Propolis-loaded liposomes (ProLip) enhance macrophage-mediated immunomodulation and suppress breast cancer cell proliferation.
Propolis, a natural remedy derived from bee by-products, is known for its immunomodulatory and anticancer properties. However, its clinical application is hindered by poor solubility and bioavailability. This study formulated a propolis-loaded liposome (ProLip) using the thin-film hydration technique (soy phospholipid-to-cholesterol ratio 6:1) to enhance its therapeutic effect. Encapsulation reduced the particle size of propolis from 402.77 ± 7.53nm to 249.67 ± 5.79nm and enhanced physicochemical properties, including a low polydispersity index (0.098 ± 0.02), highly negative zeta potential (-50.80 ± 0.10 mV), and improved solubility (water contact angle of 50.247°). FTIR analysis confirmed intermolecular interactions between phenolic groups in propolis and phospholipid carbonyl groups, while electron microscopy and surface morphology analysis revealed uniform structure and phagosomal localization in macrophages. Functionally, ProLip enhanced anti-inflammatory cytokine secretion (IL-10: 49.429 ± 0.38 pg/mL; IL-6: 40.488 ± 0.10 pg/mL) and suppressed pro-inflammatory mediators (TNF-α and IL-1β) by > 80%, indicating immunoregulatory potential. Electron microscopy confirmed ProLip internalization within macrophage endocytic compartments and reduced macrophage morphological damage compared to unencapsulated propolis, validating targeted delivery and protection capacity. Additionally, conditioned media from ProLip-treated macrophages significantly induced apoptosis (> 50%) and inhibited migration and invasion in MCF-7 breast cancer cells, supporting immune-mediated anticancer effects. These findings highlight ProLip's potential as a nanocarrier to enhance the bioavailability, cellular targeting, and therapeutic efficacy of stingless bee propolis in cancer immunotherapy.
- # Suppress Breast Cancer Cell Proliferation
- # Thin-film Hydration Technique
- # MCF-7 Breast Cancer Cells
- # Immunoregulatory Potential
- # Cellular Targeting
- # Protection Capacity
- # Migration In Breast Cancer Cells
- # Invasion In Breast Cancer Cells
- # Pro-inflammatory Mediators IL-1β
- # Pro-inflammatory Mediators TNF-α
- Preprint Article
- 10.21203/rs.3.rs-6817917/v1
- Jun 20, 2025
Propolis, a natural remedy derived from bee by-products, is known for its immunomodulatory and anticancer properties. However, its clinical application is hindered by poor solubility and bioavailability. This study formulated a propolis-loaded liposome (ProLip) using the thin-film hydration technique (soy phospholipid-to-cholesterol ratio 6:1) to enhance its therapeutic effect. Encapsulation reduced the particle size of propolis from 402.77 ± 7.53 nm to 249.67 ± 5.79 nm and enhanced physicochemical properties, including a low polydispersity index (0.098 ± 0.02), highly negative zeta potential (-50.80 ± 0.10 mV), and improved solubility (water contact angle of 50.247°). FTIR analysis confirmed intermolecular interactions between phenolic groups in propolis and phospholipid carbonyl groups, while electron microscopy and surface morphology analysis revealed uniform structure and phagosomal localization in macrophages. Functionally, ProLip enhanced the secretion of anti-inflammatory cytokines IL-10 (49.429 ± 0.38 pg/mL) and IL-6 (40.488 ± 0.10 pg/mL), while suppressing pro-inflammatory mediators TNF-α and IL-1β by more than 80% compared to the LPS-treated group, highlighting ProLip as a potential immunoregulatory agent. Electron microscopy confirmed phagosomal localization of ProLip and reduced macrophage morphological damage compared to unencapsulated propolis, validating targeted delivery and protection capacity. Additionally, conditioned media from ProLip-treated macrophages significantly induced apoptosis (>50%) and inhibited migration and invasion in MCF-7 breast cancer cells, supporting immune-mediated anticancer effects. These findings highlight ProLip’s potential as a nanocarrier to enhance the bioavailability, cellular targeting, and therapeutic efficacy of stingless bee propolis in cancer immunotherapy.
