Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Life Sci ; 324: 121745, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37127184

ABSTRACT

AIMS: Circular RNAs (circRNAs) are important regulators in breast cancer progression. However, the underlying mechanism of circRNAs functions in breast cancer remain largely unclear. MAIN METHODS: To investigate the circRNAs expression pattern in breast cancer, high-throughput circRNA microarray assay was used. The top up-regulated circRNA, circZFAND6, was submitted to further experiments, including cell counting kit-8 (CCK-8) assay, colony formation assay, transwell assay and mouse xenograft assay. To investigate the underlying mechanism of circZFAND6 function in breast cancer progression, luciferase reporter assay and RNA immunoprecipitation (RIP) assay were conducted. KEY FINDINGS: We found a novel circRNA, circZFAND6, was up-regulated in breast cancer tissues and cell lines. Inhibition of circZFAND6 reduced proliferation and metastasis of breast cancer. Mechanically, circZFAND6 acted as a competing endogenous RNA (ceRNA) to sponge miR-647 and increase fatty acid synthase (FASN) expression. And eukaryotic translation initiation factor 4A3 (EIF4A3) was found to bind to circZFAND6 pre-mRNA transcript upstream region, leading to the high expression of circZFAND6 in breast cancer. Inhibition of EIF4A3 also suppressed proliferation and metastasis of breast cancer. SIGNIFICANCE: EIF4A3-induced circZFAND6 up-regulation promoted proliferation and metastasis of breast cancer through the miR-647/FASN axis. Our results uncovered a possible mechanism underlying breast cancer progression and might provide a breast cancer treatment target.


Subject(s)
Breast Neoplasms , MicroRNAs , Animals , Female , Humans , Mice , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Fatty Acid Synthase, Type I/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics
2.
Biochem Genet ; 61(1): 336-353, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35918619

ABSTRACT

Abnormal expression of long non-coding RNAs (lncRNAs) is involved in many pathological processes of cancers. However, the role of lncRNA LINC00052 in breast cancer progression is still unclear. Here, LINC00052 expression was detected by in situ hybridization and quantitative real-time PCR assays. Cell Counting Kit-8, wound healing, and transwell assays were used to investigate changes in the proliferation, migration, and invasion of breast cancer cells. MiR-548p was found associated with LINC00052 or Notch2 by RNA pull-down, dual-luciferase reporter, and qRT-PCR assays. The effect of LINC00052 on lung metastasis was explored through in vivo experiments. High LINC00052 expression was observed in breast cancer tissues and cells. LINC00052 silencing inhibited the proliferation, migration, and invasion of MCF7 cells, and LINC00052 overexpression produced the opposite results. MiR-548p, a target gene of LINC00052, partially rescued the effects of LINC00052 on proliferation, migration, and invasion of MCF7. Notch2 was the target of miR-548p and LINC00052 could promote Notch2 expression. Moreover, the phosphorylation of proline-rich tyrosine kinase 2 (Pyk2), a downstream factor of Notch2, was increased by LINC00052, and a Pyk2 mutant could inhibit the cell migration and invasion induced by LINC00052 overexpression in MDA-MB-468 cells, which was similar to the function of the miR-548p mimic. We further demonstrated that LINC00052 exacerbated the metastases of breast cancer cells in vivo. Our research demonstrated that LINC00052 is highly expressed in breast cancer and promotes breast cancer proliferation, migration, and invasion via the miR-548p/Notch2/Pyk2 axis. LINC00052 could serve as a potential therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Female , RNA, Long Noncoding/genetics , MicroRNAs/metabolism , Breast Neoplasms/genetics , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Neoplasm Invasiveness/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Receptor, Notch2/genetics , Receptor, Notch2/metabolism
4.
Mol Ther Oncolytics ; 25: 1-15, 2022 Jun 16.
Article in English | MEDLINE | ID: mdl-35399607

ABSTRACT

The small GTPase ADP-ribosylation factor 6 (ARF6) mediates chemokine (C-C motif) ligand 18 (CCL18)-induced activation of breast cancer (BC) metastasis through its downstream effector AMAP1. However, the molecular mechanisms underlying CCL18 up-regulating ARF6 remain largely unclear. Here, microRNAs (miRNAs) that target ARF6 were predicted and selected in high metastatic BC cells treated with CCL18. Next, we assessed the role of exosomal miR-760 in vitro and in vivo. We further analyzed the expression of ARF6, AMAP1, and phosphorylated (p)-AMAP1 in tumor and adjacent normal tissues. We first observed that CCL18 increased the expression of ARF6 and p-AMAP1 and activated the Src/phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. ARF6 knockdown significantly impaired CCL18-induced malignant cellular behaviors and the Src/PI3K/Akt signaling pathway. Next, ARF6 was confirmed as a target gene of miR-760 in exosomes derived from CCL18-stimulated high metastatic BC cells. Moreover, recipient MCF-7 cells could effectively uptake these miR-760-rich exosomes that significantly promoted proliferation, tumor growth in vivo, migration, invasion, and chemoresistance by activating ARF6-mediated Src/PI3K/Akt signaling and the epithelial-mesenchymal transition (EMT) pathway. Together, our results support that exosomal miR-760 secreted by CCL18-stimulated high metastatic BC cells promoted the malignant behaviors in low metastatic BC cells by up-regulating the ARF6-mediated Src/PI3K/Akt signaling pathway.

