Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Carcinogenesis ; 42(9): 1162-1170, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34323956

ABSTRACT

Patients with ovarian clear cell carcinoma (OCCC) experience frequent recurrence, which is most likely due to chemoresistance. We used shotgun proteomics analysis and identified upregulation of ezrin-binding phosphoprotein 50 (EBP50) in recurrent OCCC samples. Cytoplasmic and/or nuclear (Cyt/N), but not membranous, EBP50 immunoreactivity was significantly higher in recurrent OCCC as compared with that of primary tumors. OCCC cells expressing cytoplasmic EBP50 were significantly less susceptible to cisplatin (CDDP)-induced apoptosis compared with cells expressing membranous EBP50. Abrogation of resistance following knockdown of cytoplasmic EBP50 was accompanied by decreased XIAP and BCL2, increased BAX and increased caspase-3 cleavage. We found that poly (ADP-ribose) polymerase1 (PARP1), which is involved in DNA damage detection and repair, binds to EBP50 through its PDZ1 domain. CDDP treatment of cells expressing cytoplasmic (but not membranous) EBP50 increased nuclear PARP1 expression, whereas knockdown of EBP50 cells decreased PARP1 expression and activity following CDDP treatment. Finally, OCCC patients with a combination of Cyt/N EBP50 and high PARP1 score had worst the prognosis for overall and progression-free survival. Together, our data suggest that cytoplasmic EBP50 inhibits apoptosis and promotes OCCC survival through stabilization of PARP1 activity and modulation of the XIAP/BCL2/BAX axis. This may increase the likelihood of tumor recurrence, and we therefore suggest a combined analysis for EBP50 and PARP1 may have great utility in OCCC prediction and prognosis.


Subject(s)
Biomarkers, Tumor/metabolism , Cytoplasm/metabolism , Ovarian Neoplasms/metabolism , Phosphoproteins/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Sodium-Hydrogen Exchangers/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement , Female , Humans , Middle Aged , Neoplasm Recurrence, Local/metabolism , Ovarian Neoplasms/pathology , Prognosis , Protein Binding , Proteomics/methods , Survival Analysis
8.
Mol Pain ; 17: 17448069211021252, 2021.
Article in English | MEDLINE | ID: mdl-34074169

ABSTRACT

BACKGROUND: Rotator cuff tears (RCTs) are often associated with severe shoulder pain. Non-steroidal anti-inflammatory drugs, not recommended for long-term use, do not effectively manage RCT-induced pain, resulting in reduced quality of life. To improve management, a better understanding of the fundamental properties of RCT pain is needed. Here, we aimed to compare the expression levels of nerve growth factor (NGF) and cyclooxygenase-2 (COX-2) mRNA in the synovial tissues of patients with RCT-induced pain and patients with non-painful recurrent shoulder dislocation (RSD). METHODS: The study included 32 patients with RCT who underwent arthroscopic rotator cuff repair and 28 patients with non-painful RSD who underwent arthroscopic Bankart repair. Synovial tissue samples were harvested from subacromial bursa and rotator interval of RCT patients and from the rotator interval of RSD patients. Samples were analyzed quantitatively expression levels for NGF and COX2 mRNA and NGF protein. RESULTS: NGF mRNA and protein levels were significantly higher in the rotator interval of RCT patients than in the rotator interval of RSD patients (p = 0.0017, p = 0.012, respectively), while COX2 mRNA levels did not differ significantly between the two patient groups. In RCT patients, COX2 mRNA was more highly expressed in the rotator interval than in the subacromial bursa (p = 0.038), whereas the mRNA and protein levels of NGF did not differ between the two tissues. The expression of NGF mRNA in the synovium of the rotator interval was significantly correlated with the numeric rating scale of pain (ρ = 0.38, p = 0.004). CONCLUSION: NGF mRNA and protein levels were elevated in patients with painful RCT compared with those in patients with non-painful RSD, whereas COX-2 levels were comparable in the two patient groups. These findings provide insights into novel potential strategies for clinical management of RCT.


Subject(s)
Rotator Cuff Injuries , Arthroscopy , Humans , Nerve Growth Factor/genetics , Rotator Cuff , Synovial Membrane
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Pathol Int ; 68(4): 232-240, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29431238

ABSTRACT

We established the KU-Lu-8 monoclonal antibody (MoAb) using a lung cancer cell line as an antigen and a random immunization method. The KU-Lu-8 MoAb recognizes basigin (BSG), which is a transmembrane-type glycoprotein that is strongly expressed on the cell membranes of lung cancer cells. This study aimed to clarify the relationships between BSG expression and clinicopathological parameters and determine the prognostic significance of BSG expression in pulmonary adenocarcinoma (AC) patients. To evaluate the significance of BSG expression in lung cancer, we immunohistochemically analyzed 113 surgically resected pulmonary adenocarcinomas, and the associations between BSG expression and various clinicopathological parameters were evaluated. Kaplan-Meier survival analysis and Cox proportional hazards models were used to investigate the effects of BSG expression on survival. Clinicopathologically, BSG expression was significantly associated with tumor differentiation, vascular invasion, lymphatic invasion, and a poor prognosis. In particular, BSG expression was significantly correlated with poorer survival in patients with stage I AC. The high BSG expression group (compared with the low BSG expression group) exhibited adjusted hazard ratios for mortality of 4.694. BSG expression is indicative of a poor prognosis in AC patients, particularly in those with stage I disease.