- Research Article
38
- 10.1016/j.apjtm.2015.11.007
- Nov 12, 2015
- Asian Pacific Journal of Tropical Medicine
To investigate the expression of targeting protein for Xenopus kinesin-like protein 2 (TPX2) in breast cancer tissue and to explore its role in proliferation, migration and invasion of breast cancer cells. The mRNA and protein expressions of TPX2 in breast cancer tissue and cell lines were assessed by quantitative RT-PCR and Western blot. The effect of TPX2 with RNA interference on proliferation, invasion and migration of breast cancer cells was observed by MTT and Transwell assays. Both mRNA and protein expressions of TPX2 were upregulated in breast cancer tissues compared to tumor-adjacent tissue. TPX2 expression was also upregulated in breast cancer cell lines, and the TPX2 interfered by small interfering RNA could inhibit the proliferation, invasion and migration of breast cancer cells by inhibiting matrix metalloproteinase-2 and matrix metalloproteinase-9. Significantly upregulated TPX2 expression is observed in breast cancer tissue and cells, and contributes to promote the proliferation, migration and invasion of breast cancer cells.
- Research Article
35
- 10.1016/j.prp.2017.12.012
- Jan 5, 2018
- Pathology - Research and Practice
MiR-449a suppresses cell migration and invasion by targeting PLAGL2 in breast cancer
- Research Article
163
- 10.1038/onc.2009.129
- Jun 1, 2009
- Oncogene
Adiponectin is widely known as an adipocytokine with therapeutic potential for its markedly protective function in the pathogenesis of obesity-related disorders, metabolic syndrome, systemic insulin resistance, cardiovascular disease and more recently carcinogenesis. In the present study, we show that adiponectin inhibits adhesion, invasion and migration of breast cancer cells. Further analysis of the underlying molecular mechanisms revealed that adiponectin treatment increased AMP-activated protein kinase (AMPK) phosphorylation and activity as evident by increased phosphorylation of downstream target of AMPK, acetyl-coenzyme A carboxylase and inhibition of p70S6 kinase (S6K). Intriguingly, we discovered that adiponectin treatment increases the expression of tumor suppressor gene LKB1 in breast cancer cells. Overexpression of LKB1 in breast cancer cells further increased adiponectin-mediated phosphorylation of AMPK. Using isogenic LKB1 knockdown cell line pair, we found that LKB1 is required for adiponectin-mediated modulation of AMPK-S6K axis and more importantly, inhibition of adhesion, migration and invasion of breast cancer cells. Taken together these data present a novel mechanism involving specific upregulation of tumor suppressor gene LKB1 by which adiponectin inhibits adhesion, invasion and migration of breast cancer cells. Our findings indicate the possibility of using adiponectin analogues to inhibit invasion and migration of breast cancer cells.
- Research Article
11
- 10.1016/j.clinsp.2022.100115
- Jan 1, 2022
- Clinics (Sao Paulo, Brazil)
BackgroundMicroRNAs (miRNAs) are involved in the progression of diverse human cancers. This work aimed to delve into how microRNA-135a-5p (miR-135a-5p) affects the biological behaviors of Breast Cancer (BC) cells. MethodsGene Expression Omnibus (GEO) datasets were used to analyze the expression differences of miR-135a-5p in cancer tissues of BC patients. Quantitative real-time PCR and western blot were conducted to detect miR-135a-5p and Bcl-2 Associated Athanogene (BAG3) expression levels in BC tissues and cells, respectively. The proliferation, migration, invasion, and cell cycle of BC cells were detected by cell counting kit-8 assay, BrdU assay, wound healing assay, transwell assay, and flow cytometry. The targeted relationship between miR-135a-5p and BAG3 mRNA 3′UTR predicted by bioinformatics was further testified by a dual-luciferase reporter gene assay. Pearson's correlation analysis was adopted to analyze the correlation between miR-135a-5p expression and BAG3 expression. The downstream pathways of BAG3 were analyzed by the LinkedOmics database. ResultsMiR-135a-5p was significantly down-regulated and BAG3 expression was significantly raised in BC tissues. MiR-135a-5p overexpression repressed the viability, migration and invasion of BC cells, and blocked cell cycle progression in G0/G1 phase while inhibiting miR-135a-5p worked oppositely. BAG3 was verified as a target of miR-135a-5p. Overexpression of BAG3 reversed the impacts of miR-135a-5p on the malignant biological behaviors of BC cells. The high expression of BAG3 was associated with the activation of the cell cycle, mTOR and TGF-β signaling pathways. ConclusionMiR-135a-5p regulates BAG3 to repress the growth, migration, invasion, and cell cycle progression of BC cells.
