Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
      • Neuroimmunology: To Sense and Protect
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • My alerts
  • Log in
  • Log out
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

ERK5 Activates NF-κB in Leukemic T Cells and Is Essential for Their Growth In Vivo

Johan Garaude, Seyma Cherni, Sandra Kaminski, Etienne Delepine, Christine Chable-Bessia, Monsef Benkirane, Joana Borges, Atanasio Pandiella, Miguel Angel Iñiguez, Manuel Fresno, Robert A. Hipskind and Martin Villalba
J Immunol December 1, 2006, 177 (11) 7607-7617; DOI: https://doi.org/10.4049/jimmunol.177.11.7607
Johan Garaude
*Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique-Unité Mixte de Recherche 5535, Montpellier, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Seyma Cherni
*Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique-Unité Mixte de Recherche 5535, Montpellier, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sandra Kaminski
*Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique-Unité Mixte de Recherche 5535, Montpellier, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Etienne Delepine
*Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique-Unité Mixte de Recherche 5535, Montpellier, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Christine Chable-Bessia
†Laboratoire de Virologie Moleculaire, Institut de Génétique Humaine, Centre National de la Recherche Scientifique Unité Propre de Recherche 1142, Montpellier, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Monsef Benkirane
†Laboratoire de Virologie Moleculaire, Institut de Génétique Humaine, Centre National de la Recherche Scientifique Unité Propre de Recherche 1142, Montpellier, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Joana Borges
§Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Cientificas-Universidad de Salamanca, Salamanca, Spain
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Atanasio Pandiella
§Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Cientificas-Universidad de Salamanca, Salamanca, Spain
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Miguel Angel Iñiguez
‡Centro de Biología Molecular, Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, Madrid, Spain; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Manuel Fresno
‡Centro de Biología Molecular, Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, Madrid, Spain; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Robert A. Hipskind
*Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique-Unité Mixte de Recherche 5535, Montpellier, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Martin Villalba
*Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique-Unité Mixte de Recherche 5535, Montpellier, France;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

MAPK cascades play a central role in the cellular response to the environment. The pathway involving the MAPK ERK5 mediates growth factor- and stress-induced intracellular signaling that controls proliferation or survival depending upon the cell context. In this study, we show that reducing ERK5 levels with a specific small hairpin RNA 5 (shERK5) reduced cell viability, sensitized cells to death receptor-induced apoptosis, and blocked the palliative effects of phorbol ester in anti-Fas Ab-treated cells. shERK5 decreased nuclear accumulation of the NF-κB p65 subunit, and conversely, ectopic activation of ERK5 led to constitutive nuclear localization of p65 and increased its ability to trans activate specific reporter genes. Finally, the T lymphoma cell line EL-4, upon expression of shERK5, proliferated in vitro, but failed to induce s.c. tumors in mice. Our results suggest that ERK5 is essential for survival of leukemic T cells in vivo, and thus represents a promising target for therapeutic intervention in this type of malignancy.

Mitogen-activated protein kinase pathways play a central role in the signaling network that turns extracellular stimuli into distinct intracellular responses. Each MAPK cascade contains a core of sequentially activated kinases that are evolutionarily conserved in eukaryotes. Four major MAPK pathways have been identified in mammalian cells. The three canonical MAPK pathways, namely the ERK1/2, JNK, and p38 cascades, mediate proliferative and stress signaling in normal and leukemic T cells. In contrast, the role of the ERK5 cascade, which is present in normal and primary T cells (1), is unknown. The core of this pathway involves the kinases MEK kinase kinase 2 or MEK kinase kinase 3-activating MEK5, which in turn specifically activates ERK5 (2). ERK5 shares the TEY activation motif with other ERKs, whereas its other structural features are unique, such as the large regulatory C terminus that controls its nucleocytoplasmic shuttling (3). Diverse extracellular stimuli, including epidermal growth factor, serum, oxidative stress, Ag-receptor engagement, cytokines, and hyperosmolarity, induce activation of the MEK5-ERK5 module (1, 4, 5).

ERK5 mediates proliferative signaling by ErbB (6, 7), Ras (8), serum (9), insulin-like growth factor (IGF)3II (10), Bcr-Abl (11), and IL-6 (12). Accordingly, different groups have described a role for the ERK5 cascade in transformed cells: it plays a significant role in proliferation of breast cancer cells (6, 9); its overexpression is linked to bony metastases and a poor prognosis in human prostate cancer (13); it is necessary for chemoresistance in breast cancer cells (14); and it mediates cell survival in human lung cancer cells (15). More germane to our studies, ERK5 is expressed in myeloma cells, where its inhibition blocks proliferation and facilitates apoptosis induced by dexamethasone (12). Finally, ERK5 expression is essential for survival of leukemic cells expressing Bcr/Abl (11).

In vivo, invalidation of the ERK5 or MEK5 genes affects both embryonic angiogenesis and cardiac development and leads to embryonic lethality by day 11.5 (16, 17, 18). The important function of ERK5 in endothelial cells is confirmed by targeted deletion of ERK5 in adult mice, which perturbs vascular integrity and leads to death through endothelial failure (19). In cardiac tissue, activation of ERK5 inhibits injury after myocardial ischemia and reperfusion (20). ERK5 is also required for tumor-associated neoangiogenesis. In ERK5flox/flox mice carrying the Mx1-Cre transgene, deletion of the host ERK5 gene reduces the volume of tumor xenografts and leads to a significant decrease in vascular density (21). These data suggest that ERK5 plays a central role in several processes linked to tumorigenesis: transduction of intracellular oncogenic signals, tumor-associated angiogenesis, and in some cases invasion/metastasis.

Few direct targets of the ERK5 cascade have been identified. Like other MAPKs, specific docking domains mediate interations between ERK5 and its partners (22). ERK5 activates the myocyte enhancer factor (MEF)2 family of transcription factors, leading to c-Jun expression. Interestingly, ERK5 is not redundant with the ERK1/2 cascade, because the two pathways cooperate to induce a NF-κB reporter gene in NIH3T3 cells, whereas ERK5 itself had a minor effect (23). NF-κB transcription factors appeared as important players in the balance between cell death and cell survival, and constitutive NF-κB activity has been observed in many cancer cells (24).

In this study, we show that the oncogenic role of ERK5 in leukemic T cells depends on its ability to activate NF-κB through retention of p65 in the nucleus, allowing leukemic T cells to resist apoptotic stimuli in vitro and in vivo.

Materials and Methods

Abs and reagents

The anti-human CD3, anti-mouse CD3, and anti-mouse CD28 mAbs were purified, as described previously (25, 26). The anti-CD28, CD69, and anti-protein kinase C (PKC)θ mAbs were from BD Biosciences. The rabbit polyclonal Abs against ERK5 were from Cell Signaling Technology or have been described (6). The p65 and p50 rabbit polyclonal Abs were from Santa Cruz Biotechnology. The rat monoclonal anti-hemagglutinin (HA) (3F10) was from Roche. Donkey anti-rabbit or sheep anti-mouse IgG Abs were obtained from Amersham, and the goat anti-hamster IgG was from Pierce Biotechnology. Annexin V-PE was from BD Pharmingen. The anti-Fas Ab was from Upstate Biotechnology.

Plasmids

The expression vectors for ERK5 and ERK5 mutant (T218/Y220F) resistant to MEK5 activation (ERK5 AEF), constitutively active MEK1 (MEK1SS3) and MEK5 (S313D/T317D, termed MEK5DD), the IκB kinase (IKK) superrepressor (27), along with the c-jun and the TRE reporter (9), have been described. The expression vectors for ERK2, β-galactosidase, AP-1 and NF-AT reporters, and IL-2 promoter have been previously used (28, 29). The conventional NF-κB reporter is based on a NF-κB binding site in the Igκ L chain promoter (30). pRc-hcRel consists of pRcCMV with c-Rel cDNA inserted in the HindIII-XbaI site (31). The 3× κB thymidine kinase luc reporter plasmid contains a trimer of the NF-κB-binding motif of the H-2k gene upstream of the thymidine kinase minimal promoter and the luciferase reporter gene (32). The pcDNA3p65 and pcDNA3p50 expression plasmids were provided by J. Alcamí (Hospital 12 de Octubre, Madrid, Spain). The GAL4-p65 and 5×Gal4 Luc plasmids were provided by L. Schmitz (Genzentrum Ludwig-Maximilians-Universität, Munich, Germany) (33). Primers containing 19 bases of ERK5 coding sequence (AGCTGCCCTGCTCAAGTCT) or a corresponding scrambled sequence were ligated into pRetroSUPER vector to generate the small hairpin RNA ERK5 (shERK5) or control expression plasmid (11). The same primers were subcloned into pSIREN-RetroQ-puro (BD Biosciences) and used to create the EL-4 stable cell lines.

