Figures
Abstract
TYR, DCT and MITF are three important genes involved in maintaining the mature phenotype and producing melanin; they therefore participate in neural crest cell development into melanocytes. Previous studies have revealed that the Wnt signaling factor lymphoid enhancer-binding factor (LEF-1) can enhance DCT and MITF gene expression. However, whether LEF-1 also affects TYR gene expression remains unclear. In the present study, we found that LEF-1 regulated TYR transcription in vitro. LEF-1 overexpression increased TYR gene promoter activity, whereas LEF-1 knockdown by RNA interference significantly decreased TYR expression. Moreover, the core GTTTGAT sequence (-56 to -50) within the TYR promoter is essential for the effect of LEF-1 on TYR expression, and chromatin immunoprecipitation (ChIP) assay indicated that endogenous LEF-1 interacts with the TYR promoter. In addition, we observed a synergistic transactivation of the TYR promoter by LEF-1 and MITF. These data suggest that Wnt signaling plays an important role in regulating melanocyte development and differentiation.
Citation: Wang X, Liu Y, Chen H, Mei L, He C, Jiang L, et al. (2015) LEF-1 Regulates Tyrosinase Gene Transcription In Vitro. PLoS ONE 10(11): e0143142. https://doi.org/10.1371/journal.pone.0143142
Editor: Andrzej T. Slominski, University of Alabama at Birmingham, UNITED STATES
Received: May 4, 2015; Accepted: October 30, 2015; Published: November 18, 2015
Copyright: © 2015 Wang et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited
Data Availability: All relevant data are within the paper and its supporting information files.
Funding: This study was supported by National Basic Research and Development Program of China (973 Program) (No. 2014CB541702, to Yong Feng), the National Natural Science Foundation of China (Nos. 81170923, 81470705 to Yong Feng; No. 81300833 to Lu Jiang), The public welfare industry research special fund projects of ministry of health of China (No. 201302001, to Yong Feng). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing interests: The authors have declared that no competing interests exist.
Introduction
Melanocytes and their progenitor cells melanoblasts originate from multi-potential neural crest stem cells, which then migrate through the developing embryo and localize to specific sites in the body. In addition, they comprise a stem cell pool for their regeneration [1,2], which may be an important factor in melanoma development from melanocytes [3]. The visible pigmentation in skin, hair and eyes primarily depends on the presence of melanin, a macromolecule synthesized by melanocytes. Tyrosinase is considered the key enzyme in melanogenesis initiation, as normal melanin formation does not occur without tyrosinase, and the lack of this enzyme causes albinism [4–8]. Tyrosinase is encoded by the TYR gene, which maps to chromosome 11q14-21 in humans and chromosome 7 in mouse [9]. Meanwhile, different types of melanin are associated with pigmentation. The TYRP1 and TYRP2/DCT proteins, with ~40% amino acid homology with TYR, have been demonstrated to play important roles in controlling the type of melanin [10–14]. Several cis-acting elements mediate the expression of these genes. For instance, microphthalmia-associated transcription factor (MITF) is a basic helix-loop-helix transcription factor, and it has been suggested to be a key regulator of TYR and DCT transcription through the E-box (CANNTG) in their promoters [15,16].
Lymphoid enhancer-binding factor 1 (LEF-1) is a member of the LEF/T-cell-specific factor (TCF) family of the high mobility group domain transcription factors, and it is a downstream nuclear Wnt signaling pathway mediator [17]. It is well established that LEF-1 is involved in the development and malignant progression of human cancers, such as melanoma, colorectal cancer, acute myeloid leukemia and pancreatic cancer [18–22]. Moreover, LEF-1 participates in embryogenesis and postnatal development by interacting with β-catenin [23–26]. LEF-1 contains three functional domains: β-catenin binding domain, context-dependent domain and high-mobility group protein domain (HMG) [17,27,28]. LEF-1 binds to the CCTTTGWW (W, A/C/T) consensus sequence in the minor groove of DNA via its HMG domain and induces a sharp bend in the DNA helix [17]. It has been shown that LEF-1 transactivates melanocyte-specific MITF isoform (MITF-M) by physically interacting with the MITF-M promoter [29]. It has also been shown that LEF-1 and MITF-M synergism is responsible for the regulation of DCT gene transcription [29].
In this study, we investigated the role of LEF-1 in the regulation of TYR gene expression. Our data indicated that LEF-1 binds to the TYR promoter and activates TYR gene expression. Additionally, we observed synergistic transactivation of the TYR promoter by LEF-1 and MITF.
Materials and Methods
Ethics statement
This study was approved by the Expert Committee of Xiangya Hospital of Central South University (equivalent to an Institutional Review Board). All experiments were conducted in cell lines.