- Abstract
- 10.1016/j.pan.2022.06.179
- Nov 1, 2022
- Pancreatology
Impact of cancer stem cell markers Nestin and Sox2 on cell migration of epithelial and mesenchymal pancreatic cancer cells
- Research Article
- 10.1158/1538-7445.am2020-5259
- Aug 13, 2020
- Cancer Research
Background and Objective: Breast cancer is the malignant tumor with the highest incidence and the second highest mortality among women, and the incidence of breast cancer in China is increasing year by year. The treatment of breast cancer includes radiotherapy, chemotherapy, endocrine therapy, biological targeted therapy and traditional Chinese medicine adjuvant therapy; but the efficacy still needs to be further improved to benefit the patients. Thymoquinone (TQ) is a key component of black grass seed oil and has anti-cancer properties in a variety of tumors. Preliminary studies in our laboratory demonstrated that TQ has a significant inhibitory effect on the migration and invasion of triple negative breast cancer cells. Methods: To find the new target genes for the inhibition of migration and invasion of breast cancer cells by TQ, RNA-sequencing (RNA-seq) was conducted with and without TQ in human breast cancer cell line BT-549 with high invasion and migration characteristics. Quantitative and semi-quantitative RT-PCR was used to further verify the genes regulated by TQ, and then Western Blotting was used to compare the changes of the genes regulated by TQ at the protein level. Finally, the molecular mechanism of TQ inhibiting migration and invasion of human breast cancer cells was investigated by increasing or decreasing the expression of these genes. Results: Eighteen differentially expressed genes, which were closely related to the carcinomas in the experimental group and the control group, were obtained by RNA-seq. HSPA6 gene changed significantly, and showed an up-regulation after adding TQ, which was consistent with the results of RNA-seq. By RT-PCR and Western Blotting from different cell lines (BT549, MCF-7, ZR-75-30, SiHa, Caski), we found that the expression of HSPA6 in the experimental group was higher than that in the control group. The subsequent TQ treatment after knockdown / over-expression will be completed for HSPA6 protein functional effects by TQ. Conclusion: HSPA6 may be a new target gene for TQ to inhibit the migration and invasion of breast cancer cells. The success of this study will provide a new candidate target for clinical treatment of breast cancer. Funding support: National Natural Science Foundation of China (Grant No. 81672887, 81172049) Citation Format: Shiyi Shen, Ting Xiao, Ju Zhou, Chunli Wei, Junjiang Fu. HSPA6 may be a new target by thyraquinone inhibiting migration and invasion of breast cancer cells [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5259.
- Research Article
95
- 10.1186/bcr3128
- Jan 1, 2012
- Breast Cancer Research : BCR
IntroductionHonokiol, a small-molecule polyphenol isolated from magnolia species, is widely known for its therapeutic potential as an antiinflammatory, antithrombosis, and antioxidant agent, and more recently, for its protective function in the pathogenesis of carcinogenesis. In the present study, we sought to examine the effectiveness of honokiol in inhibiting migration and invasion of breast cancer cells and to elucidate the underlying molecular mechanisms.MethodsClonogenicity and three-dimensional colony-formation assays were used to examine breast cancer cell growth with honokiol treatment. The effect of honokiol on invasion and migration of breast cancer cells was evaluated by using Matrigel invasion, scratch-migration, spheroid-migration, and electric cell-substrate impedance sensing (ECIS)-based migration assays. Western blot and immunofluorescence analysis were used to examine activation of the liver kinase B1 (LKB1)-AMP-activated protein kinase (AMPK) axis. Isogenic LKB1-knockdown breast cancer cell line pairs were developed. Functional importance of AMPK activation and LKB1 overexpression in the biologic effects of honokiol was examined by using AMPK-null and AMPK-wild type (WT) immortalized mouse embryonic fibroblasts (MEFs) and isogenic LKB1-knockdown cell line pairs. Finally, mouse xenografts, immunohistochemical and Western blot analysis of tumors were used.ResultsAnalysis of the underlying molecular mechanisms revealed that honokiol treatment increases AMP-activated protein kinase (AMPK) phosphorylation and activity, as evidenced by increased phosphorylation of the downstream target of AMPK, acetyl-coenzyme A carboxylase (ACC) and inhibition of phosphorylation of p70S6kinase (pS6K) and eukaryotic translation initiation factor 4E binding protein 1 (4EBP1). By using AMPK-null and AMPK-WT (MEFs), we found that AMPK is required for honokiol-mediated modulation of pACC-pS6K. Intriguingly, we discovered that honokiol treatment increased the expression and cytoplasmic translocation of tumor-suppressor LKB1 in breast cancer cells. LKB1 knockdown inhibited honokiol-mediated activation of AMPK and, more important, inhibition of migration and invasion of breast cancer cells. Furthermore, honokiol treatment resulted in inhibition of breast tumorigenesis in vivo. Analysis of tumors showed significant increases in the levels of cytoplasmic LKB1 and phospho-AMPK in honokiol-treated tumors.ConclusionsTaken together, these data provide the first in vitro and in vivo evidence of the integral role of the LKB1-AMPK axis in honokiol-mediated inhibition of the invasion and migration of breast cancer cells. In conclusion, honokiol treatment could potentially be a rational therapeutic strategy for breast carcinoma.
- Research Article
62
- 10.4248/br201304007
- Dec 31, 2013
- Bone Research
The aim of this study was to investigate the effects of plumbagin (PL), a naphthoquinone derived from the medicinal plant plumbago zeylanica, on the invasion and migration of human breast cancer cells. Human breast cancer MDA-MB-231SArfp cells were treated with different concentrations of plumbagin for 24 h. The effects of plumbagin on the migration and invasion were observed by a transwell method. The expressions of IL-1α, IL-1β, IL-6, IL-8, TGF-β, TNFα, MMP-2 and MMP-9 mRNA in MDA-MB-231SArfp cells were detected using Real-Time PCR. MDA-MB-231SArfp cells were treated with plumbagin at different concentrations for 45 minutes. The activation of STAT3 was detected by western blot. Following this analysis, STAT3 in MDA-MB-231SArfp cells was knocked out using specific siRNA. mRNA levels of IL-1α, TGF-β, MMP-2 and MMP-9 were then detected. Consequently, MDA-MB-231SArfp cells were injected intracardially into BALB/c nude mice to construct a breast cancer bone metastatic model. The mice were injected intraperitoneally with plumbagin. Non-invasive in vivo monitoring, X-ray imaging and histological staining were performed to investigate the effects of plumbagin on the invasion and migration of breast cancer cells in vivo. The in vitro results showed that plumbagin could suppress the migration and invasion of breast cancer cells and down-regulate mRNA expressions of IL-1α, TGF-β, MMP-2 and MMP-9. Western blotting demonstrated that plumbagin inhibited the activation of STAT3 signaling in MDA-MB-231SArfp cells. The inactivation of STAT3 was found to have an inhibitory effect on the expressions of IL-1α, TGF-β, MMP-2 and MMP-9. In vivo studies showed that plumbagin inhibited the metastasis of breast cancer cells and decreased osteolytic bone metastases, as well as the secretion of MMP-2 and MMP-9 by tumor cells at metastatic lesions. Plumbagin can suppress the invasion and migration of breast cancer cells via the inhibition of STAT3 signaling and by downregulation of IL-1α, TGF-β, MMP-2 and MMP-9.
