Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01.12.2018 | Research

RETRACTED ARTICLE: LncGPR107 drives the self-renewal of liver tumor initiating cells and liver tumorigenesis through GPR107-dependent manner

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2018

Abstract

Background

With self-renewal and differentiation properties, liver tumor initiating cells (TICs) are the reasons for tumor initiation, metastasis and drug resistance. G protein coupled receptors (GPCR) are critical modulators in many physiological and pathological processes. While, their roles in liver TICs are unknown.

Methods

An unbiased screening was performed using online-available data dataset. Liver TICs were sorted by FACS with surface marker CD133, or enriched by oncosphere formation. TIC self-renewal was examined by oncosphere formation and tumor initiation assay. Loss of function and gain of function assays were performed to examine the role of lncRNA. RNA pulldown, RNA immunoprecipitation, ChIP, western blot and double FISH were used explore the molecular mechanism of lncRNA.

Results

We performed an unbiased screening for GPCR expression in liver cancers, and found GPR107 was the most highly expressed GPCR in liver cancer and liver TICs. GPR107 was essential for the self-renewal of liver TICs. The expression of GPR107 was regulated by a long noncoding RNA lncGPR107. LncGPR107 was also highly expressed in liver cancers and liver TICs. LncGPR107 drove the self-renewal of liver TICs through GPR107. Moreover, lncGPR107 recruited SRCAP complex to GPR107 promoter to drive its transcriptional activation. LncGPR107 depletion inhibited the binding of SRCAP complex and GPR107 promoter and subsequent GPR107 expression. Moreover, LncGPR107-SRCAP-GPR107 can be targeted for liver TIC elimination.

Conclusion

GPR107 was the most highly expressed GPCR in liver cancer and liver TICs. LncGPR107 participated in the transcriptional regulation of GPR107 in cis, through recruiting SRCAP remodeling complex to GPR107 promoter. This work revealed the important role of GPCR signaling in liver TIC self-renewal and added a new layer for liver TIC and GPCR regulation.
Hinweise
Guanqun Huang and Hui Jiang contributed equally to this work.
A Retraction Note to this article is available online at https://​doi.​org/​10.​1186/​s13046-022-02414-1.
This article has been retracted. Please see the retraction notice for more detail:https://​doi.​org/​10.​1186/​s13046-022-02414-1
Abkürzungen
ASO
Antisense oligo
ChIP
Chromatin immunoprecipitation
EMSA
Electrophoretic mobility shift assay
GPCR
G protein coupled receptor
HCC
Hepatocellular carcinoma
lncRNA
long noncoding RNA
TIC
Tumor initiating cell

Background

Liver tumor initiating cells (TICs), also termed as liver cancer stem cells (CSCs), are the reason of liver tumorigenesis, metastasis, drug resistance and relapse [1]. Because these cells are in quiescent state and highly express many drug pumps (including ABCG2), liver TICs can survive during drug treatment, and initiate new tumors once drugs are removed [2]. Liver TICs have self-renewal and differentiate capacities, and can form many cell types along with tumorigenesis [3]. Based these specific characteristics, several functional systems have been developed for liver TICs, including sphere formation, side population, colony formation, transwell assay and tumor initiation assay [46]. Recent years, various surface markers of liver TICs were found, including CD13, CD133, EPCAM, CD24 and CD90 [710]. Some transcription factors were also found to participate in the self-renewal regulation of liver TICs, including Sox4, Zic2 and Oct4 [6, 11]. Although the importance of liver TICs is well-known, the self-renewal regulation of liver TICs are still unclear.
The activation of liver TICs is finely regulated. Wnt/β-catenin, Notch, Hedgehog signaling pathways play critical roles in liver TICs [12, 13]. G protein coupled receptors (GPCR) are critical modulators in signal transduction [14]. GPCRs play critical roles in many disease, including tumorigenesis, conferring GPCRs the most widely-used drug-targets [15]. While, the exact roles of GPCRs in liver TIC self-renewal are still elusive. Here we performed an unbiased screening and found GPR107 was highly expressed in liver cancer and liver TICs. GPR107 plays an essential role in the internalization of pseudomonas aeruginosa exotoxin A and can interacts with neuronostatin [16, 17]. The role of GPR107 in tumorigenesis hasn’t been reported. Here, we found GPR107 was required for the self-renewal of liver TICs and liver tumorigenesis.
Long noncoding RNAs (LncRNAs) are RNA molecules longer than 200 nt but have no coding potential [18]. LncRNAs were considered as byproductions of transcription process. Recently, the biological roles of lncRNAs are emerging. LncRNAs play critical roles in immunological regulation [19], development [20], tumorigenesis [21] and so on. LncRNAs exert their roles through binding with remodeling complex and recruiting them to the promoter of target genes, and also can interact with proteins to modulate their stabilities or activities [22]. According to the relative location between lncRNAs and their target genes, two kinds of function pattern of lncRNAs have been discovered: in cis and in trans [23]. As one of the common mechanisms, many lncRNAs regulate the transcription activity of nearby genes through recruiting chromatin remodeling complex [24]. Here we found a lncRNA was located near from GPR107 locus, and required for the transcription initiation of GPR107.

Methods

Reagents

Anti-β-actin (cat. no. A1978) and DAPI (cat. no. 28718–90-3) were from Sigma-Aldrich. Anti-GPR107 (25076–1-AP) antibody was from Proteintech Company. Anti-SRCAP (cat. no. PAB22228) antibody was from abnova. Phycoerythrin (PE)-conjugated CD133 (cat. no. 130098826) was from Miltenyi Biotec. The Chemiluminescent Nucleic Acid Detection Module (cat. no. 89880) and LightShift Chemiluminescent RNA EMSA kit (cat. no. 20158) were from Thermo Scientific. Biotin RNA Labeling Mix (cat. no. 11685597910) and T7 RNA polymerase (cat. no. 10881767001) were from Roche Life Science.