Reporter and expression plasmid construction
The luciferase reporter construct containing the human TYR promoter (-300 bp to +80 bp from the transcription start site) and human pCMV-3×Flag-MITF plasmid were generated as described previously [30]. The luciferase reporter constructs containing various fragments of the human TYR promoter (TYR1-Luc (-187 bp to +80 bp), TYR2-Luc (-23 bp to +80 bp), TYR3-Luc (-46 bp to +80 bp), TYR4-Luc (-90 bp to +80 bp)) were generated by inserting the corresponding DNA fragments into the pGL3-basic vector. A luciferase reporter construct containing the human TYR promoter with a mutation in the LEF-1 binding sites (TYR5-Luc) and with a mutation in the MITF binding sites (TYR6-Luc) were generated using the QuikChange II Site-Directed Mutagenesis Kit (GE Healthcare, Chalfont St. Giles, Buckinghamshire, UK)). The pcDNA3.1-LEF-1-HA expression vector containing full-length human LEF-1 cDNA (GenBank Accession No: NM_016269.4) or a dominant-negative LEF-1 (DNLEF-1) lacking the β-catenin-binding domain (amino acid residue 2–37) [27] was generated by the Nanjing Genescript Biotechnology Company (China). All plasmids were confirmed by automatic sequencing analysis.
Cell culture, transfectionand luciferase reporter assays
Human embryonic kidney 293T (HEK293T) cells [31], human cervical carcinoma HeLa cells [32] and NIH3T3 cells [30,31] were kindly provided by JD Li (State Key Laboratory of Medical Genetics of China). Human melanoma UACC903 cell lines were purchased from the University of Arizona Cancer Center [33]. All cells were cultured in DMEM/HIGH GLUCOSE (Hyclone, Logan, UT, USA) supplemented with 10% fetal bovine serum (FBS) (Invitrogen, Carlsbad, CA, USA) at 37°C in 5% CO2. For some experiments, 293T cells were cultured in two different types of culture medium: serum-free DMEM and RPMI 1640 (Hyclone, Logan, UT, USA) supplemented with 10% FBS and 2 mM L-glutamine. All of the above media were supplemented with 100 U/ml penicillin/streptomycin (Invitrogen, Carlsbad, CA, USA). Transient transfection assays were conducted using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s instructions. Cells were seeded at 50% confluence in 24-well plates 24 h before transfection. Cells were transfected with 5 ng reporter plasmids, 20 ng expression vector and 5 ng pCMV-β-gal (BD Bioscience/ Clontech, Palo Alto, CA, USA). The final DNA concentration added to each well was adjusted to 200 ng with empty vectors. Cells were washed with PBS and lysed with Reporter Lysis Buffer at 48 h after transfection (Promega, Madison, WI, USA). Extracts were used to determine luciferase and β-galactosidase activity. A luciferase reporter assay was conducted using the Luciferase Assay System (Promega, Madison, WI, USA) according to the manufacturer’s protocol. Luciferase activity was determined using a SIRIUS luminometer (Berthold Detection Systems, GmbH. Pforzheim, Germany) and normalized by measuring β-galactosidase activity. The relative luciferase activities were shown as the ratio of each normalized luciferase activity to the value obtained with pGL3-TYR-Luc and empty vector.
Small interfering RNA of LEF-1 and MITF knockdown
SiRNAs specifically targeting the human LEF-1 gene (siLEF-1) or MITF gene (siMITF) were designed and synthesized by RiboBioCo. Ltd. (Guangzhou, China) as follows: siLEF-1: (Lsi1: 5’- GGAA AAGAUCU U CGCCGAGdTdT-3’ (sense) and 5’-CUCGGCGAAGAUCUUUUCCdTdT-3’ (antisense); Lsi2: 5’-GC AAGAGACAAUUAUGGUAdTdT-3’ (sense) and 5’-UACCAUAAUUGUCUCUUGCdTdT-3’ (antisense); Lsi3: 5’-GAAAGGAGCAGGAGCCAAAdTdT- 3’ (sense) and 5’-UUUGGCUCCUGCUCCUUUCdTdT -3’ (antisense)), and siMITF: (Msi1: 5‘ GUACCUUUCUACCACUUUAdTdT 3‘ (sense) and 3‘ dTdTC AUGG AAAGAUGGUGAAAU 5‘ (antisense); Msi2: 5‘ GCUUGCCAUGUCCAAACCAdTdT 3‘(sense) and 3‘ dTdTCGAACGGUACAGGUUUGGU 5’; Msi3: 5‘GGCUAUGCUUACGCUUAACdTdT 3‘ and 3‘ dT dTCCGAUACGAAUGCGAAUUG 5‘). The negative control siRNA was purchased from Guangzhou RiboBioCo. Ltd. UACC903 cells were transfected with negative control siRNA (100 nM) or target gene siRNA (siLEF-1 or siMITF) (100 nM) using RiboFect CP Transfection Kit (166 T) (RiboBioCo. Ltd. Guangzhou, China) according to the manufacturer’s instructions. After transfection for 72 h, cells were subjected to either Western blotting or reverse transcription polymerase chain reaction (RT-PCR).
Reverse transcription polymerase chain reaction (RT-PCR)
Total RNA was extracted from cells using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. A total of 2 μg RNA was used for reverse transcription with RevertAid First Strand cDNA Synthesis Kit (Thermo Fisher Scientific, NY, USA) according to the manufacturer’s protocol. Real-time PCR was performed using SYBR Premix Ex Taqll (TakaRa, Japan). The primers against LEF-1 were: 5’-acagatcaccccacctcttg-3’ (Forward) and 5’-tgatgggaaaacctggacat-3’ (Reverse). The primers against MITF were: 5’-gggagctcacagcgtgtatt-3’ (Forward) and 5’-atggttcgttccttccagcg-3’. The primers against β-actin were: 5’-CCCATCTATGAGGGTTACGC-3’ (Forward) and 5’-TTTAATGTCACGCACGATTTC-3’ (Reverse). Primers were designed and synthesized by Sangon Biotecl Company (Shanghai, China). β-actin was used as an internal reference for normalizing mRNA expression of target genes. The PCR cycling conditions were as follows: 95°C for 5 min, followed by 40 cycles at 94°C for 30 s, 60°C for 30 s and 72°C for 30 s. The PCR reaction was performed using an iCycler IQ Multicolor Reverse-Transcription Detection System (Bio-Rad systems, Hercules, CA, USA).The relative expression of target genes was calculated by the 2-△△ct method [34]. The significant differences were analyzed by SPSS 19.0 software.
Western blots
UACC903 cells in 6-well plates were transfected with 100 μM siLEF-1 or negative control siRNA using a RiboFect CP Transfection Kit (166 T) (RiboBioCo. Ltd., Guangzhou, China) according to the manufacturer’s instructions. Cells were lysed in 2× SDS loading buffer containing 1 mM phenylmethanesulfonyl fluoride (PMSF) (Sigma, St. Louis, WA, USA), 0.2 mM β-mercaptoethanol and protease inhibitor cocktail (Sigma)72 h after transfection. A total of 10 μg protein was separated on 10% sodium dodecyl sulfate polyacrylamide gels (SDS-PAGE), which was then transferred onto a polyvinylidene fluoride (PVDF) membrane (Millipore, Billerica, MA, USA). The membrane was blocked in Tris-buffered saline supplemented with 5% non-fat milk for 2 h at room temperature and then incubated overnight at 4°C with ChIP Ab+ LEF-1 mouse monoclonal antibody (1:1000 dilution, Millipore, Billerica, MA, USA 17604), anti-MITF mouse monoclonal antibody (1:1000 dilution, Sigma, St. Louis, WA, USA M6065), anti-TYR mouse monoclonal antibody (1:1000 dilution, Cell Signaling Technology, Boston, MA, USA 9416s) or anti-GAPDH antibody (1:5000 dilution, Abcam, Cambridge, UK, ab8245). After washing in Tris-buffered saline plus 0.1% Tween 20, the membrane was incubated with a horseradish peroxidase-conjugated secondary anti-mouse IgG antibody (1:10000, Sigma, St. Louis, WA, USA) for 1 h at room temperature. Detection was performed using the ECL plus Western blotting detection system (GE Healthcare, UK) according to the manufacturer’s instructions.
Chromatin Immunoprecipitation (ChIP) assays
UACC903 cells were cross-linked with 1% formaldehyde and incubated at 37°C for 10 min. ChIP assay was performed according to the Millipore Biotechnology protocol with minor modifications. Cell lysates were sonicated by Covaris S2 (USA). Equal aliquots of isolated chromatin were subjected to immunoprecipitation with a ChIP Ab+ LEF-1 mouse monoclonal antibody (1:200 Millipore, Billerica, MA, USA 17604) or normal mouse IgG (Sigma, St. Louis, WA, USA) as a negative control. The immunoprecipitated DNA was used to PCR to amplify the TYR promoter sequence. The primers used were as follows: forward, 5’-TAACTGGGT TTGCTTAGGT-3’; reverse, 5’ -TAATACCACTCCCACCTCC-3’. The PCR products were subjected to 2% agarose gel electrophoresis.
Co-Immunoprecipitation Assays
HeLa cells were seeded in a 100-mm plate. When cells reached 80% confluence, they were transfected with 6 μg pcDNA3.1-LEF-1-HA and 6 μg pCMV-3×Flag-MITF or its mutants using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA), according to the manufacturer’s instructions. At 36 to 48 h after transfection, cells were lysed in RIPA buffer [150 mM NaCl, 50 mM Tris-His (pH, 8.0), 1% NonidetP-40, 2 mM EDTA, 10% Glycerol] with protease inhibitor cocktail and 1 mM PMSF. Cell lysis buffer (30 μl) was extracted and supplemented with 30 μl 2× SDS lysis buffer/0.2 mM β-mercaptoethanol for immunoblotting. The remaining lysis buffer was divided into two equal parts. Two parts were adjusted to a total volume of 1 ml with RIPA buffer, one of which was incubated with 1 μg anti-Flag M2 monoclonal antibody (Sigma, St. Louis, WA, USA, F1804) for IP with rotation overnight at 4°C, and the other aliquot was incubated with 1 μg normal mouse IgG as a negative control. A total of 30 μl protein G agarose beads were added to the samples, and the samples were incubated for another 2 h at 4°C with nutation. The beads were then washed five times in1 ml ice-cold PBS and were resuspended in 2× SDS lysis buffer/0.2 mM β-mercaptoethanol. Samples were boiled for 10 min and subjected to 12% SDS-PAGE gel electrophoresis. Immunoblot analysis was performed using an anti-HA rabbit polyclonal antibody (1:1000, Cell Signaling Technology, Boston, MA, USA 3724s).
Statistics analysis
Data are presented as the mean ± standard deviation (SD) and were analyzed using GraphPad Prism version 5.0 software (GraphPad Software Inc. San Diego, CA, USA) unless noted otherwise. The statistical difference between two groups was analyzed using a Student’s t-test. Comparisons between more than two groups were analyzed using one-way ANOVA. A p-value less than 0.05 was considered statistically significant. All experiments were performed at least three times in triplicate on different days using different batches of cells.
Results and Discussion
LEF-1 knockdown decreased TYR expression
Wnt signaling plays a critical role in regulating melanocyte development and differentiation. It has been demonstrated that Wnt3a can increase melanin synthesis and upregulate MITF expression and its downstream target genes, including TYR, DCT and TYRP1 [35]. Moreover, Wnt signaling inhibition suppresses melanin synthesis and MITF and TYR expression [36,37]. Wnt1- or Wnt3a-deficient mutant mice are almost devoid of pigment cells [38]. In addition, Wnt signaling activated target gene expression through the interaction of β-catenin and a LEF-1/TCF transcription factor family member [39,40].
To investigate whether LEF-1 endogenously affected TYR gene expression, we down-regulated LEF-1 expression by using small interference RNA (siRNA). As indicated in Fig 1A, Lsi1, Lsi2 and Lsi3 each effectively decreased LEF-1 mRNA levels. However, only Lsi2 and Lsi3 significantly down-regulated LEF-1 protein levels (Fig 1B). Moreover, we showed that LEF-1 knockdown led to a significant decrease in TYR protein levels (Fig 1C).
A, RT-qPCR data showed that transfection with LEF-1 specific siRNAs led to a significant decrease in LEF-1 expression compared to the control group, especially Lsi1 and Lsi2. An asterisk indicated statistical significance (**p<0.01, ***p<0.001 by unpaired t-test). B, C Western blotting was performed to examine LEF-1 and TYR protein levels in each group using a ChIP Ab+ LEF-1 mouse monoclonal antibody or anti-TYR mouse monoclonal antibody, respectively. GAPDH was used as an internal reference.
LEF-1 enhanced the human TYR promoter activity
Because LEF-1 has been demonstrated to enhance MITF expression, which can in turn activate TYR expression [29,30,41], we examined whether LEF-1 directly or indirectly affected TYR expression, we employed the web-based bioinformatics analysis program Genomatrix browser (http://www.genomatix.de/) and identified several LEF-1 binding sites in the TYR promoter, suggesting that LEF-1 may directly promote TYR transcription. We performed a luciferase assay with a luciferase reporter construct containing -300 bp to 80 bp of the TYR promoter (Fig 2A). As shown in Fig 2B, LEF-1 dramatically increased TYR promoter activity in a dose-dependent manner. We also observed a LEF-1-induced increase in TYR promoter activity in several other cell lines, including HEK293T, HeLa and NIH3T3 cells (S1A Fig). Because DMEM itself and calf serum can upregulate tyrosinase or melanogenesis [42–44], we repeated the luciferase assay using serum-free DMEM or RPMI medium in UACC903 cells. As shown in S1B Fig and S1C Fig, TYR expression mildly decreased compared to its expression in DMEM medium, but the trend was similar.
A Schematic representation of the human TYR promoter sequence containing 380 bp (-300 bp to +80 bp) together with the potential LEF-1 binding sites (double underlined) and their flanking sequences. B TYR transcriptional activity determined by Luciferase activity assay. The TYR-Luc luciferase reporter plasmid was transiently transfected into melanoma UACC903 cells in combination with increasing amounts of LEF-1 expression vector. The basal luciferase level was set to 1. Data from all other transfections are presented as fold induction above this level. Luciferase activity was normalized by measuring β-galactosidase activity. Each value represents the mean ± SD of three replicates from a single assay. The results shown were representative of at least three independent experiments. (**p<0.01, ***p<0.001 compared to basal activity, unpaired Student’s t-test).
To localize the cis-acting region within the TYR promoter required for LEF-1-induced activation, we generated a series of reporter constructs containing a truncated TYR promoter. As indicated in Fig 3A, deletion of the fragment from -300 bp to -188 bp (TYR1-Luc) did not affect LEF-1-induced TYR promoter activity; however, LEF-1 failed to activate the TYR promoter containing only the fragment from -23 bp to + 80 bp (TYR2-Luc). In addition, LEF-1 activated the TYR4-Luc construct (-90 bp to +80 bp), but not the TYR3-Luc construct (-44 bp to +80 bp), suggesting that there was a LEF-1- interacting site in the region from -90 bp to -44 bp.
A, B Transcriptional activity of LEF-1 on various TYR deleted or mutated promoters (TYR5) by luciferase assay. UACC903 cells were transiently transfected with each reporter plasmid (5 ng) and the LEF-1 expression plasmid (20 ng). Cells were lysed and luciferase activity was detected 48 h after transfection. (***p<0.001 by one-way ANOVA with Dunnett’s multiple comparison tests compared to basal activity; ###p<0.001; ns, not significant compared to the value from the TYR promoter and LEF-1 by an unpaired Student’s t-test). C Binding of LEF-1 to the TYR promoter was analyzed by ChIP assay by precipitating with the indicated antibodies and normal mouse IgG, which was used as the negative control. The crosslinks were reversed at 65°C for 4 h and digested with proteinase K (Sigma-Aldrich) for 1 h at 45°C to remove proteins. A 230-bp TYR promoter fragment (-28 to -257 bp) was amplified by PCR, and the products were subjected to 2% agarose gel electrophoresis. Ten percent of the chromatin DNA used for immunoprecipitation was subjected to PCR and is indicated as ‘input’. The results were representative of at least three independent experiments.
The region from -90 bp to -44 bp contained one putative LEF-1 binding site (AAGTGTTTGATGCTG, -60 bp to -46 bp). Accordingly, we generated a TYR5-Luc construct in which the core sequence GTTTGAT (-56 bp to -50 bp) was mutated to GAAAAGA. As indicated in Fig 3B, LEF-1 failed to activate TYR5-Luc, indicating that the core GTTTGAT sequence within the TYR promoter is a bona fide LEF-1 binding site.
We further performed a ChIP assay to confirm whether endogenous LEF-1 binds to the TYR promoter. We designed a primer pair containing the TYR promoter region from -28 bp to -257 bp. As shown in Fig 3C, the LEF-1 antibody, but not the control IgG, immunoprecipitated the DNA fragment containing the TYR promoter. Based on these findings, we demonstrated that LEF-1 could transactivate the TYR gene by binding to a conserved site in the TYR promoter region from -60 bp to -46 bp.
β-catenin is required for LEF-1 to activate the TYR promoter
β-catenin is a key downstream effector of the Wnt signaling pathway and functions as a transcriptional co-activator by interacting with TCF/LEF transcription factors [27,45]. Previous studies have shown that β-catenin is involved in LEF-1-mediated MITF-M gene expression [29,46]. Therefore, we studied whether β-catenin was also required for TYR promoter enhancement by LEF-1. We performed a luciferase assay with the TYR-Luc reporter construct and a dominant negative LEF-1 (DNLEF-1) that lacks the β-catenin-binding sites (2–37 amino acid). As shown in Fig 4A, TYR promoter transactivation by DNLEF-1 was dramatically reduced compared to wild type LEF-1, indicating that β-catenin is indeed required for TYR gene expression.
A, B Synergism between MITF and LEF-1 and the effect of β-catenin on TYR promoter determined by luciferase activity assay. UACC903 cells were transfected with the TYR promoter-reporter plasmid and the indicated effector plasmid(s). A luciferase assay was then conducted. Relative luciferase activities are expressed as the mean ± SD from three independent experiments each performed in triplicate. (**p<0.01 by unpaired t-test).C Interaction between LEF-1 and MITF. Flag-tagged full-length MITF (MITF-Flag) was expressed in UACC903 cells and immunoprecipitated by the anti-Flag M2 antibody and Protein G agarose. The interaction between HA-tagged-full-length LEF-1 (LEF-1-HA) and MITF-Flag was examined by Western blotting using an anti-HA rabbit polyclonal antibody in the Flag-IP sample. MITF-Flag and LEF-1-HA protein levels were assessed by Western blotting using the corresponding antibodies.
These findings indicated that LEF-1 played an important role in keeping the transcription of TYR gene when even lacking of Wnt signaling.
TYR promoter transactivation by MITF and LEF-1
It has been reported that LEF-1 can interact with MITF-M to enhance DCT and MITF-M gene transcription [29,46]. Thus, we transfected MITF and LEF-1 with the TYR-Luc reporter construct into HeLa cells, which lack endogenous LEF-1 and MITF-M expression [29,47,48]. As shown in Fig 4B, we observed a synergistic transactivation of the TYR promoter by MITF and LEF-1. As expected, LEF-1 and MITF could be co-immunoprecipitated when co-expressed in HeLa cells (Fig 4C). Nevertheless, transfection with MITF and DNLEF-1 lacking β-catenin-binding sites led to an obvious decrease in TYR promoter transactivation by cooperation between MITF and LEF-1 (Fig 4B). These finding indicated that β-catenin is required for the efficient cooperation of LEF-1 with MITF-M on the TYR promoter.
MITF is considered a key transcription factor for melanocyte differentiation, and it controls TYR expression [30,41]. MITF transcriptionally regulates TYR, and LEF-1 regulates MITF [29]. To further demonstrate the direct effect of LEF-1 on tyrosinase transcription, we conducted a luciferase assay using a TYR-Luc reporter with the core MITF binding sequence CATGTG mutated to CATTTG (TYR6-luc). As shown in Fig 5A, MITF itself transactivated TYR-Luc but not TYR6-Luc; however, LEF-1 activated both promoters. Furthermore, we transfected cells with MITF-specific siRNAs, which effectively decreased MITF mRNA and protein levels (Fig 5B and 5C). Although MITF knockdown mildly decreased basal luciferase activity, LEF-1 significantly enhanced tyrosinase promoter activity in the presence of MITF siRNA (Fig 5D). Our results demonstrated that LEF-1 directly bound to and enhanced tyrosinase promoter activity.
A LEF-1-mediated tyrosinase promoter activity determined by luciferase activity assay when MITF expression was inhibited. UACC903 cells were co-transfected with the indicated promoter reporter plasmid and effector plasmid (s). A luciferase assay was performed 48 h after transfection. Relative luciferase activities are expressed as the mean ± SD from three independent experiments each performed in triplicate. (***p<0.001, compared to the value from the TYR6 promoter and empty vector; ns, not significant, ###p<0.001, compared to WT; unpaired Student’s t-test). B, C Examination of MITF-specific siRNAs. Melanoma UACC903 cells were transfected with MITF-specific siRNAs. RT-qPCR data showed a significant decrease in MITF expression compared to the control group, especially Msi1. An asterisk indicated statistical significance (**p<0.01, ***p<0.001 by an unpaired t-test) (B).Western blotting was performed to examine MITF and TYR protein levels in each group using an anti-MITF mouse monoclonal antibody or anti-TYR mouse monoclonal antibody, respectively. GAPDH was used as an internal reference (C). D LEF-1 significantly enhanced tyrosinase promoter activity in the presence of MITF siRNA. UACC903 cells were transiently transfected by the TYR promoter reporter in combination with LEF-1 or Msi1.Cells were lysed and luciferase assays were conducted 48 h after transfection. Basal luciferase activitywas set to 1. Data from all other transfections are presented as fold induction above basal levels. Luciferase activity was normalized by measuring β-galactosidase activity. Each value shown was the mean ± SD of three replicates from a single assay. The results shown were representative of at least three independent experiments. (**p<0.01, ***p<0.001 compared with basal activity, unpaired Student’s t-test).
In summary, our study demonstrates that LEF-1can positively mediate TYR expression. Moreover, LEF-1 is involved in the transcriptional regulation of DCT and MITF-M [29,46]. In addition, TYR, DCT and MITF-M are all regulated by synergism between MITF and LEF-1, and β-catenin is required for their efficient expression [29,46]. Accordingly, our study highlights an important role of Wnt signaling in the melanocyte development and differentiation.
Supporting Information
S1 Fig. TYR promoter activities determined by luciferase activity assay in different cells or medium.
A Luciferase activity was detected when LEF-1 and the TYR promoter (5 ng) were co-transfected into HEK293T, HeLa or NIH3T3 cells. The basal level of all luciferase activity from the three cells was set to 1. Data from other transfections are presented as fold induction above the basal level. Luciferase activity was normalized by measuring β-galactosidase activity. The data are presented as the mean ± SD from three independent experiments each performed in triplicate. (**p<0.01, ***p<0.001 by one-way ANOVA with Dunnett’s multiple comparison tests). B, C Luciferase assays were repeated when LEF-1 was co-transfected with the TYR promoter (5 ng) into 293T cells in RPMI or serum-free DMEM cultures.
https://doi.org/10.1371/journal.pone.0143142.s001
(TIF)
Acknowledgments
We would like to thank all of the technical staff from the State Key Laboratory of Medical Genetics and Province Key Laboratory of Otolaryngology Critical Disease, for their support. We would also like to thank Dr. Hua Zhang for generously supplying the materials.
Author Contributions
Conceived and designed the experiments: XPW JDL YF. Performed the experiments: XPW. Analyzed the data: XPW ZJN. Contributed reagents/materials/analysis tools: XPW JDL YF YLL HSC LYM CFH JS ZJN LJ HJL. Wrote the paper: XPW JDL YF. Obtained permission for use of cell line: JDL.
References
- 1. Rawles ME (1947) Origin of pigment cells from the neural crest in the mouse embryo. Physiol Zool 20: 248–266. pmid:20256541
- 2. Nishimura EK, Granter SR, Fisher DE (2005) Mechanisms of hair graying: incomplete melanocyte stem cell maintenance in the niche. Science 307: 720–724. pmid:15618488
- 3. Fukunaga-Kalabis M, Hristova DM, Wang JX, Li L, Heppt MV, Wei Z, et al. (2015) UV-Induced Wnt7a in the Human Skin Microenvironment Specifies the Fate of Neural Crest-Like Cells via Suppression of Notch. J Invest Dermatol 135: 1521–1532. pmid:25705850
- 4. Spritz RA (1994) Molecular genetics of oculocutaneous albinism. Hum Mol Genet 3 Spec No: 1469–1475.
- 5. Oetting WS, King RA (1999) Molecular basis of albinism: mutations and polymorphisms of pigmentation genes associated with albinism. Hum Mutat 13: 99–115. pmid:10094567
- 6. Takeda A, Tomita Y, Matsunaga J, Tagami H, Shibahara S (1990) Molecular basis of tyrosinase-negative oculocutaneous albinism. A single base mutation in the tyrosinase gene causing arginine to glutamine substitution at position 59. J Biol Chem 265: 17792–17797. pmid:2120217
- 7. Tomita Y, Takeda A, Okinaga S, Tagami H, Shibahara S (1989) Human oculocutaneous albinism caused by single base insertion in the tyrosinase gene. Biochem Biophys Res Commun 164: 990–996. pmid:2511845
- 8. Land EJ, Ramsden CA, Riley PA (2004) Quinone chemistry and melanogenesis. Methods Enzymol 378: 88–109. pmid:15038959
- 9. Barton DE, Kwon BS, Francke U (1988) Human tyrosinase gene, mapped to chromosome 11 (q14——q21), defines second region of homology with mouse chromosome 7. Genomics 3: 17–24. pmid:3146546
- 10. Budd PS, Jackson IJ (1995) Structure of the mouse tyrosinase-related protein-2/dopachrome tautomerase (Tyrp2/Dct) gene and sequence of two novel slaty alleles. Genomics 29: 35–43. pmid:8530099
- 11. Jackson IJ, Chambers DM, Budd PS, Johnson R (1991) The tyrosinase-related protein-1 gene has a structure and promoter sequence very different from tyrosinase. Nucleic Acids Res 19: 3799–3804. pmid:1907365
- 12. Kwon BS (1993) Pigmentation genes: the tyrosinase gene family and the pmel 17 gene family. J Invest Dermatol 100: 134s–140s. pmid:8432998
- 13. Shibahara S, Taguchi H, Muller RM, Shibata K, Cohen T, Tomita Y, et al. (1991) Structural organization of the pigment cell-specific gene located at the brown locus in mouse. Its promoter activity and alternatively spliced transcript. J Biol Chem 266: 15895–15901. pmid:1908461
- 14. Kobayashi T, Vieira WD, Potterf B, Sakai C, Imokawa G, Hearing VJ (1995) Modulation of melanogenic protein expression during the switch from eu- to pheomelanogenesis. J Cell Sci 108 (Pt 6): 2301–2309. pmid:7673350
- 15. Yavuzer U, Keenan E, Lowings P, Vachtenheim J, Currie G, Goding CR (1995) The Microphthalmia gene product interacts with the retinoblastoma protein in vitro and is a target for deregulation of melanocyte-specific transcription. Oncogene 10: 123–134. pmid:7824265
- 16. Bertolotto C, Busca R, Abbe P, Bille K, Aberdam E, Ortonne JP, et al. (1998) Different cis-acting elements are involved in the regulation of TRP1 and TRP2 promoter activities by cyclic AMP: pivotal role of M boxes (GTCATGTGCT) and of microphthalmia. Mol Cell Biol 18: 694–702. pmid:9447965
- 17. Love JJ, Li X, Case DA, Giese K, Grosschedl R, Wright PE (1995) Structural basis for DNA bending by the architectural transcription factor LEF-1. Nature 376: 791–795. pmid:7651541
- 18. Eastman Q, Grosschedl R (1999) Regulation of LEF-1/TCF transcription factors by Wnt and other signals. Curr Opin Cell Biol 11: 233–240. pmid:10209158
- 19. Xu S, Yang Z, Zhang J, Jiang Y, Chen Y, Li H, et al. (2015) Increased levels of beta-catenin, LEF-1, and HPA-1 correlate with poor prognosis for acral melanoma with negative BRAF and NRAS mutation in BRAF exons 11 and 15 and NRAS exons 1 and 2. DNA Cell Biol 34: 69–77. pmid:25343173
- 20. Wang WJ, Yao Y, Jiang LL, Hu TH, Ma JQ, Ruan ZP, et al. (2013) Increased LEF1 expression and decreased Notch2 expression are strong predictors of poor outcomes in colorectal cancer patients. Dis Markers 35: 395–405. pmid:24223455
- 21. Metzeler KH, Heilmeier B, Edmaier KE, Rawat VP, Dufour A, Dohner K, et al. (2012) High expression of lymphoid enhancer-binding factor-1 (LEF1) is a novel favorable prognostic factor in cytogenetically normal acute myeloid leukemia. Blood 120: 2118–2126. pmid:22815426
- 22. Singhi AD, Lilo M, Hruban RH, Cressman KL, Fuhrer K, Seethala RR (2014) Overexpression of lymphoid enhancer-binding factor 1 (LEF1) in solid-pseudopapillary neoplasms of the pancreas. Mod Pathol 27: 1355–1363. pmid:24658583
- 23. Goding CR (2000) Mitf from neural crest to melanoma: signal transduction and transcription in the melanocyte lineage. Genes Dev 14: 1712–1728. pmid:10898786
- 24. Kuhl M, Sheldahl LC, Park M, Miller JR, Moon RT (2000) The Wnt/Ca2+ pathway: a new vertebrate Wnt signaling pathway takes shape. Trends Genet 16: 279–283. pmid:10858654
- 25. Boras-Granic K, Chang H, Grosschedl R, Hamel PA (2006) Lef1 is required for the transition of Wnt signaling from mesenchymal to epithelial cells in the mouse embryonic mammary gland. Dev Biol 295: 219–231. pmid:16678815
- 26. Driskell RR, Goodheart M, Neff T, Liu X, Luo M, Moothart C, et al. (2007) Wnt3a regulates Lef-1 expression during airway submucosal gland morphogenesis. Dev Biol 305: 90–102. pmid:17335794
- 27. Behrens J, von Kries JP, Kuhl M, Bruhn L, Wedlich D, Grosschedl R, et al. (1996) Functional interaction of beta-catenin with the transcription factor LEF-1. Nature 382: 638–642. pmid:8757136
- 28. Bruhn L, Munnerlyn A, Grosschedl R (1997) ALY, a context-dependent coactivator of LEF-1 and AML-1, is required for TCRalpha enhancer function. Genes Dev 11: 640–653. pmid:9119228
- 29. Yasumoto K, Takeda K, Saito H, Watanabe K, Takahashi K, Shibahara S (2002) Microphthalmia-associated transcription factor interacts with LEF-1, a mediator of Wnt signaling. EMBO J 21: 2703–2714. pmid:12032083
- 30. Zhang H, Luo H, Chen H, Mei L, He C, Jiang L, et al. (2012) Functional analysis of MITF gene mutations associated with Waardenburg syndrome type 2. FEBS Lett 586: 4126–4131. pmid:23098757
- 31. Wang HH, Chen HS, Li HB, Zhang H, Mei LY, He CF, et al. (2014) Identification and functional analysis of a novel mutation in the SOX10 gene associated with Waardenburg syndrome type IV. Gene 538: 36–41. pmid:24440785
- 32. Zhou W, Li J, Wang X, Hu R (2010) Stable knockdown of TPPP3 by RNA interference in Lewis lung carcinoma cell inhibits tumor growth and metastasis. Mol Cell Biochem 343: 231–238. pmid:20571904
- 33. Trent JM, Stanbridge EJ, McBride HL, Meese EU, Casey G, Araujo DE, et al. (1990) Tumorigenicity in human melanoma cell lines controlled by introduction of human chromosome 6. Science 247: 568–571. pmid:2300817
- 34. Schmittgen TD, Livak KJ (2008) Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 3: 1101–1108. pmid:18546601
- 35. Guo H, Yang K, Deng F, Ye J, Xing Y, Li Y, et al. (2012) Wnt3a promotes melanin synthesis of mouse hair follicle melanocytes. Biochem Biophys Res Commun 420: 799–804. pmid:22465131
- 36. Cho M, Ryu M, Jeong Y, Chung YH, Kim DE, Cho HS, et al. (2009) Cardamonin suppresses melanogenesis by inhibition of Wnt/beta-catenin signaling. Biochem Biophys Res Commun 390: 500–505. pmid:19800318
- 37. Yamaguchi Y, Passeron T, Hoashi T, Watabe H, Rouzaud F, Yasumoto K, et al. (2008) Dickkopf 1 (DKK1) regulates skin pigmentation and thickness by affecting Wnt/beta-catenin signaling in keratinocytes. FASEB J 22: 1009–1020. pmid:17984176
- 38. Ikeya M, Lee SM, Johnson JE, McMahon AP, Takada S (1997) Wnt signalling required for expansion of neural crest and CNS progenitors. Nature 389: 966–970. pmid:9353119
- 39. Cadigan KM, Nusse R (1997) Wnt signaling: a common theme in animal development. Genes Dev 11: 3286–3305. pmid:9407023
- 40. Barker N, Morin PJ, Clevers H (2000) The Yin-Yang of TCF/beta-catenin signaling. Adv Cancer Res 77: 1–24. pmid:10549354
- 41. Yasumoto K, Yokoyama K, Shibata K, Tomita Y, Shibahara S (1994) Microphthalmia-associated transcription factor as a regulator for melanocyte-specific transcription of the human tyrosinase gene. Mol Cell Biol 14: 8058–8070. pmid:7969144
- 42. Wolnicka-Glubisz A, Nogal K, Zadlo A, Plonka PM (2015) Curcumin does not switch melanin synthesis towards pheomelanin in B16F10 cells. Arch Dermatol Res 307: 89–98. pmid:25398276
- 43. Slominski A (1985) Some properties of Bomirski Ab amelanotic melanoma cells, which underwent spontaneous melanization in primary cell culture. Growth kinetics, cell morphology, melanin content and tumorigenicity. J Cancer Res Clin Oncol 109: 29–37. pmid:3972882
- 44. Slominski A, Moellmann G, Kuklinska E (1989) L-tyrosine, L-dopa, and tyrosinase as positive regulators of the subcellular apparatus of melanogenesis in Bomirski Ab amelanotic melanoma cells. Pigment Cell Res 2: 109–116. pmid:2497448
- 45. Dunn KJ, Williams BO, Li Y, Pavan WJ (2000) Neural crest-directed gene transfer demonstrates Wnt1 role in melanocyte expansion and differentiation during mouse development. Proc Natl Acad Sci U S A 97: 10050–10055. pmid:10963668
- 46. Saito H, Yasumoto K, Takeda K, Takahashi K, Fukuzaki A, Orikasa S, et al. (2002) Melanocyte-specific microphthalmia-associated transcription factor isoform activates its own gene promoter through physical interaction with lymphoid-enhancing factor 1. J Biol Chem 277: 28787–28794. pmid:12048204
- 47. Amae S, Fuse N, Yasumoto K, Sato S, Yajima I, Yamamoto H, et al. (1998) Identification of a novel isoform of microphthalmia-associated transcription factor that is enriched in retinal pigment epithelium. Biochem Biophys Res Commun 247: 710–715. pmid:9647758
- 48. Giese K, Kingsley C, Kirshner JR, Grosschedl R (1995) Assembly and function of a TCR alpha enhancer complex is dependent on LEF-1-induced DNA bending and multiple protein-protein interactions. Genes Dev 9: 995–1008. pmid:7774816