Open Access

A regulation loop between Nrf1α and MRTF-A controls migration and invasion in MDA-MB-231 breast cancer cells

  • Authors:
    • Yao Xu
    • Ying Luo
    • Chen Liang
    • Weibing Xing
    • Tongcun Zhang
  • View Affiliations

  • Published online on: August 9, 2018     https://doi.org/10.3892/ijmm.2018.3816
  • Pages: 2459-2468
  • Copyright: © Xu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

As a strong transactivator of promoters containing CarG boxes, myocardin‑related transcription factor A (MRTF‑A) is critical for the process of metastasis in tumor cells. Nuclear factor erythroid 2‑like 1 (Nrf1) is well known as an important regulator of oxidative stress, which exists in multiple splicing forms with many unknown functions. The present study demonstrated a novel regulation loop between Nrf1α (the longest splicing form of Nrf1) and MRTF‑A that regulated the migration and invasion of breast cancer MDA‑MB‑231 cells. The underlying mechanism of this regulation look was further investigated. In particular, Nrf1α inhibited migration and invasion of breast cancer cells through inhibiting the expression of MRTF‑A via miR‑219. The current results revealed that miR‑219 could bind to the MRTF‑A 3'‑UTR to directly regulate its expression. However, MRTF‑A could reverse activate the Nrf1α expression through binding to the CarG box in the Nrf1α promoter. It can be speculated that this regulation loop may be a homeostasis mechanism in cells against tumorigenesis.

Introduction

Despite the increasing level of modern medical care, cancer remains difficult to be targeted effectively and cancer-related mortality remains high worldwide. Metastasis, one of the features of advanced cancer, is the predominant reason for the high mortality. Therefore, potential inhibition of metastasis and invasion of tumor cells has become the research direction and goal for many researchers. In recent years, more and more transcription factors, long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) associated with tumor metastasis have been revealed. Among them, myocardin-related transcription factor A (MRTF-A) is a very important transcription factor which can promote tumor metastasis.

MRTF-A consists of 807 amino acid residues, and it belongs to the family of serum amyloid P-component serum amyloid P (SAP) proteins. MRTF-A can promote serum response factor (SRF) protein binding to the conserved cis regulatory element CC(A/T)6GG (known as CarG box), thus regulating the transcription of target genes. It has an important role in the growth and development of the organism (1-3). The activation of transforming growth factor (TGF) β-related signaling pathways can be very effective to induce MRTF-A translocation into the nucleus. Once there, MRTF-A promotes the transcription of epithelial-mesenchymal transition (EMT)-related molecules, such as α-smooth muscle actin (α-SMA), and enhances the migration and metastasis of tumor cells (4-10).

Other studies have reported that the activation of MRTF-A-mediated Rho associated coiled-coil containing protein kinase (ROCK) signaling pathway can regulate the phosphorylation of E-cadherin and decrease the adhesion ability of renal tubular epithelial cells, resulting in the occurrence of EMT (11). In the present study, while examining the role of MRTF-A in inducing the migration of breast cancer cells, it was observed that the transcription factor nuclear factor erythroid 2-like 1 (Nrf1) could regulate the above process. Nrf1 (also known as NFE2L1, LCRF1 or TCF11) belongs to the nuclear factor erythroid 2-related factor (NRF) family. It is ubiquitously expressed and essential for maintaining cellular homeostasis, organ integrity and oxidative stress during development and growth (12-17).

There are multiple splicing isoforms for Nrf1 in cells, such as the full-length Nrf1α, as well as the LCR-F1/Nrf1β, Nrf1γ and Nrf1δ isoforms. To date, the specific biological function of each isoform remains unclear. The present study aimed to explore the possible mechanisms of Nrf1α and MRTF-A in regulating migration and invasion of MDA-MB-231 breast cancer cells.

Materials and methods

Cell culture

MDA-MB-231 cells used in the present study were purchased from American Type Culture Collection (cat. no. HTB-26; Manassas, VA, USA). The cells were seeded in Dulbecco's modified Eagle's medium-high glucose (DMEM-HG; Hyclone; GE Healthcare Life Sciences, Logan, UT, USA) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C in humidified air with 5% CO2. Cos-7 cells (American Type Culture Collection; cat. no. CRL-1651) and 293T cells (American Type Culture Collection; cat. no. CRL-3216) were cultured in DMEM containing 10% FBS, penicillin (100 U/ml) and streptomycin (100 μg/ml).

Plasmid construction and standard lentivirus production

Nrf1α (Gene ID, 4779) and MRTF-A (Gene ID, 57591) were inserted into the lentivirus vector pCDH-CMV. The plasmids pCDH-Nrf1α, pCDH-MRTF-A or pCDH-CMV (empty vector control) were cotransfected with the psPAX2 and pMG2.G into 293T cells (at ~70-80% confluency) using Lipofectamine 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. The medium was changed after 8 h, and 48 h later the supernatant was collected and filtered. The target short hairpin (sh) RNAs against Nrf1α gene and MRTF-A gene were inserted into the pLKO.1 vector. As a negative control, a non-targeting sequence that had no significant homology to any mouse or human gene was inserted into pLKO.1. pCDH-Nrf1α, shNrf1α and their corresponding controls were a gift from Professor Jian Dong (North Carolina State University, Raleigh, NC, USA). The shMRTF-A plasmid has been previously described (18). shRNA lentiviral particles were produced by co-transfecting 293T cells using Lipofectamine 3000 with the lentivirus expression plasmids and packaging plasmids. Silencing efficiency was detected using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analyses. miR-219 complementary sequence (AGA ATT GCG TTT GGA CAA TCA) was inserted into pcDNA3.1(−) vector to silence the function of miR-219 and pcDNA3.1(−) vector was used as a control.

Lentivirus transduction of MDA-MB-231 cells

MDA-MB-231 cells were cultured in high DMEM supplemented with 10% FBS and lentivirus was added at a multiplicity of infection (MOI) of 5. Following overnight incubation, the media containing the lentivirus was removed and fresh media was added.

RT-qPCR

Total RNA, including miRNA, was extracted by using the miRNA kit (Omega Bio-Tek, Inc., Norcross, GA, USA), according to the manufacturer's protocol. The samples were reverse-transcribed using M-MLV Reverse Transcriptase (Promega Corporation, Madison, WI, USA). qPCR was performed in an StepOne Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). Fast SYBR Green Master Mix was obtained from Applied Biosystems (Thermo Fisher Scientific, Inc.). The relative expression levels of MRTF-A and Nrf1α were normalized to GAPDH. The primers for the qPCR analysis are listed in Table I. Amplification of U6 small nuclear RNA served as an endogenous control to normalize miR-219 expression data. The primers for the miR-219 analysis are listed in Table II. Thermocycling conditions were as follows: 95°C for 5 min followed by 40 cycles of 95°C for 10 sec and 60°C for 30 sec, then a melting curve analysis from 60 to 95°C every 0.2°C for 1.5 min was obtained. Each sample was analyzed in triplicate and quantified using the 2−∆∆Cq method (19).

Table I

Sequences of primers used in reverse transcription-quantitative polymerase chain reaction analysis.

Table I

Sequences of primers used in reverse transcription-quantitative polymerase chain reaction analysis.

GenePrimerSequence (5′-3′)
MRTF-AForward AAGGAACCACCTGGCTATGA
Reverse C TCCGCTCTGAATGAGAATGT
Nrf1Forward GCTGGACACCATCCTGAATC
Reverse GTAGGGTCGTCCGTTCTCAT
GAPDHForward TCAAGAAGGTGGTGAAGCAG
Reverse AGGTGGAGGAGTGGGTGTCG

[i] MRTF-A, myocardin-related transcription factor A; Nrf1, nuclear factor erythroid 2-like 1.

Table II

Sequences of primers used in reverse transcription-quantitative polymerase chain reaction analysis.

Table II

Sequences of primers used in reverse transcription-quantitative polymerase chain reaction analysis.

GenePrimerSequence (5′-3′)
miR-219Reverse transcription CTCAACTGGTGTCGTGGAGTCGGCAATTCAGTTGAGAGAATTGC
Forward ACACTCCAGCTGGGTGATTGTCCAAACGC
Reverse TGGTGTCGTGGAGTCG
U6Reverse transcription AACGCTTCACGAATTTGCGT
Forward CTCGCTTCGGCAGCACA
Reverse AACGCTTCACGAATTTGCGT
Protein extraction and western blotting

For western blot analysis, protein samples were extracted from the cells with Protein Extraction Reagent (Pierce; Thermo Fisher Scientific, Inc.). The concentrations of proteins were determined using a bicinchoninic acid quantification kit (Beyotime Institute of Biotechnology, Haimen, China). The proteins (20 μg) were separated by SDS PAGE (10% gel) and transferred onto a polyvinylidene difluoride membrane. The membrane was blocked using 5% non fat milk at 25°C for 1 h, and then incubated with primary antibodies overnight at 4°C. The antibodies used were as follows: Anti-human GAPDH antibody (cat. no. 97166; 1:2,000, Cell Signaling Technology, Inc., Danvers, MA, USA), anti-human Nrf1 antibody (cat. no. 46743; 1:1,000, Cell Signaling Technology, Inc.), anti-human MRTF-A antibody (cat. no. ab49311; 1:1,000, Abcam, Cambridge, UK). Then, the membrane was incubated with IRDyeTM-800 conjugated anti-mouse or anti-rabbit secondary antibodies (cat. no. 115-005-146 and 115-005-144; 1:5,000, Jackson ImmunoResearch Laboratories, Inc., West Grove, PA, USA) at 25°C for 1 h at room temperature. The protein signals were visualized with the Odyssey Infrared Imaging System (LI-COR Biosciences, Lincoln, NE, USA). GAPDH expression was used as an internal control. The western blotting results were quantified using ImageJ software (version 2.0; National Institutes of Health, Bethesda, MD, USA).

Colony formation assay

Cells were transduced with Nrf1α-expressing lentivirus or with shNrf1α lentivirus or their corresponding controls, as indicated. Twenty-four hours later, transfected cells were trypsinized, counted and replated at a density of 200 cells per 6-cm dish. Ten days later, colonies resulting from the surviving cells were fixed with 3.7% methanol, and stained with 0.1% crystal violet. Following capturing photos, the crystal violet stain was washed with 33% acetic acid and the absorbance was measured at 570 nm. Each assay was performed in triplicate.

Wound healing assay

Cells were subcultured in 6-well plates at a density of 1×105 cells/well. Upon >80% confluence, the cell monolayer was gently scraped with a yellow pipette tip to generate a linear wound and washed twice with serum-free medium to remove cell debris. Images were subsequently captured at 0 and 24 h. The closure of the wounds was quantified by the distance of cells moved into the wounded area. The experiment was repeated twice with triplicate measurements in each experiment. The results were quantified using ImageJ software (20).

Transwell invasion assay

The invasion assay was performed using transwell chambers (Corning Incorporated, Corning, NY, USA), that had Matrigel (50 μl; BD Biosciences, San Jose, CA, USA) pre-coated polycarbonate membranes (8.0 μm pore size). A total of 1×104 cells were suspended in 200 μl FBS-free DMEM and added to the upper chamber. The lower chamber was filled with 500 μl DMEM containing 10% FBS. Following incubation for 24 h, cells on the lower surface of the membrane were fixed in 4% paraformaldehyde and stained with 0.1% crystal violet. Cells in four random microscopic fields (magnification, ×20) were counted in triplicates. After capturing photos, the crystal violet was washed with 33% acetic acid and the absorbance was measured at 570 nm.

Luciferase constructs, site-mutation, and luciferase assay

The human miR-219 promoter was fused to the coding sequence of the pGL-3 luciferase reporter vector (Promega Corporation). The mutant (mut)-miR-219 promoter was identical to the pGL-3-miR-219 promoter, except that the Nrf1α binding antioxidant response element (ARE) site was cut down (the sequence was AGTGGAAGC). The human Nrf1 promoter luciferase reporter plasmids were also constructed in the same way. The 3′-untranslated region (UTR) of the human MRTF-A was amplified from human genomic DNA and individually inserted into the pmiGLO vector (Promega Corporation). The primers for constructing the luciferase reporter plasmids are listed in Table III. Cells (2×105/well) were plated in 24-well plates. Cos-7 cells were cotransfected with Nrf1α expression plasmids (pcDNA3.1(-)-myocardin) or control vector (pcDNA3.1-) in combination with miR-219-luc or mut-miR-219-luc. The transfection of these plasmids was performed using Lipofectamine 3000 according to the manufacturer's protocols. Cells were harvested 24 h following transfection and luciferase activity was measured using the Dual luciferase Assay System (Promega Corporation). Results were expressed as a fold induction relative to the cells transfected with the control vector (pcDNA3.1-) after normalization to Renilla activity. In the results from the dual luciferase assays, the columns represent the mean value of three independent experiments and the error bars represent the standard deviation.

Table III

Primers used to generate the luciferase reporter plasmids by polymerase chain reaction.

Table III

Primers used to generate the luciferase reporter plasmids by polymerase chain reaction.

Plasmid namePrimerSequence (5′-3′)
miR219 promoter-WTF GGGACTCGAGTTGCCCAGTCCATCTTGGTTGTGTT
R AAGTCTCGAGTTTGAATAACGCCACGGGGCCATCA
miR219 promoter cut downF TAGGCTCGAGGCTCCAGAGGCCTTTGGTTTCCATG
R AAGTCTCGAGTTTGAATAACGCCACGGGGCCATCA
Nrf1 promoter-WTF GCGCGCTAGCAATTCCATGAGTGGTTTGCTG
R ATTAGCTAGCGCTGCCTCCACAGCAGGCC
Nrf1 promoter-cut CarG 1F GTGCTAGCCCGGGCTCGAGCGCAAGCACAAAATGGACTCG
R ACTTAGATCGCAGATCTCGAGCACAGCAGGCCCTAAGCCC
Nrf1 promoter-cut CarG 1,2F GTGCTAGCCCGGGCTCGAGGGGGTCCTTTGGGCTGTTTC
R ACTTAGATCGCAGATCTCGAGGAGCTCGGAGCCTCCGCTTA
MRTF-A 3′UTR-WTF ATTCGCTAGCAAGACGGGGTGGGGAAGGG
R GGGGTCTAGACAGCTGCTCTCCTCTGCCCTG
MRTF-A 3′UTR-MUTFTCCACATGGT TGTGAGTCTTTGGGGGGGCA GCCCCTGCTT TTTCCC
R GGGAAAAAGCAGGGGCTGCCCCCCCAAAGACTCACAACCATGTGGA

[i] F, forward; R, reverse; Nrf1, nuclear factor erythroid 2-like 1; MRTF-A, myocardin-related transcription factor A; UTR, untranslated region; WT, wild-type; MUT, mutant.

Chromatin immunoprecipitation (ChIP) assay

A ChIP Assay kit (Merck KGaA, Darmstadt, Germany) was used, following the manufacturer's instructions. Following treatments as indicated, each experimental group was incubated with 1% formaldehyde to cross-link DNA-protein complexes. After washing with ice-cold PBS for three times, cells were lysed in SDS lysis buffer. Then, the lysate was sonicated to shear DNA to 200-1,000 bp fragments. Anti-human Nrf1 antibody (cat. no. 46743; Cell Signaling Technology, Inc.) or anti-human MRTF-A antibody (cat. no. ab49311; Abcam) were used to immunoprecipitate the cross-linked proteins at 4°C overnight. Immunoglobulin G (cat. no. ab172730; Abcam) acted as the negative control. The DNA was used as a template for PCR, where the myocardin binding sites were utilized. The PCR products were separated on 1% agarose gel. The PCR primer sequences are listed in Table IV.

Table IV

Sequences of primers used in chromatin immunoprecipitation assay.

Table IV

Sequences of primers used in chromatin immunoprecipitation assay.

Target regionPrimerSequence (5′-3′)
miR-219 promoter AREF TTCAGCATGGTCTTCTCAG
R AACCAAAGGCCTCTGGAG
Nrf1 promoter CarG box1F GTACTTAATCTGCAAACC
R TGAGTCATTAGTCCCTGT
Nrf1 promoter CarG box2F AGATGGGACTGGAGAAAT
R GTAGAAACAGCCCAAAGG

[i] F, forward; R, reverse; ARE, antioxidant response element; Nrf1, nuclear factor erythroid 2-like 1.

Statistical analysis

Quantitative data are expressed as mean ± standard error of the mean. Statistical analysis of differences between two groups was performed by Student's t-test. A one-way analysis of variance followed by Tukey's test was used for comparing differences among multiple groups. Statistical analysis was performed with GraphPad Prism 5 (GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Nrf1α inhibits migration and invasion in MDA-MB-231 breast cancer cells

To investigate the effect of Nrf1α in breast cancer cells, the MDA-MB-231 breast cancer cell line was transduced to overexpress Nrf1α (Nrf1α group), or to knockdown the expression of Nrf1α (shNrf1α group). Western blot analysis was used to evaluate the expression of Nrf1α in each group (Fig. 1A and B). Then, by using colony formation, wound healing and transwell invasion assays, the growth, migration and invasion capacities were measured in each cell group, respectively. Notably, when Nrf1α was overexpressed, the proliferation, migration and invasion ability of tumor cells was significantly decreased compared with the control group (Fig. 1C, E and G). By contrast, when endogenous Nrf1α was silenced by shRNA, the tumor cells exhibited increased proliferation, migration and invasion compared with the control group (Fig. 1D, F and H). These findings suggested that Nrf1α was negatively associated with the migration and invasion of MDA-MB-231 cells. Nrf1α expression could inhibit migration and invasion of MDA-MB-231 breast cancer cells.

Nrf1α inhibits migration and invasion by suppressing endogenous MRTF-A expression

Previous studies had demonstrated that MRTF-A promotes the migration and invasion of tumor cells, including breast cancer (21-23). As an important transcription factor associated with tumor metastasis, MRTF-A was a focus of study in our laboratory. While exploring the potential molecular mechanisms by which Nrf1α regulates the function of tumor cells, a regulatory relationship between Nrf1α and MRTF-A was discovered. As presented in Fig. 2A-D, the expression of endogenous MRTF-A was negatively associated with the expression of Nrf1α, at both the mRNA and protein level. Subsequently, the endogenous MRTF-A expression was silenced by shRNA (Fig. 2E and F), and then Nrf1α was overexpressed or knocked down on this setting. The results demonstrated that, upon MRTF-A silencing, Nrf1α lost the ability of regulating MDA-MB-231 cell migration and invasion (Fig. 2G and H).

Nrf1α inhibits the expression of MRTF-A via miR-219

The aforementioned results demonstrated that Nrf1α inhibited the expression of MRTF-A to regulate migration and invasion in breast cancer cells. However, the mechanism by which Nrf1α regulated the expression of MRTF-A was unclear. It was hypothesized that miRNAs may have participated in this process. Through miRBase and TargetScan prediction programs analysis (24,25), miR-219 was selected. Analysis of miR-219 expression levels demonstrated that miR-219 was upregulated when Nrf1α overexpressed (Fig. 3A), while when Nrf1α was silenced, miR-219 levels were decreased (Fig. 3B). Next, the miR-219 complementary sequence was used to silence the function of miR-219 in the MDA-MB-231 cells, and then Nrf1α was overexpressed or knocked down. RT-qPCR and western blot assays were used to detect the expression of endogenous MRTF-A. The results demonstrated that Nrf1α lost its function to regulate MRTF-A expression, following miR-219 silencing (Fig. 3C and D). To further investigate the molecular mechanism by which Nrf1α regulates miR-219, luciferase assays were used to directly examine the effect of Nrf1α on the transcriptional activity of the miR-219 promoter, which contains a predicted ARE site. As illustrated in Fig. 3E, Nrf1α could bind to the ARE site to activate the transcriptional activity of the miR-219 promoter. In addition, ChIP assay confirmed the direct binding of Nrf1 to the miR-219 promoter (Fig. 3F).

It is well known that the most common method by which miRNAs regulate gene expression is to act on their 3′-UTR and degrade their mRNA. As illustrated in Fig. 3G, binding sites of miR-219 were predicted to exist in the 3′-UTR of MRTF-A. Therefore, the effect of miR-219 on the MRTF-A 3′-UTR was examined by luciferase assay. The results confirmed that miR-219 could bind to the 3′-UTR region of MRTF-A directly, reducing the mRNA levels of MRTF-A (Fig. 3H).

MRTF-A affects Nrf1α expression through binding to the CarG box in the Nrf1α promoter

Previous studies have demonstrated that Nrf1α could inhibit the migration and invasion of breast cancer cells via MRTF-A. The present study revealed the potential molecular interplay between these two factors in this process. Notably, MRTF-A was demonstrated to negatively regulate the expression of Nrf1α. To further explore the relationship between Nrf1α and MRTF-A, a MRTF-A-overexpressing MDA-MB-231 line was established (Fig. 4A and B). As presented in Fig. 4C and E, the results of RT-qPCR analysis indicated that the mRNA levels of Nrf1α presented a positive correlation with the mRNA levels of MRTF-A, whether MRTF-A was overexpressed or knocked down. The western blot assay results also demonstrated that the protein expression of these two factors was similar to the mRNA expression (Fig. 4D and F). As a strong drive factor of containing the CarG locus genes, MRTF-A may activate Nrf1α through this pathway. Two potential CarG boxes were observed on the Nrf1α promoter region. The results from the luciferase assay indicated that the transcriptional activity of Nrf1α promoter could be upregulated by MRTF-A (Fig. 4G). However, the Nrf1α transcriptional activity was not affected when the far CarG box (CarG 1) was removed, or when both CarG boxes were removed (Fig. 4G). These results might indicate that MRTF-A affected the Nrf1α transcriptional activity through binding to the far CarG box (CarG 1). To further explore the mechanism of this regulation, ChIP was used. The results demonstrated that MRTF-A was bound to the far CarG box (CarG 1), but not the near CarG box (CarG 2), which was consistent with the results from the luciferase assay (Fig. 4H).

Discussion

The present study demonstrated that Nrf1α regulated migration and invasion of breast cancer cells by inhibiting the expression of MRTF-A. Notably, MRTF-A can reverse activate the expression of Nrf1α by forming a complex with SRF binding to the CarG box in the promoter of Nrf1α. Thus, a regulation loop exists between the two factors in the breast cancer cell line MDA-MB-231 (Fig. 5).

The transcription factor Nrf1 has an important role in upregulating the antioxidant response by increasing glutathione biosynthesis (13,14,16,26). Nrf1 is also known to regulate a variety of antioxidant genes through the combination with ARE (27). In the endoplasmic reticulum (ER), Nrf1 is cleaved into many forms, such as Nrf1α, LCR-F1/Nrf1β, Nrf1γ and Nrf1δ, and translocated from the ER to the nucleus in response to ER stress (28,29). To date, the specific biological function of each subtype remains unclear. Therefore, it is helpful to explore the functional differences of each subtype. In the present study, it was demonstrated that the migration and invasion of breast cancer cells were inhibited following overexpression of Nrf1α. By contrast, the ability of breast cancer cells to migrate and invade was improved when the expression of Nrf1α was silenced. Similar findings have been previously reported in HepG2 cells following Nrf1α knock down (30). This may suggest that the effect of Nrf1α in inhibiting migration and invasion may be common in multiple types of cancer. Subsequently, the present study attempted to reveal the molecular mechanism by which Nrf1α inhibited migration and invasion in breast cancer cells.

MRTF-A is an important transcription factor associated with tumor migration and invasion. The present study explored the hypothesis that a regulatory relationship may exist between the two factors. Nrf1α could indeed inhibit the expression of MRTF-A. Nrf1α, is known to positively regulate genes through ARE sites. Therefore, it was speculated that miRNAs may exist that have a role in the Nrf1α/MRTF-A regulation loop. Bioinformatics analysis was used to discover potential miRNAs that may regulate MRTF-A expression. In addition, the promoter regions of these miRNAs were examined for the presence of ARE sites. Following these criteria, miR-219 was identified as a potential target. Results from luciferase and ChIP assays demonstrated that Nrf1α indeed regulated MRTF-A expression via miR-219, which could directly bind to the 3′-UTR of MRTF-A.

In addition, MRTF-A was demonstrated to directly upregulate the expression of Nrf1α by forming a complex with SRF binding to the CarG box. Previous studies have also demonstrated that MRTF-A is associated with cancer-related processes by the SRF/MRTF-A signaling for the induction of target genes (31-35).

In conclusion, the present study demonstrated that a regulation loop exists between Nrf1α and MRTF-A, and that this loop controls the process of breast cancer cell migration and invasion. Furthermore, the potential underlying mechanism was explored. The present findings may provide a theoretical reference for the clinical inhibition of tumor metastasis. Further confirmation of these results with mouse models or patient tissues will be required in future studies.

Funding

This study was financially supported by the National Natural Science Foundation of China (grant nos. 31471282 and 31570764).

Availability of data and materials

The analyzed datasets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

YX, WX and TZ designed the experiments. YX, YL and CL performed the experiments, analyzed and interpreted the data. YX and YL were major contributors in writing the manuscript. The final version of the manuscript has been read and approved by all authors, and each author believes that the manuscript represents honest work.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Liao XH, Wang N, Liu QX, Qin T, Cao B, Cao DS and Zhang TC: Myocardin-related transcription factor-A induces cardiomyocyte hypertrophy. IUBMB Life. 63:54–61. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Parmacek MS: Myocardin-related transcription factor-a: Mending a broken heart. Circ Res. 107:168–170. 2007. View Article : Google Scholar

3 

Small EM, Thatcher JE, Sutherland LB, Kinoshita H, Gerard RD, Richardson JA, Dimaio JM, Sadek H, Kuwahara K and Olson EN: Myocardin-related transcription factor-a controls myofibroblast activation and fibrosis in response to myocardial infarction. Circ Res. 107:294–304. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Fan L, Sebe A, Péterfi Z, Masszi A, Thirone AC, Rotstein OD, Nakano H, McCulloch CA, Szászi K, Mucsi I and Kapus A: Cell contact-dependent regulation of epithelial-myofibroblast transition via the Rho-Rho kinase-phosphomyosin pathway. Mol Biol Cell. 18:1083–1097. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Miano JM, Long X and Fujiwara K: Serum response factor: Master regulator of the actin cytoskeleton and contractile apparatus. Am J Physiol Cell Physiol. 292:C70–C81. 2007. View Article : Google Scholar

6 

Sebe A, Masszi A, Zulys M, Yeung T, Speight P, Rotstein O, Nakano H, Mucsi I, Szászi K and Kapus A: Rac, PAK and p38 regulate cell contact-dependent nuclear translocation of myocardin-related transcription factor. FEBS Lett. 582:291–298. 2008. View Article : Google Scholar

7 

Zhao XH, Laschinger C, Arora P, Szászi K, Kapus A and McCulloch CA: Force activates smooth muscle α-actin promoter activity through the Rho signaling pathway. J Cell Sci. 120:1801–1809. 2007. View Article : Google Scholar : PubMed/NCBI

8 

O'Connor JW, Riley PN, Nalluri SM, Ashar PK and Gomez EW: Matrix rigidity mediates TGFβ1-induced epithelial-myofibroblast transition by controlling cytoskeletal organization and MRTF-A localization. J Cell Physiol. 230:1829–1839. 2015. View Article : Google Scholar

9 

Fan WH and Karnovsky MJ: Increased MMP-2 expression in connective tissue growth factor over-expression vascular smooth muscle cells. J Biol Chem. 277:9800–9805. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Hinson JS, Medlin MD, Lockman K, Taylor JM and Mack CP: Smooth muscle cell-specific transcription is regulated by nuclear localization of the myocardin-related transcription factors. Am J Physiol Heart Circ Physiol. 292:H1170–H1180. 2007. View Article : Google Scholar

11 

Tian YC, Fraser D, Attisano L and Phillips AO: TGF-beta1-mediated alterations of renal proximal tubular epithelial cell phenotype. Am J Physiol Renal Physiol. 285:F130–F142. 2007. View Article : Google Scholar

12 

Farmer SC, Sun CW, Winnier GE, Hogan BL and Townes TM: The bZIP transcription factor LCR-F1 is essential for mesoderm formation in mouse development. Genes Dev. 11:786–798. 1997. View Article : Google Scholar : PubMed/NCBI

13 

Chan JY, Kwong M, Lu R, Chang J, Wang B, Yen TB and Kan YW: Targeted disruption of the ubiquitous CNC-bZIP transcription factor, Nrf-1, results in anemia and embryonic lethality in mice. EMBO J. 17:1779–1787. 1998. View Article : Google Scholar : PubMed/NCBI

14 

Kwong M, Kan YW and Chan JY: The CNC basic leucine zipper factor, nrf1, is essential for cell survival in response to oxidative stress-inducing agents role for nrf1 in gamma-gcs(l) and gss expression in mouse fibroblasts. J Biol Chem. 274:37491–37498. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Xu Z, Chen L, Leung L, Yen TB, Lee C and Chan JY: Liver-specific inactivation of the Nrf1 gene in adult mouse leads to nonalcoholic steatohepatitis and hepatic neoplasia. Proc Natl Acad Sci USA. 102:4120–4125. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Ohtsuji M, Katsuoka F, Kobayashi A, Aburatani H, Hayes JD and Yamamoto M: Nrf1 and Nrf2 play distinct roles in activation of antioxidant response element-dependent genes. J Biol Chem. 283:33554–33562. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Tsujita T, Peirce V, Baird L, Matsuyama Y, Takaku M, Walsh SV, Griffin JL, Uruno A, Yamamoto M and Hayes JD: Transcription factor Nrf1 negatively regulates the cystine/glutamate transporter and lipid-metabolizing enzymes. Mol Cell Biol. 34:3800–3816. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Xu Y, Luo Y, Wang ZY, Li X, Zheng P and Zhang TC: MRTF-A can activate Nrf2 to increase the resistance to doxorubicin. Oncotarget. 8:8436–8446. 2017.

