Differential expression of CXCR4 and CXCR7 with various stem cell markers in paired human primary and recurrent glioblastomas

  • Authors:
    • Charlotte Flüh
    • Kirsten Hattermann
    • H. Maximilian Mehdorn
    • Michael Synowitz
    • Janka Held-Feindt
  • View Affiliations

  • Published online on: January 25, 2016     https://doi.org/10.3892/ijo.2016.3354
  • Pages: 1408-1416
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The chemokine CXCL12 (also termed SDF-1, stromal cell-derived factor-1) and its receptors CXCR4 and CXCR7 are known to play a pivotal role in tumor progression including glioblastomas (GBM). Previous investigations focused on the expression and functional roles of CXCR4 and CXCR7 in different GBM cell subpopulations, but comparative analysis in matched primary versus recurrent GBM samples are still lacking. Thus, here we investigated the expression of CXCR4 and CXCR7 on mRNA and protein level using matched primary and recurrent GBM pairs. Additionally, as GBM CXCR4-positive stem-like cells are supposed to give rise to recurrence, we compared the expression of both receptors in primary and recurrent GBM cells expressing either neural (MUSASHI-1) or embryonic stem cell markers (KLF-4, OCT-4, SOX-2, NANOG). We were able to show that both CXCR4 and CXCR7 were expressed at considerable mRNA and protein levels. CXCR7 was downregulated in relapse cases, and different groups regarding CXCR4/CXCR7 expression differences between primary and recurrent samples could be distinguished. A co-expression of both receptors was rare. In line with this, CXCR4 was co-expressed with all investigated neural and embryonic stem cell markers in both primary and recurrent tissues, whereas CXCR7 was mostly found on stem cell marker-negative cells, but was co-expressed with KLF-4 on a distinct GBM cell subpopulation. These results point to an individual role of CXCR4 and CXCR7 in stem cell marker-positive GBM cells in glioma progression and underline the opportunity to develop new therapeutic tools for GBM intervention.

Introduction

Gliomas are the most common type of primary brain tumors. The most malignant form, glioblastoma multiforme (GBM), accounts for ~15% of all brain tumors, and >50% of all astrocytomas (1). Current standard treatment of GBM is surgical resection of the tumor, followed by adjuvant radio- and chemotherapy. However, median survival time for GBM patients is still poor, ~12–15 months despite multimodal therapy (2), and currently there are no effective long-term treatments for this malignancy. One of the primary reasons for the poor prognosis is the development of recurrence composed of highly proliferative and infiltrative tumor cells which massively invade into the surrounding brain parenchyma and contribute to the fatal progression (2).

Chemokines, small chemotactic cytokines, are known to contribute to a broad spectrum of physiological and pathological processes, including angiogenesis (3), haematopoiesis (4), development (5,6) and also tumor initiation, survival and progression (7,8).

In particular, the chemokine CXCL12 (also termed SDF-1, stromal cell-derived factor-1) and its receptors CXCR4 and CXCR7 seem to play a pivotal role in tumor progression, as described for different tumor types including GBM (911). In GBM, CXCL12 and CXCR4 are overexpressed in tumor tissues when compared to normal brain parenchyma and their expression level correlates with tumor grade and poor prognosis (12). The CXCL12/CXCR4 activation in glioma cells and specific cells of the surrounding microenvironment (e.g., microglia, endothelial cells, and mesenchymal cells) contributes to GBM proliferation, spreading, and chemoresistance as reviewed for example by Würth et al (13). The long known receptor CXCR4 is expressed on glioma cells with stem cell properties (14,15). These cells are able to perform self-renewal, to recapitulate the whole tumor and to differentiate into specific GBM subpopulations. Thus, they are likely responsible for the development of glioblastoma relapses and the poor prognosis of recurrent GBM (16). Nevertheless, not only CXCR4, but also CXCR7, which has been described in tumors, is a regulator of GBM growth (13). For example, CXCR7 is highly expressed in tumor endothelial, microglial and GBM cells (15,17), controls tumor diffusion through CXCL12 gradients and is frequently detected in GBM-associated vasculature (18). Interestingly, in contrast to CXCR4, CXCR7 was detected on more differentiated GBM cells (15). However, a significant correlation between CXCR4 and CXCR7 in GBM was observed (19). These results are fascinating and point to a pivotal role of CXCR4 and CXCR7 in glioma progression including especially the development of recurrences.