- Supplementary Content
26
- 10.1089/cbr.2017.2432
- Jul 26, 2018
- Cancer Biotherapy and Radiopharmaceuticals
The long noncoding RNA HOTAIR (HOX transcript antisense intergenic RNA) has been reported to be a biomarker for various malignant tumors; however, its involvement in breast cancer is not fully understood. The aim of this study was to investigate the effects involved with long noncoding RNA HOTAIR and EZH2 (enhancer of zeste homologue 2) on the processes of proliferation, invasion, migration, and apoptosis of breast cancer cells. The expressions of HOTAIR and EZH2 in both normal human mammary epithelial cell (HBL-100) and breast cancer cell lines (MCF-7, MDA-MB-231, and SKBR-3) were detected by means of reverse transcription-quantitative polymerase chain reaction. The MCF-7 cells that exhibited the highest HOTAIR expressions were selected for further studies and divided into the control, negative control, and small interfering RNA-HOTAIR groups. The proliferation, invasion, migration, and apoptosis of breast cancer cells were evaluated by MTT assay, Scratch test, Transwell assay, and flow cytometry, respectively. The combination of HOTAIR with EZH2 and PTEN was predicted by bioinformation, with a dual-luciferase reporter gene assay providing further verification. Initially, lower expressions of HOTAIR and EZH2 in the normal human mammary epithelial cells, while higher expressions in the breast cancer cells of MCF-7, MDA-MB-231, and SKBR-3 were detected. In addition, the downregulation of HOTAIR or silencing of EZH2 was revealed to repress the proliferation, invasion, and migration, while acting to promote the apoptosis of the breast cancer cells. Furthermore, HOTAIR could bind specifically to EZH2 and PTEN, highlighting the capability of HOTAIR to inhibit the expression of PTEN by recruiting EZH2 in breast cancer, while the TCGA database demonstrated the expressions of PTEN were lower in breast cancer cells. The study suggests the higher expressions of HOTAIR and EZH2 among three breast cancer cells. Furthermore, the downregulation of HOTAIR or silencing of EZH2 was noted to inhibit the proliferation, invasion, and migration of breast cancer cells, while promoting their apoptosis.
- Research Article
- 10.12122/j.issn.1673-4254.2025.08.13
- Aug 20, 2025
- Nan fang yi ke da xue xue bao = Journal of Southern Medical University
To investigate the role of circular RNA circ_0000437 in regulating biological behaviors of breast cancer cells and the molecular mechanism. Breast cancer MCF-7 and MDA-MB-231 cells were transfected with sh-circ_0000437, mimics, inhibitor, si-CTPS1, or their respective negative controls. qRT-PCR was used to detect the expression levels of circ_0000437, let-7b-5p, CTPS1, Notch1, Hes1, and Numb in breast cancer cell lines and tissues. RNase R digestion was used to confirm the circular structure of circ_0000437 and its subcellular localization in the breast cancer cells was determined by cellular distribution analysis. The changes in proliferation, invasion and migration of the transfected cells were assessed using CCK-8 assay, Transwell assay and scratch assay. Dual-luciferase reporter gene and RNA immunoprecipitation assays were employed to validate binding interactions among circ_0000437, let-7b-5p, and CTPS1. The cellular expressions of CTPS1, E-cadherin, N-cadherin, and vimentin proteins were detected with Western blotting. A tumor-bearing mouse model was used to verify the oncogenic mechanism of circ_0000437 and CTPS1. Circ_0000437 and CTPS1 were upregulated while let-7b-5p was downregulated in breast cancer tissues and cell lines. Circ_0000437 or CTPS1 knockdown obviously suppressed breast cancer cell proliferation, invasion, migration and epithelial-mesenchymal transition (EMT). Overexpression of let-7b-5p produced similar inhibitory effects, whereas inhibition of let-7b-5p significantly enhanced malignant behaviors of the cells. In the tumor-bearing mouse models, circ_0000437 knockdown significantly suppressed tumor growth, but co-transfection of the cells with pcDNA-CTPS1 accelerated tumor growth. Binding sites were identified between circ_0000437 and let-7b-5p and between let-7b-5p and CTPS1, and circ_0000437, let-7b-5p, and CTPS1 showed functional interactions in breast cancer cells. Circ_0000437 is upregulated in breast cancer tissues and cells, and its high expression promotes proliferation, invasion, migration and EMT of breast cancer cells through the let-7b-5p/CTPS1 axis.
