Next Article in Journal
Arsenic Activates the ER Stress-Associated Unfolded Protein Response via the Activating Transcription Factor 6 in Human Bronchial Epithelial Cells
Next Article in Special Issue
CK2 and the Hallmarks of Cancer
Previous Article in Journal
Recent Advances in Biocatalysis for Drug Synthesis
Previous Article in Special Issue
The Immune Regulatory Role of Protein Kinase CK2 and Its Implications for Treatment of Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Inhibition of CK2 Reduces NG2 Expression in Juvenile Angiofibroma

1
Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
2
Department of Otolaryngology, Saarland University Medical Center, 66421 Homburg, Germany
3
Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(5), 966; https://doi.org/10.3390/biomedicines10050966
Submission received: 11 January 2022 / Revised: 19 April 2022 / Accepted: 20 April 2022 / Published: 21 April 2022
(This article belongs to the Special Issue CK2 Regulation of Cell Death and Targeting in Cancer Treatment)

Abstract

:
Juvenile angiofibroma (JA) is a rare fibrovascular neoplasm predominately found within the posterior nasal cavity of adolescent males. JA expresses the proteoglycan nerve–glial antigen (NG)2, which crucially determines the migratory capacity of distinct cancer cells. Moreover, it is known that the protein kinase CK2 regulates NG2 gene expression. Therefore, in the present study, we analyzed whether the inhibition of CK2 suppresses NG2-dependent JA cell proliferation and migration. For this purpose, we assessed the expression of NG2 and CK2 in patient-derived JA tissue samples, as well as in patient-derived JA cell cultures by Western blot, immunohistochemistry, flow cytometry and quantitative real-time PCR. The mitochondrial activity, proliferation and migratory capacity of the JA cells were determined by water-soluble tetrazolium (WST)-1, 5-bromo-2′-deoxyuridine (BrdU) and collagen sprouting assays. We found that NG2 and CK2 were expressed in both the JA tissue samples and cell cultures. The treatment of the JA cells with the two CK2 inhibitors, CX-4945 and SGC-CK2-1, significantly reduced NG2 gene and protein expression when compared to the vehicle-treated cells. In addition, the loss of CK2 activity suppressed the JA cell proliferation and migration. These findings indicate that the inhibition of CK2 may represent a promising therapeutic approach for the treatment of NG2-expressing JA.

1. Introduction

Juvenile angiofibroma (JA) is a pseudo-encapsulated mass, consisting of vascular and fibrous stromal tissue. Although it is a benign tumor, JA is capable of spreading through natural foraminas and fissures affecting the pterygopalatine and intratemporal fossa with erosion of the medial pterygoid lamina [1,2]. In addition, the extensive vascularization of this tumor and extension into the orbits and skull base may complicate surgical interventions due to a substantial risk of intraoperative bleeding [3,4].
In the last years, intensive research efforts have been made to find alternative strategies for the treatment of JA [5,6,7,8]. Targeting specific proteins that control JA growth and invasion may represent a particularly effective therapeutic approach [7,9]. In this context, we already showed a higher expression of the proteoglycan nerve glial (NG)2, also known as chondroitin sulfate proteoglycan 4 (CSPG4), in JA when compared to the control nasal mucosa tissue samples [10]. NG2 is a type-1 transmembrane protein. Its expression is restricted to distinct cell types, including pericytes, different progenitor cells and glioblastoma multiforme (GBM) cells [11]. The extracellular domain of the proteoglycan interacts with components of the extracellular matrix (ECM) [12], as well as with other membrane proteins, such as integrin β1, platelet-derived growth factor (PDGF)-α and fibroblast growth factor (FGF)2 [13]. These interactions trigger signaling transductions promoting cell proliferation and cell motility [14,15,16,17,18].
Of interest, we have recently identified the protein kinase CK2 as a novel regulator of NG2-dependent signaling pathways in pericytes and GBM [19,20]. This kinase is ubiquitously expressed and consists of two catalytic (CK2α or CK2α′) and two regulatory (CK2β) subunits [21]. CK2 has more than 500 substrates and, thus, is involved in various cellular processes, such as differentiation, cell cycle regulation and energy metabolism [22]. Moreover, the overexpression of CK2 promotes the development and progression of benign and malignant tumors [22,23,24,25]. Hence, several CK2 inhibitors have been developed as potential anti-cancer drugs, including CIGB-300 [26], SGC-CK2-1 [27] and CX-4945 [28].
In the present study, we analyzed whether CK2 inhibition reduces the cell proliferation and cell migration of JA by downregulating NG2 expression. To test this, we examined the effect of the two CK2 inhibitors, SGC-CK2-1 and CX-4945, on NG2 expression, mitochondrial activity, cell proliferation and cell migration in patient-derived JA cell cultures.

2. Materials and Methods

2.1. Antibodies

The anti-NG2 antibody (sc-166251) and the anti-CK2β antibody (E9) were from Santa Cruz Biotechnology (Heidelberg, Germany). The anti-α-tubulin antibody (66031) was from Proteintech Germany GmbH (St. Leon-Rot, Germany). The anti-AKT1/2/3 antibodies (11E7) and poly (ADP-ribose) polymerase (PARP) (9542) were from Cell Signaling (Frankfurt am Main, Germany). The anti-pAKTS129 antibody (ab133458) and the anti-NG2 antibody (ab129051) were from Abcam (Cambridge, UK). The anti-CD31 and anti-vimentin antibodies were from Dako Agilent (Hamburg, Germany). The anti-CK2α and anti-CK2β antibodies were generated, as described previously [29]. The peroxidase-labeled anti-rabbit antibody (NIF 824) and the peroxidase-labeled anti-mouse antibody (NIF 825) were from GE healthcare (Freiburg, Germany). The anti-chondroitin sulfate proteoglycan 4 (NG2; 562415) was from BD Biosciences (Heidelberg, Germany). The anti-5-bromo-2′-deoxyuridine (BrdU) antibody was from eBioscience (Thermo Fisher Scientific, Karlsruhe, Germany).

2.2. Patient-Derived JA Tissue Samples

Paraffin-embedded archived tissue samples of 5 male patients (age: 13–20 years) operated and diagnosed at Saarland University Medical Center between 2016–2018 were analyzed in this study. The tumor stage ranged from I1 to III3. Written informed consent was obtained from all the patients and the use of the human tissues was in accordance with the code of ethics of the World Medical Association (Declaration of Helsinki), as well as approved by the Institutional Review Board (#218/10) at Saarland University.

2.3. Cell Culture

The JA cell cultures were generated from at least 1 cm3 of native tumor samples, as described previously [30]. Briefly, the tissue samples were mechanically dissected and cultivated in DMEM F/12 Glutamax (Thermo Fisher Scientific, Darmstadt, Germany) supplemented with 10% fetal calf serum (FCS) (Thermo Fisher Scientific, Darmstadt, Germany), 1% penicillin–streptomycin, 1% sodium pyruvate, 10 µg/mL gentamycin (PAN-Biotech GmbH, Aidenbach, Germany) and 2.5 µg/mL amphotericin B (Thermo Fisher Scientific, Darmstadt, Germany) at 37 °C and 5% CO2. The medium was changed twice a week and the cells were passaged at 70% confluence by a split ratio of 1:3 with accutase solution (PromoCell, Heidelberg, Germany).

2.4. Immunohistology

For the preparation of immunohistological sections, the specimens of JA tissue were fixed for 24 h in 4% formalin. Thereafter, the sections were stained with the antibodies anti-CD31, anti-vimentin, anti-NG2, anti-CK2α and anti-CK2β. The DAKO Fast Red Kit (K5005, Agilent, Frankfurt, Germany) was used for detection according to the manufacturer’s instructions. Afterwards, the sections were counterstained with Mayer’s Hematoxylin (MHS32, Merck, Darmstadt, Germany). The quantification of the positively stained cells was performed per section by FIJI software (NIH, Bethesda, Maryland, USA) and is given as a % of all the cells per section.
For further immunohistochemical analyses, the sections were stained with anti-NG2 and anti-vimentin antibodies, which were detected by their corresponding fluorescence-coupled secondary antibodies. Hoechst 33342 was used to stain the cell nuclei (2 µg/mL). The sections were analyzed by means of fluorescence microscopy (BX61; Olympus, Hamburg, Germany).

2.5. Western Blot Analysis

The JA tissue samples, untreated JA cells and JA cells treated with CX-4945 (10 µM), SGC-CK2-1 (5 µM) or DMSO were lysed and the protein concentration was measured using a Bradford protein assay (Bio-Rad Laboratories, Feldkirchen, Germany). Bovine serum albumin (BSA) was used as standard. The JA whole cell extracts were separated through a 7.5% and 12% SDS-polyacrylamide gel and subsequently transferred onto a polyvinylidene difluoride (PVDF) membrane (Bio-Rad Laboratories, Feldkirchen, Germany). After blocking with 5% BSA in tris-buffered saline (TBS) (0.1% Tween20) for 1 h, the membrane was incubated with the primary antibodies against NG2, CK2α, CK2β, α-tubulin, pAKTS129 and AKT (dilution 1:100) in TBS (0.1% Tween20; 1% BSA) overnight followed by the corresponding secondary antibodies (1:1000). The protein expression was visualized by luminol-enhanced chemiluminescence (ECL) Western blotting substrate (Bio-Rad Laboratories, Feldkirchen, Germany) in an ECL ChemoCam Imager (Intas, Göttingen, Germany). The intensity of the measured signals was quantified using ImageJ software (NIH, Bethesda, MD, USA) and normalized to the loading control (α-tubulin).

2.6. CK2 Phosphorylation Assay

Cell extracts from the JA tissue samples were prepared for kinase filter assays, as previously described in detail [31]. The incorporation rate of [32P] phosphate into the CK2-specific substrate peptide with the sequence RRRDDDSDDD was measured. Twenty µL of kinase buffer (50 mM Tris/HCl, pH 7.5, 100 mM NaCl, 10 mM MgCl2, 1 mM DTT) containing 20 µg of proteins was mixed with 30 µL of CK2 mix (25 mM Tris/HCl, pH 8.5, 150 mM NaCl, 5 mM MgCl2, 50 µM ATP, 1 mM DTT, 0.19 mM substrate peptide) containing 10 µCi/500 µL [32P]yATP. The mixture was spotted onto a P81 ion exchange paper. The paper was washed three times with 85 mM H3PO4. After treatment with ethanol, the paper was dried and the Čerenkov radiation was determined in a scintillation counter.

2.7. Flow Cytometry

The untreated JA cells were washed with phosphate-buffered saline (PBS) and harvested with an enzyme-free dissociation buffer (Thermo Fisher Scientific, Darmstadt, Germany). The cells were incubated with a phycoerythrin (PE)-labeled primary anti-NG2 antibody for 30 min at room temperature. Thereafter, the cells were washed with PBS and the mean fluorescence intensity (MFI) of 1000 cells was analyzed by a FACSLyric™ flow cytometer (BD, Heidelberg, Germany). The non-expressing NG2 cell line HEK293 was used as a negative control.
To determine cell proliferation, the JA cells were treated with CX-4945 (10 µM), SGC-CK2-1 (5 µM) or DMSO and exposed to 10 µM BrdU for 48 h. Thereafter, the cells were fixed, permeabilized and incubated with an anti-BrdU antibody. The cells were then washed with PBS and the MFI of 500 cells was analyzed by a FACSLyric™ flow cytometer (BD, Heidelberg, Germany).

2.8. Water-Soluble Tetrazolium (WST)-1 Assay

A WST-1 assay (Roche, Mannheim, Germany) was used to analyze the effect of the inhibitors CX-4945 (ActivateScientific, Prien, Germany) and SGC-CK2-1 (Merk, Darmstadt, Germany) on the mitochondrial activity of the JA cells. Briefly, the cells were seeded in a 96-well culture plate at a density of 2 × 103 cells/well. Thereafter, they were treated with 1 µM, 5 µM and 10 µM of CX-4945 and SGC-CK2-1 or the equivalent concentrations of the vehicle (DMSO). After 24 h and 48 h, 10 μL of WST-1 reagent was added into each well and the absorbance was measured at 450 nm in a microplate photometer.

2.9. Quantitative Real-Time PCR (qRT-PCR)

Total RNA was extracted with a QIAzol lysis reagent and transcribed into cDNA by using qScriber (highQu, Kraichtal, Germany) according to the manufacturer’s protocol. Briefly, the qRT-PCR analysis was performed by using ORA™ SEE qPCR Green ROX L Mix (highQu, Kraichtal, Germany). Forward and reverse primers (NG2 forward 5′-GCAAGCCGATGTGGATTC-3′ and reverse 5′-ATGGCGGATGGTAGGATG-3′; Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) forward 5′-CCACCCATGGCAAATTCC-3′ and reverse 5′-ACTCCACGACGTACTCAG-3′) were used at a concentration of 500 nM. GAPDH was used as an endogenous control. Data collection was performed by means of a CFX96™ real-time system (Bio-Rad Laboratories, Feldkirchen, Germany) and the 2−ΔΔct method.

2.10. Spheroid Sprouting Assay

The migratory capacity of the JA cells was determined by means of a sprouting assay. For this, 500 cells per well were seeded into 96-well plates coated with 100 µL 1% agarose to form spheroids. After 24 h, 30 spheroids were collected, resuspended in a collagen solution and transferred into a well of a 24-well plate. DMEM was added to the wells (0 h) and the spheroids were incubated for 48 h in the presence of DMSO, CX-4945 (10 µM) and SGC-CK2-1 (5 µM) at 37 °C and 5% CO2. The sprouting capacity was visualized by bright field microscopy. The sprouting area was assessed by means of the ImageJ software (NIH, Bethesda, Maryland, USA).

2.11. Statistical Analysis

After testing the data for normal distribution and equal variance, the differences between the two groups were assessed by the unpaired Student’s t-test. Statistics were performed by GraphPad Prism (Prism 8 software, GraphPad, San Diego, CA, USA). All the values are expressed as the mean ± SD. Statistical significance was accepted for p < 0.05.

