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.

  • Letter
  • Published:

Duplication of Atxn1l suppresses SCA1 neuropathology by decreasing incorporation of polyglutamine-expanded ataxin-1 into native complexes

Abstract

Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disease caused by expansion of a glutamine tract in ataxin-1 (ATXN1). SCA1 pathogenesis studies support a model in which the expanded glutamine tract causes toxicity by modulating the normal activities of ATXN1. To explore native interactions that modify the toxicity of ATXN1, we generated a targeted duplication of the mouse ataxin-1-like (Atxn1l, also known as Boat) locus, a highly conserved paralog of SCA1, and tested the role of this protein in SCA1 pathology. Using a knock-in mouse model of SCA1 that recapitulates the selective neurodegeneration seen in affected individuals, we found that elevated Atxn1l levels suppress neuropathology by displacing mutant Atxn1 from its native complex with Capicua (CIC). Our results provide genetic evidence that the selective neuropathology of SCA1 arises from modulation of a core functional activity of ATXN1, and they underscore the importance of studying the paralogs of genes mutated in neurodegenerative diseases to gain insight into mechanisms of pathogenesis.

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: Atxn1l associates with Atxn1; also shown here is a schematic representation of generation of Atxn1l duplication.
Figure 2: Atxn1l duplication suppresses Sca1154Q/+ cerebellar pathology.
Figure 3: Atxn1l enhances nuclear inclusion formation of polyglutamine-expanded Atxn1 in Purkinje cells and Neuro2A cells.
Figure 4: Elevated Atxn1l levels decrease the association of Atxn1[154Q] with CIC in cerebellar extracts.
Figure 5: Atxn1[154Q] is displaced from its larger endogenous complexes containing CIC as a result of Atxn1l duplication.
Figure 6: Proposed model for suppression of SCA1 pathology by the Atxn1l duplication.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Tsuda, H. et al. The AXH domain of Ataxin-1 mediates neurodegeneration through its interaction with Gfi-1/Senseless proteins. Cell 122, 633–644 (2005).

    Article  CAS  Google Scholar 

  2. Tsai, C.C. et al. Ataxin 1, a SCA1 neurodegenerative disorder protein, is functionally linked to the silencing mediator of retinoid and thyroid hormone receptors. Proc. Natl. Acad. Sci. USA 101, 4047–4052 (2004).

    Article  CAS  Google Scholar 

  3. Mizutani, A. et al. Boat, an AXH domain protein, suppresses the cytotoxicity of mutant ataxin-1. EMBO J. 24, 3339–3351 (2005).

    Article  CAS  Google Scholar 

  4. Lam, Y.C. et al. ATAXIN-1 interacts with the repressor Capicua in its native complex to cause SCA1 neuropathology. Cell 127, 1335–1347 (2006).

    Article  CAS  Google Scholar 

  5. Matilla, A. et al. Mice lacking ataxin-1 display learning deficits and decreased hippocampal paired-pulse facilitation. J. Neurosci. 18, 5508–5516 (1998).

    Article  CAS  Google Scholar 

  6. Lim, J. et al. A protein-protein interaction network for human inherited ataxias and disorders of Purkinje cell degeneration. Cell 125, 801–814 (2006).

    Article  CAS  Google Scholar 

  7. Watase, K. et al. A long CAG repeat in the mouse Sca1 locus replicates SCA1 features and reveals the impact of protein solubility on selective neurodegeneration. Neuron 34, 905–919 (2002).

    Article  CAS  Google Scholar 

  8. Arrasate, M., Mitra, S., Schweitzer, E.S., Segal, M.R. & Finkbeiner, S. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature 431, 805–810 (2004).

    Article  CAS  Google Scholar 

  9. Bowman, A.B., Yoo, S.Y., Dantuma, N.P. & Zoghbi, H.Y. Neuronal dysfunction in a polyglutamine disease model occurs in the absence of ubiquitin-proteasome system impairment and inversely correlates with the degree of nuclear inclusion formation. Hum. Mol. Genet. 14, 679–691 (2005).

    Article  CAS  Google Scholar 

  10. Slow, E.J. et al. Absence of behavioral abnormalities and neurodegeneration in vivo despite widespread neuronal huntingtin inclusions. Proc. Natl. Acad. Sci. USA 102, 11402–11407 (2005).

    Article  CAS  Google Scholar 

  11. Zhang, X. et al. A potent small molecule inhibits polyglutamine aggregation in Huntington's disease neurons and suppresses neurodegeneration in vivo. Proc. Natl. Acad. Sci. USA 102, 892–897 (2005).

    Article  CAS  Google Scholar 

  12. Bodner, R.A. et al. Pharmacological promotion of inclusion formation: a therapeutic approach for Huntington's and Parkinson's diseases. Proc. Natl. Acad. Sci. USA 103, 4246–4251 (2006).

    Article  CAS  Google Scholar 

  13. Taylor, J.P. et al. Aggresomes protect cells by enhancing the degradation of toxic polyglutamine-containing protein. Hum. Mol. Genet. 12, 749–757 (2003).

    Article  CAS  Google Scholar 

  14. Klement, I.A. et al. Ataxin-1 nuclear localization and aggregation: role in polyglutamine-induced disease in SCA1 transgenic mice. Cell 95, 41–53 (1998).

    Article  CAS  Google Scholar 

  15. Cattaneo, E., Zuccato, C. & Tartari, M. Normal huntingtin function: an alternative approach to Huntington's disease. Nat. Rev. Neurosci. 6, 919–930 (2005).

    Article  CAS  Google Scholar 

  16. Cui, L. et al. Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell 127, 59–69 (2006).

    Article  CAS  Google Scholar 

  17. Zhai, W., Jeong, H., Cui, L., Krainc, D. & Tjian, R. In vitro analysis of huntingtin-mediated transcriptional repression reveals multiple transcription factor targets. Cell 123, 1241–1253 (2005).

    Article  CAS  Google Scholar 

  18. Dragatsis, I., Levine, M.S. & Zeitlin, S. Inactivation of Hdh in the brain and testis results in progressive neurodegeneration and sterility in mice. Nat. Genet. 26, 300–306 (2000).

    Article  CAS  Google Scholar 

  19. Van Raamsdonk, J.M. et al. Loss of wild-type huntingtin influences motor dysfunction and survival in the YAC128 mouse model of Huntington disease. Hum. Mol. Genet. 14, 1379–1392 (2005).

    Article  CAS  Google Scholar 

  20. Auerbach, W. et al. The HD mutation causes progressive lethal neurological disease in mice expressing reduced levels of huntingtin. Hum. Mol. Genet. 10, 2515–2523 (2001).

    Article  CAS  Google Scholar 

  21. Leavitt, B.R. et al. Wild-type huntingtin reduces the cellular toxicity of mutant huntingtin in vivo. Am. J. Hum. Genet. 68, 313–324 (2001).

    Article  CAS  Google Scholar 

  22. Zuccato, C. et al. Huntingtin interacts with REST/NRSF to modulate the transcription of NRSE-controlled neuronal genes. Nat. Genet. 35, 76–83 (2003).

    Article  CAS  Google Scholar 

  23. Ho, L.W., Brown, R., Maxwell, M., Wyttenbach, A. & Rubinsztein, D.C. Wild type Huntingtin reduces the cellular toxicity of mutant Huntingtin in mammalian cell models of Huntington's disease. J. Med. Genet. 38, 450–452 (2001).

    Article  CAS  Google Scholar 

  24. Graham, R.K. et al. Cleavage at the caspase-6 site is required for neuronal dysfunction and degeneration due to mutant huntingtin. Cell 125, 1179–1191 (2006).

    Article  CAS  Google Scholar 

  25. Gauthier, L.R. et al. Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell 118, 127–138 (2004).

    Article  CAS  Google Scholar 

  26. da Costa, C.A., Masliah, E. & Checler, F. Beta-synuclein displays an antiapoptotic p53-dependent phenotype and protects neurons from 6-hydroxydopamine-induced caspase 3 activation: cross-talk with alpha-synuclein and implication for Parkinson's disease. J. Biol. Chem. 278, 37330–37335 (2003).

    Article  Google Scholar 

  27. Yoo, S.Y. et al. SCA7 knockin mice model human SCA7 and reveal gradual accumulation of mutant ataxin-7 in neurons and abnormalities in short-term plasticity. Neuron 37, 383–401 (2003).

    Article  CAS  Google Scholar 

  28. Chen, H.K. et al. Interaction of Akt-phosphorylated ataxin-1 with 14–3-3 mediates neurodegeneration in spinocerebellar ataxia type 1. Cell 113, 457–468 (2003).

    Article  CAS  Google Scholar 

  29. Copps, K. et al. Complex formation by the Drosophila MSL proteins: role of the MSL2 RING finger in protein complex assembly. EMBO J. 17, 5409–5417 (1998).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to C.-C. Tsai (University of Medicine and Dentistry of New Jersey—Robert Wood Johnson Medical School) for providing ATXN1L antiserum; S. Vaishnav for mouse genotyping; A. McCall for isolating and characterizing Atxn1l clones; G. Schuster for ES cell work and generation of chimeric animals; K. Schulze for artwork (Fig. 6) and A. Flora, J. Lim, J. Gatchel, P. Moretti, E. Hyun and J. Crespo-Barreto for many discussions. This work was supported by the following US National Institutes of Health grants: National Institute for Neurological Disorders and Stroke grants NS27699 (to H.Y.Z.), NS22920 and NS45667 (to H.T.O.) and National Institute of Child Health and Human Development grant HD024064 (to the Baylor College of Medicine Mental Retardation and Developmental Disabilities Research Center). A.B.B. was a postdoctoral fellow of the Hereditary Disease Foundation. H.Y.Z. is a Howard Hughes Medical Institute investigator.

Author information

Authors and Affiliations

Authors

Contributions

A.B. designed, performed or assisted in most of the experiments; analyzed data; characterized the Atxn1l duplication allele and wrote the manuscript. Y.L. performed ATXN1L immunoprecipitation experiments, identified the CIC protein interaction, designed and characterized the CIC antisera and edited the manuscript. P.J. designed and performed cerebellar Atxn1 immunoprecipitation experiments, characterized Atxn1l sera and provided critical commentary. H.C. designed and constructed the targeting construct. R.R. performed cerebellar extraction, protein blots and HPLC chromatography and characterized Atxn1l sera. R.S. performed the in situ hybridization experiments. J.F. performed cerebellar CIC immunoprecipitation experiments, provided critical commentary and edited the manuscript. J.K. made the ATXN1L cDNA expression vector. H.O. assisted in data analysis and interpretation. H.Z. led the project by inspiring the search for ATXN1 paralogs, generated major hypotheses and formulated the genetic and biochemical experiments, interpreted data, provided experimental direction and edited the manuscript.

Corresponding author

Correspondence to Huda Y Zoghbi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Fig. 1

Atxn1l expression in Purkinje cells of adult mouse cerebellum. (PDF 527 kb)

Supplementary Fig. 2

Fractionation of ATXN1L, ATXN1 and CIC by gel filtration chromatography of mouse cerebellar extracts. (PDF 176 kb)

Supplementary Fig. 3

ATXN1 and ATXN1L compete for interaction with CIC. (PDF 106 kb)

Supplementary Fig. 4

Atxn1l duplication suppresses Sca1154Q/+ cerebellar pathology. (PDF 735 kb)

Supplementary Fig. 5

Atxn1l duplication suppresses Sca1154Q/+ lethality. (PDF 341 kb)

Supplementary Fig. 6

ATXN1L levels are similar between wild-type and Sca1154Q/+ animals. (PDF 131 kb)

Supplementary Table 1

ANOVA post hoc pairwise comparisons. (PDF 55 kb)

Supplementary Methods (PDF 52 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bowman, A., Lam, Y., Jafar-Nejad, P. et al. Duplication of Atxn1l suppresses SCA1 neuropathology by decreasing incorporation of polyglutamine-expanded ataxin-1 into native complexes. Nat Genet 39, 373–379 (2007). https://doi.org/10.1038/ng1977

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng1977

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