Resistance to TRAIL in mantle cell lymphoma cells is associated with the decreased expression of purine metabolism enzymes

  • Authors:
    • Jana Pospisilova
    • Ondrej Vit
    • Lucie Lorkova
    • Magdalena Klanova
    • Jan Zivny
    • Pavel Klener
    • Jiri Petrak
  • View Affiliations

  • Published online on: March 13, 2013     https://doi.org/10.3892/ijmm.2013.1302
  • Pages: 1273-1279
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Mantle cell lymphoma (MCL) is a rare aggressive type of B-cell non-Hodgkin's lymphoma. Response to chemotherapy tends to be short and virtually all patients sooner or later relapse. The prognosis of relapsed patients is extremely poor. The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is considered one of the novel experimental molecules with strong antitumor effects. TRAIL triggers extrinsic apoptotis in tumor cells by binding to TRAIL ‘death receptors’ on the cell surface. Recombinant TRAIL has shown promising pro-apoptotic effects in a variety of malignancies including lymphoma. However, as with other drugs, lymphoma cells can develop resistance to TRAIL. Therefore, the aim of this study was to identify the molecular mechanisms responsible for, and associated with TRAIL resistance in MCL cells. If identified, these features may be used as molecular targets for the effective elimination of TRAIL-resistant lymphoma cells. From an established TRAIL-sensitive mantle cell lymphoma cell line (HBL-2) we derived a TRAIL-resistant HBL-2/R subclone. By TRAIL receptor analysis and differential proteomic analysis of HBL-2 and HBL-2/R cells we revealed a marked downregulation of all TRAIL receptors and, among others, the decreased expression of 3 key enzymes of purine nucleotide metabolism, namely purine nucleoside phosphorylase, adenine phosphoribosyltransferase and inosine-5'-monophosphate dehydrogenase 2, in the resistant HBL-2/R cells. The downregulation of the 3 key enzymes of purine metabolism can have profound effects on nucleotide homeostasis in TRAIL-resistant lymphoma cells and can render such cells vulnerable to any further disruption of purine nucleotide metabolism. This pathway represents a ‘weakness’ of the TRAIL-resistant MCL cells and has potential as a therapeutic target for the selective elimination of such cells.

Introduction

Mantle cell lymphoma (MCL) is a rare agressive type of B-cell non-Hodgkin’s lymphoma with an estimated annual incidence in Europe of 0.45/100,000 individuals (1). MCL is a biologically and clinically heterogeneous disease; the immunophenotype of neoplastic cells reflects the phenotype of cells similar to lymphocytes in the mantle zone of normal germinal follicles (2). The genetic hallmark of MCL cells is a translocation between chromosomes 11 and 14, t(11;14)(q13;q32), juxtaposing the gene for immunoglobulin heavy chain and the gene encoding cyclin D1. This results in cyclin D1 overexpression (3,4).

The standard of care for newly diagnosed MCL patients is combined immunochemotherapy alternating rituximab-CHOP (R-CHOP; cyclophosphamide, vincristine, doxorubicin and prednisone) and R-HDAC (high-dose cytarabine). The addition of rituximab and HDAC to CHOP has improved the survival of MCL patients in the last 2 decades from 3 to 5 years. However, the response to therapy tends to be short and virtually all patients sooner or later relapse. There is no standard of care for relapsed or refractory MCL patients. Salvage therapy usually comprises diverse regimens based on fludarabine, gemcitabine, cisplatin, bendamustine, bortezomib (inhibitor of 26S proteasome) or temsirolimus (inhibitor of mTOR). Recently, several new experimental molecules have shown promise in the therapy of relapsed or resistant MCL, including lenalidomide (immunomodulatory agent), ibrutinib (PCI-32765, inhibitor of Bruton’s tyrosine-kinase), new monoclonal antibodies (e.g., anti-CD20 ofatumumab), as well as other agents (5). Combination therapies are currently being evaluated in clinical trials; however, novel drugs are required.

The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is considered one of the novel experimental molecules with strong antitumor effects. TRAIL is a type II transmembrane protein from the tumor necrosis factor superfamily (6,7) expressed mostly by cells of the immune system (natural killer cells, cytotoxic T-cells, macrophages and dendritic cells). The main function of this molecule is thought to be in tumor immunosurveillance, but its actual molecular role remains to be elucidated.

