Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Cancer ; 119: 182-192, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29937544

ABSTRACT

BACKGROUND: Surgery combined with new adjuvant chemotherapy is the primary treatment for early stage invasive and advanced stage breast cancer. Growing evidence indicates that patients with ERα-positive breast cancer show poor response to chemotherapeutics. However, ERα-mediated drug-resistant mechanisms remain unclear. METHODS: Levels of WW domain-binding protein 2 (WBP2) and drug-resistant gene were determined by western blotting and RT-PCR, respectively. Cell viability was measured by preforming MTT assay. CD243 expression and apoptosis rate were evaluated by flow cytometry. Interactions of WBP2/ERα and ERα/MDR1 were detected by co-immunoprecipitation and chromatin immunoprecipitation (ChIP) assay, respectively. RESULTS: There was an intrinsic link between WBP2 and ERα in drug-resistant cancer cells. Upregulation of WBP2 in MCF7 cells increased the chemoresistance to doxorubicin, while RNAi-mediated knockdown of WBP2 in MCF7/ADR cells sensitised the cancer cells to doxorubicin. Further investigation in in vitro and in vivo models demonstrated that WBP2 expression was directly correlated with MDR1, and WBP2 could directly modulate MDR1 transcription through binding to ERα, resulting in increased chemotherapy drug resistance. CONCLUSIONS: Our finding provides a new mechanism for the chemotherapy response of ERα-positive breast tumours, and WBP2 might be a key molecule for developing new therapeutic strategies to treat chemoresistance in breast cancer patients.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Breast Neoplasms/drug therapy , Estrogen Receptor alpha/physiology , ATP Binding Cassette Transporter, Subfamily B/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Breast Neoplasms/pathology , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Estrogen Receptor alpha/genetics , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Trans-Activators , Transcription, Genetic
2.
Cancer Res ; 78(17): 4853-4864, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29898994

ABSTRACT

p62 is a receptor that facilitates selective autophagy by interacting simultaneously with cargoes and LC3 protein on the autophagosome to maintain cellular homeostasis. However, the regulatory mechanism(s) behind this process and its association with breast cancer remain to be elucidated. Here, we report that Flightless-I (FliI), a novel p62-interacting protein, promotes breast cancer progression by impeding selective autophagy. FliI was highly expressed in clinical breast cancer samples, and heterozygous deletion of FliI retarded the development of mammary tumors in PyVT mice. FliI induced p62-recruited cargoes into Triton X-100 insoluble fractions (TI) to form aggregates, thereby blocking p62 recognition of LC3 and hindering p62-dependent selective autophagy. This function of Flil was reinforced by Akt-mediated phosphorylation at Ser436 and inhibited by phosphorylation of Ulk1 at Ser64. Obstruction of autophagic clearance of p62-recruited cargoes by FliI was associated with the accumulation of oxidative damage on proteins and DNA, which could contribute to the development of cancer. Heterozygous knockout of FliI facilitated selectively autophagic clearance of aggregates, abatement of ROS levels, and protein oxidative damage, ultimately retarding mammary cancer progression. In clinical breast cancer samples, Akt-mediated phosphorylation of FliI at Ser436 negatively correlated with long-term prognosis, while Ulk1-induced FliI phosphorylation at Ser64 positively correlated with clinical outcome. Together, this work demonstrates that FliI functions as a checkpoint protein for selective autophagy in the crosstalk between FliI and p62-recruited cargoes, and its phosphorylation may serve as a prognostic marker for breast cancer.Significance: Flightless-I functions as a checkpoint protein for selective autophagy by interacting with p62 to block its recognition of LC3, leading to tumorigenesis in breast cancer.Cancer Res; 78(17); 4853-64. ©2018 AACR.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , RNA-Binding Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Adult , Aged , Animals , Autophagosomes/metabolism , Autophagosomes/pathology , Autophagy/genetics , Autophagy-Related Protein-1 Homolog/genetics , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Disease Progression , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Middle Aged , Phosphorylation , Protein Binding/genetics , Trans-Activators
3.
Oncol Rep ; 38(3): 1533-1542, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28731154

