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
  • Log in

Search

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

Leflunomide Suppresses TNF-Induced Cellular Responses: Effects on NF-κB, Activator Protein-1, c-Jun N-Terminal Protein Kinase, and Apoptosis

Sunil K. Manna, Asok Mukhopadhyay and Bharat B. Aggarwal
J Immunol November 15, 2000, 165 (10) 5962-5969; DOI: https://doi.org/10.4049/jimmunol.165.10.5962
Sunil K. Manna
Cytokine Research Laboratory, Department of Bioimmunotherapy, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Asok Mukhopadhyay
Cytokine Research Laboratory, Department of Bioimmunotherapy, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Bharat B. Aggarwal
Cytokine Research Laboratory, Department of Bioimmunotherapy, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030
  • 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

Leflunomide is a pyrimidine biosynthesis inhibitor that has recently been approved for treatment of rheumatoid arthritis. However, the mechanism of leflunomide’s antiarthritis activity and is not fully understood. The critical role that TNF plays in rheumatoid arthritis led us to postulate that leflunomide blocks TNF signaling. Previously, we have demonstrated that leflunomide inhibits TNF-induced NF-κB activation by suppressing I-κBα (inhibitory subunit of NF-κB) degradation. We in this study show that leflunomide also blocks NF-κB reporter gene expression induced by TNFR1, TNFR-associated factor 2, and NF-κB-inducing kinase (NIK), but not that activated by the p65 subunit of NF-κB, suggesting that leflunomide acts downstream of NIK. Leflunomide suppressed TNF-induced phosphorylation of I-κBα, as well as activation of I-κBα kinase-β located downstream to NIK. Leflunomide also inhibited TNF-induced activation of AP-1 and the c-Jun N-terminal protein kinase activation. TNF-mediated cytotoxicity and caspase-induced poly(ADP-ribose) polymerase cleavage were also completely abrogated by treatment of Jurkat T cells with leflunomide. Leflunomide suppressed TNF-induced reactive oxygen intermediate generation and lipid peroxidation, which may explain most of its effects on TNF signaling. The suppressive effects of leflunomide on TNF signaling were completely reversible by uridine, indicating a critical role for pyrimidine biosynthesis in TNF-mediated cellular responses. Overall, our results suggest that suppression of TNF signaling is one of the possible mechanisms for inhibitory activity of leflunomide against rheumatoid arthritis.

Leflunomide (HWA-486), or n-(4-trifluoromethylphenyl)-5-methylisoxazol-4-carboxamide, exhibits antiinflammatory, antiproliferative, and immunosuppressive effects through mechanisms that are not fully understood (1, 2, 3, 4, 5). HWA-486 is a prodrug that is rapidly converted in the cell to an active metabolite, N-(4-trifloromethylphenyl-2,2-cyano-3-hydroxy crotoamide), named A77 1726. The initial conversion involves the opening of the isoxazole ring to produce A77 1726, which constitutes more than 95% of the drug in the circulation. Early experiments suggest that A77 1726 blocks T cell proliferation stimulated by anti-CD28 and PMA, by anti-CD3, and by IL-2 (3, 6). It also prevents the proliferation of B cells and Ab production by B cells (2). How leflunomide suppresses transplant rejection (7, 8, 9), adjuvant arthritis (1), proliferation of B and T cells and smooth muscle cells (6, 7, 10), and IL-2R expression (6) is not understood, but a role for inhibition of NF-κB activation has been suggested (11).

Recent studies have begun to clarify its mechanism of action. Leflunomide is a potent inhibitor of dihydroorotate dehydrogenase (DHODH),3 a rate-limiting enzyme in the biosynthetic pathway of pyrimidines (12, 13, 14, 15, 16). In vitro the Ki of inhibition of DHODH by leflunomide ranges from 179 nM to 2.7 μM (13, 14). Its ability to suppress proliferation of T and B cells (2, 3, 4) has been suggested to be due to inhibition of DHODH (12, 13, 14, 15), a pathway critical for the proliferation of these cells. The reversal of antiproliferative effects of leflunomide by uridine further suggests the critical role of DHODH (12, 15).

Leflunomide can also inhibit several protein tyrosine kinases, including those of src family (p59fyn and p56lck) (6, 12), the Janus kinase (JAK) family (JAK1 and JAK3) (10), and epidermal growth factor receptor kinase (17). The concentration of leflunomide required to inhibit these protein tyrosine kinases is 10–500 times higher than that required to inhibit DHODH. Inhibition of tyrosine phosphorylation of JAK3 and STAT6 by leflunomide has been implicated in inhibition of IgG1 secretion (18). While leflunomide suppresses proliferation of cells by inhibiting DHODH, protein tyrosine kinase inhibition was implicated in its ability to suppress autoimmune and lymphoproliferative disorders (16).

Leflunomide has been recently approved for treatment of rheumatoid arthritis (RA) (19), but how its antiarthritis effects are mediated is not known. Some recent studies suggest TNF may be the mediator. TNF has been implicated in causing RA (20), and agents that can down-regulate TNF-mediated cellular responses, such as TNF-soluble receptors or Ab against TNF, have been approved for treatment of RA (21, 22). We have previously shown that leflunomide blocks TNF-mediated NF-κB activation by suppressing I-κBα degradation (11). We now further extend these studies to the effects of leflunomide on TNF signaling. In our study, leflunomide blocked TNF-induced NF-κB activation, I-κBα (inhibitory subunit of NF-κB) phosphorylation, I-κBα kinase (IKK) activation, and activation of AP-1, c-Jun N-terminal protein kinase, and suppressed TNF-induced apoptosis. Leflunomide also suppressed TNF-induced reactive oxygen intermediate (ROI) generation and lipid peroxidation. We also demonstrate that the suppressive effects of leflunomide on TNF signaling can be reversed by uridine, suggesting an essential role for pyrimidine biosynthesis.

Materials and Methods

Materials

Leflunomide (A77 1726), a generous gift from Dr. Robert R. Bartlett (Hoechst AG, Weisbaden, Germany), was made up as 5 mM solution in water. Penicillin, streptomycin, RPMI 1640 medium, and FCS were obtained from Life Technologies (Grand Island, NY). Glycine, NaCl, and BSA were obtained from Sigma (St. Louis, MO). Bacteria-derived human rTNF, purified to homogeneity with a sp. act. of 5 × 107 U/mg, was kindly provided by Genentech (South San Francisco, CA). Abs against I-κBα, JNK1, cyclin D1, c-Jun, c-Fos, and p50 were obtained from Santa Cruz Biotechnology (Santa Cruz, CA). Poly(ADP-ribose) polymerase (PARP) Ab was purchased from PharMingen (San Diego, CA). Phospho-I-κBα (Ser32) Ab was purchased from New England BioLabs (Beverly, MA). Anti-IKKα or anti-IKKβ Abs were kindly provided by Imgenex (San Diego, CA). Expression plasmids encoding FLAG-tagged NF-κB-inducing kinase (NIK) (23) were kindly provided by Dr. David Wallach (Weizmann Institute of Science, Rehovot, Israel). The expression plasmid encoding myc-tagged TNFR-associated factor 2 (TRAF2) has been previously described (24).

Cell lines

HeLa (human epithelial cells) and T-Jurkat (T cells) were obtained from American Type Culture Collection (Manassas, VA). Cells were cultured in RPMI 1640 medium supplemented with 10% FBS, penicillin (100 U/ml), and streptomycin (100 μg/ml).

Isolation of PBLs

Freshly drawn human blood was incubated with 2.5% gelatin in saline (1:1 ratio) for 30 min at 37°C. The supernatant was layered on Histopaque 1077 (from Sigma) and centrifuged at 1500 rpm for 30 min at room temperature. The cells were then collected from the top layer of Histopaque, diluted with Dulbecco’s PBS, and centrifuged at 2000 rpm for 10 min. To get rid of mixed reticulocytes, pellet was suspended in 0.2% NaCl for 1 min, immediately diluted with equal volume of 1.6% NaCl, and centrifuged at 1000 rpm. To remove macrophages by adherence, the pellet was suspended in RPMI 1640 medium supplemented with 10% FBS and cultured for 2 h at 37°C, CO2 incubator in a petri dish. Then the lymphocytes were harvested from the medium by centrifugation at 1000 rpm.

NF-κB activation assays

To determine NF-κB activation, EMSA were conducted essentially as described (25). Briefly, nuclear extracts prepared from TNF-treated cells (2 × 106/ml) were incubated with 32P end-labeled 45-mer double-stranded NF-κB oligonucleotide (4 μg protein with 16 fmol DNA) from the HIV long terminal repeat, 5′-TTGTTACAAGGGACTTTCCGCTGGGGACTTTCCAGGGAGGCGTGG-3′ (underline indicates NF-κB binding sites) for 15 min at 37°C, and the DNA-protein complex formed was separated from free oligonucleotide on 6.6% native polyacrylamide gels. A double-stranded mutated oligonucleotide, 5′-TTGTTACAACTCACTTTCCGCTGCTCACTTTCCAGGGAGGCGTGG-3′, was used to examine the specificity of binding of NF-κB to the DNA. The specificity of binding was also examined by competition with the unlabeled oligonucleotide. The dried gels were visualized, and radioactive bands were quantitated by a PhosphorImager (Molecular Dynamics, Sunnyvale, CA) using ImageQuant software.

NF-κB-dependent reporter gene transcription

The effect of leflunomide on TNF-, TRAF-2-, NIK-, and p65 (transactivation subunit of NF-κB)-induced NF-κB-dependent reporter gene transcription was measured as previously described (24). Briefly, HeLa cells (0.1 × 106 cells/well) were plated in six-well plates, pretreated with 10 μM leflunomide for 2 h, and then transfected by the calcium phosphate method with medium (1 ml) containing plasmid DNAs for TRAF2, NIK, or p65 (1 μg each) along with 0.5 μg NF-κB promoter DNA linked to heat-stable secretory alkaline phosphatase (SEAP) gene. The total final amount of DNA was maintained at 3 μg by the addition of the control plasmid pCMVFLAG1 DNA.

To examine TNF-induced reporter gene expression, we transfected cells with the SEAP expression plasmid for 10 h before treating them with TNF (1 nM). Treatment with leflunomide was continued during the transfection reaction. Twenty-four hours after transfection, cell culture-conditioned medium was harvested and analyzed (25 μl) for alkaline phosphatase activity essentially as described by the protocol of Clontech (Palo Alto, CA). The activity of SEAP was assayed on a 96-well fluorescent plate reader (Fluoroscan II; Labsystems, Chicago, IL) with excitation set at 360 nm and emission at 460 nm. This reporter system was specific because TNF-induced NF-κB SEAP activity was inhibited by overexpression of either an I-κBα mutant lacking Ser32/36, a kinase-inactive NIK, or a dominant-negative TRAF2 mutant (24).

I-κBα phosphorylation

To determine the effect of leflunomide on I-κBα phosphorylation, cytoplasmic extracts were prepared from cells (2 × 106/ml) treated with leflunomide (10 μM) for 2 h, then with N-acetyl leucyl leucyl nonleucinal (ALLN) (100 μg/ml) for an additional 1 h, and then with TNF (1 nM) for 15 min, resolved on 10% SDS-PAGE, and then analyzed by Western blot using Abs against either I-κBα or phosphorylated I-κBα (26). After electrophoresis, the proteins were detected by chemiluminescence (ECL; Amersham, Arlington Heights, IL).

I-κBα kinase assay

The IKK assays were performed as described (27). Briefly, IKK signalosomes were precipitated by treating 300 μg cytoplasmic extracts with 1μg anti-IKKα Ab (IMG-136) overnight at 4°C, followed by treatment with 20 μl protein A/G-Sepharose (Pierce, Rockford, IL). After 2 h, the beads were washed three times with lysis buffer and three times with the kinase assay buffer, and then resuspended in 20 μl of kinase assay mixture containing 50 mM HEPES (pH 7.4), 20 mM MgCl2, 2 mM DTT, 20 μCi γ-ATP, 10 mM unlabeled ATP, and 2 μg of substrate GST-I-κBα 1–54(1–54). After incubation at 30°C for 30 min, the reaction was terminated by boiling with 5 μl of 5× SDS sample buffer for 5 min. Finally, the protein was resolved on 10% polyacrylamide gel under reducing conditions, the gel was dried, and the radioactive bands were visualized by PhosphorImager. To determine the total amounts of IKKα and IKKβ in each sample, 60 μg of the cytoplasmic protein was resolved on 7.5% acrylamide gels, electrotransferred to a nitrocellulose membrane, blocked the membrane with 5% nonfat milk protein for 1 h, and then incubated with either anti-IKKα or anti-IKKβ (IMG-129) Abs (at 1/500 dilution) for 1 h. The membrane was then washed and reacted with HRP-conjugated secondary anti-mouse IgG Ab, and finally detected by chemiluminescence (ECL; Amersham).

AP-1 activation assay

The activation of AP-1 was determined as described (28). Briefly, 6 μg of nuclear extract prepared as indicated above was incubated with 16 fmol of the 32P end-labeled AP-1 consensus oligonucleotide 5′-CGCTTGATGACTCAGCCGGAA-3′ (bold indicates AP-1 binding site) for 15 min at 37°C and analyzed by using 6% native polyacrylamide gel. The specificity of binding was examined by supershift with anti-c-fos and anti-c-jun Abs and by competition with unlabeled oligonucleotide. Visualization and quantitation of radioactive bands were done as indicated above.

c-Jun kinase assay

The c-Jun kinase assay was performed by a modified method, as described earlier (29). Briefly, after treatment of cells with TNF for 10 min, cell extracts were prepared, immunoprecipitated with anti-JNK Ab, collected the immune complexes by incubation with protein A/G-Sepharose beads, and performed the in vitro kinase assays using GST-Jun 1–79(1–79) as a substrate and [γ-32P]ATP. Reactions were stopped by the addition of SDS sample buffer and subjected the samples to SDS-PAGE. GST-Jun 1–79(1–79) was visualized by staining with Coomassie blue, and the dried gel was analyzed by a PhosphorImager (Molecular Dynamics).

Cytotoxicity assay

TNF-induced cytotoxicity was measured by the MTT assay (28). Briefly, 5000 cells/well were incubated in the presence or absence of the indicated test sample in a final volume of 0.1 ml for 72 h at 37°C. Thereafter, 0.025 ml of MTT solution (5 mg/ml in PBS) was added to each well. After a 2-h incubation at 37°C, 0.1 ml of the extraction buffer (20% SDS, 50% dimethyl formamide) was added. After an overnight incubation at 37°C, the OD at 590 nm were measured using a 96-well multiscanner autoreader (Dynatech MR 5000), with the extraction buffer as a blank.

Immunoblot analysis of PARP degradation

TNF-induced apoptosis was examined by proteolytic cleavage of PARP (30). Briefly, cells (2 × 106/ml) were treated with different concentrations of leflunomide at 37°C for 2 h and then stimulated with 1 nM TNF with cycloheximide (2 μg/ml) for 2 h at 37°C. The cells were then washed and extracted by treatment for 30 min on ice with 0.05 ml buffer containing 20 mM HEPES, pH 7.4, 2 mM EDTA, 250 mM NaCl, 0.1% Nonidet P-40, 2 μg/ml leupeptin, 2 μg/ml aprotinin, 1 mM PMSF, 0.5 μg/ml benzamidine, 1 mM DTT, and 1 mM sodium vanadate. The lysate was centrifuged, and the supernatant was collected. Cell extract protein (50 μg) was resolved on 7.5% SDS-PAGE, electrotransferred onto a nitrocellulose membrane, blotted with mouse anti-PARP Ab, and then detected by chemiluminescence (ECL; Amersham). Apoptosis was represented by the cleavage of 116-kDa PARP into a 85-kDa product.

Determination of lipid peroxidation

TNF-induced lipid peroxidation was determined by detection of thiobarbituric acid-reactive malondialdehyde (MDA), an end product of the peroxidation of polyunsaturated fatty acids and related esters, as described (26). Results were normalized with the amount of MDA equivalents/mg of protein and expressed as a percentage of thiobarbituric acid-reactive substances above control values. Untreated cells showed 0.571 ± 0.126 nmol of MDA equivalents/mg of protein.

Measurement of ROI

The production of ROI upon treatment of cells with TNF was determined by flow cytometry, as described (26).

Radiolabeling of TNF and receptor-binding assay

Human TNF was iodinated with [125I]Na by the Iodogen method, purified, and examined for cell surface receptors, as described previously (30).

Results

In this study, we examined the effect of leflunomide on TNF-induced signal transduction. For most studies, Jurkat cells were used because these T cells express both types of TNFR, and we have previously demonstrated that leflunomide blocks TNF-induced NF-κB activation in this cell (11). The concentration of leflunomide and its time of exposure had no effect on the viability of these cells (data not shown).

Leflunomide represses TNF-induced NF-κB-dependent reporter gene expression

Previously, we have shown that leflunomide blocks TNF-induced NF-κB activation, I-κBα degradation, and NF-κB-dependent reporter gene expression (11). TNF-induced NF-κB activation is mediated through sequential interaction of the TNFR (TNFR1) with TNFR-associated death domain, TRAF2, NIK, and IKK-β, resulting in phosphorylation of I-κBa (31, 32). To delineate the site of action of leflunomide in the TNF-signaling pathway leading to NF-κB activation, HeLa cells were transfected with TNFR1, TRAF2, NIK, and p65 plasmids, and then NF-κB-dependent SEAP expression monitored in leflunomide-untreated and -treated cells. Due to higher transfection efficiency, HeLa cells were used. As shown in Fig. 1⇓, TNFR1, TRAF2, NIK, and p65 plasmids induced gene expression, and leflunomide suppressed TNFR1-, TRAF2-, and NIK-induced expression, but had little effect on p65-induced NF-κB reporter expression. RANK, another NF-κB-inducing receptor, which is a member of the TNFR1 family, was minimally affected by leflunomide, indicating the specificity. Specificity of the assay results is also indicated by suppression of TNF-induced NF-κB reporter activity by the dominant-negative I-κBα plasmid. Thus, leflunomide must act at a step downstream from IKK-β. Because NIK is known to activate IKK-β, which in turn phosphorylates I-κBα, it appears that leflunomide blocked the activity of IKK-β, a kinase that phosphorylates I-κBα directly.

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

Leflunomide inhibits NF-κB-dependent reporter gene expression induced by and activated by TNF, TNFR1, TRAF2, and NIK. Leflunomide-treated HeLa cells were transiently transfected with indicated plasmids along with the NF-κB-containing plasmid linked to the SEAP gene. Where indicated, cells were exposed to 1 nM TNF also. After 24 h in culture, cell supernatants were collected, and 25 μl of it was assayed for secreted alkaline phosphatase activity, as described in Materials and Methods. Results are expressed as fold activity over the nontransfected control. The specificity of the assay was examined by suppression of TNF-induced NF-κB reporter activity by I-κBα dominant-negative mutant plasmid.

Leflunomide inhibits TNF-dependent phosphorylation I-κBα

To determine whether inhibition of TNF-induced I-κBα degradation by leflunomide was due to suppression of I-κBα phosphorylation, cells were treated with the proteosome inhibitor ALLN (33) for 1 h, and then the hyperphosphorylated form of I-κBα was assayed by Western blot using Abs that detect only the serine-phosphorylated form of I-κBα. The results clearly show that TNF induced the phosphorylation of I-κBα and leflunomide suppressed it (Fig. 2⇓A, upper panel). The hyperphosphorylated form of I-κBα also appeared as a slow-migrating band on SDS-PAGE (Fig. 2⇓A, lower panel). The lack of a slow-migrating band in leflunomide-treated cells indicates that leflunomide blocked TNF-induced I-κBα phosphorylation.

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

Leflunomide inhibits TNF-induced phosphorylation of I-κBα. A, Jurkat cells (2 × 106/ml) were incubated first with leflunomide (10 μM) for 2 h and then with ALLN (100 μg/ml) for an additional 1 h before treatment with TNF (1 nM) for 15 min, and then analyzed by Western blot using Abs against either phosphorylated I-κBα (upper panel) or I-κBα (lower panel). S, Indicates slow-migrating band, and N is normal-migrating band. B, Jurkat cells (5 × 106 cells), untreated or pretreated with 10 μM leflunomide for 2 h, were activated with 0.1 nM TNF for different times, thereafter prepared the cytoplasmic extracts and assayed for IKK by the immune complex kinase assay (upper panel) and for IKKα (middle panel) and IKKβ (lower panel) protein by the Western blot analysis, as described in Materials and Methods. C, Effect of leflunomide on TNF-induced activation of NF-κB in lymphocytes. Human lymphocytes were isolated from fresh blood and 2 × 106cells/ml were preincubated for 2 h at 37°C with 10 or 25 μM leflunomide, followed by TNF (0.1 nM) for 30 min, and then tested for NF-κB activation, as described in Materials and Methods.

Leflunomide inhibits TNF-induced IKK-β activation

Because TNF-induced phosphorylation of I-κBα is mediated through IKK-β, these results suggest that leflunomide must inhibit IKK-β activation. Therefore, we investigated the effect of leflunomide on TNF-induced IKK-β activation. As shown in Fig. 2⇑B (upper panel), in the immune complex kinase assays, TNF activated IKK-β in a time-dependent manner and leflunomide treatment completely suppressed the activation. Under these conditions, leflunomide had no effect on the IKK-α (middle panel) and IKK-β (lower panel) protein levels.

Leflunomide inhibits TNF-induced NF-κB activation in normal cells

All the experiments described above were performed with the Jurkat cell lines. Previously, we have shown that leflunomide also blocks TNF-induced NF-κB activation in U-937 cells (myeloid) and epithelial (HeLa) and glioma (H4) cells (11). Whether leflunomide also affects NF-κB in normal cells was examined. As shown in Fig. 2⇑C, TNF activated NF-κB by 4-fold in normal human PBLs and the pretreatment with leflunomide abolished the activation in a dose-dependent manner. These results suggest that the suppressive effect of leflunomide is not restricted to tumor cells.

Leflunomide inhibits TNF-induced AP-1 activation

Previously, we have shown that leflunomide has no effect on the constitutive levels of AP-1 (11). TNF is also one of the most potent activators of AP-1 (34). TNF induced AP-1 expression by 5-fold in Jurkat cells at 1 nM concentration (Fig. 3⇓A). The activation of AP-1 was completely inhibited by leflunomide in a concentration-dependent manner, with maximum suppression occurring at 5 μM (Fig. 3⇓A). Supershift analysis with specific Abs against c-fos and c-jun indicates that TNF-induced AP-1 consisted of fos and jun (Fig. 3⇓B). The lack of supershift by unrelated Abs and disappearance of the AP-1 band by competition with unlabeled oligonucleotide indicate the inhibition was specific (Fig. 3⇓B).

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

Leflunomide inhibits TNF-dependent AP-1 activation. A, Jurkat cells (2 × 106) were pretreated with the indicated concentrations of leflunomide for 2 h at 37°C. Then cells were stimulated with 1 nM TNF for 1 h and assayed for AP-1, as described in Materials and Methods. B, Supershift and specificity of AP-1 activation. Nuclear extracts were prepared from untreated or TNF (1 nM)-treated Jurkat cells (2 × 106/ml), incubated for 15 min with either different Abs or unlabeled AP-1 oligo, and then assayed for AP-1 by EMSA, as described in Materials and Methods. C, Leflunomide inhibits TNF-induced c-Jun-kinase activation. Jurkat cells were pretreated with different concentrations of leflunomide, as indicated, for 2 h and then stimulated with 1 nM TNF at 37°C for 10 min. Thereafter, the cells were washed, pellets were extracted, and c-Jun-kinase activation was detected from the extract, as described in Materials and Methods.

Leflunomide inhibits TNF-induced JNK activation

Previously, we have shown that leflunomide blocks TNF-induced mitogen-activated protein/extracellular signal-related kinase kinase activation (11). JNK, the downstream kinase, is known to be required for the activation of AP-1 (35, 36, 37). Whether JNK is also modulated by leflunomide was examined. Jurkat cells were pretreated with different concentrations of leflunomide for 2 h and then stimulated with TNF (1 nM) for 10 min; activation of JNK was then measured. TNF activated JNK by about 13-fold, an activation that gradually decreased with increasing concentrations of leflunomide. A 10 μM concentration of leflunomide inhibited most of the JNK induced by TNF (Fig. 3⇑C). Thus, it is possible that leflunomide blocks TNF-induced AP-1 activation through suppression of JNK activation.

Leflunomide blocks TNF-induced cytotoxicity and caspase activation

Among the cytokines, TNF is one of the most potent inducers of apoptosis (for references, see Ref. 38). Whether leflunomide modulates TNF-induced apoptosis was also investigated. Jurkat cells were treated with various concentrations of leflunomide either in the absence or presence of TNF and then examined for cytotoxicity by the MTT method (Fig. 4⇓A). TNF was cytotoxic to Jurkat cells, and leflunomide abolished TNF-induced cytotoxicity in a dose-dependent manner, reaching complete inhibition at 10 μM leflunomide. We also examined the cytotoxic effect of various concentrations of TNF either in the absence or presence of leflunomide (Fig. 4⇓B). The cytotoxic effects of TNF in Jurkat cells were dose dependent, with almost 60% killing occurring at 0.5 nM concentration of the cytokine. This cytotoxicity was nearly completely inhibited at all TNF doses by treatment of cells with 10 μM leflunomide.

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

Leflunomide blocks TNF-induced cytotoxicity and PARP degradation. A, Jurkat cells were treated with different concentrations of leflunomide for 2 h at 37°C and then incubated with 1 nM TNF for 72 h at 37°C, in a CO2 incubator. Thereafter, cell viability was examined by the MTT method, as described in Materials and Methods, and expressed as mean OD of triplicate assays. B, Jurkat cells were treated with 10 μM leflunomide for 2 h at 37°C and then incubated with different concentrations of TNF for 72 h at 37°C in a CO2 incubator. Thereafter, cell viability was examined by the MTT method. C, Jurkat cells were incubated with different concentrations of leflunomide for 2 h at 37°C, CO2 incubator and then treated with 2 μg/ml cycloheximide and 1 nM TNF for 2 h at 37°C and washed. The pellet was extracted, and Western blot conducted using anti-PARP mAb. The bands were located at 116 and 80 kDa.

Because the cytotoxic effects of TNF are mediated through the activation of caspases, we also examined the effect of leflunomide on TNF-induced caspase activation. Activated caspase-2, -3, and -7 are known to cleave PARP protein. As shown in Fig. 4⇑C, TNF induced complete cleavage of PARP, and this cleavage was inhibited in a dose-dependent manner by treatment of cells with leflunomide, with maximum effect at 10 μM concentration. Thus, leflunomide also blocked TNF-induced apoptosis.

Leflunomide blocks TNF-induced ROI generation and lipid peroxidation

Previous reports have shown that TNF activates NF-κB, AP-1, JNK, and apoptosis through generation of ROI (27, 39, 40, 41). Whether leflunomide suppresses TNF signaling through suppression of ROI generation was examined by flow cytometry. As shown in Fig. 5⇓A, TNF induced ROI generation in a time-dependent manner, and this was suppressed on pretreatment of cells with leflunomide. Because lipid peroxidation has also been implicated in TNF signaling (42), we also examined the effect of leflunomide on TNF-induced lipid peroxidation. Results in Fig. 5⇓B show that TNF induced lipid peroxidation in Jurkat cells, and it was completely suppressed by leflunomide. Thus, it is quite likely that leflunomide blocks TNF signaling through suppression of ROI generation and lipid peroxidation. Although earliest time we examined ROI generation is at 1 h, whereas NF-κB and AP-1 activation can be seen at 15 min, it is possible that low levels of ROI generated at early times are either not detectable or other mechanisms are involved in TNF signaling.

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

Leflunomide inhibits TNF-induced ROI generation (A) and lipid peroxidation (B). For A, Jurkat cells (5 × 105/ml) were treated with 10 μM leflunomide for 2 h and then exposed to TNF (0.1 nM) for indicated times at 37°C in a CO2 incubator. ROI production was then determined by the flow cytometry method, as described in Materials and Methods. The results shown are representative of two independent experiments. For B, Jurkat cells (3 × 106 in 1 ml) were pretreated with different concentrations of leflunomide for 2 h and then incubated with 1 nM TNF for 1 h and assayed for lipid peroxidation, as described in Materials and Methods.

Uridine reverses the suppressive effects of leflunomide on TNF signaling

The antiproliferative effects of leflunomide on B and T cells can be reversed by uridine, suggesting the critical role of DHODH (12, 15). To determine whether uridine reverses the suppression of TNF-induced NF-κB activation in Jurkat cells, we pretreated cells with leflunomide in the presence of various concentrations of uridine and then treated them with TNF. As shown in Fig. 6⇓A, TNF-induced NF-κB activation was suppressed by leflunomide, and uridine reversed the suppression in a dose-dependent manner. Uridine or leflunomide by themselves did not activate NF-κB, and uridine alone had minimal effect on TNF-mediated NF-κB activation. Thus, leflunomide mediated its effects on TNF-induced NF-κB activation by interfering with the pyrimidine biosynthesis pathway. We also examined the effect of uridine on the leflunomide-induced suppression of TNF-mediated cytotoxicity. As shown in Fig. 6⇓B, 50 μM uridine completely reversed the effects of leflunomide on the cytotoxicity caused by TNF. Thus, pyrimidine biosynthesis plays a critical role in the TNF signaling.

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

Uridine reverses leflunomide-mediated inhibition of NF-κB activation and cytotoxicity induced by TNF. A, Jurkat cells (2 × 106/ml) were pretreated with different concentrations of uridine for 1 h and then treated with 10 μM leflunomide for 2 h at 37°C. Thereafter, cells were activated with TNF (100 pM) for 30 min, and nuclear extracts prepared and analyzed by EMSA. B, Jurkat cells (5 × 103/0.1 ml) were pretreated with 50 μM uridine for 1 h and then treated with 10 μM leflunomide for 2 h at 37°C. Thereafter, cells were incubated with different concentrations of TNF for 72 h and examined for cell viability by the MTT method, as described in Materials and Methods.

Leflunomide has no effect on TNFR

Because leflunomide blocked a variety of signals activated by TNF, it is possible that leflunomide down-regulated TNFR. Therefore, we examined the effect of leflunomide on TNFR by receptor-binding assays. TNF bound to Jurkat cells, and this binding was completely unaffected by pretreatment of cells with leflunomide (data not shown), suggesting that the effects of leflunomide were not due to down-regulation of TNFR.

Discussion

The aim of the present studies was to investigate whether leflunomide modulates TNF-mediated cellular responses. Our results show that leflunomide blocked TNF-induced NF-κB activation, I-κBα phosphorylation, and activation of IKK, JNK, and AP-1, and suppressed TNF-induced apoptosis. None of these effects were mediated through the down-regulation of TNFR. Leflunomide suppressed TNF-induced ROI generation and lipid peroxidation. The suppressive effects of leflunomide on TNF signaling could be reversed by uridine.

There are several ways to explain how leflunomide might inhibit TNF-induced NF-κB activation. One is by suppressing TNF-induced phosphorylation of I-κBα. The phosphorylation of I-κBα is regulated by a large number of kinases, including IKK-α, IKK-β, IKK-γ, NIK, TGF-β-activated kinase-1, AKT, and mitogen-activated protein/extracellular signal-related kinase kinase kinase 1 (MEKK1), MEKK2, and MEKK3 (43). However, only IKK-β mediates TNF-induced phosphorylation of I-κBα at positions 32 and 36. Thus, leflunomide may suppress NF-κB activation through suppression of IKK-β. That leflunomide blocked TNFR1-, TRAF2-, and NIK-induced NF-κB-mediated reporter gene expression, but not that activated by p65 also suggests that the site of action of leflunomide is downstream from NIK. The latter is known to activate IKK-β, thus leading to I-κBα phosphorylation.

Our results indicate that leflunomide also blocked TNF-induced JNK and AP-1 activation. TRAF2, which is known to bind to TNFR through TNFR-associated death domain, is also required for AP-1 and JNK activation (31). The suppression of TRAF2 activity may explain how leflunomide inhibits NF-κB, AP-1, and JNK. While MEKK1, MEKK2, and MEKK3 have been implicated in NF-κB activation, MEKK4 activates JNK (44). There are some reports that indicate that AKT and NIK activate IKK-α, whereas MEKK1 and atypical PKC activate IKK-β (for references, see Ref. 45). Thus, it is possible that leflunomide inhibited I-κBα phosphorylation by inhibiting the activity of IKK-β.

Our results also indicate that leflunomide is a potent inhibitor of AP-1. This is not too surprising because most agents that activate NF-κB also activate AP-1. Activation of AP-1 requires JNK, another kinase of the mitogen-activated protein kinase family, so it is possible that AP-1 is suppressed through inhibition of JNK. Recent studies from our laboratory showed that overexpression of cells with either superoxide dismutase (27) or with γ-glutamylcysteine synthetase, a rate-limiting enzyme in the glutathione biosynthesis pathway (40), blocked both NF-κB and AP-1 activation induced by TNF, indicating a similar mechanism of activation of both transcription factors. These results also suggest that leflunomide may suppress these factors by regulating the redox status of the cells. We did indeed find that leflunomide blocks TNF-induced ROI production and lipid peroxidation.

TNF-induced cyotoxicity and caspase activation were also blocked by leflunomide. Because NF-κB activation has been shown to play an antiapoptotic role (59), the suppression of apoptosis by leflunomide may seem paradoxical. The overexpression of the antioxidant enzymes manganous superoxide dismutase or γ-glutamylcysteine synthetase has also has been shown to suppress TNF-induced apoptosis and NF-κB (27, 40), suggesting that the mechanisms of activation of apoptosis and NF-κB are very similar. Our discovery that leflunomide blocks TNF-induced ROI generation and lipid peroxidation may explain the mechanism by which leflunomide exerts some of its effects.

The suppressive effects of leflunomide on TNF signaling were reversible by uridine. These results are consistent with previous reports (12, 15). Leflunomide is a potent inhibitor of DHODH, a rate-limiting enzyme in the biosynthesis pathway of pyrimidine (12, 13, 14, 15). In vitro the Ki of inhibition of DHODH by leflunomide ranges from 179 nM to 2.7 μM (13, 14). The ability of leflunomide to suppress proliferation of T and B cells (2, 3, 4) has been suggested to be due to inhibition of DHODH (12, 13, 14, 15), a pathway critical for the proliferation of these cells. Our results are the first to demonstrate the critical role of this pathway in TNF signaling.

We found that leflunomide blocked NF-κB-dependent reporter gene expression. Several genes that are involved in RA are regulated by NF-κB (19). These include inflammatory cytokines, cyclooxygenase-2, metalloproteinases, urinary plasminogen activator, NO synthase, and cell surface adhesion molecules (47, 48, 49, 50, 51, 52). Thus, it is possible that leflunomide mediates its effects against RA through suppression of NF-κB-regulated genes. Indeed, the modulation by leflunomide of cyclooxygenase-2, NO synthase, inflammatory cytokines, and their receptors has recently been reported (53, 54, 55). Because NF-κB-regulated gene products have also been implicated in tumorigenesis, leflunomide may prove useful in suppressing tumorigenesis (56). Indeed, in vivo antitumor activity against C6 glioma has been recently assigned to leflunomide (57).

Adenoviral I-κBα, an NF-κB inhibitor, has been used for the treatment of RA and tumorigenesis (58, 59). Because leflunomide lacks delivery problems, the suppression of NF-κB activation by leflunomide is preferable. Leflunomide is also preferable over soluble TNFR or anti-TNF Abs, because formation of Abs against either of the proteins can have serious side effects. Adenovirus-enforced overexpression of mitochondrial superoxide dismutase has also been used as gene therapy for ischemia/reperfusion injury of the liver through the down-regulation of NF-κB and AP-1 activation (60). Our results indicate that suppressive effects of leflunomide on NF-κB and AP-1 activation and on other TNF-mediated cellular responses may have protective effects on liver and against cardiovascular diseases.

Acknowledgments

We thank Walter Pagel for critically reading this manuscript.

Footnotes

  • ↵1 This research was supported by The Clayton Foundation for Research.

  • ↵2 Address correspondence and reprint requests to Dr. Bharat B. Aggarwal, Cytokine Research Laboratory, Department of Bioimmunotherapy, Box 143, 1515 Holcombe Boulevard, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030. E-mail address: aggarwal{at}utmdacc.mda.uth.tmc.edu

  • ↵3 Abbreviations used in this paper: DHODH, dihydroorotate dehydrogenase; ALLN, N-acetyl leucyl leucyl nonleucinal; I-κB, inhibitory subunit of NF-κB; IKK, I-κBα kinase; JAK, Janus kinase; JNK, c-jun N-terminal protein kinase; MDA, malondialdehyde; MEKK, mitogen-activated protein/extracellular signal-related kinase kinase kinase; NIK, NF-κB-inducing kinase; PARP, poly(ADP-ribose) polymerase; RA, rheumatoid arthritis; ROI, reactive oxygen intermediate; SEAP, secretory alkaline phosphatase; TRAF, TNFR-associated factor.

  • Received April 7, 2000.
  • Accepted August 21, 2000.
  • Copyright © 2000 by The American Association of Immunologists

References

  1. ↵
    Bartlett, R. R., R. Schleyerbach. 1985. Immunopharmacological profile of a novel isoxazole derivative, HWA486, with potential antirheumatic activity. I. Disease modifying action on adjuvant arthritis in rat. Int. J. Immunopharmacol. 7: 7
    OpenUrlCrossRefPubMed
  2. ↵
    Siemasko, K. F., A. S. F. Chong, J. W. Williams, E. G. Bremer, A. Finnegan. 1996. Regulation of B cell function by the immunosuppressive agent leflunomide. Transplantation 61: 635
    OpenUrlCrossRefPubMed
  3. ↵
    Chog, A. S.-F., K. Rezai, H. M. Gebel, A. Finnegan, P. Foster, X.-L. Xu, J. W. Williams. 1996. Effects of leflunomide and other immunosuppressive agents on T cell proliferation in vitro. Transplantation 61: 140
    OpenUrlCrossRefPubMed
  4. ↵
    Cherwinski, H. M., R. G. Cohn, P. Cheung, D. J. Webster, Y.-Z. Xu, J. P. Caulfield, J. M. Young, G. Nakano, J. T. Ransom. 1995. The immunosuppressant leflunomide inhibits lymphocyte proliferation by inhibiting pyrimidine biosynthesis. J. Pharmacol. Exp. Ther. 275: 1043
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Lin, Y., H. Sobis, M. Vandeputte, M. Waer. 1995. Mechanism of leflunomide-induced prevention of xenoantibody formation and xenograft rejection in the hamster to rat heart transplantation model. Transplant. Proc. 27: 305
    OpenUrlPubMed
  6. ↵
    Xu, X.-L., J. M. Williams, E. G. Bremer, A. Finnegan, A. S. F. Chong. 1995. Inhibition of tyrosine phosphorylation in T cells by a novel immunosuppressive agent, leflunomide. J. Biol. Chem. 270: 12398
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Xiao, F., A. S. F. Chong, J. Shen, J. Yang, J. Short, P. Foster, H. Sankary, S. Jensik, D. Mital, L. McChesney, G. Koukoulis, J. W. Williams. 1995. Pharmacologically induced regression of chronic transplant rejection. Transplantation 60: 1065
    OpenUrlPubMed
  8. ↵
    Xiao, F., A. S. F. Chong, P. Foster, H. Sankary, J. L. McChesney, G. Koukoulis, J. Yang, D. Frieders, J. W. Williams. 1994. Leflunomide controls rejection in hamster to rat cardiac xenografts. Transplantation 58: 828
    OpenUrlCrossRefPubMed
  9. ↵
    Chong, A. S.-F., J. Shen, F. Xiao, L. Blinder, L. Wei, H. Sankary, P. Foster, J. Williams. 1996. Delayed xenograft rejection in the concordant hamster heart to Lewis rat model. Transplantation 62: 90
    OpenUrlPubMed
  10. ↵
    Elder, R. T., X. Xu, J. W. Williams, H. Gong, A. Finnegan, A. S. F. Chong. 1997. The immunosuppressive metabolite of leflunomide, A771726, affects murine T cells through two biochemical mechanisms. J. Immunol. 159: 22
    OpenUrlAbstract
  11. ↵
    Manna, S., B. B. Aggarwal. 1999. Immunosuppressive leflunomide metabolite (A77 1726) blocks TNF-dependent nuclear factor-κB activation and gene expression. J. Immunol. 162: 2095
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Xu, X., J. W. Williams, H. Gong, A. Finnegan, A. S. F. Chong. 1996. Two activities of immunosuppressive metabolite of leflunomide, A771726. Biochem. Pharmacol. 52: 527
    OpenUrlCrossRefPubMed
  13. ↵
    Davis, J. P., G. A. Cain, W. J. Pitts, R. L. Magolda, R. A. Copeland. 1996. The immunosuppressive metabolite of leflunomide is a potent inhibitor of human dihydroorotate dehydrogenase. Biochemistry 35: 1270
    OpenUrlCrossRefPubMed
  14. ↵
    Greene, S., K. Watanabe, J. Braatz-Trulson, L. Lou. 1995. Inhibition of dihydroorotate dehydrogenase by the immunosuppressive agent leflunomide. Biochem. Pharmacol. 50: 861
    OpenUrlCrossRefPubMed
  15. ↵
    Williamson, R. A., C. M. Yea, P. A. Robson, A. P. Curnock, S. Gadher, A. B. Hambleton, K. Woodward, J.-M. Bruneau, P. Hambleton, D. Moss, et al 1995. Dihydroorotate dehydrogenase is a high affinity binding protein for A771726 and mediator of a range of biological effects of the immunomodulatory compound. J. Biol. Chem. 270: 22467
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Xu, X., L. Blinder, J. Shen, H. Gong, A. Finnegan, J. W. Williams, A. S. F. Chong. 1997. In vivo mechanism by which leflunomide controls lymphoproliferative and autoimmune disease in MRL/MpJ-lpr/lpr mice. J. Immunol. 159: 167
    OpenUrlAbstract
  17. ↵
    Matter, T., K. Kochhar, R. Bartlett, E. G. Bremer, A. Finnegan. 1993. Inhibition of the epidermal growth factor receptor tyrosine kinase activity by leflunomide. FEBS Lett. 334: 161
    OpenUrlCrossRefPubMed
  18. ↵
    Siemasko, K., A. S. F. Chong. H. M. Jack, H. Gong, J. W. Williams, A. Finnegan. 1998. Inhibition of JAK3 and STAT6 tyrosine phosphorylation by the immunosuppressive drug leflunomide leads to a block in IgG1 production. J. Immunol. 160: 1581
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Smolen, J. S., J. R. Kalden, D. L. Scott, B. Rozman, T. K. Kvien, A. Larsen, I. Loew-Friedrich, C. Oed, R. Rosenburg. 1999. Efficacy and safety of leflunomide compared with placebo and sulphasalazine in active rheumatoid arthritis: a double-blind, randomized, multicenter trial: European Leflunomide Study Group. Lancet 353: 259
    OpenUrlCrossRefPubMed
  20. ↵
    Feldmann, M., M. J. Elliott, J. N. Woody, R. N. Maini. 1997. Antitumor necrosis factor therapy of rheumatoid arthritis. Adv. Immunol. 64: 283
    OpenUrlCrossRefPubMed
  21. ↵
    Maini, R. N., F. C. Breedveld, J. R. Kalden, J. S. Smolen, D. Davis, J. D. Macfarlane, C. Antoni, B. Leeb, M. J. Elliott, J. N. Woody, et al 1998. Randomized placebo-controlled trial of multiple intravenous infusions of anti-TNF monoclonal antibody with or without weekly methotrexate in rheumatoid arthritis. Arthritis Rheum. 41: 1552
    OpenUrlCrossRefPubMed
  22. ↵
    Moreland, L. W., S. W. Baumgartner, M. H. Schiff, E. A. Tindall, R. M. Fleischmann, A. L. Weaver, R. E. Ettlinger, S. Cohen, W. J. Koopman, K. Mohler, et al 1997. Treatment of rheumatoid arthritis with a recombinant human tumor necrosis factor receptor (p75)-Fc fusion protein. N. Engl. J. Med. 337: 141
    OpenUrlCrossRefPubMed
  23. ↵
    Malinin, N. L., M. P. Boldin, A. V. Kovalenko, D. Wallach. 1997. MAP3K-related kinase involved in NF-κB induction by TNF, CD95 and IL-1. Nature 385: 540
    OpenUrlCrossRefPubMed
  24. ↵
    Darnay, B., J. Ni, P. A. Moore, B. B. Aggarwal. 1999. Activation of NF-κB by RANK requires TRAF6 and NF-κB-inducing kinase (NIK): identification of a novel TRAF6 interaction motif. J. Biol. Chem. 274: 7724
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Chaturvedi, M. M., R. LaPushin, B. B. Aggarwal. 1994. Tumor necrosis factor and lymphotoxin: qualitative and quantitative differences in the mediation of early and late cellular responses. J. Biol. Chem. 269: 14575
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Manna, S. K., A. Mukhopadhyay, N. T. Van, B. B. Aggarwal. 1999. Silymarin suppresses TNF-induced activation of nuclear transcription factor-κB, c-Jun N-terminal kinase and apoptosis. J. Immunol. 163: 6800
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Hu, M. C.-T., Y.-P. Wang, W. R. Qui, A. Mikhail, C. F. Meyer, T.-H. Tan. 1999. Hematopoietic progenitor kinase (HPK1) stress response signaling pathway activates IκB kinases (IKK-a/b) and IKK-b is a developmentally regulated protein kinase. Oncogene 18: 5514
    OpenUrlCrossRefPubMed
  28. ↵
    Manna, S. K., H. J. Zhang, T. Yan, L. W. Oberley, B. B. Aggarwal. 1998. Overexpression of Mn-superoxide dismutase suppresses TNF induced apoptosis and activation of nuclear transcription factor-κB and activated protein-1. J. Biol. Chem. 273: 13245
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Kumar, A., B. B. Aggarwal. 1999. Assay for redox sensitive kinases. L. Packer, ed. In Methods Enzymology Vol. 300: 339 Academic Press, New York.
    OpenUrlCrossRefPubMed
  30. ↵
    Haridas, V., B. G. Darnay, K. Natarajan, R. Heller, B. B. Aggarwal. 1998. Overexpression of the p80 form of the TNF receptor induces apoptosis, NF-κB activation and c-Jun kinase activation: comparison with the endogenous receptor. J. Immunol. 160: 3152
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Hsu, H., H. B. Shu, M. G. Pan, D. V. Goeddel. 1996. TRADD-TRAF2 and TRADD-FADD interactions define two distinct TNF receptor 1 signal transduction pathways. Cell 84: 299
    OpenUrlCrossRefPubMed
  32. ↵
    Simeonidis, S., D. Stauber, G. Chen, W. A. Hendrickson, D. Thanos. 1999. Mechanisms by which IκB proteins control NF-κB activity. Proc. Natl. Acad. Sci. USA 96: 49
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Whiteside, S. T., M. K. Ernst, O. LeBail, C. Laurent-Winter, N. R. Rice, A. Israel. 1995. N- and C-terminal sequences control degradation of MAD3/IκBα in response to inducers of NF-κB activity. Mol. Cell. Biol. 15: 5339
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Karin, M., Z. G. Liu, E. Zandi. 1997. AP-1 function and regulation. Curr. Opin. Cell Biol. 9: 240
    OpenUrlCrossRefPubMed
  35. ↵
    Karin, M.. 1995. The regulation of AP-1 activity by mitogen-activated protein kinases. J. Biol. Chem. 270: 16483
    OpenUrlFREE Full Text
  36. ↵
    Karin, M., M. Delhase. 1998. JNK or IKK, AP-1 or NF-κB, which are the targets for MEK kinase 1 action?. Proc. Natl. Acad. Sci. USA 95: 9067
    OpenUrlFREE Full Text
  37. ↵
    Lee, F. S., J. Hagler, Z. J. Chen, T. Maniatis. 1997. Activation of the IκBα kinase complex by MEKK1, a kinase of the JNK pathway. Cell 88: 213
    OpenUrlCrossRefPubMed
  38. ↵
    Rath, P. C., B. B. Aggarwal. 1999. TNF-induced signaling in apoptosis. J. Clin. Immunol. 19: 350
    OpenUrlCrossRefPubMed
  39. ↵
    Lo, Y. Y. C., J. M. S. Wong, T. F. Cruz. 1996. Reactive oxygen species mediate cytokine activation of c-Jun NH2-terminal kinases. J. Biol. Chem. 271: 15703
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Manna, S. K., M. T. Kuo, B. B. Aggarwal. 1999. Overexpression of γ-glutamylcysteine synthetase abolishes tumor necrosis factor-induced apoptosis and activation of nuclear transcription factor-κB and activator protein-1. Oncogene 18: 4371
    OpenUrlCrossRefPubMed
  41. ↵
    Li, N., M. Karin. 1999. Is NF-κB the sensor of oxidative stress?. FASEB J. 13: 1137
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Bowie, A. G., P. N. Moynagh, L. A. J. O’Neill. 1997. Lipid peroxidation is involved in the activation of NF-κB by tumor necrosis factor but not interleukin-1 in the human endothelial cell line ECV304: lack of involvement of H2O2 in NF-κB activation by either cytokine in both primary and transformed endothelial cells. J. Biol. Chem. 272: 25941
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Karin, M.. 1999. How NF-B is activated: the role of the IB kinase (IKK) complex. Oncogene 18: 6867
    OpenUrlCrossRefPubMed
  44. ↵
    Zhao, Q., F. S. Lee. 1999. Mitogen-activated protein kinase/ERK kinase kinases 2 and 3 activate nuclear factor-κB through IκB kinase-α and IκB kinase-β. J. Biol. Chem. 274: 8355
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Mercurio, F., A. M. Manning. 1999. Multiple signals converging on NF-κB. Curr. Opin. Cell Biol. 11: 226
    OpenUrlCrossRefPubMed
  46. Baichwal, V. R., P. A. Baeuerle. 1997. Activate NF-κB or die?. Curr. Biol. 7: R94
    OpenUrlCrossRefPubMed
  47. ↵
    Hwang, D., B. C. Jang, G. Yu, M. Boudreau. 1997. Expression of mitogen-inducible cyclooxygenase induced by lipopolysaccharide: mediation through both mitogen-activated protein kinase and NF-κB signaling pathways in macrophages. Biochem. Pharmacol. 54: 87
    OpenUrlCrossRefPubMed
  48. ↵
    Von Knethen, A., D. Callsen, B. Brune. 1999. Superoxide attenuates macrophage apoptosis by NF-κB and AP-1 activation that promotes cyclooxygenase-2 expression. J. Immunol. 163: 2858
    OpenUrlAbstract/FREE Full Text
  49. ↵
    Taylor, B. S., M. E. de Vera, R. W. Ganster, Q. Wang, R. A. Shapiro, S. M. Morris, Jr, T. R. Billiar, D. A. Geller. 1998. Multiple NF-κB enhancer elements regulate cytokine induction of the human inducible nitric oxide synthase gene. J. Biol. Chem. 273: 15148
    OpenUrlAbstract/FREE Full Text
  50. ↵
    Sato, H., M. Seiki. 1993. Regulatory mechanism of 92 kDa type IV collagenase gene expression which is associated with invasiveness of tumor cells. Oncogene 8: 395
    OpenUrlPubMed
  51. ↵
    Collins, T., M. A. Read, A. S. Neish, M. Z. Whitley, D. Thanos, T. Maniatis. 1995. Transcriptional regulation of endothelial cell adhesion molecules: NF-κB and cytokine-inducible enhancers. FASEB J. 9: 899
    OpenUrlAbstract
  52. ↵
    Iademarco, M. F., J. J. McQuillan, G. D. Rosen, D. C. Dean. 1992. Characterization of the promoter for vascular cell adhesion molecule-1 (VCAM-1). J. Biol. Chem. 267: 16323
    OpenUrlAbstract/FREE Full Text
  53. ↵
    Hamilton, L. C., I. Vojnovic, T. D. Warner. 1999. A771726, the active metabolite of leflunomide, directly inhibits the activity of cyclo-oxygenase-2 in vitro and in vivo in a substrate-sensitive manner. Br. J. Pharmacol. 127: 1589
    OpenUrlCrossRefPubMed
  54. ↵
    Mirmohammadsadegh, A., B. Homey, H. F. Abts, K. Kohrer, T. Ruzicka, G. Michel. 1998. Differential modulation of pro- and anti-inflammatory cytokine receptors by N-(4-trifluoromethylphenyl)-2-cyano-3-hydroxy-crotonic acid amide (A77 1726), the physiologically active metabolite of the novel immunomodulator leflunomide. Biochem. Pharmacol. 55: 1523
    OpenUrlCrossRefPubMed
  55. ↵
    Deage, V., D. Burger, J. M. Dayer. 1998. Exposure of T lymphocytes to leflunomide but not to dexamethasone favors the production by monocytic cells of interleukin-1 receptor antagonist and the tissue-inhibitor of metalloproteinases-1 over that of interleukin-1β and metalloproteinases. Eur. Cytokine Network 9: 663
    OpenUrlPubMed
  56. ↵
    Waddick, K. G., F. M. Uckun. 1999. Innovative treatment programs against cancer. II. Nuclear factor-κB (NF-κB) as a molecular target. Biochem. Pharmacol. 57: 9
    OpenUrlCrossRefPubMed
  57. ↵
    Xu, X., J. Shen, J. W. Mall, J. A. Myers, W. Huang, L. Blinder, T. J. Saclarides, J. W. Williams, A. S. Chong. 1999. In vitro and in vivo antitumor activity of a novel immunomodulatory drug, leflunomide: mechanisms of action. Biochem. Pharmacol. 58: 1405
    OpenUrlCrossRefPubMed
  58. ↵
    Foxwell, B., K. Browne, J. Bondeson, C. Clarke, R. de Martin, F. Brennan, M. Feldmann. 1998. Efficient adenoviral infection with IκBα reveals that macrophage tumor necrosis factor α production in rheumatoid arthritis is NF-κB dependent. Proc. Natl. Acad. Sci. USA 95: 8211
    OpenUrlAbstract/FREE Full Text
  59. ↵
    Wang, C. Y., J. C. Cusack, Jr, R. Liu, A. S. Baldwin, Jr. 1999. Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-κB. Nat. Med. 5: 412
    OpenUrlCrossRefPubMed
  60. ↵
    Epperly, M., J. Bray, S. Kraeger, R. Zwacka, J. Engelhardt, E. Travis, J. Greenberger. 1998. Prevention of late effects of irradiation lung damage by manganese superoxide dismutase gene therapy. Gene Ther. 5: 196
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

The Journal of Immunology: 165 (10)
The Journal of Immunology
Vol. 165, Issue 10
15 Nov 2000
  • Table of Contents
  • About the Cover
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.
Leflunomide Suppresses TNF-Induced Cellular Responses: Effects on NF-κB, Activator Protein-1, c-Jun N-Terminal Protein Kinase, and Apoptosis
(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
Leflunomide Suppresses TNF-Induced Cellular Responses: Effects on NF-κB, Activator Protein-1, c-Jun N-Terminal Protein Kinase, and Apoptosis
Sunil K. Manna, Asok Mukhopadhyay, Bharat B. Aggarwal
The Journal of Immunology November 15, 2000, 165 (10) 5962-5969; DOI: 10.4049/jimmunol.165.10.5962

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Leflunomide Suppresses TNF-Induced Cellular Responses: Effects on NF-κB, Activator Protein-1, c-Jun N-Terminal Protein Kinase, and Apoptosis
Sunil K. Manna, Asok Mukhopadhyay, Bharat B. Aggarwal
The Journal of Immunology November 15, 2000, 165 (10) 5962-5969; DOI: 10.4049/jimmunol.165.10.5962
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
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Differential Susceptibility to Staphylococcal Superantigen (SsAg)-Induced Apoptosis of CD4+ T Cells from Atopic Dermatitis Patients and Healthy Subjects: The Inhibitory Effect of IL-4 on SsAg-Induced Apoptosis
  • HIV-1 Vaccination Administered Intramuscularly Can Induce Both Systemic and Mucosal T Cell Immunity in HIV-1-Uninfected Individuals
  • Osteopontin (Eta-1) and Fibroblast Growth Factor-2 Cross-Talk in Angiogenesis
Show more Clinical Immunology

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