Skip to main content
Erschienen in: Cancer Cell International 1/2015

Open Access 01.12.2015 | Primary research

RB mutation and RAS overexpression induce resistance to NK cell-mediated cytotoxicity in glioma cells

verfasst von: Mario Orozco-Morales, Francisco Javier Sánchez-García, Irene Golán-Cancela, Norma Hernández-Pedro, Jose A. Costoya, Verónica Pérez de la Cruz, Sergio Moreno-Jiménez, Julio Sotelo, Benjamín Pineda

Erschienen in: Cancer Cell International | Ausgabe 1/2015

Abstract

Several theories aim to explain the malignant transformation of cells, including the mutation of tumor suppressors and proto-oncogenes. Deletion of Rb (a tumor suppressor), overexpression of mutated Ras (a proto-oncogene), or both, are sufficient for in vitro gliomagenesis, and these genetic traits are associated with their proliferative capacity. An emerging hallmark of cancer is the ability of tumor cells to evade the immune system. Whether specific mutations are related with this, remains to be analyzed. To address this issue, three transformed glioma cell lines were obtained (Rb−/−, RasV12, and Rb−/−/RasV12) by in vitro retroviral transformation of astrocytes, as previously reported. In addition, RasV12 and Rb−/−/RasV12 transformed cells were injected into SCID mice and after tumor growth two stable glioma cell lines were derived. All these cells were characterized in terms of Rb and Ras gene expression, morphology, proliferative capacity, expression of MHC I, Rae1δ, and Rae1αβγδε, mult1, H60a, H60b, H60c, as ligands for NK cell receptors, and their susceptibility to NK cell-mediated cytotoxicity. Our results show that transformation of astrocytes (Rb loss, Ras overexpression, or both) induced phenotypical and functional changes associated with resistance to NK cell-mediated cytotoxicity. Moreover, the transfer of cell lines of transformed astrocytes into SCID mice increased resistance to NK cell-mediated cytotoxicity, thus suggesting that specific changes in a tumor suppressor (Rb) and a proto-oncogene (Ras) are enough to confer resistance to NK cell-mediated cytotoxicity in glioma cells and therefore provide some insight into the ability of tumor cells to evade immune responses.
Hinweise

Competing interests

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Authors’ contributions

Conception and design: MOM; JACP; FJSG; BP. Collection and assembly of data: MOM, IGC; NHP, VPdlC, SMJ Data analysis and interpretation: MOM; JACP; FJSG; JS; BP. Manuscript writing: MOM; FJSG and BP. All authors read and approved the final manuscript.
Abkürzungen
Rb
Retinoblastoma
NK
Natural killer
MHC
Major Histocompatibility Complex

Background

Tumorigenesis is a multiple step process in which genetic alterations drive the progressive transformation of normal cells into highly malignant derivatives with well-known hallmarks [1, 2]. In addition, two “emerging hallmarks” of cancer have recently been proposed, namely deregulation of cellular energetics, and avoiding immune destruction [3]. Furthermore, neoplastic transformation drives genome instability and mutation, and tumor-induced inflammation [3, 4].
The idea that tumors must escape from immune recognition implies that tumors can be destroyed by the immune response [5]. However, some tumors generate an immune suppressive environment, thus evading immune destruction [6]. Gliomas are the most common primary tumors in the brain and are divided into four clinical grades on histopathological and prognosis basis [7]. Several gene expression alterations and chromosomal abnormalities are commonly found in gliomas and, in some instances, these mutations correlate with the clinical grade [8].
In most cancers, the oncogenic Ras is activated, and 20-30 % of all tumors harbor oncogenic point mutations in Ras. Moreover, if Ras is not mutated, such as in gliomas, it is frequently found that the Ras signaling pathway is disrupted [2].
On the other hand, the tumor suppressor Rb regulates cell cycle, inhibiting progression into the S phase, by inactivating the E2F transcription factor, which is critical for DNA replication. The Cancer Genome Atlas (TCGA) project has shown that CDKN2A/p16-CDK4/6-RB pathway is altered in nearly 80 % of primary GBMs with the most frequent genetic alterations being CDKN2A gene deletion or mutation, CDK4 amplification, and RB1 mutation or deletion [9, 10].
Natural killer cells (NK) are regarded as the first line of defense against tumors [11]. Therefore, taking advantage of an oncogenic Ras expression and Rb inactivation-based in vitro model of gliomagenesis, as previously reported [12], we explored whether these specific genetic alterations induce a cell phenotype compatible with glioma cell evasion from NK cell-mediated cytotoxicity. In addition, in vitro transformed glioma cells were injected into SCID mice and after tumor growth, two cell lines that survived the cytotoxic effect of mice NK cells were also analyzed and showed increased resistance to NK cell-mediated cytotoxicity. Together, our results suggest that overexpression of mutated Ras, down-regulation of Rb, or both genetic traits, confer in vitro resistance to NK cells and that in vivo NK cell-based selective pressure, selected cells with an increased in vitro resistance to NK cells.

Results

Characterization of in vitro transformed astrocytes

Four types of transformed astrocytes were obtained, named as cRb loxP/loxP , RasV12, cRb −/− , and cRb −/− /RasV12. Overexpression of Ras induced cell morphology heterogeneity, including elongated cytoplasm and multinucleated cells, and loss of contact inhibition, all characteristic traits of transformed cells. Primary astrocytes (cRb loxP/loxP ), and Rb-deficient astrocytes showed no significant morphologic alterations. All the four cell types tested positive for glial fibrillary acidic protein (GFAP), thus demonstrating their glial nature.
As expected, the presence of RasV12 was observed in the cell types in which RasV12 was constitutively activated (RasV12, and cRb -/ - /RasV12), and the lack of the Rb protein was observed in the cell types in which Rb gene was removed by the Cre recombinase (cRb −/− , cRb −/− /RasV12). Likewise, the activation of the DNA damage response, as assessed by the expression of p53, p-p53, and p-H2AX was higher in the cRb −/− /RasV12 cells.
No significant cell senescence, as assessed by SA-β-gal expression was observed in any of the transformed astrocytes, and the maximal proliferation rate was observed in the cRb −/− /RasV12 cells. All these results, shown in Fig. 1, were very similar to the reported by Seoane et al. [12], in addition to confirming previous data, the cells that were specifically derived for this work were characterized.

Rb mutation and overexpression of Ras modify the expression of ligands for NK cell receptors

To gain some insight into the mechanisms that confer tumor cells the ability to avoid immune destruction. We tested the expression of defined ligands for NK cell receptors, including MHC class I (an NK inhibiting receptor) and Rae1δ, Rae1αβγδε, mult1, H60a, H60b, H60c, as well as two molecules involved in programed cell death (Fas, and FasL); MHC class I, Rae1δ, and Rae1αβγδε, were analyzed by Western blot, whereas mult1 and H60a, H60b and H60c expression was analyzed by real time PCR. Figure 2a shows the normalized expression of MHC class I (a), Rae1δ (b), Rae1αβγδε (c), Fas (d), and FasL (e). Ligand expression is presented as the fold change, as compared to the expression of untransformed astrocytes. MHC class I expression was higher in cRb −/− and lower in Rb −/− /RasV12 astrocytes; Rae1δ expression was higher in cRb −/− , and lower in Rb −/− /RasV12 astrocytes; Rae1αβγδε expression was higher in cRb −/− , and lower in Rb −/− /RasV12 astrocytes; FasL expression was lower in RasV12 astrocytes, and Fas expression was lower in all transformed astrocytes. Figure 2b shows the mRNA expression of mult-1, H60a, H60b, and H60c. Mult1 expression was higher in RasV12 and Rb −/− /RasV12 than in T731 astrocytes, H60a and H60b expression was higher in cRb loxP/loxP , cRb −/− , Rb −/− /RasV12 and T731 astrocytes than in RasV12 astrocytes, H60b expression was higher in cRb loxP/loxP , cRb −/− , Rb −/− /RasV12 and T731 astrocytes than in RasV12 astrocytes, and H60c expression was higher in RasV12, Rb −/− /RasV12, T651 and T731 than in cRb loxP/loxP and cRb −/− astrocytes.

Rb mutation and overexpression of Ras induces resistance to NK cell mediated cytotoxicity

Transformed astrocytes were exposed to murine NK cells in order to assess their susceptibility to NK cell-mediated cytotoxicity. Figure 3 shows that in an effector to target ratio of 10:1, approximately 30 % of untransformed astrocytes were lysed by NK cells, whereas all transformed astrocytes tested were more resistant to NK cell-mediated cytotoxicity, as shown by the reduced percentages of specific lysis.

RasV12 and cRb −/− /RasV12 astrocytes produce tumors in FVB immunocompetent mice

To assess the capacity of transformed glioma cell lines to produce tumors in syngeneic immunocompetent mice, we injected 1x106 cells from either cRb loxP/loxP , RasV12, cRb -/ -, or cRb −/− /RasV12 astrocytes in FVB mice (10 animals per group). As shown in Fig. 4, only RasV12 and cRb −/− /RasV12 astrocytes formed tumors. Tumors formed by injection with RasV12 cells grew during the first week, and after that, all the tumors formed were reabsorbed. cRb −/− /RasV12 tumors grew during the first two weeks and then began to involute until the fourth week. At about this time, tumors were completely reabsorbed.

Effect of Rb deleted and/or RASV12 overexpressed tumor cells on immune cell phenotype in the peripheral blood

In order to analyze the immune response against transformed glioma cell lines with Rb deletion and/or RASV12 overexpression in an homologous syngeneic model of tumor transplantation, the percentages of different immune cell subpopulations were quantified in the peripheral blood of mice in which tumor cells had been injected 28 days earlier. Figure 5 shows that mice injected with the cRb −/− /RasV12 astrocytes increase the percentage of cytotoxic CD8+ lymphocytes, as compared to the other groups of mice (p = 0.026); a significant reduction in the percentage of regulatory CD4 + CD25+ lymphocytes was observed in mice injected with RasV12 or cRb −/− (p = 0.001). In addition, the group implanted with RasV12 cells developed a significant increment in the percentage of activated T cells, both CD4 + CD69+ and CD8 + CD69+ cells (p = 0.016 and p = 0.001 respectively). In all groups of transformed glioma cell line-injected mice, the percentage of granzyme-expressing cells increased as compared to the mice injected with cRb loxP/loxP astrocytes.

Discussion

The immune system is thought to be constantly surveying for the arising of malignant cells that would in turn be eliminated by the immune response [13]. An “emerging hallmark” of cancer states that tumor cells are capable of avoiding immune destruction [3].
In gliomas from human origin, the formed tumor is immunosuppressive [14, 15]. How these tumors reach that immunosuppressive characteristic at the early stages of malignant transformation, and whether specific mutations are associated with the ability to escape from the immune response is poorly understood. Here, by using a previously reported in vitro model of gliomagenesis [12], we tested the hypothesis that defined changes in the expression of a proto-oncogene (Ras) and a tumor suppressor (Rb) confer tumor cells the ability to avoid immune destruction. In particular, we addressed the resistance to NK cell-mediated cytotoxicity. Since NK-cell mediated cytotoxicity is dependent on the tumor cell membrane expression of several ligands that upon engagement with specific NK cell receptors either inhibit or activate NK cell cytotoxic function, the expression of MHC class I (ligand for inhibiting NK receptor Ly49D), Rae1δ, Rae1αβδγε, mult1, H60a, H60b, and H60c (ligands for NKG2D, an activating NK cell receptor) [1619], as well as of Fas and FasL (molecules involved in programed cell death) was assessed in vitro transformed astrocytes. In gliomas from human origin, deregulated expression of MHC-I has been associated with NK cell-mediated cytotoxicity [20], and stem cells from glioma patients do not express protective levels of MHC-I molecules, but they express several ligands that activate NK cells [21].
These studies assessed the susceptibility of both stem cells and tumor cells to NK cell-mediated cytotoxicity, after the onset of malignant transformation and therefore after immunoediting had likely already taken place [5, 22]. Although, down-regulation of MHC-I makes tumor cells susceptible to NK cell-mediated cytotoxicity [22], high expression of MHC-I does not necessarily guarantee resistance to NK cells [23]. Here, we assessed the expression of MHC-I on various in vitro transformed astrocytes that therefore were not subjected to any immune-based selective pressure. Results showed a significant increase in the expression of MHC-I in the cRb−/− cells and a significant decrease in the cRb−/−/RasV12 cells (Fig. 1a). When Rb−/−/RasV12 cells were inoculated into SCID mice, and the T731 tumor cell line was derived, the expression of MHC-I was similar to that of cRbloxP/loxP cells (Fig. 1a). This MHC-I expression recovery may be the result of selective pressure exerted by NK cells in the SCID mice that selected cells with the highest levels of MHC-I, conferring them an advantage to evade the NK cell anti-tumor response. In this regard, the T731 cell line also showed the lowest percentage of NK cell-mediated cytotoxicity (Fig. 2). However, the lower expression of MHC-1 observed in Rb −/− /RasV12 cells, and the higher expression of Rae1α and Rae 1αβγδε observed in cRb −/− cells seem to be in contradiction with their increased resistance to NK cells. Rae1 expression is low or absent in normal tissues and it is constitutively expressed on some tumor cells. Upon engagement with the NKG2D receptor on NK cells, it activates their cytotoxic activity [24, 25]. Rae1 expression has been associated with cell proliferation [26], and Rae1 gene family members are activated by the E2F transcription factor, which plays a central role in regulating cell cycle entry [27]. A significant increase in the expression of Rae1δ and Rae1αβγδε was found in Rb −/− astrocytes, as compared with that of parental cRb loxP/loxP astrocytes (Fig. 2b and c). Ras induces the expression of Rae1 [28]. However, in this case, overexpression of Ras (RasV12 astrocytes) did not correlate with an increase in Rae1δ or Rae1 αβγδε expression. Moreover, in cRb −/− /RasV12 astrocytes a significant reduction in the expression of both Rae1δ, and Rae1αβγδε was observed (Fig. 1). The T653 and T731 astrocyte lines did not show significant changes in Rae1δ, or Rae1αβγδε expression, as compared with parental cRb loxP/loxP astrocytes. This would suggest that loss of Rb promotes the expression of Rae1δ, and Rae1αβγδε. However, since these two ligands would engage with the NK cell activating NKG2D receptor, increased NK cell-mediated cytotoxicity against cRb−/− astrocytes, as compared to that of cRb loxP/loxP astrocytes would be expected. This was clearly not the case, since the percentage of NK cell-mediated cytotoxicity was in fact lower than that for cRb loxP/loxP astrocytes (Fig. 3). The mRNA expression of four other ligands for NKG2D cell receptors was also assessed (mult1, H60a, H60b, and H60c). These ligands are poorly expressed on most normal cells, but are upregulated on tumor cells, which is keeping with the finding that mult1 mRNA expression was comparatively higher in RasV12 and cRb−/−/ RasV12 cells than in cRbloxP/loxP cells. On the other hand, the mRNA expression of mult1 was similar between cRbloxP/loxP and cRb−/− cells, thus suggesting that transformation due to RasV12 over expression but not to Rb deletion would render transformed astrocytes more susceptible to NK cytotoxicity. Again, this was not the case, since both RasV12 and cRb−/−/ RasV12 astrocytes were more resistant to NK cytotoxicity. However, in analyzing the mRNA expression of H60a and H60b, results evident that their expression is lower in RasV12 and cRb−/−, as compared to cRbloxP/loxP astrocytes. In addition, mRNA expression of H60a and H60b is similar in cRb−/−/ RasV12 and cRbloxP/loxP astrocytes. Therefore H60a and H60b expression would imply resistance to NK cell cytotoxicity, as it actually happens.
The molecule Fas induces cell death upon engagement with FasL (Fas ligand) [29]. FasL is expressed on activated T and NK cells [30] and thus FasL induces apoptotic cell death on Fas-expressing cells [28]. Accordingly, a decrease in Fas expression would protect cells from cell-mediated cytotoxicity. The loss of Fas expression has been observed in melanoma, breast cancer, leukemia, and lymphoma cells [3133]. A variety of malignant tumors show increased expression of FasL, thus allowing tumor cells to induce apoptosis on cytotoxic cells, in a process known as “tumor counterattack” [3436]. Here we showed that overexpression of Ras, deletion of Rb, or both, are sufficient to decrease the expression of Fas (Fig. 1d). Lower expression of Fas was concomitant to increased resistance to NK cell-mediated cytotoxicity (Fig. 2). No significant increase in the expression of FasL was observed in any of the transformed astrocytes. The FasL expression data suggest that genetic alterations other than just Ras overexpression and Rb deletion are required for the “tumor counterattack” phenotype acquisition. It is tempting to speculate that this phenotype only takes place after acquisition of resistance to NK cells and further genome instability, a property of neoplastic transformation [3].
Additional experiments were designed to evaluate the tumor growth in a syngeneic model (FVB mice) and the possible contribution of other immune cells in the tumor implantation outcome. cRb−/−/RasV12, followed by cRb−/− astrocytes were successfully implanted. This could be explained in part by the mRNA expression of H60a and H60b, as mentioned before, and the consequent resistance to NK cell-mediated cytotoxicity (Fig. 3). However, the expression of other ligands for NKG2D is contradictory, mult1 for instance. If tumor implantation can be attributed to resistance to NK cell cytotoxicity, tumor remission could then be attributed to the increase in the percentages of CD8+ and granzyme+ cells in the case of cRb−/−/RasV12 tumor, and to the increase in the percentages of CD8+CD69+, CD4+CD69+ and granzyme+ cells, and to the decrease in the percentage of regulatory CD4+CD25+ cells in the case of RasV12 tumor. It is worth noting that the most resistant tumor is the one with the two mutations and also the one that after 28 days post implantation only induced an increase in the percentage of CD8+ and granzyme+ cells as compared with the RasV12 that induces phenotype changes more consistent with anti-tumor immunity.
Taken together, our results suggest that Ras overexpression and Rb deletion are sufficient for the malignant transformation of astrocytes and that these genetic alterations confer transformed cells resistance to NK cell-mediated cytotoxicity, by altering the expression of NK cell receptor ligands, such as the higher expression of MHC-I observed in cRb −/− cells, or the lower expression of Rae1α and Rae1αβγδε observed in Rb −/− /RasV12 cells, and also by altering the expression of cell death associated molecules such as Fas, as observed in RasV12 cells. Its seems that the innate immune system deals with small antigenic differences between the normal and transformed tumor cells that allow tumor cells to initiate proliferation and to develop a number of oncogenic stages, however further analyses will be required.

Methods

Cell culture and cell proliferation assays

Astrocytes were isolated from 3 days old Rb floxed mice, as previously described [12]. Animal care and use of all experimental animals were performed in accordance with institutional ethical guidelines. In order to introduce an active Ras allele or to promote the Rb loss in astrocytes, Phoenix-Eco packaging cells (a kind gift from G.P. Nolan) were transfected with empty pBABE, pBABE-HRasV12, empty PIG, and PIG-CRE retroviral plasmids (a kind gift from P.P. Pandolfi), and in vitro transformation was achieved by retroviral infection. The resulting cells were denominated; cRb loxP/loxP , RasV12, cRb −/− , and cRb −/− /RasV12. The T653 and T731 cell lines were derived from tumors formed by inoculation of Rasv12 and cRb −/− /RasV12 cells in SCID mice, as previously shown [12]. A feature of the SCID mice is a deficiency in the recombination of genes needed for full maturation of T and lymphocytes. However, SCID mice harbor functional NK cells. Cells were maintained in Dulbecco Modified Eagle Medium (DMEM) (Sigma-Aldrich, St Louis, MO) supplemented with 10 % FBS and antibiotic-antimycotic solution (Bio West, Nuaillé, France). For cell growth analysis, 5x103 cells were plated into 24-well culture plates (Corning, NY, USA) and then fixed with methanol/acetic acid (3:1) on days 1, 3, 5, 7 of culture, for subsequent staining with crystal violet (0.1 % in PBS) and distaining with 10 % acetic acid. The relative cell number was assessed by spectrophotometry.

Syngeneic model

The performance of tumor growth in the syngeneic model was evaluated by subcutaneously injecting 1x106 cRbloxP/loxP, RasV12, cRb−/−, or cRb−/−/RasV12 astrocytes previously obtained from FVB mice (n = 10). Tumours from all animals (10 per group) were measured weekly, and their volumes (in cubic millimeters) were determined with the formula 6/π × L × W × H. After 28 days, animals from all groups were anaesthetized and sacrificed by exsanguination.

Flow Cytometry of T lymphocytes, macrophages and granzyme

Immunofluorescence using monoclonal antibodies was used to determine the percentages of CD68+, CD4+, CD8+, CD4+/CD25+, CD8/CD25+, CD8+/CD69+, CD4+/CD69+ and granzyme+ cells in the peripheral blood samples, (Biolegend, USA). Briefly, 30 μl of blood were incubated for 30 min with 5 μl of the corresponding monoclonal antibody (1:100 dilution) afterwards, 200 μl of lysis solution were added (Becton Dickinson, California), incubated in darkness for 10 min and washed twice with 0.1 M PBS (pH 7.2), 0.1 % BSA and 0.1 % NaN3. The cells were then fixed in 1 % paraformaldehyde solution and stored at 4 °C until examination by flow cytometry (FACSCalibur, Becton Dickinson) using the Cell Quest software. 10,000 events in the region corresponding to lymphocytes were analyzed. From this region, the percentage of positive cells from each sample was determined. Results were expressed as means (±SD) for each experimental group.

Senescence assay

Cell senescence was assessed by the expression of β-galactosidase, by using a β-galactosidase staining kit (Cell Signaling, Danvers, MA). Cells (5x103) were plated in triplicate into 24-well culture plates (Corning, NY, USA) and then fixed on day 6 of culture for subsequent β-galactosidase staining.

Immunoblot

Cell proteins were extracted in RIPA buffer (1 % Nonidet P-40, 0.5 % sodium deoxycholate, 0.1 % SDS in PBS) in the presence of 40 μg/ml of aprotinin, 10 μg/ml PMSF and 100 mM orthovanadate. 40 μg of total protein were separated by 8 % or 12 % SDS-PAGE and transferred to nitrocellulose membranes. Western blot were developed with antibodies against p-p53 (Cell signaling, Danvers, MA), p-H2AX (Millipore, MA, USA), p53 (cell signaling, Danvers, MA), pan-Ras-V12 (Calbiochem, MA, USA), p-Rb (BD Biosciences, San Jose, CA, USA), or α-tubulin (Sigma-Aldrich, St Louis, MO).

Phenotypic analysis (by Immunofluorescence)

Cells were growth in 8-well polystyrene chambers (BD Falcon, San Jose, CA, USA) until 80 % confluence, fixed with 4 % paraformaldehyde in PBS, blocked with 1 % BSA in PBS, and labeled overnight at 4 °C with anti-GFAP antibody (Millipore, MA, USA) followed by Alexa fluor-594-conjugated anti-rabbit IgG (Invitrogen). Cells were mounted in Vectashield (Vector, CA, USA) and analyzed by confocal microscopy (LSMS Pascal, Zeiss).

Phenotypic analysis (by Flow cytometry)

Cells were suspended in blocking buffer (0.5 % BSA/ 2 mM EDTA, in PBS) and then labeled with fluorochrome-conjugated antibodies: PE-conjugated anti-Fas (BD Biosciences, San Jose, CA, USA), PE-conjugated anti-FasL (Biolegend, San Diego, CA) (1 μg/ml), or anti-MHC-I (Biolegend, San Diego, CA), anti-RAE-1δ (Biolegend, San Diego, CA), anti-Rae-1αβγδε (scbt, CA, USA) (1:50), followed by APC-conjugated anti-mouse IgG (scbt, CA, USA) or APC-conjugated anti-rabbit IgG (scbt, CA, USA) (1:50) secondary antibodies, as appropriate. Cell membrane expression of these molecules was assessed by Flow Cytometry (FACS Aria III, BD Biosciences). Raw data was further analyzed by using Flow Jo software (Tree Star, Inc. Ashland OR).

NK cytotoxicity assays

NK cells were purified from mice spleens (C57 strain), hosted in the INNN animal house, in accordance with institutional guidelines, by using the NK cell isolation kit II (Myltenyi Biotech, Germany), following the manufacturer”s instructions. NK cell cytotoxicity against tumor cells was evaluated by using Lecoeur et al. method [37]. Since the tumor cells here used express green fluorescent protein (GFP) due to the transformation procedure, there was no need to label them. Isolated NK cells and tumor cells were co-cultured in 10 % FBS/DMEM at 1:10 target/effector cell ratio, for 4 h. After which, cells were labeled for 15 min with 7-aminoactinomycin D (BD Pharmingen, San Jose, CA, USA) at a final concentration of 20 μl/ml. Flow cytometry analysis was used to calculate the percentage of green fluorescent cells (tumor cells) that were stained by 7-AAD (dead cells). Results are expressed as the percentage of specific lysis calculated by the following formula:
$$ \%\ \mathrm{specific}\ \mathrm{lysis} = 100 \times \left(\%\kern0.5em \mathrm{sample}\ \mathrm{lysis} - \%\ \mathrm{basal}\ \mathrm{lysis}\right)/100 - \%\ \mathrm{basal}\ \mathrm{lysis} $$

Quantitative polymerase chain reaction

Total RNA was extracted via the phenol/chloroform method using TRIzol reagent (Invitrogen). Quantitative polymerase chain reaction (PCR) was performed using EXPRESS One-Step SYBR® GreenER™ Kit (Invitrogen, USA). Emissions from the SYBR Green reporter dye were monitored with an ABI Prism 7500 Real Time PCR (Applied Biosystems). The primer sequences used were as follows:
Mult1, 5′-CAATGTCTCTGTCCTCGGAA-3′ (sense), Mult1, 5′-CTGAACACGTCTCAGGCACT-3′ (antisense); H60a, 5′-TGCCTGATTCTGAGCCTTTTCA-3′ (sense), H60a, 5′-ATTCACTGAGCACTGTCCATGTAGAT-3′ (antisense); H60b, 5′-AGCCTTTTGGTCCTGCTGAAT-3′ (sense), H60b, 5′-ATGTTTTTTATCACCAAAATCAAGGAGT-3′ (antisense); H60c, 5′-CTTCTCTTGATCCTGGAGTCCTGTAGT-3′ (sense), H60c, 5′-GAGAGTCTTTCCATTCACTGAGCAC-3′ (antisense); β-actin, 5′-TTCTACAATGAGCTGCGT-3′ (sense), β-actin, 5′-ATCACAATGCCTGTGGTA-3′ (antisense). All expression levels of interested genes were normalized to the housekeeping gene β-actin. Gene expression values were then calculated based on the ΔΔCt method.

Statistical analyses

Data was summarized as arithmetic means and standard deviations (SD). One-way analysis of variance (ANOVA) and post-hoc (Tukey) test were conducted. Statistical significance was set at p < 0.05 in a two-sided test. SPSS software package V 18.0 for Windows; (SPSS Inc., Chicago, IL) was employed for data analysis.

Acknowledgements

We thank the members of the Molecular Oncology Laboratory for helpful discussions. This work was supported by grants PXIB208091PR (to J.A. Costoya), from Xunta de Galicia. SAF2008-00543 and SAF2009-08629 (to J.A. Costoya), from Ministerio de Ciencia e Innovación. CB158340 (to F.J. Sánchez-García), from Consejo Nacional de Ciencia y Tecnología, Mexico (CONACYT) and CB180851 (to B. Pineda), from CONACYT, and FOSSIS 182362 (to B. Pineda and S. Moreno).
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Authors’ contributions

Conception and design: MOM; JACP; FJSG; BP. Collection and assembly of data: MOM, IGC; NHP, VPdlC, SMJ Data analysis and interpretation: MOM; JACP; FJSG; JS; BP. Manuscript writing: MOM; FJSG and BP. All authors read and approved the final manuscript.
Literatur
2.
Zurück zum Zitat Ancrile BB, O'Hayer KM, Counter CM. Oncogenic ras-induced expression of cytokines: a new target of anti-cancer therapeutics. Mol Interv. 2008;8(1):22–7.PubMedCentralPubMedCrossRef Ancrile BB, O'Hayer KM, Counter CM. Oncogenic ras-induced expression of cytokines: a new target of anti-cancer therapeutics. Mol Interv. 2008;8(1):22–7.PubMedCentralPubMedCrossRef
3.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef
4.
Zurück zum Zitat Trinchieri G. Cancer and inflammation: an old intuition with rapidly evolving new concepts. Annu Rev Immunol. 2012;30:677–706.PubMedCrossRef Trinchieri G. Cancer and inflammation: an old intuition with rapidly evolving new concepts. Annu Rev Immunol. 2012;30:677–706.PubMedCrossRef
5.
Zurück zum Zitat Khong HT, Restifo NP. Natural selection of tumor variants in the generation of “tumor escape” phenotypes. Nat Immunol. 2002;3(11):999–1005.PubMedCentralPubMedCrossRef Khong HT, Restifo NP. Natural selection of tumor variants in the generation of “tumor escape” phenotypes. Nat Immunol. 2002;3(11):999–1005.PubMedCentralPubMedCrossRef
7.
Zurück zum Zitat Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114(2):97–109.PubMedCentralPubMedCrossRef Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114(2):97–109.PubMedCentralPubMedCrossRef
9.
Zurück zum Zitat Cancer Genome Atlas Research N. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–8.CrossRef Cancer Genome Atlas Research N. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–8.CrossRef
10.
Zurück zum Zitat Yin S, Van Meir EG. p53 Pathway Alterations in Brain Tumors. In: Van Meir EG, editor. CNS Cancer: Models, Markers, Prognostic Factors, Targets and Therapeutic Approaches. New York: Humana Press (Springer); 2009. Yin S, Van Meir EG. p53 Pathway Alterations in Brain Tumors. In: Van Meir EG, editor. CNS Cancer: Models, Markers, Prognostic Factors, Targets and Therapeutic Approaches. New York: Humana Press (Springer); 2009.
11.
Zurück zum Zitat Alizadeh D, Zhang L, Brown CE, Farrukh O, Jensen MC, Badie B. Induction of anti-glioma natural killer cell response following multiple low-dose intracerebral CpG therapy. Clin Cancer Res. 2010;16(13):3399–408.PubMedCentralPubMedCrossRef Alizadeh D, Zhang L, Brown CE, Farrukh O, Jensen MC, Badie B. Induction of anti-glioma natural killer cell response following multiple low-dose intracerebral CpG therapy. Clin Cancer Res. 2010;16(13):3399–408.PubMedCentralPubMedCrossRef
12.
Zurück zum Zitat Seoane M, Iglesias P, Gonzalez T, Dominguez F, Fraga M, Aliste C, et al. Retinoblastoma loss modulates DNA damage response favoring tumor progression. PLoS One. 2008;3(11), e3632.PubMedCentralPubMedCrossRef Seoane M, Iglesias P, Gonzalez T, Dominguez F, Fraga M, Aliste C, et al. Retinoblastoma loss modulates DNA damage response favoring tumor progression. PLoS One. 2008;3(11), e3632.PubMedCentralPubMedCrossRef
13.
Zurück zum Zitat Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21(2):137–48.PubMedCrossRef Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21(2):137–48.PubMedCrossRef
14.
Zurück zum Zitat Parney IF, Farr-Jones MA, Chang LJ, Petruk KC. Human glioma immunobiology in vitro: implications for immunogene therapy. Neurosurgery. 2000;46(5):1169–77. discussion 1177–1168.PubMedCrossRef Parney IF, Farr-Jones MA, Chang LJ, Petruk KC. Human glioma immunobiology in vitro: implications for immunogene therapy. Neurosurgery. 2000;46(5):1169–77. discussion 1177–1168.PubMedCrossRef
15.
Zurück zum Zitat Fenstermaker RA, Ciesielski MJ. Immunotherapeutic strategies for malignant glioma. Cancer Control. 2004;11(3):181–91.PubMed Fenstermaker RA, Ciesielski MJ. Immunotherapeutic strategies for malignant glioma. Cancer Control. 2004;11(3):181–91.PubMed
16.
Zurück zum Zitat Diefenbach A, Hsia JK, Hsiung MY, Raulet DH. A novel ligand for the NKG2D receptor activates NK cells and macrophages and induces tumor immunity. Eur J Immunol. 2003;33(2):381–91.PubMedCrossRef Diefenbach A, Hsia JK, Hsiung MY, Raulet DH. A novel ligand for the NKG2D receptor activates NK cells and macrophages and induces tumor immunity. Eur J Immunol. 2003;33(2):381–91.PubMedCrossRef
17.
Zurück zum Zitat Takada A, Yoshida S, Kajikawa M, Miyatake Y, Tomaru U, Sakai M, et al. Two novel NKG2D ligands of the mouse H60 family with differential expression patterns and binding affinities to NKG2D. J Immunol. 2008;180(3):1678–85.PubMedCrossRef Takada A, Yoshida S, Kajikawa M, Miyatake Y, Tomaru U, Sakai M, et al. Two novel NKG2D ligands of the mouse H60 family with differential expression patterns and binding affinities to NKG2D. J Immunol. 2008;180(3):1678–85.PubMedCrossRef
19.
Zurück zum Zitat Karlhofer FM, Ribaudo RK, Yokoyama WM. MHC class I alloantigen specificity of Ly-49+ IL-2-activated natural killer cells. Nature. 1992;358(6381):66–70.PubMedCrossRef Karlhofer FM, Ribaudo RK, Yokoyama WM. MHC class I alloantigen specificity of Ly-49+ IL-2-activated natural killer cells. Nature. 1992;358(6381):66–70.PubMedCrossRef
20.
Zurück zum Zitat Ogbomo H, Cinatl Jr J, Mody CH, Forsyth PA. Immunotherapy in gliomas: limitations and potential of natural killer (NK) cell therapy. Trends Mol Med. 2011;17(8):433–41.PubMedCrossRef Ogbomo H, Cinatl Jr J, Mody CH, Forsyth PA. Immunotherapy in gliomas: limitations and potential of natural killer (NK) cell therapy. Trends Mol Med. 2011;17(8):433–41.PubMedCrossRef
21.
Zurück zum Zitat Castriconi R, Daga A, Dondero A, Zona G, Poliani PL, Melotti A, et al. NK cells recognize and kill human glioblastoma cells with stem cell-like properties. J Immunol. 2009;182(6):3530–9.PubMedCrossRef Castriconi R, Daga A, Dondero A, Zona G, Poliani PL, Melotti A, et al. NK cells recognize and kill human glioblastoma cells with stem cell-like properties. J Immunol. 2009;182(6):3530–9.PubMedCrossRef
22.
Zurück zum Zitat Karre K, Ljunggren HG, Piontek G, Kiessling R. Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature. 1986;319(6055):675–8.PubMedCrossRef Karre K, Ljunggren HG, Piontek G, Kiessling R. Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature. 1986;319(6055):675–8.PubMedCrossRef
23.
Zurück zum Zitat Holscher M, Givan AL, Brooks CG. The effect of transfected MHC class I genes on sensitivity to natural killer cells. Immunology. 1991;73(1):44–51.PubMedCentralPubMed Holscher M, Givan AL, Brooks CG. The effect of transfected MHC class I genes on sensitivity to natural killer cells. Immunology. 1991;73(1):44–51.PubMedCentralPubMed
24.
Zurück zum Zitat Pellegatta S, Cuppini L, Finocchiaro G. Brain cancer immunoediting: novel examples provided by immunotherapy of malignant gliomas. Expert Rev Anticancer Ther. 2011;11(11):1759–74.PubMedCrossRef Pellegatta S, Cuppini L, Finocchiaro G. Brain cancer immunoediting: novel examples provided by immunotherapy of malignant gliomas. Expert Rev Anticancer Ther. 2011;11(11):1759–74.PubMedCrossRef
25.
Zurück zum Zitat Andre P, Castriconi R, Espeli M, Anfossi N, Juarez T, Hue S, et al. Comparative analysis of human NK cell activation induced by NKG2D and natural cytotoxicity receptors. Eur J Immunol. 2004;34(4):961–71.PubMedCrossRef Andre P, Castriconi R, Espeli M, Anfossi N, Juarez T, Hue S, et al. Comparative analysis of human NK cell activation induced by NKG2D and natural cytotoxicity receptors. Eur J Immunol. 2004;34(4):961–71.PubMedCrossRef
26.
Zurück zum Zitat Cerwenka A, Bakker AB, McClanahan T, Wagner J, Wu J, Phillips JH, et al. Retinoic acid early inducible genes define a ligand family for the activating NKG2D receptor in mice. Immunity. 2000;12(6):721–7.PubMedCrossRef Cerwenka A, Bakker AB, McClanahan T, Wagner J, Wu J, Phillips JH, et al. Retinoic acid early inducible genes define a ligand family for the activating NKG2D receptor in mice. Immunity. 2000;12(6):721–7.PubMedCrossRef
27.
Zurück zum Zitat Popa N, Cedile O, Pollet-Villard X, Bagnis C, Durbec P, Boucraut J. RAE-1 is expressed in the adult subventricular zone and controls cell proliferation of neurospheres. Glia. 2011;59(1):35–44.PubMedCrossRef Popa N, Cedile O, Pollet-Villard X, Bagnis C, Durbec P, Boucraut J. RAE-1 is expressed in the adult subventricular zone and controls cell proliferation of neurospheres. Glia. 2011;59(1):35–44.PubMedCrossRef
28.
Zurück zum Zitat Jung H, Hsiung B, Pestal K, Procyk E, Raulet DH. RAE-1 ligands for the NKG2D receptor are regulated by E2F transcription factors, which control cell cycle entry. J Exp Med. 2012;209(13):2409–22.PubMedCentralPubMedCrossRef Jung H, Hsiung B, Pestal K, Procyk E, Raulet DH. RAE-1 ligands for the NKG2D receptor are regulated by E2F transcription factors, which control cell cycle entry. J Exp Med. 2012;209(13):2409–22.PubMedCentralPubMedCrossRef
29.
Zurück zum Zitat Liu XV, Ho SS, Tan JJ, Kamran N, Gasser S. Ras activation induces expression of Raet1 family NK receptor ligands. J Immunol. 2012;189(4):1826–34.PubMedCrossRef Liu XV, Ho SS, Tan JJ, Kamran N, Gasser S. Ras activation induces expression of Raet1 family NK receptor ligands. J Immunol. 2012;189(4):1826–34.PubMedCrossRef
30.
Zurück zum Zitat Walczak H, Krammer PH. The CD95 (APO-1/Fas) and the TRAIL (APO-2 L) apoptosis systems. Exp Cell Res. 2000;256(1):58–66.PubMedCrossRef Walczak H, Krammer PH. The CD95 (APO-1/Fas) and the TRAIL (APO-2 L) apoptosis systems. Exp Cell Res. 2000;256(1):58–66.PubMedCrossRef
31.
Zurück zum Zitat Bullani RR, Wehrli P, Viard-Leveugle I, Rimoldi D, Cerottini JC, Saurat JH, et al. Frequent downregulation of Fas (CD95) expression and function in melanoma. Melanoma Res. 2002;12(3):263–70.PubMedCrossRef Bullani RR, Wehrli P, Viard-Leveugle I, Rimoldi D, Cerottini JC, Saurat JH, et al. Frequent downregulation of Fas (CD95) expression and function in melanoma. Melanoma Res. 2002;12(3):263–70.PubMedCrossRef
32.
Zurück zum Zitat Hahne M, Rimoldi D, Schroter M, Romero P, Schreier M, French LE, et al. Melanoma cell expression of Fas(Apo-1/CD95) ligand: implications for tumor immune escape. Science. 1996;274(5291):1363–6.PubMedCrossRef Hahne M, Rimoldi D, Schroter M, Romero P, Schreier M, French LE, et al. Melanoma cell expression of Fas(Apo-1/CD95) ligand: implications for tumor immune escape. Science. 1996;274(5291):1363–6.PubMedCrossRef
33.
Zurück zum Zitat Koyama S, Koike N, Adachi S. Fas receptor counterattack against tumor-infiltrating lymphocytes in vivo as a mechanism of immune escape in gastric carcinoma. J Cancer Res Clin Oncol. 2001;127(1):20–6.PubMedCrossRef Koyama S, Koike N, Adachi S. Fas receptor counterattack against tumor-infiltrating lymphocytes in vivo as a mechanism of immune escape in gastric carcinoma. J Cancer Res Clin Oncol. 2001;127(1):20–6.PubMedCrossRef
34.
Zurück zum Zitat Reimer T, Herrnring C, Koczan D, Richter D, Gerber B, Kabelitz D, et al. FasL:Fas ratio–a prognostic factor in breast carcinomas. Cancer Res. 2000;60(4):822–8.PubMed Reimer T, Herrnring C, Koczan D, Richter D, Gerber B, Kabelitz D, et al. FasL:Fas ratio–a prognostic factor in breast carcinomas. Cancer Res. 2000;60(4):822–8.PubMed
35.
Zurück zum Zitat Niehans GA, Brunner T, Frizelle SP, Liston JC, Salerno CT, Knapp DJ, et al. Human lung carcinomas express Fas ligand. Cancer Res. 1997;57(6):1007–12.PubMed Niehans GA, Brunner T, Frizelle SP, Liston JC, Salerno CT, Knapp DJ, et al. Human lung carcinomas express Fas ligand. Cancer Res. 1997;57(6):1007–12.PubMed
36.
Zurück zum Zitat O'Connell J, O'Sullivan GC, Collins JK, Shanahan F. The Fas counterattack: Fas-mediated T cell killing by colon cancer cells expressing Fas ligand. J Exp Med. 1996;184(3):1075–82.PubMedCrossRef O'Connell J, O'Sullivan GC, Collins JK, Shanahan F. The Fas counterattack: Fas-mediated T cell killing by colon cancer cells expressing Fas ligand. J Exp Med. 1996;184(3):1075–82.PubMedCrossRef
37.
Zurück zum Zitat Lecoeur H, Fevrier M, Garcia S, Riviere Y, Gougeon ML. A novel flow cytometric assay for quantitation and multiparametric characterization of cell-mediated cytotoxicity. J Immunol Methods. 2001;253(1–2):177–87.PubMedCrossRef Lecoeur H, Fevrier M, Garcia S, Riviere Y, Gougeon ML. A novel flow cytometric assay for quantitation and multiparametric characterization of cell-mediated cytotoxicity. J Immunol Methods. 2001;253(1–2):177–87.PubMedCrossRef
Metadaten
Titel
RB mutation and RAS overexpression induce resistance to NK cell-mediated cytotoxicity in glioma cells
verfasst von
Mario Orozco-Morales
Francisco Javier Sánchez-García
Irene Golán-Cancela
Norma Hernández-Pedro
Jose A. Costoya
Verónica Pérez de la Cruz
Sergio Moreno-Jiménez
Julio Sotelo
Benjamín Pineda
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2015
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-015-0209-x

Weitere Artikel der Ausgabe 1/2015

Cancer Cell International 1/2015 Zur Ausgabe

Update Onkologie

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