The TNF superfamily ligands APRIL and BAFF bind with different affinity to two receptors, BCMA and TACI, and induce cell survival and/or proliferation, whereas BAFF also binds specifically to BAFFR. These molecules were considered specific for the immune system. Recently, however, they were also found in epithelial and mesenchymal noncancerous and cancerous tissues and cell lines. In this article, we report that hepatocellular carcinoma (HCC) cell lines HepG2 and Hep3B and HCC specimens express APRIL and BAFF and their receptors BCMA and BAFFR, but not TACI; APRIL/BCMA is enhanced in HCC, compared with normal liver tissue. In contrast to previous reports, APRIL binding to BCMA decreases cell proliferation by inducing G2/M cell cycle arrest, whereas BAFF has no effect on cell growth. HCC cells therefore represent a rare system in which these two ligands (APRIL and BAFF) exert a differential effect and may serve as a model for specific APRIL/BCMA actions. We show that the effect of APRIL is mediated via BCMA, which does not activate the classical NF-κB pathway, whereas it induces a novel signaling pathway, which involves JNK2 phosphorylation, FOXO3A activation, and GADD45 transcription. In addition, JNK2 mediates the phosphorylation of Akt, which is activated but does not participate in the antiproliferative effect of APRIL. Furthermore, transcriptome analysis revealed that APRIL modifies genes specifically related to cell cycle modulation, including MCM2/4/5/6, CDC6, PCNA, and POLE2. Our data, therefore, identify a novel APRIL/BCMA signaling pathway in HCC and suggest that APRIL could have a pleiotropic role in tumor biology.

APRIL (TNFSF13) (1) is a member of the TNF superfamily, produced by macrophages, monocytes, and dendritic cells (2). APRIL and BAFF (TNFSF13B) (3) bind to two receptors (47), BCMA (TNRSF17) (8, 9) and TACI (TNFRSF13B) (10), but with a different affinity (1113); BAFF also binds to its specific receptor, BAFFR (TNFRSF13C) (14, 15). APRIL has been reported to bind also the proteoglycan syndecan-1 (CD138) (16, 17), and a not-yet-identified APRIL receptor has been advanced, as well (18, 19). This complex system of two ligands and (at least) three receptors was considered exclusive to B lymphocytes; however, recently, all or some of these five molecules have been identified in mesenchymal cells (20), normal tissues (2024), and epithelial tumors (20, 21, 2426), suggesting additional non–immune cell-related functions for these ligands and receptors, in both physiological and malignant tissues. APRIL has also been found to be expressed in some normal germinal-center B cells, myeloid and dendritic cells, malignant B cells and cell lines, activated T cells, glioblastoma cell lines, and the placenta (1, 18, 2733). BAFF has also been detected in some of these tissues, but no significant differences between normal and neoplastic tissue are usually reported; the expression of its cognate receptor BAFFR is either missing or occurs independently of tumor evolution (20, 21, 2426). Binding of APRIL or BAFF to BCMA or TACI triggers diverse signaling pathways in engineered cells, including nuclear translocation of NF-κB, activation of p38 mitogen-activated kinase and JNK phosphorylation for BCMA (34), and NF-κB nuclear translocation and JNK phosphorylation for TACI (11).

Previous investigations identified the presence of BCMA transcripts in liver tissue (35). In this article, we report that APRIL and BCMA are expressed in hepatocellular carcinoma (HCC) specimens and two HCC cell lines (HepG2, Hep3B), as well as in normal liver and in a small series of human HCCs, whereas TACI is absent. We further show that APRIL, which is usually an inducer of cell growth, binds to BCMA and inhibits cell proliferation in HCC cells, through G2/M cell cycle arrest, whereas BAFF has no effect on the proliferation of these cells. These data suggest that HepG2 and Hep3B cells could be a convenient model for analyzing specific events mediated exclusively by BCMA in nonengineered cells. We further report that the APRIL/BCMA antiproliferative action is mediated via a novel signaling pathway that involves JNK2 phosphorylation, FOXO3A activation, and GADD45 transcription.

Human HCC-derived lines HepG2 and Hep3B were purchased from DSMZ (Braunschweig, Germany); they were cultured in RPMI 1640 and DMEM, respectively, supplemented with 10% FBS (Invitrogen, Carlsbad, CA), at 37°C, 5% CO2. Wortmannin, LY-294002 (PI3K/Akt inhibitors), and SP600125 (pan-JNK inhibitor) were purchased from Calbiochem (San Diego, CA). All other chemicals were purchased from Sigma-Aldrich (St. Louis, MO), unless stated otherwise. To exclude the possibility of a batch/production effect on our experiments, we have used for almost all experiments recombinant APRIL purchased from either Abcam (Cambridge, U.K.) or ROCHE Hellas (Marousi, Greece).

Twelve histologically verified HCC samples were retrieved from the Pathology Department (University of Crete, School of Medicine) tissue archives, after formal approval from the University Hospital Research and Ethics Committee. Seven normal liver samples acquired from autopsies after car accidents, with consent of the families, were also used. Immunohistochemistry in pathology samples and in HCC cell lines was performed as previously described (24, 36), in 3-μm sections. Abs against APRIL (ALX-804-149-C100, mouse monoclonal), BAFF (ALX-804-131-C100, rat monoclonal), and BCMA (ALX-804-151-C100, rat monoclonal) were from Alexis (Lausanne, Switzerland). The Ab against BAFFR (AF1162, goat polyclonal) was from R&D Systems (Minneapolis, MN), whereas the Ab against TACI (Sc-80335, mouse monoclonal) was from Santa Cruz Biotechnology (Santa Cruz, CA). Detection systems were TL-125-AL, Lab Vision Fast Red from Thermo Fisher Scientific (Fremont, CA), or K1500 (DAB) and K0689 (Fast Red) from Dako (Glostrup, Denmark). Positive controls according to our previous studies in human cells and tissues [described in previous publications (20, 24, 36, 37)] are presented in Supplemental Fig. 1A.

A hybrid construct encoding the signal peptide sequence of human H chain γ 1 fused to the extracellular part of human BCMA, connected in-phase with the Fc segment of human IgG1, was introduced into the plasmid p7055, as described previously (18). The secreted BCMA-Fc protein was collected in the supernatant and purified on protein A–agarose beads.

RT-PCR was performed as described previously (24). Positive controls were run in parallel with samples included in the study. We used adipose-derived mesenchymal stem cells as positive controls of BAFF and APRIL (20) and isolated human lymphocytes as positive controls for BAFFR, TACI, and BCMA. Quantitative RT-PCR (qRT-PCR) was performed as described previously (38). Changes were normalized according to 18S RNA expression. The primers used for the study were as follows: BAFF forward: 5′-TTC TAG GGC ACT TCC CCT TT-3′, reverse: 5′-CTC AAG ACT GCT TGC AAC TGA-3′; APRIL forward: 5′-TCT CCT TTT CCG GGA TCT CT-3′, reverse: 5′-CCA GAA TGG GGA AGG GTA TC-3′; BAFFR forward: 5′-AGG ACG CCC CAG AGC C-3′, reverse: 5′-AGT GTC TGT GCT TCT GCA GG-3′; TACI forward: 5′-AGT GAA CCT TCC ACC AGA GC-3′, reverse: 5′-CTC TTC TTG AGG AAG CAG GC-3′; BCMA forward: 5′-GTC AGC GTT ATT GTA ATG CAA GTG T-3′, reverse: 5′-TCT TTT CCA GGT CAA TGT TAG CC-3′; MCM2 forward: 5′-CAC CAC GTA CCT TGT GCT TG-3′, reverse: 5′-AAG GAT CAG CAG ATC GGA GA-3′; MCM4 forward: 5′-ACG TTT TGC ATC CGT TTT C-3′, reverse: 5′-CCT GGG GAC AGA GTG AAT GT-3′; MCM5 forward: 5′-AGG GAT CTT CAC CAG GTG TG-3′, reverse: 5′-GCC AAG GAG GTA GCT GAT GA-3′; MCM6 forward: 5′-CGA GGG AAT AGA CAC GAT CA-3′, reverse: 5′-CAT GTC CCG ATT CGA TCT CT-3′; PCNA forward: 5′-TCA GGT ACC TCA GTG CAA AAG-3′, reverse: 5′-TGC AAG TGG AGA ACT TGG AA-3′; RPA3 forward: 5′-CTC CCT TAG CTG CGT CAC TC-3′, reverse: 5′-GAG AAA GTG CTC AAG AAT GTT CA-3′; POLE2 forward: 5′-TTT TGC AGA AGT CTT CAC AGA TG-3′, reverse: 5′-GCA GAA GGT TGG TTT GAA GA-3′; GADD45 forward: 5′-GCA GGA TCC TTC CAT TGA GA-3′, reverse: 5′-CTC TTG GAG ACC GAC GCT G-3′; JNK1 forward: 5′-GCC AGA CCG AAG TCA AGA AT-3′, reverse: 5′-CAA GCA CCT TCA TTC TGC TG-3′; JNK2 forward: 5′-TTC AGG GTG CAG TCT GAT TTC-3′, reverse: 5′-TTC TTT ACC AGA TGC TTT GTG G-3′; 18S RNA forward: 5′-ATG GTC AAC CCC ACC GTG T-3′, reverse: 5′-TTC TGC TGT CTT TGG AAC TTT GTC-3′. They were selected from qPrimer Depot (qPrimerDepot, http://primerdepot.nci.nih.gov) and synthesized by VBC Biotech (Vienna, Austria).

Centrifugation-cleared supernatants from cells incubated for 24 h in six-well plates were collected, and cells were lysed and used for protein determination-normalization. APRIL and BAFF were assayed on pooled, 5×-concentrated media, using ELISA kits (Bender MedSystems, Vienna, Austria), according to the manufacturer’s instructions. The effect of the medium was systematically subtracted. Results are expressed as nanograms per milligram of total cellular proteins (measured with the Bradford Assay) per 24 h.

Short hairpin RNA (shRNA) against JNK1 and JNK2 was prepared with the use of the psiRNA-h7SKGFPzeo Kit (Invivogen, San Diego, CA), according to the manufacturer’s instructions. Briefly, psiRNA-h7SKGFPzeo plasmid was digested with BbsI (New England Biolabs, Ipswitch, MA) and was gel-purified with Extract II columns (Macherey-Nagel, Duren, Germany). The following oligonucleotides were used: JNK1 oligo1: 5′-ACC TCG AGT CGG TTA GTC ATT GAT AGT CAA GAG CTA TCA ATG ACT AAC CGA CTC TT-3′; JNK1 oligo2: 5′-CAA AAA GAG TCG GTT AGT CAT TGA TAG CTC TTG ACT ATC AAT GAC TAA CCG ACT CG-3′; JNK2 oligo1: 5′-ACC TCG AGC AGT TAG AGT AGG TGA ATT CAA GAG ATT CAC CTA CTC TAA CTG CTC TT-3′; JNK2 oligo2: 5′-CAA AAA GAG CAG TTA GAG TAG GTG AAT CTC TTG AAT TCA CCT ACT CTA ACT GCT CG-3′; scrambled shRNA oligo-1: 5′-ACC TCG GGT ATT TAG GCT ACG ATA GTT CAA GAG ACT ATC GTA GCC TAA ATA CCC TT-3′; scrambled shRNA oligo-2: 5′-CAA AAA GGG TAT TTA GGC TAC GAT AGT CTC TTG AAC TAT CGT AGC CTA AAT ACC CG-3′. Annealed oligonucleotides (95°C for 5 min and left to cool slowly to 35°C) were ligated with T4-DNA ligase (TAKARA, Otsu, Shiga, Japan) with the digested psiRNA-h7SKGFPzeo plasmid and used for the transformation of LyoComp GT116 cells, plated on Fast-Media Zeo X–Gal plates. After 24 h incubation at 37°C, white colonies were picked, and minipreps, positive for the insert, were prepared by incubation of isolated DNA, and digested with SpeI. A single positive miniprep was selected for expansion, and the presence of the proper insert was verified with sequencing.

Cells were grown for 6 d, with a change of medium and fresh APRIL or BAFF at day 3. Cell proliferation/viability was measured by the MTT assay. All substances were added to cultures 1 d after seeding (designated day 0). APRIL concentrations ranging from 1 to 200 ng/ml were used for these experiments.

For lentiviral shRNA studies, cells were plated at an initial density of 1.5 × 104 cells in 24-well plates, with 0.5 ml medium per well. They were treated with MISSION anti-BCMA shRNA Lentiviral Transduction Particles (Sigma-Aldrich), according to the manufacturer’s instruction, in a hexadimethrine bromide environment (final concentration 8 μg/ml). The shRNA nontarget control particles were used as a negative control, and Mission TurboGFP Control Particles were used for assessment of transfection efficiency (>80% in all cases). The effectiveness of BCMA knockdown was verified with qRT-PCR (Supplemental Fig. 1B).

For JNK1 and two knockdown studies, cells were plated in either six-well or 24-well plates (2 × 106 and 1.5 × 104 cells/well, respectively) and left to adhere for 24 h. The medium was changed, and transfection was performed with a standard Lipofectamine 2000 protocol (Invitrogen; 0.8 μg DNA, 1 μl Lipofectamine 2000 in Optimem medium, for each well of a 24-well plate, scaling up for 6-well plates) and used, as described in 14Results, after 48 h. Transfection efficiency was >85%, as estimated based on GFP-positive cells. JNK1 and JNK2 knockdown was verified with qRT-PCR (Supplemental Fig. 1C) and Western blot (see 14Results).

Apoptosis of treated cells (APRIL, 200 ng/ml) was evaluated with (1) the Annexin VFITC Apoptosis Detection Kit and (2) the Caspase-3 Activity Assay (both from BD Biosciences, San Diego, CA), by flow cytometry on a FACSCalibur (BD Biosciences), according to the manufacturer’s instructions. Treated cells were compared with control (nontreated) samples.

For the assay of APRIL-induced modification of the cell cycle, treated (APRIL, 200 ng/ml) and control cells (1 × 106 per well) were incubated for a minimum of 2 h at 4°C in the dark, with 1 ml of appropriate buffer (3.4 mM citrate, 25 mM NaCl, pH 7.6, 10% Nonidet P-40, 10 mg/ml RNase A, 10 mg/ml propidium iodide); measured by flow cytometry; and analyzed with the CELLQuest (BD) and ModFit LT (Verify Software, Topsham, MN) software. Experiments were performed three times in triplicates.

Cells, in 24-well plates, were transfected with 0.5 μg per well of pNFκB-Luc plasmid (Clontech, Mountain View, CA), carrying NF-κB response elements, in front of the 5′ end of the firefly luciferase gene, together with 0.5 μg per well of a Renilla luciferase vector (pRL-CMV; Promega, Fitchburg, WI), using Lipofectamine 2000 (Invitrogen, 1 μl per well) in Optimem medium. Cells were incubated for 24 h and then treated with APRIL (200 ng/ml, 24 h). Luciferase activity was assayed with a Dual-Luciferase Reporter 1000 Assay System (Promega), in a Berthold FB12 Luminometer (Bad Wildbad, Germany).

FOXO3A and E2F activation was performed with the Cignal Reporter Assay plasmids (SA Biosciences, Frederick, MD), according to the manufacturer’s instructions. Briefly, 105 cells in 24-well plates were transfected with the reporter, or the negative or positive control (250ng/well), according to a standard Lipofectamine 2000 protocol. After a 24-h incubation, cells were treated (APRIL, 200 ng/ml), and 24 h later, luciferase activity was assayed.

Treated cells (APRIL, 200 ng/ml) in six-well plates were lysed in Laemmli's buffer; electrophoresis and blotting were performed according to standard procedures. The following Abs were used: phospho-JNK1/2 and total JNK (Abcam; 1/200), phospho-p38 [Thr180, Tyr182; (Cell Signaling Technology, Danvers, MA; 1/1000)], phospho-Akt and total Akt (Cell Signaling Technology; 1/1000), and anti-actin (Santa Cruz Biotechnology; 1/2000). At least three independent experiments were performed in each case.

HepG2 cells were treated with APRIL (200 ng/ml) for 2, 6, and 12 h. Total RNA was labeled and hybridized according to the Affymetrix protocol, using the Human Genome U133A Plus 2.0 Affymetrix GeneChip (54,675 transcripts). Normalization of signal and analysis were performed with Genespring GX V11.0 (Agilent, Foster City, CA). Transcripts modified by a factor of 2 in either direction (APRIL/vehicle) were extracted and further analyzed by use of the online resource gene set enrichment analysis (http://www.broadinstitute.org/gsea), followed by leading edge analysis (39). Finally, genes modified by APRIL after a 12-h incubation were analyzed for the reverse detection of putative transcription factors, by using the Web resource TFactS (http://www.tfacts.org) (40). Gene array data have been stored at the National Institutes of Health Gene Expression Omnibus repository (http://www.ncbi.nlm.nih.gov/geo/) under accession number GSE29375.

Statistical analysis was performed with the PASW v. 19.0 (SPSS, Chicago, IL) program. A statistical threshold of p < 0.05 was retained for significance.

APRIL and BCMA show an intense immunocytochemical expression in HepG2 and Hep3B cells (Fig. 1A). BCMA presents an intracellular and peripheral expression, compatible with a membrane or submembrane localization; in contrast, TACI immunostaining is absent, whereas BAFF and BAFFR are also expressed. A dotted expression of APRIL is observed, compatible with its mode of synthesis and secretion (2).

FIGURE 1.

Expression of APRIL and BCMA in HCC-derived cell lines. (A) APRIL and BCMA are expressed in HCC-derived cell lines. TACI immunoreactivity is absent, whereas staining for BAFF and BAFFR is also observed. A typical IHC is presented, repeated three times, in different cell preparations, with similar results. HepG2 cells are presented in the left column and Hep3B in the right. Original magnification ×200 in all panels. APRIL, TACI, BAFF, and BAFFR were detected with Fast Red and BCMA with DAB (see Materials and Methods for details). (B) RT-PCR of ligands (APRIL, BAFF) and receptors (BCMA, BAFFR, TACI) in HepG2 and Hep3B cells. Positive controls (adipose tissue-derived mesenchymal cells for APRIL and BAFF and isolated human lymphocytes for BCMA, TACI, and BAFFR) are also included. (C) Flow cytometry detection of membrane-expressed BCMA and BAFFR and absence of TACI staining in HepG2 and Hep3B cells, identified by FITC-labeled anti-receptor Abs. Mean fluorescence intensity (MFI) is also reported for unstained (control) and stained cells.

FIGURE 1.

Expression of APRIL and BCMA in HCC-derived cell lines. (A) APRIL and BCMA are expressed in HCC-derived cell lines. TACI immunoreactivity is absent, whereas staining for BAFF and BAFFR is also observed. A typical IHC is presented, repeated three times, in different cell preparations, with similar results. HepG2 cells are presented in the left column and Hep3B in the right. Original magnification ×200 in all panels. APRIL, TACI, BAFF, and BAFFR were detected with Fast Red and BCMA with DAB (see Materials and Methods for details). (B) RT-PCR of ligands (APRIL, BAFF) and receptors (BCMA, BAFFR, TACI) in HepG2 and Hep3B cells. Positive controls (adipose tissue-derived mesenchymal cells for APRIL and BAFF and isolated human lymphocytes for BCMA, TACI, and BAFFR) are also included. (C) Flow cytometry detection of membrane-expressed BCMA and BAFFR and absence of TACI staining in HepG2 and Hep3B cells, identified by FITC-labeled anti-receptor Abs. Mean fluorescence intensity (MFI) is also reported for unstained (control) and stained cells.

Close modal

Immunocytochemical data were further confirmed by RT-PCR (Fig. 1B), revealing expression of APRIL, BCMA, BAFF, and BAFFR, whereas TACI expression was not evidenced. Relative quantification of gene expression by qRT-PCR analysis (41) revealed that BAFFR is expressed 10.98 ± 2.34-fold and 6.34 ± 1.23-fold more than BCMA in HepG2 and Hep3B cells, respectively. In addition, flow cytometry in nonpermeabilized cells (Fig. 1C) confirmed the absence of TACI and identified relatively low membrane expression of BAFFR and BCMA. Further, APRIL and BAFF were also found to be secreted to culture medium (HepG2, 2.35 and 5.08 ng/mg protein per 24 h, and Hep3B, 7.23 and 1.49 ng/mg protein per 24 h, respectively). This low level of secreted BAFF makes improbable its interaction with BCMA, in view of BAFF’s substantially lower affinity for the receptor (1113).

APRIL and BAFF are considered cell proliferation promoters (4245). However, in HCC cells, a differential effect was found: BAFF has no effect on cell proliferation (Supplemental Fig. 1D), whereas addition of APRIL results in a dose- and time-dependent, moderate inhibition of cell growth (Fig. 2A). This effect is specific, as the addition of Fc-BCMA, a soluble form of BCMA, results in a reversion of APRIL action (Fig. 2B). The exclusive implication of BCMA in this action is verified by the use of anti-BCMA shRNA. BCMA-specific, but not nontarget shRNA, made cells insensitive to APRIL (Fig. 2C). The APRIL effect is not due to apoptosis, assayed by Annexin V/propidium iodide staining (control and APRIL-treated cells, 2.61 and 1.38% in HepG2, and 2.96 and 0.75% in Hep3B cells, respectively; Supplemental Fig. 1E) or cleaved caspase-3 activity (4.82% versus 3.89% in HepG2 and 3.71% versus 3.52% in Hep3B cells for control and APRIL-incubated cells, respectively). We further report that APRIL induced a marginal but significant arrest of cells at the G2/M phase of the cell cycle (Fig. 2D), a result that could explain the accumulated effect seen in our 3- and 6-d proliferation experiments. However, minor changes in the ratio phospho/total cdc2, or cyclin B1 levels are observed (∼20% reduction of p-cdc2/cdc2; data not shown), suggesting an alternative mechanism for APRIL-induced G2/M cell cycle growth arrest.

FIGURE 2.

APRIL reduces cell growth and induces a blockade in the G2/M phase of the cell cycle, acting through BCMA. (A) Dose and time effect of APRIL (200 ng/ml) on cell growth of HepG2 and Hep3B cell lines. Cells were incubated for 3 and 6 d; the medium was changed once, at day 3, and supplemented with fresh APRIL. Mean ± SEM of three different assays is presented, performed in triplicates. (B) Addition of Fc-BCMA, a soluble form of the receptor binding APRIL, reverts the effect of APRIL (200 ng/ml) in both cell lines, whereas Fc-BCMA alone or nonspecific IgG had no effect. Mean ± SEM of three independent assays in triplicate. Incubation time was 6 d. (C) Transfection of cells with shBCMA RNA reverts the effect of APRIL (200 ng/ml) on cell growth, after 6 d of incubation. In contrast, addition of nontarget shRNA has no effect. Results (mean ± SEM of three experiments in quadruplicate) are normalized compared with control (non-APRIL or shRNA-treated) cells. (D) Effect of APRIL (200 ng/ml) on the cell cycle of HepG2 (upper row) and Hep3B cells (lower row). Cells were synchronized by serum starvation for 24 h and then treated with 200 ng/ml APRIL in full culture medium for 24 additional hours. Annexin/propidium iodine staining and FACS analysis were performed as described in 2Materials and Methods. Similar results were obtained in cells treated for 48 h with APRIL (not shown). This figure presents a typical example, as analyzed with the ModFit program, and the table below shows mean ± SEM of three repetitions. *p < 0.05, **p < 0.01.

FIGURE 2.

APRIL reduces cell growth and induces a blockade in the G2/M phase of the cell cycle, acting through BCMA. (A) Dose and time effect of APRIL (200 ng/ml) on cell growth of HepG2 and Hep3B cell lines. Cells were incubated for 3 and 6 d; the medium was changed once, at day 3, and supplemented with fresh APRIL. Mean ± SEM of three different assays is presented, performed in triplicates. (B) Addition of Fc-BCMA, a soluble form of the receptor binding APRIL, reverts the effect of APRIL (200 ng/ml) in both cell lines, whereas Fc-BCMA alone or nonspecific IgG had no effect. Mean ± SEM of three independent assays in triplicate. Incubation time was 6 d. (C) Transfection of cells with shBCMA RNA reverts the effect of APRIL (200 ng/ml) on cell growth, after 6 d of incubation. In contrast, addition of nontarget shRNA has no effect. Results (mean ± SEM of three experiments in quadruplicate) are normalized compared with control (non-APRIL or shRNA-treated) cells. (D) Effect of APRIL (200 ng/ml) on the cell cycle of HepG2 (upper row) and Hep3B cells (lower row). Cells were synchronized by serum starvation for 24 h and then treated with 200 ng/ml APRIL in full culture medium for 24 additional hours. Annexin/propidium iodine staining and FACS analysis were performed as described in 2Materials and Methods. Similar results were obtained in cells treated for 48 h with APRIL (not shown). This figure presents a typical example, as analyzed with the ModFit program, and the table below shows mean ± SEM of three repetitions. *p < 0.05, **p < 0.01.

Close modal

TRAF/NF-κB activation was reported to mediate the effects of APRIL on transcription, proliferation, survival, and differentiation (34). However, in our system, incubation with APRIL reveals no induction of NF-κB activity (Fig. 3A), in contrast to TNF-α (positive control). We have therefore investigated major intracellular signaling pathways (Akt, p38K, ERK, and JNK) as possible mediators of APRIL effects, as described previously (11, 34). Our data show that APRIL induces JNK and Akt phosphorylation, whereas it has no effect on ERK and p38 (Fig. 3B).

FIGURE 3.

BCMA signaling in HCC-derived cells. (A) Addition of 200 ng/ml APRIL in HepG2 or Hep3B cells, transfected with the pNFκB-Luc vector, carrying NF-κB response elements in front of the firefly luciferase gene, does not modify the transcriptional activity of NF-κB, after a 24-h incubation. In contrast, an induction is obtained with TNF-α, used as a positive control. Results (mean ± SEM of three different experiments, in five replicates each) are normalized per the Renilla reporter gene. (B) Western blot of different signaling mediators (JNK, Akt, p38 MAPK and ERK phosphorylation) during a time course of HepG2 (left panel) and Hep3B cells (right panel) treated with 200 ng/ml APRIL. The ligand induces JNK and Akt phosphorylation, whereas p38 MAPK and ERK remain unaltered. A typical blot is presented, repeated three times with similar results. (C) The PI3K/Akt pathway does not participate in the antiproliferative action of APRIL. Treatment of HepG2 and Hep3B cells with LY-294002 (LY, 40 μM), 30 min prior to the addition of APRIL (200 ng/ml, a 6-d incubation, with one change of the medium at day 3 and addition of fresh APRIL and LY), does not block APRIL-induced cell growth arrest. Results (mean ± SEM of three assays in triplicate) are expressed compared with control (nontreated) cells. *p < 0.05, **p < 0.01. Similar results, with the use of wortmannin as a PI3K/Akt inhibitor are shown in Supplemental Fig. 3A. (D) JNK2 is implicated in APRIL-induced growth arrest in HepG2 cells. Incubation of cells with shRNA against JNK2 reverts the antiproliferative effect of APRIL (200 ng/ml) after 6 d of incubation. Figure presents the mean ± SEM of three different experiments in triplicate.

FIGURE 3.

BCMA signaling in HCC-derived cells. (A) Addition of 200 ng/ml APRIL in HepG2 or Hep3B cells, transfected with the pNFκB-Luc vector, carrying NF-κB response elements in front of the firefly luciferase gene, does not modify the transcriptional activity of NF-κB, after a 24-h incubation. In contrast, an induction is obtained with TNF-α, used as a positive control. Results (mean ± SEM of three different experiments, in five replicates each) are normalized per the Renilla reporter gene. (B) Western blot of different signaling mediators (JNK, Akt, p38 MAPK and ERK phosphorylation) during a time course of HepG2 (left panel) and Hep3B cells (right panel) treated with 200 ng/ml APRIL. The ligand induces JNK and Akt phosphorylation, whereas p38 MAPK and ERK remain unaltered. A typical blot is presented, repeated three times with similar results. (C) The PI3K/Akt pathway does not participate in the antiproliferative action of APRIL. Treatment of HepG2 and Hep3B cells with LY-294002 (LY, 40 μM), 30 min prior to the addition of APRIL (200 ng/ml, a 6-d incubation, with one change of the medium at day 3 and addition of fresh APRIL and LY), does not block APRIL-induced cell growth arrest. Results (mean ± SEM of three assays in triplicate) are expressed compared with control (nontreated) cells. *p < 0.05, **p < 0.01. Similar results, with the use of wortmannin as a PI3K/Akt inhibitor are shown in Supplemental Fig. 3A. (D) JNK2 is implicated in APRIL-induced growth arrest in HepG2 cells. Incubation of cells with shRNA against JNK2 reverts the antiproliferative effect of APRIL (200 ng/ml) after 6 d of incubation. Figure presents the mean ± SEM of three different experiments in triplicate.

Close modal

We have verified that this effect was BCMA-dependent, because in anti-BCMA shRNA-transfected cells APRIL was ineffective in inducing phosphorylation of either Akt or JNK molecules (Supplemental Fig. 2). We then focused on the role of Akt and JNK as possible mediators of APRIL antiproliferative action. Incubation of cells with APRIL and the PI3K/Akt inhibitor LY-294002 (40 μM; Fig. 3C) or wortmannin (1 μM; Supplemental Fig. 3A) results in no significant modification of the ligand effect on cell growth, suggesting that neither PI3K nor Akt mediates BCMA-dependent inhibition of HepG2/Hep3B proliferation. In contrast, SP600125 (a pan-JNK inhibitor, 10 μM) (46), had a significant antiproliferative effect of its own (Supplemental Fig. 3B), but no further reduction of cellular proliferation by APRIL was observed.

In view of our finding of increased JNK phosphorylation by APRIL and considering the fact that JNK1 has been demonstrated to be only a positive regulator of proliferation of HCC cells (47), we hypothesized that JNK2 might be the molecule regulating the antiproliferative effect of APRIL. Such an effect has been reported previously in murine embryonic fibroblasts, 3T3 fibroblasts, and erythroblasts (48), in which Jnk2−/− cells were found to exhibit a proliferative advantage, compared with their wild-type counterparts. To test our hypothesis, we separately knocked down each JNK isoform, with the use of shRNAs against JNK1 and JNK2. JNK1 inhibition led to a massive reduction of cell proliferation, comparable to the effect of SP600125 (Supplemental Fig. 3C), whereas APRIL had no additional effect. In contrast, JNK2 inhibition significantly attenuated the antiproliferative effect of APRIL in HepG2 cells (Fig. 3D), suggesting that JNK2 is implicated in the BCMA-mediated antiproliferative effect of APRIL in HCC cells.

To further decipher the mechanisms underlying these findings, we performed a kinetic transcriptome analysis, after 2, 6, and 12 h of APRIL incubation in HepG2 cells (Fig. 4A), resulting in a time-dependent modification of up- and downregulated transcripts. Gene set enrichment analysis (39) of time-modified genes reveals one significantly inhibited pathway related to immune signaling and nine induced pathways related to the cell cycle and DNA replication (Fig. 4B). Leading edge analysis (39) (Fig. 4B) of the amplified pathways revealed a number of common transcripts implicated in all pathways, a result verified by qRT-PCR (Fig. 4C). They include the following: 1) Minichromosome maintenance (MCM) 2, 4–6, and CDC6. MCM proteins participate in DNA unfolding, recruitment of other proteins, and DNA doubling; their activity is decreased by their phosphorylation by CDC6 (49). These transcripts are decreased specifically after APRIL incubation. 2) PCNA, a cofactor of the DNA polymerase δ complex downstream of the MCM complex, as well as a cofactor of DNA polymerase ε upstream of the MCM complex (50), is modified in seven of nine processes. 3) RPA3 (modified in seven of nine processes) is involved in the S phase of the cell cycle (50). Finally, 4) DNA polymerase ε itself (POLE2) is involved in six of nine processes. The effect of APRIL on these genes could be blocked with the use of shRNA against BCMA (Fig. 4C). These elements, therefore, might represent the primary target of the transcriptional effect of APRIL on the genome via BCMA, explaining the effect of the ligand on the cell cycle.

FIGURE 4.

Transcriptional effects of APRIL. (A) APRIL (200 ng/ml) incubation of HepG2 cells induces a time-related increase of upregulated transcripts; downregulated transcripts remain almost constant after 6 h of incubation. (B) Heat plot of leading edge analysis (39) of genes involved in significantly modified pathways. This plot permits the identification of genes modified by APRIL, in the majority of identified pathways. (C) Common genes modified by APRIL in the pathways identified by gene set enrichment analysis were assayed by qRT-PCR after 2, 6, and 12 h of APRIL incubation (200 ng/ml) in the presence or absence of shRNA against BCMA and nontarget shRNA. Mean ± SEM of two independent experiments assayed in triplicate. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 4.

Transcriptional effects of APRIL. (A) APRIL (200 ng/ml) incubation of HepG2 cells induces a time-related increase of upregulated transcripts; downregulated transcripts remain almost constant after 6 h of incubation. (B) Heat plot of leading edge analysis (39) of genes involved in significantly modified pathways. This plot permits the identification of genes modified by APRIL, in the majority of identified pathways. (C) Common genes modified by APRIL in the pathways identified by gene set enrichment analysis were assayed by qRT-PCR after 2, 6, and 12 h of APRIL incubation (200 ng/ml) in the presence or absence of shRNA against BCMA and nontarget shRNA. Mean ± SEM of two independent experiments assayed in triplicate. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

An alternative analysis of transcriptome data permits the reverse in silico identification of transcription factors potentially modified by an agent (40). In this analysis, by applying the Web resource TFactS (http://www.tfacts.org), we identified only two transcription factors significantly modified by APRIL: FOXO3A (positive association, p = 0.0068) and E2F (negative association, p = 0.0019). Of interest, the regulatory/promoter regions of all identified genes (MCM 2, 4–6, CDC6, PCNA, RPA3, and POLE2) contain putative FOXO3 binding sites, as detected with the online resource multiTF (http://multitf.dcode.org).

To further validate this result, we transfected cells with constructs of FOXO3A or E2F response elements, cloned upstream of the luciferase gene. Unfortunately, both cell lines display very low E2F activity, not permitting analysis of further inhibition. In contrast, FOXO3A transcriptional activity is dose-dependently increased by the addition of APRIL and inhibited by shRNA against BCMA (Fig. 5A, Supplemental Fig. 3D).

FIGURE 5.

Validation of JNK implication in FOXO3A, Akt, and GADD45 activation by APRIL. (A) Effect of APRIL on FOXO3A activation. HepG2 (upper panel) and Hep3B (lower panel) cells were transfected with a construct in which FOXO3A interaction sites have been cloned upstream of the 5′ end of the luciferase gene. They were then incubated with 200 ng/ml APRIL, in the absence or presence of LY-294002 (LY, 40 μM) or shRNA against BCMA, for 24 h. Protein was collected and used for the luciferase assay. Figure presents graphs normalized per control (nontreated) cells. (B) Effect of JNK1 and JNK2 inhibition on FOXO3A activation in HepG2 and Hep3B cells. Only shRNA against JNK2 blocks APRIL-induced FOXO3A activation (*p < 0.05 compared with scrambled shRNA-transfected plus APRIL-treated cells). (C) Time course of JNK (left panel) or Akt (right panel) phosphorylation after incubation of HepG2 cells with 200 ng/ml APRIL, in the absence (squares) or presence (circles) of 10 μM SP600125. Quantitation of Western blot results is presented as mean ± SEM of three independent assays. Similar results were obtained with Hep3B cells (not shown). (D) GADD45 transcription after incubation of HepG2 cells with APRIL, 200 ng/ml, for 2, 6, and 12 h. GADD45 was assayed by qRT-PCR. Anti-BCMA shRNA inhibited APRIL-induced GADD45 expression. In all panels, the mean ± SEM of at least two assays performed in triplicate is presented.

FIGURE 5.

Validation of JNK implication in FOXO3A, Akt, and GADD45 activation by APRIL. (A) Effect of APRIL on FOXO3A activation. HepG2 (upper panel) and Hep3B (lower panel) cells were transfected with a construct in which FOXO3A interaction sites have been cloned upstream of the 5′ end of the luciferase gene. They were then incubated with 200 ng/ml APRIL, in the absence or presence of LY-294002 (LY, 40 μM) or shRNA against BCMA, for 24 h. Protein was collected and used for the luciferase assay. Figure presents graphs normalized per control (nontreated) cells. (B) Effect of JNK1 and JNK2 inhibition on FOXO3A activation in HepG2 and Hep3B cells. Only shRNA against JNK2 blocks APRIL-induced FOXO3A activation (*p < 0.05 compared with scrambled shRNA-transfected plus APRIL-treated cells). (C) Time course of JNK (left panel) or Akt (right panel) phosphorylation after incubation of HepG2 cells with 200 ng/ml APRIL, in the absence (squares) or presence (circles) of 10 μM SP600125. Quantitation of Western blot results is presented as mean ± SEM of three independent assays. Similar results were obtained with Hep3B cells (not shown). (D) GADD45 transcription after incubation of HepG2 cells with APRIL, 200 ng/ml, for 2, 6, and 12 h. GADD45 was assayed by qRT-PCR. Anti-BCMA shRNA inhibited APRIL-induced GADD45 expression. In all panels, the mean ± SEM of at least two assays performed in triplicate is presented.

Close modal

Previous reports have shown that Akt induces specific site phosphorylation of FOXO3, reducing its activity (51, 52), in contrast to an activating effect of JNK (52). As expected, addition of the JNK inhibitor SP600125 in APRIL-treated cells resulted in inhibiting APRIL-induced FOXO3 activation in HCC cells (Supplemental Fig. 3D), suggesting that FOXO3 activation is downstream of BCMA-induced JNK phosphorylation; in contrast, the PI3K inhibitor LY-294002 had no effect (Fig. 5A). Furthermore, FOXO3A activity was not modified by the transfection of cells with shRNA against JNK1, whereas shRNA against JNK2 reverted APRIL-induced transcriptional activity of FOXO3A (Fig. 5B). These data suggest that FOXO3A activation is a downstream effector of APRIL/BCMA-induced JNK2 phosphorylation.

A recent report indicates that JNK could directly phosphorylate Akt (53), whereas an interplay between Akt and JNK2 has been reported previously in neuronal cells (54). To study whether Akt activation relies on JNK phosphorylation, we assayed JNK and Akt phosphorylation, after APRIL stimulation, in the absence (control) or presence of the JNK inhibitor SP600125. Our results (Fig. 5C) suggest that inhibition of JNK by SP600125 results in a substantial inhibition of Akt. To further verify our results, we transfected cells with shRNA against JNK1 and JNK2 and studied Akt phosphorylation after APRIL stimulation (Fig. 6). shRNA against JNK2 (but not against JNK1) significantly reduced both APRIL-induced and basal Akt phosphorylation. Furthermore, JNK2 inhibition led to a more intense loss of the 54-kDa isoforms of JNK1/2, whereas JNK1 inhibition had a more prominent effect on the 45-kDa isoforms, in line with previous results in 3T3 fibroblasts (47).

FIGURE 6.

JNK2 mediates APRIL-induced phosphorylation of Akt. (A) Typical Western blots from HepG2 cells transfected with shRNA against JNK1 and JNK2 (48 h) and treated with APRIL (200 ng/ml) for 10 min. The experiment was performed three times with identical results. (B) Quantification of pAkt/total-Akt from Western blots performed in three different experiments. Only JNK2 inhibition could block APRIL/BCMA-induced Akt phosphorylation. (C and D) JNK1 knockdown led to preferential inhibition of the 45-kDa JNK1/2 isoforms, whereas JNK2 knockdown led to inhibition of the 54-kDa isoforms of these molecules, a phenomenon suggestive of a “predominant” isoform expression from these genes in HepG2 cells. Mean ± SEM of three independent experiments. *p < 0.05, **p < 0.01 denote significance compared with scrambled shRNA-transfected cells, treated or not with 200 ng/ml APRIL.

FIGURE 6.

JNK2 mediates APRIL-induced phosphorylation of Akt. (A) Typical Western blots from HepG2 cells transfected with shRNA against JNK1 and JNK2 (48 h) and treated with APRIL (200 ng/ml) for 10 min. The experiment was performed three times with identical results. (B) Quantification of pAkt/total-Akt from Western blots performed in three different experiments. Only JNK2 inhibition could block APRIL/BCMA-induced Akt phosphorylation. (C and D) JNK1 knockdown led to preferential inhibition of the 45-kDa JNK1/2 isoforms, whereas JNK2 knockdown led to inhibition of the 54-kDa isoforms of these molecules, a phenomenon suggestive of a “predominant” isoform expression from these genes in HepG2 cells. Mean ± SEM of three independent experiments. *p < 0.05, **p < 0.01 denote significance compared with scrambled shRNA-transfected cells, treated or not with 200 ng/ml APRIL.

Close modal

Finally, we report that APRIL enhanced GADD45 mRNA transcription in a time-dependent manner (Figs. 4C, 5D). The increase of GADD45 mRNA transcription induced by APRIL was almost abolished in cells transfected with anti-BCMA shRNA, relaying its transcriptional enhancement to APRIL/BCMA stimulation. GADD45 is a DNA damage-inducible gene, inducing G2 phase growth arrest via cdc2 inhibition (55). However, as reported above, we could identify only minor changes in cdc2 phosphorylation (∼20% reduction of the p-cdc2/cdc2 ratio in APRIL-treated cells). Interestingly enough, in a very recent study, JNK2 has also been reported to phosphorylate Bcl-xL, which binds and colocalizes with cdc2, leading to G2 cell cycle arrest (56), whereas GADD45 is also directly induced by FOXO3 expression, through JNK activation (57). GADD45 activation by APRIL binding to BCMA suggests that G2/M growth arrest might be regulated by one of these mechanisms.

In an attempt to identify whether the APRIL/BCMA system might have a potential implication in HCC, we assayed by immunocytochemistry the expression of APRIL, BAFF, and their receptors in 12 HCC cases and 7 normal livers (Fig. 7). In normal liver specimens, APRIL is detected only in hepatocytes; in contrast, BAFF stains homogeneously the hepatic parenchyma. TACI stains rare occasional hepatic parenchymal cells, whereas BCMA exhibits moderate intracellular hepatocyte immunoreactivity; BAFFR stained preferentially hepatic blood vessels. This profile is modified in HCC: Staining of both APRIL and BCMA shows remarkable enhancement, with all epithelial cells heavily stained. BCMA presents a discrete peripheral staining, suggestive of membrane localization. In contrast, BAFF staining is not modified, whereas BAFFR staining is reduced, concentrated in perivascular cells, and TACI staining is absent. It should be noted that the profile of APRIL/BAFF and their receptors in the HCC samples studied is comparable to that of HepG2 and Hep3B cells, presented in Fig. 1.

FIGURE 7.

Expression of APRIL and BCMA is present in normal liver and enhanced in HCC. APRIL and BCMA are expressed in normal liver and are enhanced in HCC. Typical cases are shown. TACI immunoreactivity is absent, whereas BAFF and BAFFR immunoreactivity is present both in normal liver and in HCC. Scale bar, 100 μm. APRIL, TACI, BAFF, and BAFFR were detected with Fast Red and BCMA with DAB (see Materials and Methods for details).

FIGURE 7.

Expression of APRIL and BCMA is present in normal liver and enhanced in HCC. APRIL and BCMA are expressed in normal liver and are enhanced in HCC. Typical cases are shown. TACI immunoreactivity is absent, whereas BAFF and BAFFR immunoreactivity is present both in normal liver and in HCC. Scale bar, 100 μm. APRIL, TACI, BAFF, and BAFFR were detected with Fast Red and BCMA with DAB (see Materials and Methods for details).

Close modal

APRIL, BAFF, and their receptors (BCMA, TACI, BAFFR) for a long time were considered to be specifically involved in B lymphocyte proliferation and differentiation (5861). Abnormal expression of APRIL and BAFF has been found to regulate malignant B lymphocyte proliferation, suggesting a role for these ligands in the growth of neoplastic cells (43, 44). However, many authors have reported the expression of APRIL and BAFF in an array of normal and neoplastic cell lines (1, 18, 2733) and tissues (2026, 36). The discovery of BCMA and TACI expression in normal tissues other than B lymphocytes (2024, 37) suggests that these two receptors may also have a function outside the immune system. In this report, we present evidence that APRIL and BCMA are expressed in normal liver tissue and massively induced in HCC specimens and derived cell lines, whereas BAFF and BAFFR are also expressed, but not modified, and TACI is absent. We used HepG2 and Hep3B cells as an in vitro model to assay the exclusive role of APRIL and BCMA. These cells represent a rare system in which BAFF (which is phenotypically ineffective) and APRIL (inducing cell growth arrest exclusively through BCMA) present divergent results, whereas low secretion of BAFF, expressing a 5- to 100-fold lower affinity for BCMA (1113), compared with that of APRIL, makes improbable its interaction with BCMA, in this model.

In most cases, the effects of ligands acting on BCMA are indistinguishable from those of TACI, which is usually equally present in the studied systems. In previous work, APRIL was reported to stimulate the proliferation of several nonimmune neoplastic cells (19, 26, 62). This effect was attributed to stimulation of the cell cycle, through cyclin D (63, 64), whereas different signaling pathways have been reported in engineered cells with an overexpression of the receptors involving ERK or JNK (34, 65), in addition to the canonical NF-κB activation (66, 67). As reported in this article, in nonengineered HCC cells, APRIL did not modify NF-κB activity; hence, we have assayed the main intracellular pathways, previously described to be induced upon BCMA activation. We report that p38 and ERK are not modified, whereas JNK and Akt were significantly activated by APRIL; however, PI3K inhibition did not reverse this effect. In contrast, JNK1 inhibition led to decreased proliferation, as it was previously reported (47); however, the APRIL antiproliferative effect is not mediated by JNK1. We therefore focused on JNK2, and we report that JNK2 mediates the decrease of cell growth induced by APRIL. Of interest, this differential effect of JNK1 and JNK2 has been reported previously in mouse embryonic and 3T3 fibroblasts and erythroblasts (48, 68). In a very elegant study, Sabapathy et al. (48) showed that JNK1 was proproliferative via c-Jun phosphorylation, whereas JNK2 was antiproliferative through negative regulation of c-jun expression/phosphorylation and ubiquitin binding to c-jun. Furthermore, in a tissue type- and cell type-specific manner, JNK2 has an important role in regulating several molecules related to cell cycle progression checkpoints, especially under DNA damage conditions, leading to cell cycle arrest (6972). Hepatocytes from Jnk2−/− mice, however, exhibit increased proliferation rates compared with their wild-type counterparts (73), a finding in line with our results in HCC cells. We further show that APRIL, through BCMA, induces Akt phosphorylation, in a JNK2-dependent manner, suggesting that JNKs may modify the activity of the PI3K pathway, a finding that has been only recently reported (74). Although previous reports came to this conclusion with the use of JNK inhibitors like SP600125, whose specificity has been questioned recently (46), our knockdown experiments clearly confirm this interplay between the two pathways, establishing Akt phosphorylation as a downstream effect of JNK2 activation.

Our observations support the pivotal role of the new pathway reported in this article (APRIL/BCMA/JNK2/FOXO3A/GADD45) in the negative regulation of HCC cellular proliferation. The FOXO family of transcription factors gained increased interest as pivotal elements of cell fate, implicated in diverse functions, from development, longevity and aging, to control of cell survival or apoptosis (75). Some members are directly implicated in cell cycle control, through G1-related p130 and cyclin G2, or GADD45 resulting in G2 cycle arrest (76), as equally shown in this study. Of interest, APRIL-regulated transcripts (MCM 2, 4–6, and CDC6), coding for proteins participating in DNA unfolding and doubling, have in their promoter region putative FOXO3A binding sites. GADD45 also participates in G2 cell cycle arrest, via cdc2 inhibition (77), and its expression has been reported previously to be JNK1- and JNK2-dependent (78). However, in the current study, we could identify only minor changes in cdc2 phosphorylation (∼20% reduction of the p-cdc2/cdc2 ratio in APRIL-treated cells). Interestingly enough, in a very recent work, JNK2 has also been reported to phosphorylate Bcl-xL, which binds and colocalizes with cdc2, leading to G2 cell cycle arrest (56).

Targeted therapy represents a significant advance in HCC management. Different targets are actually under investigation, including members of the TNF superfamily (reviewed in Ref. 79). Data reported in this article show that APRIL/BCMA might also have potential diagnostic or therapeutic significance. However, an apparent contradiction derives from our findings: We show that the ligand decreases cell growth, whereas it is massively expressed in a highly invasive tumor. Two hypotheses could explain this finding: 1) The space limits of the tumor may provoke a cellular signaling “switch,” providing a local APRIL/BCMA “brake” for further tumor expansion, a result previously reported for other cardinal molecules (80); and 2) in the tumor environment, APRIL, besides regulating the cell cycle, may also act as a proinflammatory regulator of hepatocytes, as we have recently reported in keratinocytes (37). Of interest, a recent report suggests that FOXO3, activated equally by JNK, is responsible for the maintenance of acute myeloid leukemia, initiating cells in their immature state, thereby promoting maintenance of the disease (81). If a similar mechanism is also active in HCC cells, it could equally explain the increased levels of FOXO3-promoting APRIL in HCC specimens. These novel elements of APRIL effects could reorient our understanding of APRIL in HCC tumor biology.

In conclusion, in this study we report a novel signaling pathway for APRIL/BCMA via JNK2 and FOXO3A that regulates HCC cellular proliferation. Furthermore, we report that interplay exists between the MAPK pathway and the PI3K pathway whose importance should be further studied. Finally, our data suggest that BCMA, BAFFR, and TACI may also play an important role in non–immune-related tissues and they can therefore be considered TNF superfamily receptors not unique to B cell-related functions, but with a much broader implication in human physiology and disease.

This work was supported in part by European Union Programs Regional Potential/Translational Potential Grant 285948 and Steroid Receptors in Non-Small Cell Lung Cancer European Union-FP7 Marie Curie Actions-Career Reintegration Grants 2012-2015 and PCIG-GA-2011-303723 (to V.P.).

The microarray data presented in this article have been submitted to the National Institutes of Health Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/) under accession number GSE29375.

The online version of this article contains supplemental material.

Abbreviations used in this article:

HCC

hepatocellular carcinoma

MCM

minichromosome maintenance

qRT-PCR

quantitative RT-PCR

shRNA

short hairpin RNA.

1
Hahne
M.
,
Kataoka
T.
,
Schröter
M.
,
Hofmann
K.
,
Irmler
M.
,
Bodmer
J. L.
,
Schneider
P.
,
Bornand
T.
,
Holler
N.
,
French
L. E.
, et al
.
1998
.
APRIL, a new ligand of the tumor necrosis factor family, stimulates tumor cell growth.
J. Exp. Med.
188
:
1185
1190
.
2
López-Fraga
M.
,
Fernández
R.
,
Albar
J. P.
,
Hahne
M.
.
2001
.
Biologically active APRIL is secreted following intracellular processing in the Golgi apparatus by furin convertase.
EMBO Rep.
2
:
945
951
.
3
Schneider
P.
,
MacKay
F.
,
Steiner
V.
,
Hofmann
K.
,
Bodmer
J. L.
,
Holler
N.
,
Ambrose
C.
,
Lawton
P.
,
Bixler
S.
,
Acha-Orbea
H.
, et al
.
1999
.
BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth.
J. Exp. Med.
189
:
1747
1756
.
4
Marsters
S. A.
,
Yan
M.
,
Pitti
R. M.
,
Haas
P. E.
,
Dixit
V. M.
,
Ashkenazi
A.
.
2000
.
Interaction of the TNF homologues BLyS and APRIL with the TNF receptor homologues BCMA and TACI.
Curr. Biol.
10
:
785
788
.
5
Wu
Y.
,
Bressette
D.
,
Carrell
J. A.
,
Kaufman
T.
,
Feng
P.
,
Taylor
K.
,
Gan
Y.
,
Cho
Y. H.
,
Garcia
A. D.
,
Gollatz
E.
, et al
.
2000
.
Tumor necrosis factor (TNF) receptor superfamily member TACI is a high affinity receptor for TNF family members APRIL and BLyS.
J. Biol. Chem.
275
:
35478
35485
.
6
Gross
J. A.
,
Johnston
J.
,
Mudri
S.
,
Enselman
R.
,
Dillon
S. R.
,
Madden
K.
,
Xu
W.
,
Parrish-Novak
J.
,
Foster
D.
,
Lofton-Day
C.
, et al
.
2000
.
TACI and BCMA are receptors for a TNF homologue implicated in B-cell autoimmune disease.
Nature
404
:
995
999
.
7
Thompson
J. S.
,
Schneider
P.
,
Kalled
S. L.
,
Wang
L.
,
Lefevre
E. A.
,
Cachero
T. G.
,
MacKay
F.
,
Bixler
S. A.
,
Zafari
M.
,
Liu
Z. Y.
, et al
.
2000
.
BAFF binds to the tumor necrosis factor receptor-like molecule B cell maturation antigen and is important for maintaining the peripheral B cell population.
J. Exp. Med.
192
:
129
135
.
8
Laâbi
Y.
,
Gras
M. P.
,
Carbonnel
F.
,
Brouet
J. C.
,
Berger
R.
,
Larsen
C. J.
,
Tsapis
A.
.
1992
.
A new gene, BCM, on chromosome 16 is fused to the interleukin 2 gene by a t(4;16)(q26;p13) translocation in a malignant T cell lymphoma.
EMBO J.
11
:
3897
3904
.
9
Madry
C.
,
Laabi
Y.
,
Callebaut
I.
,
Roussel
J.
,
Hatzoglou
A.
,
Le Coniat
M.
,
Mornon
J. P.
,
Berger
R.
,
Tsapis
A.
.
1998
.
The characterization of murine BCMA gene defines it as a new member of the tumor necrosis factor receptor superfamily.
Int. Immunol.
10
:
1693
1702
.
10
von Bülow
G. U.
,
Bram
R. J.
.
1997
.
NF-AT activation induced by a CAML-interacting member of the tumor necrosis factor receptor superfamily.
Science
278
:
138
141
.
11
Yu
G.
,
Boone
T.
,
Delaney
J.
,
Hawkins
N.
,
Kelley
M.
,
Ramakrishnan
M.
,
McCabe
S.
,
Qiu
W. R.
,
Kornuc
M.
,
Xia
X. Z.
, et al
.
2000
.
APRIL and TALL-I and receptors BCMA and TACI: system for regulating humoral immunity.
Nat. Immunol.
1
:
252
256
.
12
Kimberley
F. C.
,
van Bostelen
L.
,
Cameron
K.
,
Hardenberg
G.
,
Marquart
J. A.
,
Hahne
M.
,
Medema
J. P.
.
2009
.
The proteoglycan (heparan sulfate proteoglycan) binding domain of APRIL serves as a platform for ligand multimerization and cross-linking.
FASEB J.
23
:
1584
1595
.
13
Day
E. S.
,
Cachero
T. G.
,
Qian
F.
,
Sun
Y.
,
Wen
D.
,
Pelletier
M.
,
Hsu
Y. M.
,
Whitty
A.
.
2005
.
Selectivity of BAFF/BLyS and APRIL for binding to the TNF family receptors BAFFR/BR3 and BCMA.
Biochemistry
44
:
1919
1931
.
14
Thompson
J. S.
,
Bixler
S. A.
,
Qian
F.
,
Vora
K.
,
Scott
M. L.
,
Cachero
T. G.
,
Hession
C.
,
Schneider
P.
,
Sizing
I. D.
,
Mullen
C.
, et al
.
2001
.
BAFF-R, a newly identified TNF receptor that specifically interacts with BAFF.
Science
293
:
2108
2111
.
15
Yan
M.
,
Brady
J. R.
,
Chan
B.
,
Lee
W. P.
,
Hsu
B.
,
Harless
S.
,
Cancro
M.
,
Grewal
I. S.
,
Dixit
V. M.
.
2001
.
Identification of a novel receptor for B lymphocyte stimulator that is mutated in a mouse strain with severe B cell deficiency.
Curr. Biol.
11
:
1547
1552
.
16
Hendriks
J.
,
Planelles
L.
,
de Jong-Odding
J.
,
Hardenberg
G.
,
Pals
S. T.
,
Hahne
M.
,
Spaargaren
M.
,
Medema
J. P.
.
2005
.
Heparan sulfate proteoglycan binding promotes APRIL-induced tumor cell proliferation.
Cell Death Differ.
12
:
637
648
.
17
Ingold
K.
,
Zumsteg
A.
,
Tardivel
A.
,
Huard
B.
,
Steiner
Q. G.
,
Cachero
T. G.
,
Qiang
F.
,
Gorelik
L.
,
Kalled
S. L.
,
Acha-Orbea
H.
, et al
.
2005
.
Identification of proteoglycans as the APRIL-specific binding partners.
J. Exp. Med.
201
:
1375
1383
.
18
Deshayes
F.
,
Laprée
G.
,
Portier
A.
,
Richard
Y.
,
Pencalet
P.
,
Mahieu-Caputo
D.
,
Horellou
P.
,
Tsapis
A.
.
2004
.
Abnormal production of the TNF-homologue APRIL increases the proliferation of human malignant glioblastoma cell lines via a specific receptor.
Oncogene
23
:
3005
3012
.
19
Rennert
P.
,
Schneider
P.
,
Cachero
T. G.
,
Thompson
J.
,
Trabach
L.
,
Hertig
S.
,
Holler
N.
,
Qian
F.
,
Mullen
C.
,
Strauch
K.
, et al
.
2000
.
A soluble form of B cell maturation antigen, a receptor for the tumor necrosis factor family member APRIL, inhibits tumor cell growth.
J. Exp. Med.
192
:
1677
1684
.
20
Alexaki
V. I.
,
Notas
G.
,
Pelekanou
V.
,
Kampa
M.
,
Valkanou
M.
,
Theodoropoulos
P.
,
Stathopoulos
E. N.
,
Tsapis
A.
,
Castanas
E.
.
2009
.
Adipocytes as immune cells: differential expression of TWEAK, BAFF, and APRIL and their receptors (Fn14, BAFF-R, TACI, and BCMA) at different stages of normal and pathological adipose tissue development.
J. Immunol.
183
:
5948
5956
.
21
Pelekanou
V.
,
Kampa
M.
,
Kafousi
M.
,
Darivianaki
K.
,
Sanidas
E.
,
Tsiftsis
D. D.
,
Stathopoulos
E. N.
,
Tsapis
A.
,
Castanas
E.
.
2008
.
Expression of TNF-superfamily members BAFF and APRIL in breast cancer: immunohistochemical study in 52 invasive ductal breast carcinomas.
BMC Cancer
8
:
76
.
22
Sandberg
W. J.
,
Otterdal
K.
,
Gullestad
L.
,
Halvorsen
B.
,
Ragnarsson
A.
,
Frøland
S. S.
,
Damås
J. K.
,
Oie
E.
,
Aukrust
P.
,
Hansson
G. K.
,
Yndestad
A.
.
2009
.
The tumour necrosis factor superfamily ligand APRIL (TNFSF13) is released upon platelet activation and expressed in atherosclerosis.
Thromb. Haemost.
102
:
704
710
.
23
Langat
D. L.
,
Wheaton
D. A.
,
Platt
J. S.
,
Sifers
T.
,
Hunt
J. S.
.
2008
.
Signaling pathways for B cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL) in human placenta.
Am. J. Pathol.
172
:
1303
1311
.
24
Pelekanou
V.
,
Notas
G.
,
Theodoropoulou
K.
,
Kampa
M.
,
Takos
D.
,
Alexaki
V. I.
,
Radojicic
J.
,
Sofras
F.
,
Tsapis
A.
,
Stathopoulos
E. N.
,
Castanas
E.
.
2011
.
Detection of the TNFSF members BAFF, APRIL, TWEAK and their receptors in normal kidney and renal cell carcinomas.
Anal Cell Pathol (Amst)
34
:
49
60
.
25
Sun
B.
,
Wang
H.
,
Wang
X.
,
Huang
H.
,
Ding
W.
,
Jing
R.
,
Shi
G.
,
Zhu
L.
.
2009
.
A proliferation-inducing ligand: a new biomarker for non-small cell lung cancer.
Exp. Lung Res.
35
:
486
500
.
26
Petty
R. D.
,
Samuel
L. M.
,
Murray
G. I.
,
MacDonald
G.
,
O’Kelly
T.
,
Loudon
M.
,
Binnie
N.
,
Aly
E.
,
McKinlay
A.
,
Wang
W.
, et al
.
2009
.
APRIL is a novel clinical chemo-resistance biomarker in colorectal adenocarcinoma identified by gene expression profiling.
BMC Cancer
9
:
434
.
27
Shu
H. B.
,
Hu
W. H.
,
Johnson
H.
.
1999
.
TALL-1 is a novel member of the TNF family that is down-regulated by mitogens.
J. Leukoc. Biol.
65
:
680
683
.
28
Kelly
K.
,
Manos
E.
,
Jensen
G.
,
Nadauld
L.
,
Jones
D. A.
.
2000
.
APRIL/TRDL-1, a tumor necrosis factor-like ligand, stimulates cell death.
Cancer Res.
60
:
1021
1027
.
29
Roth
W.
,
Wagenknecht
B.
,
Klumpp
A.
,
Naumann
U.
,
Hahne
M.
,
Tschopp
J.
,
Weller
M.
.
2001
.
APRIL, a new member of the tumor necrosis factor family, modulates death ligand-induced apoptosis.
Cell Death Differ.
8
:
403
410
.
30
Litinskiy
M. B.
,
Nardelli
B.
,
Hilbert
D. M.
,
He
B.
,
Schaffer
A.
,
Casali
P.
,
Cerutti
A.
.
2002
.
DCs induce CD40-independent immunoglobulin class switching through BLyS and APRIL.
Nat. Immunol.
3
:
822
829
.
31
He
B.
,
Chadburn
A.
,
Jou
E.
,
Schattner
E. J.
,
Knowles
D. M.
,
Cerutti
A.
.
2004
.
Lymphoma B cells evade apoptosis through the TNF family members BAFF/BLyS and APRIL.
J. Immunol.
172
:
3268
3279
.
32
He
B.
,
Raab-Traub
N.
,
Casali
P.
,
Cerutti
A.
.
2003
.
EBV-encoded latent membrane protein 1 cooperates with BAFF/BLyS and APRIL to induce T cell-independent Ig heavy chain class switching.
J. Immunol.
171
:
5215
5224
.
33
Phillips
T. A.
,
Ni
J.
,
Hunt
J. S.
.
2003
.
Cell-specific expression of B lymphocyte (APRIL, BLyS)- and Th2 (CD30L/CD153)-promoting tumor necrosis factor superfamily ligands in human placentas.
J. Leukoc. Biol.
74
:
81
87
.
34
Hatzoglou
A.
,
Roussel
J.
,
Bourgeade
M. F.
,
Rogier
E.
,
Madry
C.
,
Inoue
J.
,
Devergne
O.
,
Tsapis
A.
.
2000
.
TNF receptor family member BCMA (B cell maturation) associates with TNF receptor-associated factor (TRAF) 1, TRAF2, and TRAF3 and activates NF-kappa B, elk-1, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase.
J. Immunol.
165
:
1322
1330
.
35
Laabi
Y.
,
Gras
M. P.
,
Brouet
J. C.
,
Berger
R.
,
Larsen
C. J.
,
Tsapis
A.
.
1994
.
The BCMA gene, preferentially expressed during B lymphoid maturation, is bidirectionally transcribed.
Nucleic Acids Res.
22
:
1147
1154
.
36
Sabour Alaoui
S.
,
Dessirier
V.
,
de Araujo
E.
,
Alexaki
V. I.
,
Pelekanou
V.
,
Lkhider
M.
,
Stathopoulos
E. N.
,
Castanas
E.
,
Bagot
M.
,
Bensussan
A.
,
Tsapis
A.
.
2012
.
TWEAK affects keratinocyte G2/M growth arrest and induces apoptosis through the translocation of the AIF protein to the nucleus.
PLoS ONE
7
:
e33609
.
37
Alexaki
V. I.
,
Pelekanou
V.
,
Notas
G.
,
Venihaki
M.
,
Kampa
M.
,
Dessirier
V.
,
Sabour-Alaoui
S.
,
Stathopoulos
E. N.
,
Tsapis
A.
,
Castanas
E.
.
2012
.
B-cell maturation antigen (BCMA) activation exerts specific proinflammatory effects in normal human keratinocytes and is preferentially expressed in inflammatory skin pathologies.
Endocrinology
153
:
739
749
.
38
Notas
G.
,
Nifli
A. P.
,
Kampa
M.
,
Pelekanou
V.
,
Alexaki
V. I.
,
Theodoropoulos
P.
,
Vercauteren
J.
,
Castanas
E.
.
2012
.
Quercetin accumulates in nuclear structures and triggers specific gene expression in epithelial cells.
J. Nutr. Biochem.
23
:
656
666
.
39
Subramanian
A.
,
Tamayo
P.
,
Mootha
V. K.
,
Mukherjee
S.
,
Ebert
B. L.
,
Gillette
M. A.
,
Paulovich
A.
,
Pomeroy
S. L.
,
Golub
T. R.
,
Lander
E. S.
,
Mesirov
J. P.
.
2005
.
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles.
Proc. Natl. Acad. Sci. USA
102
:
15545
15550
.
40
Essaghir
A.
,
Toffalini
F.
,
Knoops
L.
,
Kallin
A.
,
van Helden
J.
,
Demoulin
J. B.
.
2010
.
Transcription factor regulation can be accurately predicted from the presence of target gene signatures in microarray gene expression data.
Nucleic Acids Res.
38
:
e120
.
41
Zhou
D.
,
Lobo-Ruppert
S. M.
.
2001
.
Transcription of the Schizosaccharomyces pombe U2 gene in vivo and in vitro is directed by two essential promoter elements.
Nucleic Acids Res.
29
:
2003
2011
.
42
Kern
C.
,
Cornuel
J. F.
,
Billard
C.
,
Tang
R.
,
Rouillard
D.
,
Stenou
V.
,
Defrance
T.
,
Ajchenbaum-Cymbalista
F.
,
Simonin
P. Y.
,
Feldblum
S.
,
Kolb
J. P.
.
2004
.
Involvement of BAFF and APRIL in the resistance to apoptosis of B-CLL through an autocrine pathway.
Blood
103
:
679
688
.
43
Novak
A. J.
,
Bram
R. J.
,
Kay
N. E.
,
Jelinek
D. F.
.
2002
.
Aberrant expression of B-lymphocyte stimulator by B chronic lymphocytic leukemia cells: a mechanism for survival.
Blood
100
:
2973
2979
.
44
Novak
A. J.
,
Darce
J. R.
,
Arendt
B. K.
,
Harder
B.
,
Henderson
K.
,
Kindsvogel
W.
,
Gross
J. A.
,
Greipp
P. R.
,
Jelinek
D. F.
.
2004
.
Expression of BCMA, TACI, and BAFF-R in multiple myeloma: a mechanism for growth and survival.
Blood
103
:
689
694
.
45
Novak
A. J.
,
Grote
D. M.
,
Stenson
M.
,
Ziesmer
S. C.
,
Witzig
T. E.
,
Habermann
T. M.
,
Harder
B.
,
Ristow
K. M.
,
Bram
R. J.
,
Jelinek
D. F.
, et al
.
2004
.
Expression of BLyS and its receptors in B-cell non-Hodgkin lymphoma: correlation with disease activity and patient outcome.
Blood
104
:
2247
2253
.
46
Tanemura
S.
,
Momose
H.
,
Shimizu
N.
,
Kitagawa
D.
,
Seo
J.
,
Yamasaki
T.
,
Nakagawa
K.
,
Kajiho
H.
,
Penninger
J. M.
,
Katada
T.
,
Nishina
H.
.
2009
.
Blockage by SP600125 of Fcepsilon receptor-induced degranulation and cytokine gene expression in mast cells is mediated through inhibition of phosphatidylinositol 3-kinase signalling pathway.
J. Biochem.
145
:
345
354
.
47
Hui
L.
,
Zatloukal
K.
,
Scheuch
H.
,
Stepniak
E.
,
Wagner
E. F.
.
2008
.
Proliferation of human HCC cells and chemically induced mouse liver cancers requires JNK1-dependent p21 downregulation.
J. Clin. Invest.
118
:
3943
3953
.
48
Sabapathy
K.
,
Hochedlinger
K.
,
Nam
S. Y.
,
Bauer
A.
,
Karin
M.
,
Wagner
E. F.
.
2004
.
Distinct roles for JNK1 and JNK2 in regulating JNK activity and c-Jun-dependent cell proliferation.
Mol. Cell
15
:
713
725
.
49
Bell
S. P.
,
Dutta
A.
.
2002
.
DNA replication in eukaryotic cells.
Annu. Rev. Biochem.
71
:
333
374
.
50
Ulrich
H. D.
2009
.
Regulating post-translational modifications of the eukaryotic replication clamp PCNA.
DNA Repair (Amst.)
8
:
461
469
.
51
Essaghir
A.
,
Dif
N.
,
Marbehant
C. Y.
,
Coffer
P. J.
,
Demoulin
J. B.
.
2009
.
The transcription of FOXO genes is stimulated by FOXO3 and repressed by growth factors.
J. Biol. Chem.
284
:
10334
10342
.
52
Huang
H.
,
Tindall
D. J.
.
2007
.
Dynamic FoxO transcription factors.
J. Cell Sci.
120
:
2479
2487
.
53
Wei
L.
,
Liu
Y.
,
Kaneto
H.
,
Fanburg
B. L.
.
2010
.
JNK regulates serotonin-mediated proliferation and migration of pulmonary artery smooth muscle cells.
Am. J. Physiol. Lung Cell. Mol. Physiol.
298
:
L863
L869
.
54
Dávila
D.
,
Torres-Aleman
I.
.
2008
.
Neuronal death by oxidative stress involves activation of FOXO3 through a two-arm pathway that activates stress kinases and attenuates insulin-like growth factor I signaling.
Mol. Biol. Cell
19
:
2014
2025
.
55
Tran
H.
,
Brunet
A.
,
Grenier
J. M.
,
Datta
S. R.
,
Fornace
A. J.
 Jr.
,
DiStefano
P. S.
,
Chiang
L. W.
,
Greenberg
M. E.
.
2002
.
DNA repair pathway stimulated by the forkhead transcription factor FOXO3a through the Gadd45 protein.
Science
296
:
530
534
.
56
Wang
J.
,
Beauchemin
M.
,
Bertrand
R.
.
2012
.
Phospho-Bcl-x(L)(Ser62) plays a key role at DNA damage-induced G(2) checkpoint.
Cell Cycle
11
:
2159
2169
.
57
Yin
F.
,
Bruemmer
D.
,
Blaschke
F.
,
Hsueh
W. A.
,
Law
R. E.
,
Herle
A. J.
.
2004
.
Signaling pathways involved in induction of GADD45 gene expression and apoptosis by troglitazone in human MCF-7 breast carcinoma cells.
Oncogene
23
:
4614
4623
.
58
Benson
M. J.
,
Elgueta
R.
,
Noelle
R. J.
.
2008
.
B cell survival: an unexpected mechanism of lymphocyte vitality.
Immunol. Cell Biol.
86
:
485
486
.
59
Doreau
A.
,
Belot
A.
,
Bastid
J.
,
Riche
B.
,
Trescol-Biemont
M. C.
,
Ranchin
B.
,
Fabien
N.
,
Cochat
P.
,
Pouteil-Noble
C.
,
Trolliet
P.
, et al
.
2009
.
Interleukin 17 acts in synergy with B cell-activating factor to influence B cell biology and the pathophysiology of systemic lupus erythematosus.
Nat. Immunol.
10
:
778
785
.
60
Zhang
X.
,
Park
C. S.
,
Yoon
S. O.
,
Li
L.
,
Hsu
Y. M.
,
Ambrose
C.
,
Choi
Y. S.
.
2005
.
BAFF supports human B cell differentiation in the lymphoid follicles through distinct receptors.
Int. Immunol.
17
:
779
788
.
61
Zhang
J. X.
,
Ma
H. W.
,
Sang
M.
,
Hu
Y. S.
,
Liang
Z. N.
,
Ai
H. X.
,
Zhang
J.
,
Cui
X. W.
,
Zhang
S. Q.
.
2010
.
Molecular structure, expression, cell and tissue distribution, immune evolution and cell proliferation of the gene encoding bovine (Bos taurus) TNFSF13 (APRIL).
Dev. Comp. Immunol.
34
:
1199
1208
.
62
Okano
H.
,
Shiraki
K.
,
Yamanaka
Y.
,
Inoue
H.
,
Kawakita
T.
,
Saitou
Y.
,
Yamaguchi
Y.
,
Enokimura
N.
,
Ito
K.
,
Yamamoto
N.
, et al
.
2005
.
Functional expression of a proliferation-related ligand in hepatocellular carcinoma and its implications for neovascularization.
World J. Gastroenterol.
11
:
4650
4654
.
63
Gupta
M.
,
Dillon
S. R.
,
Ziesmer
S. C.
,
Feldman
A. L.
,
Witzig
T. E.
,
Ansell
S. M.
,
Cerhan
J. R.
,
Novak
A. J.
.
2009
.
A proliferation-inducing ligand mediates follicular lymphoma B-cell proliferation and cyclin D1 expression through phosphatidylinositol 3-kinase-regulated mammalian target of rapamycin activation.
Blood
113
:
5206
5216
.
64
Quinn
J.
,
Glassford
J.
,
Percy
L.
,
Munson
P.
,
Marafioti
T.
,
Rodriguez-Justo
M.
,
Yong
K.
.
2011
.
APRIL promotes cell-cycle progression in primary multiple myeloma cells: influence of D-type cyclin group and translocation status.
Blood
117
:
890
901
.
65
Dai
S.
,
Zheng
Y.
,
Chen
B.
,
Gao
M.
,
Zhang
Y.
,
Zhang
L.
,
Gong
W.
,
He
F.
.
2009
.
Two Gln187 mutants of human soluble APRIL inhibit proliferation of lung carcinoma A549 cells.
Acta Biochim. Pol.
56
:
703
710
.
66
Weber
G.
2007
.
Molecular mechanisms of cancer.
Springer
,
Dordrecht, The Netherlands
.
67
Dempsey
P. W.
,
Doyle
S. E.
,
He
J. Q.
,
Cheng
G.
.
2003
.
The signaling adaptors and pathways activated by TNF superfamily.
Cytokine Growth Factor Rev.
14
:
193
209
.
68
Tournier
C.
,
Hess
P.
,
Yang
D. D.
,
Xu
J.
,
Turner
T. K.
,
Nimnual
A.
,
Bar-Sagi
D.
,
Jones
S. N.
,
Flavell
R. A.
,
Davis
R. J.
.
2000
.
Requirement of JNK for stress-induced activation of the cytochrome c-mediated death pathway.
Science
288
:
870
874
.
69
Chen
P.
,
O’Neal
J. F.
,
Ebelt
N. D.
,
Cantrell
M. A.
,
Mitra
S.
,
Nasrazadani
A.
,
Vandenbroek
T. L.
,
Heasley
L. E.
,
Van Den Berg
C. L.
.
2010
.
Jnk2 effects on tumor development, genetic instability and replicative stress in an oncogene-driven mouse mammary tumor model.
PLoS ONE
5
:
e10443
.
70
Mayer
C.
,
Bierhoff
H.
,
Grummt
I.
.
2005
.
The nucleolus as a stress sensor: JNK2 inactivates the transcription factor TIF-IA and down-regulates rRNA synthesis.
Genes Dev.
19
:
933
941
.
71
Mayer
C.
,
Grummt
I.
.
2005
.
Cellular stress and nucleolar function.
Cell Cycle
4
:
1036
1038
.
72
Das
M.
,
Bouchey
D. M.
,
Moore
M. J.
,
Hopkins
D. C.
,
Nemenoff
R. A.
,
Stenmark
K. R.
.
2001
.
Hypoxia-induced proliferative response of vascular adventitial fibroblasts is dependent on g protein-mediated activation of mitogen-activated protein kinases.
J. Biol. Chem.
276
:
15631
15640
.
73
Sabapathy
K.
,
Wagner
E. F.
.
2004
.
JNK2: a negative regulator of cellular proliferation.
Cell Cycle
3
:
1520
1523
.
74
Wei
J.
,
Wang
W.
,
Chopra
I.
,
Li
H. F.
,
Dougherty
C. J.
,
Adi
J.
,
Adi
N.
,
Wang
H.
,
Webster
K. A.
.
2011
.
C-Jun N-terminal kinase (JNK-1) confers protection against brief but not extended ischemia during acute myocardial infarction.
J. Biol. Chem.
286
:
13995
14006
.
75
Greer
E. L.
,
Brunet
A.
.
2005
.
FOXO transcription factors at the interface between longevity and tumor suppression.
Oncogene
24
:
7410
7425
.
76
Carter
M. E.
,
Brunet
A.
.
2007
.
FOXO transcription factors.
Curr. Biol.
17
:
R113
R114
.
77
Stein
J. V.
,
López-Fraga
M.
,
Elustondo
F. A.
,
Carvalho-Pinto
C. E.
,
Rodríguez
D.
,
Gómez-Caro
R.
,
De Jong
J.
,
Martínez-A
C.
,
Medema
J. P.
,
Hahne
M.
.
2002
.
APRIL modulates B and T cell immunity.
J. Clin. Invest.
109
:
1587
1598
.
78
Zhang
D.
,
Song
L.
,
Li
J.
,
Wu
K.
,
Huang
C.
.
2006
.
Coordination of JNK1 and JNK2 is critical for GADD45alpha induction and its mediated cell apoptosis in arsenite responses.
J. Biol. Chem.
281
:
34113
34123
.
79
Villanueva
A.
,
Llovet
J. M.
.
2011
.
Targeted therapies for hepatocellular carcinoma.
Gastroenterology
140
:
1410
1426
.
80
Romagosa
C.
,
Simonetti
S.
,
Lopez-Vicente
L.
,
Mazo
A.
,
Lleonart
M. E.
,
Castellvi
J.
,
Ramon
Y. C. S.
.
2011
.
p16(Ink4a) overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors. Oncogene
30
:
2087
2097
.
81
Sykes
S. M.
,
Lane
S. W.
,
Bullinger
L.
,
Kalaitzidis
D.
,
Yusuf
R.
,
Saez
B.
,
Ferraro
F.
,
Mercier
F.
,
Singh
H.
,
Brumme
K. M.
, et al
.
2011
.
AKT/FOXO signaling enforces reversible differentiation blockade in myeloid leukemias.
Cell
146
:
697
708
.

The authors have no financial conflicts of interest.