19 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

20 

Schneider CA, Rasband WS and Eliceiri KW: NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 9:671–675. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Hermann MR, Jakobson M, Colo GP, Rognoni E, Jakobson M, Kupatt C, Posern G and Fässler R: Integrins synergise to induce expression of the MRTF-A-SRF target gene ISG15 for promoting cancer cell invasion. J Cell Sci. 129:1391–1403. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Eisenach PA, Schikora F and Posern G: Inhibition of arginyltransferase 1 induces transcriptional activityof myocardin-related transcription factor A (MRTF-A) and promotes directional migration. J Biol Chem. 289:35376–35387. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Zhang WL, Lv W, Sun SZ, Wu XZ and Zhang JH: miR-206 inhibits metastasis-relevant traits by degrading MRTF-A in anaplastic thyroid cancer. Int J Oncol. 47:133–142. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Griffiths-Jones S, Saini HK, Van Dongen S and Enright AJ: miRBase: Tools for microRNA genomics. Nucleic Acids Res. 36:D154–D158. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Edris B: A comparison of the oligomap and targetscan algorithms for miRNA target analysis. BMI. 231:2011.

26 

Chen L, Kwong M, Lu R, Ginzinger D, Lee C, Leung L and Chan JY: Nrf1 is critical for redox balance and survival of liver cells during development. Mol Cell Biol. 23:4673–4686. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Gęgotek A and Skrzydlewska E: CNC proteins in physiology and pathology. Postepy Hig Med Dosw (Online). 69:729–743. 2015.In Polish. View Article : Google Scholar

28 

Zhang Q, Pi J, Woods CG and Andersen ME: A systems biology perspective on Nrf2-mediated antioxidant response. Toxicol Appl Pharmacol. 244:84–97. 2010. View Article : Google Scholar :

29 

Zhang Y, Li S, Xiang Y, Qiu L, Zhao H and Hayes JD: The selective post-translational processing of transcription factor Nrf1 yields distinct isoforms that dictate its ability to differentially regulate gene expression. Sci Rep. 5:129832015. View Article : Google Scholar : PubMed/NCBI

30 

Ren Y, Qiu L, Lü F, Ru X, Li S, Xiang Y, Yu S and Zhang Y: TALENs-directed knockout of the full-length transcription factor Nrf1α that represses malignant behaviour of human hepatocellular carcinoma (HepG2) cells. Sci Rep. 6:237752016. View Article : Google Scholar

31 

Scharenberg MA, Chiquet-Ehrismann R and Asparuhova MB: Megakaryoblastic leukemia protein-1 (MKL1): Increasing evidence for an involvement in cancer progression and metastasis. Int J Biochem Cell Biol. 42:1911–1914. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Medjkane S, Perez-Sanchez C, Gaggioli C, Sahai E and Treisman R: Myocardin-related transcription factors and SRF are required for cytoskeletal dynamics and experimental metastasis. Nat Cell Biol. 11:257–268. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Descot A, Hoffmann R, Shaposhnikov D, Reschke M, Ullrich A and Posern G: Negative regulation of the EGFR-MAPK cascade by actin-MAL-mediated Mig6/Errfi-1 induction. Mol Cell. 35:291–304. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Leitner L, Shaposhnikov D, Descot A, Hoffmann R and Posern G: Epithelial protein lost in neoplasm α (Eplin-alpha) is transcriptionally regulated by G-actin and MAL/MRTF coactivators. Mol Cancer. 9:602010. View Article : Google Scholar

35 

Yoshio T, Morita T, Tsujii M, Hayashi N and Sobue K: MRTF-A/B suppress the oncogenic properties of v-ras- and v-src-mediated transformants. Carcinogenesis. 31:1185–1193. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 42 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu Y, Luo Y, Liang C, Xing W and Zhang T: A regulation loop between Nrf1α and MRTF-A controls migration and invasion in MDA-MB-231 breast cancer cells. Int J Mol Med 42: 2459-2468, 2018
APA
Xu, Y., Luo, Y., Liang, C., Xing, W., & Zhang, T. (2018). A regulation loop between Nrf1α and MRTF-A controls migration and invasion in MDA-MB-231 breast cancer cells. International Journal of Molecular Medicine, 42, 2459-2468. https://doi.org/10.3892/ijmm.2018.3816
MLA
Xu, Y., Luo, Y., Liang, C., Xing, W., Zhang, T."A regulation loop between Nrf1α and MRTF-A controls migration and invasion in MDA-MB-231 breast cancer cells". International Journal of Molecular Medicine 42.5 (2018): 2459-2468.
Chicago
Xu, Y., Luo, Y., Liang, C., Xing, W., Zhang, T."A regulation loop between Nrf1α and MRTF-A controls migration and invasion in MDA-MB-231 breast cancer cells". International Journal of Molecular Medicine 42, no. 5 (2018): 2459-2468. https://doi.org/10.3892/ijmm.2018.3816