Subject(s)
Adenocarcinoma/pathology , Basigin/biosynthesis , Biomarkers, Tumor/analysis , Lung Neoplasms/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Basigin/analysis , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Middle Aged , Prognosis , Proportional Hazards Models
16.
BMC Musculoskelet Disord ; 19(1): 204, 2018 Jun 26.
Article in English | MEDLINE | ID: mdl-29945585

ABSTRACT

BACKGROUND: Research suggests that vascular endothelial growth factor (VEGF) levels in the synovial fluid of knee osteoarthritis (KOA) patients are positively correlated with KOA severity. The relationship between synovial VEGF levels and pain in human KOA patients is not fully understood, and the role of VEGF in the pain pathway remains unclear. METHODS: We harvested synovial membrane (SM) from 102 patients with radiographic evidence of KOA (unilateral Kellgren/Lawrence [K/L] grade 2-4) during total knee arthroplasty. Patients scored their pain on a 0 to 10 cm visual analog scale (VAS). VEGF levels in the SM of KOA patients with strong/severe (VAS ≥ 6) and mild/moderate pain (VAS < 6) were compared. Correlations between VAS and VEGF mRNA expression were investigated. To investigate a possible mechanism for VEGF-induced pain, the distribution of VEGF and the neuropeptide apelin was determined by immunohistochemical analyses. To investigate the role of VEGF in regulating apelin expression, SM cells were exposed to VEGF. RESULTS: VEGF expression in the VAS ≥ 6 group was significantly greater than expression in the VAS < 6 group. Expression levels of VEGF were also positively correlated with VAS. VEGF-positive cells were identified in the lining of the SM. Expression of apelin mRNA and protein were significantly elevated in SM cells treated with exogenous VEGF compared to those treated with vehicle. CONCLUSION: Synovial VEGF may be involved in pain pathways in KOA and its action may be mediated by apelin.


Subject(s)
Osteoarthritis, Knee/metabolism , Pain/metabolism , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/pharmacology , Aged , Aged, 80 and over , Biomarkers/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Female , Gene Expression , Humans , Male , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/surgery , Pain/pathology , Pain Measurement/methods , Synovial Membrane/pathology , Vascular Endothelial Growth Factor A/genetics
17.
Histopathology ; 71(2): 227-237, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28370249

ABSTRACT

AIMS: ß-Catenin signalling participates in the regulation of epithelial-mesenchymal transition (EMT)/cancer stem cell (CSC) properties. The aim of this study was to investigate the role of ß-catenin in resistance to neoadjuvant chemoradiotherapy in patients with rectal cancer, especially pertaining to its association with EMT/CSC features. METHODS AND RESULTS: A total of 109 cases of locally advanced rectal cancer, along with a colon cancer cell line, were investigated. Nuclear ß-catenin accumulation in pretreatment-biopsied samples was inversely associated with the therapeutic efficacy of chemoradiotherapy in resected rectal cancer. In resected tumours, nuclear ß-catenin was predominantly observed in EMT-like lesions with decreased E-cadherin and increased Snail expression, along with expression of CSC-related markers. The EMT-like lesions also showed significant decreases in both apoptosis and cell proliferation as compared with non-EMT lesions. In-vitro culture of a colon cancer cell line in STK2 was sufficient to induce EMT/CSC properties together with nuclear ß-catenin accumulation, and showed inhibition of cell proliferation and resistance to doxorubicin treatment. CONCLUSION: Nuclear ß-catenin accumulation may contribute to chemoradioresistance in locally advanced rectal cancer, probably through its regulation of EMT/CSC properties. In addition, nuclear ß-catenin in pretreatment-biopsied samples is useful in predicting the efficacy of chemoradiotherapy in patients with rectal cancer.


Subject(s)
Biomarkers, Tumor/analysis , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Rectal Neoplasms/pathology , beta Catenin/metabolism , Chemoradiotherapy, Adjuvant , Humans , Neoadjuvant Therapy
18.
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
19.
BMC Musculoskelet Disord ; 18(1): 283, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28668088

ABSTRACT

BACKGROUND: Recent studies have suggested that the tumor necrosis factor-α (TNF-α) pathway is a potential target for the management of osteoarthritis (OA). Transforming growth factor (TGF)-ß-activated kinase 1 (TAK1) is essential in several cytokine-mediated cascades, including the TNF-α, interleukin-1 (IL-1), and TGF-ß pathways. The role of TAK1 in synovial tissue in OA is not fully understood. Using synovial cells harvested from OA patients during surgery, we investigated whether TAK1 inhibition suppresses production of TNF-α-induced extracellular matrix degrading enzymes and expression of pain-related molecules. METHODS: Synovial tissues were harvested from ten subjects with radiographic evidence of osteoarthritis (OA) during total knee arthroplasty. Synovial cells were cultured and stimulated with control (culture media), 10 ng/mL human recombinant TNF-α, or 10 ng/mL TNF-α and 10 µM of the TAK1 inhibitor (5Z)-7-oxozeaenol for 24 h. Real-time polymerase chain reaction (PCR) analysis was used to monitor expression of mRNA of the extracellular matrix degrading enzymes matrix metalloproteinase-3 (MMP-3) and a disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1 motif, 4 (ADAMTS-4); and of the pain-related molecules cyclooxygenase-2 (COX-2), microsomal prostaglandin E synthase-1 (mPGES-1), and nerve growth factor (NGF). MMP-3 and NGF protein concentrations in cell supernatant were measured by enzyme-linked immunosorbent assay (ELISA). COX-2, mPGES-1 and ADAMTS-4 protein expression was also evaluated by western blotting. RESULTS: TNF-α stimulated increases in ADAMTS-4 and MMP3 mRNA (2.0-fold and 1.6-fold, respectively, p < 0.05) and protein expression (21.5-fold and 2.0-fold, respectively). Treatment with the TAK1 inihibitor (5Z)-7-oxozeaenol reduced ADAMTS-4 and MMP3 mRNA (0.5-fold and 0.6-fold, respectively) and protein expression (1.4-fold and 0.5-fold, respectively) in OA synovial cells. COX-2, mPGES-1 and NGF mRNA (11.2-fold, 3.1-fold and 2.7-fold, respectively) and protein expression (3.0-fold, 2.7-fold and 2.2-fold, respectively) were increased by TNF-α. (5Z)-7-oxozeaenol treatment reduced mPGES1 and NGF mRNA (1.5-fold and 0.8-fold, respectively) and protein (1.5-fold and 0.5-fold, respectively). CONCLUSION: TAK1 plays an important role in the regulation of TNF-α induced extracellular matrix degrading enzymes and pain-related molecule expression. TAK1 may be a potential target for therapeutic strategies aimed at preventing osteoarthritis progression and pain.


Subject(s)
Arthralgia/metabolism , Extracellular Matrix/enzymology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/physiology , Synovial Fluid/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Aged , Aged, 80 and over , Arthralgia/surgery , Arthroplasty, Replacement, Knee/trends , Cells, Cultured , Extracellular Matrix/drug effects , Female , Gene Expression , Humans , Lactones/pharmacology , Male , Middle Aged , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/surgery , Resorcinols/pharmacology , Synovial Fluid/drug effects
20.
BMC Cancer ; 16: 53, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26841870

ABSTRACT

BACKGROUND: Uterine carcinosarcoma (UCS) represents a true example of cancer associated with epithelial-mesenchymal transition (EMT), which exhibits cancer stem cell (CSC)-like traits. Both Sox and ß-catenin signal transductions play key roles in the regulation of EMT/CSC properties, but little is known about their involvement in UCS tumorigenesis. Herein, we focused on the functional roles of the Sox/ß-catenin pathway in UCSs. METHODS: EMT/CSC tests and transfection experiments were carried out using three endometrial carcinoma (Em Ca) cell lines. Immunohistochemical investigation was also applied for a total of 32 UCSs. RESULTS: Em Ca cells cultured in STK2, a serum-free medium for mesenchymal stem cells, underwent changes in morphology toward an EMT appearance through downregulation of E-cadherin, along with upregulation of Slug, known as a target gene of ß-catenin. The cells also showed CSC properties with an increase in the aldehyde dehydrogenase (ALDH) 1(high) activity population and spheroid formation, as well as upregulation of Sox4, Sox7, and Sox9. Of these Sox factors, overexpression of Sox4 dramatically led to transactivation of the Slug promoter, and the effects were further enhanced by cotransfection of Sox7 or Sox9. Sox4 was also able to promote ß-catenin-mediated transcription of the Slug gene through formation of transcriptional complexes with ß-catenin and p300, independent of TCF4 status. In clinical samples, both nuclear ß-catenin and Slug scores were significantly higher in the sarcomatous elements as compared to carcinomatous components in UCSs, and were positively correlated with Sox4, Sox7, and Sox9 scores. CONCLUSIONS: These findings suggested that Sox4, as well as Sox7 and Sox9, may contribute to regulation of EMT/CSC properties to promote development of sarcomatous components in UCSs through transcriptional regulation of the Slug gene by cooperating with the ß-catenin/p300 signal pathway.


Subject(s)
Carcinosarcoma/genetics , E1A-Associated p300 Protein/biosynthesis , SOXC Transcription Factors/biosynthesis , Transcription Factors/biosynthesis , Uterine Neoplasms/genetics , beta Catenin/genetics , Animals , Carcinosarcoma/pathology , Cell Line, Tumor , E1A-Associated p300 Protein/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics , SOXC Transcription Factors/genetics , SOXF Transcription Factors/biosynthesis , SOXF Transcription Factors/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Uterine Neoplasms/pathology , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL