Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

A role for mDia, a Rho-regulated actin nucleator, in tangential migration of interneuron precursors

Abstract

In brain development, distinct types of migration, radial migration and tangential migration, are shown by excitatory and inhibitory neurons, respectively. Whether these two types of migration operate by similar cellular mechanisms remains unclear. We examined neuronal migration in mice deficient in mDia1 (also known as Diap1) and mDia3 (also known as Diap2), which encode the Rho-regulated actin nucleators mammalian diaphanous homolog 1 (mDia1) and mDia3. mDia deficiency impaired tangential migration of cortical and olfactory inhibitory interneurons, whereas radial migration and consequent layer formation of cortical excitatory neurons were unaffected. mDia-deficient neuroblasts exhibited reduced separation of the centrosome from the nucleus and retarded nuclear translocation. Concomitantly, anterograde F-actin movement and F-actin condensation at the rear, which occur during centrosomal and nuclear movement of wild-type cells, respectively, were impaired in mDia-deficient neuroblasts. Blockade of Rho-associated protein kinase (ROCK), which regulates myosin II, also impaired nuclear translocation. These results suggest that Rho signaling via mDia and ROCK critically regulates nuclear translocation through F-actin dynamics in tangential migration, whereas this mechanism is dispensable in radial migration.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Radial migration of excitatory cortical precursors is not impaired in mDia DKO mice.
Figure 2: Tangential migration of cortical interneurons is impaired in mDia DKO mice.
Figure 3: Migration of SVZ neuroblasts to the granule cell layer of the olfactory bulb is interrupted in mDia DKO mice.
Figure 4: mDia deficiency causes impaired migration from MGE and SVZ explants in vitro.
Figure 5: Fluorescent imaging analyses of cell morphology and movement of SVZ neuroblasts.
Figure 6: mDia DKO neuroblasts show reduced movement of the centrosome and the cell body.
Figure 7: mDia-dependent F-actin dynamics and localization of mDia in migrating SVZ neuroblasts.
Figure 8: Roles of ROCK and myosin II in migration of SVZ neuroblasts.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Ghashghaei, H.T., Lai, C. & Anton, E.S. Neuronal migration in the adult brain: are we there yet? Nat. Rev. Neurosci. 8, 141–151 (2007).

    Article  CAS  Google Scholar 

  2. Marín, O. & Rubenstein, J.L. Cell migration in the forebrain. Annu. Rev. Neurosci. 26, 441–483 (2003).

    Article  Google Scholar 

  3. Métin, C., Vallee, R.B., Rakic, P. & Bhide, P.G. Modes and mishaps of neuronal migration in the mammalian brain. J. Neurosci. 28, 11746–11752 (2008).

    Article  Google Scholar 

  4. Alvarez-Buylla, A. & Garcia-Verdugo, J.M. Neurogenesis in adult subventricular zone. J. Neurosci. 22, 629–634 (2002).

    Article  CAS  Google Scholar 

  5. Lledo, P.M., Merkle, F.T. & Alvarez-Buylla, A. Origin and function of olfactory bulb interneuron diversity. Trends Neurosci. 31, 392–400 (2008).

    Article  CAS  Google Scholar 

  6. Kawauchi, T. & Hoshino, M. Molecular pathways regulating cytoskeletal organization and morphological changes in migrating neurons. Dev. Neurosci. 30, 36–46 (2008).

    Article  CAS  Google Scholar 

  7. Marín, O., Valdeolmillos, M. & Moya, F. Neurons in motion: same principles for different shapes? Trends Neurosci. 29, 655–661 (2006).

    Article  Google Scholar 

  8. Xie, Z., Sanada, K., Samuels, B.A., Shih, H. & Tsai, L.H. Serine 732 phosphorylation of FAK by Cdk5 is important for microtubule organization, nuclear movement, and neuronal migration. Cell 114, 469–482 (2003).

    Article  CAS  Google Scholar 

  9. Solecki, D.J., Model, L., Gaetz, J., Kapoor, T.M. & Hatten, M.E. Par6α signaling controls glial-guided neuronal migration. Nat. Neurosci. 7, 1195–1203 (2004).

    Article  CAS  Google Scholar 

  10. Tsai, J.W., Bremner, K.H. & Vallee, R.B. Dual subcellular roles for LIS1 and dynein in radial neuronal migration in live brain tissue. Nat. Neurosci. 10, 970–979 (2007).

    Article  CAS  Google Scholar 

  11. O'Rourke, N.A., Dailey, M.E., Smith, S.J. & McConnell, S.K. Diverse migratory pathways in the developing cerebral cortex. Science 258, 299–302 (1992).