5.
J Biol Chem ; 298(4): 101753, 2022 04.
Article in English | MEDLINE | ID: mdl-35189139

ABSTRACT

Despite the enormous successes of anti-PD-1/PD-L1 immunotherapy in multiple other cancer types, the overall response rates of breast cancer remain suboptimal. Therefore, exploring additional immune checkpoint molecules for potential cancer treatment is crucial. B7H3, a T-cell coinhibitory molecule, is specifically overexpressed in breast cancer compared with normal breast tissue and benign lesions, making it an attractive therapeutic target. However, the mechanism by which B7H3 contributes to the cancer phenotype is unclear. Here we show that the expression of B7H3 is negatively related to the number of CD8+ T cells in breast tumor sites. In addition, analysis of the differentially expressed B7H3 reveals that it is inversely correlated to autophagic flux both in breast cancer cell lines and clinical tumor tissues. Furthermore, block of autophagy by bafilomycin A1 (Baf A1) increases B7H3 levels and attenuates CD8+ T cell activation, while promotion of autophagy by V9302, a small-molecule inhibitor of glutamine metabolism, decreases B7H3 expression and enhances granzyme B (GzB) production of CD8+ T cells via regulation of reactive oxygen species (ROS) accumulation. We demonstrate that combined treatment with V9302 and anti-PD-1 monoclonal antibody (mAb) enhances antitumor immunity in syngeneic mouse models. Collectively, our findings unveil the beneficial effect of V9302 in boosting antitumor immune response in breast cancer and illustrate that anti-PD-1 together with V9302 treatment may provide synergistic effects in the treatment of patients insensitive to anti-PD-1 therapy.


Subject(s)
B7 Antigens , Breast Neoplasms , CD8-Positive T-Lymphocytes , Glutamine , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Autophagy , B7 Antigens/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Cell Line, Tumor , Female , Glutamine/antagonists & inhibitors , Humans , Mice , Reactive Oxygen Species
6.
Front Oncol ; 11: 681905, 2021.
Article in English | MEDLINE | ID: mdl-34692474

ABSTRACT

BACKGROUND: Breast cancer patients who achieve pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) have favorable outcomes. Reliable predictors for pCR help to identify patients who will benefit most from NAC. The pretreatment serum albumin-to-alkaline phosphatase ratio (AAPR) has been shown to be a prognostic predictor in several malignancies, but its predictive value for pCR in breast cancer is still unknown. This study aims to investigate the predictive role of AAPR in breast cancer patients and develop an AAPR-based nomogram for pCR rate prediction. METHODS: A total of 780 patients who received anthracycline and taxane-based NAC from January 2012 to March 2018 were retrospectively analyzed. Univariate and multivariate analyses were performed to assess the predictive value of AAPR and other clinicopathological factors. A nomogram was developed and calibrated based on multivariate logistic regression. A validation cohort of 234 patients was utilized to further validate the predictive performance of the model. The C-index, calibration plots and decision curve analysis (DCA) were used to evaluate the discrimination, calibration and clinical value of the model. RESULTS: Patients with a lower AAPR (<0.583) had a significantly reduced pCR rate (OR 2.228, 95% CI 1.246-3.986, p=0.007). Tumor size, clinical nodal status, histological grade, PR, Ki67 and AAPR were identified as independent predictors and included in the final model. The nomogram was used as a graphical representation of the model. The nomogram had satisfactory calibration and discrimination in both the training cohort and validation cohort (the C-index was 0.792 in the training cohort and 0.790 in the validation cohort). Furthermore, DCA indicated a clinical net benefit from the nomogram. CONCLUSIONS: Pretreatment serum AAPR is a potentially valuable predictor for pCR in breast cancer patients who receive NAC. The AAPR-based nomogram is a noninvasive tool with favorable predictive accuracy for pCR, which helps to make individualized treatment strategy decisions.

SELECTION OF CITATIONS
SEARCH DETAIL
...