Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2016

Open Access 01.12.2016 | Research

LRG1 modulates epithelial-mesenchymal transition and angiogenesis in colorectal cancer via HIF-1α activation

verfasst von: Jingjing Zhang, Lingyin Zhu, Jingyuan Fang, Zhizheng Ge, Xiaobo Li

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2016

Abstract

Background

Leucine-rich-alpha-2-glycoprotein 1 (LRG1) has been reported to be involved in several tumors, whether it participates in colorectal cancer (CRC) progression remains unclear. Here, we investigated the biological function and underlying molecular mechanisms of LRG1 in CRC.

Methods

The mRNA and protein levels of LRG1 were assessed in CRC tissues through RT-PCR and immunohistochemistry, respectively. HCT116 and SW480 cells were treated with LRG1 siRNA, control siRNA, or recombinant LRG1. Transwell invasion assays and wound healing assays were performed to evaluate the invasion and migration of CRC cells. Epithelial-to-mesenchymal transition (EMT) markers of E-cadherin, VDR, N-cadherin, α-SMA, Vimentin and Twist1 were detected by RT-PCR and western blot. Enzyme-linked immunosorbent assay was used to measure the secretion level of VEGF-A. Conditioned medium from CRC cells was collected for endothelial cell migration, tube formation and aortic ring sprouting assays.

Results

LRG1 was overexpressed in CRC tissues and associated with cancer aggressiveness. LRG1 was further found to induce the EMT process, as well as CRC cell migration and invasion capacity. In addition, LRG1 promoted VEGF-A expression in CRC cells and contributed to tumor angiogenesis. Furthermore, HIF-1α could be induced by LRG1 in a concentration- and time-dependent manner, which was responsible for LRG1-induced VEGF-A expression and EMT.

Conclusions

The present study suggests that LRG1 plays a crucial role in the progression of CRC by regulating HIF-1α expression, thereby may be a promising therapeutic target of CRC.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

JJZ and LYZ conceived and designed the experiments. JJZ performed the experiments and drafted the manuscript. JYF, ZZG and XBL contributed to the design of this study and helped to draft the manuscript. XBL supervised the whole work and revised the manuscript. All authors read and approved the manuscript.
Abkürzungen
LRG1
leucine-rich-alpha-2-glycoprotein 1
CRC
colorectal cancer
EMT
epithelial-to- mesenchymal transition
HIF-1α
hypoxia-inducible factor-1α
VEGF-A
vascular endothecial growth factor A

Background

Despite advances in early diagnosis and comprehensive therapy, colorectal cancer (CRC) remains one of the leading causes of cancer death worldwide. The prognoses of CRC patients were often poor due to recurrence and metastasis, especially for those diagnosed at advanced stages. The 5-year survival rate is only 12 % for CRC patients with distant metastasis [1]. Hence, elucidating the molecular events involved in CRC and identifying novel biomarker and therapeutic targets is indispensible and urgent for the clinical outcome of CRC.
Leucine-rich-alpha-2-glycoprotein1 (LRG1) is the founding member of leucine-rich repeat (LRR) family, which was first isolated from human serum in 1977 [2]. It is a secreted glycoprotein and contains eight repeating consensus sequences, each of which consists of 24 amino acid residues [3]. LRG1 has been reported to be involved in immune response, cell proliferation, cell migration, cell apoptosis and neovascularization [47]. LRG1 is overexpressed in several types of carcinomas, including pancreatic, bladder, ovarian, and biliary tract cancer [811]. LRG1 was shown to bind to the transforming growth factor-beta (TGF-β) accessory receptor and modulate Smad1/5/8 signalling pathway, resulting in promotion of angiogenesis in endothelial cells [7]. It was reported that LRG1 was a target of miR-335 and contribute to the migratory and invasive ability of neuroblastoma cell [5]. However, little is known about the biological function of LRG1 in colorectal cancer.
To reveal the signaling proteins downstream of LRG1, we performed a gene microarray and found that several key proteins involved in epithelial-to-mesenchymal transition (EMT) were affected by depletion of LRG1 in CRC cells. EMT is a pathological process that epithelial cells lose their characteristic of cell polarity and adhesion, while acquire a mesenchymal phenotype [12]. EMT process results in enhanced ability of mobility and invasiveness for carcinoma cells, which is considered as a crucial early step in cancer progression and metastasis [1315]. Besides, hypoxia-inducible factor-1α (HIF-1α) and vascular endothecial growth factor A (VEGF-A) was significantly downregulated in LRG1-knockdown CRC cells. HIF-1α is the oxygen-regulated subunit of HIF-1, which is the most important transcriptional regulator in response to hypoxia. HIF-1α participates in the key steps in carcinogenesis such as cell survival, angiogenesis and metastasis, through transcriptional activation of targeted genes [16, 17]. Notably, HIF1α has been identified as an important mediator of EMT in tumor cells via activation of Twist, Snail, and SIP1 [18, 19]. The mechanisms underlying HIF-1α-induced EMT in CRC have not been completely determined. Therefore, LRG1 might be associated with the EMT and angiogenesis in CRC.
In the present study, we investigated the expression level of LRG1 in CRC tissues and explored the role of LRG1 in CRC cell invasion, EMT, and endothelial cell activities. We also aimed to validate the promotion effect of LRG1 on HIF-1α expression and test the hypothesis that HIF-1α is involved in LRG1-induced EMT and angiogenesis in CRC.

Methods

Cell culture and LRG1 treatment

Human colorectal carcinoma cell lines, SW480 and HCT116, were cultured in RPMI 1640 medium supplemented with 10 % fetal bovine serum (FBS) in humidified 5.0 % CO2 atmosphere at 37 °C. Human umbilical vein endothelial cells (HUVEC) were maintained in DMEM containing 10 % FBS. Recombinant LRG1 was purchased from R&D Systems and added into the culture medium at concentration of 50–1000 ng/ml for indicated time before harvesting.

Small interfering RNA silencing

Transfection of siRNA was performed using Lipofectamine 2000 (Invitrogen, USA) according to the manufacturer’s protocol. The transfection reagent was replaced by complete medium after incubation for 6 h, and cells were harvested 24 h or 48 h later. The siRNA oligos for LRG1 (#1: sense, 5′-CCUCUAAGCUCCAAGAAUUTT-3′ and antisense, 5′-AAUUCUUGGAGCUUAGAGGTT-3′; #2: sense, 5′-GCAAUUAGAACGGCUACAUT T-3′ and antisense, 5′-AUGUAGCCGUUCUAAUUGCTT-3′), HIF-1ɑ (#1: sense, 5′-GGAA AGAGACUCAUAGAAA-3′ and antisense, 5′-UUUCUAUGACUCUCUUUCC-3′; #2: sense, 5′-GCACAGGCCACATTCACGTATAT-3′ and antisense, 5′- GGTTCACAAATCAGC ACCAAGC-3′), and a non-targeting control siRNA were purchased from GenePharma (China).

RNA extraction and quantitative real-time PCR

Total RNA was extracted by Trizol (Invitrogen, USA), and cDNA was synthesized using the PrimeScript TM RT Reagent Kit (Perfect Real Time, TaKaRa, Japan). Quantitative real-time PCR was performed in a 10ul total volume containing SYBR Green (SYBR® Premix Ex Taq TM II, TaKaRa, Japan) on an Applied Biosystems 7900 quantitative PCR system. The primers used were as follows: LRG1, 5′-GTTGGAGACCTTGCCACCT-3′ and 5′-GCTTGTTGCCGTTCAGGA-3′; HIF-1α, 5′-TGCTAATGCCACCACTACC-3′ and 5′-TG ACTCCTTTTCCTGCTCTG-3′; VEGF-A, 5′-CTTTCTGCTGTCTTGGGTG-3′ and 5′-ACT TCGTGATGATTCTGCC-3′; Twist1, 5′-AGTCCGCAGTCTTACGAGGA-3′ and 5′-GCCAG CTTGAGGGTCTGAAT-3′; E-cadherin, 5′-TACACTGCCCAGGAGCCAGA-3′ and 5′-TGG CACCAGTGTCCGGATTA-3′; N-cadherin, 5′-TTTGATGGAGGTCTCCTAACACC-3′ and 5′-ACGTTTAACACGTTGGAAATGTG-3′; VDR, 5′-GATGCCCACCACAAGACCTA-3′ and 5′-CGGTTCCATCATGTCCAGTG-3′; Vimentin, 5′-TGAGTACCGGAGACAGGTGCA G-3′ and 5′-TAGCAGCTTCAACGGCAAAGTTC-3′; α-SMA, 5′-CGTGGCTACTCCTTCGT G-3′ and 5′-TGATGACCTGCCCGTCT-3′; β-actin, 5′-TGGCACCCAGCACAATGAA-3′ and 5′-CTAAGTCATAGTCCGCCTAGAAGCA-3′. Relative expression of each specific gene was determined in accordance with the 2−ΔΔCt method, using β-actin as the internal standard. Each experiment was performed as triplicate, and data was presented as mean ± SEM.

Clinical specimens and immunohistochemistry

Human CRC tissues and adjacent non-cancerous tissues were obtained from patients who underwent surgical resection at Renji Hospital. Histological diagnoses were performed by expert pathologists. The study was approved by the Ethics Committee of Renji hospital, and written informed consent was obtained from all patients included in this study.
Paraffin-embedded specimens of 68 colorectal cancers and 32 normal colorectal tissues were selected for immunohistochemistry. Briefly, antigen retrieval was performed with 10 mM citrate buffer (Ph 6.0) using microwave. Sections were incubated with anti-LRG1 antibody (1:100, Abcam, USA) overnight at 4 °C, followed by HRP-conjugated secondary antibody. LRG1 expression was quantified based on the intensity of staining (scored as: 0, no staining; 1, weak staining; 2, moderate staining; 3, strong staining) and the percentage of positive tumor cells (scored as: 0, less than 5 %; 1, 5–25 %; 2, 26–50 %; 3, > 51 %). The final score was calculated as the product of two parameters, and at least 3 points was considered as positive.

Invasion and migration assay in vitro

The cell invasion assay was carried out using chambers with filters (pore size of 8-um), coated with Matrigel. The cells (2*105 cells per well) were seeded into the upper chamber in serum-free medium, while medium with 20 % FBS was applied to the lower chamber. For endothelial cells, conditioned medium was added to the lower chamber. After incubation for 48 h, invasive cells on the bottom surface of the membrane were fixed with 4 % formaldehyde, stained with crystal violet, and counted in five random microscopic fields for each replicate (original magnification, 200×).
The migration ability of cells was measured through wound healing assay. Cells were cultured in 6-well plates to reach 90 % confluence. The cell monolayers were scraped with a 100-ul pipette tip, washed twice with PBS, and cultured in serum-free medium. After 24 h and 48 h, the scratch area was photographed (original magnification, 200×).

Conditioned medium and enzyme-linked immunosorbent assay

CRC cells cultured in 6-well plates were treated with LRG1 for 24 h. Thereafter, cells were washed three times and changed to fresh serum-free medium for additional 24 h. The supernatants were harvested, centrifuged at 3500 rpm for 5 min, and stored at−80 °C until used as conditioned medium (CM). Tumor-derived VEGF-A in the medium was quantified by enzyme-linked immunosorbent assay (ELISA). VEGF-A concentration was determined using ELISA Kit (R&D Systerms Europe, UK) according to the manufacturer’s instructions.

Endothelial cell tube formation assay

Matrigel (BD Biosciences, CA) was laid into a 48-well plate and polymerized at 37 °C for 30 min. Then, 3 × 104 HUVECs were seeded into each well of pre-coated 48-well plate and incubated with conditioned medium. After 8 h, capillary-like tubes were photographed (original magnification, 100×) from four randomly chosen fields, and the total number of complete tubular structures was quantified.

Aortic ring sprouting assay

Aortas were excised from 8-week-old Sprague–Dawley rats and dissected into rings of 1 mm. Aortic rings were embedded in Matrigel (BD Biosciences, California, USA) in a 48-well culture plate. CM was added to the wells in a final volume of 200ul culture medium. The aortic rings were incubated at 37 °C for 6 days with medium replaced every other day. On day 6, the microvessel sprouting was photographed and scored from 0 (least positive) to 5 (most positive) in a double-blinded manner as previously described [20]. Three independent experiments were carried out with five rings per group in each assay. Representative micrographs were shown.

Western blot

Whole-cell protein extracts were prepared with RIPA buffer containing a protease inhibitor mixture. Protein concentrations were determined using BCA Protein Assay. Equal amounts of total protein were separated by SDS–polyacrylamide gel electrophoresis, transferred onto the polyvinylidene difluoride membrane, and blocked with 5 % fat-free milk. The membranes were incubated with primary antibodies overnight at 4 °C, followed by HRP-conjugated secondary antibodies for 1 h at room temperature. The immune complexs were detected using a chemiluminescence kit (SuperSignal ECL Kit, Thermo Fisher, USA). Antibody for LRG1 was purchased from Abcam (USA), and other antibodies were all purchased from Cell Signaling Technology Inc (USA). Intensity of the protein bands was quantified by Image J software and normalized to that of β-actin.

Statistical analysis

Statistical analysis was carried out using SPSS 20.0 software (SPSS, Chicago, IL, USA). Quantitative data were expressed as the means ± SEM, and comparisons between every two groups were performed with Student t test or paired t test. The association between LRG1 staining and the clinicopathologic features of CRC patients were examined by Chi-square tests. P-value <0.05 was considered statistically significant.

Results

LRG1 expression was increased in human CRCs and correlated with tumor progression

To investigate the mRNA expression level of LRG1 in CRC, we analysed the microarray data from two Oncomine Cancer Microarray databases (GSE20916 and GSE20842) and revealed remarkable overexpression of LRG1 in CRC tissues than normal tissues (Fig. 1a). Next, we performed real-time RT-PCR to assess LRG1 expression in 30 cases of CRC tissues and matched normal tissues. Similar result was observed that LRG1 was significantly overexpressed in CRC tissues, compared with corresponding normal tissues (P < 0.0001, Fig. 1b). Immunohistochemistry was performed to examine LRG1 protein in 68 CRC and 32 normal paraffin-embedded tissues. Positive staining of LRG1 was mainly distributed in the cytoplasm and cytomembrane of CRC cells (Fig. 1c). Positive expression rate of LRG1 was 66.18 % (45/68) in CRC tissues, significantly higher than that of normal colorectal mucosa (31.25 %, 12/32) (P = 0.007). The clinicopathologic features of the 68 CRCs were summarized in Table 1. LRG1 expression was significantly correlated with deeper invasion depth (P = 0.032) and lymph node metastasis (P = 0.013). There was no significant correlation between LRG1 expression and other clinicopathological characteristics, including age, gender, location, tumor size and distal metastasis.
Table 1
Correlation between LRG1 expression and clinicopathological parameters of CRCs
 
n
LRG1 expression
P values
Negative
Positive
Age
    
  < 60
37
12
25
0.791
  ≥ 60
31
11
20
 
Gender
    
 Male
41
13
28
0.649
 Female
27
10
17
 
Location
    
 Right colon
21
9
12
0.396
 Left colon
18
4
14
 
 Rectum
29
10
19
 
Tumor diameter
    
  < 5 cm
35
11
24
0.667
  ≥ 5 cm
33
12
21
 
T stage
    
 T1 + T2
32
15
17
0.032
 T3 + T4
36
8
28
 
Lymph nodes metastasis
    
 Negative
39
18
21
0.013
 Positive
29
5
24
 
Distant metastasis
    
 Negative
63
23
40
0.242
 Positive
5
0
5
 

LRG1 influenced CRC cell migration, invasion and EMT

The overexpression of LRG1 in CRCs suggested that LRG1 might play a role in its migration and invasion capability. To investigate the function of LRG1 in CRC cells, we knocked down LRG1 by transfection of specific siRNA. Both mRNA and protein levels of LRG1 were efficiently depleted in HCT116 and SW480 cells (Fig. 2a). Transwell and wound healing assay were used to examine invasion and migration of CRC cells. In transwell assays, knockdown of LRG1 significantly reduced the number of cells invaded through the membrane compared with the control (Fig. 2b). Likewise, wound closure ratio was markedly reduced to 48 and 61 % respectively (P both <0.05) in LRG1-siRNA transfected HCT116 and SW480 cells (Fig. 2c). Next, we examined the expression of EMT markers following knockdown of LRG1. Real-time RT-PCR and western blot analysis showed that epithelial biomarkers of E-cadherin and VDR were increased in LRG1-depleted CRC cells, whereas mesenchymal biomarkers of N-cadherin, α-SMA and Vimentin were decreased. Furthermore, we investigated whether the EMT-related transcription factors were involved in LRG1-promoted EMT. Results showed that Twist1 expression was significantly decreased by LRG1 knockdown in both cell lines (Fig. 3a and b). These results suggested that LRG1 can enhance CRC cell invasion and induce EMT.

LRG1 supplementation enhanced expression and secretion of VEGF-A and promotes endothelial cells migration and tube formation

Angiogenesis is an essential step in tumor metastasis. Given that LRG1 was reported to be a new regulator of angiogenesis, we determined whether LRG1 was involved in CRC angiogenesis. Our data implied that treatment with human rLRG1 for 24 h concentration- dependently increased VEGF-A mRNA expression and protein secretion level in both SW480 and HCT116 cells (Fig. 4a). Similarly, VEGF-A expression was elevated by stimulation with rLRG1 in a time-dependent manner (Fig. 4b). To further investigate the role of LRG1 in angiogenesis, we collected the CM derived from HCT116 cells incubated in the presence or absence of rLRG1 (500 ng/ml). The CM was then applied to HUVECs for transwell migration, tube formation and aortic ring sprouting assays. As shown in Fig. 4c, CM from CRC cells exposed to LRG1 promoted endothelial cell migration. In tube formation assays, the cumulative number of tubular structures formed by HUVECs in CM from LRG1-treated CRC cells was significantly increased (2.6 fold, Fig. 4d). Microvessel sprouting from aortic rings was significantly stimulated in the presence of CM from LRG1-treated CRC cells (Fig. 4e). In these assays, the promotion effect of LRG1 on endothelial cell behaviors was blocked by LRG1 mAb. These results provided evidence for the role of LRG1 in promotion of angiogenic capability in colorectal carcinomas.

HIF-1α was essential for LRG1-mediated EMT and angiogenesis

HIF-1α was a critical regulator in both tumor angiogenesis and hypoxia-induced EMT. To examine whether LRG1 would enhance HIF-1α expression, CRC cells were treated with rLRG1 for 24 h. HIF-1α mRNA expression was induced at 100 ng/ml and reached the maximum level at 1000 ng/ml (Fig. 5a). Protein expression of HIF-1α was also increased following stimulation with rLRG1 in a concentration-dependent manner in SW480 cells. LRG1 stimulation also resulted in increased expression of VEGF-A, Twist1 and N-cadherin, while decreased E-cadherin expression (Fig. 5b). Further we determined the role of HIF-1α in LRG1-induced VEGF-A overexpression and EMT. Transfection with siRNA successfully silenced the mRNA and protein levels of HIF-1α expression in SW480 cells (Fig. 5c). Secretion of VEGF-A by SW480 cells was elevated in the presence of LRG1, and the effect was abolished by knockdown of HIF-1α (Fig. 5d). Knockdown of HIF-1α also reversed LRG1-induced EMT phenotype, increasing E-cadherin expression, and decreasing Twist1 and N-cadherin expression (Fig. 5e). As shown in Fig. 5f, silencing HIF-1α inhibited the enhanced invasion ability of CRC cells induced by LRG1. These results strongly implied that HIF-1α is implicated in LRG1-induced CRC cells invasiveness, EMT and angiogenesis.

Discussion

It was reported that plasma levels of LRG1 in patients with CRC were higher than those with adenomatous polyps, which might be a novel biomarker for the progression from colorectal adenoma to carcinoma [21]. In our study, we demonstrated that LRG1 expression was significantly increased in CRC tissues compared with normal tissues. In addition, expression level of LRG1 was positively associated with deeper invasion depth and lymphatic metastasis. These results imply that LRG1 may be involved in colorectal cancer initiation and progression, which is in agreement with published studies of ovarian cancer and biliary tract cancer [10, 11]. However, LRG1 has been reported to act as a tumor suppressor in hepatocellular carcinoma and endometrial carcinoma [22, 23]. The discrepancy of LRG1 function in certain kinds of cancers may be due to its tissue specificity.
Although LRG1 is found to be implicated in various kinds of malignant carcinomas and benign diseases, the biological functions of LRG1 have not been fully elucidated. LRG1 has been predicted to play a role in cell adhesion and cell migration due to its leucine-rich repeats and tendency to bind extracellular matrix proteins [24]. A recent study showed that LRG1 promoted proliferation, migration and invasion of glioma cells, as well as downregulated the expression of E-cadherin [25]. Our experiments demonstrated that knockdown of LRG1 by siRNA significantly attenuated the migratory and invasive potency of CRC cells. Moreover, depletion of LRG1 caused a shift from mesenchymal markers to epithelial markers, which was upregulation of E-cadherin and VDR, and downregulation of N-cadherin, α-SMA and Vimentin. Meanwhile, stimulation with recombinant LRG1 induced EMT and enhanced cell invasion. The expression of Twist1, a well known EMT-promoting transcriptional factor, was also found to be positively regulated by LRG1. Our findings identified LRG1 as a novel inducer of EMT and a potential enhancer of the metastatic potential in colorectal cancer.
Sustained angiogenesis is known as an essential part in the unrestrained growth of tumors and metastasis of cancer cells to distant organs. It has been reported that LRG1 influences endothelial cells proliferation, migration, tube formation, and promotes blood vessel growth in ex vivo models of angiogenesis [7]. Whether LRG1 could regulate angiogenesis and be a promising anti-angiogenic therapy target in colorectal cancer were unknown. Our study revealed that treatment of CRC cells with rLRG1 induced the expression of critical pro-angiogenic molecules including HIF-1α and VEGF-A. Cytokines derived from the tumor microenvironment could facilitate the activities of endothelial cells and promote angiogenesis [26, 27]. Therefore, we used the conditional medium from CRC cells to determine whether LRG1 expression modulated endothelial cell migration and tube formation, both of which are critical indicators of angiogenic ability. We found that endothelial cell cultured with CM from rLRG1-stimulated CRC cells exhibited an increased migration and tube formation capacity. Taken together, our results indicated that overexpression of LRG1 in CRC cells might promote the secretion of pro-angiogenic factors like VEGF-A, thus contributing to CRC angiogenesis. Further studies with in vivo experiments are needed to demonstrate the biological role of LRG1 in CRC angiogenesis and metastasis.
In this study, we found that treatment of CRC cells with LRG1 promoted the mRNA and protein expression of HIF-1α. Hypoxia or overexpression of HIF-1α is associated with resistance to cancer chemotherapy and increased patient mortality [28]. HIF-1α was reported to be overexpressed in CRC and correlated with poor prognosis [29, 30]. Since the critical role of HIF-1α in tumor progression and metastasis, inhibition of HIF-1α was considered as a promising therapeutic strategy. HIF-1α, the regulatory subunit of HIF-1, is mainly regulated in an oxygen-dependent manner. HIF-1α is rapidly degraded under normoxic conditions through various post-translational modifications of hydroxylation, ubiquitination and ultimate proteasomal degradation [31]. However, several stimuli such as EGF, TGF-β, and IGF-1 can induce HIF-1α accumulation under normoxia, at various levels of transcription, translation, and protein stability in a cell-type-specific manner [3234]. Consistent with the role of HIF-1α in inducing EMT and metastatic phenotypes, our results indicated HIF-1α to be involved in LRG1-induced EMT, since knockdown of HIF-1α blocked the mesenchymal phenotype and invasive capacity induced by LRG1. Moreover, HIF-1α knockdown abrogated the LRG1-induced VEGF-A expression, which acts as a key downstream target of HIF-1α. These data suggested that LRG1 may target the HIF-1α pathway to induce CRC cell invasion and angiogenesis. A recent study showed that LRG1 overexpression activated the canonical TGF-β signaling pathway and enhanced the invasiveness of glioma cells, which could be reversed by TGF-β signaling pathway inhibitor [24]. It has been shown that TGF-β stimulates HIF-1α accumulation and that HIF-1α mediates TGF-β-induced EMT [35]. It is not clear whether LRG1 activates the transcription of HIF-1α directly or via TGF-β pathway.

Conclusion

In summary, this is a preliminary study to investigate the role of LRG1 in colorectal cancer. Our results provided evidence for LRG1 function as a novel inducer of EMT and angiogenesis in colorectal cancer, which was at least partially through promotion of HIF-1α expression. Thus, LRG1 may be a potential biomarker and therapeutic target for colorectal cancer. Further researches are still needed to clarify the molecular mechanisms and therapeutic benefits of LRG1 on CRC and other malignancies.

Acknowledgments

This study was supported by Innovation Program of Shanghai Municipal Education Commission. (No.12YZ036).
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.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

JJZ and LYZ conceived and designed the experiments. JJZ performed the experiments and drafted the manuscript. JYF, ZZG and XBL contributed to the design of this study and helped to draft the manuscript. XBL supervised the whole work and revised the manuscript. All authors read and approved the manuscript.
Literatur
1.
2.
Zurück zum Zitat Haupt H, Baudner S. Isolation and characterization of an unknown, leucine-rich 3.1-S- alpha2-glycoprotein from human serum. Hoppe Seyler Z Physiol Chem. 1977;358(6):639–46.CrossRefPubMed Haupt H, Baudner S. Isolation and characterization of an unknown, leucine-rich 3.1-S- alpha2-glycoprotein from human serum. Hoppe Seyler Z Physiol Chem. 1977;358(6):639–46.CrossRefPubMed
3.
Zurück zum Zitat Takahashi N, Takahashi Y, Putnam FW. Periodicity of leucine and tandem repetition of a 24-amino acid segment in the primary structure of leucine-rich alpha 2-glycoprotein of human serum. Proc Natl Acad Sci U S A. 1985;82(7):1906–10.PubMedCentralCrossRefPubMed Takahashi N, Takahashi Y, Putnam FW. Periodicity of leucine and tandem repetition of a 24-amino acid segment in the primary structure of leucine-rich alpha 2-glycoprotein of human serum. Proc Natl Acad Sci U S A. 1985;82(7):1906–10.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Serada S, Fujimoto M, Terabe F, Iijima H, Shinzaki S, Matsuzaki S, et al. Serum leucine- rich alpha-2 glycoprotein is a disease activity biomarker in ulcerative colitis. Inflamm Bowel Dis. 2012;18:2169–79.CrossRefPubMed Serada S, Fujimoto M, Terabe F, Iijima H, Shinzaki S, Matsuzaki S, et al. Serum leucine- rich alpha-2 glycoprotein is a disease activity biomarker in ulcerative colitis. Inflamm Bowel Dis. 2012;18:2169–79.CrossRefPubMed
5.
Zurück zum Zitat Lynch J, Fay J, Meehan M, Bryan K, Watters KM, Murphy DM, et al. MiRNA-335 suppresses neuroblastoma cell invasiveness by direct targeting of multiple genes from the non-canonical TGF-β signalling pathway. Carcinogenesis. 2012;33(5):976–85.PubMedCentralCrossRefPubMed Lynch J, Fay J, Meehan M, Bryan K, Watters KM, Murphy DM, et al. MiRNA-335 suppresses neuroblastoma cell invasiveness by direct targeting of multiple genes from the non-canonical TGF-β signalling pathway. Carcinogenesis. 2012;33(5):976–85.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Zhong D, Zhao S, He G, Li J, Lang Y, Ye W, et al. Stable knockdown of LRG1 by RNA interference inhibits growth and promotes apoptosis of glioblastoma cells in vitro and in vivo. Tumor Biol. 2015;36(6):4271–8.CrossRef Zhong D, Zhao S, He G, Li J, Lang Y, Ye W, et al. Stable knockdown of LRG1 by RNA interference inhibits growth and promotes apoptosis of glioblastoma cells in vitro and in vivo. Tumor Biol. 2015;36(6):4271–8.CrossRef
7.
Zurück zum Zitat Wang X, Abraham S, McKenzie JA, Jeffs N, Swire M, Tripathi VB, et al. LRG1 promotes angiogenesis by modulating endothelial TGF-β signalling. Nature. 2013;499:306–11.CrossRefPubMed Wang X, Abraham S, McKenzie JA, Jeffs N, Swire M, Tripathi VB, et al. LRG1 promotes angiogenesis by modulating endothelial TGF-β signalling. Nature. 2013;499:306–11.CrossRefPubMed
8.
Zurück zum Zitat Furukawa K, Kawamoto K, Eguchi H, Tanemura M, Tanida T, Tomimaru Y, et al. Clinicopathological Significance of Leucine-Rich α2-Glycoprotein-1 in Sera of Patients With Pancreatic Cancer. Pancreas. 2015;44:93–8.CrossRefPubMed Furukawa K, Kawamoto K, Eguchi H, Tanemura M, Tanida T, Tomimaru Y, et al. Clinicopathological Significance of Leucine-Rich α2-Glycoprotein-1 in Sera of Patients With Pancreatic Cancer. Pancreas. 2015;44:93–8.CrossRefPubMed
9.
Zurück zum Zitat Lindén M, Lind SB, Mayrhofer C, Seqersten U, Wester K, Lyutvinskiy Y, et al. Proteomic analysis of urinary biomarker candidates for nonmuscle invasive bladder cancer. Proteomics. 2012;12:135–44.CrossRefPubMed Lindén M, Lind SB, Mayrhofer C, Seqersten U, Wester K, Lyutvinskiy Y, et al. Proteomic analysis of urinary biomarker candidates for nonmuscle invasive bladder cancer. Proteomics. 2012;12:135–44.CrossRefPubMed
10.
Zurück zum Zitat Andersen JD, Boylan KL, Jemmerson R, Geller MA, Misemer B, Harrington KM, et al. Leucine-rich alpha-2-glycoprotein-1 is upregulated in sera and tumors of ovarian cancer patients. J Ovarian Res. 2010;3:21. Andersen JD, Boylan KL, Jemmerson R, Geller MA, Misemer B, Harrington KM, et al. Leucine-rich alpha-2-glycoprotein-1 is upregulated in sera and tumors of ovarian cancer patients. J Ovarian Res. 2010;3:21.
11.
Zurück zum Zitat Sandanayake NS, Sinclair J, Andreola F, Chapman MH, Xue A, Webster GJ, et al. A combination of serum leucine-rich α-2-glycoprotein 1, CA19-9 and interleukin-6 differentiate biliary tract cancer from benign biliary strictures. Br J Cancer. 2011;105(9):1370–8.PubMedCentralCrossRefPubMed Sandanayake NS, Sinclair J, Andreola F, Chapman MH, Xue A, Webster GJ, et al. A combination of serum leucine-rich α-2-glycoprotein 1, CA19-9 and interleukin-6 differentiate biliary tract cancer from benign biliary strictures. Br J Cancer. 2011;105(9):1370–8.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;199:1420–8.CrossRef Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;199:1420–8.CrossRef
13.
Zurück zum Zitat Bates RC, Mercurio AM. The epithelial-mesenchymal transition (EMT) and colorectal cancer progression. Cancer Biol Ther. 2005;4(4):365–70.CrossRefPubMed Bates RC, Mercurio AM. The epithelial-mesenchymal transition (EMT) and colorectal cancer progression. Cancer Biol Ther. 2005;4(4):365–70.CrossRefPubMed
14.
Zurück zum Zitat Li L, Li W. Epithelial-mesenchymal transition in human cancer: comprehensive reprogramming of metabolism, epigenetics, and differentiation. Pharmacol Ther. 2015;150:33–46.CrossRefPubMed Li L, Li W. Epithelial-mesenchymal transition in human cancer: comprehensive reprogramming of metabolism, epigenetics, and differentiation. Pharmacol Ther. 2015;150:33–46.CrossRefPubMed
15.
Zurück zum Zitat Yang K, Jiang L, Hu Y, Yu J, Chen H, Yao Y, et al. Short hairpin RNA-mediated gene knockdown of FOXM1 inhibits the proliferation and metastasis of human colon cancer cells through reversal of epithelial-to-mesenchymal transformation. J Exp Clin Cancer Res. 2015;34:40.PubMedCentralCrossRefPubMed Yang K, Jiang L, Hu Y, Yu J, Chen H, Yao Y, et al. Short hairpin RNA-mediated gene knockdown of FOXM1 inhibits the proliferation and metastasis of human colon cancer cells through reversal of epithelial-to-mesenchymal transformation. J Exp Clin Cancer Res. 2015;34:40.PubMedCentralCrossRefPubMed
16.
17.
Zurück zum Zitat Kim SH, Xia D, Kim SW, Holla V, Menter DG, Dubois RN. Human enhancer of filamentation 1 is a mediator of hypoxia-inducible factor-1α-mediated migration in colorectal carcinoma cells. Cancer Res. 2010;70(10):4054–63.PubMedCentralCrossRefPubMed Kim SH, Xia D, Kim SW, Holla V, Menter DG, Dubois RN. Human enhancer of filamentation 1 is a mediator of hypoxia-inducible factor-1α-mediated migration in colorectal carcinoma cells. Cancer Res. 2010;70(10):4054–63.PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Yang MH, Wu MZ, Chiou SH, Chen PM, Chang SY, Liu CJ, et al. Direct regulation of TWIST by HIF-1alpha promotes metastasis. Nat Cell Biol. 2008;10:295–305.CrossRefPubMed Yang MH, Wu MZ, Chiou SH, Chen PM, Chang SY, Liu CJ, et al. Direct regulation of TWIST by HIF-1alpha promotes metastasis. Nat Cell Biol. 2008;10:295–305.CrossRefPubMed
19.
Zurück zum Zitat Evans AJ, Russell RC, Roche O, Burry TN, Fish JE, Chow VW, et al. VHL promotes E2 boxdependent E-cadherin transcription by HIF-mediated regulation of SIP1 and snail. Mol Cell Biol. 2007;27:157–69.PubMedCentralCrossRefPubMed Evans AJ, Russell RC, Roche O, Burry TN, Fish JE, Chow VW, et al. VHL promotes E2 boxdependent E-cadherin transcription by HIF-mediated regulation of SIP1 and snail. Mol Cell Biol. 2007;27:157–69.PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Hong SW, Jung KH, Lee HS, Son MK, Yan HH, Kang NS, et al. SB365, Pulsatilla saponin D, targets c-Met and exerts antiangiogenic and antitumor activities. Carcinogenesis. 2013;34(9):2156–69.CrossRefPubMed Hong SW, Jung KH, Lee HS, Son MK, Yan HH, Kang NS, et al. SB365, Pulsatilla saponin D, targets c-Met and exerts antiangiogenic and antitumor activities. Carcinogenesis. 2013;34(9):2156–69.CrossRefPubMed
21.
Zurück zum Zitat Choi JW, Liu H, Shin DH, Yu G, Hwang JS, Kim ES. Proteomic and cytokine plasma biomarkers for predicting progression from colorectal adenoma to carcinoma in human patients. Proteomics. 2013;13:2361–74.CrossRefPubMed Choi JW, Liu H, Shin DH, Yu G, Hwang JS, Kim ES. Proteomic and cytokine plasma biomarkers for predicting progression from colorectal adenoma to carcinoma in human patients. Proteomics. 2013;13:2361–74.CrossRefPubMed
22.
Zurück zum Zitat Zhang Y, Luo Q, Wang N, Hu F, Jin H, Ge T, et al. LRG1 suppresses the migration and invasion of hepatocellular carcinoma cells. Med Oncol. 2015;32:146.CrossRefPubMed Zhang Y, Luo Q, Wang N, Hu F, Jin H, Ge T, et al. LRG1 suppresses the migration and invasion of hepatocellular carcinoma cells. Med Oncol. 2015;32:146.CrossRefPubMed
23.
Zurück zum Zitat Wen SY, Zhang LN, Yang XM, Zhang YL, Ma L, Ge QL, et al. LRG1 is an independent prognostic factor for endometrial carcinoma. Tumour Biol. 2014;35:7125–33.CrossRefPubMed Wen SY, Zhang LN, Yang XM, Zhang YL, Ma L, Ge QL, et al. LRG1 is an independent prognostic factor for endometrial carcinoma. Tumour Biol. 2014;35:7125–33.CrossRefPubMed
24.
Zurück zum Zitat Saito K, Tanaka T, Kanda H, Ebisuno Y, Izawa D, Kawamoto S, et al. Gene expression profiling of mucosal addressin cell adhesion molecule-1+ high endothelial venule cells (HEV) and identification of a leucine-rich HEV glycoprotein as a HEV marker. J Immunol. 2002;168:1050–9.CrossRefPubMed Saito K, Tanaka T, Kanda H, Ebisuno Y, Izawa D, Kawamoto S, et al. Gene expression profiling of mucosal addressin cell adhesion molecule-1+ high endothelial venule cells (HEV) and identification of a leucine-rich HEV glycoprotein as a HEV marker. J Immunol. 2002;168:1050–9.CrossRefPubMed
25.
Zurück zum Zitat Zhong D, He G, Zhao S, Li J, Lang Y, Ye W, et al. LRG1 modulates invasion and migration of glioma cell lines through TGF-β signaling pathway. Acta Histochem. 2015;117:551–8.CrossRefPubMed Zhong D, He G, Zhao S, Li J, Lang Y, Ye W, et al. LRG1 modulates invasion and migration of glioma cell lines through TGF-β signaling pathway. Acta Histochem. 2015;117:551–8.CrossRefPubMed
26.
Zurück zum Zitat Weis SM, Cheresh DA. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med. 2011;17:1359–70.CrossRefPubMed Weis SM, Cheresh DA. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med. 2011;17:1359–70.CrossRefPubMed
27.
Zurück zum Zitat Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science. 1989;246:1306–9.CrossRefPubMed Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science. 1989;246:1306–9.CrossRefPubMed
28.
Zurück zum Zitat Shannon AM, Bouchier-Hayes DJ, Condron CM, Toomey D. Tumour hypoxia, chemotherapeutic resistance and hypoxia-related therapies. Cancer Treat Rev. 2003;29(4):297–307.CrossRefPubMed Shannon AM, Bouchier-Hayes DJ, Condron CM, Toomey D. Tumour hypoxia, chemotherapeutic resistance and hypoxia-related therapies. Cancer Treat Rev. 2003;29(4):297–307.CrossRefPubMed
29.
Zurück zum Zitat Baba Y, Nosho K, Shima K, Irahara N, Chan AT, Meyerhardt JA, et al. HIF1A overexpression is associated with poor prognosis in a cohort of 731 colorectal cancers. Am J Pathol. 2010;176:2292–301.PubMedCentralCrossRefPubMed Baba Y, Nosho K, Shima K, Irahara N, Chan AT, Meyerhardt JA, et al. HIF1A overexpression is associated with poor prognosis in a cohort of 731 colorectal cancers. Am J Pathol. 2010;176:2292–301.PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Rasheed S, Harris AL, Tekkis PP, Turley H, Silver A, McDonald PJ, et al. Hypoxia- inducible factor-1alpha and -2alpha are expressed in most rectal cancers but only hypoxia-inducible factor-1alpha is associated with prognosis. Br J Cancer. 2009;100:1666–73.PubMedCentralCrossRefPubMed Rasheed S, Harris AL, Tekkis PP, Turley H, Silver A, McDonald PJ, et al. Hypoxia- inducible factor-1alpha and -2alpha are expressed in most rectal cancers but only hypoxia-inducible factor-1alpha is associated with prognosis. Br J Cancer. 2009;100:1666–73.PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat Brahimi-Horn C, Mazure N, Pouysségur J. Signalling via the hypoxia-inducible factor-1a requires multiple posttranslational modifications. Cell Signal. 2005;17:1–9.CrossRefPubMed Brahimi-Horn C, Mazure N, Pouysségur J. Signalling via the hypoxia-inducible factor-1a requires multiple posttranslational modifications. Cell Signal. 2005;17:1–9.CrossRefPubMed
32.
Zurück zum Zitat Fukuda R, Hirota K, Fan F, Jung YD, Ellis LM, Semenza GL. Insulin-like growth factor 1 induces hypoxia-inducible factor 1-mediated vascular endothelial growth factorexpression, which is dependent on MAP kinase and phosphatidylinositol 3-kinase signaling in colon cancer cells. J Biol Chem. 2002;277:38205–11.CrossRefPubMed Fukuda R, Hirota K, Fan F, Jung YD, Ellis LM, Semenza GL. Insulin-like growth factor 1 induces hypoxia-inducible factor 1-mediated vascular endothelial growth factorexpression, which is dependent on MAP kinase and phosphatidylinositol 3-kinase signaling in colon cancer cells. J Biol Chem. 2002;277:38205–11.CrossRefPubMed
33.
Zurück zum Zitat Zhong H, Chiles K, Feldser D, Laughner E, Hanrahan C, Georgescu MM, et al. Modulation of HIF-1α expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res. 2000;60:1541–5.PubMed Zhong H, Chiles K, Feldser D, Laughner E, Hanrahan C, Georgescu MM, et al. Modulation of HIF-1α expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res. 2000;60:1541–5.PubMed
34.
Zurück zum Zitat McMahon S, Charbonneau M, Grandmont S, Richard DE, Dubois CM. Transforming Growth Factor 1 Induces Hypoxia-inducible Factor-1 Stabilization through Selective Inhibition of PHD2 Expression. J Biol Chem. 2006;281:171–81. McMahon S, Charbonneau M, Grandmont S, Richard DE, Dubois CM. Transforming Growth Factor 1 Induces Hypoxia-inducible Factor-1 Stabilization through Selective Inhibition of PHD2 Expression. J Biol Chem. 2006;281:171–81.
35.
Zurück zum Zitat Han WQ, Zhu Q, Hu J, Li PL, Zhang F, Li N. Hypoxia-inducible factor prolyl-hydroxylase-2 mediates transforming growth factor beta 1-induced epithelial–mesenchymal transition in renal tubular cells. Biochim Biophys Acta. 1833;2013:1454–62. Han WQ, Zhu Q, Hu J, Li PL, Zhang F, Li N. Hypoxia-inducible factor prolyl-hydroxylase-2 mediates transforming growth factor beta 1-induced epithelial–mesenchymal transition in renal tubular cells. Biochim Biophys Acta. 1833;2013:1454–62.
Metadaten
Titel
LRG1 modulates epithelial-mesenchymal transition and angiogenesis in colorectal cancer via HIF-1α activation
verfasst von
Jingjing Zhang
Lingyin Zhu
Jingyuan Fang
Zhizheng Ge
Xiaobo Li
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2016
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-016-0306-2

Weitere Artikel der Ausgabe 1/2016

Journal of Experimental & Clinical Cancer Research 1/2016 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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