Advanced search
1 file | 2.94 MB Add to list

Small heat-shock protein HSPB1 mutants stabilize microtubules in Charcot-Marie-Tooth neuropathy

(2011) JOURNAL OF NEUROSCIENCE. 31(43). p.15320-15328
Author
Organization
Project
Abstract
Mutations in the small heat shock protein HSPB1 (HSP27) are causative for Charcot-Marie-Tooth (CMT) neuropathy. We previously showed that a subset of these mutations displays higher chaperone activity and enhanced affinity to client proteins. We hypothesized that this excessive binding property might cause the HSPB1 mutant proteins to disturb the function of proteins essential for the maintenance or survival of peripheral neurons. In the present work, we explored this hypothesis further and compared the protein complexes formed by wild-type and mutant HSPB1. Tubulin came out as the most striking differential interacting protein, with hyperactive mutants binding more strongly to both tubulin and microtubules. This anomalous binding leads to a stabilization of the microtubule network in a microtubule-associated protein-like manner as reflected by resistance to cold depolymerization, faster network recovery after nocodazole treatment, and decreased rescue and catastrophe rates of individual microtubules. In a transgenic mouse model for mutant HSPB1 that recapitulates all features of CMT, we could confirm the enhanced interaction of mutant HSPB1 with tubulin. Increased stability of the microtubule network was also clear in neurons isolated from these mice. Since neuronal cells are particularly vulnerable to disturbances in microtubule dynamics, this mechanism might explain the neuron-specific CMT phenotype caused by HSPB1 mutations.
Keywords
DISEASE, MUTATIONS, PHOSPHORYLATION, HSP27, NEURODEGENERATION, CELL-PROLIFERATION, DYNAMICS, PERIPHERAL NEUROPATHY

Downloads

  • extra11AlmeidaSouza.pdf
    • full text
    • |
    • open access
    • |
    • PDF
    • |
    • 2.94 MB

Citation

Please use this url to cite or link to this publication:

MLA
Almeida-Souza, Leonardo, et al. “Small Heat-Shock Protein HSPB1 Mutants Stabilize Microtubules in Charcot-Marie-Tooth Neuropathy.” JOURNAL OF NEUROSCIENCE, vol. 31, no. 43, 2011, pp. 15320–28, doi:10.1523/JNEUROSCI.3266-11.2011.
APA
Almeida-Souza, L., Asselbergh, B., d’Ydewalle, C., Moonens, K., Goethals, S., de Winter, V., … Janssens, S. (2011). Small heat-shock protein HSPB1 mutants stabilize microtubules in Charcot-Marie-Tooth neuropathy. JOURNAL OF NEUROSCIENCE, 31(43), 15320–15328. https://doi.org/10.1523/JNEUROSCI.3266-11.2011
Chicago author-date
Almeida-Souza, Leonardo, Bob Asselbergh, Constantin d’Ydewalle, Kristof Moonens, Sofie Goethals, Vicky de Winter, Abdelkarim Azmi, et al. 2011. “Small Heat-Shock Protein HSPB1 Mutants Stabilize Microtubules in Charcot-Marie-Tooth Neuropathy.” JOURNAL OF NEUROSCIENCE 31 (43): 15320–28. https://doi.org/10.1523/JNEUROSCI.3266-11.2011.
Chicago author-date (all authors)
Almeida-Souza, Leonardo, Bob Asselbergh, Constantin d’Ydewalle, Kristof Moonens, Sofie Goethals, Vicky de Winter, Abdelkarim Azmi, Joy Irobi, Jean-Pierre Timmermans, Kris Gevaert, Han Remaut, Ludo Van den Bosch, Vincent Timmerman, and Sophie Janssens. 2011. “Small Heat-Shock Protein HSPB1 Mutants Stabilize Microtubules in Charcot-Marie-Tooth Neuropathy.” JOURNAL OF NEUROSCIENCE 31 (43): 15320–15328. doi:10.1523/JNEUROSCI.3266-11.2011.
Vancouver
1.
Almeida-Souza L, Asselbergh B, d’Ydewalle C, Moonens K, Goethals S, de Winter V, et al. Small heat-shock protein HSPB1 mutants stabilize microtubules in Charcot-Marie-Tooth neuropathy. JOURNAL OF NEUROSCIENCE. 2011;31(43):15320–8.
IEEE
[1]
L. Almeida-Souza et al., “Small heat-shock protein HSPB1 mutants stabilize microtubules in Charcot-Marie-Tooth neuropathy,” JOURNAL OF NEUROSCIENCE, vol. 31, no. 43, pp. 15320–15328, 2011.
@article{2022794,
  abstract     = {{Mutations in the small heat shock protein HSPB1 (HSP27) are causative for Charcot-Marie-Tooth (CMT) neuropathy. We previously showed that a subset of these mutations displays higher chaperone activity and enhanced affinity to client proteins. We hypothesized that this excessive binding property might cause the HSPB1 mutant proteins to disturb the function of proteins essential for the maintenance or survival of peripheral neurons. In the present work, we explored this hypothesis further and compared the protein complexes formed by wild-type and mutant HSPB1. Tubulin came out as the most striking differential interacting protein, with hyperactive mutants binding more strongly to both tubulin and microtubules. This anomalous binding leads to a stabilization of the microtubule network in a microtubule-associated protein-like manner as reflected by resistance to cold depolymerization, faster network recovery after nocodazole treatment, and decreased rescue and catastrophe rates of individual microtubules. In a transgenic mouse model for mutant HSPB1 that recapitulates all features of CMT, we could confirm the enhanced interaction of mutant HSPB1 with tubulin. Increased stability of the microtubule network was also clear in neurons isolated from these mice. Since neuronal cells are particularly vulnerable to disturbances in microtubule dynamics, this mechanism might explain the neuron-specific CMT phenotype caused by HSPB1 mutations.}},
  author       = {{Almeida-Souza, Leonardo and Asselbergh, Bob and d'Ydewalle, Constantin and Moonens, Kristof and Goethals, Sofie and de Winter, Vicky and Azmi, Abdelkarim and Irobi, Joy and Timmermans, Jean-Pierre and Gevaert, Kris and Remaut, Han and Van den Bosch, Ludo and Timmerman, Vincent and Janssens, Sophie}},
  issn         = {{0270-6474}},
  journal      = {{JOURNAL OF NEUROSCIENCE}},
  keywords     = {{DISEASE,MUTATIONS,PHOSPHORYLATION,HSP27,NEURODEGENERATION,CELL-PROLIFERATION,DYNAMICS,PERIPHERAL NEUROPATHY}},
  language     = {{eng}},
  number       = {{43}},
  pages        = {{15320--15328}},
  title        = {{Small heat-shock protein HSPB1 mutants stabilize microtubules in Charcot-Marie-Tooth neuropathy}},
  url          = {{http://doi.org/10.1523/JNEUROSCI.3266-11.2011}},
  volume       = {{31}},
  year         = {{2011}},
}

Altmetric
View in Altmetric
Web of Science
Times cited: