Open Access

SDF1α/CXCR4 axis may be associated with the malignant progression of gastric cancer in the hypoxic tumor microenvironment

  • Authors:
    • Masakazu Yashiro
    • Haruhito Kinoshita
    • Gen Tsujio
    • Tatsunari Fukuoka
    • Yurie Yamamoto
    • Tomohiro Sera
    • Atsushi Sugimoto
    • Sadaaki Nishimura
    • Shuhei Kushiyama
    • Shingo Togano
    • Kenji Kuroda
    • Takahiro Toyokawa
    • Masaichi Ohira
  • View Affiliations

  • Published online on: November 12, 2020     https://doi.org/10.3892/ol.2020.12299
  • Article Number: 38
  • Copyright: © Yashiro et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Stromal cell‑derived factor 1α (SDF1α) and its receptor C‑X‑C chemokine receptor type 4 (CXCR4) have been reported to form an important chemokine signaling pathway. Our previous study reported that SDF1α from tumor stromal cells may stimulate the proliferation of gastric cancer (GC) cells through the CXCR4 axis in a hypoxic microenvironment. However, a limited number of studies have addressed the clinicopathological significance of the expression of SDF1α and CXCR4 in GC, particularly at hypoxic regions. Immunohistochemistry was used to investigate the expression levels of SDF1α, CXCR4 and the hypoxic marker carbonic anhydrase 9 (CA9) in 185 patients with stage II and III GC. The results demonstrated that CA9 was expressed on cancer and stromal cells in hypoxic lesions, CXCR4 was mainly expressed in cancer cells, and SDFα was mainly expressed in stromal cells. CXCR4 expression in cancer cells and SDFα expression in stromal cells were associated with the hypoxic regions with CA9 expression. The CA9 and CXCR4 expression in the cancer cells, and the SDF1α expression in the stromal cells (CA9/CXCR4/SDF1α) was significantly associated with macroscopic type 4 tumor (P=0.012) and the pattern of tumor infiltration into the surrounding tissue (P<0.001). The prognosis of the all CA9/CXCR4/SDF1α‑positive patients was significantly poorer compared with that of patients with CA9‑, CXCR4‑ or SDF1α‑negative GC at Stage III (P=0.041). These results indicated that hypoxia may upregulate SDFα production in stromal cells and CXCR4 expression in cancer cells. The SDF1α/CXCR4 axis may serve an important role in the progression of GC.

Introduction

Gastric cancer (GC) has the fifth highest cancer incidence and second highest rate of cancer-associated mortalities among all malignant neoplasms worldwide (1). Although curative resection (R0) with lymph node dissection and adjuvant chemotherapy has prolonged the survival of patients with GC, the recurrence rate of R0 cases remains at ~30% in patients with stage II/III GC (2). Peritoneal recurrence is the most frequent recurrence pattern in patients with GC following curative resection, and as such, peritoneal recurrence is the most common cause of subsequent cancer-associated mortality (3).

Stromal cell-derived factor 1α (SDF1α, also termed CXC ligand 12) and its receptor C-X-C chemokine receptor type 4 (CXCR4) have been known to serve a critical role in cancer cell migration and proliferation in solid tumors, including GC (4,5), breast (6), esophageal (7), prostate tumor (8), pancreatic cancer (9,10), melanoma (11), colon (12), ovarian (13) and lung cancer (14).

Various types of solid tumors, including GC have a heterogeneously hypoxic environment which is currently thought to be associated with aggressive tumor phenotypes (1519). Clinical and experimental data on GC also provide evidence of an association between the hypoxic environment and a poor prognosis (16,18). Therefore, a hypoxic environment has been considered to be associated with aggressive tumor phenotypes of gastric carcinomas (20,21), including the metastatic ability of cancer cells (22).

Our recent study reported that the progression of GC may be recognized as the product of evolving crosstalk between the cancer cells and their surrounding tumor stroma (23,24). The results of our previous study reported that SDF1 from tumor stromal cells may stimulate the proliferation of GC cells through the CXCR4 axis in hypoxic microenvironments (4). Certain studies also reported that the expression of CXCR4 in cancer cells has been upregulated under hypoxia (25,26). However, the clinical association between the expression of SDF1α/CXCR4 and hypoxic conditions in GC has been unclear. The present study investigated the clinicopathological significance of SDF1α and CXCR4 expression and a hypoxic environment in GC at stage II and III.

Materials and methods

Clinical materials

Human GC tissues were obtained from a total of 185 patients with stage II or III GC, who had undergone resection of a primary GC at Osaka City University Hospital. Patients with stage I or stage IV GC were excluded. None of the patients had undergone preoperative radiation and/or chemotherapy. The pathological diagnoses and classifications were made according to classified by the Japanese Classification of Gastric Carcinoma 3rd English edition (27) or the Union for International Cancer Control Tumor-Node-Metastasis classification of malignant tumors (28). Table I shows the clinicopathological characteristics of 185 patients with stage II and III GC. The study protocol conformed to the ethical guidelines of the Declaration of Helsinki (29). The present study was conducted with the approval of the Ethical Committee of Osaka City University (reference number 924). Written informed consent was obtained from all patients prior to treatment.

Table I.

Clinicopathological features of 185 patients with stage II or III gastric cancer.

Table I.

Clinicopathological features of 185 patients with stage II or III gastric cancer.

Clinicopathological featuren (n=185)
Sex
  Female76
  Male109
Age, years
  <70102
  ≥7083
Macroscopic type
  Type 423
  Other162
Histological type
  Intestinal85
  Diffuse100
Infiltration pattern
  a/b123
  c58
Lymph node metastasis
  Negative44
  Positive141
Stage78
  II
  III107
Lymphatic invasion
  Negative29
  Positive155
Venous invasion
  Negative130
  Positive55
Immunohistochemical techniques

The GC tissue was preserved by fixing in a solution of 10% neutral-buffered formalin for ~24 h at room temperature. Immunohistochemical staining was performed on 4-µm sections of formalin-fixed paraffin-embedded tissue. The slides were deparaffinized in xylene and rehydrated in decreasing concentrations of ethyl alcohol. The sections were heated for 10 min at 105°C by autoclave in Target Retrieval Solution (Dako; Agilent Technologies, Inc.). The sections were blocked for 10 min at room temperature with 10% normal goat serum (Histofine Simple Stain™ MAX-PO; Nichirei Biosciences Inc.) and subsequently incubated with 3% hydrogen peroxide to block endogenous peroxidase activity. Immunohistochemistry was performed using the following antibodies: Anti-CXCR4 (cat. no. ab124824; dilution 1:100; Abcam), anti-SDF1α (cat. no. MAB350; dilution 1:200; R&D Systems, Inc.), and a hypoxic marker, carbonic anhydrase 9 (CA9; clone; cat. no. M75; dilution 1:1,000; Novus Biologicals, LLC). The specimens were incubated with the antibodies at 4°C overnight, followed by three washes with PBS. The slides were treated with streptavidin-peroxidase reagent and were incubated in PBS diaminobenzidine and 1% hydrogen peroxide vol/vol, followed by counterstaining with Mayer's hematoxylin for 1 min at room temperature and analysis of three fields per sample under a light microscope (magnification, ×100).

Immunohistochemical determination of SDF1α, CXCR4 and CA9

Positive immunostaining was evaluated by two independent investigators who were blinded to patient outcomes and clinicopathological features. A numerical scoring system with two categories was used to assess the intensity and the extent of immunoreactivity. The proportion score was an estimate of the proportion of positive cells: 0, no immunoreactive cells; 1, <20% immunoreactive cells; 2, 20–50% immunoreactive cells; and 3, ≥50% immunoreactive cells. The intensity score estimates the average staining intensity of positive tumor cells: 0, no staining; 1, weak positive membrane staining; 2, moderate; and 3, strong staining. The two scores were multiplied together to give a final numerical score ranging between 0 and 9. The cases were considered positive if the score was 5 or more.

Statistical analysis

The χ2 test or Fisher's exact test were used to determine the significance of the difference between the covariates. Survival curves were constructed using Kaplan-Meier survival analysis and compared using the log-rank test. The influence of each prognostic factor on patient survival was evaluated using Cox regression analysis. All analyses were performed using SPSS software version 22.0 (IBM Corp.). P<0.05 was considered to indicate a statistically significant difference.

Results

Association between clinicopathological features and CA9 expression, CXCR4 expression in cancer cells, and SDF1α expression in the stromal cells

Representative images of CA9, CXCR4 and SDF1α immunostaining are presented in Fig. 1. CA9 was heterogeneously expressed on stromal cells (arrows) and GC cells (arrowheads). SDF1α expression in stromal cells was observed primarily in the cytoplasm of fibroblast-like stromal cells (arrows). CXCR4 expression was observed primarily in cancer cells (arrowheads). CA9 expression was significantly associated with CXCR4 expression in the cancer cells and SDF1α expression in the stromal cells (P=0.001), and was significantly associated with macroscopic type 4 tumor (P=0.021), and a pattern of tumor infiltration into the surrounding tissue (P=0.005). CA9 expression, CXCR4 expression in the cancer cells, and SDF1α expression in the stromal cells (CA9/CXCR4/SDF1α) were significantly associated with macroscopic type 4 (P=0.012) and a pattern of tumor infiltration into the surrounding tissue (P<0.001; Table II).

Table II.

Association between clinicopathological features and CA9/CXCR4/SDF1α expression in stage II and III gastric cancer.

Table II.

Association between clinicopathological features and CA9/CXCR4/SDF1α expression in stage II and III gastric cancer.

CA9 expression CA9/CXCR4/SDF1α expression


FactorsPositive n=96 (%)Negative n=89 (%)P-valuePositive n=20 (%)Negative n=165 (%)P-value
Age, years
  ≥7041 (49.4)42 (50.6)0.6329 (10.8)74 (89.2)1.000
  <7054 (52.9)48 (47.1) 11 (10.8)91 (89.2)
Sex
  Female32 (42.1)44 (57.9)0.03610 (13.2)66 (86.8)0.391
  Male63 (57.8)46 (42.2) 10 (9.2)99 (90.8)
Macroscopic type
  Type 417 (73.9)6 (26.1)0.0216 (26.1)17 (73.9)0.012
  Other78 (48.1)84 (51.9) 14 (8.6)165 (91.4)
Tumor size, mm
  ≥5060 (52.6)54 (47.4)0.65910 (8.8)104 (91.2)0.258
  <5035 (49.3)36 (50.7) 10 (14.1)61 (85.9)
Histological type
  Diffuse58 (58.0)42 (42)0.05014 (14)86 (86)0.508
  Intestinal37 (43.5)48 (56.5) 6 (7.1)79 (92.9)
aInfiltration pattern
  INF a/b53 (43.1)70 (56.9)0.0057 (5.7)116 (94.3)<0.001
  INF c38 (65.5)20 (34.5) 12 (20.7)46 (79.3)
Stage
  II35 (44.9)43 (55.1)0.1328 (13.0)70 (87.0)0.836
  III60 (56.1)47 (43.9) 12 (26.8)95 (73.2)
Lymph node metastasis
  Positive73 (51.8)68 (48.2)0.83715 (10.6)126 (89.4)0.727
  Negative22 (50.0)22 (50.0) 5 (11.4)39 (88.6)
Lymphatic invasion
  Positive76 (49.0)79 (51.0)0.10315 (9.7)140 (90.3)0.230
  Negative19 (65.5)10 (34.5) 5 (17.2)24 (82.8)
Venous invasion
  Positive28 (50.9)27 (49.1)0.9385 (9.1)50 (90.9)0.624
  Negative67 (51.5)63 (48.5) 15 (11.5)115 (88.5)
CXCR4/SDF1α expression
  Positive20 (83.3)4 (16.7)0.001
  Negative75 (46.6)86 (53.4)

a INF, pattern of tumor infiltration into the surrounding tissue. The predominant pattern of infiltrating growth into the surrounding tissue is classified as follows; INF a, the tumor shows expanding growth and a distinct border with the surrounding tissue; INF b, this category is between INF a and INF b; INF c, the tumor shows infiltrating growth and an indistinct border with the surrounding tissue. SDF1α, stromal cell-derived factor 1α; CXCR4, C-X-C chemokine receptor type 4.

Survival analysis

Fig. 2 shows the Kaplan-Meier survival curve for all 185 patients according to CXCR4 and SDF1α expression. The patients who were positive for all CA9, CXCR4, and SDF1α were defined as the CA9/CXCR4/SDF1α-positive group, whereas the those who were negative for CA9, CXCR4 or SDF1α were termed CA9/CXCR4/SDF1α-negative. The prognosis of the CA9/CXCR4/SDF1α-positive group tended to be poorer compared with that of the CA9/CXCR4/SDF1α-negative patients with stage II or III GC (Fig. 2A; P=0.0826). The prognosis of patients with stage II GC was not different between the all CA9/CXCR4/SDF1α-positive and -negative groups (Fig. 2B). By contrast, the prognosis of the CA9/CXCR4/SDF1α-positive group was significantly poorer compared with that of the CA9/CXCR4/SDF1α-negative patients with stage III GC (Fig. 2C; P=0.041).

As presented in Table III, the univariate analysis revealed that CA9/CXCR4/SDF1α, age, macroscopic type and tumor size were each significantly associated with a poor prognosis. The multivariate analysis revealed that macroscopic type was independent prognostic factor, whereas CA9/CXCR4/SDF1α expression was not.

Table III.

Univariate and multivariate analysis with respect to overall survival in gastric cancer.

Table III.

Univariate and multivariate analysis with respect to overall survival in gastric cancer.

Univariate analysisMultivariate analysis


VariablesHazard ratio95% CIP-valueHazard ratio95% CIP-value
CA9/CXCR4/SDF1α
  Either negative vs. all positive1.8601.094–3.1630.0221.5830.925–2.7100.094
Age, years
  >70 vs. <701.6591.050–2.6220.0301.3790.852–2.2330.191
Sex
  Female vs. male1.1790.733–1.8960.497
Macroscopic type
  Type 4 vs. other types3.7792.219–6.434<0.0012.6851.475–4.8860.001
Tumor size, mm
  <50 vs. ≥502.3851.414–4.024<0.0011.5930.894–2.8370.114
Histological type
  Intestinal vs. diffuse1.3410.840–2.1410.219
Lymphatic invasion
  Negative vs. positive1.7790.816–3.8800.147

[i] CI, confidence interval; CXCR4, C-X-C chemokine receptor type 4; SDF1α, stromal cell-derived factor 1α.

Discussion

CA9 is upregulated under hypoxic conditions through the upregulation and stabilization of hypoxia-inducible factor 1α (HIF-1α), which binds to the hypoxia-responsive element present in the promoter regions of CA9 (30). Therefore, CA9 was considered to indicate hypoxic loci, and was used as a hypoxic marker in the present study.

SDF1α was expressed in GC cells and stromal cells, as previously reported (31,32). In the GC microenvironment, SDF1α expression was observed mainly in the cytoplasm of fibroblast-like stromal cells, particularly frequently in the macroscopic type 4 or diffuse-type GC with abundant stromal cells. By contrast, the SDF1α expression on the cancer cells was observed primarily at the cell membrane. The SDF1α expression on the cancer cells was significantly associated with SDF1α expression on the stromal cells. SDF1α was first cloned from bone marrow-derived stromal cells (33) and was reported to be expressed on various stromal cells (34,35). These results suggested that SDF1α on the membrane of cancer cells may be derived from fibroblast-like stromal cells.

SDF1α was not expressed by any gastric or pancreatic cancer cell lines (36,37). Therefore, in the present study the SDF1α expression on stromal cells was investigated. Orimo et al (38) also demonstrated that SDF1α released by stromal fibroblasts directed the paracrine stimulation of tumor cells through CXCR4 expressed on breast cancer cells. SDF1α signaling may be associated with the malignant progression of cancer cells. It was also observed that the SDF1α expression on the tumor stromal cells was associated with the diffuse type, while that on the cancer cells was associated with the intestinal type (data not shown). SDF1α signaling may be different between the histological types of GC.

In the present study, CXCR4 expression on cancer cells was associated with macroscopic type 4, lymph node metastasis and peritoneal metastasis. It has been reported that CXCR4 expression was associated with lymph node or liver metastasis in GC (34,35), and was a prognostic factor in GC (3941). In the present study, patients with CXCR4 and SDF1α expression exhibited significantly poorer prognoses. The results of the present study suggested that the SDF1α/CXCR4 axis may serve an important role in the progression of cancer, and that the expression of these molecules may be a useful prognostic factor for patients with stage III GC.

Hypoxia is thought to be associated with aggressive tumor phenotypes of gastric carcinomas (42,43), including the metastatic ability of cancer cells (44,45). Clinical and experimental data have also provided evidence of an association between the hypoxic environment and a poor prognosis (45,46). In the present study, CA9, which was used to investigate the hypoxic cells, was demonstrated to be expressed heterogeneously in a gastric tumor, and it was found that the CA9 expression was significantly associated with the CXCR4 expression on the cancer cells and the SDF1α expression on the stromal cells. These results suggested that hypoxia, which was evaluated by CA9 staining, may induce SDF1a and CXCR4. Recent studies have demonstrated that SDF1α is upregulated in fibroblasts to fulfill its role in cell protection against hypoxia (4,32). These results suggested that the heterogeneous hypoxic environment in cancer may be one of the reasons for cancer heterogenicity, which is associated with tumor resistance for various types of therapy (15,47,48).

SDF1α may serve as a protective factor to promote cell repair following hypoxic injury via its main receptor, CXCR4 (49). Our previous study demonstrated that the hypoxic condition affected the expression level of certain receptors of cancer cells (17,18,50). The results of our present study suggested that these results indicated that hypoxia may upregulate SDFα production from stromal cells and CXCR4 expression in cancer cells. Therefore, the SDF1α/CXCR4 axis may serve an important role in the progression of GC cells in hypoxia.

In conclusion, the SDF1α/CXCR4 axis may be involved in the progression of GC at stage II and III, particularly under hypoxic conditions.

Acknowledgements

Not applicable.

Funding

The present study was partially founded by the KAKENHI (Grant-in-Aid for Scientific Research; nos. 18H02883 and 23390329) from the Ministry of Education, Science, Sports, Culture and Technology of Japan.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

MY and HK designed, performed the experiments and co-wrote the manuscript. GT, TF, YY, TS, ST and AS prepared the samples. SN, SK, MY and TT accumulated the data. TS, KK, ST and MO sampled the material and MO reviewed manuscript. All authors read and approved the final manuscript.

Ethics statement and consent to participate

The present study was conducted with the approval of the Ethical Committee of Osaka City University (reference no. 924). Written informed consent was obtained from all patients prior to treatment.

Patient consent for publication

Written informed consent was obtained from all patients.

Competing interests

The authors declare that they have no competing interests.

References

1 

Global Burden of Disease Cancer Collaboration, ; Fitzmaurice C, Dicker D, Pain A, Hamavid H, Moradi-Lakeh M, MacIntyre MF, Allen C, Hansen G, Woodbrook R, et al: The global burden of cancer 2013. JAMA Oncol. 1:505–527. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Miki Y, Yashiro M, Ando K, Okuno T, Kitayama K, Masuda G, Tamura T, Sakurai K, Toyokawa T, Kubo N, et al: Examination of cancer cells exposed to gastric serosa by serosal stamp cytology plus RT-PCR is useful for the identification of gastric cancer patients at high risk of peritoneal recurrence. Surg Oncol. 26:352–358. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Togano S, Yashiro M, Miki Y, Yamamoto Y, Sera T, Kushitani Y, Sugimoto A, Kushiyama S, Nishimura S, Kuroda K, et al: Microscopic distance from tumor invasion front to serosa might be a useful predictive factor for peritoneal recurrence after curative resection of T3-gastric cancer. PLoS One. 15:e02259582020. View Article : Google Scholar : PubMed/NCBI

4 

Kinoshita H, Yashiro M, Fukuoka T, Hasegawa T, Morisaki T, Kasashima H, Masuda G, Noda S and Hirakawa K: Diffuse-type gastric cancer cells switch their driver pathways from FGFR2 signaling to SDF1/CXCR4 axis in hypoxic tumor microenvironments. Carcinogenesis. 36:1511–1520. 2015.PubMed/NCBI

5 

Lee HJ and Jo DY: The role of the CXCR4/CXCL12 axis and its clinical implications in gastric cancer. Histol Histopathol. 27:1155–1161. 2012.PubMed/NCBI

6 

Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, et al: Involvement of chemokine receptors in breast cancer metastasis. Nature. 410:50–56. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Gockel I, Schimanski CC, Heinrich C, Wehler T, Frerichs K, Drescher D, von Langsdorff C, Domeyer M, Biesterfeld S, Galle PR, et al: Expression of chemokine receptor CXCR4 in esophageal squamous cell and adenocarcinoma. BMC Cancer. 6:2902006. View Article : Google Scholar : PubMed/NCBI

8 

Engl T, Relja B, Blumenberg C, Müller I, Ringel EM, Beecken WD, Jonas D and Blaheta RA: Prostate tumor CXC-chemokine profile correlates with cell adhesion to endothelium and extracellular matrix. Life Sci. 78:1784–1793. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Wu QY, Yang CK, Rong LJ, Li JC and Lei LM: Investigation of the association between C-X-C motif chemokine receptor subunits and tumor infiltration levels and prognosis in patients with early-stage pancreatic ductal adenocarcinoma. Oncol Lett. 20:162020.PubMed/NCBI

10 

Katsumoto K and Kume S: The role of CXCL12-CXCR4 signaling pathway in pancreatic development. Theranostics. 3:11–17. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Scala S, Giuliano P, Ascierto PA, Ieranò C, Franco R, Napolitano M, Ottaiano A, Lombardi ML, Luongo M, Simeone E, et al: Human melanoma metastases express functional CXCR4. Clin Cancer Res. 12:2427–2433. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Zhang SS, Han ZP, Jing YY, Tao SF, Li TJ, Wang H, Wang Y, Li R, Yang Y, Zhao X, et al: CD133+CXCR4+ colon cancer cells exhibit metastatic potential and predict poor prognosis of patients. BMC Med. 10:852012. View Article : Google Scholar : PubMed/NCBI

13 

Chiaramonte R, Colombo M, Bulfamante G, Falleni M, Tosi D, Garavelli S, De Simone D, Vigolo E, Todoerti K, Neri A and Platonova N: Notch pathway promotes ovarian cancer growth and migration via CXCR4/SDF1alpha chemokine system. Int J Biochem Cell Biol. 66:134–140. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Cavallaro S: CXCR4/CXCL12 in non-small-cell lung cancer metastasis to the brain. Int J Mol Sci. 14:1713–1727. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Kitayama K, Yashiro M, Morisaki T, Miki Y, Okuno T, Kinoshita H, Fukuoka T, Kasashima H, Masuda G, Hasegawa T, et al: Pyruvate kinase isozyme M2 and glutaminase might be promising molecular targets for the treatment of gastric cancer. Cancer Sci. 108:2462–2469. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Kato Y, Yashiro M, Noda S, Kashiwagi S, Matsuoka J, Fuyuhiro Y, Doi Y and Hirakawa K: Expression of a hypoxia-associated protein, carbonic anhydrase-9, correlates with malignant phenotypes of gastric carcinoma. Digestion. 82:246–251. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Noda S, Yashiro M, Nshii T and Hirakawa K: Hypoxia upregulates adhesion ability to peritoneum through a transforming growth factor-beta-dependent mechanism in diffuse-type gastric cancer cells. Eur J Cancer. 46:995–1005. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Matsuoka J, Yashiro M, Doi Y, Fuyuhiro Y, Kato Y, Shinto O, Noda S, Kashiwagi S, Aomatsu N, Hirakawa T, et al: Hypoxia stimulates the EMT of gastric cancer cells through autocrine TGFβ signaling. PLoS One. 8:e623102013. View Article : Google Scholar : PubMed/NCBI

19 

Hirakawa T, Yashiro M, Doi Y, Kinoshita H, Morisaki T, Fukuoka T, Hasegawa T, Kimura K, Amano R and Hirakawa K: Pancreatic fibroblasts stimulate the motility of pancreatic cancer cells through IGF1/IGF1R signaling under hypoxia. PLoS One. 11:e01599122016. View Article : Google Scholar : PubMed/NCBI

20 

Kasashima H, Yashiro M, Kinoshita H, Fukuoka T, Morisaki T, Masuda G, Sakurai K, Kubo N, Ohira M and Hirakawa K: Lysyl oxidase is associated with the epithelial-mesenchymal transition of gastric cancer cells in hypoxia. Gastric Cancer. 19:431–442. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Kasashima H, Yashiro M, Nakamae H, Kitayama K, Masuda G, Kinoshita H, Fukuoka T, Hasegawa T, Nakane T, Hino M, et al: CXCL1-Chemokine (C-X-C Motif) receptor 2 signaling stimulates the recruitment of bone marrow-derived mesenchymal cells into diffuse-type gastric cancer stroma. Am J Pathol. 186:3028–3039. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Kasashima H, Yashiro M, Nakamae H, Masuda G, Kinoshita H, Morisaki T, Fukuoka T, Hasegawa T, Sakurai K, Toyokawa T, et al: Bone marrow-derived stromal cells are associated with gastric cancer progression. Br J Cancer. 113:443–452. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Yashiro M, Matsuoka T and Ohira M: The significance of scirrhous gastric cancer cell lines: The molecular characterization using cell lines and mouse models. Hum Cell. 31:271–281. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Okuno T, Yashiro M, Masuda G, Togano S, Kuroda K, Miki Y, Hirakawa K, Ohsawa M, Wanibuchi H and Ohira M: Establishment of a new scirrhous gastric cancer cell line with FGFR2 overexpression, OCUM-14. Ann Surg Oncol. 26:1093–1102. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Oh YS, Kim HY, Song IC, Yun HJ, Jo DY, Kim S and Lee HY: Hypoxia induces CXCR4 expression and biological activity in gastric cancer cells through activation of hypoxia-inducible factor-1α. Oncol Rep. 28:2239–2246. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Romain B, Hachet-Haas M, Rohr S, Brigand C, Galzi JL, Gaub MP, Pencreach E and Guenot D: Hypoxia differentially regulated CXCR4 and CXCR7 signaling in colon cancer. Mol Cancer. 13:582014. View Article : Google Scholar : PubMed/NCBI

27 

Japanese Gastric Cancer Association: Japanese classification of gastric carcinoma: 3rd English edition. Gastric Cancer. 14:101–112. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Greene FL and Sobin LH: A worldwide approach to the TNM staging system: Collaborative efforts of the AJCC and UICC. J Surg Oncol. 99:269–272. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Shephard DA: The 1975 declaration of helsinki and consent. Can Med Assoc J. 115:1191–1192. 1976.PubMed/NCBI

30 

Eckert AW, Horter S, Bethmann D, Kotrba J, Kaune T, Rot S, Bache M, Bilkenroth U, Reich W, Greither T, et al: Investigation of the prognostic role of carbonic anhydrase 9 (CAIX) of the cellular mRNA/protein level or soluble CAIX protein in patients with oral squamous cell carcinoma. Int J Mol Sci. 20:3752019. View Article : Google Scholar

31 

Hitchon C, Wong K, Ma G, Reed J, Lyttle D and El-Gabalawy H: Hypoxia-induced production of stromal cell-derived factor 1 (CXCL12) and vascular endothelial growth factor by synovial fibroblasts. Arthritis Rheum. 46:2587–2597. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Liu H, Liu S, Li Y, Wang X, Xue W, Ge G and Luo X: The role of SDF-1-CXCR4/CXCR7 axis in the therapeutic effects of hypoxia-preconditioned mesenchymal stem cells for renal ischemia/reperfusion injury. PLoS One. 7:e346082012. View Article : Google Scholar : PubMed/NCBI

33 

Tashiro K, Tada H, Heilker R, Shirozu M, Nakano T and Honjo T: Signal sequence trap: A cloning strategy for secreted proteins and type I membrane proteins. Science. 261:600–603. 1993. View Article : Google Scholar : PubMed/NCBI

34 

Iwasa S, Yanagawa T, Fan J and Katoh R: Expression of CXCR4 and its ligand SDF-1 in intestinal-type gastric cancer is associated with lymph node and liver metastasis. Anticancer Res. 29:4751–4758. 2009.PubMed/NCBI

35 

Zhao BC, Wang ZJ, Mao WZ, Ma HC, Han JG, Zhao B and Xu HM: CXCR4/SDF-1 axis is involved in lymph node metastasis of gastric carcinoma. World J Gastroenterol. 17:2389–2396. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Yasumoto K, Koizumi K, Kawashima A, Saitoh Y, Arita Y, Shinohara K, Minami T, Nakayama T, Sakurai H, Takahashi Y, et al: Role of the CXCL12/CXCR4 axis in peritoneal carcinomatosis of gastric cancer. Cancer Res. 66:2181–2187. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Koshiba T, Hosotani R, Miyamoto Y, Ida J, Tsuji S, Nakajima S, Kawaguchi M, Kobayashi H, Doi R, Hori T, et al: Expression of stromal cell-derived factor 1 and CXCR4 ligand receptor system in pancreatic cancer: A possible role for tumor progression. Clin Cancer Res. 6:3530–3535. 2000.PubMed/NCBI

38 

Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL and Weinberg RA: Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 121:335–348. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Jiang Q, Sun Y and Liu X: CXCR4 as a prognostic biomarker in gastrointestinal cancer: A meta-analysis. Biomarkers. 24:510–516. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Yu C and Zhang Y: Characterization of the prognostic values of CXCR family in gastric cancer. Cytokine. 123:1547852019. View Article : Google Scholar : PubMed/NCBI

41 

Ishigami S, Natsugoe S, Okumura H, Matsumoto M, Nakajo A, Uenosono Y, Arigami T, Uchikado Y, Setoyama T, Arima H, et al: Clinical implication of CXCL12 expression in gastric cancer. Ann Surg Oncol. 14:3154–3158. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Nayak A, Roy AD, Rout N, Singh SP, Bhattacharyya A and Roychowdhury A: HIF1α-dependent upregulation of ATAD2 promotes proliferation and migration of stomach cancer cells in response to hypoxia. Biochem Biophys Res Commun. 523:916–923. 2020. View Article : Google Scholar : PubMed/NCBI

43 

de Barros Moreira Beltrão H, de Paula Cerroni M, de Freitas DR, das Neves Pinto AY, da Costa Valente V, Valente SA, de Góes Costa E and Sobel J: Investigation of two outbreaks of suspected oral transmission of acute chagas disease in the amazon region, para state, Brazil, in 2007. Trop Doct. 39:231–232. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Li Q, Zhu CC, Ni B, Zhang ZZ, Jiang SH, Hu LP, Wang X, Zhang XX, Huang PQ, Yang Q, et al: Lysyl oxidase promotes liver metastasis of gastric cancer via facilitating the reciprocal interactions between tumor cells and cancer associated fibroblasts. EBioMedicine. 49:157–171. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Bubnovskaya L and Osinsky D: Tumor microenvironment and metabolic factors: Contribution to gastric cancer. Exp Oncol. 42:2–10. 2020.PubMed/NCBI

46 

Zhang WJ, Chen C, Zhou ZH, Gao ST, Tee TJ, Yang LQ, Xu YX, Pang TH, Xu XY, Sun Q, et al: Hypoxia-inducible factor-1 alpha correlates with tumor-associated macrophages infiltration, influences survival of gastric cancer patients. J Cancer. 8:1818–1825. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Kato Y, Yashiro M, Fuyuhiro Y, Kashiwagi S, Matsuoka J, Hirakawa T, Noda S, Aomatsu N, Hasegawa T, Matsuzak T, et al: Effects of acute and chronic hypoxia on the radiosensitivity of gastric and esophageal cancer cells. Anticancer Res. 31:3369–3375. 2011.PubMed/NCBI

48 

Al-Juboori SI, Vadakekolathu J, Idri S, Wagner S, Zafeiris D, Rd Pearson J, Almshayakhchi R, Caraglia M, Desiderio V, Miles AK, et al: PYK2 promotes HER2-positive breast cancer invasion. J Exp Clin Cancer Res. 38:2102019. View Article : Google Scholar : PubMed/NCBI

49 

Liu S, Jia X, Li C, Han X, Yan W and Xing Y: CXCR7 silencing attenuates cell adaptive response to stromal cell derived factor 1alpha after hypoxia. PLoS One. 8:e552902013. View Article : Google Scholar : PubMed/NCBI

50 

Kato Y, Yashiro M, Noda S, Tendo M, Kashiwagi S, Doi Y, Nishii T, Matsuoka J, Fuyuhiro Y, Shinto O, et al: Establishment and characterization of a new hypoxia-resistant cancer cell line, OCUM-12/hypo, derived from a scirrhous gastric carcinoma. Br J Cancer. 102:898–907. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 21 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yashiro M, Kinoshita H, Tsujio G, Fukuoka T, Yamamoto Y, Sera T, Sugimoto A, Nishimura S, Kushiyama S, Togano S, Togano S, et al: SDF1&alpha;/CXCR4 axis may be associated with the malignant progression of gastric cancer in the hypoxic tumor microenvironment. Oncol Lett 21: 38, 2021
APA
Yashiro, M., Kinoshita, H., Tsujio, G., Fukuoka, T., Yamamoto, Y., Sera, T. ... Ohira, M. (2021). SDF1&alpha;/CXCR4 axis may be associated with the malignant progression of gastric cancer in the hypoxic tumor microenvironment. Oncology Letters, 21, 38. https://doi.org/10.3892/ol.2020.12299
MLA
Yashiro, M., Kinoshita, H., Tsujio, G., Fukuoka, T., Yamamoto, Y., Sera, T., Sugimoto, A., Nishimura, S., Kushiyama, S., Togano, S., Kuroda, K., Toyokawa, T., Ohira, M."SDF1&alpha;/CXCR4 axis may be associated with the malignant progression of gastric cancer in the hypoxic tumor microenvironment". Oncology Letters 21.1 (2021): 38.
Chicago
Yashiro, M., Kinoshita, H., Tsujio, G., Fukuoka, T., Yamamoto, Y., Sera, T., Sugimoto, A., Nishimura, S., Kushiyama, S., Togano, S., Kuroda, K., Toyokawa, T., Ohira, M."SDF1&alpha;/CXCR4 axis may be associated with the malignant progression of gastric cancer in the hypoxic tumor microenvironment". Oncology Letters 21, no. 1 (2021): 38. https://doi.org/10.3892/ol.2020.12299