Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia

Han-Xiang Deng, Wenjie Chen, Seong-Tshool Hong, Kym M Boycott, George H Gorrie, Nailah Siddique, Yi Yang, Faisal Fecto, Yong Shi, Hong Zhai, Hujun Jiang, Makito Hirano, Evadnie Rampersaud, Gerard H Jansen, Sandra Donkervoort, Eileen H Bigio, Benjamin R Brooks, Kaouther Ajroud, Robert L Sufit, Jonathan L Haines, Enrico Mugnaini, Margaret A Pericak-Vance, Teepu Siddique, Han-Xiang Deng, Wenjie Chen, Seong-Tshool Hong, Kym M Boycott, George H Gorrie, Nailah Siddique, Yi Yang, Faisal Fecto, Yong Shi, Hong Zhai, Hujun Jiang, Makito Hirano, Evadnie Rampersaud, Gerard H Jansen, Sandra Donkervoort, Eileen H Bigio, Benjamin R Brooks, Kaouther Ajroud, Robert L Sufit, Jonathan L Haines, Enrico Mugnaini, Margaret A Pericak-Vance, Teepu Siddique

Abstract

Amyotrophic lateral sclerosis (ALS) is a paralytic and usually fatal disorder caused by motor-neuron degeneration in the brain and spinal cord. Most cases of ALS are sporadic but about 5-10% are familial. Mutations in superoxide dismutase 1 (SOD1), TAR DNA-binding protein (TARDBP, also known as TDP43) and fused in sarcoma (FUS, also known as translocated in liposarcoma (TLS)) account for approximately 30% of classic familial ALS. Mutations in several other genes have also been reported as rare causes of ALS or ALS-like syndromes. The causes of the remaining cases of familial ALS and of the vast majority of sporadic ALS are unknown. Despite extensive studies of previously identified ALS-causing genes, the pathogenic mechanism underlying motor-neuron degeneration in ALS remains largely obscure. Dementia, usually of the frontotemporal lobar type, may occur in some ALS cases. It is unclear whether ALS and dementia share common aetiology and pathogenesis in ALS/dementia. Here we show that mutations in UBQLN2, which encodes the ubiquitin-like protein ubiquilin 2, cause dominantly inherited, chromosome-X-linked ALS and ALS/dementia. We describe novel ubiquilin 2 pathology in the spinal cords of ALS cases and in the brains of ALS/dementia cases with or without UBQLN2 mutations. Ubiquilin 2 is a member of the ubiquilin family, which regulates the degradation of ubiquitinated proteins. Functional analysis showed that mutations in UBQLN2 lead to an impairment of protein degradation. Therefore, our findings link abnormalities in ubiquilin 2 to defects in the protein degradation pathway, abnormal protein aggregation and neurodegeneration, indicating a common pathogenic mechanism that can be exploited for therapeutic intervention.

Conflict of interest statement

Competing interest declaration: None

Figures

Fig. 1
Fig. 1
Mutations of UBQLN2 in patients with ALS and ALS/dementia. (a) A mutation, c.1490C>A, resulting in p.P497H, was identified in a large family with ALS (F#186). This family was used to map X-chromosome-linked ALS. The pedigree is shown on the left and sequences are shown on the right. The wild-type sequence is shown in the upper panel. A representative hemizygous mutation in a male patient (V3) is shown in the lower panel. All of the affected members whose DNA samples were available for sequencing analysis had the mutation. Two obligate carriers (III4 and IV2) were identified to have the same mutation. For simplicity and clarity, more than one unaffected individual of both genders is represented by a single diamond and more than one unaffected male individual is represented by a single square. Individuals with mutation in the UBQLN2 are labeled by (m) and those without mutation are labeled by (n). (b) A mutation c.1516C>A (p.P506T) was identified in F#6316. Shown in the right lower panel is a heterozygous mutation from a female obligate carrier (II1). (a–b) Probands are indicated with arrows and patients with dementia are indicated with asterisks. (c) Evolutionary conservation of amino acids in the mutated region of ubiquilin2 in different species. Comparison of human (H. sapiens) ubiquilin and its orthologues in chimpanzee (P. troglodytes), dog (C. lupus familiaris), cattle (B. taurus), mouse (M. musculus) and rat (R. norvegicus). Amino acids identical to human UBQLN2 are in black letters and non-identical ones are denoted in red letters. The positions of the C-terminal amino acids are shown on the right. The mutated amino acids are indicated by arrows on the top. (d) Predicted structural and functional domains of ubiquilin2. Ubiquilin2 is a protein of 624 amino acids. Predicted structural and functional domains include a UBL (ubiquitin-like domain, 33–103), four STI1 (heat shock chaperonin-binding motif), a 12 PXX repeats (491–526) and a UBA (ubiquitin-associated domain). ALS- and ALS/dementia-linked mutations are clustered in the 12 PXX repeats.
Fig. 2
Fig. 2
Ubiquilin2-immunoreactive inclusions in the spinal cord and hippocampus. Spinal cord (a–c) and hippocampal (d–g) sections from a patient with a UBQLN2P506T mutation were analyzed with confocal microscopy (a–c) and immunohistochemistry (d–g) using a monoclonal antibody against ubiquilin2 (ubiquilin2-C). The ubiquilin2-positive and skein-like inclusions (arrowhead) are shown in a spinal motor neuron (a). These inclusions are also ubiquitin-positive (b and c). In the hippocampus, the ubiquilin2-positive inclusions are shown in the molecular layer of the fascia dentate (d and e), CA3 (f) and CA1 (g). White arrows in the panel (d) indicate the middle region of the molecular layer with ubiquilin2-positive inclusions. The higher magnification image of the boxed area in panel (d) is shown in panel (e). Black arrows indicate the representative inclusions in neurites (e–g), and arrowheads indicate cytoplasmic inclusions in the cell bodies (f and g). Scale bar, 200μm in panel (d), 50μm in panel (e) and 25 μm in panels (f and g).
Fig. 3
Fig. 3
Co-localization of ubiquilin2 and ALS- and dementia-linked TDP43. Neuro2a cells were transfected with various combinations of wild-type ubiquilin2, wild-type TDP43, mutant ubiquilin2 (P497H), and C-terminal fragment of TDP43 that is linked to ALS and FTLD. Ubiquilin2 is GFP-tagged and TDP43 is mCherry-tagged. Wild-type and mutant ubiquilin2 are largely cytoplasmic. WtTDP43 are almost exclusively distributed in the nuclei. The C-TDP43, an ALS- and dementia-linked TDP43 fragment (aa218-414) is almost exclusively cytoplasmic. TDP43 inclusions are co-localized with wild-type (g–i) and mutant (P497H) ubiquilin2 (j–l) (arrows). Some ubiquilin2-positive inclusions are TDP43-negative (arrowhead).
Fig. 4
Fig. 4
Mutations in ubiquilin2 lead to ubiquitin-mediated impairment of proteasomal degradation. UbG76V-GFP fluorescence intensity was quantified by FACS 48 hours post-transfection in Neuro2a cells (a) transiently transfected with either wild-type (WT) or mutant ubiquilin2. The dynamics of UbG76V-GFP reporter degradation after blockage of protein synthesis with cycloheximide for 0, 2, 4, and 6 hours in Neuro2a cells are shown in panel (b). The rates of UPS reporter degradation were significantly slower in both ubiquilin2-P497H and ubiquilin2-P506T mutants when compared to the wild-type ubiquilin2 at 4 and 6 hours (b). The mean fluorescence intensity was determined at the indicated time points by flow cytometry. Mean fluorescence before cycloheximide administration was standardized as 100%. Data are averaged from at least three independent experiments. *p <0.05, **p <0.01, and ***p <0.001 indicating significant differences when compared to WT-UBQLN2 (two-tailed Student t test). Error bars, means ± s.e.m.

References

    1. Deng HX, et al. Amyotrophic lateral sclerosis and structural defects in Cu, Zn superoxide dismutase. Science. 1993;261:1047–1051.
    1. Rosen DR, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;362:59–62.
    1. Kabashi E, et al. TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat Genet. 2008;40:572–574. doi: 10.1038/ng.132. ng.132 [pii]
    1. Sreedharan J, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319:1668–1672. doi: 10.1126/science.1154584. 1154584 [pii]
    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. doi: 10.1126/science.1166066. 323/5918/1205 [pii]
    1. Vance C, et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science. 2009;323:1208–1211. doi: 10.1126/science.1165942. 323/5918/1208 [pii]
    1. Chen YZ, et al. DNA/RNA helicase gene mutations in a form of juvenile amyotrophic lateral sclerosis (ALS4) Am J Hum Genet. 2004;74:1128–1135. doi: 10.1086/421054. S0002-9297(07)62840-7 [pii]
    1. Greenway MJ, et al. ANG mutations segregate with familial and ‘sporadic’ amyotrophic lateral sclerosis. Nat Genet. 2006;38:411–413. doi: 10.1038/ng1742. ng1742 [pii]
    1. Nishimura AL, et al. A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. Am J Hum Genet. 2004;75:822–831. doi: 10.1086/425287. S0002-9297(07)63787-2 [pii]
    1. Yang Y, et al. The gene encoding alsin, a protein with three guanine-nucleotide exchange factor domains, is mutated in a form of recessive amyotrophic lateral sclerosis. Nat Genet. 2001;29:160–165.
    1. Chow CY, et al. Deleterious variants of FIG4, a phosphoinositide phosphatase, in patients with ALS. Am J Hum Genet. 2009;84:85–88. doi: 10.1016/j.ajhg.2008.12.010. S0002-9297(08)00631-9 [pii]
    1. Maruyama H, et al. Mutations of optineurin in amyotrophic lateral sclerosis. Nature. 2010;465:223–226. doi: 10.1038/nature08971. nature08971 [pii]
    1. Ticozzi N, et al. Paraoxonase gene mutations in amyotrophic lateral sclerosis. Ann Neurol. 2010;68:102–107. doi: 10.1002/ana.21993.
    1. Mitchell J, et al. Familial amyotrophic lateral sclerosis is associated with a mutation in D-amino acid oxidase. Proc Natl Acad Sci U S A. 2010;107:7556–7561. doi: 10.1073/pnas.0914128107. 0914128107 [pii]
    1. Johnson JO, et al. Exome Sequencing Reveals VCP Mutations as a Cause of Familial ALS. Neuron. 2010;68:857–864. doi: 10.1016/j.neuron.2010.11.036. S0896-6273(10)00978-5 [pii]
    1. Lansbury PT, Lashuel HA. A century-old debate on protein aggregation and neurodegeneration enters the clinic. Nature. 2006;443:774–779. doi: 10.1038/nature05290. nature05290 [pii]
    1. Deng HX, et al. FUS-immunoreactive inclusions are a common feature in sporadic and non-SOD1 familial amyotrophic lateral sclerosis. Ann Neurol. 2010;67:739–748. doi: 10.1002/ana.22051.
    1. Neumann M, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–133. doi: 10.1126/science.1134108. 314/5796/130 [pii]
    1. Shibata N, et al. Intense superoxide dismutase-1 immunoreactivity in intracytoplasmic hyaline inclusions of familial amyotrophic lateral sclerosis with posterior column involvement. J Neuropathol Exp Neurol. 1996;55:481–490.
    1. Mackenzie IR, et al. Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol. 2007;61:427–434. doi: 10.1002/ana.21147.
    1. Neumann M, et al. Frontotemporal lobar degeneration with FUS pathology. Brain. 2009 doi: 10.1093/brain/awp214. awp214 [pii]
    1. Urwin H, et al. FUS pathology defines the majority of tau- and TDP-43-negative frontotemporal lobar degeneration. Acta Neuropathol. 2010;120:33–41. doi: 10.1007/s00401-010-0698-6.
    1. Nonaka T, Kametani F, Arai T, Akiyama H, Hasegawa M. Truncation and pathogenic mutations facilitate the formation of intracellular aggregates of TDP-43. Hum Mol Genet. 2009;18:3353–3364. doi: 10.1093/hmg/ddp275. ddp275 [pii]
    1. Ko HS, Uehara T, Tsuruma K, Nomura Y. Ubiquilin interacts with ubiquitylated proteins and proteasome through its ubiquitin-associated and ubiquitin-like domains. FEBS Lett. 2004;566:110–114. doi: 10.1016/j.febslet.2004.04.031. S0014579304004612 [pii]
    1. Dantuma NP, Lindsten K, Glas R, Jellne M, Masucci MG. Short-lived green fluorescent proteins for quantifying ubiquitin/proteasome-dependent proteolysis in living cells. Nat Biotechnol. 2000;18:538–543. doi: 10.1038/75406.
    1. Kay BK, Williamson MP, Sudol M. The importance of being proline: the interaction of proline-rich motifs in signaling proteins with their cognate domains. FASEB J. 2000;14:231–241.
    1. Aitio O, et al. Recognition of tandem PxxP motifs as a unique Src homology 3-binding mode triggers pathogen-driven actin assembly. Proc Natl Acad Sci U S A. 2010;107:21743–21748. doi: 10.1073/pnas.1010243107. 1010243107 [pii]
    1. Haapasalo A, et al. Emerging role of Alzheimer’s disease-associated ubiquilin-1 in protein aggregation. Biochem Soc Trans. 2010;38:150–155. doi: 10.1042/BST0380150. BST0380150 [pii]
    1. Kim SH, et al. Potentiation of amyotrophic lateral sclerosis (ALS)-associated TDP-43 aggregation by the proteasome-targeting factor, ubiquilin 1. J Biol Chem. 2009;284:8083–8092. doi: 10.1074/jbc.M808064200. M808064200 [pii]
    1. Aguzzi A, O’Connor T. Protein aggregation diseases: pathogenicity and therapeutic perspectives. Nat Rev Drug Discov. 2010;9:237–248. doi: 10.1038/nrd3050. nrd3050 [pii]
    1. Brooks BR, Miller RG, Swash M, Munsat TL. El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord. 2000;1:293–299.
    1. Neary D, et al. Frontotemporal lobar degeneration: a consensus on clinical diagnostic criteria. Neurology. 1998;51:1546–1554.
    1. Cairns NJ, et al. Neuropathologic diagnostic and nosologic criteria for frontotemporal lobar degeneration: consensus of the Consortium for Frontotemporal Lobar Degeneration. Acta Neuropathol. 2007;114:5–22. doi: 10.1007/s00401-007-0237-2.
    1. Mackenzie IR, et al. Heterogeneity of ubiquitin pathology in frontotemporal lobar degeneration: classification and relation to clinical phenotype. Acta Neuropathol. 2006;112:539–549. doi: 10.1007/s00401-006-0138-9.
    1. Deng HX, et al. Conversion to the amyotrophic lateral sclerosis phenotype is associated with intermolecular linked insoluble aggregates of SOD1 in mitochondria. Proc Natl Acad Sci U S A. 2006;103:7142–7147. doi: 10.1073/pnas.0602046103. 0602046103 [pii]

Source: PubMed

3
Se inscrever