Abstract

The NF-ĸB transcription factor is a critical regulator of immune homeostasis and inflammatory responses and is a critical factor in the pathogenesis of inflammatory disease. The pathways to NF-ĸB activation are paradigms for signal-induced ubiquitination and proteasomal degradation, control of transcription factor function by subcellular localisation, and the control of gene transcription and physiological processes by signal transduction mechanisms. Despite the importance of NF-ĸB in disease, the NF-ĸB pathway remains unexploited for the treatment of inflammatory disease. Our understanding of NF-ĸB comes mostly from studies of transgenic mice and cell lines where components of the pathway have been deleted or over expressed. Recent advances in quantitative proteomics offer new opportunities to understand the NF-ĸB pathway using the absolute abundance of individual pathway components. We have analysed available quantitative proteomic datasets to establish the structure of the NF-ĸB pathway in human immune cells under both steady state and activated conditions. This reveals a conserved NF-κB pathway structure across different immune cell lineages and identifies important differences to the current model of the NF-ĸB pathway. These include the findings that the IKK complex in most cells is likely to consist predominantly of IKKβ homodimers, that the relative abundancies of IκB proteins show strong cell type variation, and that the components of the non-canonical NF-ĸB pathway are significantly increased in activated immune cells. These findings challenge aspects of our current view of the NF-κB pathway and identify outstanding questions important for defining the role of key components in regulating inflammation and immunity.

Highlights

  • Since its discovery over 30 years ago, NF-κB has served as a model for inducible transcription factors, signal-directed ubiquitination, and the impact of signal transduction pathways on gene expression and physi­ ological processes [1]

  • To determine the structure of the NF-κB pathway in human immune cells we analysed the quantitative proteomic dataset recently generated by Reickman and colleagues [11]

  • Analysis of IKK component copy numbers across all immune cell types reveals that IKKβ is the most abundant component of the IKK complex, followed by NEMO and IKKα, which is a relatively minor component by abun­ dance (Fig. 2B). This pattern of relative abundance is typified in CD4+ T cells, naïve B cells, monocytes and myeloid dendritic cells (Fig. 2C)

Read more

Summary

Introduction

Since its discovery over 30 years ago, NF-κB has served as a model for inducible transcription factors, signal-directed ubiquitination, and the impact of signal transduction pathways on gene expression and physi­ ological processes [1]. NF-κB dimers are sequestered in the cytoplasm through interac­ tion with the IκB family of proteins that include IκBα, IκBβ and IκBε [4]. Included in this family of NF-κB inhibitory proteins are the p105 and p100 precursor proteins which sequester NF-κB dimers in via a C terminal ankyrin repeat domain homologous to the central ankyrin repeat domains found in IκBα, IκBβ and IκBε [4]

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.