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.

Mechanism for the learning deficits in a mouse model of neurofibromatosis type 1

Abstract

Neurofibromatosis type I (NF1) is one of the most common single-gene disorders that causes learning deficits in humans1. Mice carrying a heterozygous null mutation of the Nf1 gene (Nf1+/−) show important features of the learning deficits associated with NF1 (ref. 2). Although neurofibromin has several known properties and functions, including Ras GTPase-activating protein activity3,4, adenylyl cyclase modulation5,6 and microtubule binding7, it is unclear which of these are essential for learning in mice and humans. Here we show that the learning deficits of Nf1+/− mice can be rescued by genetic and pharmacological manipulations that decrease Ras function. We also show that the Nf1+/− mice have increased GABA (γ-amino butyric acid)-mediated inhibition and specific deficits in long-term potentiation, both of which can be reversed by decreasing Ras function. Our results indicate that the learning deficits associated with NF1 may be caused by excessive Ras activity, which leads to impairments in long-term potentiation caused by increased GABA-mediated inhibition. Our findings have implications for the development of treatments for learning deficits associated with NF1.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Learning deficits of Nf1+/− mice are Ras dependent.
Figure 2: Ras-dependent long-term potentiation deficits in Nf1+/− animals.
Figure 3: Ras-dependent enhanced inhibition in Nf1+/− mice.
Figure 4: Long-term potentiation deficits in Nf1+/− mice are caused by increased inhibition.

Similar content being viewed by others

References

  1. North, K. Neurofibromatosis type 1. Am. J. Med. Genet. 97, 119–127 (2000)

    Article  CAS  Google Scholar 

  2. Silva, A. J. et al. A mouse model for the learning and memory deficits associated with neurofibromatosis type I. Nature Genet. 15, 281–284 (1997)

    Article  CAS  Google Scholar 

  3. Ballester, R. et al. The NF1 locus encodes a protein functionally related to mammalian GAP and yeast IRA proteins. Cell 63, 851–859 (1990)

    Article  CAS  Google Scholar 

  4. Xu, G. F. et al. The catalytic domain of the neurofibromatosis type 1 gene product stimulates ras GTPase and complements ira mutants of S. cerevisiae. Cell 63, 835–841 (1990)

    Article  CAS  Google Scholar 

  5. Guo, H. F., The, I., Hannan, F., Bernards, A. & Zhong, Y. Requirement of Drosophila NF1 for activation of adenylyl cyclase by PACAP38-like neuropeptides. Science 276, 795–798 (1997)

    Article  CAS  Google Scholar 

  6. Guo, H. F., Tong, J., Hannan, F., Luo, L. & Zhong, Y. A neurofibromatosis-1-regulated pathway is required for learning in Drosophila. Nature 403, 895–898 (2000)

    Article  ADS  CAS  Google Scholar 

  7. Xu, H. & Gutmann, D. H. Mutations in the GAP-related domain impair the ability of neurofibromin to associate with microtubules. Brain Res. 759, 149–152 (1997)

    Article  CAS  Google Scholar 

  8. Ozonoff, S. Cognitive impairment in neurofibromatosis type 1. Am. J. Med. Genet. 89, 45–52 (1999)

    Article  CAS  Google Scholar 

  9. Morris, R. G., Garrud, P., Rawlins, J. N. & O'Keefe, J. Place navigation impaired in rats with hippocampal lesions. Nature 297, 681–683 (1982)

    Article  ADS  CAS  Google Scholar 

  10. Costa, R. M. et al. Learning deficits, but normal development and tumour predisposition, in mice lacking exon 23a of Nf1. Nature Genet. 27, 399–405 (2001)

    Article  CAS  Google Scholar 

  11. Klose, A. et al. Selective disactivation of neurofibromin GAP activity in neurofibromatosis type 1. Hum. Mol. Genet. 7, 1261–1268 (1998)

    Article  CAS  Google Scholar 

  12. Johnson, L. K-r. et al. ras is an essential gene in the mouse with partial functional overlap with N-ras. Genes Dev. 11, 2468–2481 (1997)

    Article  CAS  Google Scholar 

  13. Brannan, C. I. et al. Targeted disruption of the neurofibromatosis type-1 gene leads to developmental abnormalities in heart and various neural crest-derived tissues. Genes Dev. 8, 1019–1029 (1994)

    Article  ADS  CAS  Google Scholar 

  14. Brandeis, R., Brandys, Y. & Yehuda, S. The use of the Morris water maze in the study of memory and learning. Int. J. Neurosci. 48, 29–69 (1989)

    Article  CAS  Google Scholar 

  15. Gallagher, M., Burwell, R. & Burchinal, M. Severity of spatial learning impairment in aging: Development of a learning index for performance in the Morris water maze. Behav. Neurosci. 107, 618–626 (1993)

    Article  CAS  Google Scholar 

  16. Muthalif, M. M. et al. Contribution of Ras GTPase/MAP kinase and cytochrome P450 metabolites to deoxycorticosterone-salt-induced hypertension. Hypertension 35, 457–463 (2000)

    Article  CAS  Google Scholar 

  17. Gibbs, J. B. et al. Farnesyltransferase inhibitors versus Ras inhibitors. Curr. Opin. Chem. Biol. 1, 197–203 (1997)

    Article  CAS  Google Scholar 

  18. Yan, N. et al. Farnesyltransferase inhibitors block the neurofibromatosis type I (NF1) malignant phenotype. Cancer Res. 55, 3569–3575 (1995)

    CAS  PubMed  Google Scholar 

  19. Kim, H. A., Ling, B. & Ratner, N. Nf1-deficient mouse Schwann cells are angiogenic and invasive and can be induced to hyperproliferate: reversion of some phenotypes by an inhibitor of farnesyl protein transferase. Mol. Cell. Biol. 17, 862–872 (1997)

    Article  CAS  Google Scholar 

  20. Abbott, L. F. & Nelson, S. B. Synaptic plasticity: taming the beast. Nature Neurosci. 3, 1178–1183 (2000)

    Article  CAS  Google Scholar 

  21. Larson, J., Wong, D. & Lynch, G. Patterned stimulation at the theta frequency is optimal for the induction of hippocampal long-term potentiation. Brain Res. 368, 347–350 (1986)

    Article  CAS  Google Scholar 

  22. Hessler, N. A., Shirke, A. M. & Malinow, R. The probability of transmitter release at a mammalian central synapse. Nature 366, 569–572 (1993)

    Article  ADS  CAS  Google Scholar 

  23. Chapman, C. A., Perez, Y. & Lacaille, J. C. Effects of GABAA inhibition on the expression of long-term potentiation in CA1 pyramidal cells are dependent on tetanization parameters. Hippocampus 8, 289–298 (1998)

    Article  CAS  Google Scholar 

  24. Chorvatova, A., Gendron, L., Bilodeau, L., Gallo-Payet, N. & Payet, M. D. A Ras-dependent chloride current activated by adrenocorticotropin in rat adrenal zona glomerulosa cells. Endocrinology 141, 684–692 (2000)

    Article  CAS  Google Scholar 

  25. Tong, J. et al. NF1-regulated adenylyl cyclase pathway. Soc. Neurosci. Abstr. abstract no. 345.9 (Society for Neuroscience, New Orleans, 2000).

  26. Ingram, D. A. et al. Hyperactivation of p21(ras) and the hematopoietic-specific Rho GTPase, Rac2, cooperate to alter the proliferation of neurofibromin-deficient mast cells in vivo and in vitro. J. Exp. Med. 194, 57–69 (2001)

    Article  CAS  Google Scholar 

  27. Jacks, T. et al. Tumour predisposition in mice heterozygous for a targeted mutation in Nf1. Nature Genet. 7, 353–361 (1994)

    Article  CAS  Google Scholar 

  28. Umanoff, H., Edelmann, W., Pellicer, A. & Kucherlapati, R. The murine N-ras gene is not essential for growth and development. Proc. Natl Acad. Sci. USA 92, 1709–1713 (1995)

    Article  ADS  CAS  Google Scholar 

  29. Voikar, V., Koks, S., Vasar, E. & Rauvala, H. Strain and gender differences in the behaviour of mouse lines commonly used in transgenic studies. Physiol. Behav. 72, 271–281 (2001)

    Article  CAS  Google Scholar 

  30. Blanton, M. G., Lo Turco, J. J. & Kriegstein, A. R. Whole cell recording from neurons in slices of reptilian and mammalian cerebral cortex. J. Neurosci. Methods 30, 203–210 (1989)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank V. Manne for the BMS191563, and E. Friedman for technical assistance in earlier experiments. We are grateful to M. Barad, D. Buonomano, T. Cannon, J. Colicelli, P. Frankland, L. Kaczmarek, A. Matynia, M. Sanders and D. Smith for discussions, and to C. Brannan and S. Schlussel for encouragement. R.M.C. received support from the Graduated Program in Basic and Applied Biology (GABBA) of the University of Oporto, the Portuguese Foundation for Science and Technology (FCT) and the National Neurofibromatosis Foundation (NNF). This work was also supported by a generous donation from K. M. Spivak, and by grants from the NIH, Neurofibromatosis Inc. (National, Illinois, Mass Bay Area, Minnesota, Arizona, Kansas and Central Plains, Mid-Atlantic, and Texas chapters), the Merck and the NNF foundations to A.J.S.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alcino J. Silva.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Costa, R., Federov, N., Kogan, J. et al. Mechanism for the learning deficits in a mouse model of neurofibromatosis type 1. Nature 415, 526–530 (2002). https://doi.org/10.1038/nature711

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature711

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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