Cell culture and transfection

The cell lines EL-4, Jurkat T, and Jurkat T stably expressing IκB-DN (dominant negative) (34) were grown in RPMI 1640 (Invitrogen Life Technologies) supplemented with 10% FBS, 2 mM glutamine, 1 mM sodium pyruvate, 10 mM HEPES, 1× MEM nonessential amino acid solution (Invitrogen Life Technologies), 50 μM 2-ME, and 100 U/ml each of penicillin G and streptomycin. Jurkat cells in logarithmic growth phase were transfected with the indicated amounts of plasmid by electroporation, as described (26, 28). In each experiment, cells were transfected with the same total amount of DNA by supplementing expression vector DNA with empty vector. Cells were incubated for 10 min at room temperature with the DNA mix and electroporated at 260 mV, 960 μF in 400 μl of RPMI 1640. GFP was detected by excitation with 488 Blue Laser and emission FL1 530/30 by FACS.

T cell activation

Plates were coated with 5 μg/ml of each anti-human or anti-mouse CD3 and CD28 Abs in 50 mM Tris (pH 9), left 1 h at 37°C, and at least 4 h at 4°C. After three washes with PBS, Jurkat T cells were incubated on these plates at the indicated times. Plates were placed on ice, and directly lysed with 1× SDS-PAGE sample buffer. PMA, ionomycin, etoposide, taxol, cycloheximide (CHX), and TNF-α were added to the cell suspension for the indicated times. Cells were placed on ice, washed once with cold PBS, and processed.

Immunoprecipitation of HA-tagged proteins

A total of 2 × 107 transfected Jurkat Tag cells was washed twice with cold PBS, lysed in 300 μl of immunoprecipitation buffer (0.5% Nonidet P-40, 50 mM Tris-HCl (pH 8), 5 mM MgCl2, 0.5 mM EDTA, 1 mM DTT, and 10% glycerol), and incubated on ice for 10 min. The lysate was centrifuged at 14,000 rpm for 10 min at 4°C. The supernatant was precleared by incubation 30 min at 4°C with protein G agarose beads and incubated 90 min at room temperature with 1 μg of anti-HA (3F10) Ab. Twenty microliters of a 50% slurry of protein G agarose beads was added to the reaction and incubated for additional 30 min. Beads were washed three times with immunoprecipitation buffer and resuspended in 30 μl of SDS-PAGE sample buffer, denatured, separated by SDS-PAGE, and immunoblotted, as described below.

Reporter assays

In all experiments, cells were transfected with a β-galactosidase reporter plasmid (28). Transfected cells (1 × 106) were harvested after 2 days and washed twice with PBS. Cells were lysed in 100 μl of luciferase lysis buffer (Promega), and luciferase assays (40 μl) were performed, according to the manufacturer’s instructions (Promega), using a Berthold luminometer (9). For β-galactosidase assays, 40 μl of lysates was added to 200 μl of β-galactosidase assay buffer (50 mM phosphate buffer (pH 7.4), 200 μg of O-nitrophenyl-β-galactopyranoside, 1 mM MgCl2, and 50 mM 2-ME), and the absorbance was measured at 400 nm. The results are expressed as luciferase units normalized to the corresponding β-galactosidase activity. The expression level of the transfected proteins was routinely control by immunoblot analysis.

IL-2 measurement

Cells were cultured at 1 × 106 cells/ml in RPMI 1640 and stimulated as indicated with anti-CD3 Abs or PMA plus ionomycin. Supernatants were harvested, and IL-2 was measured using detection kit (BD Pharmingen). The cytokines’ values were expressed as picograms per milliliter.

Immunoblotting

Cells were washed with PBS and lysed in SDS-PAGE sample buffer. Proteins were separated by SDS-PAGE on minigels and processed for immunoblot analysis, as described previously (28).

Cell cycle

Cells were washed and resuspended in 900 μl of propidium iodide staining solution (50 μg/ml propidium iodide, 0.1% trisodium citrate dihydrate, 0.1 mg/ml RNase A, and 0.1% Triton X-100). After an overnight incubation, the cell cycle profile was determined with a FACSCalibur flow cytometer (BD Biosciences).

Subcellular fractionation

For preparation of nuclear extracts, cells were washed twice in cold PBS, resuspended in 400 μl of 10 mM HEPES (pH 7.4), 10 mM KCl, 2 mM MgCl2, 0.1 mM EDTA, and protease inhibitors (Roche), and incubated on ice for 10 min. Nonidet P-40 was added to the cells to a final concentration of 0.5%, followed by mixing and centrifugation at 1,000 × g for 20 s at 4°C to collect nuclei. Nuclear extracts were prepared by resuspending the pellets in 50 μl of 50 mM HEPES (pH 7.8), 50 mM KCl, 300 mM NaCl, 0.1 mM EDTA, 10% glycerol, and protease inhibitors (Roche). Proteins were extracted by agitation for 20 min at 4°C, the insoluble fraction was removed by centrifugation at 13,000 × g for 10 min, and the soluble proteins were analyzed by immunoblotting.

Generation of stable cell lines

A total of 5 × 106 EL-4 cells was cultured at 1.5 × 106 cells/ml. One milliliter of supernatant from 293T cells expressing the retroviral vector pSIREN for shERK5 or control plasmid was added 1 h after plating. The same protocol was repeated 1 day later. Three days later, cells were cultured with 2.5 μg/ml puromycin (Sigma-Aldrich). After 1 wk of selection, surviving cells were isolated and kept on selection medium until used.

Cell survival and apoptosis assays

Jurkat Tag cells were transfected with pSUPER-GFP or pSUPER-GFP-siERK5. Two days later, GFP+ cells were sorted and left in culture to recover for 1 additional day. These cells or EL-4 cells were treated with the indicated apoptotic agents. Where appropriate, PMA (100 ng/ml) was added to one-half of the cells 20 min before anti-Fas treatment. Cell survival was analyzed by forward/side scatter (FSC/SSC) or by annexin V staining, following the manufacturer’s instructions (BD Pharmingen), as previously described (35).

CD69 and CD25 expression

Jurkat Tag cells were transfected with pSUPER-GFP or pSUPER-GFP-siERK5. Two days later, GFP+ cells were sorted and left in culture to recover for 1 additional day. CD69 expression was measured essentially as previously described (29). Briefly, cells were transfected with the indicated vectors and harvested 2 days later. GFP+ cells were purified by FACS, washed in RPMI 1640, and suspended in 500 μl of RPMI 1640 plus 20 μl of PE-conjugated anti-human CD69 mAb for 15 min, as suggested by the manufacturer, washed twice on RPMI 1640, and analyzed by flow cytometry (FACScan; BD Biosciences) after gating for GFP-positive cells.

Tumor progression in vivo

EL-4 cells were washed in PBS and resuspended to 1.5 × 106 cells/ml. A total of 2.5 × 105 cells was injected s.c. in the flank of C57B6 mice. Tumor development was analyzed every 2 days. Tumors were measured, and the volume was calculated following the formula: V = length × (weight)2/2. All the experiments involving animals were performed according to the guidelines and regulations of the Centre Nationale de la Recherche Scientifique. J. Garaude, S. Cherni, R. Hipskind, and M. Villalba have the official degree for animal experimentation delivered by the Ministere de la Recherche (France).

Statistical analysis

The statistical analysis of the difference between means of paired samples was performed using the paired t test. The results are given as the confidence interval (p).

Results

Inhibition of ERK5 expression induces apoptosis in Jurkat cells

The ERK5 cascade has been linked to several different types of cancer. To examine its role in leukemogenesis, we reduced ERK5 levels in leukemic Jurkat T cells by electroporation of a bicistronic vector that expresses shERK5 (11) and GFP. As a control, we used a scrambled version of this sequence. Two days after transfection, >50% of the living cells expressed GFP (Fig. 1⇓A, first lane). These cells were purified by FACS and placed in culture for 1 day. At this point, >90% of the cells were GFP+ (Fig. 1⇓A, second lane), and the expression of ERK5 was largely reduced, unlike that of ERK2 (Fig. 1⇓B). At later times, the percentage of GFP+ Jurkat cells transfected wtih shERK5 was substantially lower than in control shRNA-transfected cells (13.6 vs 42.7% at 4 days, and 7.2 vs 23.7% at 6 days for Jurkat-shERK5 or Jurkat-control-shRNA cells, respectively). These data suggested that expression of shERK5 inhibited cell proliferation, induced cell death, or both. In the next set of experiments, we used cells 1 day after cell sorting, i.e., 3 days after transfection.

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Inhibition of ERK5-induced apoptosis in Jurkat cells. A, Twenty million Jurkat cells were transfected with 10 μg of pSUPER-GFP containing a shERK5 sequence or a control sequence. Two days after transfection, cells were analyzed for GFP expression (first lane) and purified by cell sorting. These purified cells were analyzed for GFP expression at different days. GFP was detected using flow cytometry by excitation with a 488-nm Blue Laser and by emission FL1 530/30. B, An aliquot of purified GFP+ cells (1 day after purification) was subjected to Western blotting to analyze the expression of ERK5 and ERK2. C, 5 × 105 GFP+ Jurkat cells were stimulated with PMA (100 ng/ml) 20 min before Fas-Ab (5 ng/ml CH11) and harvested 24 h later. The FSC and SSC profiles were analyzed by flow cytometry. Average percentages of alive cells from three independent experiments are presented with SD in the right-side graphic. D, Jurkat cells treated and stimulated as in C were stained with annexin V-PE, and the positive cells were analyzed by flow cytometry for increase in fluorescence intensity (FL-2 height). Average percentages of apoptotic cells from three independent experiments are presented with SD in the right-side graphic. E, Jurkat T cells were treated with PMA (100 ng/ml) for 20 h, and cell survival and apoptosis were analyzed as in C and D.

We tested the effect of shERK5 on various parameters of cell viability. First, we examined cell morphology by flow cytometric analysis of cell size (FSC) and granularity (SSC). Dying, or apoptotic, cells are smaller and more granular than viable cells. Dot blots from one experiment are shown in Fig. 1⇑C, and the average of three independent experiments is presented in the graphic. In the absence of stimulation, shERK5-transfected cells showed a lower rate of survival (76%) compared with control-transfected cells (89%). In early stages of apoptosis, different changes occur at the cell membrane. One involves the translocation of phosphatidylserine to the outer side of the plasma membrane, which can be detected with Annexin V. In agreement with the results of Fig. 1⇑C, shERK5-transfected cells showed increased staining by Annexin V and therefore a higher percentage of apoptotic cells (17%) than control-transfected cells (5%). This suggests that ERK5 played a role in the survival of leukemic T cells and protected them from spontaneous apoptosis.

A classical mechanism to induce apoptosis in leukemic T cell is the engagement of death receptors, of which Fas is well characterized (36). shERK5-transfected cells were more sensitive to anti-Fas treatment, developing characteristic features of apoptosis like higher granularity and Annexin V staining (Fig. 1⇑, C and D). Therefore, blocking ERK5 expression facilitated Fas-induced apoptosis.

Treatment with the phorbol ester PMA blocks Fas-induced apoptosis in different cell types. In T cells, activation of PKCθ by PMA inhibits anti-Fas-induced cell death (35). Interestingly, shERK5-treated cells were less effectively protected from this type of apoptosis by PMA than control cells (Fig. 1⇑, C and D), suggesting that the intracellular pathways involved in protecting leukemic T cells were also impaired in shERK5-treated cells. In fact, PMA alone induced apoptosis in a small, but consistent number of shERK5-transfected cells (Fig. 1⇑E).

ERK5 knockdown renders leukemic Jurkat T cells more sensitive to TNF-α

The transcription factor NF-κB plays an antiapoptotic role in different cell lines and mediates, in part, the protective effect of phorbol esters on Fas-induced apoptosis (34, 37). Interestingly, PMA becomes proapoptotic if NF-κB activation is blocked in Jurkat T cells (34), suggesting that blocking ERK5 activity would have a similar proapoptotic effect in leukemic cells as blocking NF-κB activation. TNF-α only induces apoptosis in Jurkat cells that fail to activate NF-κB (38). Moreover, p65−/− mice die from the increased sensitivity of hepatocytes to TNF-α, an effect that is reversed by inactivation of the type I TNF receptor (39). This protective effect of NF-κB requires gene expression, because compounds that block transcription or translation sensitize cells to TNF-α-induced apoptosis. Therefore, we investigated whether shERK5 sensitized Jurkat T cells to TNF-α. Because shERK5 induced apoptosis per se, we calculated the specific apoptosis induced by the different treatments by subtracting the percentage of apoptotic cells under resting conditions from that observed after treatment. In Jurkat cells transfected with the control scrambled shRNA, TNF-α or the protein synthesis inhibitor CHX induced apoptosis in 10% of the cells (Fig. 2⇓A), whereas the two together induced >60% apoptosis. In contrast, TNF-α alone induced apoptosis in >50% of the shERK5-transfected cells. These data suggested that ERK5 was implicated in NF-κB activation.

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

ERK5 expression is essential for NF-κB-induced survival. A, shERK5 sensitizes Jurkat cells to TNF-α. A half million GFP+ Jurkat cells were transfected as in Fig. 1⇑, sorted at day 2, and treated the next day with TNF-α (100 ng/ml), CHX (2 μg/ml), or a combination of both. Twenty-four hours later, cells were stained with annexin V-PE and analyzed by flow cytometry for increase in fluorescence intensity (FL-2 height). Mean percentages ± SEM of specific apoptosis from three independent experiments are presented in the right-side graphic, Student’s t test: ∗, p < 0.01; compare with control cells. The specific apoptosis was calculated by subtracting the percentage of apoptosis under resting conditions from the percentage of apoptosis after treatment. These values were 6 and 17% for control or shERK5 cells, respectively. B, Inhibition of ERK5 expression does not cooperate with taxol or etoposide. A total of 5 × 105 GFP+ Jurkat cells was treated for 24 h with anti-Fas (5 ng/ml), etoposide (0.5 μM), or taxol (0.3 μM), and analyzed as in A.

Interestingly, CHX alone induced a similar rate of specific apoptosis in control and shERK5 cells. Because CHX blocks protein expression, we suspected that ERK5-induced NF-κB activation did not play a major role in CHX-induced apoptosis. Therefore, we tested two anticancer drugs whose proapoptotic effects were largely independent of NF-κB activation: etoposide, an antineoplastic agent that targets the DNA-unwinding enzyme topoisomerase II (40, 41), and taxol, which targets microtubules and causes mitotic arrest leading to apoptosis (42). Consistent with the idea that shERK5 targets NF-κB activation, shERK5 did not sensitize Jurkat cells to apoptosis induced by these two compounds (Fig. 2⇑B). In contrast, it sensitizes Jurkat cells to Fas engagement, which cooperates with NF-κB inhibition to induce apoptosis (Figs. 1⇑ and 2⇑B) (43, 44, 45). Importantly, this effect was also observed in the mouse leukemic EL-4 T cell line (see Fig. 8⇓).

ERK5 knockdown does not affect cell cycle progression or activation of leukemic T cells

The role of ERK5 in tumorigenesis could also be linked to cell proliferation. shERK5 expression in leukemic cells did not alter the cell cycle (Fig. 3⇓A). Activation of T cells with PMA/ionomycin induced cell cycle arrest at GO/G1, which was unaffected by ERK5 knockdown. Activation-induced IL-2 production was not inhibited in shERK5-expressing cells (Fig. 3⇓B); similarly, these cells showed no changes in other activation markers like IL-2Rα (CD25) or CD69 (Fig. 3⇓C). These results suggested that ERK5 did not play a central role in T cell activation and excluded that defects in IL-2 production or in the up-regulation of its receptor could underlie the decrease in survival of shERK5-transfected cells.

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

The ERK5 pathway is not necessary for Jurkat T cell activation and proliferation. A, 106 Jurkat cells transfected as in Fig. 1⇑ were activated with PMA (100 ng/ml) plus ionomycin (1 μg/ml) during 24 h, and the cell cycle was analyzed by propidium iodide staining and FACS analysis. B, The level of IL-2 expression in cell treated with CD3 (plate-bound), PMA (100 ng/ml), or ionomycin (1 μg/ml) during 24 h was analyzed by ELISA. C, Jurkat cells were treated as in A, and the levels of expression of CD69 and CD25 were analyzed by FACS. The percentage of positive cells for each marker is depicted in the graphic. The percentage of cells reacting with the control IgG (data not shown) was almost identical with that labeled by CD25 in resting cells. The experiments were repeated three times with similar results.

Therefore, ERK5 plays a significant role in leukemic T cell survival, but not in leukemic T cell activation or proliferation, suggesting that shERK5 did not block all T cell functions. Nevertheless, we cannot formally exclude that a low residual level of ERK5 is sufficient to mediate T cell activation and proliferation, but not T cell survival.

The ERK5 pathway activates NF-κB, but not AP-1 or NF-AT, in leukemic T cells

To test the possibility that the ERK5 pathway activates NF-κB in Jurkat cells, we used an ERK5 expression vector together with one encoding a constitutively active mutant of the upstream kinase MEK5 (MEK5DD) that heavily phosphorylates and activates ERK5 in Jurkat cells (Fig. 4⇓A) (1). Neither vector alone activated a NF-κB reporter gene in Jurkat cells. However, the combination of MEK5DD and ERK5 induced NF-κB trans activation in resting, PMA-, or PMA/ionomycin-stimulated cells (Fig. 4⇓B). Similar results were obtained with the mouse leukemic T cell line EL-4, although the induction was lower (data not shown).

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

The ERK5 module targets NF-κB, but not AP-1 or NF-AT, in Jurkat cells. B and C, 107 Jurkat-TAg cells were cotransfected with 3 μg of each ERK5 and/or MEK5DD, 2 μg of the NF-κB or TRE luciferase reporters, and 1 μg of β-galactosidase expression vector. Control cells represent cells transfected with equal amounts of an empty vector. Two days later, equal numbers of cells were left unstimulated or stimulated for 4 h with PMA (100 ng/ml) or PMA plus ionomycin (1 μg/ml), and luciferase and β-galactosidase activities were evaluated. A, 106 cells were analyzed by immunoblotting after anti-HA immunoprecipitation. The data represent means ± SD, n = 8, Student’s t test: ∗∗, p < 0.0005; compare with control cells.

Next, we examined whether ERK5 activated other transcription factors in Jurkat T cells. In fact, we have shown previously that selective activation of the ERK5 pathway targets the c-jun promoter in leukemic T cells (1). AP-1 is composed of heterodimers of the Fos and Jun families of transcription factors. We cotransfected ERK5 and/or MEK5DD and investigated the effect of these plasmids on a PMA response element (TRE)-luciferase reporter gene (46) that is driven by four canonical AP-1 binding sites (Fig. 4⇑C). The expression of either kinase alone showed no effect in resting or stimulated cells. Cotransfection of both kinases induced a 2-fold increase in resting cells, but had no significant effect in stimulated cells. Similar observations were obtained in Jurkat cells using a reporter driven by the collagenase promoter (47) that contains a single AP-1 response element (data not shown). ERK5 also failed to activate a NF-AT/AP-1 reporter derived from the IL-2 promoter (data not shown). These results show that ERK5 plays at best a minor role in AP-1 activation in T cells, probably through induction of c-Jun expression, suggesting that the antiapoptotic effect of ERK5 involves NF-κB activation.

ERK5 activates NF-κB by a noncanonical pathway

NF-κB is retained in the cytoplasm by the IκB family of proteins. Upon TCR stimulation, IκB proteins are phosphorylated by one of a number of IKKs, which leads to their ubiquitinylation and degradation. This frees NF-κB to translocate to the nucleus and activate its target genes. An IκB superrepressor (IκB-DN) that cannot be phosphorylated, and therefore is not degraded, retains NF-κB proteins in the cytosol and completely blocks their induction via IKK (27). NF-κB induction by PMA requires IKK activation, a process that was effectively blocked by IκB-DN (Fig. 5⇓A). Interestingly, IκB-DN reduced ERK5 + MEK5DD-induced NF-κB activation by only 30% in resting cells (Fig. 5⇓A), whereas the mutant completely abolished cooperation between these kinases and either PMA or PMA plus ionomycin. We obtained similar results in Jurkat cells that constitutively express IκB-DN (data not shown) (34). In T cells, PMA or anti-CD3 Abs activate PKCθ and thereby NF-κB. Conversely, inhibition of PKCθ function by the DN PKCθ-K409R mutant blocks PMA- or anti-CD3-induced NF-κB activation (30), but not that induced by ERK5 (data not shown). Therefore, ERK5 mainly activated NF-κB through a nonconventional pathway that cooperated with the canonical one to superinduce this transcription factor somewhere downstream of IκB.

FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

The ERK5 module activates NF-κB by a nonconventional mechanism. A, 107 Jurkat-TAg cells were cotransfected with 3 μg of each ERK5 and MEK5, 6 μg of IκB superrepressor (IκB-DN), 2 μg of the NF-κB luciferase reporter, and 1 μg of β-galactosidase expression vector. Equal numbers of cells were left unstimulated or stimulated for 4 h with PMA (100 ng/ml) or PMA plus ionomycin (1 μg/ml). The data represent means ± SD, n = 4, Student’s t test: ∗, p < 0.005; compare with ERK5/MEK5-transfected cells. B, 107 Jurkat-TAg cells were cotransfected with 3 μg of ERK5, MEK5, or ERK5 AEF expression vectors and 2 μg of the NF-κB luciferase reporter along with 1 μg of β-galactosidase expression vector. Equal numbers of cells were left unstimulated or stimulated for 4 h with PMA (100 ng/ml), anti-CD3 plus anti-CD28 Abs (1 μg/ml each), or TNF-α (20 ng/ml), and luciferase and β-galactosidase activities were evaluated. The data represent means ± SD, n = 4, Student’s t test: ∗, p < 0.01; compare with control-transfected cells. In the insert, an aliquot of the transfected cell lysate was separated on SDS-PAGE and analyzed by Western blotting with an anti-ERK5 Ab.

Next, we tested whether ERK5 cooperated with other signals that induce the canonical pathway. In T cells, PMA and CD3 engagement induce a similar cascade of intracellular events, but TNF-α induces a different intracellular signaling cascade before activating the IKK complex (48). The ERK5 pathway not only cooperated with PMA, but also anti-CD3 Abs and TNF-α (Fig. 5⇑B). This effect required MEK5-induced ERK5 phosphorylation, because a mutant of ERK5 that cannot be activated by MEK5 (ERK5 AEF; Fig. 5⇑B insert and Ref. 9) had no effect in resting cells and failed to cooperate with any treatment (Fig. 5⇑B). Therefore, the ERK5 cascade targets a pathway, activated by either TNF and/or the TCR, which lies downstream of IKKs.

The ERK5 pathway predominantly targets p65

Members of the NF-κB family bind as homo- or heterodimers to NF-κB regulatory elements, with p50/p65 representing the prototypical NF-κB complex and c-Rel playing a central role in T cell activation by TCR/CD28 costimulation. Although p65 and c-Rel contain transcriptional activation domains, p50 homodimers act as repressors (49). In Jurkat T cells, only overexpression of p65 activated our NF-κB reporter and cooperated with PMA above control levels (Fig. 6⇓A). Moreover, cotransfected ERK5 + MEK5DD cooperated with p65, but not with p50 or c-Rel, to activate NF-κB in resting or PMA-stimulated Jurkat cells (Fig. 6⇓A). c-Rel was active in our system, as it strongly induced a reporter controlled by a CD28 response element (data not shown).

FIGURE 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6.

The ERK5 module targets p65 in Jurkat T cells. A and B, 107 Jurkat-TAg cells were cotransfected with 3 μg of ERK5 and MEK5DD expression vectors and/or 1 μg of the expression vectors for p65, p50, and c-Rel, together with 2 μg of the corresponding luciferase reporters and 1 μg of the β-galactosidase expression vector. Two days later, equal numbers of cells were either left unstimulated or stimulated for 4 h with PMA (100 ng/ml), and luciferase and β-galactosidase activities were evaluated. The data represent means ± SD, n = 4, Student’s t test: ∗∗, p < 0.01; compare with p65-transfected cells.

We next investigated the effect of the ERK5 cascade on the 3× κB thymidine kinase reporter that is predominantly targeted by p65 (Fig. 6⇑B). Cotransfection of ERK5 + MEK5DD significantly activated this reporter in resting or PMA-stimulated cells, and synergized with p65 to activate this reporter in resting or PMA-stimulated cells (Fig. 6⇑B).

ERK5 controls p65 localization in the nucleus

The p65 protein shuttles in and out of the nucleus, where it exerts its biological activity (50). Proteins that induce p65 nuclear translocation or block its export to the cytosol will increase its transcriptional activity. To investigate whether ERK5 controlled p65 localization, we first blocked ERK5 expression with shERK5 (Fig. 7⇓A) and stimulated the cells to induce p65 nuclear translocation (Fig. 7⇓B). In the control scrambled shRNA-transfected cells, most of p65 localized in the nucleus after 30 min of stimulation, and half of the protein was still in the nucleus 2 h postinduction. In Jurkat cells with decreased ERK5 levels, the nuclear fraction of p65 was reduced after 30 min of stimulation, and was equal to resting cells after 120 min (Fig. 7⇓B). JunB and ERK2 showed the expected localization, indicating that the different fractions were largely pure.

FIGURE 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 7.

ERK5 controlled p65 nuclear localization. A and B, 6 × 107 Jurkat cells were transfected with shERK5 or with a scramble sequence (control). Three days later, GFP+ cells were sorted by FACS (Fig. 1⇑), and stimulated for different times with PMA (100 ng/ml) plus ionomycin (1 μg/ml). After subcellular fractionation, the presence of the different proteins in the nuclear and cytosolic fractions was analyzed by immunoblotting. C, 108 million Jurkat-TAg cells were cotransfected with 2 μg of GFP and an empty vector or ERK5 plus MEK5DD expression vectors. Two days later, GFP+ cells were sorted and equal numbers of cells were left unstimulated or, as a positive control, stimulated for 10 min with 20 ng/ml TNF-α. Cells were fractionated, and only the nuclear fractions were analyzed for enrichment of p65 or p50 by Western blotting. D, 107 Jurkat-TAg cells were cotransfected with 1 μg of ERK5 and MEK5DD expression vectors and/or 0.5 μg of the expression vectors for Gal-DBD or Gal4-p65, together with 1.5 μg of the Gal4-luciferase reporter plasmid and 1 μg of the β-galactosidase control vector. Two days later, equal numbers of cells were either left unstimulated or stimulated for 4 h with PMA (100 ng/ml) and PMA plus ionomycin (1 μg/ml), and luciferase and β-galactosidase activities were evaluated. The data represent means ± SD, n = 4, Student’s t test: ∗, p < 0.02; ∗∗, p < 0.01; ∗∗∗, p < 0.005; compare with Gal4-p65-transfected cells.

We next selectively activated the ERK5 pathway by transfection of MEK5DD and ERK5 and investigated p65 localization. As a positive control, we used TNF-α (Fig. 7⇑C). Constitutive activation of the ERK5 pathway led to nuclear localization of p65 before stimulation without affecting that of p50 (Fig. 7⇑C). As above, the pattern of JunB and ERK2 localization confirmed the purity of the different fractions (data not shown).

ERK5 induces the trans activation activity of p65

To investigate whether the intranuclear localization of p65 correlated with an increase in its transcriptional activity, we used an expression vector (Gal4 DBD-p65) that contains the DNA binding domain of the yeast GAL4 transcription factor fused to the trans activation domain of p65. The activity of this fusion protein is not blocked by IκB association or activated by its degradation. This construct, or the Gal4 DBD alone, was transfected into Jurkat T cells along with a 5×Gal4 Luc reporter gene, in which luciferase expression is controlled by five binding sites for Gal4. Cotransfection of MEK5DD + ERK5 increased trans activation by Gal4 DBD-p65 by >4-fold (Fig. 7⇑D), and was more efficient than PMA or PMA/ionomycin. Moreover, MEK5DD + ERK5 increased the trans-activating activity of p65 induced by either treatment. Taken together, our results suggest a role for ERK5 in potentiating trans activation by p65.

ERK5 is essential for tumor progression of leukemic T cells

The mouse leukemic T cell line EL-4 has been extensively used to generate tumors in C57B6 recipient mice (51, 52). We infected EL-4 cells with a viral vector expressing shERK5 and the puromycin resistance gene to obtain a stable cell line. Although the control shRNA viral vector rapidly generated puromycin-resistant cells, the shERK5 vector gave rise to very few puromycin-resistant cells. We finally obtained a cell line, called EL-4-shERK, that showed decreased ERK5 expression (Fig. 8⇓A), increased spontaneous apoptosis relative to control cells (Fig. 8⇓C), and grew in vitro at a comparable rate to control cells. shERK5-infected cells showed impaired PMA-induced p65 accumulation in the nucleus (Fig. 8⇓B) and were more sensitive to Fas ligand-induced apoptosis (Fig. 8⇓C). All of these results were in agreement with our results in Jurkat T cells, and confirmed our hypothesis that ERK5 protects leukemic T cells from death receptor-induced apoptosis. Similarly to Jurkat cells (Fig. 2⇑), taxol, CHX, and etoposide induced comparable levels of apoptosis in EL-4-control and EL-4-shERK5 cells (data not shown). Interestingly, ERK5 down-regulation in EL-4 stable cell lines, but not in transient transfected Jurkat cells, led to an increase in the ERK2 protein. We confirmed these results by preparing a second stable shERK5-expressing EL-4 cell line (shERK5 B) (Fig. 8⇓A, right panel). Whereas ERK2 overexpression in these cells might compensate for the decrease of ERK5 in certain cellular functions, it does not account for the increase in apoptosis or the decrease in p65 accumulation in the nucleus (Fig. 8⇓, B and C).

FIGURE 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 8.

ERK5 down-regulation in EL-4 cells inhibits p65 accumulation and facilitates death receptor-induced apoptosis. A, 106 control or shERK5-expressing cells were subjected to immunoblotting to analyze the expression of ERK5, ERK2, and actin. Jurkat cells were transiently transfected, and EL-4 cells were stable transfected. Right panel, Shows the results of two EL-4 stable cell lines expressing shERK5. B, 5 × 106 control or shERK5 cells were stimulated with PMA (100 ng/ml) plus ionomycin (1 μg/ml) for 30 min. After subcellular fractionation, the presence of the different proteins in the nuclear and cytosolic fractions was analyzed by immunoblotting. C, 2 × 105 wt or shERK5 were treated with Fas ligand (1 ng/ml). Twenty-four hours later, cells were stained with annexin V-PE and analyzed by flow cytometry. Mean percentages ± SEM of specific apoptosis from three independent experiments are presented. Student’s t test, ∗, p < 0.01; compare with control cells. Specific apoptosis was calculated by subtracting the percentage of apoptosis under resting conditions from the percentage of apoptosis after treatment. These values were 11.4 and 14.8% for control or shERK5 cells, respectively. The experiments were performed three times with similar results.

The immune system clears tumor cells by inducing apoptosis through different mechanisms. One involves activation of death receptors on the surface of tumor cells. In contrast, some tumor cells escape immune system surveillance by becoming resistant to apoptosis. Because shERK5 rendered leukemic T cells more sensitive to death receptor-induced apoptosis, we injected EL-4-shERK5, as well as control cells, in C57B6 mice and investigated tumor formation. Wild-type (wt) EL-4 cells induced tumors in all mice (Fig. 9⇓). The tumors were first observed 1 wk after cell injection and progressed rapidly, as shown by the plots in Fig. 9⇓. In contrast, EL-4-shERK5 cells failed to develop tumors (Fig. 9⇓) in four independent experiments (n = 20). The second EL-4-shERK5 cell line (EL-4-shERK5 B) also failed to induce tumors (data not shown). In contrast, EL-4 cells stable expressing small hairpin RNA for luciferase induced tumors like the wt cells. shERK5-injected mice sacrificed at day 17 contained no tumor cells. Moreover, mice infected with EL-4-shERK5 were healthy 2 mo after injection, showing that the absence of tumors was not related to slower proliferation, but more likely reflected clearance of tumor cells. Furthermore, these results indicate that ERK2 up-regulation in shERK5-expressing cells could not compensate for the down-regulation of ERK5 in vivo.

FIGURE 9.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 9.

Down-regulation of ERK5 blocks tumor development in mice. A, Five C57B6 mice were injected with 2.5 × 105 control or shERK5 cells. Every 2 days, tumor size was measured. The wt mice with large tumors were euthanized 17 days after inoculation. Two months after cell injection, shERK5-injected mice were still alive and healthy. This experiment was performed four times with identical results (n = 20). B, Tumors of euthanized mice injected with different EL-4 cell lines: wt, expressing small hairpin RNA for luciferase or expressing shERK5 were weight and the average ± SD is depicted in the graphic.

Discussion

The ERK5 and NF-κB pathways play a significant role in the development and survival of different kinds of tumor cells. Moreover, treatments aimed at decreasing the activity of NF-κB, such as the proteasome inhibitor bortezomib, have demonstrated clinical benefit. In this study, we show that both ERK5 and NF-κB signaling cooperate to facilitate leukemic T cell survival in vitro. Furthermore, ERK5 knockdown blocked tumor progression in mice, suggesting that ERK5, in addition to NF-κB, may be an ideal candidate for therapeutic intervention, at least in T cell leukemias.

Dysregulation of ERK5 has a significant role in maintenance of leukemic and perhaps other malignant cells, but our results do not show that ERK5 is necessarily oncogenic per se. Previous reports (12) in tumoral plasma cells indicated that the ERK5 cascade plays a role in the control of neoplastic B cells. However, the mechanism underlying its activity was not detailed in that study. In this study, using a related cellular model derived from a T cell malignancy, we confirmed that the ERK5 pathway participates in the control of survival of leukemic T cells. Moreover, studies on the mechanism by which ERK5 may influence leukemic cell survival indicated a predominant role of the NF-κB pathway as a mediator of the action of ERK5. Several findings support this conclusion. First, blocking ERK5 expression in leukemic T cells by shRNA induced spontaneous apoptosis; second, it sensitized them to TNF-α-induced apoptosis, a process that absolutely requires inhibition of NF-κB activity (38, 39); third, it failed to sensitize Jurkat T cells to apoptotic agents that do not require NF-κB.inhibition, i.e., etoposide, taxol, and CHX; fourth, it converted the survival agent PMA into an apoptotic factor, as does blocking NF-κB activity (34). Importantly, these effects were also observed in the mouse leukemic cell line EL-4. In the latter, and during the generation of the EL-4 stable cell lines, we confirmed that ERK5 was essential for EL-4 survival, because few cells carried the puromycin-resistant gene, and therefore, shERK5. We probably selected cells that expressed low levels of ERK5, but still enough to survive. Knockdown of ERK5 cooperated with Fas engagement to induce apoptosis, as occurs when NF-κB activity is inhibited (43, 44, 45). It is interesting that inhibition of NF-κB also enhances TRAIL-induced apoptosis (53). TRAIL, a member of the TNF family of receptors, induces apoptosis in many transformed cells, but not in normal cells. Our preliminary results show that shERK5 renders leukemic cells more sensitive to TRAIL. In line with these findings, Aza-Blanc et al. (54) have described that blocking ERK5 expression by siRNAs enhanced TRAIL-induced death. As TRAIL is being explored as a potential anticancer drug, the development of strategies to decrease ERK5 function may be of therapeutic value by increasing TRAIL effectiveness.

Our results indicate that NF-κΒ activation by ERK5 occurred by a nonconventional mechanism that does not totally require IKK-mediated phosphorylation of IκB. NF-κB is composed of homo- and heterodimers of Rel family proteins. All Rel proteins share a Rel homology domain controlling DNA-binding activity. In addition, p65/RelA, RelB, and c-Rel contain a C-terminal trans activation domain. A DN mutant of IκB blocks conventional activation of NF-κB, but not that induced by ERK5, suggesting that the latter does not involve phosphorylation of IκB, but rather another, perhaps later, event. This may involve nuclear accumulation of p65. In fact, reduction of ERK5 provoked a decrease in the nuclear retention of p65, indicating that ERK5 may favor NF-κB activity by enhancing p65 nuclear localization. The accumulation of p65 in the nucleus could arise from its phosphorylation, and/or by degradation of IκBα. ERK5 binds to and phosphorylates ribosomal S6 kinase 1 (RSK1) (23, 55). RSK1 phosphorylates the p65 subunit of NF-κB, decreasing the affinity of p65 for IκBα, which allows p65 to be activated in the nucleus (56). TCR-induced T cell stimulation induces RSK1 phosphorylation (57, 58) (J. Garaude and M. Villalba, unpublished observations), suggesting that RSK1 may transduce the signal(s) from ERK5 to NF-κB. We are investigating this possibility.

Although this mechanism appears attractive, other possibilities may also explain the effect of ERK5 on NF-κB activation. We have shown previously that TCR stimulation induces accumulation of phosphorylated ERK5 in the nucleus (1). Therefore, ERK5 could control p65 localization by direct interaction. Related to this is the interaction of the C-terminal region of ERK5 with MEF2 transcription factors. This domain was required for coactivation of MEF2D by ERK5, and the MEF2-ERK5 interaction was found to be dependent upon activation. The transcriptional activation domain of ERK5 is required for maximal MEF2 activity in response to calcium flux in T cells, and it can activate the endogenous Nur77 gene when constitutively recruited to the Nur77 promoter via MEF2 sites (59). Therefore, p65 could recruit ERK5, and its transcriptional activity, to p65-dependent promoters, or vice versa. More generally, the idea that p65 might interact with ERK5 raises the interesting possibility that ERK5 acts as a nuclear docking site for multiple transcription factors, extending their temporal nuclear residency, as occurs with p65, or favoring their transcriptional activity, as occurs with MEF2.

Both NF-κB and ERK5 have been linked to the proliferation/survival of other tumor cells, opening the question as to whether these pathways also collaborate in supporting tumor growth in other cell systems. Although we have not tested the role of ERK5 in NF-κB-induced survival in other cell types, our preliminary results, in agreement with previous studies (23), show that overexpression of ERK5 and MEK5DD in nonhemopoietic cells barely increases NF-κB activity. In contrast, in vascular endothelial cells, IGF-1 activates NF-κB by a PD98059-sensitive pathway (60). PD98059 inhibits both MEK1 and MEK5. Therefore, ERK5 could mediate the effect of IGF-1 on NF-κB. The vascular failure induced by targeted deletion of ERK5 in adult mice arises from massive apoptosis of endothelial cells (19). Endothelial and hemopoietic cells have the same ontogenic precursor, suggesting that they probably share certain intracellular signaling mechanisms. It will be very interesting to determine whether endothelial cells derived from ERK5-deficient mice show defects in NF-κB activation by IGF-1 or other factors that promote survival in these cells.

ERK2 and ERK5 seem likely to share some biological functions. Prolonged ERK5 down-regulation could induce an increase in ERK2 levels to compensate for the reduced ERK5 levels. In stable cell lines in which ERK5 has been down-regulated by shRNA, increased ERK2 levels might enhance survival. This might explain why we do not observe these differences in transient transfections, in which there is no long-term selection underway. The data obtained in mice injected with wt or ERK5 knockdown EL-4 cells argue in favor of an in vivo role of the ERK5 pathway in the regulation of T cell leukemias. Interestingly, the in vitro proliferation properties of EL-4-shERK5 cells did not differ from that of wt EL-4 cells. Yet, the former were unable to create tumors in mice. This discrepancy between in vitro and in vivo behavior very likely reflects the complex physiological conditions encountered by the tumor cells in vivo. One possible explanation is increased tumor cell killing by death receptor agonists available in the animal, but absent in vitro. Whatever the mechanism, our in vivo observations indicate that the ERK5-NF-κB duo is an attractive target for therapeutic intervention in leukemias.

Acknowledgments

We thank the members of the Intracellular Signaling and Gene Regulation group at the Institut de Génétique Moléculaire de Montpellier for helpful discussions and comments during the preparation of this manuscript. We specially thank the members of the Animal Facility at the Institut de Génétique Moléculaire de Montpellier and C. Duperray for their help on cell sorting.

Disclosures

The authors have no financial conflict of interest.

Footnotes

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • ↵1 This work was supported by the French Association pour la Recherche sur le Cancer (to M.V. and R.A.H.); Fondation de France (to M.V.); a Marie Curie International Reintegration Grant (to M.V.); and fellowships from La Ligue Contre le Cancer Department de l’Herault (to S.K.), Region Languedoc-Rousillon (to S.C.), and Comite National (to J.G.).

  • ↵2 Address correspondence and reprint requests to Dr. Martin Villalba, Institut de Génétique Moléculaire de Montpellier, Centre National de la Recherche Scientifique-Unité Mixte de Recherche 5535, Institut Fédératif de Recherche 122, 1919 Route de Mende, 34293 Montpellier, France. E-mail address: martin.villalba{at}igmm.cnrs.fr

  • ↵3 Abbreviations used in this paper: IGF, insulin-like growth factor; CHX, cycloheximide; DN, dominant negative; FSC, forward scatter; SSC, side scatter; HA, hemagglutinin; IKK, IκB kinase; MEF, myocyte enhancer factor; PKC, protein kinase C; RSK1, ribosomal S6 kinase 1; shERK5, small hairpin RNA ERK5; wt, wild type.

  • Received May 23, 2006.
  • Accepted August 23, 2006.
  • Copyright © 2006 by The American Association of Immunologists