ABSTRACT

Hepatocellular carcinoma (HCC) is the second leading cause of cancer mortality worldwide, which is mainly due to relapse and metastasis. Synaptophysin-like 1 (SYPL1), a member of SYP family proteins, exerts complicated functions, which prompted us to wonder whether SYPL1 contributed to HCC progress. Herein, we performed integrative experiments of quantitative real-time polymerase chain reaction (qRT-PCR), western blot analysis and immunohistochemistry (IHC), and found that SYPL1 overexpression in HCC tissues was closely correlated with several malignant clinicopathologic features of HCC. The results from IHC in serial sections of HCC tissues further indicated that SYPL1 expression was associated with epithelial-mesenchymal transition (EMT) biomarkers of HCC cells. Additionally, Kaplan-Meier survival analysis showed that SYPL1 overexpression was significantly associated with reduced overall survival (OS) (p<0.001) and disease-free survival (DFS) (p=0.002). Furthermore, univariate and multivariate Cox proportional hazards analysis identified SYPL1 as an independent prognostic factor for OS [hazard ratio (HR), 2.443, 95% confidence interval (CI), 1.429-4.177, p=0.001] and DFS (HR, 1.680, 95% CI=1.012-2.788, p=0.045) of HCC patients. Collectively, SYPL1 overexpression predicts poor prognosis of HCC and may associate with EMT of HCC cells. Therefore, SYPL1 could serve as a future novel biomarker and potential therapy target for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Epithelial-Mesenchymal Transition/genetics , Liver Neoplasms/genetics , Synaptophysin/genetics , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Kaplan-Meier Estimate , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Staging/methods , Prognosis , Proportional Hazards Models , Up-Regulation/genetics
4.
Oncotarget ; 8(24): 39401-39416, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28455959

ABSTRACT

Great progress has been achieved in the study of the role of TGF-ß signaling in triggering epithelial-mesenchymal transition (EMT) in a variety of cancers; however, the regulation of TGF-ß signaling during EMT in mammary tumor metastasis has not been completely defined. In the present study, we demonstrated that OVOL2, a zinc finger transcription factor, inhibits TGF-ß signaling-induced EMT in mouse and human mammary tumor cells, as well as in mouse tumor models. Data from the Oncomine databases indicated a strong negative relationship between OVOL2 expression and breast cancer progression. Moreover, our experiments revealed that OVOL2 inhibits TGF-ß signaling at multiple levels, including inhibiting Smad4 mRNA expression and inducing Smad7 mRNA expression, blocking the binding between Smad4 and target DNA, and interfering with complex formation between Smad4 and Smad2/3. These findings reveal a novel mechanism that controls the TGF-ß signaling output level in vitro and in vivo. The modulation of these molecular processes may represent a strategy for inhibiting breast cancer invasion by restoring OVOL2 expression.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Signal Transduction , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Biomarkers , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA-Binding Proteins/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Kaplan-Meier Estimate , Mice , Neoplasm Metastasis , Prognosis , Protein Binding , Smad4 Protein/genetics , Smad4 Protein/metabolism , Smad7 Protein/genetics , Smad7 Protein/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
5.
PLoS One ; 9(5): e97804, 2014.
Article in English | MEDLINE | ID: mdl-24845596

ABSTRACT

BACKGROUND: Matrix metalloproteinase 9 (MMP-9) is a type-IV collagenase that is highly expressed in breast cancer, but its exact role in tumor progression and metastasis is unclear. METHODS: MMP-9 mRNA and protein expression was examined by real-time reverse transcriptase PCR and immunohistochemical staining, respectively, in 41 breast cancer specimens with matched peritumoral benign breast epithelial tissue and suspicious metastatic axillary lymph nodes. Lymph vessels were labeled with D2-40 and lymphatic microvessel density (LMVD) was calculated. Correlation of MMP-9 protein expression with clinicopathological parameters and LMVD was also evaluated. RESULTS: MMP-9(+) staining in breast cancer specimens (35/41, 85.4%) was higher than in matched epithelium (21/41, 51.2%; P<0.05) and lymph nodes (13/41, 31.7%; P<0.001). Higher MMP-9 mRNA expression was also detected in tumor specimens compared with matched epithelial tissues and lymph nodes (P<0.05). Elevated MMP-9 expression was correlated with lymph node metastasis and LMVD (P<0.05). CONCLUSION: MMP-9 was overexpressed in breast cancer specimens compared with peritumoral benign breast epithelium and lymph nodes. Moreover, its expression in the matched epithelium and lymph nodes was positively associated with lymph node metastasis, and its expression in lymph nodes was positively associated with lymphangiogenesis in breast cancer. Thus, MMP-9 is a potential marker for breast cancer progression.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 9/genetics , Adult , Aged , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Lymphatic Vessels/pathology , Matrix Metalloproteinase 9/metabolism , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , RNA, Messenger/genetics
6.
J Cell Sci ; 126(Pt 13): 2877-89, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23613467

ABSTRACT

Wnt signalling through ß-catenin and the lymphoid-enhancing factor 1/T-cell factor (LEF1/TCF) family of transcription factors maintains stem cell properties in both normal and malignant tissues; however, the underlying molecular pathway involved in this process has not been completely defined. Using a microRNA microarray screening assay, we identified let-7 miRNAs as downstream targets of the Wnt-ß-catenin pathway. Expression studies indicated that the Wnt-ß-catenin pathway suppresses mature let-7 miRNAs but not the primary transcripts, which suggests a post-transcriptional regulation of repression. Furthermore, we identified Lin28, a negative let-7 biogenesis regulator, as a novel direct downstream target of the Wnt-ß-catenin pathway. Loss of function of Lin28 impairs Wnt-ß-catenin-pathway-mediated let-7 inhibition and breast cancer stem cell expansion; enforced expression of let-7 blocks the Wnt-ß-catenin pathway-stimulated breast cancer stem cell phenotype. Finally, we demonstrated that the Wnt-ß-catenin pathway induces Lin28 upregulation and let-7 downregulation in both cancer samples and mouse tumour models. Moreover, the delivery of a modified lin28 siRNA or a let-7a agomir into the premalignant mammary tissues of MMTV-wnt-1 mice resulted in a complete rescue of the stem cell phenotype driven by the Wnt-ß-catenin pathway. These findings highlight a pivotal role for Lin28/let-7 in Wnt-ß-catenin-pathway-mediated cellular phenotypes. Thus, the Wnt-ß-catenin pathway, Lin28 and let-7 miRNAs, three of the most crucial stem cell regulators, connect in one signal cascade.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/genetics , Wnt1 Protein/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Female , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , Neoplastic Stem Cells/pathology , RNA-Binding Proteins/genetics , Transcriptional Activation , Wnt1 Protein/genetics , beta Catenin/genetics
7.
BMC Cancer ; 12: 47, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22284579

ABSTRACT

BACKGROUND: Extracellular matrix protein 1 (ECM1) and vascular endothelial growth factor-C (VEGF-C) are secretory glycoproteins that are associated with lymphangiogenesis; these proteins could, therefore, play important roles in the lymphatic dissemination of tumors. However, very little is known about their potential roles in lymphangiogenesis. The aim of this study was to investigate whether correlations exist between ECM1 and VEGF-C in human breast cancer, lymphangiogenesis, and the clinicopathological characteristics of the disease. METHODS: ECM1 and VEGF-C mRNA and protein expression levels in 41 patients were investigated using real-time reverse transcriptase polymerase chain reaction (RT-PCR), or immunohistochemical (IHC) staining of breast cancer tissue, matched noncancerous breast epithelial tissues, and suspicious metastatic axillary lymph nodes. D2-40 labelled lymph vessels and lymphatic microvessel density (LMVD) were counted. Correlations between ECM1 or VEGF-C protein expression levels, LMVD, and clinicopathological parameters were statistically tested. RESULTS: The rate of ECM1 positive staining in breast cancer tissues was higher (31/41, 75.6%) than that in the corresponding epithelial tissues (4/41, 9.8%, P < 0.001) and lymph nodes (13/41, 31.7%, P < 0.001). Similarly, the VEGF-C expression rate in cancer specimens was higher (33/41, 80.5%) than in epithelial tissues (19/41, 46.3%, P < 0.01) or lymph nodes (15/41, 36.6%, P < 0.01). Higher ECM1 and VEGF-C mRNA expression levels were also detected in the tumor tissues, compared to the non-cancerous tissue types or lymph nodes (P < 0.05). ECM1 protein expression was positively correlated with the estrogen receptor status (P < 0.05) and LMVD (P < 0.05). LMVD in the ECM1- and VEGF-C-positive tumor specimens was higher than that in the tissue types with negative staining (P < 0.05). CONCLUSIONS: Both ECM1 and VEGF-C were overexpressed in breast cancer tissue samples. ECM1 expression was positively correlated with estrogen responsiveness and the metastatic properties of breast cancer. We conclude, therefore, that ECM1 and VEGF-C may have a synergistic effect on lymphangiogenesis to facilitate lymphatic metastasis of breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Extracellular Matrix Proteins/metabolism , Lymphatic Vessels/metabolism , Vascular Endothelial Growth Factor C/metabolism , Adult , Aged , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Female , Humans , Immunohistochemistry , Lymphangiogenesis , Lymphatic Metastasis , Lymphatic Vessels/pathology , Microvessels/metabolism , Microvessels/pathology , Middle Aged , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...