TRAIL can trigger extrinsic apoptotis in target cells by binding to TRAIL death receptors located on the cell surface (8). This interaction is performed by a long extracellular C-terminal region of the TRAIL molecule. There are 4 distinct cell surface TRAIL receptors in humans (DcR1, DcR2, DR4 and DR5) encoded by separate genes (9,10). However, only DR4 and DR5 contain a functional death domain (structurally conserved protein interaction domain) and are capable of signaling apoptosis. Two decoy receptors (DcR1 and DcR2) lack a functional death domain and inhibit TRAIL signaling by competing with death receptors for TRAIL (9,10). The binding of TRAIL to DR4 or DR5 leads to receptor homotrimerization and formation of the death-inducing signaling complex (DISC) (11). Through the DISC a caspase machinery is activated, which results in apoptosis (12). TRAIL death receptors DR4 and DR5 are ubiquitously expressed, indicating that most tissues and cell types are potential targets of TRAIL signaling (13). Nevertheless, TRAIL seems to induce apoptosis only in tumor cells but not in healthy tissues. Due to its selective pro-apoptotic effect, TRAIL has attracted much attention for its possible use in cancer therapy. In vitro, a recombinant soluble TRAIL molecule has shown cytostatic or cytotoxic effects in a wide variety of tumor cell lines, including leukemia and lymphoma cells, but not in normal cells (6,7,10,11,1419). The administration of a recombinant soluble TRAIL molecule has been shown to induce the regression or complete remission of tumors in tumor xenograft models (11,2026). The efficacy of recombinant TRAIL and agonistic antibodies recognizing either receptor DR4 or DR5 has been investigated in numerous clinical trials, as recently reviewed (27).

TRAIL has also shown promising pro-apoptotic effects in a variety of lymphoma cell lines including MCL (15). However, as with other drugs, cancer cells can develop resistance to TRAIL following prolonged exposure to sublethal doses of TRAIL (14,28). Resistance to TRAIL-mediated apoptosis can arise due to changes at the cell membrane level (typically by loss of expression or mutation of functional DR4 and/or DR5 at the cell surface) or on the intracellular level (such as incorrect formation of DISC and abberant expression of caspases) (29). The successful therapy of malignancies in general, and particularly those with very poor prognosis, such as MCL, depends on the effective management of drug resistance. An in-depth understanding of the processes involved in the development of drug resistance and a detailed description of secondary molecular changes associated with resistance are essential for successful cancer therapy. Specific molecular changes which occur in drug-resistant cells can confer a potential selective disadvatage to such cells and may be used as targets for the effective elimination of drug-resistant lymphoma cells.

The aim of this study was to elucidate the molecular mechanisms responsible for TRAIL resistance in MCL cells, as well as the secondary molecular alterations associated with this process. We also aimed to identify the phenotypic features specific for TRAIL-resistant MCL cells. If identified, these molecular features can be, at least theoretically, used as molecular targets for the effective elimination of TRAIL-resistant lymphoma cells in experimental therapies.

Materials and methods

Cell growth and cellular toxicity assay

HBL-2 cells were grown in Iscove’s modified Dulbecco’s medium in the presence of 10% foetal bovine serum, 1% penicillin-streptomycin solution in a 37°C humidified atmosphere with 5% CO2. TRAIL-resistant HBL-2/R cells were derived by selective pressure of increasing concentrations of human recombinant TRAIL (Apronex Biotechnologies, Czech Republic) up to 1,000 ng/ml in medium from the wild-type HBL-2 cells in 5 weeks. The toxicity of TRAIL to HBL-2 and HBL-2/R was measured using the colorimetric WST-8-based Quick Cell proliferation Assay kit II (BioVision, San Francisco, CA, USA) according to the manufacturer’s instructions. Briefly, 40,000 cells were seeded in a 96-well plate in 300 μl of medium supplemented with increased concentrations of TRAIL up to 1,000 ng/ml in medium for 1–4 days. After the addition of WST reagent, absorbance was measured on a Sunrise microplate absorbance reader (Tecan Group Ltd., Männedorf, Switzerland) with a 450 nm reading filter and 630 nm reference filter. The absorbance of free medium was used as the background level, triplicate samples were grown and measured for each cell type and TRAIL concentration. Mean values were calculated. All chemicals were purchased from Sigma-Aldrich (St. Louis, MO, USA) unless specified otherwise.

Flow cytometric analysis

HBL-2 and HBL-2/R cells (2×105 cells for each assay) were washed in PBS buffer (0.5% foetal bovine serum in PBS), stained with phycoerythrin-conjugated antibodies against TRAIL receptors DR4, DR5, DcR1 and DcR2 (anti-hTRAIL R1, anti-hTRAIL R2, anti-hTRAIL R3 and anti-hTRAIL R4; R&D Systems, Minneapolis, MN, USA) and analyzed by flow cytometry in triplicate (FASCCanto II, BD Biosciences, San Jose, CA, USA). Unstained cells and cells incubated with isotype controls served as the background fluorescence controls.

Sample preparation for two-dimensional electrophoresis

HBL-2 and HBL-2/R cells (6×107) were harvested, washed twice with PBS and cell pellets were frozen and stored at −80°C. Samples were thawed and homogenized in lysis buffer [7 M urea, 2 M thiourea, 4% CHAPS, 60 mM dithiothreitol (DTT) and 1% ampholytes (Bio-Lyte 3–10 Buffer, Bio-Rad, Hercules, CA, USA)] and protease inhibitor cocktail (Roche Diagnostics GmbH, Mannheim, Germany) for 20 min at room temperature with occasional vortexing. Samples were sedimented at 18,000 × g for 20 min at room temperature, supernatants were collected and protein concentration was determined by the Bradford assay (Bio-Rad). Protein concentrations in all samples were equalized to 3.3 mg/ml by dilution with lysis buffer.

Two-dimensional electrophoresis

IPG strips (pH 4.0–7.0, 24 cm; ReadyStrip, Bio-Rad) were rehydrated overnight in 450 μl of sample, representing 1.5 mg of protein. Isoelectric focusing was performed for 70 kVh, with maximum voltage not exceeding 5 kV, current limited to 50 μA per strip and temperature set to 20°C (Protean IEF Cell, Bio-Rad). Six replicates were run for each cell type. Focused strips were briefly rinsed in deionized water, equilibrated and reduced in equilibration buffer supplemented with DTT (6 M urea, 50 mM Tris pH 8.8, 30% glycerol, 2% SDS and 450 mg DTT per 50 ml) for 15 min and then alkylated in equilibration buffer with iodacetamide (1.125 mg iodacetamide per 50 ml of the buffer). Equilibrated strips were then secured on 10% SDS-PAGE and secured in place by molten agarose. SDS-PAGE electrophoresis was performed in a Tris-glycine-SDS system using a 12-gel Protean Dodeca Cell apparatus (Bio-Rad) with buffer circulation and external cooling (20°C). Gels were run at a constant voltage of 45 V per gel for 30 min and then at a constant voltage of 200 V for 6 h. Gels were washed 3 times for 15 min in deionized water to remove redundant SDS. Gels were then stained with colloidal Coomassie Brilliant Blue (SimplyBlue™ Safestain, Invitrogen, Carlsbad, CA, USA) overnight and briefly de-stained in deionized water.

Gel image analysis and extraction of peptides

Stained gels were scanned with GS 800 calibrated densitometer (Bio-Rad) and image analysis was performed with Progenesis™ software (Nonlinear Dynamics, Ltd., Newcastle upon Tyne, UK) in semi-manual mode with 6 gel replicates for each cell type. Normalization of gel images was based on total spot density, and integrated spot density values (spot volumes) were then calculated after background subtraction. Average spot volume values (averages from the all 6 gels in the group) for each spot were compared between the groups. Protein spots were considered differentially expressed if their average normalized spot volume difference was >1.5-fold. As determined by the Student’s t-test, a p-value <0.05 was considered to indicate a statistically significant difference.

Protein digestion and peptide extraction

Spots containing differentially expressed proteins were excised from the gels, cut into small pieces and washed 3 times with 25 mM ammonium bicarbonate in 50% acetonitrile (ACN). The gels were then dried in a SpeedVac Concentrator (Eppendorf, Hamburg, Germany). Sequencing grade modified trypsin (Promega, Madison, WI, USA) (6 ng/μl of trypsin in 25 mM ammonium bicarbonate in 5% ACN) was added. Following overnight incubation at 37°C, the resulting peptides were extracted with 50% ACN.

Matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF MS) and identification of selected proteins

Peptide samples were spotted on a polished steel target plate (Bruker Daltonics, Bremen, Germany) and allowed to dry at room temperature. Matrix solution (3 mg α-cyano-4-hydroxycinnamic acid in 1 ml of 50% ACN containing 0.1% trifluoroacetic acid) was then added. MS was performed on an Autoflex II MALDI-TOF/TOF mass spectrometer (Bruker Daltonics) using a solid nitrogen laser (337 nm) and FlexControl software (Bruker Daltonics) in reflectron mode with positive ion mass spectra detection. The mass spectrometer was externally calibrated with Peptide Calibration Standard II (Bruker Daltonics). Spectra were acquired in the mass range 800–4,000 Da. The peak lists were generated using FlexAnalysis and searched against Swiss-Prot (2011 version, 524420 sequences) using Mascot software. The peptide mass tolerance was set to 50 ppm, taxonomy Homo sapiens, missed cleavage was set to 2, fixed modification for cysteine carbamidomethylation, and variable modifications for methionine oxidation and protein N-terminal acetylation.

Western blot analysis

Cells were lysed in NHT buffer (140 mM NaCl, 10 mM HEPES, 1.5% Triton X-100, pH 7.4). Protein concentration in the collected supernatants was determined by the Bradford assay (Bio-Rad). Lysate samples (25–70 μg) were combined with SDS loading buffer containing 2-mercaptoethanol and boiled for 5 min. Triplicate samples were separated on 12% SDS-PAGE minigels in Tris-glycine buffer (Bio-Rad). Electrophoresis was performed at a constant voltage for 30 min at 45 V per gel, and then at 90 V per gel until the dye front reached the gel bottom. Proteins were transferred onto 0.45 μm PVDF membranes (Milipore, Billerica, MA, USA) in a semi-dry blotter (Hoefer, San Francisco, CA, USA) at 0.8 mA/cm2 of membrane. Membranes were incubated with blocking buffer containing PBS (Invitrogen), 0.1% Tween-20 and 5% non-fat dried milk for 1 h. As primary antibodies anti-adenine phosphoribosyltransferase (APRT; 1:1,000, rabbit polyclonal antibody), anti-purine nucleoside phosphorylase (PNP; 1:1,000, mouse monoclonal antibody) and anti-GAPDH (1:10,000, rabbit polyclonal antibody) were used. After thoroughly washing in blocking buffer, a secondary horseradish peroxidase-conjugated anti-mouse or anti-rabbit antibody was added (1:10,000). GAPDH was used as the loading control. The signal was detected using Western Blotting Luminol Reagent (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) and membranes were exposed to X-ray films (Kodak, Rochester, NY, USA). All used antibodies were from Santa Cruz Biotechnology.

Results

Molecular changes associated with the generation of drug-resistant cells can confer potential selective disadvantage. Such a ‘weakness’ may be used as druggable target for effective elimination of drug-resistant lymphoma cells. Our aim was to elucidate the molecular changes associated with the development of TRAIL resistance in (originally TRAIL-sensitive) MCL cells in order to identify such a cellular ‘weakness’ of TRAIL-resistant MCL cells. To identify the specific protein expression changes in the TRAIL-resistant cells, we derived a TRAIL-resistant HBL-2 subclone (HBL-2/R) from the originally TRAIL-sensitive HBL-2 cell line, and performed differential analysis of the surface expression of TRAIL receptors and comparative proteomic analysis of the HBL-2/R and HBL-2 cells.

TRAIL-resistant cell line

The TRAIL-resistant HBL-2 subclone (HBL-2/R) was derived from the originally TRAIL-sensitive HBL-2 cell line by selective pressure of increasing TRAIL concentration in medium over 5 weeks. While the IC50 for TRAIL in the originally sensitive HBL-2 cells was 1 ng/ml at 48 h (data not shown), the resulting HBL-2/R subclone proliferated in up to 1,000 ng/ml TRAIL concentration in medium and was therefore >1,000-fold more resistant to TRAIL than the HBL-2 cells (Fig. 1).

TRAIL receptors - flow cytometric analysis of cell surface expression

The attenuated expression of TRAIL death receptors DR4 and DR5 has been previously described as a cause of TRAIL resistance. We therefore determined the relative expression of TRAIL receptors in HBL-2 and HBL-2/R cells by flow cytometry (Fig. 2). The expression of DR4, DR5, DcR1 and DcR2 in the HBL-2/R cells was markedly decreased compared to the HBL-2 cells. The marked downregulation of death receptors DR4 and DR5 explains the resistance of the HBL-2/R cells to TRAIL, while the downregulation of decoy receptors DcR1 and DcR2 may indicate further, more complex phenotypic changes in the HBL-2/R cells.

Proteomic analysis

In order to identify specific changes in protein expression associated with TRAIL resistance in HBL-2/R cells, we performed comparative proteomic analysis of cellular homogenates of HBL-2/R and TRAIL-sensitive HBL-2 cells. Using two-dimensional electrophoresis of total cell lysates, we reproducibly detected 820 protein spots on Coomassie Brilliant Blue-stained gels. We found 21 protein spots to be significantly quantitatively changed (upregulated or downregulated, change >1.5-fold; p<0.05) in HBL-2/R cells (Fig. 3). Using MALDI-TOF/TOF mass spectrometry we identified all 21 proteins differentially expressed in HBL-2/R cells (Table I).

Table I

List of proteins differentially expressed in HBL-2/R cells (difference at least 1.5-fold and statistical significance p<0.05).

Table I

List of proteins differentially expressed in HBL-2/R cells (difference at least 1.5-fold and statistical significance p<0.05).

Spot no.Swiss-Prot no.aProtein nameFold changeMascot scorebSequence cov. (%)cMr
Proteins upregulated in HBL-2/R cells
1P04792Heat shock protein β-13.9845122826
2P42704Leucine-rich PPR motif-containing protein, mitochondrial2.610023159003
3O75351Vacuolar protein sorting-associated protein 4B2.61713249443
4P23381Tryptophanyl-tRNA synthetase, cytoplasmic2.42405453474
5P20591Interferon-induced GTP-binding protein Mx12.21764275872
6P09211Glutathione S-transferase P1.91105623569
7P06396Gelsolin1.91152286043
8P13010X-ray repair cross-complementing protein 51.72624683222
9Q9HAV7GrpE protein homolog 1, mitochondrial1.6994424492
10O43776Asparaginyl-tRNA synthetase, cytoplasmic1.52504163758
11Q15084Protein disulfide-isomerase A61.5762948490
Proteins downregulated in HBL-2/R cells
12P08559Pyruvate dehydrogenase E1 component subunit α3.21113243952
13P19338Nucleolin2.41462976625
14P07741Adenine phosphoribosyltransferase2.22277919766
15O75792Ribonuclease H2 subunit A1.73487233716
16Q07955 Serine/arginine-rich splicing factor 11.7823527842
17P00491Purine nucleoside phosphorylase1.61826832325
18P12268 Inosine-5′-monophosphate dehydrogenase 21.62304456226
19P40121Macrophage-capping protein1.61024138760
20P13674Prolyl 4-hydroxylase subunit α-11.52344861296
21Q15019Septin-21.5622641689

a Swiss-Prot no. is the code under which the identified protein is deposited in the Swiss-Prot database.

b Mascot score helps to estimate the correctness of the individual hit. It is expressed as −10 × log(P) where P is the probability that the observed match is a random event.

c Sequence coverage is the number of amino acids spanned by the assigned peptides divided by the sequence length.

Functional annotations of the identified differentially expressed proteins were analyzed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Among the 21 identified proteins we found molecules involved in diverse functions, including cytoskeleton regulation, ribosome synthesis and maturation, RNA metabolism, chromosome translocation, DNA repair and replication, as well as protein folding. However, one pathway was markedly enriched in our set (hsa00230 - purine metabolism) represented by 3 differentially expressed proteins. These 3 molecules are key enzymes of the purine nucleotide metabolism (Fig. 5) and all 3 are downregulated in TRAIL-resistant HBL-2/R cells [PNP (downregulated 1.6-fold in HBL-2/R cells), APRT (downregulated 2.2-fold in HBL-2/R cells) and inosine-5′-monophosphate dehydrogenase 2 (IMPDH2, downregulated 1.6-fold in HBL-2/R cells)].

Verification of proteomic analysis

To confirm the results of proteomic analysis by an independent method we verified the decreased expression of the 2 proteins involved in purine metabolism, namely PNP and APRT, by western blot analysis in HBL-2 and HBL-2/R cell lysates (Fig. 4).

Discussion

The downregulation of the 3 key enzymes of purine metabolism can have a profound effect on nucleotide homeostasis in TRAIL-resistant lymphoma cells. Purine nucleotides, the building blocks for synthesis of DNA, RNA and enzyme co-factors, are recruited either from de novo purine synthesis from low molecular weight precursors or by recycling of free nucleobases in the so-called salvage pathway. Both pathways lead to the production of nucleoside-5′-phosphates (Fig. 5). Both pathways can supply cellular demand independently; however, their importance in different tissues is variable. In leukemic and lymphoma cells the salvage pathway is considered the major source of purine nucleotides (30,31).

The de novo synthesis of purine nucleotides requires 5-phosphoribosyl-1-pyrophosphate (PRPP), ATP, glutamine, glycine, CO2, aspartate and formate to create the first fully formed nucleotide, inosine-5′-monophosphate (IMP). IMP represents a branch point for purine biosynthesis, since it can be converted either to guanosine-5′-monophosphate (GMP) by IMPDH2 (downregulated in HBL-2/R cells) or to adenosine-5′-monophosphate (Fig. 5).

The catabolism of purine nucleotides leads to the liberation of free purine bases by PNP (downregulated in HBL-2/R cells). In the salvage pathway the free bases are reconverted back to nucleoside-5′-monophosphates in a reaction with activated sugar (PRPP) catalyzed by APRT (downregulated in HBL-2/R cells) or hypoxanthine-guanine phosphoribosyltransferase (32) (Fig. 5). Ribonucleotides are converted by ribonucleotide reductase into the corresponding deoxyribonucleotides.

The delicate balance of enzyme activities and concentrations of products and intermediates are critical for purine (nucleotide) homeostasis. The inhibition of PNP results in the accumulation of its substrate, 2′-deoxyguanosine which is further phosphorylated to deoxyguanosine triphosphate (dGTP). A high intracellular concentration of dGTP inhibits cell proliferation and induces apoptosis (3335). If APRT is inhibited, accumulated adenine is oxidized to insoluble 2,8-dihydroxyadenine. Accumulation of this precipitate results in cell death (32). Similarly, the inhibition of IMPDH2 leads to depletion of guanosine nucleotides, which blocks DNA synthesis and cell division (36,37).

Disruption of the purine nucleotide metabolism generally results in an accumulation and/or a lack of ribonucleotides or deoxyribonucleotides or metabolic intermediates with potentially cytotoxic consequences. The observed decreased expression of the 3 purine metabolism enzymes affects both de novo synthesis and the salvage pathway of purine metabolism and may also affect purine nucleotide homeostasis in TRAIL-resistant HBL-2/R cells. Such an imbalance may represent a selective disadvantage for the affected cells. Such a ‘weakness’ may not be apparent under normal circumstances but may become critical under stress or unfavorable conditions. As the proliferation rates of HBL-2/R and HBL-2 cells are comparable, the proposed imbalance in purine nucleotide metabolism in TRAIL-resistant cells is possibly mild and/or well compensated in vitro. However, this ‘weakness’ may become apparent due to lack of building blocks for DNA and RNA synthesis in the environment or upon further disruption of purine metabolism. Since both pathways of purine metabolism are compromised in TRAIL-resistant MCL cells, these cells should be vulnerable to further inactivation of purine nucleotide metabolism enzymes. Therefore, drugs that target (already disbalanced) purine metabolism should be highly cytotoxic to TRAIL-resistant cells (compared to non-malignant cells) and may therefore be selectively effective in the elimination of TRAIL-resistant MCL cells in experimental therapy. There are several approved inhibitors of purine metabolism, such as methotrexate (inhibits purine de novo synthesis via dihydrofolate reductase) (38), ribavirin and mycophenolic acid (inhibitors of IMPDH2) (39,40) or forodesine (a novel inhibitor of PNP) (41,42), available for clinical use.

The adaptation of cancer cells to cytostatic and cytotoxic drugs is associated to a certain degree with extensive changes in the cell phenotype. Some of the molecular changes, although seemingly unrelated to the mechanism of resistance, can provide a selective disadvantage to the cells and such a ‘weakness’ may be used as a potential therapeutic target. By the presented proteomic analysis of the changes associated with resistance to TRAIL in MCL HBL-2 cells, we demonstrated the downregulation of all types of TRAIL receptors and identified the altered expression of several proteins including 3 enzymes of the purine metabolism pathway. This downregulated pathway potentially represents a ‘weakness’ of the TRAIL-resistant MCL cells and has potential as a therapeutic target for the selective elimination of such cells in the future.

Acknowledgements

This study was supported by the Grant Agency of Charles University (GAUK 251180 111210 and 253284 700712), by the Grant Agency of the Czech Republic (305/09/1390), by the Ministry of Education, Youth and Sports (PRVOUK P24/LF1/3 and SVV 2012-264507), UNCE 204021 and the Ministry of Health of the Czech Republic (IGA MZ NT12248-5, IGA-MZ NT13201-4).

References

1 

Sant M, Allemani C, Tereanu C, et al: Incidence of hematologic malignancies in Europe by morphologic subtype: results of the HAEMACARE project. Blood. 116:3724–3734. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Perez-Galan P, Dreyling M and Wiestner A: Mantle cell lymphoma: biology, pathogenesis, and the molecular basis of treatment in the genomic era. Blood. 117:26–38. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Tsujimoto Y, Yunis J, Onorato-Showe L, Erikson J, Nowell PC and Croce CM: Molecular cloning of the chromosomal breakpoint of B-cell lymphomas and leukemias with the t(11;14) chromosome translocation. Science. 224:1403–1406. 1984. View Article : Google Scholar : PubMed/NCBI

4 

Williams ME, Swerdlow SH, Rosenberg CL and Arnold A: Characterization of chromosome 11 translocation breakpoints at the bcl-1 and PRAD1 loci in centrocytic lymphoma. Cancer Res. 52:5541S–5544S. 1992.PubMed/NCBI

5 

Humala K and Younes A: Current and emerging new treatment strategies for mantle cell lymphoma. Leuk Lymphoma. Feb 19–2013.(Epub ahead of print).

6 

Wiley SR, Schooley K, Smolak PJ, et al: Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity. 3:673–682. 1995. View Article : Google Scholar : PubMed/NCBI

7 

Pitti RM, Marsters SA, Ruppert S, Donahue CJ, Moore A and Ashkenazi A: Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family. J Biol Chem. 271:12687–12690. 1996. View Article : Google Scholar : PubMed/NCBI

8 

Ashkenazi A and Dixit VM: Death receptors: signaling and modulation. Science. 281:1305–1308. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Sheridan JP, Marsters SA, Pitti RM, et al: Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors. Science. 277:818–821. 1997. View Article : Google Scholar : PubMed/NCBI

10 

Ashkenazi A: Targeting death and decoy receptors of the tumour-necrosis factor superfamily. Nat Rev Cancer. 2:420–430. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Castro Alves C, Terziyska N, Grunert M, et al: Leukemia-initiating cells of patient-derived acute lymphoblastic leukemia xenografts are sensitive toward TRAIL. Blood. 119:4224–4227. 2012.PubMed/NCBI

12 

Peter ME and Krammer PH: The CD95(APO-1/Fas) DISC and beyond. Cell Death Differ. 10:26–35. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Spierings DC: Tissue distribution of the death ligand TRAIL and its receptors. J Histochem Cytochem. 52:821–831. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Petrak J, Toman O, Simonova T, et al: Identification of molecular targets for selective elimination of TRAIL-resistant leukemia cells. From spots to in vitro assays using TOP15 charts. Proteomics. 9:5006–5015. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Molinsky J, Klanova M, Koc M, et al: Roscovitine sensitizes leukemia and lymphoma cells to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. Leuk Lymphoma. 54:372–380. 2013. View Article : Google Scholar

16 

Klener P, Leahomschi S, Molinsky J, et al: TRAIL-induced apoptosis of HL60 leukemia cells: two distinct phenotypes of acquired TRAIL resistance that are accompanied with resistance to TNFalpha but not to idarubicin and cytarabine. Blood Cells Mol Dis. 42:77–84. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Leahomschi S, Molinsky J, Klanova M, et al: Multi-level disruption of the extrinsic apoptotic pathway mediates resistance of leukemia cells to TNF-related apoptosis-inducing ligand (TRAIL). Neoplasma. 60:223–231. 2013. View Article : Google Scholar

18 

Wen J, Ramadevi N, Nguyen D, Perkins C, Worthington E and Bhalla K: Antileukemic drugs increase death receptor 5 levels and enhance Apo-2L-induced apoptosis of human acute leukemia cells. Blood. 96:3900–3906. 2000.PubMed/NCBI

19 

Plasilova M, Zivny J, Jelinek J, et al: TRAIL (Apo2L) suppresses growth of primary human leukemia and myelodysplasia progenitors. Leukemia. 16:67–73. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Ashkenazi A, Pai RC, Fong S, et al: Safety and antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest. 104:155–162. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Di Pietro R and Zauli G: Emerging non-apoptotic functions of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)/Apo2L. J Cell Physiol. 201:331–340. 2004.

22 

Kelley SK, Harris LA, Xie D, et al: Preclinical studies to predict the disposition of Apo2L/tumor necrosis factor-related apoptosis-inducing ligand in humans: characterization of in vivo efficacy, pharmacokinetics, and safety. J Pharmacol Exp Ther. 299:31–38. 2001.

23 

Lawrence D, Shahrokh Z, Marsters S, et al: Differential hepatocyte toxicity of recombinant Apo2L/TRAIL versions. Nat Med. 7:383–385. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Roth W, Isenmann S, Naumann U, et al: Locoregional Apo2L/TRAIL eradicates intracranial human malignant glioma xenografts in athymic mice in the absence of neurotoxicity. Biochem Biophys Res Commun. 265:479–483. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Walczak H, Miller RE, Ariail K, et al: Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat Med. 5:157–163. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Hylander BL, Pitoniak R, Penetrante RB, et al: The anti-tumor effect of Apo2L/TRAIL on patient pancreatic adenocarcinomas grown as xenografts in SCID mice. J Transl Med. 3:222005. View Article : Google Scholar : PubMed/NCBI

27 

Dimberg LY, Anderson CK, Camidge R, Behbakht K, Thorburn A and Ford HL: On the TRAIL to successful cancer therapy? Predicting and counteracting resistance against TRAIL-based therapeutics. Oncogene. 14–May;2012.(Epub ahead of print). View Article : Google Scholar : 1642012.PubMed/NCBI

28 

Zhang L and Fang B: Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Ther. 12:228–237. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Maksimovic-Ivanic D, Stosic-Grujicic S, Nicoletti F and Mijatovic S: Resistance to TRAIL and how to surmount it. Immunol Res. 52:157–168. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Scavennec J, Maraninchi D, Gastaut JA, Carcassonne Y and Cailla HL: Purine and pyrimidine ribonucleoside monophosphate patterns of peripheral blood and bone marrow cells in human acute leukemias. Cancer Res. 42:1326–1330. 1982.PubMed/NCBI

31 

Natsumeda Y, Prajda N, Donohue JP, Glover JL and Weber G: Enzymic capacities of purine de novo and salvage pathways for nucleotide synthesis in normal and neoplastic tissues. Cancer Res. 44:2475–2479. 1984.PubMed/NCBI

32 

Bollee G, Harambat J, Bensman A, Knebelmann B, Daudon M and Ceballos-Picot I: Adenine phosphoribosyltransferase deficiency. Clin J Am Soc Nephrol. 7:1521–1527. 2012. View Article : Google Scholar

33 

Bantia S, Miller PJ, Parker CD, et al: Purine nucleoside phosphorylase inhibitor BCX-1777 (Immucillin-H) - a novel potent and orally active immunosuppressive agent. Int Immunopharmacol. 1:1199–1210. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Bantia S, Montgomery JA, Johnson HG and Walsh GM: In vivo and in vitro pharmacologic activity of the purine nucleoside phosphorylase inhibitor BCX-34: the role of GTP and dGTP. Immunopharmacology. 35:53–63. 1996. View Article : Google Scholar : PubMed/NCBI

35 

Galmarini CM, Popowycz F and Joseph B: Cytotoxic nucleoside analogues: different strategies to improve their clinical efficacy. Curr Med Chem. 15:1072–1082. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Allison AC and Eugui EM: Mycophenolate mofetil and its mechanisms of action. Immunopharmacology. 47:85–118. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Hedstrom L: IMP dehydrogenase: structure, mechanism, and inhibition. Chem Rev. 109:2903–2928. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Fairbanks LD, Ruckemann K, Qiu Y, et al: Methotrexate inhibits the first committed step of purine biosynthesis in mitogen-stimulated human T-lymphocytes: a metabolic basis for efficacy in rheumatoid arthritis? Biochem J. 342:143–152. 1999. View Article : Google Scholar

39 

Allison AC and Eugui EM: The design and development of an immunosuppressive drug, mycophenolate mofetil. Springer Semin Immunopathol. 14:353–380. 1993. View Article : Google Scholar : PubMed/NCBI

40 

Zhou S, Liu R, Baroudy BM, Malcolm BA and Reyes GR: The effect of ribavirin and IMPDH inhibitors on hepatitis C virus subgenomic replicon RNA. Virology. 310:333–342. 2003. View Article : Google Scholar : PubMed/NCBI

41 

Gandhi V, Kilpatrick JM, Plunkett W, et al: A proof-of-principle pharmacokinetic, pharmacodynamic, and clinical study with purine nucleoside phosphorylase inhibitor immucillin-H (BCX-1777, forodesine). Blood. 106:4253–4260. 2005. View Article : Google Scholar

42 

Miles RW, Tyler PC, Furneaux RH, Bagdassarian CK and Schramm VL: One-third-the-sites transition-state inhibitors for purine nucleoside phosphorylase. Biochemistry. 37:8615–8621. 1998. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May 2013
Volume 31 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pospisilova J, Vit O, Lorkova L, Klanova M, Zivny J, Klener P and Petrak J: Resistance to TRAIL in mantle cell lymphoma cells is associated with the decreased expression of purine metabolism enzymes. Int J Mol Med 31: 1273-1279, 2013
APA
Pospisilova, J., Vit, O., Lorkova, L., Klanova, M., Zivny, J., Klener, P., & Petrak, J. (2013). Resistance to TRAIL in mantle cell lymphoma cells is associated with the decreased expression of purine metabolism enzymes. International Journal of Molecular Medicine, 31, 1273-1279. https://doi.org/10.3892/ijmm.2013.1302
MLA
Pospisilova, J., Vit, O., Lorkova, L., Klanova, M., Zivny, J., Klener, P., Petrak, J."Resistance to TRAIL in mantle cell lymphoma cells is associated with the decreased expression of purine metabolism enzymes". International Journal of Molecular Medicine 31.5 (2013): 1273-1279.
Chicago
Pospisilova, J., Vit, O., Lorkova, L., Klanova, M., Zivny, J., Klener, P., Petrak, J."Resistance to TRAIL in mantle cell lymphoma cells is associated with the decreased expression of purine metabolism enzymes". International Journal of Molecular Medicine 31, no. 5 (2013): 1273-1279. https://doi.org/10.3892/ijmm.2013.1302