Materials and methods

Tumor specimens

GBM samples were surgically dissected tissues from the Department of Neurosurgery (Kiel, Germany) and were obtained in accordance with the Helsinki Declaration of 1975 with approval of the ethics committee of the University of Kiel, Germany after written informed consent of donors (file reference: D 536/15). Tumors were classified according to the WHO criteria, and the diagnosis was established by a pathologist. A total of 28 GBM (14 primary and 14 recurrent tumors, paired samples for each single donor) was included. If enough material was available, matched probes of individual tumor samples were used for different investigations.

Reverse transcription and real-time PCR (qRT-PCR)

RNA was isolated with the TRIzol reagent (Invitrogen, Carlsbad, CA, USA), digested by DNase, cDNA was synthesized, and quantitative reverse transcription real-time PCR (qRT-PCR) was performed as described before (20) using TaqMan primer probes (Applied Biosystems, Foster City, CA, USA): glycerinaldehyde-3-phosphate-dehydrogenase (hGAPDH) (Hs99999905_m1), hCXCR4 (Hs00237052_m1), hCXCR7 (Hs00664172_s1). Fluorescent data were converted into cycle threshold (CT) measurements, and ΔCT values of each sample were calculated as CTgene of interest − CT GAPDH. Relative gene expression was calculated with 2(normalized CT non-stimulated − normalized CT stimulated) = n-fold of control. A ΔCT value of 3.33 corresponds to one magnitude lower gene expression compared to GAPDH. To visualize possible similarities in chemokine receptor expression of individual primary-recurrent GBM pairs, relative gene expression data were assigned to grey shades in Fig. 1. A relative gene expression value of 1 (= equal expression in primary and recurrent GBM) was assigned as 30% grey, lower n-fold expression values (lower expression in recurrent compared to primary) were displayed with increasing lighter shading with 0 corresponding to white. Relative expression values >1 (higher expression in recurrence compared to primary) were assigned with increasing darker grey shades until 3-fold induction (or higher) which was assigned as maximum (black). Afterwards, a ‘heatmap-like’ arrangement of individual primary-recurrent GBM pairs was performed orientating to up- or downregulation of CXCR4 and CXCR7 in recurrent samples.

Immunofluorescence

Cryostat sections of different primary and recurrent GBM tissues were fixed in acetone/methanol, and sequentially blocked with Sudan black and 0.1% bovine serum albumin as described before (20). Primary antibodies were applied overnight at 4°C, secondary antibodies were incubated at 37°C for 1 h, nuclei were counterstained with 4′,6-diamidino-2-phenylindole (DAPI; Sigma-Aldrich, Hamburg, Germany), slides were embedded with Immumount (ThermoShandon, Pittsburgh, PA, USA) and digital photography was performed using a Zeiss microscope and Zeiss camera (Zeiss, Oberkochen, Germany). Primary antibodies were anti-OCT-4 (octamer binding transcription factor 4; 1:150, rabbit; Cell Signaling, Danvers, MA, USA), anti-SOX-2 (sex determining region Y-box 2; 1:200, rabbit; Santa Cruz Biotechnology, CA, USA), anti-MUSASHI-1 [Musashi (Drosophila) homolog 1; 1:100, mouse; R&D Systems, Wiesbaden, Germany], anti-NANOG (‘Tir nan Og‘; 1:500, rabbit; Thermo Fisher Scientific, Rockford, IL, USA), anti-KLF-4 (Krüppel-like factor 4; 1:250, mouse; Thermo Fisher Scientific), anti-CXCR4 (1:200, rabbit; Imgenex IMG 125-2, San Diego, CA, USA) and anti-CXCR7 (1:100, mouse, MAB42273; R&D Systems). If primary antibodies were derived from the same species, unspecific binding was blocked by F(ab) fragments derived from this species [donkey anti-mouse and anti-rabbit F(ab) fragments, 1:100, from Dianova, Hamburg, Germany]. Primary antibodies were omitted for negative controls. As secondary antibodies donkey anti-mouse or anti-rabbit IgGs labeled with Alexa Fluor 488 or Alexa Fluor 555 (1:1,000; Invitrogen) were used.

Statistical analysis

For statistical analyses a two-tailed Student's t-test with matched samples was used. Significance level was p<0.05 (indicated by an asterisk (*) in the figures).

Results

Expression of CXCR4 and CXCR7 in human primary and recurrent GBM pairs

To evaluate mRNA expression levels of CXCR4 and CXCR7, qRT-PCR analysis was performed using matched probes of solid human primary and recurrent GBM. Results are shown in Fig. 1, where single ΔCT values for primary GBM samples are demonstrated in black circles and ΔCT values for recurrent ones in white triangles. It should be kept in mind that a ΔCT value of 3.33 corresponds to one magnitude lower gene expression.

Irrespective of the tumor identity (primary versus recurrent), both CXCR4 and CXCR7 were detected at considerable levels in solid GBM tissues with CXCR7 clearly to higher extent. Additionally, when comparing primary and recurrent GBM samples, the chemokine receptor CXCR7 was expressed in lower amounts in recurrences (mean reduction to ~40%; p<0.05), while this was not observed for CXCR4. In detail, the normalized averaged ΔCT values for investigated primary/recurrent GBM samples were: 7.74/7.91 (CXCR4), and 2.61/3.95 (CXCR7), respectively (Fig. 1, left).

To compare chemokine receptor expression differences between individual primary and recurrent GBM pairs more in detail, we arranged the n-fold expression changes of all individual primary-recurrent GBM pairs as a heatmap: equal n-fold expression in primary and recurrent GBM was assigned as 30% grey, lower n-fold expression values were displayed with increasing lighter shading with 0 corresponding to white, and relative n-fold expression values >1 were assigned with increasing darker grey shades until 3-fold induction (or higher) which was assigned as maximum (black). By this, three different GBM groups in our collective could be identified (Fig. 1, right): the first group, containing seven primary-recurrent GBM pairs, was characterized by a general lower mRNA expression of CXCR4 and CXCR7 in recurrent samples. The second group, containing five primary-recurrent GBM pairs, was characterized by higher expression of CXCR4 in combination with lower expression of CXCR7. The third group included two GBM pairs and was characterized by higher expression of both chemokine receptors in recurrences. Interestingly, a combination of increased CXCR7 and decreased CXCR4 expression was not observed in our cohort. Summarized, various combinations of loss or gain of CXCR4 and CXCR7 mRNA expression are apparently detectable in our collective of paired primary and recurrent GBM samples, and it became clear that a more precise evaluation of chemokine receptor expression during the progression from primary to recurrent GBM is possible if each individual pair is analyzed in detail.

To confirm qRT-PCR results and to evaluate CXCR4 and CXCR7 expression also on the protein level in primary-recurrent GBM pairs, we performed double-immunofluorencence staining of both chemokine receptors. Nuclei were stained with DAPI. Exemplary results of stained matched primary and recurrent samples are shown in Fig. 2. Although no clear quantitative data could be obtained using fluorescence immunostaining of cryo-sections the primary tumors showed high amounts of CXCR4 or CXCR7 positively stained cells, whereas in matched recurrent samples only few CXCR7-positive cells, but considerable amounts of CXCR4-positive cells were visible. In addition, the majority of tumor cells were solely positive for CXCR4 or CXCR7, respectively (exemplified in Fig. 2), only single cells expressed CXCR4 and CXCR7 in the same cell regions [examples for merged regions (yellow) are shown in Fig. 2].

Summarized, while both CXCR4 and CXCR7 are expressed at considerable levels in primary and recurrent GBM samples, CXCR7 expression is apparently downregulated in relapsed cases. Regarding CXCR4/CXCR7 expression differences between primary and recurrent samples, our cohort included different GBM groups of combined induction or reduction upon relapse. Expression of CXCR4 and CXCR7 was confirmed on protein level, and co-stainings revealed single as well as double-positive cells.

Cellular allocation of CXCR4 and CXCR7 in human primary and recurrent GBM pairs

We and others have shown that CXCR4 is predominately expressed in (tumor) stem-like cells (14,15), and GBM stem-like cells are supposed to give rise to the recurrences. Therefore, we analyzed in a next step in more detail which cells might account for expression of CXCR4 and CXCR7 in primary and recurrent GBM. Thus, co-stainings with neural stem/progenitor (RNA-binding protein MUSASHI-1) and different embryonic stem cell markers [KLF-4, OCT-4, SOX-2 as embryonic stem cell transcription factors, and NANOG which initially was used as readout for efficient reprogramming of iPSCs (21)] were performed and analyzed by fluorescence microscopy.

In general, CXCR4 yielded clear co-stainings with different stem cells markers in both primary and recurrent samples (exemplified in Fig. 3). However, this method does not allow for a valid quantification of both staining intensities and amounts of positively stained cells. Additionally, different antigens are not localized in the same cellular structures, thus signals do not merge in all cases, but can be found in the same regions. Nevertheless, especially in the co-staining CXCR4-SOX2 nearly all CXCR4-positive cells were also SOX-2-positive. For KLF-4, OCT-4 and MUSASHI-1 we also observed clear co-stainings with CXCR4 in both primary and recurrent GBM, with OCT-4-CXCR4 double-positive cells being remarkably localized in cluster-like structures. However, also CXCR4, KLF-4, OCT-4 and MUSASHI-1 single-positive cells existed within the sections. For NANOG, a co-staining with CXCR4 was also observed but not as prominent as detected for the other stem cell markers (exemplified in Fig. 3).

In comparison to CXCR4 the results for CXCR7 were clearly different. With exception of the combination CXCR7-KLF-4 only very few CXCR7-stem cell marker double-positive cells were found in the GBM specimens (exemplified in Fig. 4). In detail, when looking at OCT-4 and SOX-2 in CXCR7-positive regions, some OCT-4 or SOX-2-positive cells (and here more obvious in recurrences) were found, respectively, but it seems that in the majority of the cases CXCR7 was expressed in different, OCT-4 or SOX-2 negative cells. In line with these results, a clear co-staining of CXCR7 with NANOG or MUSASHI-1 was not detectable although positive cells were found in CXCR7-positive cell regions. In contrast, KLF-4 and CXCR7 were found co-expressed in the same cells and, although this method does not allow a valid quantification, this was more prominent in recurrent samples (exemplified in Fig. 4). Nevertheless, also KLF-4 and CXCR7 single-positive cells were found in the tumor sections, respectively.

Summarized, CXCR4 was clearly expressed in stem cell marker positive cells in both primary and recurrent GBM samples, whereas CXCR7, with the exception of a clear co-staining with KLF-4-positive cells especially in recurrences, was predominantly found in stem cell marker negative cells in the investigated samples.

Discussion

The chemokine CXCL12/SDF-1 is involved in glioma progression (10,13,15,17). In GBM, CXCL12 and its receptor, CXCR4, are localized in regions of necrosis and angiogenesis (22), and are supposed to mediate proliferation of GBM progenitor cells (14). In contrast, the CXCL12 receptor CXCR7, which was initially regarded as a decoy receptor scavenging CXCL12 to prevent CXCR4 signaling and effects (23), mediates apoptosis resistance in human and rat glioma cells (15,17). Previous results focusing on the expression of both receptors in human GBM indeed yielded an expression of CXCR4 by a subpopulation of GBM cells with stem cell properties (14), whereas the bulk of more differentiated GBM cells express CXCR7 (15,24). Conversely, other studies reported the existence of CXCR4-CXCR7 double-positive cells and showed that this subpopulation might regulate the stem cell phenotype (25), and was able to initiate intracranial tumors in vivo (26). Nevertheless, also CXCR4 and CXCR7 single populations exist and a high level of heterogeneity in both the surface expression and functions of CXCR4 and CXCR7 in primary human GBM cells of the proliferative subclass was determined (26). In addition, GBM primary cell cultures show a heterogeneous cell surface expression of CXCR4 and CXCR7 despite similar levels of corresponding mRNAs (25), and the expression of CXCR4 and CXCR7 is significantly correlated (19).

Since comparative investigations regarding the expression of CXCR4 and CXCR7 in matched samples of primary and recurrent GBM are still lacking, we analysed this point in 14 different human primary-recurrent GBM pairs. We were able to show that irrespective of used materials (primary/recurrent GBM) both CXCR4 and CXCR7 were expressed at considerable amounts on the mRNA level with CXCR7 clearly to higher extent. Whereas CXCR4 expression was nearly equal in primary and recurrent samples, CXCR7 was found in lower amounts in recurrent GBM samples (~40%) in comparison to primary ones. Nevertheless when analyzing each individual pair in detail, two primary-recurrent pairs were observed in our GBM collective with higher CXCR7 mRNA level in the recurrent samples. Although immunofluorescence staining of cryo-sections cannot clearly deliver quantitative data, the CXCR7 expression appeared lower in recurrent samples on the protein level, which is in accordance with the results obtained by qRT-PCR.

Interestingly, Razmkhah et al (27) reported that CXCR7 was undetectable in secondary brain tumors (sarcoma and breast cancer), whereas CXCR4 was expressed. Conversely to our observations, in that glioma cohort, CXCR4 expression was ~110-fold higher than its counterpart CXCR7. However, only 7 samples of glioma (4 high-grade gliomas and 3 low-grade gliomas without any histopathological specification) were included.

Further, when investigating CXCR4 and CXCR7 expression in primary and recurrent GBM on the protein level by double-immunofluorescence technique, we found that the chemokine receptors were mainly detectable on different cell types, only very few CXCR4-CXCR7 double-positive cells existed in the samples. In accordance with this, heterogeneity of CXCR4 and CXCR7 expression was reported before for primary GBM cells pointing to the existence of different subpopulations measurable by variable percentages of CXCR4+CXCR7, CXCR4CXCR7+ and CXCR4+CXCR7+ cells in glioma samples (26). In addition, since for GBM-positive correlations not only for the expression of CXCR7 and CXCR4 but also for the expression of CXCR7 and HIF1α and CXCR7 and IDH1 were reported, these results also indicate the existence of different chemokine receptor expressing subclones (19). By co-staining of CXCR4 and CXCR7 with stem cell markers we could show in our primary-recurrent GBM cohort that CXCR4 was mostly found on stem-cell marker positive GBM cells, whereas CXCR7 was usually expressed on stem-cell marker negative cells, with exception of the combination CXCR7-KLF-4. An embryonic stem cell gene signature is well known to correlate with a more undifferentiated phenotype in various cancers (28), and a large scale tissue microarray analysis including 80 low-grade and 98 high-grade gliomas showed an upregulated protein level of NANOG, KLF-4, OCT-4 and SOX-2 in high-grade gliomas (29). Several other studies also point to the relevance of neural and embryonic stem cell markers in GBM progression (3036). In the present study we showed that both neural and embryonic stem cells markers are preferentially co-expressed with CXCR4 in matched samples of primary and recurrent GBM pairs, whereas CXCR7 was mostly found on stem cell marker negative cells. In line with these results, CXCR4 expression in neuroblastoma is associated with highly aggressive undifferentiated tumors, while CXCR7 expression was detected in more differentiated and mature neuroblastic tumors (37). In addition, CXCR4 is essential for the self-renewal of GBM stem-like cells since disruption of the CXCL12/CXCR4 pathway resulted in reduced expression of stem-like markers like Nestin and MUSASHI-1 or genes which are involved in regulating stem cell properties such as OCT-4 and NANOG (38). The miR-137 inhibited glioma stem-like cell self-renewal and promoted their differentiation by targeting RTVP-1, which downregulated CXCR4 (39). The inhibition of CXCR4 in glioma-initiating cells disrupted the SHH-GLI-NANOG network, which is involved in self-renewal and expression of the embryonic stem cell-like signature (40).

Interestingly, Chen et al (41) reported that CXCR7 can mediate neural progenitor cell migration to CXCL12 independently of CXCR4. In line with this, our primary-recurrent GBM cohort showed a distinct subpopulation of CXCR7-KLF-4 double-positive cells, which was more prominent in the recurrences. Since it has been shown that the miR-152 targets KLF-4 in GBM cells and thereby influences cell proliferation, invasion, apoptosis and cell migration processes (42), the CXCR7-KLF-4 double-positive GBM cells in our cohort could be a subpopulation of highly migratory tumor cells. However, the paracrine PGI signaling initiated by mesenchymal glioma cells also induces self-renewal and tumorigenic potential of glioma stem cells through induction of KLF-4 (43). The functional role of CXCR7 in KLF-4-positive glioma-stem cells remains still highly speculative.

Summarized, we were able to show that in a cohort of matched primary-recurrent GBM samples the CXCL12 receptors CXCR4 and CXCR7 are expressed both on the mRNA and protein levels in large amounts, with CXCR7 mRNA being statistically significantly downregulated in recurrent samples. A co-expression of both receptors was rare. In accordance with this, CXCR4 was co-expressed with all investigated neural and embryonic stem cell markers in both primary and recurrent tissues, whereas CXCR7 was mostly found on stem cell marker-negative cells but was co-expressed with KLF-4 on a distinct GBM cell subpopulation. These results point to an individual role of CXCR4 and CXCR7 in the contribution of stem cell marker-positive GBM cells in glioma progression and underline the potential of chemokine receptors as targets for new therapeutic approaches in GBM intervention.

Acknowledgements

We thank Brigitte Rehmke, Fereshteh Ebrahim and Jörg Krause for expert technical assistance. This study was supported by the University of Kiel and by the popgen 2.0 network [(P2N; supported by a grant from the German Ministry for Education and Research (01EY1103)].

Abbreviations:

CT

cycle of threshold

DAPI

4′,6-diamidino-2-phenylindole

DMEM

Dulbecco's modified Eagle's medium

FCS

fetal calf serum

GAPDH

glycerinaldehyde-3-phosphate-dehydrogenase

GBM

glioblastoma multiforme

KLF-4

Krüppel-like factor 4

MUSASHI-1

Musashi (Drosophila) homolog 1

NANOG

‘Tir nan Og’

OCT-4

octamer binding transcription factor

qRT-PCR

quantitative reverse transcription polymerase chain reaction

SOX-2

sex determining region Y-box 2

WHO

World Health Organization

References

1 

Ohgaki H and Kleihues P: Epidemiology and etiology of gliomas. Acta Neuropathol. 109:93–108. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al; European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Strieter RM, Polverini PJ, Kunkel SL, Arenberg DA, Burdick MD, Kasper J, Dzuiba J, Van Damme J, Walz A, Marriott D, et al: The functional role of the ELR motif in CXC chemokine-mediated angiogenesis. J Biol Chem. 270:27348–27357. 1995. View Article : Google Scholar : PubMed/NCBI

4 

Broxmeyer HE and Kim CH: Regulation of hematopoiesis in a sea of chemokine family members with a plethora of redundant activities. Exp Hematol. 27:1113–1123. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Dambly-Chaudière C, Cubedo N and Ghysen A: Control of cell migration in the development of the posterior lateral line: Antagonistic interactions between the chemokine receptors CXCR4 and CXCR7/RDC1. BMC Dev Biol. 7:232007. View Article : Google Scholar : PubMed/NCBI

6 

Hattermann K, Ludwig A, Gieselmann V, Held-Feindt J and Mentlein R: The chemokine CXCL16 induces migration and invasion of glial precursor cells via its receptor CXCR6. Mol Cell Neurosci. 39:133–141. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Vandercappellen J, Van Damme J and Struyf S: The role of CXC chemokines and their receptors in cancer. Cancer Lett. 267:226–244. 2008. View Article : Google Scholar : PubMed/NCBI

8 

O'Hayre M, Salanga CL, Handel TM and Allen SJ: Chemokines and cancer: Migration, intracellular signalling and intercellular communication in the microenvironment. Biochem J. 409:635–649. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Müller 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

10 

Hattermann K and Mentlein R: An infernal trio: The chemokine CXCL12 and its receptors CXCR4 and CXCR7 in tumor biology. Ann Anat. 195:103–110. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Zlotnik A, Burkhardt AM and Homey B: Homeostatic chemokine receptors and organ-specific metastasis. Nat Rev Immunol. 11:597–606. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Bian XW, Yang SX, Chen JH, Ping YF, Zhou XD, Wang QL, Jiang XF, Gong W, Xiao HL, Du LL, et al: Preferential expression of chemokine receptor CXCR4 by highly malignant human gliomas and its association with poor patient survival. Neurosurgery. 61:570–579. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Würth R, Bajetto A, Harrison JK, Barbieri F and Florio T: CXCL12 modulation of CXCR4 and CXCR7 activity in human glioblastoma stem-like cells and regulation of the tumor microenvironment. Front Cell Neurosci. 8:144eCollection. 2014.PubMed/NCBI

14 

Ehtesham M, Mapara KY, Stevenson CB and Thompson RC: CXCR4 mediates the proliferation of glioblastoma progenitor cells. Cancer Lett. 274:305–312. 2009. View Article : Google Scholar :

15 

Hattermann K, Held-Feindt J, Lucius R, Müerköster SS, Penfold ME, Schall TJ and Mentlein R: The chemokine receptor CXCR7 is highly expressed in human glioma cells and mediates antiapoptotic effects. Cancer Res. 70:3299–3308. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Auffinger B, Spencer D, Pytel P, Ahmed AU and Lesniak MS: The role of glioma stem cells in chemotherapy resistance and glioblastoma multiforme recurrence. Expert Rev Neurother. 15:741–752. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Hattermann K, Mentlein R and Held-Feindt J: CXCL12 mediates apoptosis resistance in rat C6 glioma cells. Oncol Rep. 27:1348–1352. 2012.PubMed/NCBI

18 

Liu Y, Carson-Walter EB, Cooper A, Winans BN, Johnson MD and Walter KA: Vascular gene expression patterns are conserved in primary and metastatic brain tumors. J Neurooncol. 99:13–24. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Bianco AM, Uno M, Oba-Shinjo SM, Clara CA, de Almeida Galatro TF, Rosemberg S, Teixeira MJ and Nagahashi Marie SK: CXCR7 and CXCR4 expression in infiltrative astrocytomas and their interactions with HIF1α expression and IDH1 mutation. Pathol Oncol Res. 21:229–240. 2015. View Article : Google Scholar

20 

Kubelt C, Hattermann K, Sebens S, Mehdorn HM and Held-Feindt J: Epithelial-to-mesenchymal transition in paired human primary and recurrent glioblastomas. Int J Oncol. 46:2515–2525. 2015.PubMed/NCBI

21 

Takahashi K and Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 126:663–676. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Rempel SA, Dudas S, Ge S and Gutiérrez JA: Identification and localization of the cytokine SDF1 and its receptor, CXC chemokine receptor 4, to regions of necrosis and angiogenesis in human glioblastoma. Clin Cancer Res. 6:102–111. 2000.PubMed/NCBI

23 

Naumann U, Cameroni E, Pruenster M, Mahabaleshwar H, Raz E, Zerwes HG, Rot A and Thelen M: CXCR/functions as a scavenger for CXCR12 and CXCL11. Plos One. 5:e91752010. View Article : Google Scholar

24 

Gatti M, Pattarozzi A, Bajetto A, Würth R, Daga A, Fiaschi P, Zona G, Florio T and Barbieri F: Inhibition of CXCL12/CXCR4 autocrine/paracrine loop reduces viability of human glioblastoma stem-like cells affecting self-renewal activity. Toxicology. 314:209–220. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Walters MJ, Ebsworth K, Berahovich RD, Penfold ME, Liu SC, Al Omran R, Kioi M, Chernikova SB, Tseng D, Mulkearns-Hubert EE, et al: Inhibition of CXCR7 extends survival following irradiation of brain tumours in mice and rats. Br J Cancer. 110:1179–1188. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Liu C, Pham K, Luo D, Reynolds BA, Hothi P, Foltz G and Harrison JK: Expression and functional heterogeneity of chemokine receptors CXCR4 and CXCR7 in primary patient-derived glioblastoma cells. PLoS One. 8:e597502013. View Article : Google Scholar : PubMed/NCBI

27 

Razmkhah M, Arabpour F, Taghipour M, Mehrafshan A, Chenari N and Ghaderi A: Expression of chemokines and chemokine receptors in brain tumor tissue derived cells. Asian Pac J Cancer Prev. 15:7201–7205. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A and Weinberg RA: An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 40:499–507. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Elsir T, Edqvist P-H, Carlson J, Ribom D, Bergqvist M, Ekman S, Popova SN, Alafuzoff I, Ponten F, Nistér M, et al: A study of embryonic stem cell-related proteins in human astrocytomas: Identification of Nanog as a predictor of survival. Int J Cancer. 134:1123–1131. 2014. View Article : Google Scholar

30 

Berezovsky AD, Poisson LM, Cherba D, Webb CP, Transou AD, Lemke NW, Hong X, Hasselbach LA, Irtenkauf SM, Mikkelsen T, et al: Sox2 promotes malignancy in glioblastoma by regulating plasticity and astrocytic differentiation. Neoplasia. 16:193–206. 206.e19–206.e25. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Du Z, Jia D, Liu S, Wang F, Li G, Zhang Y, Cao X, Ling EA and Hao A: Oct4 is expressed in human gliomas and promotes colony formation in glioma cells. Glia. 57:724–733. 2009. View Article : Google Scholar

32 

Galatro TF, Uno M, Oba-Shinjo SM, Almeida AN, Teixeira MJ, Rosemberg S and Marie SK: Differential expression of ID4 and its association with TP53 mutation, SOX2, SOX4 and OCT-4 expression levels. PLoS One. 8:e616052013. View Article : Google Scholar : PubMed/NCBI

33 

Guo Y, Liu S, Wang P, Zhao S, Wang F, Bing L, Zhang Y, Ling EA, Gao J and Hao A: Expression profile of embryonic stem cell-associated genes Oct4, Sox2 and Nanog in human gliomas. Histopathology. 59:763–775. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Holmberg J, He X, Peredo I, Orrego A, Hesselager G, Ericsson C, Hovatta O, Oba-Shinjo SM, Marie SK, Nistér M, et al: Activation of neural and pluripotent stem cell signatures correlates with increased malignancy in human glioma. PLoS One. 6:e184542011. View Article : Google Scholar : PubMed/NCBI

35 

Ikushima H, Todo T, Ino Y, Takahashi M, Saito N, Miyazawa K and Miyazono K: Glioma-initiating cells retain their tumorigenicity through integration of the Sox axis and Oct4 protein. J Biol Chem. 286:41434–41441. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Muto J, Imai T, Ogawa D, Nishimoto Y, Okada Y, Mabuchi Y, Kawase T, Iwanami A, Mischel PS, Saya H, et al: RNA-binding protein Musashi1 modulates glioma cell growth through the post-transcriptional regulation of Notch and PI3 kinase/Akt signaling pathways. PLoS One. 7:e334312012. View Article : Google Scholar : PubMed/NCBI

37 

Mühlethaler-Mottet A, Liberman J, Ascenção K, Flahaut M, Balmas Bourloud K, Yan P, Jauquier N, Gross N and Joseph JM: The CXCR4/CXCR7/CXCL12 axis is involved in a secondary but complex control of neuroblastoma metastatic cell homing. PLoS One. 10:e01256162015. View Article : Google Scholar : PubMed/NCBI

38 

Lee CC, Lai JH, Hueng DY, Ma HI, Chung Y, Sun YY, Tsai YJ, Wu WB and Chen CL: Disrupting the CXCL12/CXCR4 axis disturbs the characteristics of glioblastoma stem-like cells of rat RG2 glioblastoma. Cancer Cell Int. 13:852013. View Article : Google Scholar : PubMed/NCBI

39 

Bier A, Giladi N, Kronfeld N, Lee HK, Cazacu S, Finniss S, Xiang C, Poisson L, deCarvalho AC, Slavin S, et al: MicroRNA-137 is downregulated in glioblastoma and inhibits the stemness of glioma stem cells by targeting RTVP-1. Oncotarget. 4:665–676. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Fareh M, Turchi L, Virolle V, Debruyne D, Almairac F, de-la- Forest Divonne S, Paquis P, Preynat-Seauve O, Krause K-H, Chneiweiss H, et al: The miR 302–367 cluster drastically affects self-renewal and infiltration properties of glioma-initiating cells through CXCR4 repression and consequent disruption of the SHH-GLI-NANOG network. Cell Death Differ. 19:232–244. 2012. View Article : Google Scholar :

41 

Chen Q, Zhang M, Li Y, Xu D, Wang Y, Song A, Zhu B, Huang Y and Zheng JC: CXCR7 mediates neural progenitor cells migration to CXCL12 independent of CXCR4. Stem Cells. 33:2574–2585. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Ma J, Yao Y, Wang P, Liu Y, Zhao L, Li Z, Li Z and Xue Y: MiR-152 functions as a tumor suppressor in glioblastoma stem cells by targeting Krüppel-like factor 4. Cancer Lett. 355:85–95. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Zhu XY, Wang L, Luan SH, Zhang HS, Huang WT and Wang NH: The PGI-KLF4 pathway regulates self-renewal of glioma stem cells residing in the mesenchymal niches in human gliomas. Neoplasma. 61:401–410. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2016
Volume 48 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Flüh C, Hattermann K, Mehdorn HM, Synowitz M and Held-Feindt J: Differential expression of CXCR4 and CXCR7 with various stem cell markers in paired human primary and recurrent glioblastomas. Int J Oncol 48: 1408-1416, 2016
APA
Flüh, C., Hattermann, K., Mehdorn, H.M., Synowitz, M., & Held-Feindt, J. (2016). Differential expression of CXCR4 and CXCR7 with various stem cell markers in paired human primary and recurrent glioblastomas. International Journal of Oncology, 48, 1408-1416. https://doi.org/10.3892/ijo.2016.3354
MLA
Flüh, C., Hattermann, K., Mehdorn, H. M., Synowitz, M., Held-Feindt, J."Differential expression of CXCR4 and CXCR7 with various stem cell markers in paired human primary and recurrent glioblastomas". International Journal of Oncology 48.4 (2016): 1408-1416.
Chicago
Flüh, C., Hattermann, K., Mehdorn, H. M., Synowitz, M., Held-Feindt, J."Differential expression of CXCR4 and CXCR7 with various stem cell markers in paired human primary and recurrent glioblastomas". International Journal of Oncology 48, no. 4 (2016): 1408-1416. https://doi.org/10.3892/ijo.2016.3354