    Article  CAS  Google Scholar 

  12. Wichterle, H., Garcia-Verdugo, J.M. & Alvarez-Buylla, A. Direct evidence for homotypic, glia-independent neuronal migration. Neuron 18, 779–791 (1997).

    Article  CAS  Google Scholar 

  13. Marín, O. & Rubenstein, J.L. A long, remarkable journey: tangential migration in the telencephalon. Nat. Rev. Neurosci. 2, 780–790 (2001).

    Article  Google Scholar 

  14. Schaar, B.T. & McConnell, S.K. Cytoskeletal coordination during neuronal migration. Proc. Natl. Acad. Sci. USA 102, 13652–13657 (2005).

    Article  CAS  Google Scholar 

  15. Baudoin, J.P., Alvarez, C., Gaspar, P. & Metin, C. Nocodazole-induced changes in microtubule dynamics impair the morphology and directionality of migrating medial ganglionic eminence cells. Dev. Neurosci. 30, 132–143 (2008).

    Article  CAS  Google Scholar 

  16. Martini, F.J. & Valdeolmillos, M. Actomyosin contraction at the cell rear drives nuclear translocation in migrating cortical interneurons. J. Neurosci. 30, 8660–8670 (2010).

    Article  CAS  Google Scholar 

  17. Bellion, A., Baudoin, J.P., Alvarez, C., Bornens, M. & Metin, C. Nucleokinesis in tangentially migrating neurons comprises two alternating phases: forward migration of the Golgi/centrosome associated with centrosome splitting and myosin contraction at the rear. J. Neurosci. 25, 5691–5699 (2005).

    Article  CAS  Google Scholar 

  18. Hall, A. Rho GTPases and the actin cytoskeleton. Science 279, 509–514 (1998).

    Article  CAS  Google Scholar 

  19. Govek, E.E., Newey, S.E. & Van Aelst, L. The role of the Rho GTPases in neuronal development. Genes Dev. 19, 1–49 (2005).

    Article  CAS  Google Scholar 

  20. Nguyen, L., Besson, A., Roberts, J.M. & Guillemot, F. Coupling cell cycle exit, neuronal differentiation and migration in cortical neurogenesis. Cell Cycle 5, 2314–2318 (2006).

    Article  CAS  Google Scholar 

  21. Govek, E.E., Hatten, M.E. & Van Aelst, L. The role of Rho GTPase proteins in CNS neuronal migration. Dev. Neurobiol. 71, 528–553 (2011).

    Article  CAS  Google Scholar 

  22. Narumiya, S., Ishizaki, T. & Watanabe, N. Rho effectors and reorganization of actin cytoskeleton. FEBS Lett. 410, 68–72 (1997).

    Article  CAS  Google Scholar 

  23. Kimura, K. et al. Regulation of myosin phosphatase by Rho and Rho-associated kinase (Rho-kinase). Science 273, 245–248 (1996).

    Article  CAS  Google Scholar 

  24. Ishizaki, T. et al. p160ROCK, a Rho-associated coiled-coil forming protein kinase, works downstream of Rho and induces focal adhesions. FEBS Lett. 404, 118–124 (1997).

    Article  CAS  Google Scholar 

  25. Watanabe, N., Kato, T., Fujita, A., Ishizaki, T. & Narumiya, S. Cooperation between mDia1 and ROCK in Rho-induced actin reorganization. Nat. Cell Biol. 1, 136–143 (1999).

    Article  CAS  Google Scholar 

  26. Higashida, C. et al. Actin polymerization–driven molecular movement of mDia1 in living cells. Science 303, 2007–2010 (2004).

    Article  CAS  Google Scholar 

  27. Campellone, K.G. & Welch, M.D. A nucleator arms race: cellular control of actin assembly. Nat. Rev. Mol. Cell Biol. 11, 237–251 (2010).

    Article  CAS  Google Scholar 

  28. Konno, D. et al. Neuroepithelial progenitors undergo LGN-dependent planar divisions to maintain self-renewability during mammalian neurogenesis. Nat. Cell Biol. 10, 93–101 (2008).

    Article  CAS  Google Scholar 

  29. Wen, Y. et al. EB1 and APC bind to mDia to stabilize microtubules downstream of Rho and promote cell migration. Nat. Cell Biol. 6, 820–830 (2004).

    Article  CAS  Google Scholar 

  30. Ishizaki, T. et al. Coordination of microtubules and the actin cytoskeleton by the Rho effector mDia1. Nat. Cell Biol. 3, 8–14 (2001).

    Article  CAS  Google Scholar 

  31. Palazzo, A.F., Cook, T.A., Alberts, A.S. & Gundersen, G.G. mDia mediates Rho-regulated formation and orientation of stable microtubules. Nat. Cell Biol. 3, 723–729 (2001).

    Article  CAS  Google Scholar 

  32. Westermann, S. & Weber, K. Post-translational modifications regulate microtubule function. Nat. Rev. Mol. Cell Biol. 4, 938–948 (2003).

    Article  CAS  Google Scholar 

  33. Riedl, J. et al. Lifeact: a versatile marker to visualize F-actin. Nat. Methods 5, 605–607 (2008).

    Article  CAS  Google Scholar 

  34. Vaughan, S. & Dawe, H.R. Common themes in centriole and centrosome movements. Trends Cell Biol. 21, 57–66 (2011).

    Article  CAS  Google Scholar 

  35. Rosenblatt, J., Cramer, L.P., Baum, B. & McGee, K.M. Myosin II–dependent cortical movement is required for centrosome separation and positioning during mitotic spindle assembly. Cell 117, 361–372 (2004).

    Article  CAS  Google Scholar 

  36. Cao, J., Crest, J., Fasulo, B. & Sullivan, W. Cortical Actin dynamics facilitate early-stage centrosome separation. Curr. Biol. 20, 770–776 (2010).

    Article  CAS  Google Scholar 

  37. Nguyen, L. et al. p27kip1 independently promotes neuronal differentiation and migration in the cerebral cortex. Genes Dev. 20, 1511–1524 (2006).

    Article  CAS  Google Scholar 

  38. Ge, W. et al. Coupling of cell migration with neurogenesis by proneural bHLH factors. Proc. Natl. Acad. Sci. USA 103, 1319–1324 (2006).

    Article  CAS  Google Scholar 

  39. Pacary, E. et al. Proneural transcription factors regulate different steps of cortical neuron migration through Rnd-mediated inhibition of RhoA signaling. Neuron 69, 1069–1084 (2011).

    Article  CAS  Google Scholar 

  40. Patel, B.N. & Van Vactor, D.L. Axon guidance: the cytoplasmic tail. Curr. Opin. Cell Biol. 14, 221–229 (2002).

    Article  CAS  Google Scholar 

  41. Sawamoto, K. et al. New neurons follow the flow of cerebrospinal fluid in the adult brain. Science 311, 629–632 (2006).

    Article  CAS  Google Scholar 

  42. Sakata, D. et al. Impaired T lymphocyte trafficking in mice deficient in an actin-nucleating protein, mDia1. J. Exp. Med. 204, 2031–2038 (2007).

    Article  CAS  Google Scholar 

  43. Yagi, T. et al. A novel ES cell line, TT2, with high germline-differentiating potency. Anal. Biochem. 214, 70–76 (1993).

    Article  CAS  Google Scholar 

  44. Niwa, H., Yamamura, K. & Miyazaki, J. Efficient selection for high-expression transfectants with a novel eukaryotic vector. Gene 108, 193–199 (1991).

    Article  CAS  Google Scholar 

  45. Yasuda, S. et al. Cdc42 and mDia3 regulate microtubule attachment to kinetochores. Nature 428, 767–771 (2004).

    Article  CAS  Google Scholar 

  46. Watanabe, N. et al. p140mDia, a mammalian homolog of Drosophila diaphanous, is a target protein for Rho small GTPase and is a ligand for profilin. EMBO J. 16, 3044–3056 (1997).

    Article  CAS  Google Scholar 

  47. Watanabe, S. et al. mDia2 induces the actin scaffold for the contractile ring and stabilizes its position during cytokinesis in NIH 3T3 cells. Mol. Biol. Cell 19, 2328–2338 (2008).

    Article  CAS  Google Scholar 

  48. Masahira, N. et al. Olig2-positive progenitors in the embryonic spinal cord give rise not only to motoneurons and oligodendrocytes, but also to a subset of astrocytes and ependymal cells. Dev. Biol. 293, 358–369 (2006).

    Article  CAS  Google Scholar 

  49. Saito, T. & Nakatsuji, N. Efficient gene transfer into the embryonic mouse brain using in vivo electroporation. Dev. Biol. 240, 237–246 (2001).

