Overexpression of CXCR7 induces angiogenic capacity of human hepatocellular carcinoma cells via the AKT signaling pathway

  • Authors:
    • Yuhui Chen
    • Fei Teng
    • Geying Wang
    • Zhiyu Nie
  • View Affiliations

  • Published online on: August 25, 2016     https://doi.org/10.3892/or.2016.5045
  • Pages: 2275-2281
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Angiogenesis is essential for tumor growth, especially in hepatocellular carcinoma (HCC). The hypervascularity is associated with poor prognosis and highly invasive HCC. The C‑X‑C chemokine receptor type 7 (CXCR7) has been implied overexpressed in many tumor types. Our study aimed to investigate the CXCR7 function in HCC. The tube formation, Transwell migration assay of human umbilical vein endothelial cells (HUVECs) and chicken chorioallantoic membrane (CAM) assay were used. We confirmed that CXCR7 induces angiogenic capacity. Moreover, overexpressing CXCR7 increased the phosphorylated (but not total) AKT expression in HCC cells. Furthermore, overexpressing CXCR7 increased the expression of tumor necrosis factor (TNF)‑α, interleukin (IL)‑6 and IL‑8 in HCC cells. Additionally, inhibition of AKT by LY294002 abrogated CXCR7‑induced angiogenic capacity in HCC cells. Our study suggested that CXCR7 plays an important pro‑angiogenic role in HCC via activation of the AKT pathway. So CXCR7 may be a potential target for anti‑angiogenic therapy in HCC.

Introduction

Angiogenesis, the creation of new blood vessels, is an important effector of many physiological and pathological processes (1). Angiogenesis occurs to induce tumor growth and metastasis, and represents a key hallmark of tumor development (25). Tumor angiogenesis provides the nutrients and oxygen to maintain tumor growth and invasion. Thus, inhibition of tumor angiogenesis may decrease tumor cell growth and spread (68). Angiogenesis is considered to be an effective therapeutic target; hence, angiogenesis is a necessary area of biological research and clinical oncology (4,9,10). Tumor angiogenesis is an outcome of an imbalance between pro-angiogenic factors, including the vascular endothelial growth factor (VEGF) family, and anti-angiogenesis factors, such as endostatin and other related factors (1113). VEGF effects the sprouting and endothelial cell proliferation, and then VEGF can stimulate tumor angiogenesis (14). Many of anti-angiogenesis drugs serve as inhibiting pro-angiogenic factors, such as the monoclonal antibody bevacizumab binds to VEGF, or other small molecules that inhibit the binding of VEGF (15,16). Nevertheless, the mechanisms of tumor angiogenesis are not yet entirely understood and specific, effective inhibitors of angiogenesis are required for cancer therapy.

Hepatocellular carcinoma (HCC) includes >90% of primary malignant liver cancers and is one of the most common reasons for cancer-related mortality (17,18). Because of the hepatitis B virus (HBV) and hepatitis C virus (HCV) infection, the HCC incidence is increasing in Asia, especially in China (19). HCC is regarded as hypervascular, and tumor growth depends on angiogenesis (15,20). Targeting angiogenesis by pharmacologic therapy has been used in many other solid tumors. Therefore, the anti-angiogenic strategy for HCC may increase the treatment outcomes for HCC patients.

C-X-C chemokine receptor type 7 (CXCR7), a new known orphan receptor, has been shown to bind stromal cell-derived factor-1 (SDF-1) (19,21). CXCR7 also has been demonstrated important for primordial germ cell migration in zebra fish (22). Recently, high level of CXCR7 has been confirmed to be associated with aggressive tumors (23). Moreover, overexpression of CXCR7 was found to be connected to metastatic recurrence in non-small cell lung cancer. Recently, overexpression of CXCR7 was reported in tumor cell lines and tissues (24). However, the CXCR7 function in angiogenesis of HCC is not yet clear.

We demonstrated that CXCR7 is highly expressed in HCC cell lines. In addition, overexpression of CXCR7 promoted the angiogenic capacity of HCC cells via ATK signaling pathway. This study demonstrates that CXCR7 may induce angiogenesis in vivo; therefore, CXCR7 may have potential therapeutic effects in HCC.

Materials and methods

Cell lines

The HCC cell lines HCCLM3 (100% lung meta-static potential), MHCC97-L (low meta static potential) and SMMC-7721 (without lung metastatic potential) were purchased from the Chinese Academy of Science (Shanghai, China). The cells were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS) (both from Gibco, Carlsbad, CA, USA) and 100 U penicillin-streptomycin (Invitrogen, Carlsbad, CA, USA) in a 5% CO2 humidified incubator at 37°C.

Real-time reverse transcription-PCR analyses

The CXCR7 mRNA expression was calculated as follows. Reactions were executed in 20 µl volumes, every sample including 2 µl complementary DNA (cDNA) by using the primer pairs: CXCR7 sense, 5′-GGGATGCAGCGGATAGTCAA-3′ and antisense, 5′-CGGTCGTTGTCCACATCCA-3′; Taqman probe, 5′-TCGGTCTCTCCCTGCCCGTCCT-3′. Real-time reverse transcription-PCR used TaqMan PCR reagents and the ABI PRISM 7700 Sequence Detection System (Applied Biosystems, Foster City, CA, USA) followed with an initial denaturation step at 95°C for 10 min, then 28 cycles of denaturation at 95°C for 60 sec, primer annealing at 58°C for 30 sec and primer extension at 72°C for 30 sec, with a final extension step at 72°C for 5 min.

Western blotting

Western blotting was performed by using the Bio-Rad Transfer Cell System (Bio-Rad, Mississauga, ON, Canada). Rabbit anti-human CXCR7 antibody (1:200; R&D Systems, Inc., Minneapolis, MN, USA) followed by 1:3,000 horseradish peroxidase-conjugated goat anti-rabbit IgG F(ab′)2 antibody (Jackson ImmunoResearch, West Grove, PA, USA) was used.

Overexpression of CXCR7 in HCC cells

The HCC cell line HCCLM3 was transformed with human full-length CXCR7 cDNA by using Lipofectamine 2000 (Invitrogen). The HCC cell line HCCLM3 transformed with an empty plasmid was used as a negative control. Stable cell lines expressing CXCR7 were selected with G418 (K1, 250 µg/ml; TPC-1, 200 µg/ml; and B-CPAP, 300 µg/ml). The CXCR7 expression was evaluated by western blotting.

RNA interference (RNAi) in HCC cells

Downregulation of the expression of CXCR7 in HCCLM3 cells was performed using small interfering RNAs (siRNAs) as follows: siCXCR7-286, 5′-CGCUCUCCUUCAUUUACAUdTdT-3′ (at position 286); negative control siRNA, 5′-UUCUCCGAACGUGUCACGUTT-3′; glyceraldehyde 3-phosphate dehydrogenase (GAPDH) positive control siRNA, 5′-GUAUGACAACAGCCUCAAGTT-3′. siRNA transfection of HCCLM3 cells was performed using the protocol.

Human umbilical vein endothelial cell (HUVEC) tube formation assay

The HUVEC tube formation assay was performed as previously described. Firstly, 200 µl Matrigel were placed into a 96-well plate for 30 min at 37°C. HUVECs (2×104) in 200 µl conditioned medium (CM) from indicated HCC cells were added to the well and incubated for 24 h at 37°C. Images were attained by a bright-field microscope (×100), and formation of capillary tubes was quantified by measuring their total number of each image.

Chicken chorioallantoic membrane (CAM) assay

The CAM assay was performed using 8-day-old chicken embryo. About 1-cm diameter window was shaped in the shell of chicken embryo. A diameter gelatin sponge with 100 µl CM harvested from the indicated HCC cells was placed on the CAM. The windows in the chicken embryo were closed by bandages. The chicken embryos were incubated at 37°C for 48 h. Then the CAM was fixed with stationary solution (1:1 v/v mixture of methanol and acetone) for 15 min, the CAM was imaged by a digital camera. The number of second- and third-order vessels in the test samples was compared to control.

HUVEC Transwell migration assay

HUVECs (1×104) were cultured on the top of polycarbonate Transwell filters (pore size, 8.0 µm; Corning, Inc., Corning, NY, USA) in CM containing 5% FBS. The lower chamber was filled with 500 µl of media containing SDF-1α (100 ng/ml) and 15% FBS. The cells were incubated at 37°C for 8 h, and the cells that migrated to the lower membrane surface were fixed in 4% paraformaldehyde, stained using hematoxylin for 15 min, and the number of cells in 10 randomly selected ×200 fields of view per filter was counted and expressed relative to that of cells treated with CM from vector control cells.

Matrigel plug assay in mice

C57BL/6 mice were injected 50 µl of reconstituted CM with 50 U/ml heparin and SDF-1α (100 ng/ml) added to 0.6 ml Matrigel. Matrigel polymerizes to a solid gel at body temperature, and then the gel become vascularized after 10 days. The gel was removed, photographed, stained, and also diluted in water to measure the hemoglobin content by Drabkin's reagent kit (Sigma).

Enzyme-linked immunosorbent assay (ELISA)

The tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-8 ELISA assay were performed by the commercial kits. All samples were added to the 96-well in triplicate, incubated at 36°C for 90 min, washed, incubated with a specific anti-antibody (Cell Signaling Technology, Inc.) at 36°C for 1 h, washed, incubated with secondary antibody at 36°C for 1 h, and then substrate was added, incubated for 1 h and the absorbance values were read at OD450 by an ELISA plate reader.

Statistical analysis

All experimental data are presented as the mean ± SD of three independent biological replicates. Statistical analyses were performed using SPSS 13.0 (IBM Corp., Armonk, NY, USA). Analysis of variance (ANOVA) was used to evaluate the significance of the differences between two groups. P≤0.05 was considered statistically significant.

Results

CXCR7 is upregulated in HCC cell lines

Western blotting and qRT-PCR assays demonstrated CXCR7 protein and mRNA expression in HCCLM3 cells. The qRT-PCR showed that CXCR7 mRNA level was obviously increased in HCCLM3 compared to the SMMC-7721 (Fig. 1A). Consistent with the mRNA results, the protein level also was significantly upregulated in HCCLM3 compared to the SMMC-7721 (Fig. 1B). The data implied that CXCR7 is overexpressed in HCC cells.

Overexpression of CXCR7 promotes the angiogenic capacity in HCC cells

Firstly, overexpression of CXCR7 in the HCCLM3 was confirmed by western blotting (Fig. 2A). The effect of CXCR7 inducing angiogenesis in the HCCLM3 was investigated by tube formation assay. CM from CXCR7 overexpression in HCCLM3 notably induced the tube formation compared to the control (Fig. 2B). In addition, CXCR7 overexpressing in HCCLM3 significantly induced the HUVEC migration (Fig. 2C). Moreover, CM from CXCR7 overexpression of HCCLM3 cells increased the second- and third-order vessel number in the CAM (Fig. 2D). The results together indicated, that CXCR7 induced the angiogenesis capacity of HCCLM3 cells in vitro.

Silencing CXCR7 reduces the angiogenic capacity in HCC cells

To better confirm the effect of CXCR7 on angiogenesis in the HCC progression, CXCR7 was knocked down in the HCCLM3 cells and was confirmed by western blotting (Fig. 3A). Compared to the control, CM from CXCR7 knockdown cells decreased HUVEC tube formation (Fig. 3B) and migration (Fig. 3C). CM from CXCR7 knockdown cells also decreased the second- and third-order vessel number in the CAM assay (Fig. 3D). These data illustrated that CXCR7 is involved in angiogenesis of HCCLM3 cells.

Overexpression of CXCR7 induces the angiogenic capacity in Matrigel plug assay

Vascularization was calculated by Matrigel plug assay (Fig. 4A). The C57BL/6 mice were injected with Matrigel containing aliquots of control, overexpressing or silenced CXCR7 in HCCLM3, plus heparin (50 U/ml) and SDF-1α (100 ng/ml). As shown in Fig. 4B and C, the relative hemoglobin content and vessel number showed that angiogenesis was inhibited by CM containing CXCR7-silenced HCCLM3, whereas it was partially promoted by CM containing overexpressing CXCR7.

CXCR7 promotes the angiogenic capacity of HCC cells via activating the AKT signaling pathway

The CXCR7 affected the angiogenic capacity of HCCLM3 via activating AKT signaling. We used the AKT signaling inhibitor LY294002. The stimulatory effects of CM derived from CXCR7 overexpressing HCCLM3 on HUVEC tube formation (Fig. 5A), migration (Fig. 5B) and the second- and third-order vessel number in the CAM assay (Fig. 5C) was significantly reduced by using LY294002. Taken all together, these results suggested that CXCR7 enhances the angiogenic capacity of HCCLM3 cells via activation of the AKT signaling pathway. The expression of AKT target protein, including TNF-α, IL-6 and IL-8, was induced in CXCR7 overexpressing HCCLM3 and reduced in CXCR7 knockdown HCCLM3 (Fig. 5D). Moreover, western blotting demonstrated that overexpression of CXCR7 upregulated the phosphorylated AKT expression but did not significantly impact the total AKT protein expression (Fig. 6). These data implied that the AKT pathway may trigger angiogenesis of CXCR7 in HCCLM3 cells.

Discussion

There is abundant evidence to show that CXCR7 overexpression is involved in HCC progress, including breast, lung, prostate and pancreatic cancers (23,25). It has been demonstrated that knockdown of CXCR7 expression significantly reduces SMMC-7721 cell invasion, adhesion and angiogenesis. Moreover, decreased CXCR7 expression inhibited tumor growth in a mouse model of HCC. The highly specific CXCR7 antagonist CCX771 also was used. In the past report, CXCL12 induced Huh7, SNU449 and SNU475 cell migration, and the migration was blocked by CCX771/anti-CXCR7. Similarly, the CCX771 significantly inhibited glioma cell proliferation and invasion (26). The study provides new insights into the significance of CXCR7 in invasion and angiogenesis of tumors (27,28). Although the study shows the importance of CXCR7 in HCC invasion, angiogenesis and tumor growth, the role of CXCR7 effect the HUVEC function in tumor microenvironment is not fully established. In the present study, CXCR7 was confirmed to be involved in metastatic HCC. Both the past studies and our data showed overexpression of CXCR7 in a highly metastatic HCC. Also, increased expression of CXCR7 in HCCLM3 induced the angiogenic capacity in vitro. Furthermore, silence of CXCR7 decreased the angiogenic capacity of HUVECs. The data showed that CXCR7 may induce angiogenesis via activating the AKT signaling pathway. Collectively the findings suggested the potential effect of CXCR7 in angiogenic capacity of HCC cells.

The overexpression of CXCR7 suggested several potential unknown functions. Although substantial research has suggested that CXCR7 may serve as an oncogene in many tumor types, the molecular mechanism of CXCR7 has not been accurately demonstrated. In our study, we found that the overexpression of CXCR7 induced the tube formation and migration of HUVECs and significantly induced the second- and third-order vessel number of CAM. The data showed the potential effect of CXCR7 angiogenic activities in HCC cells. Furthermore, overexpression of CXCR7 notably enhanced the AKT pathway activity, suggesting that phosphorylated AKT plays a key role in the CXCR7-induced angiogenesis of HCC progression. AKT signaling pathway is known to regulate inflammatory responses, and other physiological and pathological functions including cancer progression. The findings suggested that CXCR7 induces HCC angiogenesis via activating the AKT signaling pathway.

Additionally, overexpression of CXCR7 induced TNF-α, IL-6, and IL-8 expression. TNF-α promoted angiogenesis and regulated blood vessel remodeling in vivo; IL-6 and IL-8 also promoted VEGF expression and tumor angiogenesis. Increasing and compelling epidemiological evidence has been suggested that the inflammatory microenvironment plays a key role in tumor cell proliferation, angiogenesis and metastasis (2931). TNF-α is known as a key regulator of inflammation-related cancer, including HCC (32). IL-6 is necessary for HCC progression in animal models, with hepatic-associated macro-phages representing a major paracrine IL-6 expression during HCC development and autocrine IL-6 inducing notably HCC initiation (3335). IL-8 induced HCC grade, metastasis and recurrence (36,37). It would be worth exploring the TNF-α, IL-6, and IL-8 functions in HCC angiogenesis. The TNF-α, IL-6, and IL-8 expression in HCC cells remained to be clarified and should be investigated further.

In conclusion, our study indicated that CXCR7 is upregulated and induces angiogenic capacity in HCC by activation of the AKT pathway. These data may give new insight into the angiogenesis mechanism in HCC; and CXCR7 may be a new therapeutic target for HCC.

Acknowledgments

This study was supported by grants (grant nos. 81400961 and 81301157) of the National Natural Science Foundation of China (NSFC).

References

1 

Carmeliet P: Angiogenesis in life, disease and medicine. Nature. 438:932–936. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Folkman J: Role of angiogenesis in tumor growth and metastasis. Semin Oncol. 29(Suppl 16): 15–18. 2002. View Article : Google Scholar

3 

Varinska L, Gal P, Mojzisova G, Mirossay L and Mojzis J: Soy and breast cancer: Focus on angiogenesis. Int J Mol Sci. 16:11728–11749. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Herbst RS and Fidler IJ: Angiogenesis and lung cancer: Potential for therapy. Clin Cancer Res. 6:4604–4606. 2000.

5 

Onishi M, Ichikawa T, Kurozumi K and Date I: Angiogenesis and invasion in glioma. Brain Tumor Pathol. 28:13–24. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Pollitt MJ, Hanby AM, Horgan K, Murphy CE, Jones PF and Speirs V: Angiogenesis in breast cancer: How should we measure this? (Review). Oncol Rep. 13:931–936. 2005.PubMed/NCBI

7 

Herbst RS, Onn A and Sandler A: Angiogenesis and lung cancer: Prognostic and therapeutic implications. J Clin Oncol. 23:3243–3256. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Sato Y: Molecular diagnosis of tumor angiogenesis and anti-angiogenic cancer therapy. Int J Clin Oncol. 8:200–206. 2003. View Article : Google Scholar : PubMed/NCBI

9 

van Hinsbergh VW, Collen A and Koolwijk P: Angiogenesis and anti-angiogenesis: Perspectives for the treatment of solid tumors. Ann Oncol. 10(Suppl 4): 60–63. 1999. View Article : Google Scholar : PubMed/NCBI

10 

Duarte IG, Bufkin BL, Pennington MF, Gal AA, Cohen C, Kosinski AS, Mansour KA and Miller JI: Angiogenesis as a predictor of survival after surgical resection for stage I non-small-cell lung cancer. J Thorac Cardiovasc Surg. 115:652–658; discussion 658–659. 1998. View Article : Google Scholar : PubMed/NCBI

11 

Folkman J: Endogenous inhibitors of angiogenesis. Harvey Lect. 92:65–82. 1997.

12 

Gyenge M, Amagase K, Kunimi S, Matsuoka R and Takeuchi K: Roles of pro-angiogenic and anti-angiogenic factors as well as matrix metalloproteinases in healing of NSAID-induced small intestinal ulcers in rats. Life Sci. 93:441–447. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Marjon PL, Bobrovnikova-Marjon EV and Abcouwer SF: Expression of the pro-angiogenic factors vascular endothelial growth factor and interleukin-8/CXCL8 by human breast carcinomas is responsive to nutrient deprivation and endoplasmic reticulum stress. Mol Cancer. 3:42004. View Article : Google Scholar : PubMed/NCBI

14 

Hellberg C, Ostman A and Heldin CH: PDGF and vessel maturation. Recent Results Cancer Res. 180:103–114. 2010. View Article : Google Scholar

15 

Alfaro C, Suarez N, Gonzalez A, Solano S, Erro L, Dubrot J, Palazon A, Hervas-Stubbs S, Gurpide A, Lopez-Picazo JM, et al: Influence of bevacizumab, sunitinib and sorafenib as single agents or in combination on the inhibitory effects of VEGF on human dendritic cell differentiation from monocytes. Br J Cancer. 100:1111–1119. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Thompson Coon J, Hoyle M, Green C, Liu Z, Welch K, Moxham T and Stein K: Bevacizumab, sorafenib tosylate, sunitinib and temsirolimus for renal cell carcinoma: A systematic review and economic evaluation. Health Technol Assess. 14:1–184. iii–iv. 2010. View Article : Google Scholar

17 

Willatt JM, Francis IR, Novelli PM, Vellody R, Pandya A and Krishnamurthy VN: Interventional therapies for hepatocellular carcinoma. Cancer Imaging. 12:79–88. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Zhu AX, Duda DG, Sahani DV and Jain RK: HCC and angiogenesis: Possible targets and future directions. Nat Rev Clin Oncol. 8:292–301. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Burns JM, Summers BC, Wang Y, Melikian A, Berahovich R, Miao Z, Penfold ME, Sunshine MJ, Littman DR, Kuo CJ, et al: A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. J Exp Med. 203:2201–2213. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Finn RS and Zhu AX: Targeting angiogenesis in hepatocellular carcinoma: Focus on VEGF and bevacizumab. Expert Rev Anticancer Ther. 9:503–509. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Balabanian K, Lagane B, Infantino S, Chow KY, Harriague J, Moepps B, Arenzana-Seisdedos F, Thelen M and Bachelerie F: The chemokine SDF-1/CXCL12 binds to and signals through the orphan receptor RDC1 in T lymphocytes. J Biol Chem. 280:35760–35766. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Boldajipour B, Mahabaleshwar H, Kardash E, Reichman-Fried M, Blaser H, Minina S, Wilson D, Xu Q and Raz E: Control of chemokine-guided cell migration by ligand sequestration. Cell. 132:463–473. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Wang J, Shiozawa Y, Wang J, Wang Y, Jung Y, Pienta KJ, Mehra R, Loberg R and Taichman RS: The role of CXCR7/RDC1 as a chemokine receptor for CXCL12/SDF-1 in prostate cancer. J Biol Chem. 283:4283–4294. 2008. View Article : Google Scholar

24 

Zheng K, Li HY, Su XL, Wang XY, Tian T, Li F and Ren GS: Chemokine receptor CXCR7 regulates the invasion, angiogenesis and tumor growth of human hepatocellular carcinoma cells. J Exp Clin Cancer Res. 29:312010. View Article : Google Scholar : PubMed/NCBI

25 

Iwakiri S, Mino N, Takahashi T, Sonobe M, Nagai S, Okubo K, Wada H, Date H and Miyahara R: Higher expression of chemokine receptor CXCR7 is linked to early and metastatic recurrence in pathological stage I nonsmall cell lung cancer. Cancer. 115:2580–2593. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Liu Y, Carson-Walter E and Walter KA: Targeting chemokine receptor CXCR7 inhibits glioma cell proliferation and mobility. Anticancer Res. 35:53–64. 2015.PubMed/NCBI

27 

Lin L, Han MM, Wang F, Xu LL, Yu HX and Yang PY: CXCR7 stimulates MAPK signaling to regulate hepatocellular carcinoma progression. Cell Death Dis. 5:e14882014. View Article : Google Scholar : PubMed/NCBI

28 

Xue TC, Jia QA, Bu Y, Chen RX, Cui JF, Tang ZY and Ye SL: CXCR7 correlates with the differentiation of hepatocellular carcinoma and suppresses HNF4α expression through the ERK pathway. Oncol Rep. 32:2387–2396. 2014.PubMed/NCBI

29 

Balkwill F and Mantovani A: Inflammation and cancer: Back to Virchow? Lancet. 357:539–545. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Castello G, Scala S, Palmieri G, Curley SA and Izzo F: HCV-related hepatocellular carcinoma: From chronic inflammation to cancer. Clin Immunol. 134:237–250. 2010. View Article : Google Scholar

31 

Karin M and Greten FR: NF-kappaB: Linking inflammation and immunity to cancer development and progression. Nat Rev Immunol. 5:749–759. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Feldmann M: Development of anti-TNF therapy for rheumatoid arthritis. Nat Rev Immunol. 2:364–371. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Maeda S, Hikiba Y, Sakamoto K, Nakagawa H, Hirata Y, Hayakawa Y, Yanai A, Ogura K, Karin M and Omata M: Ikappa B kinasebeta/nuclear factor-kappaB activation controls the development of liver metastasis by way of interleukin-6 expression. Hepatology. 50:1851–1860. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Naugler WE, Sakurai T, Kim S, Maeda S, Kim K, Elsharkawy AM and Karin M: Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science. 317:121–124. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Park EJ, Lee JH, Yu GY, He G, Ali SR, Holzer RG, Osterreicher CH, Takahashi H and Karin M: Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell. 140:197–208. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Ren Y, Poon RT, Tsui HT, Chen WH, Li Z, Lau C, Yu WC and Fan ST: Interleukin-8 serum levels in patients with hepatocellular carcinoma: Correlations with clinicopathological features and prognosis. Clin Cancer Res. 9:5996–6001. 2003.PubMed/NCBI

37 

Welling TH, Fu S, Wan S, Zou W and Marrero JA: Elevated serum IL-8 is associated with the presence of hepatocellular carcinoma and independently predicts survival. Cancer Invest. 30:689–697. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 36 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen Y, Teng F, Wang G and Nie Z: Overexpression of CXCR7 induces angiogenic capacity of human hepatocellular carcinoma cells via the AKT signaling pathway. Oncol Rep 36: 2275-2281, 2016
APA
Chen, Y., Teng, F., Wang, G., & Nie, Z. (2016). Overexpression of CXCR7 induces angiogenic capacity of human hepatocellular carcinoma cells via the AKT signaling pathway. Oncology Reports, 36, 2275-2281. https://doi.org/10.3892/or.2016.5045
MLA
Chen, Y., Teng, F., Wang, G., Nie, Z."Overexpression of CXCR7 induces angiogenic capacity of human hepatocellular carcinoma cells via the AKT signaling pathway". Oncology Reports 36.4 (2016): 2275-2281.
Chicago
Chen, Y., Teng, F., Wang, G., Nie, Z."Overexpression of CXCR7 induces angiogenic capacity of human hepatocellular carcinoma cells via the AKT signaling pathway". Oncology Reports 36, no. 4 (2016): 2275-2281. https://doi.org/10.3892/or.2016.5045