggest that Nodal is a multifunctional cytokine involved in the modulation of cell kinetics in ovarian endometriosis-OCCCa lesions.


Subject(s)
Adenocarcinoma, Clear Cell/metabolism , Endometriosis/metabolism , Nodal Protein/metabolism , Ovarian Neoplasms/metabolism , Up-Regulation , Adenocarcinoma, Clear Cell/genetics , Adult , Aged , Aged, 80 and over , Apoptosis , Cell Proliferation , Endometriosis/genetics , Female , Humans , Middle Aged , Nodal Protein/genetics , Ovarian Neoplasms/genetics , Phosphorylation , Signal Transduction , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism
17.
Hum Pathol ; 89: 10-23, 2019 07.
Article in English | MEDLINE | ID: mdl-31022415

ABSTRACT

We focused on specific molecular events during the development of endometriosis-associated ovarian endometrioid carcinoma (OEmCa) and ovarian clear cell carcinoma (OCCCa). Alterations in ß-catenin (encoded by CTNNB1) and microsatellite instability (MSI), as well as changes in the expression levels of HNF-1ß and DNA mismatch repair (MMR) proteins were investigated in 50 OEmCas and 21 OCCCas with endometriotic lesions. Mutations of CTNNB1 were identified in 28 (56%) of the 50 OEmCa cases and 26 (41.9%) of the 62 coexisting endometriosis lesions. MSI-high (H) was observed in 7 (14.6%) of the 48 OEmCa and 14 (23.3%) of the 60 coexisting endometriosis, and was significantly associated with loss of MMR protein expression, but not CTNNB1 mutations. Nonidentical CTNNB1 status between 2 different epithelial lesions within endometriosis was observed in 8 of 10 informative endometriosis cases that had adjacent OEmCa. Similar findings for MSI features were also found in 2 of 3 informative cases, suggesting that endometriotic lesions may predominantly consist of polyclonal cells. In contrast, high HNF-1ß expression was significantly associated with SLC3A1 expression, which plays a major role in HNF-1ß-triggered induction of reactive oxygen species in OCCCas, independent of abnormalities in both ß-catenin and MSI/MMR status. Finally, 4 inflammatory parameters associated with repeated hemorrhaging in endometriosis were significantly higher in endometriosis with MSI-high when compared with that with MSS, independent of both ß-catenin and HNF-1ß status. In conclusion, different molecular pathways including alterations in ß-catenin, MSI, and HNF-1ß levels may contribute to tumorigenesis in endometriosis-associated carcinoma.


Subject(s)
Carcinogenesis/metabolism , Endometriosis/complications , Ovarian Neoplasms/etiology , Ovarian Neoplasms/metabolism , Adenocarcinoma, Clear Cell/etiology , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/metabolism , Adult , Aged , Carcinogenesis/genetics , Carcinoma, Endometrioid/etiology , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/metabolism , Endometriosis/metabolism , Female , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 1-beta/metabolism , Humans , Microsatellite Instability , Middle Aged , Ovarian Neoplasms/genetics , Young Adult , beta Catenin/genetics , beta Catenin/metabolism
18.
Hum Pathol ; 85: 210-220, 2019 03.
Article in English | MEDLINE | ID: mdl-30448218

ABSTRACT

Although proteomic profiles for ovarian epithelial carcinoma (OECa) have been widely investigated, no single marker or set of predictors has been clinically implemented mainly because their reliability and validity have not yet been well established. To establish immunohistochemical (IHC) panels for prognosis prediction of OECa for use in daily pathology practice, the expression patterns of 12 IHC markers, p53, HNF-1ß, ARID1A, estrogen receptor-α, progesterone receptor, vimentin, PTEN, PIK3CA, WT1, left-right determination factor, ß-catenin, and Ki-67 were investigated using 282 OECas. Hierarchical clustering analysis revealed 7 major immunoprofile groups (IPGs I-VII) that could be used to categorize OECa tumors independent of histotypes. Based on the results of the cluster analysis and protein expression statuses, we further demonstrated the effective classification of OECa tumors into simplified immunoprofile panels using only 4 IHC markers including HNF-1ß, p53, ARID1A, and WT1. The tumors in IPG VII with HNF1ß+/p53+/ARID1A+ immunophenotype demonstrated a significantly worse overall survival and progression-free survival as compared with the other IPGs. Multivariate Cox regression analysis also revealed that the immunophenotype (HNF1ß+/p53+/ARID1A+) and clinical stage were significant and independent prognostic factors for overall survival and progression-free survival in advanced OECa. In conclusion, we identified immunoprofiles in OECa using a panel of 4 IHC markers, which could identify tumors by the immunophenotype that is associated with the most unfavorable prognosis and thus facilitate prognosis prediction of advanced OECa.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial/metabolism , Ovarian Neoplasms/metabolism , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/pathology , Female , Humans , Immunophenotyping , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Prognosis , Progression-Free Survival , Proteomics , Survival Rate
19.
Mol Carcinog ; 57(8): 957-967, 2018 08.
Article in English | MEDLINE | ID: mdl-29603383

ABSTRACT

Advanced ovarian clear cell carcinoma (OCCCa) shows poor prognosis with chemoresistance, which is associated with epithelial-mesenchymal transition (EMT)/cancer stem cell (CSC) features. The left-right determination factor (LEFTY), a novel member of the TGF-ß superfamily, is a marker of stemness. Here we focused on the functional roles of LEFTY in OCCCas. OCCCa cell lines that were cultured in STK2, a serum-free medium for mesenchymal stem cells, or treated with TGF-ß1 underwent morphological changes toward an EMT appearance, along with increased expression of LEFTY and Snail. The cells also showed CSC properties, as demonstrated by increases in the aldehyde dehydrogenase (ALDH)1high activity population, number of spheroid formation, and expression of several CSC markers. Inhibition of LEFTY expression induced decreases in the number of spindle-shaped cells and CSC features, while cells stably overexpressing LEFTY exhibited enhancement of such EMT/CSC properties. Finally, treatment of cells with TGF-ß1 led to increased LEFTY expression and activation of Akt, which subsequently induced inactivation of GSK-3ß, while inhibition of GSK-3ß resulted in increased expression of both LEFTY and Snail. In clinical samples, LEFTY expression showed a tendency for positive associations with expression of vimentin, as well as Sox2 and ALDH1, in OCCCas with epithelial-like morphology, indicating a possible relationship between LEFTY and the epithelial-mesenchymal hybrid stage of the tumors. In conclusion, TGF-ß-mediated LEFTY/Akt/GSK-3ß/Snail axis may contribute to the establishment and maintenance of phenotypic characteristics of OCCCas through modulation of EMT/CSC properties.


Subject(s)
Adenocarcinoma, Clear Cell/metabolism , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Aged, 80 and over , Female , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Left-Right Determination Factors/metabolism , Middle Aged , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Snail Family Transcription Factors/metabolism
20.
Cell Commun Signal ; 15(1): 56, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29268772

ABSTRACT

BACKGROUND: The left-right determination factor (LEFTY) is a novel member of the TGF-ß/Smad2 pathway and belongs to the premenstrual/menstrual repertoire in human endometrium, but little is known about its functional role in endometrial carcinomas (Em Cas). Herein, we focused on LEFTY expression and its association with progesterone therapy in Em Cas. METHODS: Regulation and function of LEFTY, as well as its associated molecules including Smad2, ovarian hormone receptors, GSK-3ß, and cell cycle-related factors, were assessed using clinical samples and cell lines of Em Cas. RESULTS: In clinical samples, LEFTY expression was positively correlated with estrogen receptor-α, but not progesterone receptor (PR), status, and was inversely related to phosphorylated (p) Smad2, cyclin A2, and Ki-67 levels. During progesterone therapy, expression of LEFTY, pSmad2, and pGSK-3ß showed stepwise increases, with significant correlations to morphological changes toward secretory features and decreased Ki-67 values. In Ishikawa cells, an Em Ca cell line that expresses PR, progesterone treatment reduced proliferation and induced increased expression of LEFTY and pGSK-3ß, although LEFTY promoter regions were inhibited by transfection of PR. Moreover, inhibition of GSK-3ß resulted in increased LEFTY expression through a decrease in its ubiquitinated form, suggesting posttranslational regulation of LEFTY protein via GSK-3ß suppression in response to progesterone. In addition, overexpression or knockdown of LEFTY led to suppression or enhancement of Smad2-dependent cyclin A2 expression, respectively. CONCLUSION: Upregulation of LEFTY may serve as a useful clinical marker for the therapeutic effects of progesterone for Em Cas, leading to inhibition of tumor cell proliferation through alteration in Smad2-dependent transcription of cyclin A2.


Subject(s)
Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Left-Right Determination Factors/metabolism , Progesterone/metabolism , Adult , Cell Cycle/drug effects , Cell Line, Tumor , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrium/drug effects , Endometrium/metabolism , Endometrium/pathology , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Left-Right Determination Factors/genetics , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Smad2 Protein/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL