m (Clara moved page Draft García 537730221 to Li et al 2016a)
 
Line 1: Line 1:
 
==Abstract==
 
==Abstract==
  
Long non-coding RNAs (lncRNAs) have been proved to play important roles in the tumorigenesis and development of several human malignancies. Our study aims to investigate the expression and function of lncRNA-UCA1 in osteosarcoma. lncRNA-UCA1 expression was detected in osteosarcoma tissues and cell lines by using qRT-PCR. Association between lncRNA-UCA1 levels and clinicopathological factors and patients prognosis was analyzed. The roles of lncRNA-UCA1 in regulating osteosarcoma cell proliferation, apoptosis, migration, and invasion were evaluated in vitro. We found that lncRNA-UCA1 expression was upregulated in osteosarcoma tissues and cell lines. High lncRNA-UCA1 expression was significantly correlated with large tumor size, high tumor grade, positive distant metastasis, and advanced clinical stage. Multivariate regression analysis identified lncRNA-UCA1 overexpression as an independent unfavorable prognostic factor. lncRNA-UCA1 knockdown inhibited osteosarcoma cell proliferation, promoted cell apoptosis, and suppressed cell invasion and migration, whereas lncRNA-UCA1 overexpression showed opposite effects. These findings suggested that lncRNA-UCA1 may contribute to osteosarcoma initiation and progression, and would be not only a novel prognostic marker but also a potential therapeutic target for this disease.
+
Liver X receptor (LXR) plays an important role in reverse cholesterol transport (RCT), and activation of LXR could reduce atherosclerosis. In the present study we used a cell-based screening method to identify new potential LXR''β'' agonists. A novel benzofuran-2-carboxylate derivative was identified with LXR''β'' agonist activity: E17110 showed a significant activation effect on LXR''β'' with an EC<sub>50</sub> value of 0.72 μmol/L. E17110 also increased the expression of ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) in RAW264.7 macrophages. Moreover, E17110 significantly reduced cellular lipid accumulation and promoted cholesterol efflux in RAW264.7 macrophages. Interestingly, we found that the key amino acids in the LXR''β'' ligand-binding domain had distinct interactions with E17110 as compared to TO901317. These results suggest that E17110 was identified as a novel compound with LXR''β'' agonist activity ''in vitro via'' screening, and could be developed as a potential anti-atherosclerotic lead compound.
 +
 
 +
==Graphical abstract==
 +
 
 +
E17110 was identified as a novel LXR''β'' agonist by using a cell-based screening method. E17110 could increase the expression of ABCA1 and ABCG1 in RAW264.7 macrophages and significantly reduce cellular lipid accumulation and promote cholesterol efflux. Interestingly, we found that LXR''β'' had distinct interactions with E17110 compared to TO901317.
 +
 
 +
 
 +
[[Image:Li_et_al_2016a-1-s2.0-S2211383516300053-fx1.jpg|center|467px|fx1]]
  
 
==Abbreviations==
 
==Abbreviations==
  
lncRNA , long noncoding RNA ; UCA1 , Urothelial carcinoma associated 1 ; RT-PCR , reverse transcription-polymerase chain reaction ; DMSO , dimethyl sulfoxide ; MTT , 3-(4,5-dimethylthiazol-2-yl)−2,5-diphenyltetrazolium bromide ; DMEM , Dulbeccos modified Eagles medium ; TNM , tumor-node-metastasis
+
ABCA1, ATP-binding cassette transporter A1; ABCG1, ATP-binding cassette transporter G1; ApoA-I, apolipoprotein A-I; GAPDH, glyceraldehyde-phosphate dehydrogenase; HDL, high-density lipoprotein; LBD, ligand-binding domain; LXR, liver X receptor; LXRE, LXR response element; NR, nuclear receptor; ox-LDL, oxidized low-density lipoprotein; RCT, reverse cholesterol transport; RXR, retinoid X receptor
  
==Keywords==
+
==Key words==
  
Osteosarcoma ; LncRNA-UCA1 ; Prognosis ; Invasion
+
LXRβ; Atherosclerosis; ABCA1; ABCG1; Reverse cholesterol transport; Cholesterol efflux
  
 
==1. Introduction==
 
==1. Introduction==
  
Osteosarcoma is one of the most common primary bone tumor and occurs predominantly in children and young adults [[#bib1|[1]]] . With the development of multiple therapeutic strategies including wide tumor excision, neoadjuvant or adjuvant chemotherapy, and radiotherapy, the 5 year survival of the non-metastatic patients has increased to 65% [[#bib2|[2]]] . However, osteosarcoma is very aggressive and approximately 40–50% of patients will eventually develop metastases, especially pulmonary metastases [[#bib3|[3]]] . The prognosis of these patients is rather poor, and the long-term survival rate is only 10–30% [[#bib4|[4]]] . The complex molecular mechanisms underlying osteosarcoma tumorigenesis and progression remain largely unclear. Identification of new candidate molecules that take part in these processes is crucial for developing new therapeutic approach for osteosarcoma and improving clinical outcomes of patients with this disease.    
+
The liver X receptors (LXR''α'' and LXR''β'') are ligand-activated transcription factors that belong to the nuclear receptor (NR) superfamily <sup>[[#bib1|1]] and [[#bib2|2]]</sup>. LXR''β'' (NR1H2) is ubiquitously expressed at a moderate level in most physiological systems, whereas LXR''α'' (NR1H3) is mainly expressed in the intestine, kidney, spleen and adipose tissue, especially in the liver [[#bib3|<sup>3</sup>]]. LXRs generally function as permissive heterodimers with retinoid X receptor (RXR) that bind to specific response elements in the regulatory region of their target genes to regulate their expression[[#bib4|<sup>4</sup>]]. LXRs sense excess cholesterol and trigger various adaptive mechanisms to protect the cells from cholesterol overload. ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) are regulated by LXRs ''via'' functional LXR response elements (LXREs) found in their genes, which play important roles in cholesterol efflux <sup>[[#bib5|5]], [[#bib6|6]] and [[#bib7|7]]</sup>. ABCA1 can transfer both cholesterol and phospholipids to lipid-free apolipoprotein A-I (apoA-I), and ABCG1 can transfer cholesterol to high-density lipoprotein (HDL) <sup>[[#bib7|7]] and [[#bib8|8]]</sup>.
  
Long noncoding RNAs (lncRNAs) are defined as RNA transcripts of more than 200 nucleotides in length with no or little protein-coding capacity [[#bib5|[5]]] . LncRNAs can regulate gene expression through diverse mechanisms including epigenetic silencing, mRNA splicing, lncRNA-miRNA interaction, lncRNA-protein interaction and lncRNA-mRNA interaction [[#bib6|[6]]] . Recent studies showed that lncRNAs are involved in a wide range of biological processes, such as embryonic development, cell proliferation, apoptosis, invasion, metastasis and angiogenesis [[#bib7|[7]]] , [[#bib8|[8]]]  and [[#bib9|[9]]] . Furthermore, lncRNAs can act as oncogenes or tumor suppressors, and play important roles in carcinogenesis and cancer development [[#bib10|[10]]]  and [[#bib11|[11]]] . Abnormal lncRNA expression and its association with various important clinicopathological parameters have been reported in many types of cancers. In terms of osteosarcoma, increased lncRNA FGFR3-AS1 expression correlated with large tumor size, advanced Enneking stage, and poor survival [[#bib12|[12]]] . Plasma lncRNA TUG1 contributed to osteosarcoma detection and dynamic surveillance [[#bib13|[13]]] . Silence of lncRNA TUSC7 promoted osteosarcoma cell proliferation and increased colony formation in vitro [[#bib14|[14]]] . LncRNA ODRUL inhibition could inhibit osteosarcoma cell proliferation and migration, and partly reversed doxorubicin resistance [[#bib15|[15]]] . Therefore, lncRNAs may be utilized for osteosarcoma diagnosis and prognosis, and serve as potential therapeutic targets.    
+
Excessive absorption of lipoproteins in macrophages causes foam cell formation within arterial walls, and these cells subsequently rupture and promote early atherosclerotic plaque formation<sup>[[#bib9|9]] and [[#bib10|10]]</sup>. The efflux of excess cellular cholesterol from peripheral tissues and its return to the liver for excretion in the bile occurs by a process referred to as reverse cholesterol transport (RCT)[[#bib11|<sup>11</sup>]]. Furthermore, RCT is regarded as a major mechanism that removes cholesterol from the cells and transports it to the liver in order to protect against atherosclerotic cardiovascular disease, and this process can be stimulated by LXRs[[#bib11|<sup>11</sup>]].
  
Urothelial carcinoma associated 1 (UCA1) is an lncRNA originally identified in human bladder carcinoma [[#bib16|[16]]] . Overexpression of UCA1 significantly enhanced bladder cancer cell proliferation and migration and confered drug resistance. Recently, UCA1 has been reported to be upregulated and exert its oncogenic activity in several cancers such as esophageal squamous cell carcinoma [[#bib17|[17]]] , breast cancer [[#bib18|[18]]] , non-small cell lung cancer [[#bib19|[19]]] , gastric cancer [[#bib20|[20]]] , colorectal cancer[[#bib21|[21]]] , hepatocellular carcinoma [[#bib22|[22]]] , renal cell carcinoma [[#bib23|[23]]] , ovarian cancer [[#bib24|[24]]] , and prostate cancer [[#bib25|[25]]] . However, the significance of UCA1 in osteosarcoma is still unclear. In the present study, we investigated the expression level of UCA1 in osteosarcoma samples and cell lines. We also investigated the correlation between UCA1 expression and clinicopathological characteristics and patients survival. Moreover, we explored the role of UCA1 in the regulation of biological behaviors of osteosarcoma cells.    
+
Previous studies showed that treatment of atherosclerotic mice with synthetic LXR ligands effectively inhibited progression and promoted regression of atherosclerotic plaques<sup>[[#bib12|12]] and [[#bib13|13]]</sup>. Meanwhile, macrophage-specific deletion of LXR in mice enhances atherogenesis[[#bib14|<sup>14</sup>]]. Several LXR ligands, such as endogenous ligand 22(''R'')-hydroxycholesterol and synthetic agonists TO901317 and GW3965, have recently been reported and investigated substantially <sup>[[#bib15|15]], [[#bib16|16]] and [[#bib17|17]]</sup>. However, these ligands have the undesirable side effect of inducing lipogenesis and hypertriglyceridemia because of their up-regulation of sterol response element binding protein 1c (SREBP-1c) transcription[[#bib18|<sup>18</sup>]]. Therefore, the identification of novel LXR''β'' agonists which could achieve beneficial effects from regulating cholesterol metabolism is necessary.
 +
 
 +
In this study, we discovered E17110 as a novel benzofuran-2-carboxylate derivative with potential LXR''β'' agonist activity using an LXR''β''-GAL4 chimera reporter assay. We then investigated the effect and mechanism of this compound on the target genes of LXR''β'' and cholesterol efflux in murine macrophages. Furthermore, based on the molecular docking of E17110 and LXR''β'' ligand-binding domain (LBD) structures, we illustrated the probable interaction mode between LXR''β'' and E17110.
  
 
==2. Materials and methods==
 
==2. Materials and methods==
  
===2.1. Patients and clinical specimens===
+
===2.1. Reagents===
  
Matched fresh osteosarcoma specimens and adjacent nontumorous tissues were acquired from 135 patients at Hanzhong Municipal Central Hospital between January 2006 and December 2010. None of the patients received chemotherapy or radiotherapy before sample collection. All specimens were frozen in liquid nitrogen immediately after collection and stored at −80 °C until use. [[#t0005|Table 1]]  showed the details of clinical and pathological characteristics of the patients. The follow-up data were available and complete for each patient. Overall survival was calculated from the day of primary surgery to death or last follow-up. This project was approved by the Clinical Research Ethics Committee of our hospital and all patients provided written informed consent.
+
The compound E17110 was donated by the National Laboratory for Screening New Microbial Drugs, Peking Union Medical College (PUMC, Beijing, China). TO901317 (also referred as T1317 in this paper), oil red O stain and phorbol-12-myristate-13-acetate (PMA) were purchased from Sigma (St. Louis, MO, USA). HEK293T cells and RAW264.7 macrophages were obtained from the Cell Center of PUMC. Fetal bovine serum (FBS) and Opti-MEM<sup>®</sup> reduced serum medium used for transfection were purchased from Gibco (Invitrogen, Carlsbad, CA, USA). Dulbecco׳s modified Eagle׳s medium (DMEM) was purchased from Hyclone (Thermo Scientific, Rockford, USA). Lipofectamine 2000 and 22-(''N''-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)−23,24-bisnor-5-cholen-3''β''-ol (22-NBD-cholesterol) was purchased from Invitrogen. ApoA-I, HDL and oxidized low-density lipoprotein (ox-LDL) was obtained from Union-Biology Company (Beijing, China).
  
<span id='t0005'></span>
+
===2.2. Plasmids construction and cell culture===
  
{| class="wikitable" style="min-width: 60%;margin-left: auto; margin-right: auto;"
+
The wild-type gene of human LXR''β''-LBD was obtained by PCR from HepG2 cells and cloned into the pBIND vector (Promega, Madison, WI, USA), which included the GAL4 DNA-binding domain (GAL4-DBD) in order to construct the pBIND-LXR''β''-LBD plasmid. The LXR''β''-LBD forward primer was 5′-ATTCGGGATCCCAGCGGCTCAA-3′, and the reverse primer was 5′-TGGGGTACCTCACTCGTGGACGT-3′. GAL4-pGL4-luc plasmid was constructed by inserting the 5×GAL4 response elements into the promoter region of the pGL4.17 reporter vector (Promega) as described previously [[#bib19|<sup>19</sup>]].
|+
+
  
Table 1.
+
Mutations in pBIND-LXR''β''-LBD were created by site-directed mutagenesis using the Fast Mutagenesis System (TransGen Biotech, Beijing, China). Several key amino acids were changed to alanines in LXR''β''-LBD. The mutated plasmids were generated as follows: F271A (Phe271 to Ala) and T316A (Thr316 to Ala).
  
Association of lncRNA-UCA1 expression with clinicopathological factors in osteosarcoma.
+
===2.3. LXR''β''-GAL4 chimera reporter assay and screening===
  
|-
+
In brief, HEK293T cells and RAW264.7 macrophages were cultured in different media separately as described previously[[#bib20|<sup>20</sup>]].
  
! Clinicopathological features
+
A synthetic compound library with 20,000 drug-like structures from the National Laboratory for Screening New Microbial Drugs was used for screening in this study. This compound library was purchased from J&K Chemical (synthesized by Enamine, Kyiv, Ukraine), and all of the compounds in this library are modified based on existing drug structures. These compounds were stocked in 96-well plates at 10 mg/mL in 100% DMSO. HEK293T cells were seeded in 96-well plates at 2×10<sup>4</sup> cells/well in 100 µL DMEM containing 10% FBS. After incubation for 12 h, the cells at 90% confluence were washed once with phosphate-buffered saline (PBS) and transfected with GAL4-pGL4-luc reporter plasmids (180 ng/well) and pBIND-LXR''β''-LBD expression plasmids (18 ng/well) using Lipofectamine 2000 (Invitrogen; 0.5 µL/well). After 6 h, the transfected cells were washed twice with PBS and the buffer replaced with 200 µL DMEM, containing the indicated screening compounds (10 µg/mL of various screening samples, 1 µmol/L TO901317 as a positive control, and 0.1% DMSO as a negative control). For initial screening, each compound was assayed only singly. For rescreening, each initially positive compound was tested in triplicate. For E17110 activity assay the cells were treated with various concentrations of E17110 in serum-free DMEM. After 18 h, the cells were washed with PBS once and lysed with 20 µL 1×CCLR (Promega) per well. The luciferase activity was measured as relative luminescence units (RLUs) in a final volume of 60 µL with the Luciferase Assay System (Promega) on a microplate reader (PerkinElmer, Waltham, MA, USA)[[#bib19|<sup>19</sup>]].
! Number of cases
+
! colspan="2" | lncRNA-UCA1 expression
+
! P
+
|-
+
  
!
+
===2.4. Real-time quantitative RT-PCR analysis===
!
+
! Low n (%)
+
! High n (%)
+
!
+
|-
+
  
! Age
+
RAW264.7 macrophages were seeded in 6-well plates at 6×10<sup>5</sup> cells/mL in DMEM containing 10% FBS. After cell attachment (24 h), compound E17110 was added at various concentrations. The cells were harvested after 18 h, total RNA was extracted from the cells using TRIzol<sup>®</sup> reagent (Invitrogen) according to the manufacturer׳s instructions, and the first-strand cDNA was synthesized from the total RNA in a 20-µL system using a reverse transcriptase kit (TransGen Biotech). Real-time quantitative PCR assay with SYBR Green (Roche Diagnostics, Lewes, UK) detection chemistry was performed on a CFX96™ Real-Time PCR Detection System (Bio-Rad, Hercules, USA). The sequences of the primers are listed in [[#t0005|Table 1]]. Melting curves were recorded, and the specificity of the PCR products was checked by agarose gel analysis. The mRNA levels of all genes were normalized for glyceraldehyde-phosphate dehydrogenase (''GAPDH'') levels, and the quantitative measurements were carried out by the <sup>Δ</sup><sup>Δ</sup>''C''<sub>t</sub> method.
|
+
|
+
|
+
|
+
|-
+
  
! <20 years
+
<span id='t0005'></span>
| 98
+
| 46(46.9%)
+
| 52(53.1%)
+
| 0.339
+
|-
+
  
! ≥20 years
+
{| class="wikitable" style="min-width: 60%;margin-left: auto; margin-right: auto;"
| 37
+
|+
| 21(56.8%)
+
| 16(43.2%)
+
|
+
|-
+
  
!
+
Table 1.
|
+
|
+
|
+
|
+
|-
+
  
! Gender
+
Primers for real-time quantitative PCR.
|
+
|
+
|
+
|
+
|-
+
  
! Male
 
| 95
 
| 49(51.6%)
 
| 46(48.4%)
 
| 0.573
 
 
|-
 
|-
  
! Female
+
! Gene
| 40
+
! Accession No.
| 18(45.0%)
+
! Forward primer
| 22(55.0%)
+
! Reverse primer
|
+
 
|-
 
|-
  
!  
+
! m''GAPDH''
|  
+
| NM_008084.2
|  
+
| 5′-AACGACCCCTTCATTGAC-3′
|  
+
| 5′-TCCACGACATACTCAGCAC-3′
|
+
 
|-
 
|-
  
! Tumor size
+
! m''ABCA1''
|
+
| NM_013454.3
|  
+
| 5′-GTTCCTGCAGAAACAGTAGCA-3′
|  
+
| 5′-ATGAGGTTGGAGATAGCAGAGA-3′
|  
+
 
|-
 
|-
  
! >8 cm
+
! m''ABCG1''
| 57
+
| NM_009593.2
| 20(35.1%)
+
| 5′-AGGTCTCAGCCTTCTAAAGTTCCTC-3′
| 37(64.9%)
+
| 5′-TCTCTCGAAGTGAATGAAATTTATCG-3′
| 0.005
+
|}
|-
+
  
! ≤8 cm
+
===2.5. Western blotting===
| 78
+
| 47(60.3%)
+
| 31(39.7%)
+
|
+
|-
+
  
!
+
RAW264.7 macrophages were seeded on 6-well plates at 6×10<sup>5</sup> cells/mL. The cells were stimulated with different concentrations E17110 for 18 h after which protein extracts were prepared as previously described[[#bib21|<sup>21</sup>]]. Protein concentrations were determined by a BCA protein assay kit (Thermo Scientific). Equal amounts of protein were analyzed by 10% SDS-PAGE gel electrophoresis and electroblotted onto a 0.45 μm polyvinylidene fluoride membranes (Millipore Corp., Bedford, MA, USA). The membranes were blocked with 5% (''w''/''v'') skimmed milk in Tris-buffered saline containing 0.2% Tween-20 (TBST) for 1 h, and then incubated with the following primary antibodies which were diluted in 5% (''w''/''v'') skimmed milk in TBST buffer: mouse anti-ABCA1 (1:1000, Novus, Littleton, CO, USA), anti-ABCG1 (1:1000, Abcam) and anti-''β''-actin (1:2000, Sigma) for 4 °C overnight. The membranes then were washed with TBST three times, followed by incubation with horseradish peroxidase-conjugated secondary antibodies: anti-mouse and anti-rabbit IgG antibodies (1:5000, Novus) for 2 h at temperature. After being washed with TBST three times, the protein bands were detected with an Enhanced Chemiluminescence (ECL) reaction kit (Millipore), and quantified by Quantity One Software (Bio-Rad). All the proteins were normalized to ''β''-actin.
|
+
|
+
|
+
|
+
|-
+
  
! Anatomic location
+
===2.6. Oil red O staining===
|
+
|
+
|
+
|
+
|-
+
  
! Tibia/femur
+
Cellular lipid accumulation was evaluated by means of oil red O staining in RAW264.7 macrophages. The cells were cultured in 96-well plates at 6×10<sup>4</sup> cells/well, and 60 μg/mL ox-LDL was added after cell attachment. After 12 h, when the cells were grown to 90%–95% confluence, they were stimulated with the compound E17110 for 18 h at various concentrations. Then the samples were treated as described in the previous methods, and observed by light microscopy<sup>[[#bib19|19]] and [[#bib22|22]]</sup>. To extract oil red O, isopropanol was added to each well which was then shaken at room temperature for 5 min. Samples were read at 510 nm using a microplate reader[[#bib23|<sup>23</sup>]].
| 93
+
| 48(51.6%)
+
| 45(48.4%)
+
| 0.512
+
|-
+
  
! Elsewhere
+
===2.7. Cholesterol efflux===
| 42
+
| 19(45.2%)
+
| 23(54.8%)
+
|
+
|-
+
  
! colspan="3" |
+
Cellular cholesterol efflux experiments were performed using 22-NBD-cholesterol in RAW264.7 macrophages[[#bib20|<sup>20</sup>]]. The cells were seeded in 96-well clear-bottom black plates (Costar) at 4×10<sup>5</sup> cells/mL. After they attached to the plates the medium was removed and the cells were labeled with 22-NBD-cholesterol (2.0 µmol/L at the final concentration) in serum-free medium containing 0.2% (''w''/''v'') bovine serum albumin (BSA) (Sigma Chemical) (medium A) for 24 h in a 37 °C 5% CO<sub>2</sub> incubator. After 24 h of labeling, cells were washed twice with PBS and incubated with 100 µL medium A containing E17110 (0, 0.3, 1, 3 and 10 µmol/L) for an additional 18 h. 10 µg/mL ApoA-I or 50 µg/mL HDL was added as the receptor protein to start the efflux experiment at 37 °C for 6 h. Then the amounts of cholesterol in medium and cells were assayed using a microplate reader respectively (PerkinElmer, excitation 485 nm, emission 535 nm). The percentage of 22-NBD-cholesterol efflux (%) was calculated as (medium)/(medium+cell)×100. Each efflux assay was performed in duplicate in three times.
|
+
|
+
|-
+
  
! colspan="3" | Tumor grade
+
===2.8. Molecular docking===
|
+
|
+
|-
+
  
! Low
+
To evaluate the activity of E17110, the docking program Discovery Studio 4.1 (Accelrys Inc., CA, USA) was used to dock the structure of LXR''β'' (PDB code: 1PQC, LXR''β'' with TO901317). First, all crystal water molecules were removed from the original structure and hydrogen was added in the DS CDOCKER module. To obtain an optimal starting conformation, the compound was minimized to reach the lowest energy state before docking.
| 45
+
| 30(66.7%)
+
| 15(33.3%)
+
| 0.004
+
|-
+
  
! High
+
===2.9. Statistical analysis===
| 90
+
| 37(41.1%)
+
| 53(58.9%)
+
|
+
|-
+
  
! colspan="3" |
+
Statistics and best-fit curves were calculated with Graphpad Prism 5.0 software (San Diego, CA, USA). The data are expressed as mean±SEM. Results were analyzed by the student׳s ''t''-test and one-way ANOVA analysis by SPSS version 11.0 (SPSS Inc., Chicago, IL, USA). All ''P'' values <0.05 were considered statistically significant (<sup>*</sup>''P''<0.05, <sup>**</sup>''P''<0.01 and <sup>***</sup>''P''<0.001).
|
+
|
+
|-
+
  
! colspan="3" | Histological type
+
==3. Results==
|
+
|
+
|-
+
  
! Osteoblastic
+
===3.1. Cell-based assay optimization===
| 52
+
| 28(53.8%)
+
| 24(46.2%)
+
| 0.774
+
|-
+
  
! Fibroblastic
+
To assess an assay system, the reproducibility and signal variation at the activity range must be evaluated. In our screening system, TO901317 (1 µmol/L) was the positive control and four assay parameters influencing the signal and noise of the cellular reaction were taken into consideration, including: DMSO concentration: 0.1%; ratio between the reporter plasmid and the expression plasmid: 10:1; cell number: 2×10<sup>4</sup> cells/well; and incubation time: 18 h ([[#s0115|Supplementary Fig. 1]]). The signal-to-noise ratio (''S''/''N''), signal-to-background ratio (''S''/''B''), coefficient of variation (CV%) and Z′ factor are classic and scientific indices for evaluation of the quality of assays, and can be utilized in assay validation and optimization [[#bib24|<sup>24</sup>]]. According to our evaluation, this transient transfection system can be used for screening ([[#t0010|Table 2]]).
| 26
+
| 11(42.3%)
+
| 15(57.7%)
+
|  
+
|-
+
  
! Chondroblastic
+
<span id='t0010'></span>
| 20
+
| 9(45.0%)
+
| 11(55.0%)
+
|
+
|-
+
  
! Telangiectatic
+
{| class="wikitable" style="min-width: 60%;margin-left: auto; margin-right: auto;"
| 21
+
|+
| 12(57.1%)
+
| 9(42.9%)
+
|
+
|-
+
  
! Others
+
Table 2.
| 16
+
| 7(43.8%)
+
| 9(56.2%)
+
|
+
|-
+
  
!
+
The parameters of the LXR''β'' screening model.
|
+
|
+
|
+
|
+
|-
+
  
! Enneking stage
 
|
 
|
 
|
 
|
 
 
|-
 
|-
  
! I
+
! Parameter
| 35
+
! LXR''β'' screening model
| 27(77.1%)
+
! High-throughput screening
| 8(22.9%)
+
| <0.001
+
 
|-
 
|-
  
! II
+
! ''S''/''B''
| 66
+
| 43
| 32(48.5%)
+
| >3
| 34(51.5%)
+
|  
+
 
|-
 
|-
  
! III
+
! ''S''/''N''
| 34
+
| 12.13
| 8(23.5%)
+
| >10
| 26(76.5%)
+
|  
+
 
|-
 
|-
  
!  
+
! CV (%)
|  
+
| 4.98
|  
+
| <10
|
+
|
+
 
|-
 
|-
  
! Distant metastasis
+
! ''Z''′ factor
|  
+
| 0.78
|
+
| >0.5
|
+
|
+
|-
+
 
+
! Absent
+
| 101
+
| 58(57.4%)
+
| 43(42.6%)
+
| 0.002
+
|-
+
 
+
! Present
+
| 34
+
| 9(26.5%)
+
| 25(73.5%)
+
|
+
 
|}
 
|}
  
===2.2. Cell culture===
+
===3.2. E17110 has LXRβ agonist activity===
  
The normal osteoblast cell line hFOB1.19 and human osteosarcoma cell lines (HOS, Saos-2, MG-63, U2OS) were purchased from the Chinese Academy of Medical Sciences and cultured in Dulbeccos modified Eagles medium (DMEM, USA) supplemented with 10% fetal bovine serum (FBS, USA), penicillin (100 U/ml), and streptomycin (100 μg/ml). All cells were incubated at 37 °C in 5% CO<sub>2</sub> .    
+
In this study we identified E17110, a structural analog of benzofuran-2-carboxylate ([[#f0005|Fig. 1]]A), with LXR''β'' agonist activity by the LXR''β''-GAL4 luciferase reporter screening as described in Materials and Methods. The chemical name of E17110 is [1-oxo-1-(2-oxoimidazolidin-1-yl)propan-2-yl]5-fluoro-3-methylbenzofuran-2-carboxylate, which has not been reported previously to possess pharmacological activity. E17110 significantly and dose-dependently induced the activation of LXR''β'' from 0.001 μmol/L to 10 μmol/L with an EC50 of 0.72 μmol/L, and showed a maximal activity of approximately 1.76-fold ([[#f0005|Fig. 1]]B). In contrast, TO901317 exhibited approximately 3-fold LXR''β'' activation, with an EC<sub>50</sub> of 0.06 μmol/L ([[#f0005|Fig. 1]]C). TO901317 is regarded as a positive control, therefore this result was consistent with other prior studies, and our cell-based screening model was stable and credible[[#bib22|<sup>22</sup>]].
  
===2.3. RNA extraction and real-time PCR===
+
<span id='f0005'></span>
  
Total RNA was extracted by Trizol reagent (Invitrogen, Carlsbad, CA, USA). The first-strand cDNA was synthesized from 1 μg of total RNA using the Reverse Transcription System Kit (Takara, Dalian, China). The real-time PCR was carried out using SYBR Premix Ex Taq kit (Takara, Dalian, China) on an ABI 7500 Real-Time PCR System (Applied Biosystems, CA, USA). Each assay was performed in triplicate, and GAPDH was used as the endogenous control gene. The primer sequences used were as follows: UCA1, 5′-TTCCTTATTATCTCTTCTG-3′ (forward) and 5′-TCCATCATACGAATAGTA-3′ (reverse); GAPDH, 5′-CTCGCTTTGGCAGCA CA-3′ (forward) and 5′-AACGCTTCACGAATTTGCGT-3′ (reverse). The relative amount of UCA1 to GAPDH was calculated using the 2<sup>–ΔCt</sup>  method, where ΔCT =(CT<sub>UCA1</sub>  - CT<sub>GAPDH</sub> ).     
+
{| style="text-align: center; border: 1px solid #BBB; margin: 1em auto; max-width: 100%;"
 +
|-
 +
|
  
===2.4. UCA1 knockdown and overexpression===
 
  
The cDNA encoding UCA1 was PCR-amplified and subcloned into the pCDNA3.1 vector (Invitrogen, Shanghai, China). The empty pcDNA3.1 vector was used as the control. UCA1 small interfering RNA (si-UCA1) and negative control siRNA (si-NC) were synthesized by GenePharma (Shanghai, China). Transfections were performed using Lipofectamine 2000 according to the manufacturers instructions (Invitrogen, CA, USA).
+
[[Image:Li_et_al_2016a-1-s2.0-S2211383516300053-gr1.jpg|center|527px|Fig. 1]]
  
===2.5. Cell proliferation assay===
 
  
Osteosarcoma cells were seeded into 96-well plates after transfection. Cell density was adjusted to 5×10<sup>3</sup> /well, and the final volume was 150 μl/well. Cell proliferation was determined every 24 h for 4 days. At the indicated time point, 20 μl of MTT (Sigma, USA) was added into each well and the cells were cultured for another 4 h at 37 °C. Then the supernatants were removed and 150 μl of DMSO was added. Optical density was detected at a wavelength of 490 nm and each assay was repeated three times.     
+
|-
 +
| <span style="text-align: center; font-size: 75%;">
  
===2.6. Detection of apoptosis by flow cytometry===
+
Figure 1.
  
Cell apoptosis was measured by using an Annexin V-FITC Apoptosis Detection kit (BD Biosciences, San Diego, CA) according to the manufacturers instructions. After transfection, osteosarcoma cells were harvested and washed twice with cold phosphate-buffered saline. Then, the cells were treated with Annexin V/propidium iodide (PI) for 15 min in the dark at room temperature, and analyzed by flow cytometry assay.
+
E17110 regulates LXR''β''. (A) Structure of E17110. (B) LXR''β'' regulatory activity of E17110. HEK293T cells were transfected with GAL4-pGL4-luc reporter plasmid and pBIND-LXR''β'' expression plasmid. E17110 showed significant LXR''β'' agonistic activity in the luciferase activity assay described in the methods section. (C) LXR''β'' regulatory activity of TO901317. Similar results were obtained in three independent experiments. Data are means±SEM (''n''=3).
  
===2.7. Transwell invasion and migration assays===
+
</span>
 +
|}
  
Cell invasion and migration assays were performed using the 24-well transwell chambers (8 µm pore size; BD Biosciences, San Jose, CA, USA). For the migration assay, about 1×10<sup>5</sup>  osteosarcoma cells were resuspended in 200 μl serum-free medium and placed in the top chambers. DMEM (600 μl) containing 10% FBS was filled into the bottom chambers. After 24 h of incubation at 37 °C, the cells that did not migrate through the pores were removed by a cotton swab, and cells on the lower surface of the membrane were stained with crystal violet and counted. The invasion assay protocol was similar to that of the migration assay except that t the cell culture insert surface was first coated with Matrigel.     
+
===3.3. E17110 induces ABCA1 and ABCG1 expression in vitro===
  
===2.8. Statistics===
+
ABCA1 and ABCG1 are crucial target genes of LXR involved in the RCT process in macrophages[[#bib17|<sup>17</sup>]]. The effects of E17110 on the expression of ABCA1 and ABCG1 in murine macrophages were first detected by western blotting. E17110 significantly increased the protein expression of ABCA1 and ABCG1 in RAW264.7 macrophages ([[#f0010|Fig. 2]]A and B). Furthermore, it up-regulated the ABCA1 and ABCG1 mRNA levels at the same time ([[#f0010|Fig. 2]]C). However, a greater effect was observed when the cells were stimulated with TO901317.
  
All statistical analyses were performed using the SPSS 17.0 software package (SPSS, Chicago, IL, USA). The significance of differences between groups was estimated by Students ''t'' -test and Chi-square test. Survival curves were constructed with the Kaplan-Meier method and compared by log-rank test. The significance of survival variables was evaluated using a multivariate Cox proportional hazards regression analysis. P<0.05 was considered statistically significant.     
+
<span id='f0010'></span>
 
+
==3. Results==
+
 
+
===3.1. Increased UCA1 expression in osteosarcoma tissues and cell lines===
+
 
+
UCA1 expression in osteosarcoma tissues and cell lines was measured by qRT-PCR. We found that UCA1 expression in osteosarcoma samples was significantly higher than in adjacent nontumorous tissues ([[#f0005|Fig. 1]] (A), P<0.001). In addition, UCA1 expression was significantly increased in four osteosarcoma cell lines compared to hFOB cells ([[#f0005|Fig. 1]] (B), P<0.001). Since MG-63 cells exhibited the highest UCA1 expression while HOS cells expressed relatively low UCA1 levels among the four osteosarcoma cell lines, these two cell lines were selected for the transfection of si-UCA1 or pcDNA-UCA1.
+
 
+
<span id='f0005'></span>
+
  
 
{| style="text-align: center; border: 1px solid #BBB; margin: 1em auto; max-width: 100%;"  
 
{| style="text-align: center; border: 1px solid #BBB; margin: 1em auto; max-width: 100%;"  
Line 327: Line 182:
  
  
[[Image:draft_García_537730221-1-s2.0-S2212137416300409-gr1.jpg|center|px|Fig. 1]]
+
[[Image:Li_et_al_2016a-1-s2.0-S2211383516300053-gr2.jpg|center|px|Fig. 2]]
  
  
Line 333: Line 188:
 
| <span style="text-align: center; font-size: 75%;">
 
| <span style="text-align: center; font-size: 75%;">
  
Fig. 1.
+
Figure 2.
  
Expression of lncRNA-UCA1 in osteosarcoma tissues and cell lines. (A) UCA1 expression was significantly higher in osteosarcoma samples than in adjacent nontumorous tissues. (B) Increased UCA1 expression in four osteosarcoma cell lines compared to normal osteoblast cells.
+
Effect of E17110 on ABCA1 and ABCG1 expression. (A and B) RAW264.7 macrophages were incubated with E17110 at various concentrations for 18 h, and the levels of ABCA1 and ABCG1 proteins were determined by western blotting. Induction factors were normalized to ''β''-actin, and the control groups were treated with DMSO (0.1%). (C) RAW264.7 macrophages were treated with E17110 at various concentrations for 18 h. Then mRNAs levels of ABCA1 and ABCG1 were measured by real-time quantitative PCR. Induction factors were normalized to ''GAPDH''. Similar results were obtained in four independent experiments. Data are means±SEM (''n''=4, <sup>*</sup>''P''<0.05, <sup>**</sup>''P''<0.01 and <sup>***</sup>''P''<0.001 ''vs''. control).
  
 
</span>
 
</span>
 
|}
 
|}
  
===3.2. Correlation between UCA1 expression and clinical features and patients prognosis===
+
===3.4. E17110 promotes cholesterol efflux from macrophages===
  
To assess the correlation of UCA1 expression with clinicopathologic characteristics, the osteosarcoma samples were classified into high UCA1 expression group (n=68) and low UCA1 expression group (n=67) according to the median UCA1 expression level of all samples. [[#t0005|Table 1]] showed that high expression of UCA1 was significantly correlated with large tumor size (P =0.005), high tumor grade (P=0.004), positive distant metastasis (P=0.002), and advanced Enneking stage (P<0.001). Kaplan-Meier survival curves indicated that patients in low UCA1 expression group had better 5-year overall survival than those in high UCA1 expression group (P<0.001, [[#f0010|Fig. 2]] ). Univariate Cox proportional hazards analysis revealed that UCA1 expression, tumor size, tumor grade, metastasis status, and clinical stage were prognostic indicators ([[#t0010|Table 2]] ). Multivariate analysis confirmed UCA1 expression (P=0.015) as an independent prognostic indicator for overall survival of osteosarcoma patients in addition to tumor grade (P=0.026), tumor stage (P=0.001), and distant metastasis (P=0.002; [[#t0010|Table 2]] ).
+
LXR activation in macrophages induced the expression of genes encoding ABCA1 and ABCG1, which facilitate cholesterol efflux from macrophages to plasma HDL and ApoA-I[[#bib25|<sup>25</sup>]]. We next determined the effect on the cholesterol efflux in RAW264.7 macrophages. ApoA-I (10 µg/mL) or HDL (50 µg/mL) was added to the medium to promote cholesterol efflux. Obviously, E17110 dose-dependently increased cholesterol efflux to ApoA-I or HDL, and reduced the cellular cholesterol concentration in this cell line ([[#f0015|Fig. 3]]).
  
<span id='f0010'></span>
+
<span id='f0015'></span>
  
 
{| style="text-align: center; border: 1px solid #BBB; margin: 1em auto; max-width: 100%;"  
 
{| style="text-align: center; border: 1px solid #BBB; margin: 1em auto; max-width: 100%;"  
Line 351: Line 206:
  
  
[[Image:draft_García_537730221-1-s2.0-S2212137416300409-gr2.jpg|center|339px|Fig. 2.]]
+
[[Image:Li_et_al_2016a-1-s2.0-S2211383516300053-gr3.jpg|center|509px|Fig. 3]]
  
  
Line 357: Line 212:
 
| <span style="text-align: center; font-size: 75%;">
 
| <span style="text-align: center; font-size: 75%;">
  
Fig. 2.
+
Figure 3.
  
Kaplan-Meier survival curves of osteosarcoma patients based on lncRNA-UCA1 expression levels. Patients in the high UCA1 expression group had significantly poorer prognosis than those in low UCA1 expression group (P<0.001, log-rank test).
+
E17110 induced cholesterol efflux in RAW264.7 macrophages. RAW264.7 macrophages were preincubated with 22-NBD-cholesterol for 24 h, after which the cells were washed with PBS and incubated with E17110 (0, 0.3, 1, 3 and 10 μmol/L). After 18 h, (A) 10 mg/mL ApoA-I or (B) 50 mg/mL HDL was added and the incubation continued for 6 h at 37 °C. The amounts of cholesterol in medium and cell were separately measured. Relative 22-NBD-cholesterol efflux to ApoA-I or HDL induced by E17110 was calculated as described in the Methods section. Similar results were obtained in three independent experiments. Data are means±SEM (''n''=3, <sup>*</sup>''P''<0.05 and <sup>**</sup>''P''<0.01 ''vs.'' control).
  
 
</span>
 
</span>
 
|}
 
|}
  
<span id='t0010'></span>
+
===3.5. E17110 reduces cellular lipid accumulation===
  
{| class="wikitable" style="min-width: 60%;margin-left: auto; margin-right: auto;"
+
Here, we investigated the potential role of E17110 to inhibit lipid accumulation and foam cell formation in RAW264.7 cells, to evaluate whether it could promote lipid fluxing from mouse peritoneal macrophages. As shown in [[#f0020|Fig. 4]]C–E, treatment of RAW264.7 cells with E17110 effectively reduced lipid accumulation compared with ox-LDL alone ([[#f0020|Fig. 4]]B). Furthermore, foam cells were obviously inhibited when stimulated with 10 μmol/L E17110 ([[#f0020|Fig. 4]]E), with levels similar to the vehicle group ([[#f0020|Fig. 4]]A). At the same time, we used a fast and simple method to quantitate the content of lipid in cells. The result showed that E17110 could significantly reduce lipid accumulation from 1 to 10 μmol/L ([[#f0020|Fig. 4]]F).
|+
+
  
Table 2.
+
<span id='f0020'></span>
 
+
Univariate and multivariate analysis of overall survival in 135 osteosarcoma patients.
+
  
 +
{| style="text-align: center; border: 1px solid #BBB; margin: 1em auto; max-width: 100%;"
 
|-
 
|-
 +
|
  
! rowspan="2" | Variable
 
! colspan="2" | Univariate analysis
 
! colspan="2" | Multivariate analysis
 
|-
 
  
! colspan="2" | HR p-value
+
[[Image:Li_et_al_2016a-1-s2.0-S2211383516300053-gr4.jpg|center|565px|Fig. 4]]
! colspan="2" | HR p-value
+
|-
+
  
! Age
 
| 1.528
 
| 0.225
 
| –
 
| –
 
|-
 
  
! Gender
 
| 1.120
 
| 0.773
 
| –
 
| –
 
 
|-
 
|-
 +
| <span style="text-align: center; font-size: 75%;">
  
! Anatomic location
+
Figure 4.
| 1.685
+
| 0.179
+
| –
+
| –
+
|-
+
  
! Tumor grade
+
E17110 reduces ox-LDL-induced lipid accumulation in RAW264.7 macrophages. RAW264.7 macrophages were preincubated with (A) PBS for vehicle and (B)–(E) ox-LDL (60 μg/mL) for samples. After 24 h, these cells were separately treated with (B) DMSO, (C)–(E) E17110 (1, 3 and 10 μmol/L) for 18 h. The cells were fixed with 4% paraformaldehyde and stained with 0.5% oil red O to detect lipid accumulation. Representative images of the five study group samples are shown (×400 magnification). Similar results were obtained in three independent experiments. (F) PBS, ox-LDL (60 μg/mL) and E17110 (1, 3 and 10 μmol/L) were added to the cultures throughout the experiment. After oil red O staining, bound dye was solubilized and quantified spectrophotometrically at 510 nm.
| 3.562
+
| 0.012
+
| 2.872
+
| 0.026
+
|-
+
  
! Tumor size
+
</span>
| 3.101
+
| 0.028
+
| 1.153
+
| 0.089
+
|-
+
 
+
! Clinical stage
+
| 5.969
+
| <0.001
+
| 5.116
+
| 0.001
+
|-
+
 
+
! Distant metastasis
+
| 4.383
+
| 0.003
+
| 4.989
+
| 0.002
+
|-
+
 
+
! lncRNA-UCA1 level
+
| 5.746
+
| <0.001
+
| 3.141
+
| 0.015
+
 
|}
 
|}
  
===3.3. Effects of UCA1 on the biological behaviors of osteosarcoma cells===
+
===3.6. E17110 docks to the LXRβ-LBD===
  
[[#f0015|Fig. 3]] (A) confirmed increased UCA1 expression in HOS cells after pcDNA-UCA1 transfection, and decreased UCA1 expression in MG-63 cells following si-UCA1 transfection. MTT assay showed that cell proliferation was significantly impaired in MG-63 cells transfected with si-UCA1, while proliferation of HOS cells was increased after pcDNA-UCA1 transfection ([[#f0015|Fig. 3]] (B)). Flow cytometry demonstrated promoted cell apoptosis after si-UCA1 transfection and reduced cell apoptosis after pcDNA-UCA1 transfection ([[#f0015|Fig. 3]] (C)). As shown in [[#f0015|Fig. 3]] (D) and (E), down-regulation of UCA1 impeded MG-63 cell invasion and migration, while transfection of HOS cells with pcDNA-UCA1 promoted cell invasion/migration ability.
+
To determine the putative binding mode and potential ligand-pocket interactions of E17110, the structure of E17110 was docked into the ligand-binding domain of LXR''β'' (PDB code: 1PQC) using the docking program DS CDOCKER. The predicted binding mode suggested that E17110 can fit nicely into the LXR''β'' ligand-binding domain ([[#f0025|Fig. 5]]A and B), and included two hydrogen bonds and two ''π''–''π'' stacking interactions with the surrounding amino acids. Specifically, one hydrogen bond formed between the oxygen atom of E17110 and the hydrogen atom of Thr316, and other one formed between the hydrogen atom of E17110 and the oxygen atom of Phe271. Meanwhile, two atypical ''π''–''π'' stacking interactions formed between E17110 and Thr272 and Met312.
  
<span id='f0015'></span>
+
<span id='f0025'></span>
  
 
{| style="text-align: center; border: 1px solid #BBB; margin: 1em auto; max-width: 100%;"  
 
{| style="text-align: center; border: 1px solid #BBB; margin: 1em auto; max-width: 100%;"  
Line 451: Line 254:
  
  
[[Image:draft_García_537730221-1-s2.0-S2212137416300409-gr3.jpg|center|476px|Fig. 3]]
+
[[Image:Li_et_al_2016a-1-s2.0-S2211383516300053-gr5.jpg|center|508px|Fig. 5]]
  
  
Line 457: Line 260:
 
| <span style="text-align: center; font-size: 75%;">
 
| <span style="text-align: center; font-size: 75%;">
  
Fig. 3.
+
Figure 5.
  
Effects of lncRNA-UCA1 on the biological behaviors of osteosarcoma cells. (A) qRT-PCR analysis confirmed increased UCA1 expression in HOS cells transfected with pcDNA-UCA1, and decreased UCA1 expression in MG-63 cells transfected with si-UCA1. **P<0.01. (B) MTT assay showed that lncRNA-UCA1 promoted cell proliferation. **P<0.01. (C) Flow cytometric analysis indicated that lncRNA-UCA1 reduced cell apoptosis. (D, E) Down-regulation of lncRNA-UCA1 impeded the invasion/migration of MG-63 cells, while up-regulation of lncRNA-UCA1 promoted cell invasion/migration ability in HOS cells. **p<0.01.
+
(A and B) The result of E17110 docking into the active site of the ligand-binding domain of LXR''β'' based on the X-ray co-crystal structure of T1317. (C) Activation of various LXR''β'' mutants by E17110, using the LXR''β''-GAL4 chimera reporter assay. (D) E17110 (3 μmol/L) showed different LXR''β'' agonist activity on the wild-type group and different mutants in the LXR''β''-GAL4 chimera reporter assays. Similar results were obtained in three independent experiments. Data are mean±SEM (''n''=3, <sup>*</sup>''P''<0.05 and <sup>**</sup>''P''<0.01 ''vs''. control).
  
 
</span>
 
</span>
 
|}
 
|}
 +
 +
===3.7. E17110 has interaction sites distinct from those of TO901317===
 +
 +
Two different amino acids of LXR''β'' that were shown as crucial residues for binding of E17110 were individually replaced with alanine residues, and the resulting LXR''β'' mutants were assayed for activation by E17110 ( [[#f0025|Fig. 5]]C and D). The F271A and T316A mutants exhibited a significant decrease of agonistic activation by E17110, indicating a crucial role for these amino acid residues in transcriptional activation. Consistent with this finding, compared with the wild-type group, the different mutants showed distinct agonist activity when treated with 3 μmol/L of E17110.
  
 
==4. Discussion==
 
==4. Discussion==
  
It is urgent to develop novel targets for the diagnosis, treatment, and prognosis of osteosarcoma. Increasing evidence has demonstrated that lncRNAs play important roles in almost every aspects of physiological and pathological processes in the human body [[#bib6|[6]]] . Changes of lncRNA expression are involved in cancer formation and progression, which may provide a new but promising way to deal with cancer [[#bib26|[26]]]  and [[#bib27|[27]]] . In the present study, we observed high UCA1 expression in osteosarcoma tissues and cell lines, providing the first evidence that UCA1 upregulation was closely associated with osteosarcoma initiation. Then we confirmed the correlation between increased UCA1 levels and aggressive clinicopathological features of osteosarcoma samples. UCA1 overexpression enhanced HOS cell proliferation, invasion and migration, and inhibited cell apoptosis, whereas UCA1 silencing in MG-63 cells showed opposite effects. These findings revealed that UCA1 might contribute to osteosarcoma progression and serve as a potential therapeutic target. Furthermore, osteosarcoma patients with high UCA1 levels tended to have shorter overall survival compared to patients with low UCA1 levels, and multivariate Cox hazard regression analysis identified UCA1 expression as an independent prognostic indicator. To our knowledge, this is the first study to analyze the expression and clinical significance of UCA1 in osteosarcoma.    
+
In our study, we identified E17110, a benzofuran-2-carboxylate derivative with LXR''β'' agonistic activity with an EC<sub>50</sub> of 0.72 μmol/L. ABCA1 and ABCG1 are major transporters involved in cholesterol efflux from macrophages and play a vital role in maintaining cellular cholesterol homeostasis. Here we demonstrated that in RAW264.7 macrophages, E17110 dose-dependently induced the expression of ABCA1 and ABCG1 proteins and mRNAs. At the same time, we found that E17110 could reduce cellular lipid accumulation in RAW264.7 macrophages. ABCA1 can transfer both cholesterol and phospholipids from plasma membranes to HDL or to lipid-free ApoA-I[[#bib26|<sup>26</sup>]], while ABCG1 only transfers cholesterol to HDL but not to lipid-free ApoA-I[[#bib27|<sup>27</sup>]]. We also performed cholesterol efflux experiments in RAW264.7 macrophages. We found that E17110 significantly increased cholesterol efflux to ApoA-I or HDL, and reduced the cellular cholesterol concentration in a dose-dependent manner. Therefore, we speculated that the cholesterol efflux induced by E17110 was related to the upregulation of ABCA1 and ABCG1 expression ''via'' activation of LXR''β'' in macrophages. This could be of benefit in the prevention of atherosclerosis.
  
Our results are consistent with previous findings in other malignancies. Functional assays revealed that ectopic expression of UCA1 promoted cell proliferation and/or reduced cell apoptosis in non-small cell lung cancer [[#bib19|[19]]] , breast cancer [[#bib18|[18]]] , colorectal cancer [[#bib28|[28]]] , and renal cell carcinoma [[#bib23|[23]]] . Knockdown of UCA1 impaired cell invasion or migration ability in breast cancer [[#bib29|[29]]] , esophageal squamous cell carcinoma [[#bib17|[17]]] , hepatocellular carcinoma[[#bib22|[22]]] , ovarian cancer [[#bib30|[30]]] , and melanoma [[#bib31|[31]]] . UCA1 also decreased chemosensitivity in colorectal cancer[[#bib28|[28]]] , ovarian cancer [[#bib24|[24]]] , and bladder cancer [[#bib32|[32]]] . Clinical investigation demonstrated that UCA1 overexpression was related to lymph node metastasis and advanced FIGO stage in ovarian cancer [[#bib24|[24]]] . Increased UCA1 expression correlated with worse differentiation, large tumor size, deep local invasion, and advanced TNM stage in gastric cancer [[#bib33|[33]]] . Further, high UCA1 expression was an unfavorable prognostic factor for overall survival of patients with esophageal squamous cell carcinoma [[#bib17|[17]]] , gastric cancer [[#bib33|[33]]] , colorectal cancer [[#bib21|[21]]] , hepatocellular carcinoma [[#bib22|[22]]] , and epithelial ovarian cancer [[#bib34|[34]]] . Taken together, these research indicated the oncogenic functions of UCA1 in human cancers and suggested a potential application for UCA1 in molecular targeted therapy.    
+
Molecular docking was carried out to analyze ligand characteristics of E17110. Several potentially crucial amino acid residues were identified from the docking results, and they were replaced with alanine residues by site-directed mutagenesis. Interestingly, we found that the amino acids in LXR''β''-LBD proposed to interact with E17110 differed from those identified for TO901317. Two amino acids (Phe271 and Thr316) formed the most important interaction forces with E17110. In contrast, H435 and W457, which are very important for binding TO901317, did not show significant impact on E17110 binding (data not shown). Thus, we suggest that E17110 has a distinct mechanism for promoting LXR''β'' agonist activity ''in vitro''.
  
We are aware of some limitations in our work. First, the clinical part was a retrospective study, and the tumor sample size was relatively small. Second, although we revealed the oncogene function of UCA1 in osteosarcoma, its probable downstream mediators are still unclear, and the underlying mechanisms by which UCA1 promotes cancer development and progression remain to be thoroughly clarified.
+
LXRs are members of the nuclear receptor superfamily and are present in two isoforms, LXR''α'' and LXR''β''<sup>[[#bib2|2]] and [[#bib28|28]]</sup>. LXRs act as cholesterol sensors that control the expression of target genes when activated by ligands. LXR activation promotes cholesterol efflux and reduces cellular lipid accumulation, to prevent macrophage foam cell formation. Recently, LXRs have been regarded as potential targets for treating atherosclerosis, and synthetic agonists have been the key subject of many studies <sup>[[#bib29|29]] and [[#bib30|30]]</sup>. However, full LXR agonists commonly lead to lipid accumulation in the liver because they activate the LXR''α'' subtype and increase the expression of SREBP-1c regulated genes in the lipogenesis pathway <sup>[[#bib25|25]] and [[#bib31|31]]</sup>. Therefore, in this study, our goal was to find a novel compound targeted to LXR''β'' with potential anti-atherosclerotic activity by screening. LXR''α'' and LXR''β'' have a similar structure in both DBD and LBD domains, so the effect of this new compound on the LXR''α'' subtype still needs to be tested and the possible effects on triglyceride metabolism evaluated.
  
In summary, our study showed increased UCA1 expression in osteosarcoma and its association with aggressive clinicopathological features and poor patients prognosis. Regulation of UCA1 expression influenced biological behaviors of osteosarcoma cells. These findings indicate that UCA1 might be an important molecular marker for prognostic evaluation of osteosarcoma and serve as a potential therapeutic target for this disease.
+
==5. Conclusions==
  
==Conflicts of interest==
+
Overall, through screening we identified E17110, a derivative of benzofuran-2-carboxylate as an anti-atherosclerotic lead compound with potential LXR''β'' agonist activity ''in vitro''. E17110 increased the expression of ABCA1 and ABCG1 dependently on LXR''β'' activation, and promoted cholesterol efflux in macrophages. Meanwhile, E17110 could reduce lipid accumulation and inhibit the foam cell formation. In summary, our study suggests that E17110 may be useful for the development of pharmaceutical agents for treating atherosclerosis.
  
The authors had no conflicts of interest to declare in relation to this article.
+
==Acknowledgments==
  
==Acknowledgement==
+
This work was kindly supported by the National Natural Science Foundation of China (Nos. [[#gs1|81273515]], [[#gs1|81321004]] and [[#gs1|81503065]]), the Key New Drug Creation and Manufacturing Program (Nos. [[#gs2|2012ZX09301002-003]] and [[#gs2|2012ZX09301002-001]]), and the Basic Scientific Research Program of Materia Medica, CAMS ([[#gs3|2014ZD03]]).
  
None.
+
<span id='s0115'></span>
 +
 
 +
==Appendix A. Supplementary material==
 +
 
 +
{| style="text-align: center; border: 1px solid #BBB; margin: 1em auto; max-width: 100%;"
 +
|-
 +
|
 +
 
 +
 
 +
[[Image:Li_et_al_2016a-mmc_zip.gif|center|px|Supplementary Fig. 1 Optimization of the cell-based screening model. (A) The ...]]
 +
 
 +
 
 +
|-
 +
| <span style="text-align: center; font-size: 75%;">
 +
 
 +
Supplementary Fig. 1 Optimization of the cell-based screening model. (A) The sensitivity to DMSO concentration. Cells were seeded at 2×10<sup>4</sup> cells/well and treated with blank control (DMEM only), or serial dilution of DMSO (0.01%, 0.05%, 0.1%, 0.5%, 1%, 5% and 10%). (B) The ratio of reporter plasmid GAL4-pGL4-luc to expression plasmid pBIND-LXR''β''-LBD. The cells were transfected with reporter plasmid to expression plasmid at the ratio of 20:1. 10:1, 5:1, 1:1, 1:5, 1:10 and 1:20 respectively, followed by 18 h treatment of positive control (1 µmol/L TO901317) and negative control (0.1% DMSO). (C) Cell number. HEK293T cells were seeded at 0.5×10<sup>4</sup>, 1×10<sup>4</sup>, 2×10<sup>4</sup>, 5×10<sup>4</sup> and 10×10<sup>4</sup> cells/well respectively, followed by 18 h treatment of positive control (1 µmol/L TO901317) and negative control (0.1% DMSO). (D) Incubation time. The cells were incubated for 6, 12, 18, 24 and 36 h respectively, with positive control (1 µmol/L TO901317) and negative control (0.1% DMSO). Data are means±SEM (<sup>''*''</sup>''P''<0.05, <sup>''**''</sup>''P''<0.01, <sup>''***''</sup>''P''<0.001 ''vs''. control).
 +
 
 +
</span>
 +
|}
  
 
==References==
 
==References==
  
 
<ol style='list-style-type: none;margin-left: 0px;'><li><span id='bib1'></span>
 
<ol style='list-style-type: none;margin-left: 0px;'><li><span id='bib1'></span>
[[#bib1|[1]]] D.S. Geller, R. Gorlick; Osteosarcoma: a review of diagnosis, management, and treatment strategies; Clin. Adv. hematol. Oncol.: HO, 8 (10) (2010), pp. 705–718</li>
+
[[#bib1|1]] G. Wojcicka, A. Jamroz-Wiśniewska, K. Horoszewicz, J. Beltowski; Liver X receptors (LXRs). Part I: structure, function, regulation of activity, and role in lipid metabolism; Postepy Hig Med Dosw, 61 (2007), pp. 736–759</li>
 
<li><span id='bib2'></span>
 
<li><span id='bib2'></span>
[[#bib2|[2]]] S.S. Bielack, B. Kempf-Bielack, G. Delling, G.U. Exner, S. Flege, K. Helmke, R. Kotz, M. Salzer-Kuntschik, M, Werner, W. Winkelmann, A. Zoubek, H. Jurgens, K. Winkler; Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols; J. Clin. Oncol.: Official J. Am. Soc. Clin. Oncol., 20 (3) (2002), pp. 776–790</li>
+
[[#bib2|2]] P.J. Willy, K. Umesono, E.S. Ong, R.M. Evans, R.A. Heyman, D.J. Mangelsdorf; LXR, a nuclear receptor that defines a distinct retinoid response pathway; Genes Dev, 9 (1995), pp. 1033–1045</li>
 
<li><span id='bib3'></span>
 
<li><span id='bib3'></span>
[[#bib3|[3]]] K. Eppert, J.S. Wunder, V. Aneliunas, R. Kandel, I.L. Andrulis; von Willebrand factor expression in osteosarcoma metastasis; Modern Pathol.: an Official J. U. S. Can. Acad. Pathol. Inc, 18 (3) (2005), pp. 388–397</li>
+
[[#bib3|3]] D. Auboeuf, J. Rieusset, L. Fajas, P. Vallier, V. Frering, J.P. Riou, ''et al.''; Tissue distribution and quantification of the expression of mRNAs of peroxisome proliferator-activated receptors and liver X receptor-''α'' in humans: no alteration in adipose tissue of obese and NIDDM patients; Diabetes, 46 (1997), pp. 1319–1327</li>
 
<li><span id='bib4'></span>
 
<li><span id='bib4'></span>
[[#bib4|[4]]] P.A. Meyers; Muramyl tripeptide (mifamurtide) for the treatment of osteosarcoma; Expert Rev. anticancer Ther., 9 (8) (2009), pp. 1035–1049</li>
+
[[#bib4|4]] K. Tamura, Y.E. Chen, M. Horiuchi, Q. Chen, L. Daviet, Z. Yang,  ''et al.''; LXR''α'' functions as a cAMP-responsive transcriptional regulator of gene expression; Proc Natl Acad Sci U S A, 97 (2000), pp. 8513–8518</li>
 
<li><span id='bib5'></span>
 
<li><span id='bib5'></span>
[[#bib5|[5]]] K.C. Wang, H.Y. Chang; Molecular mechanisms of long noncoding RNAs; Mol. Cell, 43 (6) (2011), pp. 904–914</li>
+
[[#bib5|5]] P. Costet, Y. Luo, N. Wang, A.R. Tall; Sterol-dependent transactivation of the ABC1 promoter by the liver X receptor/retinoid X receptor; J Biol Chem, 275 (2000), pp. 28240–28245</li>
 
<li><span id='bib6'></span>
 
<li><span id='bib6'></span>
[[#bib6|[6]]] C. Ernst, C.C. Morton; Identification and function of long non-coding; RNA, Front. Cell. Neurosci., 7 (2013), p. 168</li>
+
[[#bib6|6]] S.L. Sabol, H.B. Brewer Jr., S. Santamarina-Fojo; The human ABCG1 gene: identification of LXR response elements that modulate expression in macrophages and liver; J Lipid Res, 46 (2005), pp. 2151–2167</li>
 
<li><span id='bib7'></span>
 
<li><span id='bib7'></span>
[[#bib7|[7]]] A. Fatica, I. Bozzoni; Long non-coding RNAs: new players in cell differentiation and development; Nat. Rev. Genet., 15 (1) (2014), pp. 7–21</li>
+
[[#bib7|7]] M. Baranowski; Biological role of liver X receptors; J Physiol Pharmacol, 59 (Suppl. 7) (2008), pp. 31–55</li>
 
<li><span id='bib8'></span>
 
<li><span id='bib8'></span>
[[#bib8|[8]]] T. Kunej, J. Obsteter, Z. Pogacar, S. Horvat, G.A. Calin; The decalog of long non-coding RNA involvement in cancer diagnosis and monitoring; Crit. Rev. Clin. Lab. Sci., 51 (6) (2014), pp. 344–357</li>
+
[[#bib8|8]] C. Cavelier, I. Lorenzi, L. Rohrer, A. von Eckardstein; Lipid efflux by the ATP-binding cassette transporters ABCA1 and ABCG1; Biochim Biophys Acta, 1761 (2006), pp. 655–666</li>
 
<li><span id='bib9'></span>
 
<li><span id='bib9'></span>
[[#bib9|[9]]] P.G. Maass, F.C. Luft, S. Bahring; Long non-coding RNA in health and disease; J. Mol. Med., 92 (4) (2014), pp. 337–346</li>
+
[[#bib9|9]] S.U. Naik, X. Wang, J.S. Da Silva, M. Jaye, C.H. Macphee, M.P. Reilly,  ''et al.''; Pharmacological activation of liver X receptors promotes reverse cholesterol transport ''in vivo''; Circulation, 113 (2006), pp. 90–97</li>
 
<li><span id='bib10'></span>
 
<li><span id='bib10'></span>
[[#bib10|[10]]] A. Khandelwal, A. Bacolla, K.M. Vasquez, A. Jain; Long non-coding RNA: a new paradigm for lung cancer; Mol. Carcinog., 54 (11) (2015), pp. 1235–1251</li>
+
[[#bib10|10]] N. Shibata, C.K. Glass; Macrophages, oxysterols and atherosclerosis; Circ J, 74 (2010), pp. 2045–2051</li>
 
<li><span id='bib11'></span>
 
<li><span id='bib11'></span>
[[#bib11|[11]]] M. Cui, L. You, X. Ren, W. Zhao, Q. Liao, Y. Zhao; Long non-coding RNA PVT1 and cancer; Biochem. Biophys. Res. Commun., 471 (1) (2016), pp. 10–14</li>
+
[[#bib11|11]] D.J. Rader, E.T. Alexander, G.L. Weibel, J. Billheimer, G.H. Rothblat; The role of reverse cholesterol transport in animals and humans and relationship to atherosclerosis; J Lipid Res, 50 (Suppl) (2009), pp. S189–S194</li>
 
<li><span id='bib12'></span>
 
<li><span id='bib12'></span>
[[#bib12|[12]]] J. Sun, X. Wang, C. Fu, X. Wang, J. Zou, H. Hua, Z. Bi; Long noncoding RNA FGFR3-AS1 promotes osteosarcoma growth through regulating its natural antisense transcript FGFR3; Mol. Biol. Rep. (2016)</li>
+
[[#bib12|12]] S.B. Joseph, E. McKilligin, L. Pei, M.A. Watson, A.R. Collins, B.A. Laffitte, ''et al.''; Synthetic LXR ligand inhibits the development of atherosclerosis in mice; Proc Natl Acad Sci U S A, 99 (2002), pp. 7604–7609</li>
 
<li><span id='bib13'></span>
 
<li><span id='bib13'></span>
[[#bib13|[13]]] B. Ma, M. Li, L. Zhang, M. Huang, J.B. Lei, G.H. Fu, C.X. Liu, Q.W. Lai, Q.Q. Chen, Y.L. Wang; Upregulation of Long non-coding RNA TUG1 Correlates with poor prognosis and Disease Status in Osteosarcoma; Tumour Biol.: J. Int. Soc. Oncodevelopmental Biol. Med. (2015)</li>
+
[[#bib13|13]] N. Levin, E.D. Bischoff, C.L. Daige, D. Thomas, C.T. Vu, R.A. Heyman, ''et al.''; Macrophage liver X receptor is required for antiatherogenic activity of LXR agonists; Arterioscler Thromb Vasc Biol, 25 (2005), pp. 135–142</li>
 
<li><span id='bib14'></span>
 
<li><span id='bib14'></span>
[[#bib14|[14]]] M. Cong, J. Li, R. Jing, Z. Li; Long non-coding RNA tumor suppressor Candidate 7 Functions as a tumor suppressor and Inhibits proliferation in Osteosarcoma; Tumour Biol.: J. Int. Soc. Oncodevelopmental Biol. Med. (2016)</li>
+
[[#bib14|14]] R.K. Tangirala, E.D. Bischoff, S.B. Joseph, B.L. Wagner, R. Walczak, B.A. Laffitte,  ''et al.''; Identification of macrophage liver X receptors as inhibitors of atherosclerosis; Proc Natl Acad Sci U S A, 99 (2002), pp. 11896–11901</li>
 
<li><span id='bib15'></span>
 
<li><span id='bib15'></span>
[[#bib15|[15]]] C.L. Zhang, K.P. Zhu, G.Q. Shen, Z.S. Zhu; A Long non-coding RNA Contributes to Doxorubicin Resistance of Osteosarcoma; Tumour Biol.: J. Int. Soc. Oncodevelopmental Biol. Med. (2015)</li>
+
[[#bib15|15]] B.A. Janowski, P.J. Willy, T.R. Devi, J.R. Falck, D.J. Mangelsdorf; An oxysterol signalling pathway mediated by the nuclear receptor LXR''α''; Nature, 383 (1996), pp. 728–731</li>
 
<li><span id='bib16'></span>
 
<li><span id='bib16'></span>
[[#bib16|[16]]] F. Wang, X. Li, X. Xie, L. Zhao, W. Chen; UCA1, a non-protein-coding RNA up-regulated in bladder carcinoma and embryo, influencing cell growth and promoting invasion; FEBS Lett., 582 (13) (2008), pp. 1919–1927</li>
+
[[#bib16|16]] K.A. Houck, K.M. Borchert, C.D. Hepler, J.S. Thomas, K.S. Bramlett, L.F. Michael, ''et al.''; T0901317 is a dual LXR/FXR agonist; Mol Genet Metab, 83 (2004), pp. 184–187</li>
 
<li><span id='bib17'></span>
 
<li><span id='bib17'></span>
[[#bib17|[17]]] J.Y. Li, X. Ma, C.B. Zhang; Overexpression of long non-coding RNA UCA1 predicts a poor prognosis in patients with esophageal squamous cell carcinoma; Int. J. Clin. Exp. Pathol., 7 (11) (2014), pp. 7938–7944</li>
+
[[#bib17|17]] R. Geyeregger, M. Zeyda, T.M. Stulnig; Liver X receptors in cardiovascular and metabolic disease; Cell Mol Life Sci, 63 (2006), pp. 524–539</li>
 
<li><span id='bib18'></span>
 
<li><span id='bib18'></span>
[[#bib18|[18]]] Y.L. Tuo, X.M. Li, J. Luo; Long noncoding RNA UCA1 modulates breast cancer cell growth and apoptosis through decreasing tumor suppressive miR-143; Eur. Rev. Med. Pharmacol. Sci., 19 (18) (2015), pp. 3403–3411</li>
+
[[#bib18|18]] D.J. Peet, S.D. Turley, W.Z. Ma, B.A. Janowski, J.M. Lobaccaro, R.E. Hammer,  ''et al.''; Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR''α''; Cell, 93 (1998), pp. 693–704</li>
 
<li><span id='bib19'></span>
 
<li><span id='bib19'></span>
[[#bib19|[19]]] W. Nie, H.J. Ge, X.Q. Yang, X. Sun, H. Huang, X. Tao, W.S. Chen, B. Li; LncRNA-UCA1 exerts oncogenic functions in non-small cell lung cancer by targeting miR-193a-3p; Cancer Lett., 371 (1) (2016), pp. 99–106</li>
+
[[#bib19|19]] N. Li, Y. Xu, T. Feng, C. Liu, Y. Li, X. Wang, ''et al.''; Identification of a selective agonist for liver X receptor ''α'' (LXR''α'') ''via'' screening of a synthetic compound library; J Biomol Screen, 19 (2014), pp. 566–574</li>
 
<li><span id='bib20'></span>
 
<li><span id='bib20'></span>
[[#bib20|[20]]] J. Gao, R. Cao, H. Mu; Long non-coding RNA UCA1 may be a novel diagnostic and predictive biomarker in plasma for early gastric cancer; Int. J. Clin. Exp. Pathol., 8 (10) (2015)</li>
+
[[#bib20|20]] N. Li, X. Wang, J. Zhang, C. Liu, Y.Z. Li, T.T. Feng, ''et al.''; Identification of a novel partial agonist of liver X receptor ''α'' (LXR''α'') ''via'' screening; Biochem Pharmacol, 92 (2014), pp. 438–447</li>
 
<li><span id='bib21'></span>
 
<li><span id='bib21'></span>
[[#bib21|[21]]] Y. Han, Y.N. Yang, H.H. Yuan, T.T. Zhang, H. Sui, X.L. Wei, L. Liu, P. Huang, W.J. Zhang, Y.X. Bai; UCA1, a long non-coding RNA up-regulated in colorectal cancer influences cell proliferation, apoptosis and cell cycle distribution; Pathology, 46 (5) (2014), pp. 396–401</li>
+
[[#bib21|21]] A. Amoruso, C. Bardelli, G. Gunella, F. Ribichini, S. Brunelleschi; A novel activity for substance P: stimulation of peroxisome proliferator-activated receptor-''γ'' protein expression in human monocytes and macrophages; Br J Pharmacol, 154 (2008), pp. 144–152</li>
 
<li><span id='bib22'></span>
 
<li><span id='bib22'></span>
[[#bib22|[22]]] F. Wang, H.Q. Ying, B.S. He, Y.Q. Pan, Q.W. Deng, H.L. Sun, J. Chen, X. Liu, S.K. Wang; Upregulated lncRNA-UCA1 contributes to progression of hepatocellular carcinoma through inhibition of miR-216b and activation of FGFR1/ERK signaling pathway; Oncotarget, 6 (10) (2015), pp. 7899–7917</li>
+
[[#bib22|22]] M.H. Hoang, Y.Y. Jia, H.J. Jun, J.H. Lee, B.Y. Lee, S.J. Lee; Fucosterol is a selective liver X receptor modulator that regulates the expression of key genes in cholesterol homeostasis in macrophages, hepatocytes, and intestinal cells; J Agric Food Chem, 60 (2012), pp. 11567–11575</li>
 
<li><span id='bib23'></span>
 
<li><span id='bib23'></span>
[[#bib23|[23]]] Y. Li, T. Wang, Y. Li, D. Chen, Z. Yu, L. Jin, L. Ni, S. Yang, X. Mao, Y. Gui, Y. Lai; Identification of Long-non coding RNA UCA1 as an Oncogene in Renal Cell Carcinoma; Mol. Med. Rep. (2016)</li>
+
[[#bib23|23]] C.H. Zou, Z.F. Shen; One-step intracellular triglycerides extraction and quantitative measurement ''in vitro''; J Pharmacol Toxicol Methods, 56 (2007), pp. 63–66</li>
 
<li><span id='bib24'></span>
 
<li><span id='bib24'></span>
[[#bib24|[24]]] L. Zhang, X. Cao, L. Zhang, X. Zhang, H. Sheng, K. Tao; UCA1 overexpression predicts clinical outcome of patients with ovarian cancer receiving adjuvant chemotherapy; Cancer Chemother. Pharmacol., 77 (3) (2016), pp. 629–634</li>
+
[[#bib24|24]] J.H. Zhang, T.D. Chung, K.R. Oldenburg; A simple statistical parameter for use in evaluation and validation of high throughput screening assays; J Biomol Screen, 4 (1999), pp. 67–73</li>
 
<li><span id='bib25'></span>
 
<li><span id='bib25'></span>
[[#bib25|[25]]] X.Y. Na, Z.Y. Liu, P.P. Ren, R. Yu, X.S. Shang; Long non-coding RNA UCA1 contributes to the progression of prostate cancer and regulates proliferation through KLF4-KRT6/13 signaling pathway; Int. J. Clin. Exp. Med., 8 (8) (2015), pp. 12609–12616</li>
+
[[#bib25|25]] J.J. Repa, D.J. Mangelsdorf; The role of orphan nuclear receptors in the regulation of cholesterol homeostasis; Annu Rev Cell Dev Biol, 16 (2000), pp. 459–481</li>
 
<li><span id='bib26'></span>
 
<li><span id='bib26'></span>
[[#bib26|[26]]] I. Dickson; Hepatocellular carcinoma: A role for lncRNA in liver cancer; Nat. Rev. Gastroenterol. Hepatol., 13 (3) (2016), pp. 122–123</li>
+
[[#bib26|26]] M. Dean, Y. Hamon, G. Chimini; The human ATP-binding cassette (ABC) transporter superfamily; J Lipid Res, 42 (2001), pp. 1007–1017</li>
 
<li><span id='bib27'></span>
 
<li><span id='bib27'></span>
[[#bib27|[27]]] M. Sun, F.Q. Nie, Z.X. Wang; W. De, Involvement of lncRNA dysregulation in gastric cancer; Histol. Histopathol., 31 (1) (2016), pp. 33–39</li>
+
[[#bib27|27]] M.A. Kennedy, G.C. Barrera, K. Nakamura, A. Baldan, P. Tarr, M.C. Fishbein,  ''et al.''; ABCG1 has a critical role in mediating cholesterol efflux to HDL and preventing cellular lipid accumulation; Cell Metab, 1 (2005), pp. 121–131</li>
 
<li><span id='bib28'></span>
 
<li><span id='bib28'></span>
[[#bib28|[28]]] Z. Bian, L. Jin, J. Zhang, Y. Yin, C. Quan, Y. Hu, Y. Feng, H. Liu, B. Fei, Y. Mao, L. Zhou, X. Qi, S. Huang, D. Hua, C. Xing, Z. Huang; LncRNA-UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting miR-204–5p; Sci. Rep., 6 (2016), p. 23892</li>
+
[[#bib28|28]] K.S. Miyata, S.E. McCaw, H.V. Patel, R.A. Rachubinski, J.P. Capone; The orphan nuclear hormone receptor LXR''α'' interacts with the peroxisome proliferator-activated receptor and inhibits peroxisome proliferator signaling; J Biol Chem, 271 (1996), pp. 9189–9192</li>
 
<li><span id='bib29'></span>
 
<li><span id='bib29'></span>
[[#bib29|[29]]] S. Chen, C. Shao, M. Xu, J. Ji, Y. Xie, Y. Lei, X. Wang; Macrophage infiltration promotes invasiveness of breast cancer cells via activating long non-coding RNA UCA1; Int. J. Clin. Exp. Pathol., 8 (8) (2015), pp. 9052–9061</li>
+
[[#bib29|29]] C. Huang; Natural modulators of liver X receptors; J Integr Med, 12 (2014), pp. 76–85</li>
 
<li><span id='bib30'></span>
 
<li><span id='bib30'></span>
[[#bib30|[30]]] F. Wang, J. Zhou, X. Xie, J. Hu, L. Chen, Q. Hu, H. Guo, C. Yu; Involvement of SRPK1 in cisplatin resistance related to long non-coding RNA UCA1 in human ovarian cancer cells; Neoplasma (2015)</li>
+
[[#bib30|30]] C. Hong, P. Tontonoz; Liver X receptors in lipid metabolism: opportunities for drug discovery; Nat Rev Drug Discov, 13 (2014), pp. 433–444</li>
 
<li><span id='bib31'></span>
 
<li><span id='bib31'></span>
[[#bib31|[31]]] Y. Tian, X. Zhang, Y. Hao, Z. Fang, Y. He; Potential roles of abnormally expressed long noncoding RNA UCA1 and Malat-1 in metastasis of melanoma; Melanoma Res., 24 (4) (2014), pp. 335–341</li>
+
[[#bib31|31]] E.M. Quinet, D.A. Savio, A.R. Halpern, L. Chen, G.U. Schuster, J.A. Gustafsson, ''et al.''; Liver X receptor (LXR)-''β'' regulation in LXR''α''-deficient mice: implications for therapeutic targeting; Mol Pharmacol, 70 (2006), pp. 1340–1349</li>
<li><span id='bib32'></span>
+
[[#bib32|[32]]] Y. Fan, B. Shen, M. Tan, X. Mu, Y. Qin, F. Zhang, Y. Liu; Long non-coding RNA UCA1 increases chemoresistance of bladder cancer cells by regulating Wnt signaling; FEBS J., 281 (7) (2014), pp. 1750–1758</li>
+
<li><span id='bib33'></span>
+
[[#bib33|[33]]] Q. Zheng, F. Wu, W.Y. Dai, D.C. Zheng, C. Zheng, H. Ye, B. Zhou, J.J. Chen, P. Chen; Aberrant expression of UCA1 in gastric cancer and its clinical significance, Clinical and translational oncology: official publication of the Federation of Spanish; Oncol. Soc. Natl. Cancer Inst. Mex., 17 (8) (2015), pp. 640–646</li>
+
<li><span id='bib34'></span>
+
[[#bib34|[34]]] Y. Yang, Y. Jiang, Y. Wan, L. Zhang, J. Qiu, S. Zhou, W. Cheng; UCA1 Functions as a competing Endogenous RNA to Suppress epithelial Ovarian Cancer Metastasis; Tumour Biol.: J. Int. Soc. Oncodev. Biol. Med. (2016)</li>
+
 
</ol>
 
</ol>

Latest revision as of 14:34, 17 October 2016

Abstract

Liver X receptor (LXR) plays an important role in reverse cholesterol transport (RCT), and activation of LXR could reduce atherosclerosis. In the present study we used a cell-based screening method to identify new potential LXRβ agonists. A novel benzofuran-2-carboxylate derivative was identified with LXRβ agonist activity: E17110 showed a significant activation effect on LXRβ with an EC50 value of 0.72 μmol/L. E17110 also increased the expression of ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) in RAW264.7 macrophages. Moreover, E17110 significantly reduced cellular lipid accumulation and promoted cholesterol efflux in RAW264.7 macrophages. Interestingly, we found that the key amino acids in the LXRβ ligand-binding domain had distinct interactions with E17110 as compared to TO901317. These results suggest that E17110 was identified as a novel compound with LXRβ agonist activity in vitro via screening, and could be developed as a potential anti-atherosclerotic lead compound.

Graphical abstract

E17110 was identified as a novel LXRβ agonist by using a cell-based screening method. E17110 could increase the expression of ABCA1 and ABCG1 in RAW264.7 macrophages and significantly reduce cellular lipid accumulation and promote cholesterol efflux. Interestingly, we found that LXRβ had distinct interactions with E17110 compared to TO901317.


fx1

Abbreviations

ABCA1, ATP-binding cassette transporter A1; ABCG1, ATP-binding cassette transporter G1; ApoA-I, apolipoprotein A-I; GAPDH, glyceraldehyde-phosphate dehydrogenase; HDL, high-density lipoprotein; LBD, ligand-binding domain; LXR, liver X receptor; LXRE, LXR response element; NR, nuclear receptor; ox-LDL, oxidized low-density lipoprotein; RCT, reverse cholesterol transport; RXR, retinoid X receptor

Key words

LXRβ; Atherosclerosis; ABCA1; ABCG1; Reverse cholesterol transport; Cholesterol efflux

1. Introduction

The liver X receptors (LXRα and LXRβ) are ligand-activated transcription factors that belong to the nuclear receptor (NR) superfamily 1 and 2. LXRβ (NR1H2) is ubiquitously expressed at a moderate level in most physiological systems, whereas LXRα (NR1H3) is mainly expressed in the intestine, kidney, spleen and adipose tissue, especially in the liver 3. LXRs generally function as permissive heterodimers with retinoid X receptor (RXR) that bind to specific response elements in the regulatory region of their target genes to regulate their expression4. LXRs sense excess cholesterol and trigger various adaptive mechanisms to protect the cells from cholesterol overload. ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) are regulated by LXRs via functional LXR response elements (LXREs) found in their genes, which play important roles in cholesterol efflux 5, 6 and 7. ABCA1 can transfer both cholesterol and phospholipids to lipid-free apolipoprotein A-I (apoA-I), and ABCG1 can transfer cholesterol to high-density lipoprotein (HDL) 7 and 8.

Excessive absorption of lipoproteins in macrophages causes foam cell formation within arterial walls, and these cells subsequently rupture and promote early atherosclerotic plaque formation9 and 10. The efflux of excess cellular cholesterol from peripheral tissues and its return to the liver for excretion in the bile occurs by a process referred to as reverse cholesterol transport (RCT)11. Furthermore, RCT is regarded as a major mechanism that removes cholesterol from the cells and transports it to the liver in order to protect against atherosclerotic cardiovascular disease, and this process can be stimulated by LXRs11.

Previous studies showed that treatment of atherosclerotic mice with synthetic LXR ligands effectively inhibited progression and promoted regression of atherosclerotic plaques12 and 13. Meanwhile, macrophage-specific deletion of LXR in mice enhances atherogenesis14. Several LXR ligands, such as endogenous ligand 22(R)-hydroxycholesterol and synthetic agonists TO901317 and GW3965, have recently been reported and investigated substantially 15, 16 and 17. However, these ligands have the undesirable side effect of inducing lipogenesis and hypertriglyceridemia because of their up-regulation of sterol response element binding protein 1c (SREBP-1c) transcription18. Therefore, the identification of novel LXRβ agonists which could achieve beneficial effects from regulating cholesterol metabolism is necessary.

In this study, we discovered E17110 as a novel benzofuran-2-carboxylate derivative with potential LXRβ agonist activity using an LXRβ-GAL4 chimera reporter assay. We then investigated the effect and mechanism of this compound on the target genes of LXRβ and cholesterol efflux in murine macrophages. Furthermore, based on the molecular docking of E17110 and LXRβ ligand-binding domain (LBD) structures, we illustrated the probable interaction mode between LXRβ and E17110.

2. Materials and methods

2.1. Reagents

The compound E17110 was donated by the National Laboratory for Screening New Microbial Drugs, Peking Union Medical College (PUMC, Beijing, China). TO901317 (also referred as T1317 in this paper), oil red O stain and phorbol-12-myristate-13-acetate (PMA) were purchased from Sigma (St. Louis, MO, USA). HEK293T cells and RAW264.7 macrophages were obtained from the Cell Center of PUMC. Fetal bovine serum (FBS) and Opti-MEM® reduced serum medium used for transfection were purchased from Gibco (Invitrogen, Carlsbad, CA, USA). Dulbecco׳s modified Eagle׳s medium (DMEM) was purchased from Hyclone (Thermo Scientific, Rockford, USA). Lipofectamine 2000 and 22-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)−23,24-bisnor-5-cholen-3β-ol (22-NBD-cholesterol) was purchased from Invitrogen. ApoA-I, HDL and oxidized low-density lipoprotein (ox-LDL) was obtained from Union-Biology Company (Beijing, China).

2.2. Plasmids construction and cell culture

The wild-type gene of human LXRβ-LBD was obtained by PCR from HepG2 cells and cloned into the pBIND vector (Promega, Madison, WI, USA), which included the GAL4 DNA-binding domain (GAL4-DBD) in order to construct the pBIND-LXRβ-LBD plasmid. The LXRβ-LBD forward primer was 5′-ATTCGGGATCCCAGCGGCTCAA-3′, and the reverse primer was 5′-TGGGGTACCTCACTCGTGGACGT-3′. GAL4-pGL4-luc plasmid was constructed by inserting the 5×GAL4 response elements into the promoter region of the pGL4.17 reporter vector (Promega) as described previously 19.

Mutations in pBIND-LXRβ-LBD were created by site-directed mutagenesis using the Fast Mutagenesis System (TransGen Biotech, Beijing, China). Several key amino acids were changed to alanines in LXRβ-LBD. The mutated plasmids were generated as follows: F271A (Phe271 to Ala) and T316A (Thr316 to Ala).

2.3. LXRβ-GAL4 chimera reporter assay and screening

In brief, HEK293T cells and RAW264.7 macrophages were cultured in different media separately as described previously20.

A synthetic compound library with 20,000 drug-like structures from the National Laboratory for Screening New Microbial Drugs was used for screening in this study. This compound library was purchased from J&K Chemical (synthesized by Enamine, Kyiv, Ukraine), and all of the compounds in this library are modified based on existing drug structures. These compounds were stocked in 96-well plates at 10 mg/mL in 100% DMSO. HEK293T cells were seeded in 96-well plates at 2×104 cells/well in 100 µL DMEM containing 10% FBS. After incubation for 12 h, the cells at 90% confluence were washed once with phosphate-buffered saline (PBS) and transfected with GAL4-pGL4-luc reporter plasmids (180 ng/well) and pBIND-LXRβ-LBD expression plasmids (18 ng/well) using Lipofectamine 2000 (Invitrogen; 0.5 µL/well). After 6 h, the transfected cells were washed twice with PBS and the buffer replaced with 200 µL DMEM, containing the indicated screening compounds (10 µg/mL of various screening samples, 1 µmol/L TO901317 as a positive control, and 0.1% DMSO as a negative control). For initial screening, each compound was assayed only singly. For rescreening, each initially positive compound was tested in triplicate. For E17110 activity assay the cells were treated with various concentrations of E17110 in serum-free DMEM. After 18 h, the cells were washed with PBS once and lysed with 20 µL 1×CCLR (Promega) per well. The luciferase activity was measured as relative luminescence units (RLUs) in a final volume of 60 µL with the Luciferase Assay System (Promega) on a microplate reader (PerkinElmer, Waltham, MA, USA)19.

2.4. Real-time quantitative RT-PCR analysis

RAW264.7 macrophages were seeded in 6-well plates at 6×105 cells/mL in DMEM containing 10% FBS. After cell attachment (24 h), compound E17110 was added at various concentrations. The cells were harvested after 18 h, total RNA was extracted from the cells using TRIzol® reagent (Invitrogen) according to the manufacturer׳s instructions, and the first-strand cDNA was synthesized from the total RNA in a 20-µL system using a reverse transcriptase kit (TransGen Biotech). Real-time quantitative PCR assay with SYBR Green (Roche Diagnostics, Lewes, UK) detection chemistry was performed on a CFX96™ Real-Time PCR Detection System (Bio-Rad, Hercules, USA). The sequences of the primers are listed in Table 1. Melting curves were recorded, and the specificity of the PCR products was checked by agarose gel analysis. The mRNA levels of all genes were normalized for glyceraldehyde-phosphate dehydrogenase (GAPDH) levels, and the quantitative measurements were carried out by the ΔΔCt method.

Table 1. Primers for real-time quantitative PCR.
Gene Accession No. Forward primer Reverse primer
mGAPDH NM_008084.2 5′-AACGACCCCTTCATTGAC-3′ 5′-TCCACGACATACTCAGCAC-3′
mABCA1 NM_013454.3 5′-GTTCCTGCAGAAACAGTAGCA-3′ 5′-ATGAGGTTGGAGATAGCAGAGA-3′
mABCG1 NM_009593.2 5′-AGGTCTCAGCCTTCTAAAGTTCCTC-3′ 5′-TCTCTCGAAGTGAATGAAATTTATCG-3′

2.5. Western blotting

RAW264.7 macrophages were seeded on 6-well plates at 6×105 cells/mL. The cells were stimulated with different concentrations E17110 for 18 h after which protein extracts were prepared as previously described21. Protein concentrations were determined by a BCA protein assay kit (Thermo Scientific). Equal amounts of protein were analyzed by 10% SDS-PAGE gel electrophoresis and electroblotted onto a 0.45 μm polyvinylidene fluoride membranes (Millipore Corp., Bedford, MA, USA). The membranes were blocked with 5% (w/v) skimmed milk in Tris-buffered saline containing 0.2% Tween-20 (TBST) for 1 h, and then incubated with the following primary antibodies which were diluted in 5% (w/v) skimmed milk in TBST buffer: mouse anti-ABCA1 (1:1000, Novus, Littleton, CO, USA), anti-ABCG1 (1:1000, Abcam) and anti-β-actin (1:2000, Sigma) for 4 °C overnight. The membranes then were washed with TBST three times, followed by incubation with horseradish peroxidase-conjugated secondary antibodies: anti-mouse and anti-rabbit IgG antibodies (1:5000, Novus) for 2 h at temperature. After being washed with TBST three times, the protein bands were detected with an Enhanced Chemiluminescence (ECL) reaction kit (Millipore), and quantified by Quantity One Software (Bio-Rad). All the proteins were normalized to β-actin.

2.6. Oil red O staining

Cellular lipid accumulation was evaluated by means of oil red O staining in RAW264.7 macrophages. The cells were cultured in 96-well plates at 6×104 cells/well, and 60 μg/mL ox-LDL was added after cell attachment. After 12 h, when the cells were grown to 90%–95% confluence, they were stimulated with the compound E17110 for 18 h at various concentrations. Then the samples were treated as described in the previous methods, and observed by light microscopy19 and 22. To extract oil red O, isopropanol was added to each well which was then shaken at room temperature for 5 min. Samples were read at 510 nm using a microplate reader23.

2.7. Cholesterol efflux

Cellular cholesterol efflux experiments were performed using 22-NBD-cholesterol in RAW264.7 macrophages20. The cells were seeded in 96-well clear-bottom black plates (Costar) at 4×105 cells/mL. After they attached to the plates the medium was removed and the cells were labeled with 22-NBD-cholesterol (2.0 µmol/L at the final concentration) in serum-free medium containing 0.2% (w/v) bovine serum albumin (BSA) (Sigma Chemical) (medium A) for 24 h in a 37 °C 5% CO2 incubator. After 24 h of labeling, cells were washed twice with PBS and incubated with 100 µL medium A containing E17110 (0, 0.3, 1, 3 and 10 µmol/L) for an additional 18 h. 10 µg/mL ApoA-I or 50 µg/mL HDL was added as the receptor protein to start the efflux experiment at 37 °C for 6 h. Then the amounts of cholesterol in medium and cells were assayed using a microplate reader respectively (PerkinElmer, excitation 485 nm, emission 535 nm). The percentage of 22-NBD-cholesterol efflux (%) was calculated as (medium)/(medium+cell)×100. Each efflux assay was performed in duplicate in three times.

2.8. Molecular docking

To evaluate the activity of E17110, the docking program Discovery Studio 4.1 (Accelrys Inc., CA, USA) was used to dock the structure of LXRβ (PDB code: 1PQC, LXRβ with TO901317). First, all crystal water molecules were removed from the original structure and hydrogen was added in the DS CDOCKER module. To obtain an optimal starting conformation, the compound was minimized to reach the lowest energy state before docking.

2.9. Statistical analysis

Statistics and best-fit curves were calculated with Graphpad Prism 5.0 software (San Diego, CA, USA). The data are expressed as mean±SEM. Results were analyzed by the student׳s t-test and one-way ANOVA analysis by SPSS version 11.0 (SPSS Inc., Chicago, IL, USA). All P values <0.05 were considered statistically significant (*P<0.05, **P<0.01 and ***P<0.001).

3. Results

3.1. Cell-based assay optimization

To assess an assay system, the reproducibility and signal variation at the activity range must be evaluated. In our screening system, TO901317 (1 µmol/L) was the positive control and four assay parameters influencing the signal and noise of the cellular reaction were taken into consideration, including: DMSO concentration: 0.1%; ratio between the reporter plasmid and the expression plasmid: 10:1; cell number: 2×104 cells/well; and incubation time: 18 h (Supplementary Fig. 1). The signal-to-noise ratio (S/N), signal-to-background ratio (S/B), coefficient of variation (CV%) and Z′ factor are classic and scientific indices for evaluation of the quality of assays, and can be utilized in assay validation and optimization 24. According to our evaluation, this transient transfection system can be used for screening (Table 2).

Table 2. The parameters of the LXRβ screening model.
Parameter LXRβ screening model High-throughput screening
S/B 43 >3
S/N 12.13 >10
CV (%) 4.98 <10
Z′ factor 0.78 >0.5

3.2. E17110 has LXRβ agonist activity

In this study we identified E17110, a structural analog of benzofuran-2-carboxylate (Fig. 1A), with LXRβ agonist activity by the LXRβ-GAL4 luciferase reporter screening as described in Materials and Methods. The chemical name of E17110 is [1-oxo-1-(2-oxoimidazolidin-1-yl)propan-2-yl]5-fluoro-3-methylbenzofuran-2-carboxylate, which has not been reported previously to possess pharmacological activity. E17110 significantly and dose-dependently induced the activation of LXRβ from 0.001 μmol/L to 10 μmol/L with an EC50 of 0.72 μmol/L, and showed a maximal activity of approximately 1.76-fold (Fig. 1B). In contrast, TO901317 exhibited approximately 3-fold LXRβ activation, with an EC50 of 0.06 μmol/L (Fig. 1C). TO901317 is regarded as a positive control, therefore this result was consistent with other prior studies, and our cell-based screening model was stable and credible22.


Fig. 1


Figure 1.

E17110 regulates LXRβ. (A) Structure of E17110. (B) LXRβ regulatory activity of E17110. HEK293T cells were transfected with GAL4-pGL4-luc reporter plasmid and pBIND-LXRβ expression plasmid. E17110 showed significant LXRβ agonistic activity in the luciferase activity assay described in the methods section. (C) LXRβ regulatory activity of TO901317. Similar results were obtained in three independent experiments. Data are means±SEM (n=3).

3.3. E17110 induces ABCA1 and ABCG1 expression in vitro

ABCA1 and ABCG1 are crucial target genes of LXR involved in the RCT process in macrophages17. The effects of E17110 on the expression of ABCA1 and ABCG1 in murine macrophages were first detected by western blotting. E17110 significantly increased the protein expression of ABCA1 and ABCG1 in RAW264.7 macrophages (Fig. 2A and B). Furthermore, it up-regulated the ABCA1 and ABCG1 mRNA levels at the same time (Fig. 2C). However, a greater effect was observed when the cells were stimulated with TO901317.


Fig. 2


Figure 2.

Effect of E17110 on ABCA1 and ABCG1 expression. (A and B) RAW264.7 macrophages were incubated with E17110 at various concentrations for 18 h, and the levels of ABCA1 and ABCG1 proteins were determined by western blotting. Induction factors were normalized to β-actin, and the control groups were treated with DMSO (0.1%). (C) RAW264.7 macrophages were treated with E17110 at various concentrations for 18 h. Then mRNAs levels of ABCA1 and ABCG1 were measured by real-time quantitative PCR. Induction factors were normalized to GAPDH. Similar results were obtained in four independent experiments. Data are means±SEM (n=4, *P<0.05, **P<0.01 and ***P<0.001 vs. control).

3.4. E17110 promotes cholesterol efflux from macrophages

LXR activation in macrophages induced the expression of genes encoding ABCA1 and ABCG1, which facilitate cholesterol efflux from macrophages to plasma HDL and ApoA-I25. We next determined the effect on the cholesterol efflux in RAW264.7 macrophages. ApoA-I (10 µg/mL) or HDL (50 µg/mL) was added to the medium to promote cholesterol efflux. Obviously, E17110 dose-dependently increased cholesterol efflux to ApoA-I or HDL, and reduced the cellular cholesterol concentration in this cell line (Fig. 3).


Fig. 3


Figure 3.

E17110 induced cholesterol efflux in RAW264.7 macrophages. RAW264.7 macrophages were preincubated with 22-NBD-cholesterol for 24 h, after which the cells were washed with PBS and incubated with E17110 (0, 0.3, 1, 3 and 10 μmol/L). After 18 h, (A) 10 mg/mL ApoA-I or (B) 50 mg/mL HDL was added and the incubation continued for 6 h at 37 °C. The amounts of cholesterol in medium and cell were separately measured. Relative 22-NBD-cholesterol efflux to ApoA-I or HDL induced by E17110 was calculated as described in the Methods section. Similar results were obtained in three independent experiments. Data are means±SEM (n=3, *P<0.05 and **P<0.01 vs. control).

3.5. E17110 reduces cellular lipid accumulation

Here, we investigated the potential role of E17110 to inhibit lipid accumulation and foam cell formation in RAW264.7 cells, to evaluate whether it could promote lipid fluxing from mouse peritoneal macrophages. As shown in Fig. 4C–E, treatment of RAW264.7 cells with E17110 effectively reduced lipid accumulation compared with ox-LDL alone (Fig. 4B). Furthermore, foam cells were obviously inhibited when stimulated with 10 μmol/L E17110 (Fig. 4E), with levels similar to the vehicle group (Fig. 4A). At the same time, we used a fast and simple method to quantitate the content of lipid in cells. The result showed that E17110 could significantly reduce lipid accumulation from 1 to 10 μmol/L (Fig. 4F).


Fig. 4


Figure 4.

E17110 reduces ox-LDL-induced lipid accumulation in RAW264.7 macrophages. RAW264.7 macrophages were preincubated with (A) PBS for vehicle and (B)–(E) ox-LDL (60 μg/mL) for samples. After 24 h, these cells were separately treated with (B) DMSO, (C)–(E) E17110 (1, 3 and 10 μmol/L) for 18 h. The cells were fixed with 4% paraformaldehyde and stained with 0.5% oil red O to detect lipid accumulation. Representative images of the five study group samples are shown (×400 magnification). Similar results were obtained in three independent experiments. (F) PBS, ox-LDL (60 μg/mL) and E17110 (1, 3 and 10 μmol/L) were added to the cultures throughout the experiment. After oil red O staining, bound dye was solubilized and quantified spectrophotometrically at 510 nm.

3.6. E17110 docks to the LXRβ-LBD

To determine the putative binding mode and potential ligand-pocket interactions of E17110, the structure of E17110 was docked into the ligand-binding domain of LXRβ (PDB code: 1PQC) using the docking program DS CDOCKER. The predicted binding mode suggested that E17110 can fit nicely into the LXRβ ligand-binding domain (Fig. 5A and B), and included two hydrogen bonds and two ππ stacking interactions with the surrounding amino acids. Specifically, one hydrogen bond formed between the oxygen atom of E17110 and the hydrogen atom of Thr316, and other one formed between the hydrogen atom of E17110 and the oxygen atom of Phe271. Meanwhile, two atypical ππ stacking interactions formed between E17110 and Thr272 and Met312.


Fig. 5


Figure 5.

(A and B) The result of E17110 docking into the active site of the ligand-binding domain of LXRβ based on the X-ray co-crystal structure of T1317. (C) Activation of various LXRβ mutants by E17110, using the LXRβ-GAL4 chimera reporter assay. (D) E17110 (3 μmol/L) showed different LXRβ agonist activity on the wild-type group and different mutants in the LXRβ-GAL4 chimera reporter assays. Similar results were obtained in three independent experiments. Data are mean±SEM (n=3, *P<0.05 and **P<0.01 vs. control).

3.7. E17110 has interaction sites distinct from those of TO901317

Two different amino acids of LXRβ that were shown as crucial residues for binding of E17110 were individually replaced with alanine residues, and the resulting LXRβ mutants were assayed for activation by E17110 ( Fig. 5C and D). The F271A and T316A mutants exhibited a significant decrease of agonistic activation by E17110, indicating a crucial role for these amino acid residues in transcriptional activation. Consistent with this finding, compared with the wild-type group, the different mutants showed distinct agonist activity when treated with 3 μmol/L of E17110.

4. Discussion

In our study, we identified E17110, a benzofuran-2-carboxylate derivative with LXRβ agonistic activity with an EC50 of 0.72 μmol/L. ABCA1 and ABCG1 are major transporters involved in cholesterol efflux from macrophages and play a vital role in maintaining cellular cholesterol homeostasis. Here we demonstrated that in RAW264.7 macrophages, E17110 dose-dependently induced the expression of ABCA1 and ABCG1 proteins and mRNAs. At the same time, we found that E17110 could reduce cellular lipid accumulation in RAW264.7 macrophages. ABCA1 can transfer both cholesterol and phospholipids from plasma membranes to HDL or to lipid-free ApoA-I26, while ABCG1 only transfers cholesterol to HDL but not to lipid-free ApoA-I27. We also performed cholesterol efflux experiments in RAW264.7 macrophages. We found that E17110 significantly increased cholesterol efflux to ApoA-I or HDL, and reduced the cellular cholesterol concentration in a dose-dependent manner. Therefore, we speculated that the cholesterol efflux induced by E17110 was related to the upregulation of ABCA1 and ABCG1 expression via activation of LXRβ in macrophages. This could be of benefit in the prevention of atherosclerosis.

Molecular docking was carried out to analyze ligand characteristics of E17110. Several potentially crucial amino acid residues were identified from the docking results, and they were replaced with alanine residues by site-directed mutagenesis. Interestingly, we found that the amino acids in LXRβ-LBD proposed to interact with E17110 differed from those identified for TO901317. Two amino acids (Phe271 and Thr316) formed the most important interaction forces with E17110. In contrast, H435 and W457, which are very important for binding TO901317, did not show significant impact on E17110 binding (data not shown). Thus, we suggest that E17110 has a distinct mechanism for promoting LXRβ agonist activity in vitro.

LXRs are members of the nuclear receptor superfamily and are present in two isoforms, LXRα and LXRβ2 and 28. LXRs act as cholesterol sensors that control the expression of target genes when activated by ligands. LXR activation promotes cholesterol efflux and reduces cellular lipid accumulation, to prevent macrophage foam cell formation. Recently, LXRs have been regarded as potential targets for treating atherosclerosis, and synthetic agonists have been the key subject of many studies 29 and 30. However, full LXR agonists commonly lead to lipid accumulation in the liver because they activate the LXRα subtype and increase the expression of SREBP-1c regulated genes in the lipogenesis pathway 25 and 31. Therefore, in this study, our goal was to find a novel compound targeted to LXRβ with potential anti-atherosclerotic activity by screening. LXRα and LXRβ have a similar structure in both DBD and LBD domains, so the effect of this new compound on the LXRα subtype still needs to be tested and the possible effects on triglyceride metabolism evaluated.

5. Conclusions

Overall, through screening we identified E17110, a derivative of benzofuran-2-carboxylate as an anti-atherosclerotic lead compound with potential LXRβ agonist activity in vitro. E17110 increased the expression of ABCA1 and ABCG1 dependently on LXRβ activation, and promoted cholesterol efflux in macrophages. Meanwhile, E17110 could reduce lipid accumulation and inhibit the foam cell formation. In summary, our study suggests that E17110 may be useful for the development of pharmaceutical agents for treating atherosclerosis.

Acknowledgments

This work was kindly supported by the National Natural Science Foundation of China (Nos. 81273515, 81321004 and 81503065), the Key New Drug Creation and Manufacturing Program (Nos. 2012ZX09301002-003 and 2012ZX09301002-001), and the Basic Scientific Research Program of Materia Medica, CAMS (2014ZD03).

Appendix A. Supplementary material


Supplementary Fig. 1 Optimization of the cell-based screening model. (A) The ...


Supplementary Fig. 1 Optimization of the cell-based screening model. (A) The sensitivity to DMSO concentration. Cells were seeded at 2×104 cells/well and treated with blank control (DMEM only), or serial dilution of DMSO (0.01%, 0.05%, 0.1%, 0.5%, 1%, 5% and 10%). (B) The ratio of reporter plasmid GAL4-pGL4-luc to expression plasmid pBIND-LXRβ-LBD. The cells were transfected with reporter plasmid to expression plasmid at the ratio of 20:1. 10:1, 5:1, 1:1, 1:5, 1:10 and 1:20 respectively, followed by 18 h treatment of positive control (1 µmol/L TO901317) and negative control (0.1% DMSO). (C) Cell number. HEK293T cells were seeded at 0.5×104, 1×104, 2×104, 5×104 and 10×104 cells/well respectively, followed by 18 h treatment of positive control (1 µmol/L TO901317) and negative control (0.1% DMSO). (D) Incubation time. The cells were incubated for 6, 12, 18, 24 and 36 h respectively, with positive control (1 µmol/L TO901317) and negative control (0.1% DMSO). Data are means±SEM (*P<0.05, **P<0.01, ***P<0.001 vs. control).

References

  1. 1 G. Wojcicka, A. Jamroz-Wiśniewska, K. Horoszewicz, J. Beltowski; Liver X receptors (LXRs). Part I: structure, function, regulation of activity, and role in lipid metabolism; Postepy Hig Med Dosw, 61 (2007), pp. 736–759
  2. 2 P.J. Willy, K. Umesono, E.S. Ong, R.M. Evans, R.A. Heyman, D.J. Mangelsdorf; LXR, a nuclear receptor that defines a distinct retinoid response pathway; Genes Dev, 9 (1995), pp. 1033–1045
  3. 3 D. Auboeuf, J. Rieusset, L. Fajas, P. Vallier, V. Frering, J.P. Riou, et al.; Tissue distribution and quantification of the expression of mRNAs of peroxisome proliferator-activated receptors and liver X receptor-α in humans: no alteration in adipose tissue of obese and NIDDM patients; Diabetes, 46 (1997), pp. 1319–1327
  4. 4 K. Tamura, Y.E. Chen, M. Horiuchi, Q. Chen, L. Daviet, Z. Yang, et al.; LXRα functions as a cAMP-responsive transcriptional regulator of gene expression; Proc Natl Acad Sci U S A, 97 (2000), pp. 8513–8518
  5. 5 P. Costet, Y. Luo, N. Wang, A.R. Tall; Sterol-dependent transactivation of the ABC1 promoter by the liver X receptor/retinoid X receptor; J Biol Chem, 275 (2000), pp. 28240–28245
  6. 6 S.L. Sabol, H.B. Brewer Jr., S. Santamarina-Fojo; The human ABCG1 gene: identification of LXR response elements that modulate expression in macrophages and liver; J Lipid Res, 46 (2005), pp. 2151–2167
  7. 7 M. Baranowski; Biological role of liver X receptors; J Physiol Pharmacol, 59 (Suppl. 7) (2008), pp. 31–55
  8. 8 C. Cavelier, I. Lorenzi, L. Rohrer, A. von Eckardstein; Lipid efflux by the ATP-binding cassette transporters ABCA1 and ABCG1; Biochim Biophys Acta, 1761 (2006), pp. 655–666
  9. 9 S.U. Naik, X. Wang, J.S. Da Silva, M. Jaye, C.H. Macphee, M.P. Reilly, et al.; Pharmacological activation of liver X receptors promotes reverse cholesterol transport in vivo; Circulation, 113 (2006), pp. 90–97
  10. 10 N. Shibata, C.K. Glass; Macrophages, oxysterols and atherosclerosis; Circ J, 74 (2010), pp. 2045–2051
  11. 11 D.J. Rader, E.T. Alexander, G.L. Weibel, J. Billheimer, G.H. Rothblat; The role of reverse cholesterol transport in animals and humans and relationship to atherosclerosis; J Lipid Res, 50 (Suppl) (2009), pp. S189–S194
  12. 12 S.B. Joseph, E. McKilligin, L. Pei, M.A. Watson, A.R. Collins, B.A. Laffitte, et al.; Synthetic LXR ligand inhibits the development of atherosclerosis in mice; Proc Natl Acad Sci U S A, 99 (2002), pp. 7604–7609
  13. 13 N. Levin, E.D. Bischoff, C.L. Daige, D. Thomas, C.T. Vu, R.A. Heyman, et al.; Macrophage liver X receptor is required for antiatherogenic activity of LXR agonists; Arterioscler Thromb Vasc Biol, 25 (2005), pp. 135–142
  14. 14 R.K. Tangirala, E.D. Bischoff, S.B. Joseph, B.L. Wagner, R. Walczak, B.A. Laffitte, et al.; Identification of macrophage liver X receptors as inhibitors of atherosclerosis; Proc Natl Acad Sci U S A, 99 (2002), pp. 11896–11901
  15. 15 B.A. Janowski, P.J. Willy, T.R. Devi, J.R. Falck, D.J. Mangelsdorf; An oxysterol signalling pathway mediated by the nuclear receptor LXRα; Nature, 383 (1996), pp. 728–731
  16. 16 K.A. Houck, K.M. Borchert, C.D. Hepler, J.S. Thomas, K.S. Bramlett, L.F. Michael, et al.; T0901317 is a dual LXR/FXR agonist; Mol Genet Metab, 83 (2004), pp. 184–187
  17. 17 R. Geyeregger, M. Zeyda, T.M. Stulnig; Liver X receptors in cardiovascular and metabolic disease; Cell Mol Life Sci, 63 (2006), pp. 524–539
  18. 18 D.J. Peet, S.D. Turley, W.Z. Ma, B.A. Janowski, J.M. Lobaccaro, R.E. Hammer, et al.; Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXRα; Cell, 93 (1998), pp. 693–704
  19. 19 N. Li, Y. Xu, T. Feng, C. Liu, Y. Li, X. Wang, et al.; Identification of a selective agonist for liver X receptor α (LXRα) via screening of a synthetic compound library; J Biomol Screen, 19 (2014), pp. 566–574
  20. 20 N. Li, X. Wang, J. Zhang, C. Liu, Y.Z. Li, T.T. Feng, et al.; Identification of a novel partial agonist of liver X receptor α (LXRα) via screening; Biochem Pharmacol, 92 (2014), pp. 438–447
  21. 21 A. Amoruso, C. Bardelli, G. Gunella, F. Ribichini, S. Brunelleschi; A novel activity for substance P: stimulation of peroxisome proliferator-activated receptor-γ protein expression in human monocytes and macrophages; Br J Pharmacol, 154 (2008), pp. 144–152
  22. 22 M.H. Hoang, Y.Y. Jia, H.J. Jun, J.H. Lee, B.Y. Lee, S.J. Lee; Fucosterol is a selective liver X receptor modulator that regulates the expression of key genes in cholesterol homeostasis in macrophages, hepatocytes, and intestinal cells; J Agric Food Chem, 60 (2012), pp. 11567–11575
  23. 23 C.H. Zou, Z.F. Shen; One-step intracellular triglycerides extraction and quantitative measurement in vitro; J Pharmacol Toxicol Methods, 56 (2007), pp. 63–66
  24. 24 J.H. Zhang, T.D. Chung, K.R. Oldenburg; A simple statistical parameter for use in evaluation and validation of high throughput screening assays; J Biomol Screen, 4 (1999), pp. 67–73
  25. 25 J.J. Repa, D.J. Mangelsdorf; The role of orphan nuclear receptors in the regulation of cholesterol homeostasis; Annu Rev Cell Dev Biol, 16 (2000), pp. 459–481
  26. 26 M. Dean, Y. Hamon, G. Chimini; The human ATP-binding cassette (ABC) transporter superfamily; J Lipid Res, 42 (2001), pp. 1007–1017
  27. 27 M.A. Kennedy, G.C. Barrera, K. Nakamura, A. Baldan, P. Tarr, M.C. Fishbein, et al.; ABCG1 has a critical role in mediating cholesterol efflux to HDL and preventing cellular lipid accumulation; Cell Metab, 1 (2005), pp. 121–131
  28. 28 K.S. Miyata, S.E. McCaw, H.V. Patel, R.A. Rachubinski, J.P. Capone; The orphan nuclear hormone receptor LXRα interacts with the peroxisome proliferator-activated receptor and inhibits peroxisome proliferator signaling; J Biol Chem, 271 (1996), pp. 9189–9192
  29. 29 C. Huang; Natural modulators of liver X receptors; J Integr Med, 12 (2014), pp. 76–85
  30. 30 C. Hong, P. Tontonoz; Liver X receptors in lipid metabolism: opportunities for drug discovery; Nat Rev Drug Discov, 13 (2014), pp. 433–444
  31. 31 E.M. Quinet, D.A. Savio, A.R. Halpern, L. Chen, G.U. Schuster, J.A. Gustafsson, et al.; Liver X receptor (LXR)-β regulation in LXRα-deficient mice: implications for therapeutic targeting; Mol Pharmacol, 70 (2006), pp. 1340–1349
Back to Top

Document information

Published on 17/10/16

Licence: Other

Document Score

0

Views 0
Recommendations 0

Share this document

claim authorship

Are you one of the authors of this document?