Sphere formation

For liver TIC sphere formation assay, 5000 HCC cells were cultured in sphere formation medium (1 × B27, 1 × N2, 20 ng/ml EGF, 20 ng/ml bFGF, in DMEM/F12) and seeded into Ultra Low Attachment six-well plates (cat. no. 3471, Corning). Two weeks later, typical images were taken and sphere numbers were counted. Sphere initiating ratios (%) = Sphere numbers/5000 × 100%.

Primary cell isolation and culture

Primary HCC cells were obtained from hepatocellular carcinoma patients, and the information of HCC samples used in this research was listed in Table 1. For primary cell isolation, a portion of excised tumor was incubated in Hanks balanced salt solution (HBSS; Gibco) and transported to the laboratory on ice. Then the samples were cut into small fragments, and digested in HBSS containing 0.03% pronase, 0.05% type IV collagenase, and 0.01% deoxyribonuclease for 30 min at 37 °C. The sample was filtered through 100-μm-nylon filter and centrifuged for 2 min at 50 x g in 4 °C. HCC primary cells were in precipitation, and cell survival & purification were examined. For sphere formation, 5000 primary cells were seeded into sphere formation medium and cultured in Ultra Low Attachment six-well plate. For liver TIC enrichment, HCC primary cells were incubated with CD133 antibody for FACS.
Table 1
Characteristics of HCC samples
Samples
Sample 1
Sample 2
Sample 3
Sample 4
Sample 5
Age
61
72
53
74
63
Sex
Female
Male
Male
Male
Female
HBsAg
+
+
+
+
+
Serum α-fetoprotein
5.8 ng/ml
2182
3168
439
1216
Tumor bulk used (mm)
7.2 × 6.3 × 5.2
5.8 × 4.2 × 3.9
9.2 × 8.3 × 7.5
7.9 × 5.3 × 4.8
6.9 × 6.1 × 4.2
Cirrhosis
No
No
Yes
No
Yes
Vascular invasion
No
No
Yes
Yes
No
Stages
T1
T2
T3
T3
T2
Sphere formation
+
+
+
+
CD133+ cells
+
+
+
+

Liver TIC sorting

For liver TIC sorting, primary cells were incubated in PBS at 5000 cells/μl, supplementary with 2% FBS. Then Phycoerythrin (PE)-conjugated anti-CD133 antibody or control IgG were added (1:100). The samples were placed in 4 °C for 30 min, and then liver TICs were isolated by FACS. BD FACSAria was used for sorting and CD133+ cells were liver TICs.

Fluorescence in situ hybridization (FISH)

For FISH assay, fluorescence-conjugated lncGPR107 probes were generated according to the manual of Biosearch Technologies. HCC samples, liver TICs or oncospheres were treated in a non-denaturing condition, followed by hybridization with lncGPR107 probes. After RNA hybridization, then corresponding antibodies (anti-CD133, Zic2, SRCAP) or fluorescence-conjugated antibodies were added for protein observation. Finally samples were incubated in 4′,6-diamidino-2-phenylindole (DAPI, 1:1000) for 5 min’ counterstain, and observed by confocal microscopy (FV1000, Olympus). All experiments were performed according to manuals of Biosearch Technologies.

RNA pulldown

For RNA pulldown assays, biotin-labeled lncGPR107 or fragments were obtained by in vitro transcription and biotin RNA labeling mix (Roche). Then the labeled RNAs were incubated with liver oncosphere lysate for six hours. Streptavidin beads were added for another three hours’ incubation. The samples were washed for three times (five minutes once), and then incubated in loading buffer and boiled at 100 °C for 10 min. Finally the samples were subjected into SDS–PAGE. For protein identification, specific bands enriched by lncGPR107 were identified by mass spectrometry (LTQ Orbitrap XL). For Western blot, the samples in SDS-PAGE were transferred onto NC membrane and detected by antibodies.

Chromatin immunoprecipitation (ChIP)

ChIP assays were completed according to Upstate Biotechnology. Briefly, lncGPR107 silenced or lncGPR107 overexpressing spheres were fixed with 1% formaldehyde for 15 min’ crosslink, and crushed in SDS lysis buffer. The samples were then treated by ultrasonic for shearing DNA. SRCAP, H3K4Me3, RNA polymerase II or control IgG antibodies were added into samples for 6 h’ incubation. Then beads containing sonicated salmon sperm (Upstate Biotechnology) were added into samples for DNA enrichment. DNA segment enrichment of GPR107 promoter was examined by real-time PCR.

ASO treatment

For HCC propagation, primary HCC cells were injected into BALB/c nude mice for tumor propagation. Six weeks later, tumors were obtained for another round tumor propagation (if necessary) until the cells were enough for experiments in 18 mice. For each mice, 1 × 106 cells were used for subcutaneous injection. When tumors were reached to 300 mm3 (about three weeks), mice were randomly assigned into three groups (six mice per group). LncGPR107 ASOs or control ASO were injected into tumors every 4 days. Three weeks after ASO intratumoral injection, tumors were obtained, weighted and used for further experiments. GPR107 antibody was used for immunohistochemistry, Ki67 and CD133 antibodies were used for FACS.

GPCR expression analysis

For gene expression analysis, GSE14520 was selected because it contained a large number of HCC samples with detailed clinical information, and download form NCBI database. R language and Bioconductor methods were utilized for background correction, normalization, calculation and annotation. Gene lists and expression levels generated by R3.1.0 were used for further analyses. For G protein coupled receptors (GPCRs) analysis, GPCRs list was download from DAVID website. GPCRs were listed according to expression changes in liver tumorigenesis. The expression levels in every samples were normalized to the average expression of peri-tumor samples, and shown as heatmap.

Tumor propagation and initiating assay

For tumor propagation, 1 × 106 lncGPR107 silenced and control cells were subcutaneously injected into 6-week-old BALB/c nude mice. One month later, tumors were selected and tumor weight was examined. For tumor initiation, 10, 1 × 102, 1 × 103, 1 × 104 and 1 × 105 lncGPR107 silenced and control cells were subcutaneously injected into 6-week-old BALB/c nude mice for 3 months’ tumor formation. Tumor formation was observed for tumor-free ratios. Tumor initiating ratios were calculated by Extreme Limiting Dilution Analysis (ELDA) (http://​bioinf.​wehi.​edu.​au/​software/​elda/​) [25].

Immunohistochemistry

For immunohistochemistry, formalin-fixed liver cancer sections were treated with xylene and graded alcohols (100, 90% and then 70%) for hydratization. Then the samples were incubated 15 min in 3% H2O2, followed by 15 min’s boiling in citrate buffer. The tissues were incubaed with primary antibody (GPR107, 1:100) and HRP-conjugated secondary antibody. Finally the sections were counterstained with hematoxylin and dehydrated in graded alcohols, followed by observation with Nikon-EclipseTi microscopy.

Realtime PCR

Total RNA was obtained from primary samples or oncospherss by standard TRIZOL method. Briefly, 1 ml TriZol was added into samples for dissolution through 5 min’s incubation. 200ul chloroform was then added into samples and mixed vigorously, followed by separation with 12,000 g centrifugation. The RNA components in supernatant were extracted with isopropanol and then washed with 75% ethanol. RNA pallets were dissolved with RNase-free H2O. RT-PCR (revers-transcription PCR) was performed to generate cDNA from RNA, and reatime PCR was performed to examine RNA expression with ABI7300. The primer sequences for realtime PCR were: GPR107, 5’-GGGGCAGTGTCATTTCAGTTT-3′, 5’-CTGAGAGGTAGCTGTCAGGAT-3′. lncGPR107, 5’-GGCTCAGCTACTGAAGCCGG-3′, 5’-AGGGGCCTGGCTGTCCTGCT-3′.

Western blot

Liver cancer or spheres were crushed with RIPA buffer (0.5% sodium deoxycholate, 1% NP-40, 1 mM EDTA, 0.1% SDS and 50 mM Tris, 150 mM NaCl, pH 8.0) and subjected into SDS–PAGE. The protein samples were transferred onto NC membrane (Beyotime Biotechnology), followed by incubation with primary antibodies and HRP-conjugated secondary antibodies for visualization.

RNA pulldown

Biotin-labeled lncGPR107 transcript was obtained with in vitro transcription (Roche), and incubated with oncosphere lysate. Then streptavidin conjugated beads were used for pulldown and enriched components were subjected into SDS–PAGE, followed by mass spectra or Western blot analyses.

Statistical analysis

One-tailed Student’s t tests were used for statistical analysis. *P < 0.05; **P < 0.01; ***P < 0.001. P < 0.05 was defined to be statistically significant. All experiments were repeated at least three times.

Results

GPR107 was up-regulated in liver cancer and liver TICs

Liver tumorigenesis is a long-term and complex process, and many signaling pathways are involved [26]. Wnt/β-catenin and Notch pathways are well investigated in liver tumorigenesis, and the roles of GPCR signaling in liver tumorigenesis and liver TICs are elusive. Here, we analyzed online-available dataset for GPCR expression. GSE14520 contains more than 400 samples with detailed clinical characteristics, and serves as a good candidate for dataset analysis [27, 28]. We analyzed the expression profiles of 382 GPCRs and found some GPCRs were highly expressed in liver tumors and some were down-regulated (Fig. 1a). Here, we focused on GPR107 because it is the most highly expressed GPCR in liver tumor samples (Fig. 1a, b). Moreover, the expression pattern of GPR107 was associated with liver tumor clinical characteristics, including metastasis, relapse and prognosis (Fig. 1c-e). We then obtained peri-tumor, early HCC (eHCC) and advanced HCC (aHCC) samples, and examined GPR107 expression with immunohistochemistry, confirming the high expression of GPR107 along with liver tumorigenesis (Fig. 1f).
We then evaluated the expression levels of GPR107 in liver TICs. We obtained HCC primary cells (Table 1), enriched liver TICs with surface marker CD133 and examined GPR107 with realtime PCR, and found GPR107 expression was increased in liver TICs (Fig. 1g). GPR107 was also highly expressed in oncospheres (Fig. 1h). The up-regulation of GPR107 in liver TICs was also confirmed by Western blot (Fig. 1i, j). Overall, GPR107 was up-regulated in liver cancer and liver TICs.

LncGPR107 was overexpressed in liver TICs

We then focused on GPR107 loci and found a long noncoding RNA LOC401554 (hereafter termed as lncGPR107). Many lncRNAs are may co-expressed with their neighboring genes [20], so we examined lncGPR107 expression pattern. LncGPR107 was also highly expressed in liver cancer, especially in advanced liver cancer (Fig. 2a). Realtime PCR results revealed the positive relationship between the expression levels of GPR107 and lncGPR107 (Fig. 2b). Northern blot and in situ hybridization validated the high expression of lncGPR107 in liver cancer samples (Fig. 2c, d).
We then analyzed lncGPR107 expression in liver TICs. CD133 positive liver TICs and oncospheres were enriched for lncGPR107 detection, revealing that lncGPR107 was highly expressed in liver TICs (Fig. 2e, f). Fluorescence in situ hybridization (FISH) assay also confirmed the overexpression of lncGPR107 in liver TICs (Fig. 2g) and oncospheres (Fig. 2h).

LncGPR107 was required for the self-renewal of liver TICs

Then we examined the role of lncGPR107 in self-renewal regulation of liver TICs. We generated lncGPR107 silenced cells through CRISPRi strategy [29], and found decreased expression of TIC markers and impaired self-renewal capacity (Fig. 3a, b). Tumor initiation assays also confirmed the impaired tumor initiation and decreased liver TIC ratios of lncGPR107 depleted cells (Fig. 3c). Tumor invasion was an important characteristic of liver TICs, and thus we examined the invasion capacity through transwell assay, and found impaired invasion of lncGPR107 depleted cells (Fig. 3d). We also generated lncGPR107 silenced cells through antisense oligo (ASO), and also found impaired self-renewal (Fig. 3e) and invasion (Fig. 3f).
We the activated lncGPR107 expression using CRISPRa strategy and found increased expression of liver TICs (Fig. 3g) [29]. LncGPR107 activation also enhanced sphere formation indicating the sufficient role of lncGPR107 in liver TIC (Fig. 3h). Through tumor initiation assay, enhanced tumor initiation capacities of lncGPR107 activated cells were also confirmed (Fig. 3i). Altogether, lncGPR107 played a necessary and sufficient role in liver TIC self-renewal.

LncGPR107 positively regulated the self-renewal of liver TICs through GPR107

Many lncRNAs participated in the transcription regulation of nearby genes. To explore the target genes of lncGPR107, we firstly examined the expression of GPR107. As expected, lncGPR107 depletion largely impaired the expression of GPR107, and lncGPR107 overexpression promoted GPR107 expression (Fig. 4a-c).
To explored the role of GPR107 in liver TIC regulation, we generated GPR107 knockout cells, and discovered impaired sphere formation of GPR107 deficient cells (Fig. 4d). Tumor initiation assay also indicated the essential role of GPR107 in liver tumor initiation (Fig. 4e). Our results indicated that GPR107 was overexpressed in liver TIC and played an essential role in the self-renewal regulation of liver TICs.
We then investigated whether lncGPR107 regulated liver TIC through GPR107 expression. We generated lncGPR107 knockdown cells using GPR107 deficient cells, and found upon GPR107 deletion, lncGPR107 had no role in the self-renewal of liver TICs (Fig. 4f). LncGPR107 overexpression also showed no influence in sphere formation upon GPR107 knockout (Fig. 4g). Using transwell invasion assay, we also confirmed that GPR107 was necessary for the function of lncGPR107 (Fig. 4h, I). Altogether, lncGPR107 drove the self-renewal of live TICs through GPR107.

LncGPR107 recruited SRCAP complex to GPR107 promoter to drove its expression

To further explore the molecular mechanism of lncGPR107, we performed RNA pulldown assay and silver staining. A specific band in lncGPR107 eluate was identified as SCRCAP by Mass spectrum (Fig. 5a). Western blot assay also validated the interaction of lncGPR107 and SRCAP (Fig. 5b). Through region mapping and RNA pulldown assay, the eighth region of lncGPR107 was found to bind SRCAP (Fig. 5c). RNA Electrophoretic Mobility Shift Assay (EMSA) confirmed the interaction of SRCAP and lncGPR107 (Fig. 5d). Meanwhile, the binding of SRCAP and lncGPR107 was also validated by RNA immunoprecipitation and FISH assays (Fig. 5e, f). In conclusion, lncGPR107 interacted with SRCAP in liver TICs.
SRCAP plays a critical role in chromatin remodeling and transcription regulation [30]. Accordingly, we examined if lnGPR107 and SRCAP in the activation of GPR107 promoter. ChIP assay confirmed the interaction between SRCAP and GPR107 promoter (Fig. 5g). In lncGPR107 depleted cells, impaired binding of SRCAP and GPR107 promoter was found, again confirming the critical role of lncGPR107 in recruiting SRCAP to GPR107 promoter (Fig. 5g). Along with SRCAP, the enrichment of H3K4me3 and RNA polymerase II was also impaired in lncGPR107 depleted cells, confirming the essential role of lncGPR107 in the activation of GPR107 promoter (Fig. 5g). LncGPR107 overexpression showed enhanced SRCAP recruiting and stronger activity of GPR107 promoter (Fig. 5h). Altogether, lncGPR107 recruited SRCAP complex to GPR107 promoter to drive its transcription activation.

LncGPR107-SRCAP-GPR107 signaling was required for liver TIC targeting

We then examined the role of lncGPR107-SRCAP-GPR107 in liver tumor propagation and targeting. Firstly, we examined tumor propagation of lncGPR107 depleted cells, and found decreased tumor volume upon lncGPR107 inhibition (Fig. 6a). On the contrary, lncGPR107 overexpression promoted tumor propagation (Fig. 6b). SRCAP and GPR107 deleted cells were also generated for liver tumor propagation, and smaller tumors were generated upon SRCAP knockout or GPR107 knockout (Fig. 6c).
We then collected the established tumors, examined GPR107 expression and found impaired GPR107 expression upon inhibition of lncGPR107-SRCAP-GPR107 signaling (Fig. 6d). FACS examination also confirmed that blockade of lncGPR107-SRCAP-GPR107 resulted in a decreased ratios of proliferating cells (Fig. 6e) and liver TICs (Fig. 6f), confirming the critical role of lncGPR107-SRCAP-GPR107 pathway in liver tumorigenesis and TIC self-renewal.

Discussion

With tumor initiation and self-renewal capacities, liver TICs (liver CSCs) are the origin of liver tumor formation, metastasis, relapse and drug resistance [3]. Several signaling pathways have been reported in the self-renewal regulation of liver TICs, including Wnt/β-catenin, Notch, Hedgehog and Yap1 signaling [12, 13, 3133]. However, the role of G-protein-coupled receptor (GPCR) signaling in liver TICs hasn’t been explored. Here, we analyzed the expression level of GPCRs using online-available HCC dataset and found GPR107 was the most highly expressed GPCRs along with liver tumorigenesis. Moreover, GPR107 is required for the self-renewal of liver TIC, revealing the relationship of liver TICs and GPCR signaling. Besides GPR107, several other GPRs, including OPN3, GPR88, LGR5, PTH2R, ACKR3 F2RL1, GABBR2, PTCFT and GPR5B, are also highly expressed in liver cancers (Fig. 1a). As one of the most important pathways in signal transduction, it is possible that more than one GPCR member participate in the self-renewal of liver TIC and liver tumorigenesis. Here, we revealed that GPR107 and lncGRP107 was highly expressed in liver cancer, especially in liver TICs (Fig. 1 and Fig. 2). In peri-tumor, the expression levels of GPR107 and lncGRP107 were extremely low, and very few cells showed modest expression of GPR107 and lncGRP107 (Figs. 1f and 2d). These GPR107 and lncGRP107 expressed cells may also have self-renewal and propagating capacities. Some groups identified liver progenitor cells in normal tissue, with self-renewal and differentiating capacities [34, 35]. It’s an interesting issue to investigate whether liver progenitor cells also show GPR107/lncGRP107 expression and the potential role of GPR107/lncGRP107 in liver progenitor cells.
Recently, GPCR signaling was found to be related to NFkB signaling [36] and Yap/Taz signaling [37], which regulated the activation of liver TICs. NFkB and Yap signaling pathways also participated in liver tumorigenesis [31]. Here we found GPR107 was overexpressed in and required for liver TICs, how GPR107 functions is still unclear. It’ll be interesting to detect the role of GPR107 in the activation of NFkB signaling and Yap/Taz signaling.
LncRNAs were recognized as the byproduct of RNA polymerase II, while, their critical roles emerge recently. Several lncRNAs regulate the transcription activation in cis. For example, lncTCF7 recruits SWI/SNF complex to activate TCF7 expression, and finally drives the self-renewal of liver TICs [24]. Our results provided another example for in cis regulation of lncRNA. Interestingly, lncGPR107 also recruited chromatin remodeling complex to the promoter of target gene. Through this regulation mechanism, lncGPR107 was co-expressed with GPR107 in liver TICs. LncGPR107 participated in the self-renewal of liver TICs through facilitating the transcriptional activation of GPR107. Through loss-of-function assays, GPR107 was also involved in the self-renewal of liver TICs.
To some extent, tumorigenesis is a process of chromatin remodeling [38]. Chromatin remodeling is mediated by chromatin remodeling complexes, including SWI/SNF, NURD, NURF, PRC1/2 and SRCAP complexes [3941]. Usually, the key components of many chromatin remodeling complexes are dys-regulated along with tumorigenesis. For example, two components of SWI/SNF (BRG1 and BRM) were differently expressed during liver tumorigenesis. BRG1 was highly expressed and BRM was lowly expressed in liver cancer and liver TICs [31]. EZH2, the core component of PRC2 complex, was also highly expressed in liver tumorigenesis [32]. Here, we found SRCAP was required for GPR107 expression, and the binding of SRCAP complex and GPR107 promoter is lncGPR107-dependent.

Conclusion

The role of GPCRs in liver TIC self-renewal remain unclear. Here, we found GPR107 was the most highly expressed GPCR in liver cancer and liver TICs. LncGPR107 participated in the transcriptional regulation of GPR107 in cis, through recruiting SRCAP remodeling complex to GPR107 promoter. This work revealed the important role of GPCR signaling in liver TIC self-renewal and added a new layer for liver TIC and GPCR regulation.

Acknowledgements

The authors thank Min Liang (Department of Abdominal Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University) for experimental supports.

Funding

This work was supported by science and technology projects of Guangdong province (2017A020211031, 2016ZC0145), science and technology projects of Guangzhou Medical University (201624), the National Natural Science Foundation of China (81673206) and science and technology projects of Guangzhou city (201508020262), Medical Scientific Research Foundation of Guangdong province (A2015243).

Availability of data and materials

All data and materials can be provided upon request.
Primary hepatocarcinoma samples were from the Fifth Affiliated Hospital of Guangzhou Medical University and Guangdong General Hospital with informed consent. The samples were ranked according to obtained time, and some samples (#1, #4 and #5) with sphere formation capacity were selected for analyses.
The author agree for publication.

Competing interests

All authors read and approved the final version of the manuscript, and the authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Zhu PP, Fan ZS. Cancer stem cell niches and targeted interventions. Prog Biochem Biophys. 2017;44:697–708. Zhu PP, Fan ZS. Cancer stem cell niches and targeted interventions. Prog Biochem Biophys. 2017;44:697–708.
2.
Zurück zum Zitat Chen ZZ, Zhang LF, He YL, Shen YQ, Li YF. Enhanced shRNA delivery and ABCG2 silencing by charge-reversible layered Nanocarriers. Small. 2015;11:952–62.CrossRef Chen ZZ, Zhang LF, He YL, Shen YQ, Li YF. Enhanced shRNA delivery and ABCG2 silencing by charge-reversible layered Nanocarriers. Small. 2015;11:952–62.CrossRef
3.
Zurück zum Zitat Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell. 2014;14:275–91.CrossRef Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell. 2014;14:275–91.CrossRef
4.
Zurück zum Zitat Pastrana E, Silva-Vargas V, Doetsch F. Eyes wide open: a critical review of sphere-formation as an assay for stem cells. Cell Stem Cell. 2011;8:486–98.CrossRef Pastrana E, Silva-Vargas V, Doetsch F. Eyes wide open: a critical review of sphere-formation as an assay for stem cells. Cell Stem Cell. 2011;8:486–98.CrossRef
5.
Zurück zum Zitat Chiba T, Kita K, Zheng YW, Yokosuka O, Saisho H, Iwama A, Nakauchi H, Taniguchi H. Side population purified from hepatocellular carcinoma cells harbors cancer stem cell-like properties. Hepatology. 2006;44:240–51.CrossRef Chiba T, Kita K, Zheng YW, Yokosuka O, Saisho H, Iwama A, Nakauchi H, Taniguchi H. Side population purified from hepatocellular carcinoma cells harbors cancer stem cell-like properties. Hepatology. 2006;44:240–51.CrossRef
6.
Zurück zum Zitat Zhu P, Wang Y, He L, Huang G, Du Y, Zhang G, Yan X, Xia P, Ye B, Wang S, et al. ZIC2-dependent OCT4 activation drives self-renewal of human liver cancer stem cells. J Clin Invest. 2015;125:3795–808.CrossRef Zhu P, Wang Y, He L, Huang G, Du Y, Zhang G, Yan X, Xia P, Ye B, Wang S, et al. ZIC2-dependent OCT4 activation drives self-renewal of human liver cancer stem cells. J Clin Invest. 2015;125:3795–808.CrossRef
7.
Zurück zum Zitat Feng DF, Peng G, Li W, Xu HM, Zhang TQ, Wang NY. Identification and characterization of tumorigenic liver Cancer stem cells by CD133 and CD24. J Biomater Tiss Eng. 2015;5:635–46.CrossRef Feng DF, Peng G, Li W, Xu HM, Zhang TQ, Wang NY. Identification and characterization of tumorigenic liver Cancer stem cells by CD133 and CD24. J Biomater Tiss Eng. 2015;5:635–46.CrossRef
8.
Zurück zum Zitat Ma S, Chan KW, Lee TKW, Tang KH, Wo JYH, Zheng BJ, Guan XY. Aldehyde dehydrogenase discriminates the CD133 liver cancer stem cell populations. Mol Cancer Res. 2008;6:1146–53.CrossRef Ma S, Chan KW, Lee TKW, Tang KH, Wo JYH, Zheng BJ, Guan XY. Aldehyde dehydrogenase discriminates the CD133 liver cancer stem cell populations. Mol Cancer Res. 2008;6:1146–53.CrossRef
9.
Zurück zum Zitat Haraguchi N, Ishii H, Mimori K, Tanaka F, Ohkuma M, Kim HM, Akita H, Takiuchi D, Hatano H, Nagano H, et al. CD13 is a therapeutic target in human liver cancer stem cells. J Clin Invest. 2010;120:3326–39.CrossRef Haraguchi N, Ishii H, Mimori K, Tanaka F, Ohkuma M, Kim HM, Akita H, Takiuchi D, Hatano H, Nagano H, et al. CD13 is a therapeutic target in human liver cancer stem cells. J Clin Invest. 2010;120:3326–39.CrossRef
10.
Zurück zum Zitat Yang ZF, Ho DW, Ng MN, Lau CK, Yu WC, Ngai P, Chu PWK, Lam CT, Poon RTP, Fan ST. Significance of CD90(+) cancer stem cells in human liver cancer. Cancer Cell. 2008;13:153–66.CrossRef Yang ZF, Ho DW, Ng MN, Lau CK, Yu WC, Ngai P, Chu PWK, Lam CT, Poon RTP, Fan ST. Significance of CD90(+) cancer stem cells in human liver cancer. Cancer Cell. 2008;13:153–66.CrossRef
11.
Zurück zum Zitat Chen ZZ, Huang L, Wu YH, Zhai WJ, Zhu PP, Gao YF. LncSox4 promotes the self-renewal of liver tumour-initiating cells through Stat3-mediated Sox4 expression. Nat Commun. 2016;7:12598. Chen ZZ, Huang L, Wu YH, Zhai WJ, Zhu PP, Gao YF. LncSox4 promotes the self-renewal of liver tumour-initiating cells through Stat3-mediated Sox4 expression. Nat Commun. 2016;7:12598.
12.
Zurück zum Zitat Zhu P, Wang Y, Du Y, He L, Huang G, Zhang G, Yan X, Fan Z. C8orf4 negatively regulates self-renewal of liver cancer stem cells via suppression of NOTCH2 signalling. Nat Commun. 2015;6:7122.CrossRef Zhu P, Wang Y, Du Y, He L, Huang G, Zhang G, Yan X, Fan Z. C8orf4 negatively regulates self-renewal of liver cancer stem cells via suppression of NOTCH2 signalling. Nat Commun. 2015;6:7122.CrossRef
13.
Zurück zum Zitat Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, Yang S, Ivy SP. Targeting notch, hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12:445–64.CrossRef Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, Yang S, Ivy SP. Targeting notch, hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12:445–64.CrossRef
14.
Zurück zum Zitat Venkatakrishnan AJ, Deupi X, Lebon G, Tate CG, Schertler GF, Babu MM. Molecular signatures of G-protein-coupled receptors. Nature. 2013;494:185–94.CrossRef Venkatakrishnan AJ, Deupi X, Lebon G, Tate CG, Schertler GF, Babu MM. Molecular signatures of G-protein-coupled receptors. Nature. 2013;494:185–94.CrossRef
15.
Zurück zum Zitat Dror RO, Pan AC, Arlow DH, Borhani DW, Maragakis P, Shan YB, Xu HF, Shaw DE. Pathway and mechanism of drug binding to G-protein-coupled receptors. P Natl Acad Sci Usa. 2011;108:13118–23.CrossRef Dror RO, Pan AC, Arlow DH, Borhani DW, Maragakis P, Shan YB, Xu HF, Shaw DE. Pathway and mechanism of drug binding to G-protein-coupled receptors. P Natl Acad Sci Usa. 2011;108:13118–23.CrossRef
16.
Zurück zum Zitat Tafesse FG, Guimaraes CP, Maruyama T, Carette JE, Lory S, Brummelkamp TR, Ploegh HL. GPR107, a G-protein-coupled receptor essential for intoxication by Pseudomonas aeruginosa exotoxin a, localizes to the Golgi and is cleaved by Furin. J Biol Chem. 2014;289:24005–18.CrossRef Tafesse FG, Guimaraes CP, Maruyama T, Carette JE, Lory S, Brummelkamp TR, Ploegh HL. GPR107, a G-protein-coupled receptor essential for intoxication by Pseudomonas aeruginosa exotoxin a, localizes to the Golgi and is cleaved by Furin. J Biol Chem. 2014;289:24005–18.CrossRef
17.
Zurück zum Zitat Yosten GLC, Redlinger LJ, Samson WK. Evidence for an interaction of neuronostatin with the orphan G protein-coupled receptor, GPR107. Am J Phys. 2012;303:R941–9. Yosten GLC, Redlinger LJ, Samson WK. Evidence for an interaction of neuronostatin with the orphan G protein-coupled receptor, GPR107. Am J Phys. 2012;303:R941–9.
18.
Zurück zum Zitat Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136:629–41.CrossRef Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136:629–41.CrossRef
19.
Zurück zum Zitat Liu BY, Ye BQ, Yang LL, Zhu XX, Huang GL, Zhu PP, Du Y, Wu JY, Qin XW, Chen RS, et al. Long noncoding RNA lncKdm2b is required for ILC3 maintenance by initiation of Zfp292 expression. Nat Immunol. 2017;18:499–508.CrossRef Liu BY, Ye BQ, Yang LL, Zhu XX, Huang GL, Zhu PP, Du Y, Wu JY, Qin XW, Chen RS, et al. Long noncoding RNA lncKdm2b is required for ILC3 maintenance by initiation of Zfp292 expression. Nat Immunol. 2017;18:499–508.CrossRef
20.
Zurück zum Zitat Luo S, Lu JY, Liu L, Yin Y, Chen C, Han X, Wu B, Xu R, Liu W, Yan P, et al. Divergent lncRNAs regulate gene expression and lineage differentiation in pluripotent cells. Cell Stem Cell. 2016;18:637–52.CrossRef Luo S, Lu JY, Liu L, Yin Y, Chen C, Han X, Wu B, Xu R, Liu W, Yan P, et al. Divergent lncRNAs regulate gene expression and lineage differentiation in pluripotent cells. Cell Stem Cell. 2016;18:637–52.CrossRef
21.
Zurück zum Zitat Liu BD, Sun LJ, Liu Q, Gong C, Yao YD, Lv XB, Lin L, Yao HR, Su FX, Li DS, et al. A cytoplasmic NF-kappa B interacting long noncoding RNA blocks I kappa B phosphorylation and suppresses breast Cancer metastasis. Cancer Cell. 2015;27:370–81.CrossRef Liu BD, Sun LJ, Liu Q, Gong C, Yao YD, Lv XB, Lin L, Yao HR, Su FX, Li DS, et al. A cytoplasmic NF-kappa B interacting long noncoding RNA blocks I kappa B phosphorylation and suppresses breast Cancer metastasis. Cancer Cell. 2015;27:370–81.CrossRef
22.
Zurück zum Zitat Wang KC, Chang HY. Molecular mechanisms of long noncoding RNAs. Mol Cell. 2011;43:904–14.CrossRef Wang KC, Chang HY. Molecular mechanisms of long noncoding RNAs. Mol Cell. 2011;43:904–14.CrossRef
23.
Zurück zum Zitat Batista PJ, Chang HY. Long noncoding RNAs: cellular address codes in development and disease. Cell. 2013;152:1298–307.CrossRef Batista PJ, Chang HY. Long noncoding RNAs: cellular address codes in development and disease. Cell. 2013;152:1298–307.CrossRef
24.
Zurück zum Zitat Wang Y, He L, Du Y, Zhu P, Huang G, Luo J, Yan X, Ye B, Li C, Xia P, et al. The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling. Cell Stem Cell. 2015;16:413–25.CrossRef Wang Y, He L, Du Y, Zhu P, Huang G, Luo J, Yan X, Ye B, Li C, Xia P, et al. The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling. Cell Stem Cell. 2015;16:413–25.CrossRef
25.
Zurück zum Zitat Hu YF, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods. 2009;347:70–8.CrossRef Hu YF, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods. 2009;347:70–8.CrossRef
26.
Zurück zum Zitat Bosch FX, Ribes J, Diaz M, Cleries R. Primary liver cancer: worldwide incidence and trends. Gastroenterology. 2004;127:S5–S16.CrossRef Bosch FX, Ribes J, Diaz M, Cleries R. Primary liver cancer: worldwide incidence and trends. Gastroenterology. 2004;127:S5–S16.CrossRef
27.
Zurück zum Zitat Roessler S, Long EL, Budhu A, Chen YD, Zhao XL, Ji JF, Walker R, Jia HL, Ye QH, Qin LX, et al. Integrative genomic identification of genes on 8p associated with hepatocellular carcinoma progression and patient survival. Gastroenterology. 2012;142:957–U451.CrossRef Roessler S, Long EL, Budhu A, Chen YD, Zhao XL, Ji JF, Walker R, Jia HL, Ye QH, Qin LX, et al. Integrative genomic identification of genes on 8p associated with hepatocellular carcinoma progression and patient survival. Gastroenterology. 2012;142:957–U451.CrossRef
28.
Zurück zum Zitat Roessler S, Jia HL, Budhu A, Forgues M, Ye QH, Lee JS, Thorgeirsson SS, Sun ZT, Tang ZY, Qin LX, Wang XW. A unique metastasis gene signature enables prediction of tumor relapse in early-stage hepatocellular carcinoma patients. Cancer Res. 2010;70:10202–12.CrossRef Roessler S, Jia HL, Budhu A, Forgues M, Ye QH, Lee JS, Thorgeirsson SS, Sun ZT, Tang ZY, Qin LX, Wang XW. A unique metastasis gene signature enables prediction of tumor relapse in early-stage hepatocellular carcinoma patients. Cancer Res. 2010;70:10202–12.CrossRef
29.
Zurück zum Zitat Gilbert LA, Horlbeck MA, Adamson B, Villalta JE, Chen Y, Whitehead EH, Guimaraes C, Panning B, Ploegh HL, Bassik MC, et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell. 2014;159:647–61.CrossRef Gilbert LA, Horlbeck MA, Adamson B, Villalta JE, Chen Y, Whitehead EH, Guimaraes C, Panning B, Ploegh HL, Bassik MC, et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell. 2014;159:647–61.CrossRef
30.
Zurück zum Zitat Wong MM, Cox LK, Chrivia JC. The chromatin remodeling protein, SRCAP, is critical for deposition of the histone variant H2A.Z at promoters. J Biol Chem. 2007;282:26132–9.CrossRef Wong MM, Cox LK, Chrivia JC. The chromatin remodeling protein, SRCAP, is critical for deposition of the histone variant H2A.Z at promoters. J Biol Chem. 2007;282:26132–9.CrossRef
31.
Zurück zum Zitat Zhu P, Wang Y, Wu J, Huang G, Liu B, Ye B, Du Y, Gao G, Tian Y, He L, Fan Z. LncBRM initiates YAP1 signalling activation to drive self-renewal of liver cancer stem cells. Nat Commun. 2016;7:13608.CrossRef Zhu P, Wang Y, Wu J, Huang G, Liu B, Ye B, Du Y, Gao G, Tian Y, He L, Fan Z. LncBRM initiates YAP1 signalling activation to drive self-renewal of liver cancer stem cells. Nat Commun. 2016;7:13608.CrossRef
32.
Zurück zum Zitat Zhu P, Wang Y, Huang G, Ye B, Liu B, Wu J, Du Y, He L, Fan Z. Lnc-beta-Catm elicits EZH2-dependent beta-catenin stabilization and sustains liver CSC self-renewal. Nat Struct Mol Biol. 2016;23:631–9.CrossRef Zhu P, Wang Y, Huang G, Ye B, Liu B, Wu J, Du Y, He L, Fan Z. Lnc-beta-Catm elicits EZH2-dependent beta-catenin stabilization and sustains liver CSC self-renewal. Nat Struct Mol Biol. 2016;23:631–9.CrossRef
33.
Zurück zum Zitat Chen Z, Gao Y, Yao L, Liu Y, Huang L, Yan Z, Zhao W, Zhu P, Weng H. LncFZD6 initiates Wnt/beta-catenin and liver TIC self-renewal through BRG1-mediated FZD6 transcriptional activation. Oncogene. 2018;37:3098–12. Chen Z, Gao Y, Yao L, Liu Y, Huang L, Yan Z, Zhao W, Zhu P, Weng H. LncFZD6 initiates Wnt/beta-catenin and liver TIC self-renewal through BRG1-mediated FZD6 transcriptional activation. Oncogene. 2018;37:3098–12.
34.
Zurück zum Zitat Huch M, Dorrell C, Boj SF, van Es JH, Li VS, van de Wetering M, Sato T, Hamer K, Sasaki N, Finegold MJ, et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 2013;494:247–50.CrossRef Huch M, Dorrell C, Boj SF, van Es JH, Li VS, van de Wetering M, Sato T, Hamer K, Sasaki N, Finegold MJ, et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 2013;494:247–50.CrossRef
35.
Zurück zum Zitat Wang B, Zhao L, Fish M, Logan CY, Nusse R. Self-renewing diploid Axin2(+) cells fuel homeostatic renewal of the liver. Nature. 2015;524:180–5.CrossRef Wang B, Zhao L, Fish M, Logan CY, Nusse R. Self-renewing diploid Axin2(+) cells fuel homeostatic renewal of the liver. Nature. 2015;524:180–5.CrossRef
36.
Zurück zum Zitat Gao H, Sun Y, Wu YL, Luan B, Wang YY, Qu B, Pei G. Identification of beta-arrestin2 as a G protein-coupled receptor-stimulated regulator of NF-kappa B pathways. Mol Cell. 2004;14:303–17.CrossRef Gao H, Sun Y, Wu YL, Luan B, Wang YY, Qu B, Pei G. Identification of beta-arrestin2 as a G protein-coupled receptor-stimulated regulator of NF-kappa B pathways. Mol Cell. 2004;14:303–17.CrossRef
37.
Zurück zum Zitat Yu FX, Zhao B, Panupinthu N, Jewell JL, Lian I, Wang LH, Zhao JG, Yuan HX, Tumaneng K, Li HR, et al. Regulation of the hippo-YAP pathway by G-protein-coupled receptor signaling. Cell. 2012;150:780–91.CrossRef Yu FX, Zhao B, Panupinthu N, Jewell JL, Lian I, Wang LH, Zhao JG, Yuan HX, Tumaneng K, Li HR, et al. Regulation of the hippo-YAP pathway by G-protein-coupled receptor signaling. Cell. 2012;150:780–91.CrossRef
38.
Zurück zum Zitat Wang GG, Allis CD, Chi P. Chromatin remodeling and cancer, part II: ATP-dependent chromatin remodeling. Trends Mol Med. 2007;13:373–80.CrossRef Wang GG, Allis CD, Chi P. Chromatin remodeling and cancer, part II: ATP-dependent chromatin remodeling. Trends Mol Med. 2007;13:373–80.CrossRef
39.
Zurück zum Zitat Clapier CR, Cairns BR. The biology of chromatin remodeling complexes. Annu Rev Biochem. 2009;78:273–304.CrossRef Clapier CR, Cairns BR. The biology of chromatin remodeling complexes. Annu Rev Biochem. 2009;78:273–304.CrossRef
40.
Zurück zum Zitat Klochendler-Yeivin A, Muchardt C, Yaniv M. SWI/SNF chromatin remodeling and cancer. Curr Opin Genet Dev. 2002;12:73–9.CrossRef Klochendler-Yeivin A, Muchardt C, Yaniv M. SWI/SNF chromatin remodeling and cancer. Curr Opin Genet Dev. 2002;12:73–9.CrossRef
41.
Zurück zum Zitat Lai AY, Wade PA. Cancer biology and NuRD: a multifaceted chromatin remodelling complex. Nat Rev Cancer. 2011;11:588–96.CrossRef Lai AY, Wade PA. Cancer biology and NuRD: a multifaceted chromatin remodelling complex. Nat Rev Cancer. 2011;11:588–96.CrossRef
Metadaten
Titel
RETRACTED ARTICLE: LncGPR107 drives the self-renewal of liver tumor initiating cells and liver tumorigenesis through GPR107-dependent manner
Publikationsdatum
01.12.2018
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2018
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0794-3

Weitere Artikel der Ausgabe 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.