Antiproliferative and cytotoxic activity of Papaver somniferum seed oil on human breast cancer cell lines
Background: This study aimed to determine the antiproliferative and cytotoxic activities of Papaver somniferum seed oil on human breast cancer (MDA-MB-231) cell lines under in vitro conditions. Methods: Poppy seed oil extracted by cold press method were subjected to compositional analysis using GC-MS. In the study, control group, DMSO (0.1%) applied group and 5 different concentrations of Poppy seed oil applied group three main treatment groups were created. Cell viability was investigated by the trypan blue method. Some pharmacokinetic properties and toxicity profiles of the identified molecules were predicted using a Swiss ADME online server and ProTox-II webserver. Results: The effect of Poppy seed oil on breast cancer cells increased relatively starting from the lowest concentration (87 µg/ml), and it was determined that the effect became more pronounced after a concentration of 350 µg/ml. Except Hexadecanoic acid and (E)-9- Octadecanoic acid, all other compounds were predicted to be non-hepatotoxic, non-carcinogenic, non-cytotoxic, non-immunotoxic and non-mutagenic. All compounds investigated are orally absorbable, suitable for dermal trans-distribution, have high gastrointestinal absorption, however, Hexadecanoic acid is permeable to the blood-brain barrier (BBB), while the other 4 compounds are not. The bioavailability values of all compounds investigated can be considered to be very well absorbed by the body. Conclusion: In this study, it was determined that P. somniferum seed oil has antiproliferative and cytotoxic activity on breast cancer cells in vitro. P. somniforum seed oil may be a natural anticancer agent to treat breast cancer, but more studies are needed on this subject.
454
- 10.1016/j.cmet.2004.12.008
- Feb 1, 2005
- Cell Metabolism
7
- 10.3390/plants12112105
- May 25, 2023
- Plants
107
- 10.1016/0278-6915(92)90180-s
- Nov 1, 1992
- Food and Chemical Toxicology
11
- 10.18596/jotcsa.43273
- Aug 26, 2016
- Journal of the Turkish Chemical Society, Section A: Chemistry
22
- 10.1097/mol.0b013e3283613a7d
- Jun 1, 2013
- Current Opinion in Lipidology
15
- 10.1002/9780470650622.ch7
- Oct 2, 1996
4
- 10.15237/gida.gd20061
- Aug 19, 2020
- Gıda
265
- 10.1023/b:pham.0000041444.06122.8d
- Sep 1, 2004
- Pharmaceutical Research
18
- 10.1007/s11094-018-1839-9
- Aug 1, 2018
- Pharmaceutical Chemistry Journal
65
- 10.3389/fchem.2020.00737
- Aug 27, 2020
- Frontiers in Chemistry
- Research Article
46
- 10.1074/mcp.m110.002170
- Jan 1, 2011
- Molecular & Cellular Proteomics
The present study addresses, by transcriptomics and quantitative stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics, the estrogen receptor α (ERα) and β (ERβ)-mediated effects on gene and protein expression in T47D breast cancer cells exposed to the phytoestrogen genistein. Using the T47D human breast cancer cell line with tetracycline-dependent ERβ expression (T47D-ERβ), the effect of a varying intracellular ERα/ERβ ratio on genistein-induced gene and protein expression was characterized. Results obtained reveal that in ERα-expressing T47D-ERβ cells with inhibited ERβ expression genistein induces transcriptomics and proteomics signatures pointing at rapid cell growth and migration by dynamic activation of cytoskeleton remodeling. The data reveal an interplay between integrins, focal adhesion kinase, CDC42, and actin cytoskeleton signaling cascades, occurring upon genistein treatment, in the T47D-ERβ breast cancer cells with low levels of ERα and no expression of ERβ. In addition, data from our study indicate that ERβ-mediated gene and protein expression counteracts ERα-mediated effects because in T47D-ERβ cells expressing ERβ and exposed to genistein transcriptomics and proteomics signatures pointing at a clear down-regulation of cell growth and induction of cell cycle arrest and apoptosis were demonstrated. These results suggest that ERβ decreases cell motility and metastatic potential as well as cell survival of the breast cancer cell line. It is concluded that the effects of genistein on proteomics and transcriptomics end points in the T47D-ERβ cell model are comparable with those reported previously for estradiol with the ultimate estrogenic effect being dependent on the relative affinity for both receptors and on the receptor phenotype (ERα/ERβ ratio) in the cells or tissue of interest.
- Research Article
47
- 10.1006/mthe.2002.0680
- Sep 1, 2002
- Molecular Therapy
Adenoviral vectors with E1A regulated by tumor-specific promoters are selectively cytolytic for breast cancer and melanoma.
- Research Article
- 10.1158/1538-7445.sabcs17-p2-08-03
- Feb 14, 2018
- Cancer Research
Background- GRB7 gene encodes a multi-domain signal transduction molecule and is part of the core of the HER-2 amplicon. GRB7 is commonly co-amplified and over-expressed with HER-2 in human breast cancer. Earlier studies found a functional role of GRB7 in breast cancer. The role of GRB7 in HER-2 positive human breast cancer resistant to HER-2 targeted therapy remains unexplored however. Materials and Methods- HCC-1954, 21MT1 and JimT1 are human HER-2 positive breast cancer cell lines that are resistant to trastuzumab and lapatinib treatment. Transient knock down of GRB7 protein expression was achieved with siRNA transfection and stable knock down with lentiviral vector mediated shRNA over-expression. Cell lines transfected with non-targeting siRNA or shRNA serve as negative controls. Knock down of GRB7 protein expression is verified by Western blotting. The growth of human breast cancer cell lines after GRB7 knock down in vitro is measured with the CellTiter Glo assay as well as the Incucyte live cell imaging. Activation status of specific signaling pathways was examined with phospho-specific antibody by immune-blotting and immune-precipitation. To assess the growth promoting function of GRB7 in human breast cancer cell lines in vivo, polyclonal HCC-1954, 21MT1 and JimT1 cells, with GRB7 knock down or their corresponding negative control, were orthotopically injected into the mammary fat pads of female immune-deficient NSG mice. The growth rates of these tumors, measured serially with caliper, and final tumor weights were compared between GRB7 knock down and the negative control. The proliferation rate and apoptosis of these tumors were studied with ki-67 staining and Tunel assay.The effects of GRB7 knock down on signaling were investigated with a proteome profiler receptor tyrosine kinase kit (R&D). The role of signaling molecules differentially activated in the growth of breast cancer cells by GRB7 knock down was examined utilizing siRNA mediated knock down, and antibody and small molecule inhibitors. Results- GRB7 knock down decreased the growth of HCC-1954, 21MT1 and JimT1 cells in vitro and the growth of tumor xenograft these cells formed in animal models. When assayed by ki67 staining and Tunel assay, the mechanism of reduced tumor xenograft growth appeared to be distinct. Reduced proliferation and increased apoptosis were seen in 21MT1 cells, while reduced proliferation was seen in HCC-1954 cells and increased apoptosis in JimT1 cells. Protein profiling found that tyrosine phosphorylation of candidate signaling molecules was reduced with GRB7 knock down in JimT1 cells. Immuno-blotting and immuno-precipitation experiments were performed to evaluate these effects in other cell lines. The effect of targeting these molecules in breast cancer cell growth by siRNA and inhibitors is being examined. Discussion- GRB7 has essential growth promoting function in therapy resistant HER-2 positive human breast cancer cells. GRB7 knock down has pleiotropic effects on signaling in various cellular contexts. The potential of targeting GRB7 signaling in treating therapy resistant HER-2 positive breast cancer merits further study. Citation Format: Luoh S-W, Wagoner W, Lai X, Hu Z, Chin K, Ramsey B. An essential role of GRB7 in promoting the growth of therapy resistant HER-2 positive human breast cancer cells in culture and animal models [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P2-08-03.
- Research Article
- 10.1158/1538-7445.am2018-1852
- Jul 1, 2018
- Cancer Research
Background: The proliferative and pro-survival signals driven by the insulin-like growth factor (IGF) ligands, IGF-1 and IGF-2, are transmitted through their binding to the IGF-1 receptor (IGF-1R). In addition, IGF-2 promotes proliferation through activation of the insulin receptor variant A (IR-A) that is expressed during embryonic development as well as in many cancers. IGF survival signaling has been implicated in driving resistance to cancer therapies with diverse mechanisms of action, due to cross-talk between cellular signaling networks. Recent studies[1][2] suggest that the combination of IGF-1R signaling inhibitors with CDK4/6 inhibitors can result in enhanced anti-tumor activity. The aim of this study was to explore the potential of the IGF-1/-2 ligand blocking antibody, xentuzumab (BI 836845[3]), to enhance the anti-tumor activity of the CDK4/6 inhibitor abemaciclib, alone or in combination with fulvestrant, in human breast cancer (BC) cell lines. Methods: The anti-proliferative activity of the xentuzumab/abemaciclib combination was evaluated using CellTiter-Glo and propidium iodide staining in a panel of 51 and 20 BC cell lines, respectively. Detailed studies of abemaciclib (+/- fulvestrant), xentuzumab and combinations thereof were performed in MCF7 cells. Cell cycle analysis was done by FACS and BrdU ELISA, cellular signaling was assessed by Western blotting, proliferation was evaluated by Incucyte, CellTiter-Glo and alamarBlue assay. Apoptosis was measured by detection of cleaved PARP and caspase 3. Results: Among a panel of BC cell lines, enhanced anti-proliferative activity of xentuzumab+abemaciclib vs. abemaciclib alone was observed specifically in hormone receptor positive (HR+) cell lines. Combined treatment resulted in more pronounced cell cycle arrest in MCF7 cells, associated with synergistic blockade of IGF survival signaling and suppression of cell cycle genes downstream of CDK4/6. The triple combination with fulvestrant more effectively inhibited tumor cell proliferation than the doublet abemaciclib+fulvestrant, and led to induction of apoptosis. Conclusion: The study results show that addition of the IGF-1/-2 neutralizing antibody xentuzumab to abemaciclib, in the absence or presence of fulvestrant, leads to improved anti-proliferative activity and, in the triple combination, results in cellular death in MCF7 HR+ breast cancer cells. A phase Ib trial evaluating the abemaciclib+xentuzumab combination, including triplets with endocrine therapy in HR+BC patients, is currently ongoing.
- Research Article
- 10.3760/cma.j.issn.1001-9030.2013.04.009
- Apr 8, 2013
- Chinese journal of experimental surgery
Objective To explore the role of miR-483-5p in breast cancer,and investigate the expression of miR-483-5p in human normal breast skin cells (CCD-1095Sk),human breast cancer cell lines (MCF-7,MDA-MB-231),paired breast cancer tissues and adjacent non-tumor breast tissues.Methods The expression level of miR-483-5p in human normal breast skin cells (CCD-1095Sk),human breast cancer cell lines (MCF-7,MDA-MB-231),paired breast cancer tissues and adjacent non-tumor breast tissues was detected by using reverse transcription polymerase chain reaction (RT-PCR).MDA-MB-231 cells were transfected with miR-483-5p mimics and subjected to proliferation,cell cycle,apoptosis,and Transwell assays.Results The relative expression of miR-483-5p in human breast cancer specimens was 0.6333 ±0.1898,which was significantly lower than 1.4471 ±0.3908 of adjacent non-tumor breast tissues.miR-483-5p was also down-regulated in human breast cancer cell lines (MCF-7,MDA-MB-231) as compared with normal breast skin cells (CCD-1095Sk) (0.3290 ± 0.0219 and 0.2307 ± 0.0144 vs.1.0000 ±0.0000,both P < 0.05).miR-483-5p mimics-transfected cells exhibited significantly reduced cell proliferation and the proliferation rate was 65.68%.Transwell assay revealed that the amount of invaded MDA-MB-231 cells in miR-483-5p transfected groups was significantly declined as compared with the control and scramble groups (89.8 ± 12.4 vs.203.8 ± 12.0 and 199.3 ± 13.1,both P <0.05).In comparison to control group,cell cycle was arrested in G0/G1 phase [(55.1 ± 5.2) %] and there was no significant difference in apoptosis of cells transfected with miR-483-5p mimics.Conclusion miR-483-5p,which is down-regulated in human breast cancer specimens and cell lines,can suppress proliferation and invasive ability in vitro. Key words: Breast cancer; miR-483-5p; Proliferation; Invasion
- Research Article
42
- 10.1074/jbc.m113.508994
- Feb 1, 2014
- Journal of Biological Chemistry
PDGF-C, which is abundant in the malignant breast tumor microenvironment, plays an important role in cell growth and survival. Because tumor-associated macrophages (TAMs) contribute to cancer malignancy, macrophage survival mechanisms are an attractive area of research into controlling tumor progression. In this study, we investigated PDGF-C-mediated signaling pathways involved in anti-apoptotic effects in macrophages. We found that the human malignant breast cancer cell line MDA-MB-231 produced high quantities of PDGF-C, whereas benign MCF-7 cells did not. Recombinant PDGF-C induced PDGF receptor α chain phosphorylation, followed by Akt and Bad phosphorylation in THP-1-derived macrophages. MDA-MB-231 culture supernatants also activated macrophage PDGF-Rα. PDGF-C prevented staurosporine-induced macrophage apoptosis by inhibiting the activation of caspase-3, -7, -8, and -9 and cleavage of poly(ADP-ribose) polymerase. Finally, TAMs isolated from the PDGF-C knockdown murine breast cancer cell line 4T1 and PDGF-C knockdown MDA-MB-231-derived tumor mass showed higher rates of apoptosis than the respective WT controls. Collectively, our results suggest that tumor cell-derived PDGF-C enhances TAM survival, promoting tumor malignancy.
- Research Article
80
- 10.1128/mcb.21.6.2070-2084.2001
- Mar 1, 2001
- Molecular and Cellular Biology
The epidermal growth factor receptor (EGF-R) plays an important role in the growth and progression of estrogen receptor-negative human breast cancers. EGF binds with high affinity to the EGF-R and activates a variety of second messenger pathways that affect cellular proliferation. However, the underlying mechanisms involved in the regulation of EGF-R expression in breast cancer cells are yet to be described. Here we show that the EGF-induced upregulation of EGF-R mRNA in two human breast cancer cell lines that overexpress EGF-R (MDA-MB-468 and BT-20) is accompanied by stabilization (>2-fold) of EGF-R mRNA. Transient transfections using a luciferase reporter identified a novel EGF-regulated approximately 260-nucleotide (nt) cis-acting element in the 3' untranslated region (3'-UTR) of EGF-R mRNA. This cis element contains two distinct AU-rich sequences (~75 nt), EGF-R1A with two AUUUA pentamers and EGF-R2A with two AUUUUUA extended pentamers. Each independently regulated the mRNA stability of the heterologous reporter. Analysis of mutants of the EGF-R2A AU-rich sequence demonstrated a role for the 3' extended pentamer in regulating basal turnover. RNA gel shift analysis identified cytoplasmic proteins (~55 to 80 kDa) from breast cancer cells that bound specifically to the EGF-R1A and EGF-R2A cis-acting elements and whose binding activity was rapidly downregulated by EGF and phorbol esters. RNA gel shift analysis of EGF-R2A mutants identified a role for the 3' extended AU pentamer, but not the 5' extended pentamer, in binding proteins. These EGF-R mRNA-binding proteins were present in multiple human breast and prostate cancer cell lines. In summary, these data demonstrate a central role for mRNA stabilization in the control of EGF-R gene expression in breast cancer cells. EGF-R mRNA contains a novel complex AU-rich 260-nt cis-acting destabilizing element in the 3'-UTR that is bound by specific and EGF-regulated trans-acting factors. Furthermore, the 3' extended AU pentamer of EGF-R2A plays a central role in regulating EGF-R mRNA stability and the binding of specific RNA-binding proteins. These findings suggest that regulated RNA-protein interactions involving this novel cis-acting element will be a major determinant of EGF-R mRNA stability.
- Research Article
- 10.1158/1538-7445.sabcs15-p2-03-07
- Feb 15, 2016
- Cancer Research
BACKGROUND: Diverse molecular mechanisms are being reported in human breast cancer (BC), which can affect the biochemical functions throughout malignant cells development. The microRNAs (miRNAs) are an emerging class of modulators of gene expression with relevant roles in several biological processes, as oncogenic, tumor-suppressive, and metastatic-influencing in BC cells. Recently, a few reports have implied the possible pattern of expression (time oscillation) of miRNAs in time that may be related to molecular changes in mammalian cells. These findings suggest a biological connection among normal and cancer cells, and rhythmic regulation of some miRNAs, but such connection has not yet been studied. In this study, we aimed to identify and compare the rhythmic expression of miRNAs in human breast epithelial normal and cancer cell lines. METHODS: We used cell culture to explore three cell lines, one breast epithelial normal (MCF10A) and two cancer (MCF-7 and MDA-MB-231) cell lines under standard growth conditions in vitro. The cells were synchronized by serum shock (50% horse serum for 2 h), and we collected sample cells (triplicate) for intervals of 4 hours during 48 hours. Collected cells at 12h to 40h (8 time-points) were genome-wide analyzed of miRNA expression using high-throughput Agilent Human miRNA microarray of 2006 human miRNAs. Analysis for identification of rhythmic miRNAs was developed by cosine analysis in R software. RESULTS: We observed diverse oscillation patterns (minimum 6 patterns, i.e. cosine or sine oscillation) of miRNAs in cell lines. Each cell line shows approximately 85 miRNAs with rhythmic oscillation. These also showed distinct phases between cell lines, which could suggest as part of molecular changes in breast normal and cancer cell lines. CONCLUSIONS: Our results suggest that miRNAs may present rhythmic oscillation in the regulation of molecular changes of human breast normal and cancer cells. Citation Format: Chacolla RJ, Trevino VM, Scott SP, Moreno JE. Rhythmic time oscillations of microRNAs in human breast epithelial normal and cancer cell lines. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P2-03-07.
- Research Article
115
- 10.1038/sj.onc.1205514
- Jun 5, 2002
- Oncogene
Two human breast cancer cell lines of differing invasive and metastatic potential, MDA-MB-435 and MCF7, were examined using subtractive suppression hybridization in a search for any genes associated with metastasis. Of the 17 cDNAs identified as being differentially expressed genes, it was determined that syntenin was overexpressed in metastatic MDA-MB-435 cells. Expression analysis showed that the expression level of syntenin was well correlated with invasive and metastatic potential in various human breast and gastric cancer cell lines. Moreover, gastric tumor tissues exhibited a much higher syntenin mRNA expression than their normal counterparts. Syntenin-transfected MCF7 cells migrated more actively, and showed an increased invasion rate relative to vector-transfectants or parental MCF7 in vitro, without evidencing any effect on the adhesion to fibronectin, type I collagen and laminin. Similarly, the forced expression of syntenin to human gastric cancer cell line Az521 increased its migratory and invasive potential in vitro. Syntenin-expressing MCF7 cells were associated with the appearance of numerous cell surface extensions and with pseudopodia formation on collagen I, suggesting that syntenin may be involved in the signaling cascade to actin-reorganization. Mutation study suggested that PDZ2 domain of syntenin could be an essential role in its stimulatory effect on the cell migration. This is the first demonstration that syntenin, a PDZ motif-containing protein, can be overexpressed during the metastatic progression of human breast and gastric cancer cells and that it can function as a metastasis-inducing gene.
- Research Article
1
- 10.1155/1993/619781
- Jan 1, 1993
- Disease Markers
A panel of monoclonal antibodies (n = 72 including controls) directed against lung cancer antigens was screened immunohistochemically against a panel of seven human lung cancer cell lines (including small cell carcinoma, squamous cell carcinoma, adenocarcinoma and mesothelioma), six human breast cancer cell lines and one human colon cancer cell line. The majority of the antibodies (n = 42) reacted also with antigens present on breast and colon cancer cell lines. This cross reactivity especially between lung and breast cancer cell lines is not altogether unexpected since antigens common to breast and lung tissue including their neoplasms such as MUC1 antigen have been described. Our results indicate that epitopes shared by lung and breast cancers are probably more common than previously thought. The relevance for prognosis and therapy of these shared antigens, especially as disease markers in breast cancer, has to be investigated.
- Research Article
51
- 10.1023/a:1006135331912
- Mar 1, 1999
- Breast Cancer Research and Treatment
Our laboratory has developed twelve human breast cancer cell lines from primary and metastatic sites. In this report we demonstrate that eight of eight breast cancer cell lines examined exhibit constitutively tyrosine phosphorylated and enzymatically active endogenous pp125fak when grown in monolayer. The activation status of pp125fak in breast cancer cells in monolayer is significantly elevated over that exhibited by normal mammary epithelial cells cultured under the same conditions. Constitutive activation of pp125fak is the only characteristic so far studied that all of these breast cancer cell lines have in common. In contrast to HBC cells, tyrosine phosphorylation of pp125fak in HME cells was low or absent in monolayer culture but was induced to high levels by culturing the cells in Matrigel. Thus tyrosine phosphorylation and activation of pp125fak is a regulated process in normal mammary epithelial cells, but is constitutive in breast cancer cells. Finally, analysis of the ability of normal human mammary epithelial cells and breast cancer cell lines to grow under anchorage-independent conditions indicated that normal human mammary epithelial cells rapidly and uniformly lost viability when not substrate-attached, whereas all of the breast cancer cell lines survived for a 3-week culture period. Furthermore, a subset of the breast cancer cell lines grew to form large colonies under anchorage-independent conditions. Interestingly, pp125fak activation decreased dramatically in HBC cells cultured for two weeks in suspension, suggesting that activation of this kinase is not necessary for long-term growth under anchorage-independent conditions. These results suggest that constitutive activation of pp125fak results in preferential survival of human breast cancer cells under anchorage-independent conditions but that activation of pp125fak is not the sole mediator of anchorage-independent colony formation.
- Research Article
- 10.1158/1538-7445.am2013-1507
- Apr 15, 2013
- Cancer Research
HLA-G is a nonclassical MHC Class I molecule that protects the fetus from the maternal immune system, and it also plays a role in immune evasion of cancer. Previously we have shown that iron can suppress the cytolytic function of natural killer cells to human breast cancer cell lines. We have found that breast cancer cells over express HLA-G mRNA and proteins in both cell lines and breast carcinomas. In order to determine if iron concentration affects HLA-G expression, we cultured the human breast cancer cell line MCF-7 in media with 100 mM, 400 mM, and 1600 mM of iron for 24 hours. We used RT-PCR to examine HLA-G mRNA expression of the MCF-7 cells. The PCR primers were designed so that they could detect all alternatively spliced HLA-G mRNA isoforms, 1000 bp (HLA-G1 and HLA-G5), 600 bp (HLA-G2 and HLA-G4), and 300 bp (HLA-G3). RT-PCR showed that the MCF-7 cells cultured with control medium only had one band of 300 bp mRNA expression. However, the MCF-7 cells cultured in medium containing 100 mM, 400 mM, and 1600 mM of iron had three bands of 300 bp, 600 bp, and 1000 bp HLA-G mRNA expression. Moreover, the intensity of the 300 bp band from the iron-treated MCF-7 cells is significantly higher than the cells cultured with control medium. In summary, iron upregulates the HLA-G mRNA expression of MCF-7 cells, and this probably contributes to cancer immunosuppression and to the cytolytic dysfunction of natural killer cells. Citation Format: Jonathan F. Head, Xian P. Jiang, Robert L. Elliott. Iron upregulates HLA-G mRNA expression of the human MCF-7 breast cancer cell line. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 1507. doi:10.1158/1538-7445.AM2013-1507
- Research Article
58
- 10.1194/jlr.m500473-jlr200
- Apr 1, 2006
- Journal of Lipid Research
Bone is the most common site to which breast cancer cells metastasize. We found that osteoblast-like MG63 cells and human bone tissue contain the bile acid salt sodium deoxycholate (DC). MG63 cells take up and accumulate DC from the medium, suggesting that the bone-derived DC originates from serum. DC released from MG63 cells or bone tissue promotes cell survival and induces the migration of metastatic human breast cancer MDA-MB-231 cells. The bile acid receptor farnesoid X receptor (FXR) antagonist Z-guggulsterone prevents the migration of these cells and induces apoptosis. DC increases the gene expression of FXR and induces its translocation to the nucleus of MDA-MB-231 cells. Nuclear translocation of FXR is concurrent with the increase of urokinase-type plasminogen activator (uPA) and the formation of F-actin, two factors critical for the migration of breast cancer cells. Our results suggest a novel mechanism by which DC-induced increase of uPA and binding to the uPA receptor of the same breast cancer cell self-propel its migration and metastasis to the bone.
- Research Article
28
- 10.1002/jcb.26577
- May 8, 2018
- Journal of Cellular Biochemistry
In several cancers, microRNA (miRNAs) play vital roles in tumor initiation, drug resistance, and metastasis. The aim of this study was to examine the expression levels of miR-4301 in human breast cancer and investigate whether its potential roles involved targeting Dopamine receptor D2 (DRD2). Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) was also used to examine the expression levels of miR-4301 in human breast cancer cell lines MDA-MB-231, MCF-7, and SKBR3. In these cell lines, MTT assay, immunofluorescence staining, caspase assay, proliferation assay, and flow cytometry were conducted to explore the potential functions of miR-4301. The effects of modulating miR-4301 on transcription levels of DRD2 were subsequently confirmed via qRT-PCR. miR-4301 expression levels were significantly decreased in human breast cancer specimens and cell lines (P < 0.05). Transfection of miR-4301 in breast cancer cells suppressed cell proliferation and induced apoptosis. Expression analysis indicated that miR-4301 was inversely correlated with DRD2 expression in breast cancer specimens. qRT-PCR showed that miR-4301 negatively regulated DRD2 expression. Downregulation of DRD2 expression in MDA-MB-231, MCF-7, and SKBR3 cells suppressed cell proliferation and promoted apoptosis.
- Abstract
71
- 10.1016/s0021-9258(19)37144-3
- Sep 1, 1992
- Journal of Biological Chemistry
Estrogen receptor (ER) binding has been shown to decrease in breast cancer cell lines exposed to sodium butyrate; however, the underlying mechanisms are unknown. In MCF-7 breast cancer cells, butyrate caused a rapid time- and concentration-dependent decrease in ER mRNA levels, apparent by 3 h at 3 mM butyrate. ER gene transcription rate was decreased and cycloheximide co-treatment did not relieve this inhibitory effect, suggesting that the butyrate effect was not dependent on ongoing protein synthesis. In both MCF-7 and T-47D cells the decrease in ER mRNA was mirrored by an increase in the level of epidermal growth factor receptor (EGF-R) mRNA. A marked inverse relationship exists between ER and EGF-R in human breast cancer biopsies and cell lines, and the reciprocal modulation of these genes by butyrate suggests that the expression of ER and EGF-R may be co-regulated. This relationship was further investigated in lines expressing only one or the other receptor. In the ER-positive EGF-R-negative line, MDA-MB-134-VI, butyrate exposure decreased ER mRNA levels, implying that the regulation of ER mRNA by butyrate is independent of EGF-R expression. However, butyrate decreased EGF-R mRNA in two ER-negative lines, MDA-MB-231 and HBL-100. As this effect differed from that in ER-positive lines, the regulation of EGF-R may depend on the expression of ER. The possibility that ER and EGF-R gene expression are closely linked has implications in the understanding of progression of human breast cancers to a hormone-independent phenotype and for the use of ER and EGF-R levels as independent prognostic indicators.
- Research Article
- 10.46309/biodicon.2025.1732263
- Oct 2, 2025
- Biological Diversity and Conservation
- Research Article
- 10.46309/biodicon.2025.1786862
- Oct 2, 2025
- Biological Diversity and Conservation
- Research Article
- 10.46309/biodicon.2025.1596745
- Sep 25, 2025
- Biological Diversity and Conservation
- Research Article
- 10.46309/biodicon.2025.1621315
- Sep 25, 2025
- Biological Diversity and Conservation
- Research Article
- 10.46309/biodicon.2025.1610652
- Sep 25, 2025
- Biological Diversity and Conservation
- Research Article
- 10.46309/biodicon.2025.1588677
- Aug 15, 2025
- Biological Diversity and Conservation
- Research Article
- 10.46309/biodicon.2025.1553694
- May 21, 2025
- Biological Diversity and Conservation
- Research Article
- 10.46309/biodicon.2025.1541568
- May 21, 2025
- Biological Diversity and Conservation
- Research Article
- 10.46309/biodicon.2025.1585290
- May 21, 2025
- Biological Diversity and Conservation
- Research Article
- 10.46309/biodicon.2025.1537342
- May 21, 2025
- Biological Diversity and Conservation
- Ask R Discovery
- Chat PDF
AI summaries and top papers from 250M+ research sources.