Elsevier

Biochemical Pharmacology

Volume 59, Issue 1, 1 January 2000, Pages 7-11
Biochemical Pharmacology

BCP Symposium Presentation
Cell type-specific role for reactive oxygen species in nuclear factor-kappaB activation by Interleukin-1

Presented at Oxford, 10–12 June 1999.
https://doi.org/10.1016/S0006-2952(99)00290-7Get rights and content

Abstract

The role of reactive oxygen intermediates (ROIs) in nuclear factor-kappaB (NF-κB) activation remains a matter of controversy. We have studied whether ROIs played any role in NF-κB induction by interleukin-1β (IL-1β) in different cell types. Our studies indicated three different pathways. IL-1β stimulation of lymphoid cells generates ROIs, which are required for IκB-α degradation and NF-κB activation. The source of these ROIs is the 5-lipoxygenase (5-LOX) enzyme. In monocytic cells, ROIs are also produced in response to IL-1β and necessary for NF-κB induction, but their source appears to be the NADPH oxidase complex. Finally, epithelial cells do not generate ROIs after IL-1β stimulation, but do rapidly activate NF-κB. Interestingly, transfection of epithelial cells with the 5-LOX and 5-LOX activating protein expression vectors restored ROI production and ROI-dependent NF-κB activation in response to IL-1β. Our data thus indicate that ROIs are cell type-specific second messengers for NF-κB induction by IL-1β.

Section snippets

Lymphoid cells

Stimulation of lymphoid cells (70Z/3, Raji, or EL-4 cell lines) with IL-1β generates ROIs and rapidly induces nuclear NF-κB DNA-binding activity. We could demonstrate that ROI production was required for IL-1β-induced NF-κB DNA binding, IκB-α degradation, and NF-κB-dependent transactivation, as these activities were blocked by the antioxidants NAC and PDTC. We then identified 5-LOX as the main source of ROIs in these cells after IL-1β treatment. Indeed, the above-mentioned lymphoid cells

Monocytic cells

Stimulation of monocytic cells (U937 and THP-1 cell lines) with IL-1β also generates ROIs and potently activates NF-κB. As in lymphoid cells, preincubation of these cells with the antioxidants NAC and PDTC inhibits both the production of ROIs and NF-κB nuclear activity, thus indicating that the pathway leading to IκB-α degradation requires ROI production. However, the source of ROIs was different than in lymphoid cells. Indeed, the monocytic cell lines we analyzed do not express the 5-LOX

Epithelial cells

In the various epithelial cell lines we explored (MCF7 A/Z, OVCAR-3, HCT116, and SKOV-3), IL-1β induced a strong NF-κB nuclear activity, but did not activate ROI production. Therefore, NF-κB activation and IκB-α degradation were not affected by antioxidants in these cells. There are two possible explanations for the lack of ROI production in these cells. Epithelial cells, such as OVCAR-3 cells, express high levels of catalase activity. Moreover, none of the epithelial cell lines we analyzed

Discussion

Although the role of ROIs in NF-κB activation by proinflammatory cytokines and other stimulating agents was reported several years ago, it has become very much a matter of controversy since the cloning of the IKKs. Several authors have recently described a direct pathway for NF-κB activation by IL-1β which links the receptor, receptor-associated adaptor proteins and kinases (MyD88, Il-1 receptor-associated kinase (IRAK), and TNF receptor-associated kinase factors 6 (TRAF6)), the NIK or MEKK-1

Acknowledgements

M-P.M. and V.B. are research associates and J.P. is a research director at the National Fund for Scientific Research (FNRS, Belgium). G.B. is a fellow from the Biotechnology Programme, European Commission.

References (40)

  • P.J. Barnes et al.

    Nuclear factor-κBA pivotal transcription factor in chronic inflammatory diseases

    N Engl J Med

    (1997)
  • M.J. May et al.

    IkappeB kinasesKinsmen with different crafts

    Science

    (1999)
  • K. Brown et al.

    Control of IκB-α proteolysis by site-specific, signal-induced phosphorylation

    Science

    (1995)
  • J.A. DiDonato et al.

    A cytokine-responsive IκB kinase that activates the transcriptional factor NF-κB

    Nature

    (1997)
  • F. Mercurio et al.

    IKK-1 and IKK-2Cytokine-activated IκB kinases essential for NF-κB activation

    Science

    (1997)
  • J.D. Woronicz et al.

    IκB kinase-βNF-κB activation and complex formation with IκB kinase-α and NIK

    Science

    (1997)
  • L. Ling et al.

    NF-κB-inducing kinase activates IKK-α by phosphorylation of Ser-176

    Proc Natl Acad Sci USA

    (1998)
  • N.L. Malinin et al.

    MAP3K-related kinase involved in NF-κB induction by TNF, CD95 and IL-1

    Nature

    (1997)
  • H. Nakano et al.

    Differential regulation of IκB kinase α and β by two upstream kinases, NF-κB-inducing kinase and mitogen-activated protein kinase/ERK kinase kinase-1

    Proc Natl Acad Sci USA

    (1998)
  • Z. Chen et al.

    Signal-induced site-specific phosphorylation targets IκBα to the ubiquitin–proteasome pathway

    Genes Dev

    (1995)
  • Cited by (139)

    • Rheumatoid arthritis: From synovium biology to cell-based therapy

      2022, Cytotherapy
      Citation Excerpt :

      The predicted consequence of such oxygen fluctuations would be the cycles of HIF-1 stabilization and expression of its targets, followed by repetitive bursts of ROS. ROS are involved in NF-κB activation, presumably via effects on upstream kinases, resulting in the enhanced transcription of pro-inflammatory cytokines and anti-apoptotic mediators, many of which are also induced by HIF-1 [79,80.] Because the RA synovium is enriched with different cell types, it is fundamentally important to understand this disease in the context of the pathologic cell-to-cell communication.

    • Biomarkers of high salt intake

      2021, Advances in Clinical Chemistry
      Citation Excerpt :

      TNF-α promotes monocyte chemotaxis toward mesangial cells by regulating the expression of monocyte chemoattractant protein-1 (MCP-1) via a Rho-kinase/p38 mitogen-activated protein kinase (MAPK)-dependent pathway [124]. In addition, ROS activated p38MAPK and the transcription factor nuclear factor kappa B (NF-κB), leading to proinflammatory cytokine release and the accumulation of inflammatory cells in the kidney [125,126]. Thus, sustained inflammatory responses may contribute to the progression of renal injury [127].

    View all citing articles on Scopus
    View full text