3. Results

3.1. Expression of CK2 and NG2 in Patient-Derived JA Tissue Samples

JA consists of vascular and fibrous stromal tissue [1,2]. To assess whether the expression of CK2 and NG2 is restricted to distinct cell types, we first performed immunohistochemical stainings of a patient-derived JA tissue sample (JA1) (Figure 1A). CD31 and vimentin were stained to visualize the vascular and fibrotic areas, respectively (Figure 1A). We detected NG2 expression in the cells surrounding the blood vessels, as well as in the fibrotic tissue areas (Figure 1A). Similar expression patterns could be found for the CK2 subunits CK2α and CK2β (Figure 1A). We additionally determined the number of CD31-, vimentin-, NG2-, CK2α- and CK2β-positive cells within the tissue sample (Figure 1B). As expected, ~10% of the cells were positive for CD31 and ~90% of the cells stained positive for vimentin. The number of NG2-, CK2α- and CK2β-positive cells was ~60% (Figure 1B).
Moreover, we investigated the expression of CK2 and NG2 in five patient-derived JA tissue samples (JA1–JA5). All the tissue samples expressed the catalytic CK2α subunit, the regulatory CK2β subunits and the surface protein NG2 (Figure 1C–F). We additionally analyzed CK2 kinase activity, which was detected in all the patient-derived tissue samples (Figure 1G). Of note, the expression pattern and activity of CK2α and CK2β markedly differed between the individual tissue samples.

3.2. Expression of CK2 and NG2 in Patient-Derived JA Cells

To study the influence of CK2 inhibition on NG2 expression in vitro, we additionally established cell cultures from the patient-derived JA1–JA4 tissue samples. The cells exhibited a fibroblast-like, multipolar shape in bright-field microscopy (Figure 2A). Immunofluorescence stainings demonstrated that the cells were positive for NG2 and for the intermediate filament protein vimentin (Figure 2A). Further Western blot analyses revealed that the cells from JA1–JA4 express NG2 as well as CK2α and CK2β (Figure 2B–E). We additionally verified NG2 protein expression in all the tissue samples by flow cytometry and NG2 gene expression by qRT-PCR analyses (Figure 2F,G).

3.3. CK2 Inhibition Suppresses the Proliferation of Patient-Derived JA Cells

The effect of CK2 inhibition on NG2 expression was investigated in the two cell cultures JA1 and JA2. For this purpose, the cells were cultured for 24 h or 48 h with increasing concentrations of the CK2 inhibitors CX-4945 and SGC-CK2-1. Subsequently, the mitochondrial activity of the cells was assessed by means of a WST-1 assay (Figure 3A–D). We found that only 5 µM and 10 µM of CX-4945 attenuate the activity of the JA1 cells after 24 h when compared to the vehicle (Figure 3A). After a longer period of 48 h, both compounds significantly decreased the formazan reduction in a dose-dependent manner (Figure 3B). In contrast, treatment with CX-4945 or SGC-CK2-1 reduced the mitochondrial activity of the JA2 cells after 24 h, which was even more pronounced after 48 h (Figure 3C,D). Of note, the lowest activity after CK2 inhibition was detected in the cells treated with 10 µM CX-4945 or 5 µM SGC-CK2-1 for 48 h. Therefore, we decided to use these concentrations over 48 h to study the effect of CK2 inhibition on cell proliferation and apoptosis. First, we analyzed whether both compounds effectively inhibited the CK2 activity in the JA1 and JA2 cells. Our Western blot analyses revealed an attenuated AKTS129 phosphorylation after exposure of the cells to CX-4945 and SGC-CK2-1 (Figure 3E,F). As expected, the two inhibitors significantly reduced cell proliferation, as shown by a lower number of BrdU-positive cells (Figure 3G,H). Moreover, the Western blot analyses revealed an increased, but not significantly higher, protein level of cleaved PARP (Figure 3I,J).

3.4. CK2 Inhibition Reduces NG2 Expression and the Migratory Capacity of Patient-Derived JA Cells

We next analyzed the effect of CX-4945 and SGC-CK2-1 on NG2 expression. We detected a markedly decreased expression of NG2 in the JA1 and JA2 cells (Figure 4A–C). In contrast, the expression of CK2α and CK2β was not affected by CX-4945 or SGC-CK2-1 treatment (Figure 4A,D–G). Additional analyses of the NG2 gene expression in these cells revealed that the loss of CK2 activity significantly attenuates NG2 mRNA levels (Figure 4H,I).
The binding of NG2 to proteins of the ECM triggers cell migration [12,32,33]. To analyze the influence of CK2 on NG2-dependent JA cell migration, we finally performed a collagen sprouting assay. For this purpose, 500 JA1 or JA2 cells were cultivated by using the liquid overlay technique to generate spheroids, which were subsequently embedded within a collagen matrix. The sprouting capacity of the cells was assessed over 48 h. We found that the cells of the vehicle-treated JA1 and JA2 spheroids rapidly grew out. This outgrowth was more pronounced in the JA1 spheroids compared to the JA2 spheroids (Figure 5A). Notably, in both groups, CK2 inhibition with CX-4945 or SGC-CK2-1 significantly reduced the sprouting capacity of the spheroids (Figure 5B,C).

4. Discussion

In the present study, we demonstrated that the pharmacological inhibition of CK2 by SGC-CK2-1 and CX-4945 significantly reduces NG2 gene expression in the patient-derived JA cells. Moreover, we detected a markedly decreased proliferation and migration in the JA cells after CK2 inhibition. JA are pseudo-encapsulated, heterogeneous benign tumors consisting of abundant vascular channels lacking the normal muscular layer in the channel wall and a network of fibrocollagenous tissue [34]. We already showed that NG2 mRNA expression is upregulated in JA when compared to the control nasal mucosa tissues [10]. This indicates that this proteoglycan may play an important role in JA cell growth and migration. In contrast, the expression pattern of CK2 in JA has not been analyzed so far. Of interest, our results demonstrate for the first time that NG2, as well as the CK2 subunits CK2α and CK2β, are expressed in all the patient-derived JA tissue samples.
During the last years, several studies reported that the selective inhibition of CK2 induces apoptosis in tumor cells relative to normal cells [35]. Accordingly, many CK2 inhibitors have been generated as anti-cancer drugs [28,36,37,38,39]. For instance, CX-4945 (IC50 = 360 nM) is currently being tested in phase I and II clinical trials for the treatment of various cancer types, including medulloblastoma (NCT03904862), cholangiocarcinoma (NCT02128282) and multiple myeloma (NCT01199718). Recently, Wells et al. [27] suggested SGC-CK2-1 (IC50 = 36 nM) as an efficient CK2 inhibitor [27]. This molecule is considered as non-toxic and, thus, has been proposed to be suitable not only for the treatment of cancers, but also for other pathological conditions, such as neurodegenerative diseases [27,40].
To study the effects of CX-4945 and SGC-CK2-1 on NG2 expression in JA, we first established cell cultures from the JA tissue samples, which is quite challenging due to the scarcity and the heterogeneity of this tumor type [10]. We showed that the JA cells expressed both NG2 and CK2. We next determined suitable concentrations that efficiently suppress CK2 activity. The exposure of the JA cells to 10 µM of CX-4945 and 5 µM of SGC-CK2-1 reduced the phosphorylation of AKT at serine 129, a specific CK2 phosphorylation site [41]. More importantly, both inhibitors significantly reduced the gene expression of NG2. This is in line with our previous studies showing that the loss of CK2 activity decreases NG2 gene expression in human placenta pericytes and in human NG2-positive GBM cells due to the inhibitory action of the transcription factor Sp1 [19,20,42,43]. Sp1 regulates the expression of multiple genes and its overexpression contributes to the malignant phenotype of a variety of human cancers by upregulating genes that enhance proliferation and migration [44]. The expression of Sp1 in JA has not been investigated so far. Hence, we can only assume that CK2 also regulates the expression of NG2 via Sp1 in JA.
NG2 promotes cell migration via its binding to the ECM [12,32,33]. Hence, it is conceivable that a diminished NG2 expression caused by CK2 inhibition results in a markedly decreased cell migration. In fact, our collagen matrix-based spheroid sprouting assays revealed that treatment with CX-4945 and SGC-CK2-1 significantly reduces the sprouting capacity of the JA cells. Notably, JA contains a higher amount of collagen type VI when compared to the control nasal mucosa [10]. Moreover, it is known that the specific binding of NG2 to collagen type VI triggers cell migration by convergent signal transduction pathways [45]. These findings indicate that the inhibition of CK2 and the resulting downregulation of NG2 expression may be highly effective for the selective treatment of JA.
In the last decade, several studies described the involvement of specific genes and signaling pathways, such as Wnt, in JA progression [46]. In particular, Wnt5B, a member of the Wnt family, is overexpressed in JA [47]. Of note, CK2 acts as a positive regulator of Wnt signaling [48] and several studies have already suggested that pharmacological CK2 inhibition may be a promising therapeutic approach for the treatment of tumors with high Wnt activity, such as chronic lymphocytic leukemia and B-cell lymphomas [49,50]. Accordingly, it may be speculated that besides the downregulation of NG2, the suppression of Wnt signaling by CK2 inhibition also contributed to the reduced migratory activity of the JA cells in our spheroid sprouting assays.
Besides the effect of NG2 on cell migration, the proteoglycan also promotes cancer growth by altering the expression of genes regulating cell proliferation and apoptosis [32,51]. Accordingly, the inhibition of NG2-dependent pathways is thought to be a promising therapeutic approach for the treatment of NG2-positive tumors [52]. In the present study, we detected a significantly reduced cell proliferation of and a slightly increased induction of apoptosis in the JA cells after CK2 inhibition, which may be caused by the reduced NG2 expression. However, it should be noted that CK2 regulates cell cycle progression and apoptosis via the phosphorylation of different proteins [53,54,55,56,57]. Therefore, further detailed analyses are required to study the effects of the reduced NG2 expression after CK2 inhibition on these processes.
Treatment with the cytotoxic CK2 inhibitors 4,5,6,7-tetrabromobenzotriazole (TBB), 2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole (DMAT) and CX-4945 typically leads to massive necrotic and apoptotic cell death [58]. Although the effectiveness and benefit of these compounds have been acknowledged in numerous cancer studies, they have also been recognized as hazardous substances due to their potential mutagenic, carcinogenic and reproductive toxicity properties [28,38,58,59,60,61]. In contrast, the novel inhibitor SGC-CK2-1 is not only highly specific for CK2, but also non-toxic [27]. Considering our novel finding that the inhibition of CK2 reduces NG2 expression and NG2-dependent cell migration in JA, SGC-CK2-1 may, thus, be a promising candidate for the future treatment of this benign tumor type, which is found particularly in young patients.

Author Contributions

A.S.B., S.W. (Silke Wemmert), S.W. (Selina Wrublewsky), C.G., M.M. and E.A. conceived and designed the experiments. A.S.B., S.W. (Silke Wemmert), C.G., P.K., S.W. (Selina Wrublewsky) and E.A. performed the experiments. A.S.B., S.W. (Silke Wemmert), B.S., M.M., S.W. (Selina Wrublewsky), C.G. and E.A. analyzed the data. E.A., M.M., B.S., M.D.M. and M.W.L. contributed the reagents, materials and analysis tools. A.S.B., M.W.L., B.S., M.D.M. and E.A. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Rolf M. Schwiete Stiftung, Mannheim, Germany, project Nr. 29/2019 to E.A. and the APC was funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) and Saarland University within the funding programme Open Access Publishing.

Institutional Review Board Statement

The study was approved by the local German ethical board (Ethikkommission der Ärztekammer des Saarlandes, Saarbrücken, Germany, General Medical Council of the State Saarland, NO 218/10).

Informed Consent Statement

Informed consent was obtained from all the subjects involved in the study.

Data Availability Statement

Data are contained within the article.

Acknowledgments

We are grateful for the excellent technical assistance of Caroline Bickelmann (Institute for Clinical and Experimental Surgery, Saarland University), and Carolin Bick and Ulrike Bechtel (Department of Otolaryngology, Saarland University Medical Center).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Li, W.; Ni, Y.; Lu, H.; Hu, L.; Wang, D. Current perspectives on the origin theory of juvenile nasopharyngeal angiofibroma. Discov. Med. 2019, 27, 245–254. [Google Scholar]
  2. Alshaikh, N.A.; Eleftheriadou, A. Juvenile nasopharyngeal angiofibroma staging: An overview. Ear Nose Throat J. 2015, 94, E12–E22. [Google Scholar] [CrossRef] [Green Version]
  3. Overdevest, J.B.; Amans, M.R.; Zaki, P.; Pletcher, S.D.; El-Sayed, I.H. Patterns of vascularization and surgical morbidity in juvenile nasopharyngeal angiofibroma: A case series, systematic review, and meta-analysis. Head Neck 2018, 40, 428–443. [Google Scholar] [CrossRef]
  4. Mehan, R.; Rupa, V.; Lukka, V.K.; Ahmed, M.; Moses, V.; Shyam Kumar, N.K. Association between vascular supply, stage and tumour size of juvenile nasopharyngeal angiofibroma. Eur. Arch. Oto-Rhino-Laryngol. 2016, 273, 4295–4303. [Google Scholar] [CrossRef]
  5. Min, H.J.; Chung, H.J.; Kim, C.H. Delayed cerebrospinal fluid rhinorrhea four years after gamma knife surgery for juvenile angiofibroma. J. Craniofacial Surg. 2014, 25, e565–e567. [Google Scholar] [CrossRef]
  6. Sitenga, G.; Granger, P.; Hepola, K.; Aird, J.; Silberstein, P.T. The use of flutamide for the neoadjuvant treatment of juvenile nasopharyngeal angiofibroma: A review of the literature comparing results by pubertal status and tumor stage. Int. J. Dermatol. 2021. online ahead of print. [Google Scholar] [CrossRef]
  7. Le, T.; New, J.; Jones, J.W.; Usman, S.; Yalamanchali, S.; Tawfik, O.; Hoover, L.; Bruegger, D.E.; Thomas, S.M. Inhibition of fibroblast growth factor receptor with AZD4547 mitigates juvenile nasopharyngeal angiofibroma. Int. Forum Allergy Rhinol. 2017, 7, 973–979. [Google Scholar] [CrossRef]
  8. Wendler, O.; Dlugaiczyk, J.; Birk, S.; Schick, B. Anti-proliferative effect of glucocorticoids on mesenchymal cells in juvenile angiofibromas. Head Neck 2012, 34, 1615–1621. [Google Scholar] [CrossRef]
  9. Saylam, G.; Yucel, O.T.; Sungur, A.; Onerci, M. Proliferation, angiogenesis and hormonal markers in juvenile nasopharyngeal angiofibroma. Int. J. Pediatr. Otorhinolaryngol. 2006, 70, 227–234. [Google Scholar] [CrossRef]
  10. Gramann, M.; Wendler, O.; Haeberle, L.; Schick, B. Prominent collagen type VI expression in juvenile angiofibromas. Histochem. Cell Biol. 2009, 131, 155–164. [Google Scholar] [CrossRef]
  11. Ampofo, E.; Schmitt, B.M.; Menger, M.D.; Laschke, M.W. The regulatory mechanisms of NG2/CSPG4 expression. Cell Mol. Biol. Lett. 2017, 22, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Burg, M.A.; Nishiyama, A.; Stallcup, W.B. A central segment of the NG2 proteoglycan is critical for the ability of glioma cells to bind and migrate toward type VI collagen. Exp. Cell Res. 1997, 235, 254–264. [Google Scholar] [CrossRef] [PubMed]
  13. Stallcup, W.B.; Huang, F.J. A role for the NG2 proteoglycan in glioma progression. Cell Adhes. Migr. 2008, 2, 192–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ferrara, G.; Errede, M.; Girolamo, F.; Morando, S.; Ivaldi, F.; Panini, N.; Bendotti, C.; Perris, R.; Furlan, R.; Virgintino, D.; et al. NG2, a common denominator for neuroinflammation, blood-brain barrier alteration, and oligodendrocyte precursor response in EAE, plays a role in dendritic cell activation. Acta Neuropathol. 2016, 132, 23–42. [Google Scholar] [CrossRef] [Green Version]
  15. Fukushi, J.; Makagiansar, I.T.; Stallcup, W.B. NG2 proteoglycan promotes endothelial cell motility and angiogenesis via engagement of galectin-3 and alpha3beta1 integrin. Mol. Biol. Cell 2004, 15, 3580–3590. [Google Scholar] [CrossRef] [Green Version]
  16. Ozerdem, U.; Stallcup, W.B. Pathological angiogenesis is reduced by targeting pericytes via the NG2 proteoglycan. Angiogenesis 2004, 7, 269–276. [Google Scholar] [CrossRef] [Green Version]
  17. Rivera, Z.; Ferrone, S.; Wang, X.; Jube, S.; Yang, H.; Pass, H.I.; Kanodia, S.; Gaudino, G.; Carbone, M. CSPG4 as a target of antibody-based immunotherapy for malignant mesothelioma. Clin. Cancer Res. 2012, 18, 5352–5363. [Google Scholar] [CrossRef] [Green Version]
  18. Schmitt, B.M.; Laschke, M.W.; Roessler, O.G.; Huang, W.; Scheller, A.; Menger, M.D.; Ampofo, E. Nerve/glial antigen (NG) 2 is a crucial regulator of intercellular adhesion molecule (ICAM)-1 expression. Biochim. Biophys. Acta 2018, 1865, 57–66. [Google Scholar] [CrossRef]
  19. Schmitt, B.M.; Boewe, A.S.; Becker, V.; Nalbach, L.; Gu, Y.; Götz, C.; Menger, M.D.; Laschke, M.W.; Ampofo, E. Protein Kinase CK2 Regulates Nerve/Glial Antigen (NG)2-Mediated Angiogenic Activity of Human Pericytes. Cells 2020, 9, 1546. [Google Scholar] [CrossRef]
  20. Schmitt, B.M.; Boewe, A.S.; Götz, C.; Philipp, S.E.; Urbschat, S.; Oertel, J.; Menger, M.D.; Laschke, M.W.; Ampofo, E. CK2 Activity Mediates the Aggressive Molecular Signature of Glioblastoma Multiforme by Inducing Nerve/Glial Antigen (NG)2 Expression. Cancers 2021, 13, 1678. [Google Scholar] [CrossRef]
  21. Castello, J.; Ragnauth, A.; Friedman, E.; Rebholz, H. CK2-An Emerging Target for Neurological and Psychiatric Disorders. Pharmaceuticals 2017, 10, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Roffey, S.E.; Litchfield, D.W. CK2 Regulation: Perspectives in 2021. Biomedicines 2021, 9, 1361. [Google Scholar] [CrossRef] [PubMed]
  23. Chua, M.M.J.; Lee, M.; Dominguez, I. Cancer-type dependent expression of CK2 transcripts. PLoS ONE 2017, 12, e0188854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zheng, Y.; McFarland, B.C.; Drygin, D.; Yu, H.; Bellis, S.L.; Kim, H.; Bredel, M.; Benveniste, E.N. Targeting protein kinase CK2 suppresses prosurvival signaling pathways and growth of glioblastoma. Clin. Cancer Res. 2013, 19, 6484–6494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yenice, S.; Davis, A.T.; Goueli, S.A.; Akdas, A.; Limas, C.; Ahmed, K. Nuclear casein kinase 2 (CK-2) activity in human normal, benign hyperplastic, and cancerous prostate. Prostate 1994, 24, 11–16. [Google Scholar] [CrossRef] [PubMed]
  26. Perea, S.E.; Baladron, I.; Garcia, Y.; Perera, Y.; Lopez, A.; Soriano, J.L.; Batista, N.; Palau, A.; Hernandez, I.; Farina, H.; et al. CIGB-300, a synthetic peptide-based drug that targets the CK2 phosphoaceptor domain. Translational and clinical research. Mol. Cell. Biochem. 2011, 356, 45–50. [Google Scholar] [CrossRef] [PubMed]
  27. Wells, C.I.; Drewry, D.H.; Pickett, J.E.; Tjaden, A.; Kramer, A.; Muller, S.; Gyenis, L.; Menyhart, D.; Litchfield, D.W.; Knapp, S.; et al. Development of a potent and selective chemical probe for the pleiotropic kinase CK2. Cell Chem. Biol. 2021, 28, 546–558.e10. [Google Scholar] [CrossRef]
  28. Siddiqui-Jain, A.; Drygin, D.; Streiner, N.; Chua, P.; Pierre, F.; O’Brien, S.E.; Bliesath, J.; Omori, M.; Huser, N.; Ho, C.; et al. CX-4945, an orally bioavailable selective inhibitor of protein kinase CK2, inhibits prosurvival and angiogenic signaling and exhibits antitumor efficacy. Cancer Res. 2010, 70, 10288–10298. [Google Scholar] [CrossRef] [Green Version]
  29. Faust, M.; Schuster, N.; Montenarh, M. Specific binding of protein kinase CK2 catalytic subunits to tubulin. FEBS Lett. 1999, 462, 51–56. [Google Scholar] [CrossRef] [Green Version]
  30. Wemmert, S.; Willnecker, V.; Kulas, P.; Weber, S.; Lerner, C.; Berndt, S.; Wendler, O.; Schick, B. Identification of CTNNB1 mutations, CTNNB1 amplifications, and an Axin2 splice variant in juvenile angiofibromas. Tumour Biol. 2016, 37, 5539–5549. [Google Scholar] [CrossRef]
  31. Ampofo, E.; Rudzitis-Auth, J.; Dahmke, I.N.; Rössler, O.G.; Thiel, G.; Montenarh, M.; Menger, M.D.; Laschke, M.W. Inhibition of protein kinase CK2 suppresses tumor necrosis factor (TNF)-alpha-induced leukocyte-endothelial cell interaction. Biochim. Biophys. Acta 2015, 1852, 2123–2136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Al-Mayhani, M.T.; Grenfell, R.; Narita, M.; Piccirillo, S.; Kenney-Herbert, E.; Fawcett, J.W.; Collins, V.P.; Ichimura, K.; Watts, C. NG2 expression in glioblastoma identifies an actively proliferating population with an aggressive molecular signature. Neuro-Oncology 2011, 13, 830–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Schiffer, D.; Mellai, M.; Boldorini, R.; Bisogno, I.; Grifoni, S.; Corona, C.; Bertero, L.; Cassoni, P.; Casalone, C.; Annovazzi, L. The Significance of Chondroitin Sulfate Proteoglycan 4 (CSPG4) in Human Gliomas. Int. J. Mol. Sci. 2018, 19, 2724. [Google Scholar] [CrossRef] [Green Version]
  34. Midilli, R.; Karci, B.; Akyildiz, S. Juvenile nasopharyngeal angiofibroma: Analysis of 42 cases and important aspects of endoscopic approach. Int. J. Pediatr. Otorhinolaryngol. 2009, 73, 401–408. [Google Scholar] [CrossRef] [PubMed]
  35. Intemann, J.; Saidu, N.E.; Schwind, L.; Montenarh, M. ER stress signaling in ARPE-19 cells after inhibition of protein kinase CK2 by CX-4945. Cell. Signal. 2014, 26, 1567–1575. [Google Scholar] [CrossRef] [PubMed]
  36. Cozza, G.; Mazzorana, M.; Papinutto, E.; Bain, J.; Elliott, M.; di Maira, G.; Gianoncelli, A.; Pagano, M.A.; Sarno, S.; Ruzzene, M.; et al. Quinalizarin as a potent, selective and cell-permeable inhibitor of protein kinase CK2. Biochem. J. 2009, 421, 387–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Götz, C.; Gratz, A.; Kucklaender, U.; Jose, J. TF—A novel cell-permeable and selective inhibitor of human protein kinase CK2 induces apoptosis in the prostate cancer cell line LNCaP. Biochim. Biophys. Acta 2012, 1820, 970–977. [Google Scholar] [CrossRef]
  38. Prins, R.C.; Burke, R.T.; Tyner, J.W.; Druker, B.J.; Loriaux, M.M.; Spurgeon, S.E. CX-4945, a selective inhibitor of casein kinase-2 (CK2), exhibits anti-tumor activity in hematologic malignancies including enhanced activity in chronic lymphocytic leukemia when combined with fludarabine and inhibitors of the B-cell receptor pathway. Leukemia 2013, 27, 2094–2096. [Google Scholar] [CrossRef] [Green Version]
  39. Sarno, S.; Pinna, L.A. Protein kinase CK2 as a druggable target. Mol. Biosyst. 2008, 4, 889–894. [Google Scholar] [CrossRef]
  40. Salvi, M.; Borgo, C.; Pinna, L.A.; Ruzzene, M. Targeting CK2 in cancer: A valuable strategy or a waste of time? Cell Death Discov. 2021, 7, 325. [Google Scholar] [CrossRef]
  41. Di Maira, G.; Salvi, M.; Arrigoni, G.; Marin, O.; Sarno, S.; Brustolon, F.; Pinna, L.A.; Ruzzene, M. Protein kinase CK2 phosphorylates and upregulates Akt/PKB. Cell Death Differ. 2005, 12, 668–677. [Google Scholar] [CrossRef] [PubMed]
  42. Armstrong, S.A.; Barry, D.A.; Leggett, R.W.; Mueller, C.R. Casein kinase II-mediated phosphorylation of the C terminus of Sp1 decreases its DNA binding activity. J. Biol. Chem. 1997, 272, 13489–13495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Zhang, S.; Kim, K.H. Protein kinase CK2 down-regulates glucose-activated expression of the acetyl-CoA carboxylase gene. Arch. Biochem. Biophys. 1997, 338, 227–232. [Google Scholar] [CrossRef] [PubMed]
  44. Dupuis-Maurin, V.; Brinza, L.; Baguet, J.; Plantamura, E.; Schicklin, S.; Chambion, S.; Macari, C.; Tomkowiak, M.; Deniaud, E.; Leverrier, Y.; et al. Overexpression of the transcription factor Sp1 activates the OAS-RNAse L-RIG-I pathway. PLoS ONE 2015, 10, e0118551. [Google Scholar] [CrossRef] [PubMed]
  45. Cattaruzza, S.; Nicolosi, P.A.; Braghetta, P.; Pazzaglia, L.; Benassi, M.S.; Picci, P.; Lacrima, K.; Zanocco, D.; Rizzo, E.; Stallcup, W.B.; et al. NG2/CSPG4-collagen type VI interplays putatively involved in the microenvironmental control of tumour engraftment and local expansion. J. Mol. Cell Biol. 2013, 5, 176–193. [Google Scholar] [CrossRef] [PubMed]
  46. Ponti, G.; Losi, L.; Pellacani, G.; Rossi, G.B.; Presutti, L.; Mattioli, F.; Villari, D.; Wannesson, L.; Alicandri Ciufelli, M.; Izzo, P.; et al. Wnt pathway, angiogenetic and hormonal markers in sporadic and familial adenomatous polyposis-associated juvenile nasopharyngeal angiofibromas (JNA). Appl. Immunohistochem. Mol. Morphol. 2008, 16, 173–178. [Google Scholar] [CrossRef]
  47. Calanca, N.; Binato, S.M.S.; da Silva, S.D.; Brentani, H.P.; Sennes, L.U.; Pinto, C.A.L.; Domingues, M.A.C.; Fonseca-Alves, C.E.; Rainho, C.A.; Rogatto, S.R. Master Regulators of Epithelial-Mesenchymal Transition and WNT Signaling Pathways in Juvenile Nasopharyngeal Angiofibromas. Biomedicines 2021, 9, 1258. [Google Scholar] [CrossRef]
  48. Seldin, D.C.; Landesman-Bollag, E.; Farago, M.; Currier, N.; Lou, D.; Dominguez, I. CK2 as a positive regulator of Wnt signalling and tumourigenesis. Mol. Cell. Biochem. 2005, 274, 63–67. [Google Scholar] [CrossRef]
  49. Janovska, P.; Bryja, V. Wnt signalling pathways in chronic lymphocytic leukaemia and B-cell lymphomas. Br. J. Pharm. 2017, 174, 4701–4715. [Google Scholar] [CrossRef] [Green Version]
  50. Martins, L.R.; Perera, Y.; Lucio, P.; Silva, M.G.; Perea, S.E.; Barata, J.T. Targeting chronic lymphocytic leukemia using CIGB-300, a clinical-stage CK2-specific cell-permeable peptide inhibitor. Oncotarget 2014, 5, 258–263. [Google Scholar] [CrossRef] [Green Version]
  51. Hsu, S.C.; Nadesan, P.; Puviindran, V.; Stallcup, W.B.; Kirsch, D.G.; Alman, B.A. Effects of chondroitin sulfate proteoglycan 4 (NG2/CSPG4) on soft-tissue sarcoma growth depend on tumor developmental stage. J. Biol. Chem. 2018, 293, 2466–2475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Rolih, V.; Barutello, G.; Iussich, S.; De Maria, R.; Quaglino, E.; Buracco, P.; Cavallo, F.; Riccardo, F. CSPG4: A prototype oncoantigen for translational immunotherapy studies. J. Transl. Med. 2017, 15, 151. [Google Scholar] [CrossRef] [PubMed]
  53. Khan, D.H.; He, S.; Yu, J.; Winter, S.; Cao, W.; Seiser, C.; Davie, J.R. Protein kinase CK2 regulates the dimerization of histone deacetylase 1 (HDAC1) and HDAC2 during mitosis. J. Biol. Chem. 2013, 288, 16518–16528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Bandyopadhyay, K.; Li, P.; Gjerset, R.A. CK2-mediated hyperphosphorylation of topoisomerase I targets serine 506, enhances topoisomerase I-DNA binding, and increases cellular camptothecin sensitivity. PLoS ONE 2012, 7, e50427. [Google Scholar] [CrossRef] [Green Version]
  55. Schneider, C.C.; Götz, C.; Hessenauer, A.; Gunther, J.; Kartarius, S.; Montenarh, M. Down-regulation of CK2 activity results in a decrease in the level of cdc25C phosphatase in different prostate cancer cell lines. Mol. Cell. Biochem. 2011, 356, 177–184. [Google Scholar] [CrossRef]
  56. Ahmad, K.A.; Wang, G.; Unger, G.; Slaton, J.; Ahmed, K. Protein kinase CK2--a key suppressor of apoptosis. Adv. Enzym. Regul. 2008, 48, 179–187. [Google Scholar] [CrossRef] [Green Version]
  57. Wang, G.; Ahmad, K.A.; Ahmed, K. Role of protein kinase CK2 in the regulation of tumor necrosis factor-related apoptosis inducing ligand-induced apoptosis in prostate cancer cells. Cancer Res. 2006, 66, 2242–2249. [Google Scholar] [CrossRef] [Green Version]
  58. Strum, S.W.; Gyenis, L.; Litchfield, D.W. CSNK2 in cancer: Pathophysiology and translational applications. Br. J. Cancer 2022, 126, 994–1003. [Google Scholar] [CrossRef]
  59. Pierre, F.; Chua, P.C.; O’Brien, S.E.; Siddiqui-Jain, A.; Bourbon, P.; Haddach, M.; Michaux, J.; Nagasawa, J.; Schwaebe, M.K.; Stefan, E.; et al. Pre-clinical characterization of CX-4945, a potent and selective small molecule inhibitor of CK2 for the treatment of cancer. Mol. Cell. Biochem. 2011, 356, 37–43. [Google Scholar] [CrossRef]
  60. Pagano, M.A.; Meggio, F.; Ruzzene, M.; Andrzejewska, M.; Kazimierczuk, Z.; Pinna, L.A. 2-Dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole: A novel powerful and selective inhibitor of protein kinase CK2. Biochem. Biophys. Res. Commun. 2004, 321, 1040–1044. [Google Scholar] [CrossRef]
  61. Ruzzene, M.; Penzo, D.; Pinna, L.A. Protein kinase CK2 inhibitor 4,5,6,7-tetrabromobenzotriazole (TBB) induces apoptosis and caspase-dependent degradation of haematopoietic lineage cell-specific protein 1 (HS1) in Jurkat cells. Biochem. J. 2002, 364, 41–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Expression of CK2 and NG2 in the patient-derived JA tissue samples. (A) Representative immunohistochemical stainings of CD31, vimentin, NG2, CK2α and CK2β from JA1 (upper panel). Scale bars: 25 µm. Immunohistochemical staining without primary antibodies served as a negative control (ctrl, lower panel). Scale bars: 25 µm. (B) Quantitative analysis of (A). The number of CD31-, vimentin-, NG2-, CK2α- and CK2β-positive cells is given as % of all the cells per section. Mean ± SEM (n = 4). (C) JA tissue samples from 5 patients (JA1–JA5) were lysed and the expression of NG2, CK2α, CK2β and α-tubulin (as a loading control) was analyzed by Western blot. (DF) Quantitative analysis of (C). Data are expressed as the relative density ratio of NG2/α-tubulin (D), CK2α/α-tubulin (E) and CK2β/α-tubulin. (G) CK2 kinase activity (CPM) was measured by a CK2 kinase assay. Mean ± SD (n = 3).
Figure 1. Expression of CK2 and NG2 in the patient-derived JA tissue samples. (A) Representative immunohistochemical stainings of CD31, vimentin, NG2, CK2α and CK2β from JA1 (upper panel). Scale bars: 25 µm. Immunohistochemical staining without primary antibodies served as a negative control (ctrl, lower panel). Scale bars: 25 µm. (B) Quantitative analysis of (A). The number of CD31-, vimentin-, NG2-, CK2α- and CK2β-positive cells is given as % of all the cells per section. Mean ± SEM (n = 4). (C) JA tissue samples from 5 patients (JA1–JA5) were lysed and the expression of NG2, CK2α, CK2β and α-tubulin (as a loading control) was analyzed by Western blot. (DF) Quantitative analysis of (C). Data are expressed as the relative density ratio of NG2/α-tubulin (D), CK2α/α-tubulin (E) and CK2β/α-tubulin. (G) CK2 kinase activity (CPM) was measured by a CK2 kinase assay. Mean ± SD (n = 3).
Biomedicines 10 00966 g001
Figure 2. Expression of CK2 and NG2 in the patient-derived JA cells. (A) Bright field images (scale bars: 20 µm) and immunofluorescence stainings (scale bars: 50 µm) of NG2 (red), vimentin (green) and cell nuclei (blue) in the JA1–JA4 cells. (B) The JA1–JA4 cells were lysed and the expression of NG2, CK2α, CK2β and α-tubulin (as a loading control) was analyzed by Western blot. (CE) Quantitative analysis of (B). Data are expressed as the relative density ratio of NG2/α-tubulin (D), CK2α/α-tubulin (E) and CK2β/α-tubulin. (F) NG2 surface expression of the JA1–JA4 cells was detected by flow cytometry. Mean ± SD (n = 2). (G) RNA was extracted from the JA1–JA4 cells and the gene expression of NG2 was examined by qRT-PCR (% of JA1). Mean ± SD (n = 3).
Figure 2. Expression of CK2 and NG2 in the patient-derived JA cells. (A) Bright field images (scale bars: 20 µm) and immunofluorescence stainings (scale bars: 50 µm) of NG2 (red), vimentin (green) and cell nuclei (blue) in the JA1–JA4 cells. (B) The JA1–JA4 cells were lysed and the expression of NG2, CK2α, CK2β and α-tubulin (as a loading control) was analyzed by Western blot. (CE) Quantitative analysis of (B). Data are expressed as the relative density ratio of NG2/α-tubulin (D), CK2α/α-tubulin (E) and CK2β/α-tubulin. (F) NG2 surface expression of the JA1–JA4 cells was detected by flow cytometry. Mean ± SD (n = 2). (G) RNA was extracted from the JA1–JA4 cells and the gene expression of NG2 was examined by qRT-PCR (% of JA1). Mean ± SD (n = 3).
Biomedicines 10 00966 g002
Figure 3. CK2 inhibition suppresses the proliferation of the JA cells. (AD) The JA1 and JA2 cells were treated with the indicated concentrations of CX-4945 and SGC-CK2-1. The mitochondrial activity was measured by a WST-1 assay after 24 h and 48 h. Vehicle-treated cells served as controls and were set at 100%. Mean ± SD (n = 3). * p < 0.05 vs. vehicle. (E,F) The JA1 and JA2 cells were treated with CX-4945 (10 μM) and SGC-CK2-1 (5 μM) for 48 h. Vehicle-treated cells served as controls. The cells were lysed and the expression of AKT and pAKTS129 was assessed by Western blot. The relative density ratio of pAKTS129/AKT was determined and the vehicle-treated cells were set at 100%. Mean ± SD (n = 3). * p < 0.05 vs. vehicle. (G,H) JA1 and JA2 were treated as described in (E,F) and the incorporation of BrdU was analyzed by flow cytometry. The BrdU-positive cells were expressed to all the gated cells and the vehicle-treated cells were set at 100%. Mean ± SD (n = 3). * p < 0.05 vs. vehicle. (I,J) The JA1 and JA2 cells were treated with CX-4945 (10 μM) and SGC-CK2-1 (5 μM) for 48 h. Vehicle-treated cells served as controls. The cells were lysed and the expression of AKT and pAKTS129 was assessed by Western blot. The relative density ratio of cPARP/PARP was determined and the vehicle-treated cells were set at 100%. Mean ± SD (n = 3).
Figure 3. CK2 inhibition suppresses the proliferation of the JA cells. (AD) The JA1 and JA2 cells were treated with the indicated concentrations of CX-4945 and SGC-CK2-1. The mitochondrial activity was measured by a WST-1 assay after 24 h and 48 h. Vehicle-treated cells served as controls and were set at 100%. Mean ± SD (n = 3). * p < 0.05 vs. vehicle. (E,F) The JA1 and JA2 cells were treated with CX-4945 (10 μM) and SGC-CK2-1 (5 μM) for 48 h. Vehicle-treated cells served as controls. The cells were lysed and the expression of AKT and pAKTS129 was assessed by Western blot. The relative density ratio of pAKTS129/AKT was determined and the vehicle-treated cells were set at 100%. Mean ± SD (n = 3). * p < 0.05 vs. vehicle. (G,H) JA1 and JA2 were treated as described in (E,F) and the incorporation of BrdU was analyzed by flow cytometry. The BrdU-positive cells were expressed to all the gated cells and the vehicle-treated cells were set at 100%. Mean ± SD (n = 3). * p < 0.05 vs. vehicle. (I,J) The JA1 and JA2 cells were treated with CX-4945 (10 μM) and SGC-CK2-1 (5 μM) for 48 h. Vehicle-treated cells served as controls. The cells were lysed and the expression of AKT and pAKTS129 was assessed by Western blot. The relative density ratio of cPARP/PARP was determined and the vehicle-treated cells were set at 100%. Mean ± SD (n = 3).
Biomedicines 10 00966 g003
Figure 4. CK2 inhibition reduces the NG2 expression of the JA cells. (AD) The JA1 and JA2 cells were treated with CX-4945 (10 μM) and SGC-CK2-1 (5 μM) for 48 h. Vehicle-treated cells served as controls. The cells were lysed and the expression of NG2, CK2α, CK2β and α-tubulin (as a loading control) was assessed by Western blot. The relative density ratio of NG2/α-tubulin (B,C), CK2α/α-tubulin (D,E) and CK2β/α-tubulin (F,G) was determined and the vehicle-treated cells were set at 100%. Mean ± SD (n = 3). * p < 0.05 vs. vehicle. (H,I). The gene expression of NG2 was measured by qRT-PCR. Vehicle-treated cells were set at 100%. Mean ± SD (n = 3). * p < 0.05 vs. vehicle.
Figure 4. CK2 inhibition reduces the NG2 expression of the JA cells. (AD) The JA1 and JA2 cells were treated with CX-4945 (10 μM) and SGC-CK2-1 (5 μM) for 48 h. Vehicle-treated cells served as controls. The cells were lysed and the expression of NG2, CK2α, CK2β and α-tubulin (as a loading control) was assessed by Western blot. The relative density ratio of NG2/α-tubulin (B,C), CK2α/α-tubulin (D,E) and CK2β/α-tubulin (F,G) was determined and the vehicle-treated cells were set at 100%. Mean ± SD (n = 3). * p < 0.05 vs. vehicle. (H,I). The gene expression of NG2 was measured by qRT-PCR. Vehicle-treated cells were set at 100%. Mean ± SD (n = 3). * p < 0.05 vs. vehicle.
Biomedicines 10 00966 g004
Figure 5. CK2 inhibition reduces the migratory activity of the patient-derived JA cells. (A) The JA1 and JA2 cells were cultivated by means of the liquid overlay technique for 24 h and the resulting spheroids were embedded into a collagen matrix. Bright field images of the sprouting area around the spheroids (the border marked by a broken yellow line) were taken immediately after embedding (0 h) and 48 h after treatment with the vehicle, CX-4945 (10 µM) and SGC-CK2-1 (5 µM). Scale bars: 100 µm. (B,C) The sprouting area was assessed 48 h after the treatment of the JA1 (B) and JA2 (C) spheroids with the vehicle, CX-4945 (10 µM) and SGC-CK2-1 (5 µM). Data are expressed as a % of the vehicle-treated spheroids. Mean ± SD (n = 10). * p < 0.05 vs. vehicle.
Figure 5. CK2 inhibition reduces the migratory activity of the patient-derived JA cells. (A) The JA1 and JA2 cells were cultivated by means of the liquid overlay technique for 24 h and the resulting spheroids were embedded into a collagen matrix. Bright field images of the sprouting area around the spheroids (the border marked by a broken yellow line) were taken immediately after embedding (0 h) and 48 h after treatment with the vehicle, CX-4945 (10 µM) and SGC-CK2-1 (5 µM). Scale bars: 100 µm. (B,C) The sprouting area was assessed 48 h after the treatment of the JA1 (B) and JA2 (C) spheroids with the vehicle, CX-4945 (10 µM) and SGC-CK2-1 (5 µM). Data are expressed as a % of the vehicle-treated spheroids. Mean ± SD (n = 10). * p < 0.05 vs. vehicle.
Biomedicines 10 00966 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Boewe, A.S.; Wemmert, S.; Kulas, P.; Schick, B.; Götz, C.; Wrublewsky, S.; Montenarh, M.; Menger, M.D.; Laschke, M.W.; Ampofo, E. Inhibition of CK2 Reduces NG2 Expression in Juvenile Angiofibroma. Biomedicines 2022, 10, 966. https://doi.org/10.3390/biomedicines10050966

AMA Style

Boewe AS, Wemmert S, Kulas P, Schick B, Götz C, Wrublewsky S, Montenarh M, Menger MD, Laschke MW, Ampofo E. Inhibition of CK2 Reduces NG2 Expression in Juvenile Angiofibroma. Biomedicines. 2022; 10(5):966. https://doi.org/10.3390/biomedicines10050966

Chicago/Turabian Style

Boewe, Anne S., Silke Wemmert, Philipp Kulas, Bernhard Schick, Claudia Götz, Selina Wrublewsky, Mathias Montenarh, Michael D. Menger, Matthias W. Laschke, and Emmanuel Ampofo. 2022. "Inhibition of CK2 Reduces NG2 Expression in Juvenile Angiofibroma" Biomedicines 10, no. 5: 966. https://doi.org/10.3390/biomedicines10050966

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop