The kinesin KIF1C transports APC-dependent mRNAs to cell protrusions (original) (raw)

  1. Konstadinos Moissoglu3,12,
  2. Emeline Coleno1,2,4,
  3. Tianhong Wang3,
  4. Arthur Imbert5,6,7,
  5. Marie-Cécile Robert1,2,4,
  6. Marion Peter1,2,
  7. Racha Chouaib1,2,8,
  8. Thomas Walter5,6,7,
  9. Florian Mueller9,10,
  10. Kazem Zibara8,11,
  11. Edouard Bertrand1,2,4 and
  12. Stavroula Mili3
  13. 1Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, 34293 Montpellier, France
  14. 2Equipe labélisée Ligue Nationale Contre le Cancer, University of Montpellier, CNRS, 34000 Montpellier, France
  15. 3Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20814, USA
  16. 4Institut de Génétique Humaine, University of Montpellier, CNRS, 34396 Montpellier, France
  17. 5MINES ParisTech, PSL-Research University, CBIO-Centre for Computational Biology, 77300 Fontainebleau, France
  18. 6Institut Curie, 75248 Paris Cedex, France
  19. 7INSERM, U900, 75248 Paris Cedex, France
  20. 8Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
  21. 9Unité Imagerie et Modélisation, Institut Pasteur and CNRS UMR 3691, 75015 Paris, France
  22. 10C3BI, USR 3756 IP CNRS – Paris, France
  23. 11ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon
  24. Corresponding authors: edouard.bertrand{at}igh.cnrs.fr, voula.mili{at}nih.gov
  25. 12 These authors contributed equally to this work.

Abstract

RNA localization and local translation are important for numerous cellular functions. In mammals, a class of mRNAs localize to cytoplasmic protrusions in an APC-dependent manner, with roles during cell migration. Here, we investigated this localization mechanism. We found that the KIF1C motor interacts with APC-dependent mRNAs and is required for their localization. Live cell imaging revealed rapid, active transport of single mRNAs over long distances that requires both microtubules and KIF1C. Two-color imaging directly revealed single mRNAs transported by single KIF1C motors, with the 3′UTR being sufficient to trigger KIF1C-dependent RNA transport and localization. Moreover, KIF1C remained associated with peripheral, multimeric RNA clusters and was required for their formation. These results reveal a widespread RNA transport pathway in mammalian cells, in which the KIF1C motor has a dual role in transporting RNAs and clustering them within cytoplasmic protrusions. Interestingly, KIF1C also transports its own mRNA, suggesting a possible feedback loop acting at the level of mRNA transport.

Footnotes

OPEN ACCESS ARTICLE

This Article

  1. » AbstractFree

  2. Full TextFree

  3. Full Text (PDF)Free

  4. Supplemental Material

  5. Alert me when this article is cited

  6. Alert me if a correction is posted

  7. Similar articles in this journal

  8. Similar articles in Web of Science

  9. Similar articles in Web of Science

  10. Article Metrics

  11. Similar articles in PubMed

  12. Download to citation manager

  13. Permissions

  14. Load citing article information

  15. Citing articles via Web of Science

  16. Citing articles via Google Scholar

  17. Articles by Pichon, X.

  18. Articles by Mili, S.

  19. Search for related content

  20. PubMed citation

  21. Articles by Pichon, X.

  22. Articles by Mili, S.

  23. Profile for Pichon, X.http://orcid.org/0000-0002-0755-2305

  24. Profile for Moissoglu, K.http://orcid.org/0000-0001-7211-4320

  25. Profile for Peter, M.http://orcid.org/0000-0002-5720-4798

  26. Profile for Mueller, F.http://orcid.org/0000-0002-9622-4396

  27. Profile for Bertrand, E.http://orcid.org/0000-0002-9642-7994

  28. Profile for Mili, S.http://orcid.org/0000-0002-9161-8660