    Article  CAS  Google Scholar 

  50. Umeshima, H., Hirano, T. & Kengaku, M. Microtubule-based nuclear movement occurs independently of centrosome positioning in migrating neurons. Proc. Natl. Acad. Sci. USA 104, 16182–16187 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Okabe for providing pCX-EGFP, F. Matsuzaki for providing pCAG-EGFP-C1 and pCAG-PACT-mKO1, Y. Yanagawa for providing GAD65 and GAD67 antisense probes, Y. Deguchi for providing mDia3 riboprobes, A. Mizutani for animal care, K. Tohyama for supporting genotyping and plasmid construction, T. Arai and A. Washimi for secretarial help, and K. Nonomura for technical assistance. R.S. thanks A. Kakizuka for providing the opportunity to work in Kyoto University Graduate School of Medicine. R.S. was supported by the Japan Society for the Promotion of Science Research Fellowship. This work was supported in part by Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan and a Core Research for Evolutional Science and Technology (CREST) grant from Japan Science and Technology Agency.

Author information

Authors and Affiliations

Authors

Contributions

R.S., D.T., T.F. and S.N. designed the study. R.S. performed most of the experiments. H. Kamijo, R.S., H. Kiyonari, T.I. and S.N. generated mDia3 knockout and mDia DKO mice. D.T. and R.S. carried out in utero electroporation for live imaging. R.S. collected and analyzed the imaging data. N.K. and K.S. performed the whole-mount immunostaining. K.W. and H.T. provided essential advices in setting up and performing in utero electroporation, and conducted in situ hybridization. S.N. and T.F. supervised the project. R.S., T.F. and S.N. wrote the manuscript. All authors discussed the results and concurred on the contents of this manuscript.

Corresponding author

Correspondence to Shuh Narumiya.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 (PDF 7632 kb)

Supplementary Video 1

Time-lapse, phase-contrast images of neuroblast migration from a wild-type SVZ explant in Matrigel. Images were acquired at 5 min intervals for 8 h. (MOV 2141 kb)

Supplementary Video 2

Time-lapse, phase-contrast images of neuroblast migration from an mDia-DKO SVZ explant in Matrigel. Images were acquired at 5 min intervals for 8 h. (MOV 1343 kb)

Supplementary Video 3

Time-lapse fluorescent images of migrating SVZ neuroblasts expressing EGFP from a wild-type explant in Matrigel. Images were acquired at 3 min intervals for 45 min. (MOV 340 kb)

Supplementary Video 4

Time-lapse fluorescent images of migrating SVZ neuroblasts expressing EGFP from an mDia-DKO explant in Matrigel. Images were acquired at 3 min intervals for 45 min. (MOV 940 kb)

Supplementary Video 5

Time-lapse fluorescent images of a wild-type SVZ neuroblast during the nuclear translocation in Matrigel. Images were acquired at 3 min intervals for 15 min. Signals of EGFP and PACT-mKO1 are shown in green and purple, respectively. (MOV 566 kb)

Supplementary Video 6

Time-lapse fluorescent images of an mDia-DKO SVZ neuroblast during the nuclear translocation in Matrigel. Images were acquired at 3 min intervals for 33 min. Signals of EGFP and PACT-mKO1 are shown in green and purple, respectively. (MOV 783 kb)

Supplementary Video 7

Time-lapse fluorescent images of F-actin dynamics during the nuclear translocation in two representative wild-type SVZ neuroblasts. Images were acquired at 3 min intervals for 21 or 18 min for respective neuroblasts. Signals of Lifeact-EGFP and PACT-mKO1 are shown in green and purple, respectively. (MOV 1459 kb)

Supplementary Video 8

Time-lapse fluorescent images of F-actin dynamics during the nuclear translocation in two representative mDia-DKO SVZ neuroblasts. Images were acquired at 3 min intervals for 15 or 33 min for respective neuroblasts. Signals of Lifeact-EGFP and PACT-mKO1 are shown in green and purple, respectively. (MOV 1285 kb)

Supplementary Video 9

Time-lapse fluorescent images of a migrating SVZ neuroblast without Y-27632, a ROCK inhibitor, in Matrigel. Images were acquired at 3 min intervals for 36 min. Signals of EGFP and PACT-mKO1 are shown in green and purple, respectively. (MOV 1154 kb)

Supplementary Video 10

Time-lapse fluorescent images of a migrating SVZ neuroblast treated with Y-27632, a ROCK inhibitor, in Matrigel. Images were acquired at 3 min intervals for 30 min. Signals of EGFP and PACT-mKO1 are shown in green and purple, respectively. (MOV 2617 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Shinohara, R., Thumkeo, D., Kamijo, H. et al. A role for mDia, a Rho-regulated actin nucleator, in tangential migration of interneuron precursors. Nat Neurosci 15, 373–380 (2012). https://doi.org/10.1038/nn.3020

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3020

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing