Mutations in the profilin 1 gene cause familial amyotrophic lateral sclerosis

Chi-Hong Wu, Claudia Fallini, Nicola Ticozzi, Pamela J Keagle, Peter C Sapp, Katarzyna Piotrowska, Patrick Lowe, Max Koppers, Diane McKenna-Yasek, Desiree M Baron, Jason E Kost, Paloma Gonzalez-Perez, Andrew D Fox, Jenni Adams, Franco Taroni, Cinzia Tiloca, Ashley Lyn Leclerc, Shawn C Chafe, Dev Mangroo, Melissa J Moore, Jill A Zitzewitz, Zuo-Shang Xu, Leonard H van den Berg, Jonathan D Glass, Gabriele Siciliano, Elizabeth T Cirulli, David B Goldstein, Francois Salachas, Vincent Meininger, Wilfried Rossoll, Antonia Ratti, Cinzia Gellera, Daryl A Bosco, Gary J Bassell, Vincenzo Silani, Vivian E Drory, Robert H Brown Jr, John E Landers, Chi-Hong Wu, Claudia Fallini, Nicola Ticozzi, Pamela J Keagle, Peter C Sapp, Katarzyna Piotrowska, Patrick Lowe, Max Koppers, Diane McKenna-Yasek, Desiree M Baron, Jason E Kost, Paloma Gonzalez-Perez, Andrew D Fox, Jenni Adams, Franco Taroni, Cinzia Tiloca, Ashley Lyn Leclerc, Shawn C Chafe, Dev Mangroo, Melissa J Moore, Jill A Zitzewitz, Zuo-Shang Xu, Leonard H van den Berg, Jonathan D Glass, Gabriele Siciliano, Elizabeth T Cirulli, David B Goldstein, Francois Salachas, Vincent Meininger, Wilfried Rossoll, Antonia Ratti, Cinzia Gellera, Daryl A Bosco, Gary J Bassell, Vincenzo Silani, Vivian E Drory, Robert H Brown Jr, John E Landers

Abstract

Amyotrophic lateral sclerosis (ALS) is a late-onset neurodegenerative disorder resulting from motor neuron death. Approximately 10% of cases are familial (FALS), typically with a dominant inheritance mode. Despite numerous advances in recent years, nearly 50% of FALS cases have unknown genetic aetiology. Here we show that mutations within the profilin 1 (PFN1) gene can cause FALS. PFN1 is crucial for the conversion of monomeric (G)-actin to filamentous (F)-actin. Exome sequencing of two large ALS families showed different mutations within the PFN1 gene. Further sequence analysis identified 4 mutations in 7 out of 274 FALS cases. Cells expressing PFN1 mutants contain ubiquitinated, insoluble aggregates that in many cases contain the ALS-associated protein TDP-43. PFN1 mutants also display decreased bound actin levels and can inhibit axon outgrowth. Furthermore, primary motor neurons expressing mutant PFN1 display smaller growth cones with a reduced F/G-actin ratio. These observations further document that cytoskeletal pathway alterations contribute to ALS pathogenesis.

Figures

Figure 1. Exome sequencing identifies PFN1 gene…
Figure 1. Exome sequencing identifies PFN1 gene mutations in familial ALS
a-c, Familial ALS pedigrees harboring PFN1 mutations are shown. Asterisks indicate samples subjected to exome sequencing. To prevent identification of individual family members, the gender of each subject and information on the lower generation are withheld. Genotypes of available DNA samples for the indicated PFN1 mutation are shown (w=wild-type, m=mutant). The genotype of sample III:2 in Family #2 (+) was inferred from the genotypes of spouse and progeny (not shown). d, The evolutionary conservation of PFN1 mutations is shown. For each, the mutated amino acid is colored in red.
Figure 2. Mutant PFN1 produces ubiquitinated i…
Figure 2. Mutant PFN1 produces ubiquitinated insoluble aggregates
a, Western blot analysis of transfected N2A cells subject to NP-40-soluble (S) and insoluble (I) fractionation. b, Transfected cells were treated with MG132 and processed as in (a). Hash marks indicate 25 kDa and 37 kDa markers. Transfected N2A cells (c) and PMNs (d) were stained with V5, HA (PMNs) and ubiquitin (N2A) antibodies. Example aggregates are enlarged in the inset in (d). e, Transfected N2A cells displaying insoluble aggregates were counted and analyzed using one-way ANOVA testing with Dunnett’s multiple test comparison (n=127-135 transfected cells from 3 independent experiments). *P<0.05, ***P<0.001, n.s. P>0.05. Error bars indicate SEM. f, Transfected PMNs stained with V5 and TDP-43 antibodies. Scale bars: 5 μm (c), 10 μm (d, f)
Figure 3. Mutant PFN1 inhibits axon outgrowth
Figure 3. Mutant PFN1 inhibits axon outgrowth
a, PFN1-actin interaction region (PDB accession: 2BTF) using the PyMOL Molecular Graphics System (v. 1.4). Magenta: actin; Yellow: PFN1; Green: actin-binding PFN1 residues; Red: ALS-linked mutated PFN1 residues. b, Transfected HEK293 cells were immunoprecipitated with a V5 antibody and then immunoblotted with antibodies for either V5 or actin. c, PMNs transfected with wild-type or mutant V5-PFN1 and a GFP expressing construct were stained to detect V5-PFN1. d, Cumulative distribution of axon lengths relative to the mean of wild-type PFN1 transfected cells was plotted. The axon tip, indicated by arrows, is enlarged in the inset in (d), right panel. P values are given in the legend (n=104-161 cells from 4 independent experiments). Scale bar: 100 μm.
Figure 4. Mutant PFN1 reduces growth cone…
Figure 4. Mutant PFN1 reduces growth cone size and F-/G-actin expression
a, PMNs were transfected with either wild-type or mutant V5-PFN1. At 3 days post-transfection, cells were fixed and stained to detect V5-PFN1, F-actin (Phalloidin, red) and G-actin (DNase I, green). The growth cone region of representative cells is shown. Scale bar: 10 μm. The growth cone area (b) and F-/G-actin expression (c) of transfected cells was determined and plotted. Comparisons to the wild-type V5-PFN1 transfected cells were made using one-way ANOVA testing. *P<0.05, **P<0.01, ***P<0.001 (n=27-35 cells from 3 independent experiments). Error bars indicate SEM.

References

    1. Kabashi E, et al. TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat Genet. 2008;40:572–574.
    1. Sreedharan J, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319:1668–1672.
    1. Kwiatkowski TJ, Jr., et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009;323:1205–1208.
    1. Vance C, et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science. 2009;323:1208–1211.
    1. Johnson JO, et al. Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron. 2010;68:857–864.
    1. Dejesus-Hernandez M, et al. Expanded GGGGCC Hexanucleotide Repeat in Noncoding Region of C9ORF72 Causes Chromosome 9p-Linked FTD and ALS. Neuron. 2011;72:245–256.
    1. Deng HX, et al. Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature. 2011;477:211–215.
    1. Renton AE, et al. A Hexanucleotide Repeat Expansion in C9ORF72 Is the Cause of Chromosome 9p21-Linked ALS-FTD. Neuron. 2011;72:257–268.
    1. Gijselinck I, et al. A C9orf72 promoter repeat expansion in a Flanders-Belgian cohort with disorders of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis spectrum: a gene identification study. Lancet Neurol. 2012;11:54–65.
    1. Mockrin SC, Korn ED. Acanthamoeba profilin interacts with G-actin to increase the rate of exchange of actin-bound adenosine 5′-triphosphate. Biochemistry. 1980;19:5359–5362.
    1. Landers JE, et al. Reduced expression of the Kinesin-Associated Protein 3 (KIFAP3) gene increases survival in sporadic amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A. 2009;106:9004–9009.
    1. Suetsugu S, et al. The essential role of profilin in the assembly of actin for microspike formation. EMBO J. 1998;17:6516–6526.
    1. Giesemann T, et al. A role for polyproline motifs in the spinal muscular atrophy protein SMN. Profilins bind to and colocalize with smn in nuclear gems. J Biol Chem. 1999;274:37908–37914.
    1. Schutt CE, et al. The structure of crystalline profilin-beta-actin. Nature. 1993;365:810–816.
    1. Wills Z, et al. Profilin and the Abl tyrosine kinase are required for motor axon outgrowth in the Drosophila embryo. Neuron. 1999;22:291–299.
    1. Witke W, et al. In mouse brain profilin I and profilin II associate with regulators of the endocytic pathway and actin assembly. EMBO J. 1998;17:967–976.
    1. Braun A, et al. Genomic organization of profilin-III and evidence for a transcript expressed exclusively in testis. Gene. 2002;283:219–225.
    1. Tilney LG, et al. Actin from Thyone sperm assembles on only one end of an actin filament: a behavior regulated by profilin. J Cell Biol. 1983;97:112–124.
    1. Takeuchi H, et al. Hsp70 and Hsp40 improve neurite outgrowth and suppress intracytoplasmic aggregate formation in cultured neuronal cells expressing mutant SOD1. Brain Res. 2002;949:11–22.
    1. Duan W, et al. MG132 enhances neurite outgrowth in neurons overexpressing mutant TAR DNA-binding protein-43 via increase of HO-1. Brain Res. 2011;1397:1–9.
    1. Fujii R, et al. The RNA binding protein TLS is translocated to dendritic spines by mGluR5 activation and regulates spine morphology. Curr Biol. 2005;15:587–593.
    1. Witke W. The role of profilin complexes in cell motility and other cellular processes. Trends Cell Biol. 2004;14:461–469.
    1. Shao J, et al. Phosphorylation of profilin by ROCK1 regulates polyglutamine aggregation. Mol Cell Biol. 2008;28:5196–5208.
    1. Al-Chalabi A, et al. Deletions of the heavy neurofilament subunit tail in amyotrophic lateral sclerosis. Hum Mol Genet. 1999;8:157–164.
    1. Al-Louis F, et al. A frameshift deletion in peripherin gene associated with amyotrophic lateral sclerosis. J Biol Chem. 2004;279:45951–45956.
    1. Puls I, et al. Mutant dynactin in motor neuron disease. Nat Genet. 2003;33:455–456.
    1. Hazan J, et al. Spastin, a new AAA protein, is altered in the most frequent form of autosomal dominant spastic paraplegia. Nat Genet. 1999;23:296–303.
    1. Reid E, et al. A kinesin heavy chain (KIF5A) mutation in hereditary spastic paraplegia (SPG10) Am J Hum Genet. 2002;71:1189–1194.
    1. Mersiyanova IV, et al. A new variant of Charcot-Marie-Tooth disease type 2 is probably the result of a mutation in the neurofilament-light gene. Am J Hum Genet. 2000;67:37–46.
    1. Perrot R, Eyer J. Neuronal intermediate filaments and neurodegenerative disorders. Brain Res Bull. 2009;80:282–295.
    1. Gudbjartsson DF, et al. Allegro, a new computer program for multipoint linkage analysis. Nat Genet. 2000;25:12–13.
    1. Choi M, et al. Genetic diagnosis by whole exome capture and massively parallel DNA sequencing. Proc Natl Acad Sci U S A. 2009;106:19096–19101.
    1. Li R, et al. SOAP2: an improved ultrafast tool for short read alignment. Bioinformatics. 2009;25:1966–1967.
    1. Li R, et al. SNP detection for massively parallel whole-genome resequencing. Genome Res. 2009;19:1124–1132.
    1. Kumar P, et al. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4:1073–1081.
    1. Consortium GP. A map of human genome variation from population-scale sequencing. Nature. 2010;467:1061–1073.
    1. Exome Variant Server. NHLBI Exome Sequencing Project (ESP); Seattle, WA: [December 14, 2011 accessed]. URL:
    1. Bosco DA, et al. Wild-type and mutant SOD1 share an aberrant conformation and a common pathogenic pathway in ALS. Nat Neurosci. 2010;13:1396–1403.
    1. Fallini C, et al. High-efficiency transfection of cultured primary motor neurons to study protein localization, trafficking, and function. Mol Neurodegener. 2010;5:17.
    1. Meijering E, et al. Design and validation of a tool for neurite tracing and analysis in fluorescence microscopy images. Cytometry A. 2004;58:167–176.

Source: PubMed

3
Subskrybuj