Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
J Cancer Res Clin Oncol ; 147(11): 3225-3243, 2021 Nov.
Article En | MEDLINE | ID: mdl-34258652

PURPOSE: FOXA1, as a pioneering transcription factor, has been shown to drive prostate cancer progression. Previous studies showed that FOXA1 expression in prostate cancer was positively associated with cancer angiolymphatic invasion and metastasis. However, the mechanism underlying the correlation between FOXA1 and prostate cancer angiolymphatic invasion and metastasis remains largely unclear. METHODS: Herein, we set out to investigate the role of FOXA1 in the interactions between prostate cancer cells and endothelial cells. Endothelial cells' phenotypes were assessed through CCK-8 assay, Transwell migration assay, and tube formation assay. The angiogenic factors acting on endothelial cells mediated by FOXA1were characterized by RNA-seq, qPCR array, angiogenesis cytokines array, and ELISA assay. The impact of FOXA1 on tumor angiogenesis was examined in a xenograft model in nude mice. The effect of FOXA1 on prostate cancer angiogenesis was validated on a primary prostate cancer tissue microarray. RESULTS: FOXA1 expression in prostate cancer cells promoted endothelial cell proliferation, migration, and tube formation in vitro. Mechanistically, FOXA1 increased pro-angiogenic factors production, including EGF, Endothelin-1, and Endoglin. Moreover, in vivo study showed that FOXA1 facilitated tumor angiogenesis. Furthermore, clinical samples investigation indicated that FOXA1 enhanced prostate cancer angiogenesis. CONCLUSION: Overall, these findings illustrated a tumor angiogenesis-promoting role of FOXA1 in prostate cancer.


Angiogenesis Inducing Agents/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Prostatic Neoplasms/blood supply , Animals , Cell Communication/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Coculture Techniques , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression , HEK293 Cells , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Hepatocyte Nuclear Factor 3-alpha/genetics , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
2.
Aging (Albany NY) ; 13(2): 2264-2278, 2020 12 11.
Article En | MEDLINE | ID: mdl-33323547

Multiple studies have previously demonstrated that long intergenic non-coding RNAs (lincRNAs) play an important role in the development of bladder cancer. However, little is known regarding the underlying molecular mechanisms of LINC00482 functions in bladder cancer. The current study aimed to elucidate the role of LINC00482 in the progression of bladder cancer. The initial step was to detect the expressions of LINC00482 and MMP15 in bladder cancer cells and tissue. According to the results from the RT-qPCR, LINC00482 and MMP15 were both highly expressed in bladder cancer cells and tissue. The relationship among LINC00482, FOXA1 and MMP15 was studied via dual-luciferase reporter assay. LINC00482 was positively correlated with MMP15. LINC00482 promoted MMP15 expression by recruiting FOXA1. Using the gain- and loss-of-function approaches, silencing of LINC00482 resulted in the downregulation of VEGF and NF-κB protein levels, decreased expression of inflammatory factors, and inhibited angiogenesis. Silencing of LINC00482 also suppressed tumor-associated inflammation and angiogenesis in vivo, which was found to be reversed by the overexpression of MMP15. The present study demonstrated that LINC00482 induced the expression of MMP15 by interacting with FOXA1, thereby contributing to the inflammation and angiogenesis in bladder cancer.


Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Hepatocyte Nuclear Factor 3-alpha/genetics , Matrix Metalloproteinase 15/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , RNA, Long Noncoding/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy , Aged , Animals , Cell Line, Tumor , Computational Biology , Down-Regulation , Female , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Humans , Male , Matrix Metalloproteinase 15/biosynthesis , Mice , Mice, Inbred BALB C , Middle Aged , NF-kappa B/biosynthesis , NF-kappa B/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Xenograft Model Antitumor Assays
3.
Breast Cancer Res Treat ; 183(1): 41-48, 2020 Aug.
Article En | MEDLINE | ID: mdl-32572714

BACKGROUND: Multi-gene expression assays have been developed with the aim of predicting late recurrence in patients with estrogen receptor (ER)-positive breast cancer. However, establishment of alternative markers based on immunohistochemistry is also important for achieving practical use. Based on our previous study, forkhead box A1 (FOXA1) protein was tested as a potentially useful predictive marker for late recurrence. METHODS: 117 patients with ER-positive HER2-negative invasive breast cancer who developed distant metastasis following curative surgery were retrospectively investigated. We also evaluated responsiveness to endocrine therapy according to FOXA1 expression. Furthermore, publicly available mRNA microarray data were analyzed to examine patterns of metastasis according to FOXA1 mRNA expression, employing the Kaplan-Meier plotter. RESULTS: High expression of FOXA1 was an independent factor predicting long disease-free survival (DFS), along with small tumor size (p = 0.010 and 0.016, respectively). Discrimination of DFS was improved by combining these two factors, i.e., patients with FOXA1-high small tumors had the longest DFS while those with FOXA1-low large tumors had the shortest DFS. Moreover, we revealed that risk of distant metastasis started to increase after the completion of adjuvant endocrine therapy in patients with FOXA1-high tumors. CONCLUSION: Among patients who developed distant metastasis, those with FOXA1-high tumors had significantly longer DFS. We believe our data to raise the possibility of FOXA1 being a useful predictive marker for late recurrence and to provide new insights into the biology of FOXA1-high breast cancers.


Breast Neoplasms/metabolism , Estrogens , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Neoplasm Proteins/biosynthesis , Neoplasms, Hormone-Dependent/metabolism , Adult , Aged , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/physiology , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/chemistry , Neoplasms, Hormone-Dependent/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Recurrence , Time Factors
4.
Acta Histochem ; 120(6): 520-524, 2018 Aug.
Article En | MEDLINE | ID: mdl-29909922

Previously studies showed that Forkhead transcription factor A1 (FoxA1) was associated with sweat secretion. To investigate the expression and localization of FoxA1 in the three-dimensional (3D) reconstructed eccrine sweat glands, eccrine sweat gland cells were transplanted subcutaneously into nude mice with Matrigel, and at 2, 3, 4, 5, 6, 8, 10 and 12 weeks post-transplantation, the reconstructed eccrine sweat glands were removed and immunostained for FoxA1 and co-immunostained for FoxA1 and eccrine sweat markers, K7, carbonic anhydrase II (CA Ⅱ), gross cystic disease fluid protein-15 (GCDFP-15) and α-smooth muscle actin (α-SMA), and FoxA1 and sweat secretion-related proteins, Na+-K+-ATPase α and Na+-K+-2Cl- cotransporter 1 (NKCC1). The results showed that FoxA1-positive cells weren't detected until 3 weeks post-implantation, a time point of the differntiation of secretory coil-like structures. From the fourth week on, the number of FoxA1-positive cells increased and thereafter maintained at a high number. Double immunofluorescence staining showed that FoxA1-positive cells co-expressed dark cell marker GCDFP-15 and myoepithelial cell marker α-SMA, as well as secretion-related proteins, Na+-K+-ATPase α and NKCC1 in both the native and reconstructed eccrine sweat glands. In conclusion, FoxA1 might be related to the development and differentiation of secretory coil-like structures, as well as the secretory function of the 3D reconstructed eccrine sweat glands.


Eccrine Glands/metabolism , Gene Expression Regulation/physiology , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Animals , Eccrine Glands/cytology , Eccrine Glands/transplantation , Female , Heterografts , Humans , Male , Mice , Mice, Nude
5.
Carcinogenesis ; 38(12): 1180-1187, 2017 12 07.
Article En | MEDLINE | ID: mdl-29029032

FOXA1 (Fork-head box protein A1, HNF-3a) is a transcription factor involved in androgen signaling with relevance for lineage-specific gene expression of the prostate. The expression was analyzed by immunohistochemistry on a tissue microarray containing 11152 prostate cancer specimens. Results were compared with tumor phenotype, biochemical recurrence, androgen receptor expression, ETS-related gene (ERG) status and other recurrent genomic alterations. FOXA1 expression was detectable in 97.6% of 8227 interpretable cancers and considered strong in 28.5%, moderate in 46.2% and weak in 22.9% of cases. High FOXA1 expression was associated with TMPRSS2:ERG rearrangement and ERG expression (P < 0.0001). High FOXA1 expression was linked to high Gleason grade, advanced pathological tumor (pT) stage and early PSA recurrence in ERG negative cancers (P < 0.0001), while these associations were either weak or absent in ERG positive cancers. In ERG negative cancers, the prognostic role of FOXA1 expression was independent of Gleason grade, pathological tumor stage, lymph node stage, surgical margin status and preoperative PSA. Independent prognostic value became even more evident if the analysis was limited to preoperatively available features such as biopsy Gleason grade, preoperative PSA, cT stage and FOXA1 expression (P < 0.0001). Within ERG negative cancers, FOXA1 expression was also strongly associated with PTEN and 5q21 deletions (P < 0.0001). High expression of FOXA1 is an independent prognostic parameter in ERG negative prostate cancer. Thus, FOXA1 measurement might provide clinically useful information in prostate cancer.


Biomarkers, Tumor/analysis , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Neoplasm Recurrence, Local/pathology , Prostatic Neoplasms/pathology , Adult , Aged , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Proportional Hazards Models , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/mortality , Prostatic Neoplasms/surgery , Transcriptional Regulator ERG/genetics
6.
Virchows Arch ; 471(3): 337-345, 2017 Sep.
Article En | MEDLINE | ID: mdl-28721490

Invasive bladder cancer is diverse, and includes several named histomorphologies that differ from conventional urothelial carcinoma, termed "histologic variants." By transcriptional analysis, bladder cancers can be divided into luminal and basal subtypes. In this paper, we study associations between markers of transcriptional subtypes and variant histology in a retrospective cohort of 309 cystectomy specimens. Histology slides were methodically reviewed for all cases, and variant histology was documented. Immunohistochemistry for FOXA1 (luminal marker) and CK14 (basal maker) was performed on histologic variants and their associated conventional urothelial carcinomas. Invasive carcinoma was present in 270 of the cystectomy specimens, 35% of which contained a histologic variant. Squamous carcinomas expressed higher CK14 levels than micropapillary, nested, and plasmacytoid carcinomas (p < 0.001, Kruskal-Wallis), keeping with the basal character of squamous carcinoma. Likewise, squamous carcinomas expressed lower FOXA1 levels than micropapillary, nested, and plasmacytoid carcinomas (p < 0.001, Kruskal-Wallis), keeping with the luminal character of micropapillary carcinoma, and suggesting that nested and plasmacytoid cancers have luminal character. FOXA1 was expressed at lower levels in conventional urothelial carcinoma associated with squamous carcinoma than conventional urothelial carcinoma associated with micropapillary carcinoma (p = 0.0072, Wilcoxon rank sum). CK14 expression did not differ between conventional urothelial carcinomas associated with squamous versus micropapillary carcinoma (p = 0.89, Wilcoxon rank sum). Instead, CK14 expression was higher in squamous carcinoma than conventional urothelial carcinoma present in the same bladder (p = 0.014, Wilcoxon rank sum, paired). Overall, the findings show that squamous and micropapillary cancers have different expression patterns of CK14 and FOXA1 and suggest that they arise from distinct precursors.


Biomarkers, Tumor/analysis , Carcinoma, Transitional Cell/pathology , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Keratin-14/biosynthesis , Urinary Bladder Neoplasms/pathology , Female , Humans , Male , Retrospective Studies
7.
Nat Rev Urol ; 14(2): 98-106, 2017 Feb.
Article En | MEDLINE | ID: mdl-27898096

Genomic and transcriptional studies have identified discrete molecular subtypes of bladder cancer. These observations could be the starting point to identify new treatments. Several members of the forkhead box (FOX) superfamily of transcription factors have been found to be differentially expressed in the different bladder cancer subtypes. In addition, the FOXA protein family are key regulators of embryonic bladder development and patterning. Both experimental and clinical data support a role for FOXA1 and FOXA2 in urothelial carcinoma. FOXA1 is expressed in embryonic and adult urothelium and its expression is altered in urothelial carcinomas and across disparate molecular bladder cancer subtypes. FOXA2 is normally absent from the adult urothelium, but developmental studies identified FOXA2 as a marker of a transient urothelial progenitor cell population during bladder development. Studies also implicate FOXA2 in bladder cancer and several other FOX proteins might be involved in development and/or progression of this disease; for example, FOXA1 and FOXO3A have been associated with clinical patient outcomes. Future studies should investigate to what extent and by which mechanisms FOX proteins might be directly involved in bladder cancer pathogenesis and treatment responses.


Biomarkers, Tumor/biosynthesis , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Biomarkers, Tumor/genetics , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Urinary Bladder Neoplasms/diagnosis
8.
Hum Mol Genet ; 25(15): 3269-3283, 2016 08 01.
Article En | MEDLINE | ID: mdl-27378691

Predicting response to endocrine therapy and survival in oestrogen receptor positive breast cancer is a significant clinical challenge and novel prognostic biomarkers are needed. Long-range regulators of gene expression are emerging as promising biomarkers and therapeutic targets for human diseases, so we have explored the potential of distal enhancer elements of non-coding RNAs in the prognostication of breast cancer survival. HOTAIR is a long non-coding RNA that is overexpressed, promotes metastasis and is predictive of decreased survival. Here, we describe a long-range transcriptional enhancer of the HOTAIR gene that binds several hormone receptors and associated transcription factors, interacts with the HOTAIR promoter and augments transcription. This enhancer is dependent on Forkhead-Box transcription factors and functionally interacts with a novel alternate HOTAIR promoter. HOTAIR expression is negatively regulated by oestrogen, positively regulated by FOXA1 and FOXM1, and is inversely correlated with oestrogen receptor and directly correlated with FOXM1 in breast tumours. The combination of HOTAIR and FOXM1 enables greater discrimination of endocrine therapy responders and non-responders in patients with oestrogen receptor positive breast cancer. Consistent with this, HOTAIR expression is increased in cell-line models of endocrine resistance. Analysis of breast cancer gene expression data indicates that HOTAIR is co-expressed with FOXA1 and FOXM1 in HER2-enriched tumours, and these factors enhance the prognostic power of HOTAIR in aggressive HER2+ breast tumours. Our study elucidates the transcriptional regulation of HOTAIR, identifies HOTAIR and its regulators as novel biomarkers of patient response to endocrine therapy and corroborates the importance of transcriptional enhancers in cancer.


Biomarkers, Tumor/biosynthesis , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding/biosynthesis , RNA, Neoplasm/biosynthesis , Transcription, Genetic , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Forkhead Box Protein M1/biosynthesis , Forkhead Box Protein M1/genetics , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , MCF-7 Cells , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics
9.
Oncotarget ; 7(30): 47891-47903, 2016 Jul 26.
Article En | MEDLINE | ID: mdl-27374105

GATA transcription factors are essential in mammalian cell lineage determination and have a critical role in cancer development. In cultured prostate cancer cells, GATA2 coordinates with androgen receptor (AR) to regulate gene transcription. In the murine prostate, among six GATA members, GATA2 and GATA3 are expressed. Immunofluorescence staining revealed that both GATA factors predominantly localize in the nuclei of luminal epithelial cells. The pioneer factor FoxA1 is exclusively detected in the luminal cells, whereas AR is detected in both luminal and basal cells. Using genetic engineering, we generated prostate-specific GATA2 and GATA3 knockout (KO) mice. Ablation of single GATA gene had marginal effect on prostate morphology and AR target gene expression, likely due to their genetic compensation. Double KO mice exhibited PIN III to IV lesions, but decreased prostate to body weight ratio, altered AR target gene expression, and expansion of p63-positive basal cells. However, deletion of GATA2 and GATA3 did not reduce the mRNA or protein levels of AR or FoxA1, indicating that GATA factors are not required for AR or FoxA1 expression in adult prostate. Surprisingly, GATA2 and GATA3 exhibit minimal expression in the ventral prostatic (VP) lobe. In contrast, FoxA1 and AR expression levels in VP are at least as high as those in anterior prostatic (AP) and dorsal-lateral prostatic (DLP) lobes. Together, our results indicate that GATA2 and GATA3 are essential for adult murine prostate function and in vivo AR signaling, and the lack of the GATA factor expression in the VP suggests a fundamental difference between VP and other prostatic lobes.


GATA Transcription Factors/metabolism , Prostate/physiology , Receptors, Androgen/metabolism , Animals , GATA Transcription Factors/genetics , Genetic Engineering/methods , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Male , Mice , Mice, Knockout , Prostate/metabolism , Receptors, Androgen/genetics
10.
Urology ; 94: 314.e1-7, 2016 Aug.
Article En | MEDLINE | ID: mdl-27215483

OBJECTIVE: To determine the prognostic significance of Forkhead Box A1 (FOXA1) expression in patients with upper tract urothelial carcinoma (UTUC) undergoing radical nephroureterectomy (RNU). MATERIALS AND METHODS: A retrospective analysis of 566 patients undergoing RNU at seven academic medical centers was performed. Tissue microarrays were subjected to immunohistochemistry using a commercially available polyclonal FOXA1 antibody. Logistic regression determined the association of FOXA1 expression with pathologic features and survival outcomes. RESULTS: Three hundred twenty-two men and 244 women were included. The pathologic distribution of specimens included 53% muscle-invasive or greater (≥pT2), 74% high-grade, 16% with flat architecture, 13% with necrosis, 21% with lymphovascular invasion, 18% with concomitant carcinoma in situ, and 8% with positive lymph nodes. The median FOXA1 score was 5.0 (range: 0-8). Lower FOXA1 expression was significantly correlated with advanced pathologic stage (≥pT3) (P = .02), concomitant carcinoma in situ (P = .006), and renal pelvis (vs ureter) location (P < .0001). At a median follow-up of 27.0 months (range: 3-196), 139 patients (25%) experienced disease recurrence and 121 (21%) died from the disease. In a multivariate model, lower FOXA1 expression was independently associated with disease recurrence (hazard ratio [HR]: 1.11, 95% confidence interval [CI]: 1.05-1.62, P = .04), cancer-specific mortality (HR: 1.17, 95% CI: 1.03-1.92, P = .04), and all-cause mortality (HR: 1.08, 95% CI: 1.02-1.18, P = .05). CONCLUSION: Lower FOXA1 expression is associated with adverse pathologic features and inferior survival outcomes for UTUC patients undergoing RNU. These data indicate lower FOXA1 expression may be a marker of aggressive disease in UTUC.


Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/surgery , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Kidney Neoplasms/metabolism , Kidney Neoplasms/surgery , Nephrectomy , Transcription Factors/biosynthesis , Ureter/surgery , Ureteral Neoplasms/metabolism , Ureteral Neoplasms/surgery , Aged , Carcinoma, Transitional Cell/mortality , Female , Humans , Kidney Neoplasms/mortality , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Ureteral Neoplasms/mortality
11.
Oncotarget ; 7(11): 13139-52, 2016 Mar 15.
Article En | MEDLINE | ID: mdl-26909612

Recent studies have implied that miRNAs may play a crucial role in tumor progression and may be involved in the modulation of some drug resistance in cancer cells. Earlier studies have demonstrated that miR-194 was involved in tumor metastasis and drug resistance in non-small cell lung cancer (NSCLC), whereas their expression and roles on NSCLC still need further elucidation. In the current study, we found that miR-194 is decreased in NSCLC samples compared with adjacent non-cancerous lung samples, and low expression of miR-194 predicts poor patient survival. Both in vitro and in vivo experiments showed that ectopic stable expression miR-194 suppressed proliferation, migration, invasion and metastasis and induced apoptosis in NSCLC cells and that this suppression could be reversed by reintroducing forkhead box A1 (FOXA1), a functional target of miR-194. In addition, miR-194 was downregulated in in cisplatin-resisted human NSCLC cell line-A549/DDP and overexpression of miR-194 increases cisplatin sensitivity. These findings suggested that miR-194 inhibits proliferation and metastasis and reverses cisplatin-resistance of NSCLC cells and may be useful as a new potential therapeutic target for NSCLC.


Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm/genetics , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Lung Neoplasms/pathology , MicroRNAs/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Lung Neoplasms/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness/genetics
12.
Int J Oncol ; 45(5): 2167-75, 2014 Nov.
Article En | MEDLINE | ID: mdl-25175082

Tamoxifen­resistant breast cancer is a major clinical problem and new treatment strategies are highly warranted. In this study, the multitargeting kinase inhibitors sorafenib and nilotinib were investigated as potential new treatment options for tamoxifen­resistant breast cancer. The two compounds inhibited cell growth, reduced expression of total estrogen receptor α (ER), Ser118-phosphorylated ER, FOXA1 and AIB1 and resensitized tamoxifen­resistant cells to tamoxifen. The ER downmodulator fulvestrant exerted strong growth inhibition of tamoxifen­resistant cells and addition of sorafenib and nilotinib could not further suppress growth, showing that sorafenib and nilotinib exerted growth inhibition via ER. In support of this, estradiol prevented sorafenib and nilotinib mediated growth inhibition. These results demonstrate that sorafenib and nilotinib act via ER and ER-associated proteins, indicating that these kinase inhibitors in combination with tamoxifen may be potential new treatments for tamoxifen­resistant breast cancer.


Breast Neoplasms/drug therapy , Estrogen Receptor alpha/biosynthesis , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Pyrimidines/administration & dosage , Tamoxifen/administration & dosage , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Humans , MCF-7 Cells , Niacinamide/administration & dosage , Nuclear Receptor Coactivator 3/biosynthesis , Sorafenib
13.
Clin Lab ; 60(6): 909-18, 2014.
Article En | MEDLINE | ID: mdl-25016694

BACKGROUND: Recent study demonstrated the important contribution of SOX2 to tumorigenesis and metastasis properties of various types of cancers and strongly supported the concept that SOX2 can be used as an effective marker for diagnosis and predicting prognosis of cancer patients. However, our previous RNA-Seq results from human lung cancer cell line A549 showed that some oncogenes, including FOXA1 are negatively regulated by SOX2. METHODS: To further verify the transcriptional regulation effect of SOX2 on FOXA1 and elucidate its application in the diagnosis of human lung and breast cancer, we performed real-time RT-PCR and Western blotting to test the regulation effect of SOX2 on the expression of FOXA1 gene. OncoPrint analysis was used to reveal the alteration of SOX2 and FOXA1 genes in breast invasive carcinoma cases and lung squamous cell carcinoma cases from the Cancer Genome Atlas (TCGA) data portal. Immunohistochemistry staining was performed to test the expression of SOX2 and FOXA1 in human breast and lung carcinoma. RESULTS: The results showed that there is an inhibitory effect of SOX2 on the expression of FOXA1 gene. In addition, these two genes are altered in 5.8% of 484 breast invasive carcinoma cases and 46.4% of 179 lung squamous cell carcinoma cases from the Cancer Genome Atlas (TCGA) data portal, which showed an increased percentage of carcinoma cases when compared with single gene alteration. Immunohistochemistry staining of SOX2 and FOXA1 in human breast and lung carcinoma further revealed the mutual complementary effect of these two proteins in the diagnosis of carcinoma. CONCLUSIONS: Our study revealed SOX2 as a negative upstream regulator for FOXA1 gene and demonstrated SOX2 and FOXA1 as effective dual markers in improving the diagnosis efficiency for human lung and breast tumor.


Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/genetics , Lung Neoplasms/genetics , SOXB1 Transcription Factors/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/metabolism
14.
Breast Cancer Res Treat ; 144(2): 249-61, 2014 Apr.
Article En | MEDLINE | ID: mdl-24549642

Estrogen receptor (ER) is essential for estrogen-dependent growth, and its level of expression is considered a crucial determinant of response to endocrine therapy and prognosis in ER-positive breast cancer. On the other hand, the clinical role of progesterone receptor (PgR) in ER-positive breast cancer remains controversial, although testing of PgR by immunohistochemistry (IHC) has become routine. Recent studies indicated that plasma estradiol levels were related to the expression levels of estrogen-responsive genes in ER-positive breast cancer tissues in both pre- and postmenopausal women. In this study, we analyzed the expression levels of estrogen-responsive genes (PgR and TFF1), a progesterone-responsive gene (RANKL), ER-related genes (FOXA1 and GATA3), HER2, Ki67 and p53 in ER-positive, HER2-negative breast cancer tissues by IHC. Correlations between the expression levels of these molecular markers and clinicopathological factors, including prognosis, were compared between pre- and postmenopausal women. Serum levels of estrone, estradiol, progesterone, and testosterone were also measured. Expression levels of PgR, TFF1, RANKL, and GATA3 were significantly higher in premenopausal women than in postmenopausal women. Serum estradiol levels were positively correlated with Ki67 labeling index (LI) in premenopausal women, but not in postmenopausal women. High expression of FOXA1 and GATA3 was significantly associated with improved disease-free survival in premenopausal women, but not in postmenopausal women, whereas high expression of PgR and low expression of p53 were significantly correlated with the improved disease-free survival in postmenopausal women, but not in premenopausal women. Moreover, the best cutoff points of Ki67 LI for disease-free survival were 30 % for premenopausal women and 14 % for postmenopausal women. Expression levels of ER, TFF1, and RANKL were not associated with the disease-free survival in either pre- or postmenopausal women. Our results suggest that the mechanisms of development and estrogen-dependent growth of ER-positive breast cancer might differ according to menopausal status.


Breast Neoplasms/blood , GATA3 Transcription Factor/biosynthesis , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Disease-Free Survival , Estradiol/blood , Estrone/blood , Female , GATA3 Transcription Factor/genetics , Gene Expression , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Ki-67 Antigen/biosynthesis , Ki-67 Antigen/genetics , Middle Aged , Postmenopause/blood , Premenopause/blood , Progesterone/blood , Prognosis , RANK Ligand/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Testosterone/blood , Trefoil Factor-1 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
15.
Breast Cancer Res ; 15(1): 303, 2013 Feb 18.
Article En | MEDLINE | ID: mdl-23427833

Genome-wide association studies have revealed a multitude of breast cancer-associated SNPs. The majority of these SNPs are located in noncoding regions of the genome. Yet how they contribute to breast cancer development is unknown. Recently, a groundbreaking study by the Lupien group has shown that risk-associated SNPs of breast cancer are enriched for FOXA1 binding sites, which influences the function of this transcription factor.


Breast Neoplasms/genetics , Genome-Wide Association Study , Hepatocyte Nuclear Factor 3-alpha/genetics , Binding Sites , Breast Neoplasms/pathology , Female , Genetic Predisposition to Disease , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Humans , Polymorphism, Single Nucleotide
16.
PLoS One ; 7(5): e36669, 2012.
Article En | MEDLINE | ID: mdl-22590586

Approximately 50% of patients with muscle-invasive bladder cancer (MIBC) develop metastatic disease, which is almost invariably lethal. However, our understanding of pathways that drive aggressive behavior of MIBC is incomplete. Members of the FOXA subfamily of transcription factors are implicated in normal urogenital development and urologic malignancies. FOXA proteins are implicated in normal urothelial differentiation, but their role in bladder cancer is unknown. We examined FOXA expression in commonly used in vitro models of bladder cancer and in human bladder cancer specimens, and used a novel in vivo tissue recombination system to determine the functional significance of FOXA1 expression in bladder cancer. Logistic regression analysis showed decreased FOXA1 expression is associated with increasing tumor stage (p<0.001), and loss of FOXA1 is associated with high histologic grade (p<0.001). Also, we found that bladder urothelium that has undergone keratinizing squamous metaplasia, a precursor to the development of squamous cell carcinoma (SCC) exhibited loss of FOXA1 expression. Furthermore, 81% of cases of SCC of the bladder were negative for FOXA1 staining compared to only 40% of urothelial cell carcinomas. In addition, we showed that a subpopulation of FOXA1 negative urothelial tumor cells are highly proliferative. Knockdown of FOXA1 in RT4 bladder cancer cells resulted in increased expression of UPK1B, UPK2, UPK3A, and UPK3B, decreased E-cadherin expression and significantly increased cell proliferation, while overexpression of FOXA1 in T24 cells increased E-cadherin expression and significantly decreased cell growth and invasion. In vivo recombination of bladder cancer cells engineered to exhibit reduced FOXA1 expression with embryonic rat bladder mesenchyme and subsequent renal capsule engraftment resulted in enhanced tumor proliferation. These findings provide the first evidence linking loss of FOXA1 expression with histological subtypes of MIBC and urothelial cell proliferation, and suggest an important role for FOXA1 in the malignant phenotype of MIBC.


Antigens, Differentiation/biosynthesis , Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Urinary Bladder Neoplasms/metabolism , Urothelium/metabolism , Animals , Cadherins/biosynthesis , Carcinoma, Squamous Cell/pathology , Female , Humans , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Neoplasm Transplantation , Rats , Urinary Bladder Neoplasms/pathology , Urothelium/pathology
17.
Horm Cancer ; 3(4): 147-59, 2012 Aug.
Article En | MEDLINE | ID: mdl-22476979

Breast cancer is primarily a hormone-dependent tumor that can be regulated by the status of the steroid hormones estrogen and progesterone. Forkhead box A1 (FOXA1) is a member of the forkhead box transcription factor family and functions as a pioneer factor of the estrogen receptor (ER) in breast cancer. In the present study, we demonstrate that FOXA1 mRNA was upregulated by estrogen and that estrogen receptor-α (ERα) recruitment to ER-binding sites in the vicinity of the FOXA1 gene was increased by estrogen in ERα-positive MCF-7 breast cancer cells. The estrogen-induced FOXA1 upregulation was repressed by 4-hydroxytamoxifen treatment. We also demonstrated that the proliferation and the migration of MCF-7 cells were decreased by FOXA1-specific small interfering RNA (siRNA; siFOXA1). Furthermore, siFOXA1 decreased the estrogen response element-driven transcription and the estrogen-dependent upregulation of ERα target genes in MCF-7 cells. Next, the immunohistochemical analyses of FOXA1 were performed using two groups of breast cancer specimens. The nuclear immunoreactivity of FOXA1 was detected in 80 (74%) of 108 human invasive breast cancers and was negatively correlated with tumor grade and positively correlated with hormone receptor status, including ERα and progesterone receptor, pathological tumor size, and immunoreactivity of FOXP1, another FOX family transcription factor. FOXA1 immunoreactivity was significantly elevated in the relapse-free breast cancer patients treated with tamoxifen. Notably, the double-positive immunoreactivities of FOXA1 and FOXP1 were significantly associated with a favorable prognosis for the relapse-free and overall survival of patients with tamoxifen-treated breast cancer, with lower P values compared with FOXA1 or FOXP1 immunoreactivity alone. These results suggest that FOXA1 plays an important role in the proliferation and migration of breast cancer cells by modulating estrogen signaling and that the double-positive immunoreactivities of FOXA1 and FOXP1 are associated with a favorable prognosis of tamoxifen-treated breast cancer.


Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Forkhead Transcription Factors/biosynthesis , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Repressor Proteins/biosynthesis , Tamoxifen/therapeutic use , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Growth Processes/drug effects , Cell Movement/drug effects , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Profiling , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/immunology , Humans , Immunohistochemistry , Mice , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Prognosis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rabbits , Repressor Proteins/genetics , Repressor Proteins/immunology , Tamoxifen/pharmacology , Transfection
18.
Oncol Rep ; 24(5): 1233-9, 2010 Nov.
Article En | MEDLINE | ID: mdl-20878115

The classification of breast cancer into multiple molecular subtypes has necessitated the need for biomarkers that can assess tumor progression and the effects of chemopreventive agents on specific breast cancer subtypes. The goal of this study was to identify biomarkers whose expression are altered along with estrogen receptor α (ERα) in the polyoma middle-T antigen (PyMT) transgenic model of breast cancer and to investigate the chemopreventive activity of phenethyl isothiocyanate (PEITC). The diet of PyMT female mice was fortified with PEITC (8 mmol/kg) and the mammary streak and/or gross tumors and metastases in lungs were subjected to immunohistochemical analyses for ERα, FOXA1, and GATA-3. FOXA1 is associated with luminal type A cancers, while GATA-3 is a marker of luminal progenitor cell differentiation. In both control and PEITC-treated groups, there was a progressive loss of ERα and FOXA1 but persistence of GATA-3 expression indicating that the tumors retain luminal phenotype. Overall, the PyMT induced tumors exhibited the entire gamut of phenotypes from ERα+/FOXA1+/GATA-3+ tumors in the early stage to ERα±/FOXA1-/GATA-3+ in the late stage. Thus, PyMT model serves as an excellent model for studying progression of luminal subtype tumors. PEITC treated animals had multiple small tumors, indicating delay in tumor progression. Although these tumors were histologically similar to those in controls, there was a lower expression of these biomarkers in normal luminal cells indicating delay in tumor initiation. In in vitro studies, PEITC depleted AldeFluor-positive putative stem/progenitor cells, which may partly be responsible for the delay in tumor initiation.


Estrogen Receptor alpha/deficiency , Hepatocyte Nuclear Factor 3-alpha/deficiency , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Animals , Anticarcinogenic Agents/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , GATA3 Transcription Factor/biosynthesis , GATA3 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Hepatocyte Nuclear Factor 3-alpha/genetics , Isothiocyanates/pharmacology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Prognosis
19.
Anat Rec (Hoboken) ; 293(6): 938-46, 2010 Jun.
Article En | MEDLINE | ID: mdl-20503388

Pre- and postnatal developmental studies of the lung have provided compelling evidence demonstrating multiple factors that orchestrate alveolar epithelial cell differentiation. The extent to which reactivation of certain developmental pathways in the adult might influence the course of differentiation of alveolar type 2 cells (AT2) into AT1 cells is not known. In this study, we examined selected members of the forkhead (Fox) family of transcription factors and the Wnt (wingless) family of signaling proteins for expression during human alveolar cell differentiation in vitro and determined their potential responses to sulfated components of extracellular matrix (ECM), like those shed from cell surfaces or found in basement membrane and modeled by heparin. Isolated adult human AT2 cells cultured over a 9-day period were used to define the temporal profile of expression of targeted factors during spontaneous differentiation to AT1-like cells. FoxA1 protein was upregulated at early to intermediate time points, where it was strongly elevated by heparin. Gene expression of wnt7A increased dramatically beginning on day 3 and was enhanced even further on days 7 and 9 by heparin, whereas protein expression appeared at days 7 and 9. These temporal changes of expression suggest that sulfated ECMs may act to enhance the increase in FoxA1 at the critical juncture when AT2 cells commence the differentiation process to AT1 cells, in addition to enhancing the increase in wnt7A when the AT1 cell phenotype stabilizes. Collectively, these factors may act to modulate differentiation in the adult human pulmonary alveolus.


Heparin/physiology , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Pulmonary Alveoli/metabolism , Respiratory Mucosa/metabolism , Wnt Proteins/biosynthesis , Adult , Anticoagulants/physiology , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cell Separation , Cells, Cultured , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Time Factors , Wnt Proteins/genetics , beta Catenin/biosynthesis , beta Catenin/genetics
20.
Cancer Res ; 70(5): 2115-25, 2010 Mar 01.
Article En | MEDLINE | ID: mdl-20160041

FOXA1 and FOXA2, members of the forkhead transcription factor family, are critical for epithelial differentiation in many endoderm-derived organs, including the pancreas. However, their role in tumor progression is largely unknown. Here, we identified FOXA1 and FOXA2 as important antagonists of the epithelial-to-mesenchymal transition (EMT) in pancreatic ductal adenocarcinoma (PDA) through their positive regulation of E-cadherin and maintenance of the epithelial phenotype. In human PDA samples, FOXA1/2 are expressed in all epithelia from normal to well-differentiated cancer cells, but are lost in undifferentiated cancer cells. In PDA cell lines, FOXA1/2 expression is consistently suppressed in experimental EMT models and RNAi silencing of FOXA1/2 alone is sufficient to induce EMT. Conversely, ectopic FOXA1/2 expression can potently neutralize several EMT-related E-cadherin repressive mechanisms. Finally, ectopic FOXA2 expression could reactivate E-cadherin expression in a PDA cell line with extensive promoter hypermethylation. In fact, demethylation-mediated reactivation of E-cadherin expression in these cells required concurrent reactivation of endogenous FOXA2 expression. We conclude that suppression of FOXA1/2 expression is both necessary and sufficient for EMT during PDA malignant progression.


Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Hepatocyte Nuclear Factor 3-beta/biosynthesis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Cadherins/biosynthesis , Cadherins/genetics , Cadherins/metabolism , DNA Methylation , Epithelial Cells/pathology , Gene Silencing , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Humans , Immunohistochemistry , Mesoderm/pathology , Pancreatic Neoplasms/genetics , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Transfection
...