- Research Article
55
- 10.1002/1878-0261.12862
- Dec 7, 2020
- Molecular Oncology
Circular RNAs (circRNAs) have been shown to modulate gene expression and participate in the development of multiple malignancies. The purpose of this study was to investigate the role of circ_0008039 in breast cancer (BC). The expression of circ_0008039, miR-140-3p, and spindle and kinetochore-associated protein 2 (SKA2) was detected by qRT-PCR. Cell viability, colony formation, migration, and invasion were evaluated using methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay, colony formation assay, and transwell assay, respectively. Glucose consumption and lactate production were measured using commercial kits. Protein levels of hexokinase II (HK2) and SKA2 were determined by western blot. The interaction between miR-140-3p and circ_0008039 or SKA2 was verified by dual-luciferase reporter assay. Finally, a mouse xenograft model was established to investigate the roles of circ_0008039 in BC in vivo. We found that circ_0008039 and SKA2 were upregulated in BC tissues and cells, while miR-140-3p was downregulated. Knockdown of circ_0008039 suppressed BC cell proliferation, migration, invasion, and glycolysis. Moreover, miR-140-3p could bind to circ_0008039 and its inhibition reversed the inhibitory effect of circ_0008039 interference on proliferation, migration, invasion, and glycolysis in BC cells. SKA2 was verified as a direct target of miR-140-3p and its overexpression partially inhibited the suppressive effect of miR-140-3p restoration in BC cells. Additionally, circ_0008039 positively regulated SKA2 expression by sponging miR-140-3p. Consistently, silencing circ_0008039 restrained tumor growth via increasing miR-140-3p and decreasing SKA2. In conclusion, circ_0008039 downregulation suppressed BC cell proliferation, migration, invasion, and glycolysis partially through regulating the miR-140-3p/SKA2 axis, providing an important theoretical basis for treatment of BC.
- Research Article
128
- 10.1074/jbc.m110.193714
- Apr 1, 2011
- Journal of Biological Chemistry
Substantial data from clinical trials and epidemiological studies show promising results for use of statins in many cancers, including mammary carcinoma. Breast tumor primarily metastasizes to bone to form osteolytic lesions, causing severe pain and pathological fracture. Here, we report that simvastatin acts as an inhibitor of osteolysis in a mouse model of breast cancer skeletal metastasis of human mammary cancer cell MDA-MB-231, which expresses the mutant p53R280K. Simvastatin and lovastatin attenuated migration and invasion of MDA-MB-231 and BT-20 breast tumor cells in culture. Acquisition of phenotype to express the cancer stem cell marker, CD44, leads to invasive potential of the tumor cells. Interestingly, statins significantly decreased the expression of CD44 protein via a transcriptional mechanism. shRNA-mediated down-regulation of CD44 markedly reduced the migration and invasion of breast cancer cells in culture. We identified that in the MDA-MB-231 cells, simvastatin elevated the levels of mutated p53R280K, which was remarkably active as a transcription factor. shRNA-derived inhibition of mutant p53R280K augmented the expression of CD44, leading to increased migration and invasion. Finally, we demonstrate an inverse correlation between expression of p53 and CD44 in the tumors of mice that received simvastatin. Our results reveal a unique function of statins, which foster enhanced expression of mutant p53R280K to prevent breast cancer cell metastasis to bone.
- Research Article
83
- 10.1007/s13277-015-4342-x
- Nov 4, 2015
- Tumor Biology
Wnt/β-catenin signaling pathway plays a major role in the cancer metastasis. Several microRNAs (miRNAs) are contributed to the inhibition of breast cancer metastasis. Here, we attempted to find novel targets and mechanisms of microRNA-100 (miR-100) in regulating the migration and invasion of breast cancer cells. In this study, we found that miR-100 expression was downregulated in human breast cancer tissues and cell lines. The overexpression of miR-100 inhibited the migration and invasion of MDA-MB-231 breast cancer cells. Inversely, the downregulation of miR-100 increased the migration and invasion of MCF-7 breast cancer cells. Furthermore, FZD-8, a receptor of Wnt/β-catenin signaling pathway, was demonstrated a direct target of miR-100. The overexpression of miR-100 decreased the expression levels not only FZD-8 but also the key components of Wnt/β-catenin pathway, including β-catenin, metalloproteniase-7 (MMP-7), T-cell factor-4 (TCF-4), and lymphoid enhancing factor-1 (LEF-1), and increased the protein expression levels of GSK-3β and p-GSK-3β in MDA-MB-231 cells, and the transfection of miR-100 inhibitor in MCF-7 cells showed the opposite effects. In addition, the expression of miR-100 was negatively correlated with the FZD-8 expression in human breast cancer tissues. Overall, these findings suggest that miR-100 suppresses the migration and invasion of breast cancer cells by targeting FZD-8 and inhibiting Wnt/β-catenin signaling pathway and manipulation of miR-100 may provide a promoting therapeutic strategy for cancer breast treatment.
- Research Article
2
- 10.12122/j.issn.1673-4254.2020.12.06
- Dec 30, 2020
- Nan fang yi ke da xue xue bao = Journal of Southern Medical University
To investigate the expression of ZNF652 in breast cancer tissues and cells and explore its role in breast cancer cell proliferation, invasion and migration. We exploited the data from the TCGA database to analyze the differential expression of ZNF652 in breast cancer tissues and adjacent tissues and the correlations of ZNF652 expression with the clinicopathological characteristics of breast cancer patients including molecular subtypes, pathological types, TNM stages and clinical stages. RT-qPCR and Western blotting were used to detect the expression of ZNF652 in 5 breast cancer cell lines including MCF-7, MDA-MB-231, SK-BR-3, UACC-812 and BT-474. Using a lentivirus system and siRNA technique, we assessed the effects of ZNF652 over-expression and knockdown on proliferation, colony forming ability, migration and invasion of breast cancer cells with CCK-8 assay, clonogenic assay, Transwell assay and wound healing assay. The subcellular localization of ZNF652 in 293T cells was determined using immunofluorescence assay. ZNF652 was significantly up-regulated in breast cancer tissues (P < 0.001). In breast cancer tissues of different molecular types, ZNF652 was down-regulated in TNBC breast cancer tissues but increased in HER2+, Luminal A and Luminal B breast cancer tissues (P < 0.01 or 0.001). The expression of ZNF652 was significantly higher in breast cancer tissues of all pathological types except for mucinous carcinoma than in the adjacent tissues (P < 0.05). The high expression of ZNF652 was closely related to distant metastasis and malignancy of breast cancer (P < 0.01 or 0.001). The mRNA and protein expression levels of ZNF652 was significantly higher in the 5 breast cancer cell lines than in normal breast cells (P < 0.05 or 0.001). Overexpression of ZNF652 promoted the proliferation, invasion and migration of breast cancer cells, while ZNF652 knockdown produced the opposite effects (P < 0.05). Immunofluorescence assay identified subcellular localization of ZNF652 in the nuclei of 293T cells. ZNF652 is highly expressed in breast cancer tissues and cells to promote the development and progression of breast cancer and may serve as a potential molecular target for diagnosis and treatment of the malignancy.
- New
- Research Article
- 10.1038/s41598-025-22646-3
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22635-6
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22867-6
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22758-w
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22854-x
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22866-7
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22895-2
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22651-6
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22678-9
- Nov 6, 2025
- Scientific Reports
- New
- Research Article
- 10.1038/s41598-025-22664-1
- Nov 6, 2025
- Scientific Reports
- Ask R Discovery
- Chat PDF
AI summaries and top papers from 250M+ research sources.