4.5 Article

Nuclear relocalisation of cytoplasmic poly(A)-binding proteins PABP1 and PABP4 in response to UV irradiation reveals mRNA-dependent export of metazoan PABPs

Journal

JOURNAL OF CELL SCIENCE
Volume 124, Issue 19, Pages 3344-3355

Publisher

COMPANY BIOLOGISTS LTD
DOI: 10.1242/jcs.087692

Keywords

Translation initiation factor; Translational reprogramming; Subcellular localisation; Cellular stress response; ICP27; PABPC4

Categories

Funding

  1. MRC Unit (HGU/HRSU) [G117/564]
  2. MRC [G9826324]
  3. BBSRC [BB/I02137X/1] Funding Source: UKRI
  4. MRC [MC_U127692697, G9826324, MC_U127561111, G1002033, G117/564] Funding Source: UKRI
  5. Biotechnology and Biological Sciences Research Council [BB/I02137X/1] Funding Source: researchfish
  6. Medical Research Council [MC_U127561111, G1002033, MC_U127692697, G117/564, G9826324] Funding Source: researchfish

Ask authors/readers for more resources

Poly(A)-binding protein 1 (PABP1) has a fundamental role in the regulation of mRNA translation and stability, both of which are crucial for a wide variety of cellular processes. Although generally a diffuse cytoplasmic protein, it can be found in discrete foci such as stress and neuronal granules. Mammals encode several additional cytoplasmic PABPs that remain poorly characterised, and with the exception of PABP4, appear to be restricted in their expression to a small number of cell types. We have found that PABP4, similarly to PABP1, is a diffusely cytoplasmic protein that can be localised to stress granules. However, UV exposure unexpectedly relocalised both proteins to the nucleus. Nuclear relocalisation of PABPs was accompanied by a reduction in protein synthesis but was not linked to apoptosis. In examining the mechanism of PABP relocalisation, we found that it was related to a change in the distribution of poly(A) RNA within cells. Further investigation revealed that this change in RNA distribution was not affected by PABP knockdown but that perturbations that block mRNA export recapitulate PABP relocalisation. Our results support a model in which nuclear export of PABPs is dependent on ongoing mRNA export, and that a block in this process following UV exposure leads to accumulation of cytoplasmic PABPs in the nucleus. These data also provide mechanistic insight into reports that transcriptional inhibitors and expression of certain viral proteins cause relocation of PABP to the nucleus.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available