References

  1. ↵
    Garaude, J., S. Kaminski, S. Cherni, R. A. Hipskind, M. Villalba. 2005. The role of ERK5 in T-cell signalling. Scand. J. Immunol. 62: 515-520.
    OpenUrlCrossRefPubMed
  2. ↵
    Lee, J. D., R. J. Ulevitch, J. Han. 1995. Primary structure of BMK1: a new mammalian MAP kinase. Biochem. Biophys. Res. Commun. 213: 715-724.
    OpenUrlCrossRefPubMed
  3. ↵
    Buschbeck, M., A. Ullrich. 2005. The unique C-terminal tail of the mitogen-activated protein kinase ERK5 regulates its activation and nuclear shuttling. J. Biol. Chem. 280: 2659-2667.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Kato, Y., R. I. Tapping, S. Huang, M. H. Watson, R. J. Ulevitch, J. D. Lee. 1998. Bmk1/Erk5 is required for cell proliferation induced by epidermal growth factor. Nature 395: 713-716.
    OpenUrlCrossRefPubMed
  5. ↵
    Sohn, S. J., D. Li, L. K. Lee, A. Winoto. 2005. Transcriptional regulation of tissue-specific genes by the ERK5 mitogen-activated protein kinase. Mol. Cell. Biol. 25: 8553-8566.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Esparis-Ogando, A., E. Diaz-Rodriguez, J. C. Montero, L. Yuste, P. Crespo, A. Pandiella. 2002. Erk5 participates in neuregulin signal transduction and is constitutively active in breast cancer cells overexpressing ErbB2. Mol. Cell. Biol. 22: 270-285.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Yuste, L., J. C. Montero, A. Esparis-Ogando, A. Pandiella. 2005. Activation of ErbB2 by overexpression or by transmembrane neuregulin results in differential signaling and sensitivity to herceptin. Cancer Res. 65: 6801-6810.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    English, J. M., G. Pearson, T. Hockenberry, L. Shivakumar, M. A. White, M. H. Cobb. 1999. Contribution of the ERK5/MEK5 pathway to Ras/Raf signaling and growth control. J. Biol. Chem. 274: 31588-31592.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Mulloy, R., S. Salinas, A. Philips, R. A. Hipskind. 2003. Activation of cyclin D1 expression by the ERK5 cascade. Oncogene 22: 5387-5398.
    OpenUrlCrossRefPubMed
  10. ↵
    Linnerth, N. M., M. Baldwin, C. Campbell, M. Brown, H. McGowan, R. A. Moorehead. 2005. IGF-II induces CREB phosphorylation and cell survival in human lung cancer cells. Oncogene 24: 7310-7319.
    OpenUrlCrossRefPubMed
  11. ↵
    Buschbeck, M., S. Hofbauer, L. D. Croce, G. Keri, A. Ullrich. 2005. Abl-kinase-sensitive levels of ERK5 and its intrinsic basal activity contribute to leukemia cell survival. EMBO Rep. 6: 63-69.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Carvajal-Vergara, X., S. Tabera, J. C. Montero, A. Esparis-Ogando, R. Lopez-Perez, G. Mateo, N. Gutierrez, M. Parmo-Cabanas, J. Teixido, J. F. San Miguel, A. Pandiella. 2005. Multifunctional role of Erk5 in multiple myeloma. Blood 105: 4492-4499.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Mehta, P. B., B. L. Jenkins, L. McCarthy, L. Thilak, C. N. Robson, D. E. Neal, H. Y. Leung. 2003. MEK5 overexpression is associated with metastatic prostate cancer, and stimulates proliferation, MMP-9 expression and invasion. Oncogene 22: 1381-1389.
    OpenUrlCrossRefPubMed
  14. ↵
    Weldon, C. B., A. B. Scandurro, K. W. Rolfe, J. L. Clayton, S. Elliott, N. N. Butler, L. I. Melnik, J. Alam, J. A. McLachlan, B. M. Jaffe, et al 2002. Identification of mitogen-activated protein kinase kinase as a chemoresistant pathway in MCF-7 cells by using gene expression microarray. Surgery 132: 293-301.
    OpenUrlCrossRefPubMed
  15. ↵
    Linnerth, N. M., M. Baldwin, C. Campbell, M. Brown, H. McGowan, R. A. Moorehead. 2005. IGF-II induces CREB phosphorylation and cell survival in human lung cancer cells. Oncogene 24: 7310-7319.
    OpenUrlCrossRefPubMed
  16. ↵
    Sohn, S. J., B. K. Sarvis, D. Cado, A. Winoto. 2002. ERK5 MAPK regulates embryonic angiogenesis and acts as a hypoxia-sensitive repressor of vascular endothelial growth factor expression. J. Biol. Chem. 277: 43344-43351.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Regan, C. P., W. Li, D. M. Boucher, S. Spatz, M. S. Su, K. Kuida. 2002. Erk5 null mice display multiple extraembryonic vascular and embryonic cardiovascular defects. Proc. Natl. Acad. Sci. USA 99: 9248-9253.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Wang, X., A. J. Merritt, J. Seyfried, C. Guo, E. S. Papadakis, K. G. Finegan, M. Kayahara, J. Dixon, R. P. Boot-Handford, E. J. Cartwright, et al 2005. Targeted deletion of mek5 causes early embryonic death and defects in the extracellular signal-regulated kinase 5/myocyte enhancer factor 2 cell survival pathway. Mol. Cell. Biol. 25: 336-345.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Hayashi, M., S. W. Kim, K. Imanaka-Yoshida, T. Yoshida, E. D. Abel, B. Eliceiri, Y. Yang, R. J. Ulevitch, J. D. Lee. 2004. Targeted deletion of BMK1/ERK5 in adult mice perturbs vascular integrity and leads to endothelial failure. J. Clin. Invest. 113: 1138-1148.
    OpenUrlCrossRefPubMed
  20. ↵
    Cameron, S. J., S. Itoh, C. P. Baines, C. Zhang, S. Ohta, W. Che, M. Glassman, J. D. Lee, C. Yan, J. Yang, J. Abe. 2004. Activation of big MAP kinase 1 (BMK1/ERK5) inhibits cardiac injury after myocardial ischemia and reperfusion. FEBS Lett. 566: 255-260.
    OpenUrlCrossRefPubMed
  21. ↵
    Hayashi, M., C. Fearns, B. Eliceiri, Y. Yang, J. D. Lee. 2005. Big mitogen-activated protein kinase 1/extracellular signal-regulated kinase 5 signaling pathway is essential for tumor-associated angiogenesis. Cancer Res. 65: 7699-7706.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Barsyte-Lovejoy, D., A. Galanis, A. Clancy, A. D. Sharrocks. 2004. ERK5 is targeted to myocyte enhancer factor 2A (MEF2A) through a MAPK docking motif. Biochem. J. 381: 693-699.
    OpenUrlCrossRefPubMed
  23. ↵
    Pearson, G., J. M. English, M. A. White, M. H. Cobb. 2001. ERK5 and ERK2 cooperate to regulate NF-κB and cell transformation. J. Biol. Chem. 276: 7927-7931.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Rayet, B., C. Gelinas. 1999. Aberrant rel/nfkb genes and activity in human cancer. Oncogene 18: 6938-6947.
    OpenUrlCrossRefPubMed
  25. ↵
    Villalba, M., S. Kasibhatla, L. Genestier, A. Mahboubi, D. R. Green, A. Altman. 1999. PKCθ is a necessary component, and cooperates with calcineurin, to induce FasL expression during activation-induced T cell death. J. Immunol. 163: 5813-5819.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Villalba, M., K. Bi, J. Hu, Y. Altman, P. Bushway, E. Reits, J. Neefjes, G. Baier, R. T. Abraham, A. Altman. 2002. Translocation of PKCθ in T cells is mediated by a nonconventional, PI3-K- and Vav-dependent pathway, but does not absolutely require phospholipase C. J. Cell Biol. 157: 253-263.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Brockman, J. A., D. C. Scherer, T. A. McKinsey, S. M. Hall, X. Qi, W. Y. Lee, D. W. Ballard. 1995. Coupling of a signal response domain in IκBα to multiple pathways for NF-κB activation. Mol. Cell. Biol. 15: 2809-2818.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Villalba, M., N. Coudronniere, M. Deckert, E. Teixeiro, P. Mas, A. Altman. 2000. A novel functional interaction between Vav and PKCθ is required for TCR-induced T cell activation. Immunity 12: 151-160.
    OpenUrlCrossRefPubMed
  29. ↵
    Villalba, M., J. Hernandez, M. Deckert, A. Altman. 2000. Vav modulation of the Ras/MEK/ERK signaling pathway plays a role in NFAT activation and CD69 up-regulation. Eur. J. Immunol. 30: 1587-1596.
    OpenUrlCrossRefPubMed
  30. ↵
    Coudronniere, N., M. Villalba, N. Englund, A. Altman. 2000. NF-κB activation induced by CD28 costimulation is mediated by PKCθ. Proc. Natl. Acad. Sci. USA 97: 3394-3399.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Martin, A. G., M. Fresno. 2000. Tumor necrosis factor-α activation of NF-κB requires the phosphorylation of Ser-471 in the transactivation domain of c-Rel. J. Biol. Chem. 275: 24383-24391.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Yano, O., J. Kanellopoulos, M. Kieran, O. Le Bail, A. Israel, P. Kourilsky. 1987. Purification of KBF1, a common factor binding to both H-2 and β2-microglobulin enhancers. EMBO J. 6: 3317-3324.
    OpenUrlPubMed
  33. ↵
    Schmitz, M. L., P. A. Baeuerle. 1991. The p65 subunit is responsible for the strong transcription activating potential of NF-κB. EMBO J. 10: 3805-3817.
    OpenUrlPubMed
  34. ↵
    Busuttil, V., V. Bottero, C. Frelin, V. Imbert, J. E. Ricci, P. Auberger, J. F. Peyron. 2002. Blocking NF-κB activation in Jurkat leukemic T cells converts the survival agent and tumor promoter PMA into an apoptotic effector. Oncogene 21: 3213-3224.
    OpenUrlCrossRefPubMed
  35. ↵
    Villalba, M., P. Bushway, A. Altman. 2001. PKCθ mediates a selective T cell survival signal via phosphorylation of BAD. J. Immunol. 166: 5955-5963.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Ashkenazi, A., V. M. Dixit. 1999. Apoptosis control by death and decoy receptors. Curr. Opin. Cell Biol. 11: 255-260.
    OpenUrlCrossRefPubMed
  37. ↵
    Engedal, N., H. K. Blomhoff. 2003. Combined action of ERK and NFκB mediates the protective effect of phorbol ester on Fas-induced apoptosis in Jurkat cells. J. Biol. Chem. 278: 10934-10941.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Van Antwerp, D. J., S. J. Martin, T. Kafri, D. R. Green, I. M. Verma. 1996. Suppression of TNF-α-induced apoptosis by NF-κB. Science 274: 787-789.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Beg, A. A., D. Baltimore. 1996. An essential role for NF-κB in preventing TNF-α-induced cell death. Science 274: 782-784.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Nencioni, A., L. Wille, G. Dal Bello, D. Boy, G. Cirmena, S. Wesselborg, C. Belka, P. Brossart, F. Patrone, A. Ballestrero. 2005. Cooperative cytotoxicity of proteasome inhibitors and tumor necrosis factor-related apoptosis-inducing ligand in chemoresistant Bcl-2-overexpressing cells. Clin. Cancer Res. 11: 4259-4265.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Sabatini, N., R. Di Pietro, M. Rapino, S. Sancilio, S. Comani, A. Cataldi. 2004. PI-3-kinase/NF-κB mediated response of Jurkat T leukemic cells to two different chemotherapeutic drugs, etoposide and TRAIL. J. Cell. Biochem. 93: 301-311.
    OpenUrlCrossRefPubMed
  42. ↵
    Bellarosa, D., M. Binaschi, C. A. Maggi, C. Goso. 2005. Sabarubicin- (MEN 10755) and paclitaxel show different kinetics in nuclear factor-κB (NF-κB) activation: effect of parthenolide on their cytotoxicity. Anticancer Res. 25: 2119-2128.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Dudley, E., F. Hornung, L. Zheng, D. Scherer, D. Ballard, M. Lenardo. 1999. NF-κB regulates Fas/APO-1/CD95- and TCR-mediated apoptosis of T lymphocytes. Eur. J. Immunol. 29: 878-886.
    OpenUrlCrossRefPubMed
  44. ↵
    Boothby, M. R., A. L. Mora, D. C. Scherer, J. A. Brockman, D. W. Ballard. 1997. Perturbation of the T lymphocyte lineage in transgenic mice expressing a constitutive repressor of nuclear factor (NF)-κB. J. Exp. Med. 185: 1897-1907.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Jones, R. G., S. D. Saibil, J. M. Pun, A. R. Elford, M. Bonnard, M. Pellegrini, S. Arya, M. E. Parsons, C. M. Krawczyk, S. Gerondakis, et al 2005. NF-κB couples protein kinase B/Akt signaling to distinct survival pathways and the regulation of lymphocyte homeostasis in vivo. J. Immunol. 175: 3790-3799.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Fluhmann, B., U. Zimmermann, R. Muff, G. Bilbe, J. A. Fischer, W. Born. 1998. Parathyroid hormone responses of cyclic AMP-, serum- and phorbol ester-responsive reporter genes in osteoblast-like UMR-106 cells. Mol. Cell. Endocrinol. 139: 89-98.
    OpenUrlCrossRefPubMed
  47. ↵
    Claret, F. X., M. Hibi, S. Dhut, T. Toda, M. Karin. 1996. A new group of conserved coactivators that increase the specificity of AP-1 transcription factors. Nature 383: 453-457.
    OpenUrlCrossRefPubMed
  48. ↵
    Li, Z. W., R. C. Rickert, M. Karin. 2004. Genetic dissection of antigen receptor-induced NF-κB activation. Mol. Immunol. 41: 701-714.
    OpenUrlCrossRefPubMed
  49. ↵
    Caamano, J., C. A. Hunter. 2002. NF-κB family of transcription factors: central regulators of innate and adaptive immune functions. Clin. Microbiol. Rev. 15: 414-429.
    OpenUrlAbstract/FREE Full Text
  50. ↵
    Tam, W. F., R. Sen. 2001. IκB family members function by different mechanisms. J. Biol. Chem. 276: 7701-7704.
    OpenUrlAbstract/FREE Full Text
  51. ↵
    Dyall, R., L. V. Vasovic, R. A. Clynes, J. Nikolic-Zugic. 1999. Cellular requirements for the monoclonal antibody-mediated eradication of an established solid tumor. Eur. J. Immunol. 29: 30-37.
    OpenUrlCrossRefPubMed
  52. ↵
    Zeytun, A., M. Nagarkatti, P. S. Nagarkatti. 2000. Growth of FasL-bearing tumor cells in syngeneic murine host induces apoptosis and toxicity in Fas+ organs. Blood 95: 2111-2117.
    OpenUrl
  53. ↵
    Jeremias, I., C. Kupatt, B. Baumann, I. Herr, T. Wirth, K. M. Debatin. 1998. Inhibition of nuclear factor κB activation attenuates apoptosis resistance in lymphoid cells. Blood 91: 4624-4631.
    OpenUrlAbstract/FREE Full Text
  54. ↵
    Aza-Blanc, P., C. L. Cooper, K. Wagner, S. Batalov, Q. L. Deveraux, M. P. Cooke. 2003. Identification of modulators of TRAIL-induced apoptosis via RNAi-based phenotypic screening. Mol. Cell 12: 627-637.
    OpenUrlCrossRefPubMed
  55. ↵
    Ranganathan, A., G. W. Pearson, C. A. Chrestensen, T. W. Sturgill, M. H. Cobb. 2006. The MAP kinase ERK5 binds to and phosphorylates p90 RSK. Arch. Biochem. Biophys. 449: 8-16.
    OpenUrlCrossRefPubMed
  56. ↵
    Bohuslav, J., L. F. Chen, H. Kwon, Y. Mu, W. C. Greene. 2004. p53 induces NF-κB activation by an IκB kinase-independent mechanism involving phosphorylation of p65 by ribosomal S6 kinase 1. J. Biol. Chem. 279: 26115-26125.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    Lafont, V., E. Astoul, A. Laurence, J. Liautard, D. Cantrell. 2000. The T cell antigen receptor activates phosphatidylinositol 3-kinase-regulated serine kinases protein kinase B and ribosomal S6 kinase 1. FEBS Lett. 486: 38-42.
    OpenUrlCrossRefPubMed
  58. ↵
    Franklin, R. A., A. Tordai, H. Patel, A. M. Gardner, G. L. Johnson, E. W. Gelfand. 1994. Ligation of the T cell receptor complex results in activation of the Ras/Raf-1/MEK/MAPK cascade in human T lymphocytes. J. Clin. Invest. 93: 2134-2140.
    OpenUrlCrossRefPubMed
  59. ↵
    Kasler, H. G., J. Victoria, O. Duramad, A. Winoto. 2000. ERK5 is a novel type of mitogen-activated protein kinase containing a transcriptional activation domain. Mol. Cell. Biol. 20: 8382-8389.
    OpenUrlAbstract/FREE Full Text
  60. ↵
    Liu, W., Y. Liu, W. L. Lowe, Jr. 2001. The role of phosphatidylinositol 3-kinase and the mitogen-activated protein kinases in insulin-like growth factor-I-mediated effects in vascular endothelial cells. Endocrinology 142: 1710-1719.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

The Journal of Immunology: 177 (11)
The Journal of Immunology
Vol. 177, Issue 11
1 Dec 2006
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Advertising (PDF)
  • Back Matter (PDF)
  • Editorial Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
ERK5 Activates NF-κB in Leukemic T Cells and Is Essential for Their Growth In Vivo
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
ERK5 Activates NF-κB in Leukemic T Cells and Is Essential for Their Growth In Vivo
Johan Garaude, Seyma Cherni, Sandra Kaminski, Etienne Delepine, Christine Chable-Bessia, Monsef Benkirane, Joana Borges, Atanasio Pandiella, Miguel Angel Iñiguez, Manuel Fresno, Robert A. Hipskind, Martin Villalba
The Journal of Immunology December 1, 2006, 177 (11) 7607-7617; DOI: 10.4049/jimmunol.177.11.7607

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
ERK5 Activates NF-κB in Leukemic T Cells and Is Essential for Their Growth In Vivo
Johan Garaude, Seyma Cherni, Sandra Kaminski, Etienne Delepine, Christine Chable-Bessia, Monsef Benkirane, Joana Borges, Atanasio Pandiella, Miguel Angel Iñiguez, Manuel Fresno, Robert A. Hipskind, Martin Villalba
The Journal of Immunology December 1, 2006, 177 (11) 7607-7617; DOI: 10.4049/jimmunol.177.11.7607
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Disclosures
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Innate Immunity Together with Duration of Antigen Persistence Regulate Effector T Cell Induction
  • Regulatory Roles of IL-2 and IL-4 in H4/Inducible Costimulator Expression on Activated CD4+ T Cells During Th Cell Development
  • Induction of CD4+ T Cell Apoptosis as a Consequence of Impaired Cytoskeletal Rearrangement in UVB-Irradiated Dendritic Cells
Show more CELLULAR IMMUNOLOGY AND IMMUNE REGULATION

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • Public Access
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2021 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606