Article

Neuronal Nuclear Membrane Budding Occurs during a Developmental Window Modulated by Torsin Paralogs

Graphical Abstract Authors Lauren M. Tanabe, Chun-Chi Liang, William T. Dauer

Correspondence [email protected]

In Brief The symptoms of torsinA-related dystonia emerge during a discrete period of neurodevelopment. Tanabe et al. show that torsinA-related nuclear membrane budding similarly occurs during a critical period of CNS maturation and identify torsinB as a factor that modulates and can rescue this process.

Highlights d Dystonia-related nuclear budding occurs during a neurodevelopmental critical period d TorsinB modulates the opening and closing of the nuclear budding critical period d Dystonia-related neuronal nuclear budding can be modeled in vitro d Increasing torsinB levels rescues the formation of dystonia- related nuclear budding

Tanabe et al., 2016, Cell Reports 16, 3322–3333 September 20, 2016 ª 2016 The Author(s). http://dx.doi.org/10.1016/j.celrep.2016.08.044 Cell Reports Article

Neuronal Nuclear Membrane Budding Occurs during a Developmental Window Modulated by Torsin Paralogs

Lauren M. Tanabe,1,3 Chun-Chi Liang,1,3 and William T. Dauer1,2,4,* 1Department of Neurology 2Department of Cell and Developmental Biology University of Michigan, Ann Arbor, MI 48109, USA 3Co-first author 4Lead Contact *Correspondence: [email protected] http://dx.doi.org/10.1016/j.celrep.2016.08.044

SUMMARY The DYT1 mutation is a 3-bp in-frame deletion in TOR1A, which removes a single glutamic acid residue (‘‘DE’’) from tor- DYT1 dystonia is a neurodevelopmental disease that sinA (Ozelius et al., 1997). TorsinA is a membrane-associated manifests during a discrete period of childhood. The that resides within the ER/nuclear envelope (NE) endo- disease is caused by impaired function of torsinA, membrane space. Several observations support a normal role a protein linked to nuclear membrane budding. The for torsinA at the NE that is disrupted by the DYT1 mutation. relationship of NE budding to neural development An engineered mutation that blocks torsinA ATP hydrolysis and CNS function is unclear, however, obscuring also causes the protein to accumulate abnormally in the perinu- clear space (Gonzalez-Alegre and Paulson, 2004; Goodchild and its potential role in dystonia pathogenesis. We find Dauer, 2004; Naismith et al., 2004). Wild-type torsinA interacts NE budding begins and resolves during a discrete with the NE lamina-associated polypeptide 1 (LAP1), neurodevelopmental window in torsinA null neurons SUN1, and several nesprins (Goodchild et al., 2005; Jungwirth in vivo. The developmental resolution of NE budding et al., 2011; Nery et al., 2008). LAP1 forms a hetero-oligomer corresponds to increased torsinB protein, while with torsinA and activates its enzymatic (ATPase) activity (Brown ablating torsinB from torsinA null neurons prevents et al., 2014; Sosa et al., 2014; Zhao et al., 2013). The DYT1 budding resolution and causes lethal neural dysfunc- mutation impairs binding of torsinA to LAP1 and causes torsinA tion. Developmental changes in torsinB also corre- to accumulate abnormally within the perinuclear space, a late with NE bud formation in differentiating DYT1 phenomenon that requires SUN1 (Jungwirth et al., 2011; Nai- embryonic stem cells, and overexpression of torsinA smith et al., 2009; Zhao et al., 2013; Goodchild and Dauer, or torsinB rescues NE bud formation in this system. 2004). The relevance of these events to dystonia is supported by the finding that a mutation in LAP1 causes a severe form of These findings identify a torsinA neurodevelopmen- childhood dystonia (Dorboz et al., 2014). tal window that is essential for normal CNS function Mouse genetic experiments provide strong in vivo support and have important implications for dystonia patho- for a potential link between torsinA activity, nuclear membrane genesis and therapeutics. function, and dystonic movements. These experiments demon- strate that the DYT1 mutation impairs torsinA function, causing INTRODUCTION abnormal twisting movements and striking morphological abnor- malities of the nuclear membrane selectively in post-migratory An important challenge to unraveling the pathogenesis of in- neurons (Goodchild et al., 2005; Liang et al., 2014; Weisheit herited neurodevelopmental diseases is determining whether and Dauer, 2015). These nuclear abnormalities are membranous pathogenic proteins play unique roles during CNS development out-pouchings (termed ‘‘buds’’) that emerge from the inner (e.g., during a neurodevelopmental window). This challenge is nuclear membrane and protrude into the perinuclear space. exemplified by torsinA, a AAA+ protein that causes DYT1 dysto- The exclusive appearance of buds in neurons and their apparent nia when its is mutated. The abnormal twisting movements relationship to neuronal maturation are consistent with a role for that characterize DYT1 dystonia emerge during a discrete period nuclear membrane dysfunction in the pathogenesis of DYT1 of childhood, but mutation carriers who do not manifest dystonia dystonia (Goodchild et al., 2005; Liang et al., 2014; Weisheit during this window typically remain symptom free for life. Deter- and Dauer, 2015). mining whether there is a unique temporal requirement for Structures similar to neuronal NE buds have been observed normal torsinA function during CNS development is essential in several developmental contexts in diverse wild-type organ- for the identification of key pathophysiological events, accurate isms (Clark, 1960; Hadek and Swift, 1962; Hochstrasser and Se- disease modeling, and targeted therapeutic development. dat, 1987a; Szo¨ llo¨ si, 1965; Szo¨ llo¨ si and Szo¨ llo¨ si, 1988). Some

3322 Cell Reports 16, 3322–3333, September 20, 2016 ª 2016 The Author(s). This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). observations indicate that these buds may represent structural NE budding. We reported recently that conditional deletion of intermediates of a poorly understood form of nuclear pore- torsinA from the CNS (using nestin-Cre; ‘‘N-CKO’’ mice) is independent nucleo-cytosolic transport, a process possibly compatible with life for up to 16 days of age (Liang et al., analogous to herpesvirus nuclear egress (Maric et al., 2014). In 2014), enabling us to advance understanding of this question. Drosophila, bud-like structures are implicated in the normal We first confirmed that deletion of torsinA from neural progenitor transport of ‘‘mega’’ ribonucleoprotein complexes containing cells (the stage of nestin-Cre expression) caused nuclear buds Wingless signaling components (e.g., the C-terminal cleavage indistinguishable from those previously reported in constitutive product of DFrizzled-2) across the post-synaptic nuclear mem- null and DYT1 knockin mice (Goodchild et al., 2005; Tanabe brane (Speese et al., 2012). Drosophila torsin protein appears et al., 2012)(Figure 1A). To explore the relationship between localized to the necks of these structures, and Drosophila torsin NE budding and CNS maturation, we quantified the percentage mutants show defects in transport of mRNAs contained within of neurons with NE buds in several brain regions on the day of the vesicles, and related abnormalities of neuromuscular junc- birth (P0) and in the more mature CNS (P8). All brain regions tion development (Jokhi et al., 2013). examined developed NE buds (Figures 1A, 1B, and S1), and The observation of similar nuclear membrane out-pouchings the developmental age of the brain region strongly influenced in early developmental contexts has led to the suggestion the percentage of affected neurons observed (Figures 1B–1E). that this process may be required specifically when bursts of co- At P0, earlier maturing caudal regions such as spinal cord and ordinated protein synthesis are required to support critically brainstem (pons) contained a considerably higher percentage timed differentiation or maturation events (Strambio-De-Castilla, of NE bud-bearing neurons (and NE buds/neuron) than later 2013). Indeed, buds in the neuronal nuclei of torsinA null (or ho- maturing rostral regions, such as the striatum (Figures 1B and mozygous DYT1 DE knockin) mice appear selectively in post- 1C). In all regions examined, the percentage of affected neurons migratory maturing neurons (Goodchild et al., 2005), cells highly (and NE buds/neuron) increased with ongoing CNS maturation utilizing the machinery of mRNA synthesis and transport. Other (i.e., from P0 to P8; Figures 1B, 1D, and 1E). Additionally, the work links torsinA loss of function to abnormalities of nuclear relationship between NE buds and developmental age was pore biogenesis and function (VanGompel et al., 2015), suggest- observed within brain regions containing neural classes of ing that the dramatic increase in neuronal nuclear pore number differing maturational ages. For example, at P8, 80% of in maturing neurons (Lodin et al., 1978) may also contribute to earlier-born Purkinje cells (born  embryonic day, E12.5) con- NE budding. These mechanisms imply a relationship between tained NE buds, whereas only 6% later-born granule cells are bud formation and CNS maturation. It has not been possible to affected (birthdate E13–E17; post-mitotic from P3 onward; Fig- explore this question directly, however, because of the perinatal ure 1B) (Machold and Fishell, 2005; Mizuhara et al., 2010). lethality of torsinA mutant mice and absence of an in vitro model Considered together with our previous findings, these observa- of this process (Goodchild et al., 2005; Liang et al., 2014; Tanabe tions indicate that NE bud formation, which begins following et al., 2012). neuronal migration (Goodchild et al., 2005), continues during Here, we demonstrate that neuronal nuclear membrane post-natal CNS maturation. budding is a developmentally regulated event and identify a Little is understood about the cellular or molecular require- molecular component of this process that controls its timing. ments for neuronal NE budding. To examine whether NE bud for- Neuronal NE budding in torsin mutant neurons occurs initially mation is a neural cell autonomous process, we deleted torsinA following neuronal migration and peaks during a period corre- selectively from neurons by intercrossing floxed torsinA and sponding to synaptic integration and circuit formation. Remark- Synapsin 1-Cre transgenic mice (herein Syn-CKO mice). The ably, NE buds then disappear completely as neurons fully Syn-Cre transgene begins to express in maturing neurons, as mature. This developmental window is strongly modulated by early as E12.5 (Zhu et al., 2001). Syn-CKO mice showed near the torsinA paralog, torsinB. The developmental upregulation complete deletion of torsinA in the spinal cord and brainstem, of endogenous torsinB levels corresponds to the disappearance efficient deletion in cortex, and little or no deletion in cerebellum of buds and, in mice null for both torsinA and torsinB, NE buds and striatum, a pattern corresponding to transgene expression develop earlier (in migrating neurons) and persist into adulthood. (Figures 2A, 2B, and S2A). All regions showing efficient torsinA We model this process in vitro during the neural specification of deletion developed NE buds indistinguishable from those seen DYT1 mutant mouse embryonic stem cells and demonstrate that in N-CKO mice (Figures 2C and S2B), demonstrating that NE overexpression of torsinB (or torsinA) can rescue NE bud forma- budding is a neuronal cell autonomous process. tion. These findings link neuronal NE budding to a discrete neu- To examine the developmental dependence of NE budding rodevelopmental window, provide molecular insight into this during the complete process of neural maturation, we examined process, and have important implications for understanding multiple ages in the striata of Dlx5/6-Cre conditional torsinA mu- the pathogenesis of DYT1 dystonia. tants that live into adulthood (herein Dlx5/6 conditional KO ‘‘Dlx- CKO’’; Pappas et al., 2015). Additional advantages of this model RESULTS are the relative homogeneity of the striatum (95% of striatal cells are medium spiny neurons) and the strong links between striatal Neuronal NE Budding Is a Cell Autonomous Process that dysfunction and dystonia pathogenesis (Pappas et al., 2015; Ta- Occurs during a Discrete Neurodevelopmental Window nabe et al., 2012). These mice exhibited essentially complete Perinatal lethality of constitutive torsinA null mice has limited our deletion of torsinA from the striatum (Figure 2D). Despite early understanding of the relationship between CNS maturation and (E12.5) prenatal deletion of torsinA by Dlx-5/6-Cre (Monory

Cell Reports 16, 3322–3333, September 20, 2016 3323 A Figure 1. Neuronal Nuclear Membrane Budding Increases during Early Post-natal CNS Maturation Conditional CNS deletion of torsinA caused neuronal NE buds in all CNS regions examined. (A) Ultrastructural images of normal control and N-CKO neuronal nuclei with NE budding from P8 cortex (Ctx; first row). The subse- quent rows are examples from spinal cord (SC), pons, Purkinje cells of cerebellum (Cbl), and striatum (Str). The nuclear membrane buds are denoted by arrows (Nucleus = N and Cytosol = C). The scale bar represents 500 nm. (B) Percentage of nuclei with NE buds in the CNS regions shown in (A) at P0 and P8 (n = 2, per region/age) (Purkinje cells = PC and granule cells = GC). The results are expressed as mean ± SEM (one- way Anova; * = p < 0.05; ** = p < 0.01; and **** = p < 0.0001). (C) Stacked bar graphs showing percentage of nuclei (at P0) with different numbers of buds/nucleus by region in all regions examined. (D and E) The number of NE buds/nucleus continues to accumulate from P0 to P8. The stacked bar graphs show percentage of nuclei with different numbers of buds/nucleus in pons (D) and striatum (E) (n = 2, per region/age).

B C

DE

3324 Cell Reports 16, 3322–3333, September 20, 2016 A

B CD

EF G

Figure 2. Neuronal Nuclear Membrane Budding Is a Cell Autonomous Process that Occurs during a Neurodevelopmental Window (A) Western blot demonstrating efficient deletion of torsinA protein in Syn-CKO mice in cortex (Ctx), pons, and spinal cord (SC), but not striatum (Str), cerebellum (Cbl), or liver (n = 2, per region). (B) Quantification of the torsinA western blots displayed in (A). (C) Percentage of nuclei with NE buds in P10 Syn-CKO mice. The results are expressed as mean ± SEM. (D) Western blot showing efficient deletion of torsinA protein from striatum in Dlx-CKO mice. (E) Developmental time course of NE bud frequency in striatum of Dlx-CKO mice from E18.5 to P42 reveals the trajectory of NE bud development, peak, and resolution. (F and G) Striatal lysates from wild-type mice demonstrate that endogenous levels of torsinB levels are low as NE buds accumulate (P0 to P7), and that torsinA and torsin B exhibit opposite changes in levels of expression as NE buds resolve (P7 to P28). The levels of the torsinA-interacting proteins LAP1 and LULL1 remain largely unchanged during this period of CNS maturation (F). The changes for torsinA, torsinB, LAP1, and LULL1 are quantified in (G) as a relative percentage to calnexin control. The results are expressed as mean ± SEM. All lysates in duplicate for western blot. et al., 2006; Pappas et al., 2015), striatal NE buds did not begin to of NE bud-bearing neurons then decreased rapidly during the appear until P3. Similar to N-CKO and Syn-CKO mice, NE second post-natal week (P7–P14) and NE buds were essentially budding frequency increased during CNS maturation, reaching undetectable by P42 (Figure 2E). This disappearance of NE blebs a peak of 35% of neurons at P7. Surprisingly, the percentage is not due to cell death, as striatal size and the number of medium

Cell Reports 16, 3322–3333, September 20, 2016 3325 spiny neurons are normal in Dlx-CKO mice (Pappas et al., 2015), To overcome the embryonic lethality of dKO mice and and assessment by TEM showed no apoptotic nuclei or other ul- examine a potential role for torsinB in modifying torsinA-related trastructural evidence of cell toxicity. Similar to Dlx-CKO mice, NE budding, we used nestin-Cre to conditionally delete these analysis of older Syn-CKO mice (P23) also demonstrated com- from neural progenitor cells (nestin double conditional plete absence of NE buds (Figure S2C). These data demonstrate knockout, herein ‘‘N-dCKO’’; Figure 3B). N-dCKO mice were that neuronal NE budding does not continue to progressively in- recovered at reduced Mendelian ratios in newborn litters, but crease with age as implied by findings in N-CKO mice (Figure 1B) at normal Mendelian ratios at E16.5, indicating that many die and in previous work (Goodchild et al., 2005; Kim et al., 2010). during late gestation (Table S1). All N-dCKO mice identified in Rather, NE budding is confined to a discrete neurodevelopmen- newborn litters died in the first 12 hr of life. Standard histological tal window, implying the existence of factors that modulate this analysis (e.g., Nissl and H&E staining) of the brains of these mice process. did not reveal any gross abnormalities (data not shown). An intriguing candidate as a regulator of NE budding is the In striking contrast to the neurodevelopmental dependence torsinA paralog, torsinB. We reported previously that in vitro of NE buds observed in N-CKO mice (Figure 1B), essentially all torsinA and torsinB have conserved function at the NE and that N-dCKO neurons exhibited NE buds, regardless of maturational in torsinA null tissue, levels of torsinB strongly influence the state (Figures 3C–3H). N-dCKO nuclei (Figure 3D) also exhibited susceptibility of different cell types to the formation of NE buds greater numbers of buds than N-CKO nuclei (Figure 1C). Strik- (e.g., in neuronal versus non-neuronal cells; Kim et al., 2010). ingly, NE buds were also observed in N-dCKO migrating neurons Other work demonstrates differential expression of torsinB (Figures 3E–3H), a developmental stage at which NE buds are both spatially and temporally in mouse and human brain (Bahn never seen in N-CKO (data not shown) or Tor1a null or DE-torsinA et al., 2006; Jungwirth et al., 2010; Vasudevan et al., 2006). knock-in mice (Goodchild et al., 2005). This observation sug- Therefore, we explored whether physiological changes in the gests that torsinB participates in timing of initiation of NE developmental expression of torsinB might correspond to the budding during neurodevelopment. appearance and disappearance of NE buds in Dlx-CKO mice. To determine whether the later increase in torsinB levels (Figures Consistent with this notion, we found that in wild-type mice, 2F and 2G) might likewise be involved in the developmental down- striatal torsinB levels are low during the first post-natal week regulation of NE budding, we examined a role for this protein in when NE buds are accumulating and then increase by more later maturing neurons. This question could not be addressed in than 3-fold during the second post-natal week coincident with N-dCKO (Table S1)orDlx-dCKO(Figure S4A) mice because of NE bud disappearance (Figures 2E–2G). TorsinA levels show the early lethality in these lines. We therefore utilized mice in which the converse pattern, decreasing after the first post-natal week both torsinA and torsinB were deleted using Synapsin 1-Cre (‘‘Syn- (Figures 2F and 2G). Levels of torsinA interacting proteins dCKO’’) (Figure 4A). Syn-dCKO mice were born in normal Mende- LAP1 and LULL1 remain largely unchanged in this time frame lian ratios and survived for up to 6 weeks (median age = 23 days; (Figures 2F and 2G). These observations implicate torsinB in Figure S4B). Whereas NE buds are absent by P23 in Syn-CKO the developmental regulation of neuronal NE budding and sug- mice (Figure S2C),NEbudsremainorcontinuetoincreaseinall gest a potential molecular mechanism underlying the selective structures examined in Syn-dCKO animals (Figures 4B and 4C). vulnerability of immature neurons to torsinA loss of function. Importantly, the early lethality of Syn-dCKO mice—in which dele- tion is exclusively neuronal—links dysregulation of NE-budding TorsinB Modulates the Developmental Dependence of to neuronal dysfunction. Considered together, these data identify Neuronal NE Budding torsinB as a regulator of the opening and closing of the neurodeve- To investigate directly a role for torsinB in neuronal NE budding, lopmental window for NE budding in torsinA mutant mice. we used gene targeting to explore the consequences of Tor1b deletion and the effect of altering torsinB function in the context A DYT1 Mutant Embryonic Stem Cell Model of torsinA loss of function (Figure S3). Intercrossing germline Recapitulates Neurodevelopmental Onset of NE Tor1b+/À mice generated all expected genotypes at normal Budding and Provides a Platform for Molecular Mendelian frequency. Tor1bÀ/À mice were indistinguishable Therapeutics from their littermate controls and showed no NE buds in the Having established in vivo an essential role for torsinB in the clos- CNS or peripheral tissues (including heart, tongue, liver, and kid- ing of the NE budding window (Figure 4B), we next pursued a se- ney; data not shown). Tor1a and Tor1b are adjacently located, ries of experiments to further explore the role of torsinB during precluding the possibility of intercrossing the individual mutant the opening of the NE budding window. Critically, we sought to alleles to create torsinA and torsinB double null mice. We there- explore whether overexpression of torsinA or torsinB could fore generated a floxed allele in which the LoxP sites encompass ‘‘rescue’’ NE budding. In an attempt to translate our findings both Tor1a and Tor1b (Figure 3A). No germline torsinA/torsinB into a more disease-relevant context, we pursued these studies double knockout mice (Tor1(ab) À/À or ‘‘dKO’’) were recovered in the context of DE-torsinA (as exists in DYT1 dystonia sub- from the litters of Tor1(ab)+/À intercrosses. Assessment of prena- jects). Neuronal NE budding of Tor1aDE/DE mice is indistinguish- tal time points demonstrated that germline dKO embryos devel- able from that of torsinA null mice, and NE buds are not observed opmentally arrest at E8.5 (data not shown). Considering the in Tor1a+/+ or Tor1aDE/+ littermate controls (Goodchild et al., lack of phenotype in Tor1bÀ/À mice, this finding is consistent 2005). We therefore derived multiple independent mouse embry- with previous work indicating that torsinA and torsinB share onic stem cell (mESC) lines from an allelic series of DE-torsinA conserved function (Kim et al., 2010). knock-in mice. To test whether neuronal differentiation of the

3326 Cell Reports 16, 3322–3333, September 20, 2016 A B

C DE

FGH

Figure 3. TorsinB Modulates the Timing of Critical Period Opening for Neuronal NE Budding (A) Schematic of the floxed Tor1(ab) allele and product of Cre recombination. (B) Western blot demonstrating deletion of torsinA and torsinB from brain (Br), but not liver (Lv), in N-dCKO mice. (C) Neurons of N-dCKO mice exhibit similarly high levels NE bud formation in areas of differing developmental age/maturational state (P0; n = 2, per region/age). (D) Stacked bar graphs show percentage of nuclei with differing numbers of buds/nucleus by region (P0). (E) N-dCKO mice exhibit NE budding in migrating cortical neurons, a developmental stage in which budding does not occur in N-CKO mice. The quantification of percentage of affected nuclei (n = 2 per age) is shown. (F–H) Ultrastructural images of migrating cortical neurons of N-dCKO animals at E16.5. (F) Low magnification image of several migrating nuclei (N). (G) Higher magnification of elongated migrating nuclei with NE buds. (H) Boxed NE from (G) shows arrows pointing at NE buds at greater magnification. The scale bars represent 1 mm. The results are expressed as mean ± SEM.

Cell Reports 16, 3322–3333, September 20, 2016 3327 A SC Pons Ctx Figure 4. TorsinB Modulates the Timing of Neurodevelopmental Window Closure for Syn- Syn- Syn- NE Budding ctrl ctrl ctrl dCKO dCKO dCKO (A) Western blot demonstrating conditional dele- tion of torsinA and torsinB protein in spinal cord 1212 1212 1212 (SC), pons, and cortex (Ctx) in Syn-dCKO mice. Note that Syn-CKO expresses exclusively within TorsinA - neurons, so much of the remaining protein likely derives from non-neuronal cells. (B) NE budding continues to increase in Syn- TorsinB - dCKO neurons from P10 to P23, when it resolves completely in Syn-CKO mice (n = 2, per region/ age) (one-way Anova; * = p < 0.05). Calnexin - (C) Stacked bar graph shows percentage of nuclei (for torsinA) with differing numbers of buds/nucleus (P10 and P23). Note that the NE buds/nucleus continue Calnexin - to accumulate during this time. The results are (for torsinB) expressed as mean ± SEM.

B C 1-20 21-40 >40 80 * P10 100 logically indistinguishable from those P23 observed in vivo (Figure 5C), and the percentage of affected cells increased 80 60 with neural maturation (Figure 5D; day 5 5% and day 13 60%). Similar to the 60 neural specificity of NE buds in vivo, 40 no nuclear buds were observed when 40 Tor1aDE/DE mESCs were driven down the cardiomyocyte lineage (Figure 5C). 20 Strikingly, opposite patterns of torsinB % Nuclei w/ NE buds NE w/ % Nuclei 20 expression were observed in these two Frequency buds/nucleus NE 0 0 lineages. TorsinB levels decreased dur- SC Pons Ctx ing neural differentiation, but increased during cardiomyocyte differentiation (Fig- ures 5E and 5F), establishing another SC (P10)SC (P23)ons (P10)ons (P23)Ctx (P10)Ctx (P23) P P context in which levels of torsinB anti- correlate with the formation of NE buds. These data demonstrate that the mESC mESC clones replicates a maturation-related budding phenom- differentiation paradigms recapitulate fundamental attributes enon selectively in neurons, we drove Tor1a+/+ and Tor1aDE/DE of torsinA-associated NE budding seen in vivo: neuronal cell mESC clones down either the neuronal or cardiomyocyte line- specificity and autonomy, developmental dependence, and ages (Figure 5A). We utilized a paradigm in which the mESC anti-correlation with levels of torsinB. were differentiated into embryoid bodies (EB), treated with either Having established this in vitro platform, we tested whether retinoic acid (neural lineage) or ascorbic acid (cardiomyocyte increasing levels of torsinB could suppress NE bud formation. lineage), and subsequently plated onto an appropriate substrate This experiment further tests the role of torsinB in NE bud dy- for further maturation (PDL/laminin for neurons or gelatin for namics and—in the context of DE-torsinA—is relevant to the cardiomyocytes; Figure 5A). No differences in maturation were development of DYT1 therapeutics. We transduced EBs with observed between Tor1a+/+ and Tor1aDE/DE when driven down GFP-tagged-torsinA, -torsinB, or -torsin2-expressing lentivi- either lineage. After 5 days of RA exposure, both genotypes ruses on RA-differentiation day 2, when EBs are beginning to ac- differentiated into GABAergic neurons expressing medium chain quire neuroprogenitor characteristics (Bain et al., 1996). Both neurofilament and GAD65/67 (Figure 5B) and developed exten- torsinA and torsinB significantly decreased the percentage of sive neuronal processes following plating onto PDL/laminin. nuclei with NE buds (by nearly 50%). Torsin2 had no significant Similarly, cultures of both genotypes developed foci of contract- effect (Figure 5G), but also appeared to potentially reduce NE ing cells 2 days following ascorbic acid exposure (Movie S1). buds, raising the possibility that other torsin proteins may be In vivo, NE buds emerge selectively in post-migratory maturing involved in this process. Transduction of EBs did not alter the neurons (Goodchild et al., 2005) and increase during neuronal trajectory of neuronal differentiation, as all virally transduced maturation (Figures 1 and 2). We observed a similar pattern of EBs expressed NeuN and GABA at the same rate (Figure S5A) NE bud development during in vitro neuronal differentiation: and to the same extent as non-transduced EBs, and transduc- Tor1aDE/DE neuronal EBs developed NE buds that were morpho- tion efficiency was similar among the different torsin isoforms

3328 Cell Reports 16, 3322–3333, September 20, 2016 A C D TEM 70 +RA N ****

ES ) C

+/+ 60 +AA CM Neuron Tor1a

( 50 Time (d) 0 3 5 7 10 15 N RA N 40

PDL + laminin ) **** E C AA CM E/ Gelatin 30 Neuron Tor1a

+/+ E/ E ( B Tor1a Tor1a % Nuclei w/ NE buds 20 Time (d) 035710035710 n.s. ) NF-M - E C 10 E/ GAD65/67 - 0 5 7 10 13 Tor1a -tubulin - (

Cardiomyocyte Age (d)

EFG Neuron Cardiomyocyte TorsinA (neuron) Time (d) 034 715 ES RA (+RA) (+AA) TorsinB (neuron) TorsinA (CM) Lentivirus Time (d) 0235103510 TorsinB (CM) PDL + laminin 600 n.s. TorsinA - 50 ** 500 40 **** TorsinB - 400 30 300 LAP1 - 20 200 * 100 10

LULL1 - % Nuclei w/ NE buds Relative Percentage (%) 0 0 0235 10 Calnexin - vehicle Age (d) GFP-torsinAGFP-torsinBGFP-torsin2A

Figure 5. TorsinB Overexpression Suppresses NE Budding during the Maturation of DYT1 Mutant Neurons (A) Diagrammatic overview of experimental design. The wild-type and DYT1 mutant mESCs were grown in suspension for 2 days to form EBs. They were then driven down either a neural or cardiomyocyte lineage. For the neural lineage, they were exposed to RA for 5 days and subsequently plated on poly-D-lysine (PDL) and laminin for up to 8 days, during which time they extended processes and neuronal (N) markers. For the cardiomyocyte (CM) lineage, they were exposed to ascorbic acid (AA) for 5 days and subsequently plated on gelatin for up to 8 days; these cultures developed beating foci within 48 hr of AA exposure. (B) Western blots of NF-M and GAD65/67 demonstrating that control and DYT1 mutant mESCs differentiate into neurons with a similar time course following RA exposure. g-tubulin was used as loading control. (C) Ultrastructural analysis of mutant neurons and muscle demonstrates the development of NE buds selectively in the neural lineage that are indistinguishable from those observed in mouse brain. (D) Percentage of neuronal nuclei with buds increases as the cells mature on PDL/laminin (n = 2 per time point). (E) Expression of torsinA, torsinB, LAP1, and LULL1 demonstrates a unique divergence of torsinB expression levels during neural and cardiomyocyte differ- entiation. Note that the asterisk denotes the major immunoreactive band of LULL1 in the cardiomyocyte lysate. (F) Relative quantification of torsinA and torsinB levels shown in (E). (G) Introduction of lentiviral GFP-torsinA, GFP-torsinB, or GFP-torsin2A midway through neuronal differentiation. Both torsinA and torsinB rescued NE budding phenotype in mutant neuronal EBs (Mann-Whitney: compared to vehicle, GFP-torsinA: p = 0.000094 and GFP-torsinB: p = 0.006365; p = 0.112278; ** = p < 0.01; and **** = p < 0.0001). The results are expressed as mean ± SEM.

(Figure S5B). These further support an inverse relationship DISCUSSION between torsinB levels and NE bud formation. Considered together, these results establish the utility of this model system Our studies establish a neurodevelopmental window for torsinA- for identifying factors that can mitigate or prevent the deleterious related NE budding during CNS maturation in vivo. These effects of torsinA loss of function at the NE. data demonstrate that NE budding is a neurodevelopmentally

Cell Reports 16, 3322–3333, September 20, 2016 3329 regulated process and describe the lethal consequences for the disorders (Connors et al., 2008; Hoftman and Lewis, 2011; Shaw organism when it is allowed to continue unabated. A regulated et al., 2010). Our current findings, previous work identifying other role for NE budding during development is consistent with torsinA loss-of-function pathologies confined to early post-natal several previous observations of NE buds in developmental development (Liang et al., 2014; Pappas et al., 2015; Weisheit contexts (Gay, 1956; Hadek and Swift, 1962; Hochstrasser and Dauer, 2015), and the clinical phenomenon of delayed-onset and Sedat, 1987b; Speese et al., 2012; Szo¨ llo¨ si and Szo¨ llo¨ si, dystonia (Burke et al., 1980), suggest that this model of disease 1988). Considered together, these observations suggest that pathogenesis may also apply to DYT1 and other childhood- NE budding may be a normal cellular process, and the appear- onset primary dystonias. ance of buds may reflect a stalled structural intermediate, the To provide mechanistic understanding of NE budding and progression of which is slowed or blocked by the absence of identify therapeutic targets for DYT1 dystonia, we next sought torsinA function. The lethality that occurs when this process factors that modulate this neurodevelopmental window. We is continually dysregulated—including selectively within neu- and others have documented marked differences in the levels rons—support its essential role in CNS function. We also identify of torsinB between neural and non-neural tissue that correlate torsinB as a potent molecular modifier of torsinA-dependent NE with susceptibility to NE bud formation (Jungwirth et al., 2010; budding, demonstrating its ability to strongly influence both the Kim et al., 2010). Data from rodent and human brains indicate opening and closing of the neurodevelopmental window for that levels of torsinB are low prenatally and increase substan- this process, and show that overexpression of torsinB can sup- tially thereafter (Bahn et al., 2006; Vasudevan et al., 2006). The press NE bud formation in an in vitro model of DYT1 dystonia. timing of increased torsinB expression presented an ideal oppor- These findings provide a powerful platform for further elucidation tunity to study the relationship of torsinB levels and NE buds in of the biological roles of NE budding during neural development striatum, a structure strongly implicated in dystonia. Consistent and highlight the importance of studying the developing CNS with a potential role for torsinB in regulating NE bud dynamics, to identify torsinA processes relevant to the pathogenesis of torsinB levels remain low during the peak of NE bud formation, dystonia. but increase by 3-fold coincident with NE bud disappearance DYT1 dystonia is 30% penetrant and essentially no mutation (Figures 2F and 2G). carriers that remain unaffected by their 20’s subsequently To directly test the role of torsinB in NE bud dynamics, we develop symptoms. This pattern suggests that pathogenic experimentally manipulated torsinB levels in several in vivo and torsinA is capable of disrupting motor circuits only during an in vitro contexts. In contrast to the developmental gradient early neurodevelopmental window. We explored this possibility observed in N-CKO mice, nearly all N-dCKO neurons exhibited by conditionally deleting torsinA from the murine CNS and buds, regardless of developmental age (Figures 3C and 3D). analyzing NE budding throughout CNS maturation. Using this Notably, loss of torsinB on the torsinA null background dramat- approach, we identify a strong relationship between develop- ically lengthened the neurodevelopmental window, causing NE mental age and susceptibility to torsinA loss of function. buds to form in migrating neurons (Figures 3E–3H) and to persist Whereas previous work implied that NE buds continue to accu- in mature neurons, maturational states in which NE buds are mulate without end, we now demonstrate that NE buds disap- never seen in torsinA null mice. The neurodevelopmental window pear in all mutant lines that survive into adulthood (Syn-CKO failed to close in the absence of torsinB (Figures 4B and 4C), and Dlx-CKO; Figures 2E and S2C). Considered together, these causing a progressive increase in NE buds and early lethality. data establish an essential requirement for torsinA function Importantly, Syn-CKO mice show normal viability, whereas during a neurodevelopmental window during CNS maturation. Syn-dCKO mice die by 6 weeks of age. Markedly earlier lethality Considerable uncertainty exists about the human age that cor- was also seen in all other dKO mice compared to torsinA deletion responds to the early post-natal stage we identify as critically alone, strengthening the connection between NE buds and dependent upon normal torsinA function. This period, however, neuronal function. Conversely, we demonstrate that experimen- is likely earlier than the early childhood period when dystonic tally increasing levels of torsinB can suppress NE bud formation symptoms emerge in DYT1 subjects. We propose a model in an in vitro model of DYT1 dystonia. These data establish the whereby early abnormalities of neural circuits consequent to changes during CNS maturation of endogenous torsinB levels torsinA loss of function disrupt the highly dynamic develop- as a physiological mechanism modulating the window of vulner- mental trajectory that occurs throughout childhood, resulting in ability to torsinA loss of function. Considered with previous work the later emergence of dystonic symptoms. Consistent with (Kim et al., 2010; Zhao et al., 2013), it is most likely that torsinA this model, behavioral abnormalities emerge and subsequently and torsinB share a redundant function at the NE and that a worsen when NE buds have largely resolved in Dlx-CKO striatum developmental window for NE budding reflects a time when (Pappas et al., 2015). NE buds peak at P7, yet are largely absent aggregate torsin protein function (represented by the combina- by P14 (Figure 2E). However, Dlx-CKO mice are indistinguish- tion of torsinA and torsinB levels) dips below a critical threshold. able from littermate controls until P14, when abnormal twisting The ability of torsinA or torsinB to suppress NE budding in movements emerge, and progressive abnormalities on the grid immature neurons contrasts with our previous work (Kim et al., hang test do not emerge until P21, and then worsen over the 2010), demonstrating that overexpression of these proteins in- next 2–3 weeks. The concept of early deleterious events causing creases NE budding in more mature primary neuronal cultures, symptoms to emerge much later, consequent to altered further highlighting a critical role for developmental timing in tor- neurodevelopmental trajectories, is well established in studies sin protein family function emphasized by our in vivo studies. The of autism and schizophrenia, among other neurodevelopmental finding that torsinB can effect rescue is critical, because any

3330 Cell Reports 16, 3322–3333, September 20, 2016 therapeutic strategy based on increasing endogenous torsinA pattern of torsinB as a molecular mechanism governing the tem- levels will necessarily increase DYT1 mutant torsinA, which poral window of neural susceptibility to torsinA loss of function. could negate any beneficial increases from the wild-type protein The importance of this process for dystonia pathogenesis is through dominant-negative effects. In contrast, modulation of supported by the presence of NE budding in LAP1 null mice torsinB should overcome this obstacle. Considered together, and the recent connection of LAP1 to human dystonia (Dorboz our observations demonstrate that torsinB is a potent modifier et al., 2014). Our development of an in vitro system modeling of torsinA phenotypes and a potential therapeutic target. Future this phenomenon, and the demonstration that NE budding can studies will be required to determine the precise role of torsinB in be rescued by torsinB, establish a powerful platform to further this process, whether overexpression of torsinB can suppress dissect this biological process. NE bud formation in vivo, and if this will ameliorate the behavioral phenotypes in torsinA mutant mice. EXPERIMENTAL PROCEDURES The discrete window during which NE buds form implicates several developmental events as potentially related to this pro- Mouse Breeding and Genotyping cess. One intriguing possibility is that torsin proteins function in Animal testing was conducted in accord with the NIH laboratory animal care guidelines and with the Institutional Animal Care and Use Committee interphase nuclear pore insertion, a process that is markedly up- at the University of Michigan. Prenatal birthdating of mice was determined regulated in maturing neurons (Lodin et al., 1978). Several yeast by the presence of a vaginal plug, designated as E0.5. For post-natal birth- strains mutant for nucleoporins develop structures similar to NE dating, the day of birth was designated as P0. Nestin-Cre, Syn-Cre, and buds (Siniossoglou et al., 1996; Wente and Blobel, 1994; Zabel Dlx5/6-Cre mice were obtained from The Jackson Laboratory. Tor1a floxed et al., 1996), and torsinA loss of function in C. elegans causes nu- mice were described previously (Liang et al., 2014). Tor1b floxed mice were clear pore abnormalities and NE bud-like structures (VanGompel generated by introducing LoxP sites in the Tor1b introns in between exons 2and3andafterexon5(Figure S3). Tor1(ab) floxed animals contain LoxP et al., 2015), potentially linking these processes. In the striatum, sites in the Tor1a intron between exons 2 and 3 and the Tor1b intron be- the period of NE bud formation corresponds to the time that neu- tween exons 2 and 3 (Figure 3). Both Tor1b and Tor1(ab) floxed mice are rons are integrating into circuits, when profound changes in neu- generated by the standard method, as described previously (Liang et al., ral physiology occur, including the formation of projections and 2014). Mouse genotyping and breeding is described in detail in Supple- synapses (Uryu et al., 1999). These events impact the NE by up- mental Information. regulating transcription—visually apparent from the transition from hetero- to euchromatin in maturing neurons—and transla- Generation of mESC Clones +/+ DE/DE tion, increasing the need for nucleo-cytosolic transport to deliver The Tor1a and Tor1a mESC lines were generated by superovulating Tor1aDE/+ female mice and mating them with Tor1aDE/+ male mice. At 3.5 mRNAs to the cytosol. Transcription and translation are associ- dpc, blastocysts were flushed from the ovaries of female mice that displayed ated with an increase in nuclear pore number in interphase vaginal plugs. Blastocysts were plated on a mitomycin-C treated mouse em- cells (Doucet and Hetzer, 2010; Maul et al., 1980). In the CNS, bryonic fibroblast (MEF) feeder layer. During the next 4 days blastocysts the number of nuclear pores increases dramatically during hatched and attached to the plate. More MEFs were added on day 4 of plating. cortical neuron maturation (roughly 5-fold; Lodin et al., 1978) The following day and every day after that, cells were fed with fresh ES medium and following increased synaptic activity (Wittmann et al., containing a Mek1/2 inhibitor (U0126; Cell Signaling Technology) until ES colonies were observed and plentiful. At day 8 and every few days after that 2009). Synaptic activity increases nuclear membrane surface the well was trypsinized and passed to another well with an established feeder area as well as nuclear pore number, a form of structural plas- layer for each blastocyst. This was done repeatedly and the clones were ticity that may reflect adaptation to a metabolically more active expanded to a 10-cm plate and frozen down. state (Wittmann et al., 2009). Increasing nuclear membrane sur- face area and related neurodevelopmental events (e.g., process mESC Culture and In Vitro Differentiation formation) require membrane synthesis. TorsinA has been linked Neural Lineage to lipid metabolism (Grillet et al., 2016) and several observations mESCs were plated and maintained for two passages on treated tissue culture in yeast demonstrate that disruption of phospolipid and related plates coated with 0.1% gelatin and mitotically inactivated MEF cells, followed by two additional passages on gelatin coated plates without MEF feeders pathways can cause NE abnormalities, some of which resemble (Millipore) in ES medium (DMEM with 4,500 mg/l glucose and 2,250 mg/l NE buds (Schneiter et al., 1996; Siniossoglou, 2009). Finally, a Na-bicarbonate supplemented with 13 non-essential amino acids, 13 nucle- series of observations suggests the presence of a nuclear osides [Specialty Media], 0.1 mM b-mercaptoethanol [Sigma], 2 mM L-gluta- pore-independent form of nucleo-cytosolic transport at specific mine [Gibco], penicillin/streptomycin [Gibco], 15% FBS [HyClone], and developmental stages and at times of high metabolic demand 1,000 units/ml LIF [Chemicon]). Prior to differentiation, mESCs were trypsi- 3 6 (Speese et al., 2012). This process appears to transport large nized, dissociated, and plated as single cells in suspension (2.0 10 cells in 15 ml serum free differentiation medium). These cells were grown in aggre- ribonuclear particles directly through the membrane in a manner gate cultures for 2 days to form EBs in serum-free medium (Advanced DMEM/ akin to herpesvirus budding (Speese et al., 2012), but likely F12 [Gibco] and Neurobasal [Gibco] media in a 1:1 ratio supplemented with differs in important respects since herpesvirus budding pro- KnockOut Serum Replacement [10% by volume; Invitrogen], penicillin/strep- ceeds normally in the absence of torsin proteins (Turner et al., tomycin [1.0% by volume; Sigma], L-glutamine (200 mM), and b-mercaptoe- 2015). TorsinA, a membrane associated protein, may participate thanol [0.1 mM; [Sigma]). Following 2 days in suspension culture, EBs were in any of these processes, which are likely strongly upregulated split 1:2 and treated with retinoic acid (RA; Sigma) dissolved in dimethyl-sulf- oxide (1 mM; Sigma; 0.1% of total volume). EBs were cultured for a total of in maturing neurons. 5 days in RA-containing medium. They were then transferred to PDL/laminin Our findings fundamentally advance understanding of torsinA- (Sigma) coated plates in non-RA containing maturation medium (Advanced related NE budding, linking this process to a discrete neurodeve- DMEM/F12 and Neurobasal media, B27 supplement [2.5% by volume; Invitro- lopmental window, and identify the developmental expression gen], L-glutamine [200 mM], penicillin/streptomycin [1.0% by volume]).

Cell Reports 16, 3322–3333, September 20, 2016 3331 Cardiomyocyte Lineage ACKNOWLEDGMENTS Following EB formation (48 hr), EBs were split 1:2 and replated in suspension culture plates in the same media described above, except with 10% FBS and We thank Roger Albin and members of the Dauer lab for their careful reading of the addition of 0.1 mg/ml ascorbic acid (Sigma). EBs were exposed to this and suggestions for this manuscript. We also thank Rose Goodchild and Chris- formulation for 5 days. EBs were then transferred to gelatin-coated tissue cul- tian Schlieker for the kind gift of antibodies to torsinB and Christian Schlieker ture treated plates in the same media as used for differentiation except without for antibodies to LAP1 and LULL1. We thank Hynek Wichterle for early help 0.1 mg/ml ascorbic acid. EBs adhered to plates after 24 hr and beating foci with neural differentiation of mESC. This research project was supported in were detected after 48 hr. part by Bachmann-Strauss Dystonia and Parkinson Disease Foundation, a Fellowship from the Dystonia Medical Research Foundation (to L.M.T.), the Protein Extraction from Tissue and Cells and Western Blotting Robert P. Apkarian Integrated Electron Microscopy Core of Emory University, For tissue extraction, animals were heavily anesthetized with ketamine and xy- and a grant from the National Institute of Neurological Disorders and Stroke lazine, intraperitoneally (i.p.), and transcardially perfused with ice cold PBS. (1R01NS077730 to W.T.D.). Tissue was lysed by sonicating with 1.0% SDS lysis buffer containing 100 mM Tris-HCl (pH 8.0), 10% glycerol, 150 mM NaCl, 2 mM EDTA, and pro- Received: March 9, 2016 tease inhibitor cocktail (Sigma). Protein concentration was determined with Revised: June 19, 2016 BCA assay (Pierce). Proteins were denatured prior to SDS-PAGE by boiling Accepted: August 14, 2016 with Laemmli’s sample buffer (Bio-Rad) containing b-mercaptoethanol for Published: September 20, 2016 5 min. After SDS-PAGE, proteins were transferred to nitrocellulose membrane, which was blocked with blocking buffer (5.0% skim milk in PBS with 0.2% REFERENCES Tween) for 1 hr at room temperature. Blocked membrane was incubated in  blocking buffer containing primary antibody overnight at 4 C. Membrane Bahn, E., Siegert, S., Pfander, T., Kramer, M.L., Schulz-Schaeffer, W.J., He- was washed with washing buffer (PBS with 0.2% Tween; PBS-T) and then wett, J.W., Breakefield, X.O., Hedreen, J.C., and Rosta´ sy, K.M. (2006). TorsinB incubated in blocking buffer with HRP-conjugated secondary antibody for expression in the developing human brain. Brain Res. 1116, 112–119. 1 hr at room temperature. The washed membrane was developed using Bain, G., Ray, W.J., Yao, M., and Gottlieb, D.I. (1996). Retinoic acid promotes chemiluminescent substrate for HRP (Pierce) and exposed on film (Kodak neural and represses mesodermal in mouse embryonic stem X-omat LS films or Amersham Hyperfilm). All western blots were quantified cells in culture. Biochem. Biophys. Res. Commun. 223, 691–694. with densitometry using ImageJ. All levels were calculated relative to calnexin loading control. Brown, R.S.H., Zhao, C., Chase, A.R., Wang, J., and Schlieker, C. (2014). The mechanism of Torsin ATPase activation. Proc. Natl. Acad. Sci. USA 111, E4822–E4831. Antibodies The dilution rates for each antibody used in this study are listed as follows. anti- Burke, R.E., Fahn, S., and Gold, A.P. (1980). Delayed-onset dystonia in torsinA (rabbit polyclonal; Abcam; 1:10,000); anti-torsinB (rabbit polyclonal; patients with ‘‘static’’ encephalopathy. J. Neurol. Neurosurg. Psychiatry 43, kind gifts of Dr. Rose Goodchild; VIB Center for the Biology of Disease and 789–797. Dr. Christian Schlieker; Yale University; 1:1,000); anti-LAP1 and anti-LULL1 Clark, W.H., Jr. (1960). Electron microscope studies of nuclear extrusions in (rabbit polyclonal; kind gift of Dr. Christian Schlieker; Yale University; pancreatic acinar cells of the rat. J. Biophys. Biochem. Cytol. 7, 345–352. 1:1,000); anti-calnexin (rabbit polyclonal; Stressgen; 1:10,000); anti-GAD65/ Connors, S.L., Levitt, P., Matthews, S.G., Slotkin, T.A., Johnston, M.V., Kinney, 67 (rabbit polyclonal; Millipore; 1:2,000); anti-neurofilament-M (rabbit poly- H.C., Johnson, W.G., Dailey, R.M., and Zimmerman, A.W. (2008). Fetal mech- clonal; Millipore; 1:200); g-tubulin (monoclonal; Sigma; 1:2,000); anti-NeuN anisms in neurodevelopmental disorders. Pediatr. Neurol. 38, 163–176. (Monoclonal A-60; Millipore; 1:200); anti-GABA (rabbit polyclonal; Sigma; Dorboz, I., Coutelier, M., Bertrand, A.T., Caberg, J.-H., Elmaleh-Berge` s, M., 1:200); anti-GFP (rabbit polyclonal; Abcam; 1:200); Goat anti-Rabbit IgG Horse Laine´ , J., Stevanin, G., Bonne, G., Boespflug-Tanguy, O., and Servais, L. Radish Peroxidase and Goat anti-Mouse IgG Horse Radish Peroxidase (Cell (2014). Severe dystonia, cerebellar atrophy, and cardiomyopathy likely caused signaling; 1:10,000); and anti-mouse Alexa Fluor 488, anti-rabbit Alexa Fluor by a missense mutation in TOR1AIP1. Orphanet J. Rare Dis. 9, 174. 488, and anti-rabbit Alexa Fluor 633 (Invitrogen; 1:400). Doucet, C.M., and Hetzer, M.W. (2010). Nuclear pore biogenesis into an intact Statistics nuclear envelope. Chromosoma 119, 469–477. GraphPad Prism software (GraphPad Software) was used to analyze all data. Gay, H. (1956). Nucleocytoplasmic relations in Drosophila. Cold Spring Harb. Means are displayed with error bars representing SEM. One-way ANOVA Symp. Quant. Biol. 21, 257–269. tests were followed by post hoc Dunnett test, where appropriate. Mouse Gonzalez-Alegre, P., and Paulson, H.L. (2004). Aberrant cellular behavior of survival probability was analyzed using the Kaplan and Meier method. mutant torsinA implicates nuclear envelope dysfunction in DYT1 dystonia. Analyses of NE buds quantification for the control condition of wild-type J. Neurosci. 24, 2593–2601. mice is not included because we never observe NE budding in wild-type tis- Goodchild, R.E., and Dauer, W.T. (2004). Mislocalization to the nuclear enve- sue or cells, as initially reported (Goodchild et al., 2005), and confirmed in lope: an effect of the dystonia-causing torsinA mutation. Proc. Natl. Acad. Sci. extensive experiments since (Kim et al., 2010; Weisheit and Dauer, 2015). USA 101, 847–852. Similarly, we never observe NE buds in wild-type ESCs driven down a neural lineage. Goodchild, R.E., Kim, C.E., and Dauer, W.T. (2005). Loss of the dystonia-asso- ciated protein torsinA selectively disrupts the neuronal nuclear envelope. Neuron 48, 923–932. SUPPLEMENTAL INFORMATION Grillet, M., Dominguez Gonzalez, B., Sicart, A., Po¨ ttler, M., Cascalho, A., Supplemental Information includes Supplemental Experimental Procedures, Billion, K., Hernandez Diaz, S., Swerts, J., Naismith, T.V., Gounko, N.V., five figures, one table, and one movie and can be found with this article online et al. (2016). Torsins are essential regulators of cellular lipid metabolism. at http://dx.doi.org/10.1016/j.celrep.2016.08.044. Dev. Cell 38, 235–247. Hadek, R., and Swift, H. (1962). Nuclear extrusion and intracisternal inclusions AUTHOR CONTRIBUTIONS in the rabbit blastocyst. J. Cell Biol. 13, 445–451. Hochstrasser, M., and Sedat, J.W. (1987a). Three-dimensional organization of L.M.T., C.-C.L., and W.T.D. designed the experiments and wrote the paper. Drosophila melanogaster interphase nuclei. II. spatial organiza- L.M.T. and C.-C.L. conducted experiments. tion and gene regulation. J. Cell Biol. 104, 1471–1483.

3332 Cell Reports 16, 3322–3333, September 20, 2016 Hochstrasser, M., and Sedat, J.W. (1987b). Three-dimensional organization of long-chain fatty acid synthesis to the structure and function of the nuclear Drosophila melanogaster interphase nuclei. I. Tissue-specific aspects of poly- membrane-pore complex. Mol. Cell. Biol. 16, 7161–7172. tene nuclear architecture. J. Cell Biol. 104, 1455–1470. Shaw, P., Gogtay, N., and Rapoport, J. (2010). Childhood psychiatric disor- Hoftman, G.D., and Lewis, D.A. (2011). Postnatal developmental trajectories of ders as anomalies in neurodevelopmental trajectories. Hum. Brain Mapp. neural circuits in the primate prefrontal cortex: identifying sensitive periods for 31, 917–925. vulnerability to schizophrenia. Schizophr. Bull. 37, 493–503. Siniossoglou, S. (2009). Lipins, lipids and nuclear envelope structure. Traffic Jokhi, V., Ashley, J., Nunnari, J., Noma, A., Ito, N., Wakabayashi-Ito, N., 10, 1181–1187. Moore, M.J., and Budnik, V. (2013). Torsin mediates primary envelopment Siniossoglou, S., Wimmer, C., Rieger, M., Doye, V., Tekotte, H., Weise, C., of large ribonucleoprotein granules at the nuclear envelope. Cell Rep. 3, Emig, S., Segref, A., and Hurt, E.C. (1996). A novel complex of nucleoporins, 988–995. which includes Sec13p and a Sec13p homolog, is essential for normal nuclear Jungwirth, M., Dear, M.L., Brown, P., Holbrook, K., and Goodchild, R. (2010). pores. Cell 84, 265–275. Relative tissue expression of homologous torsinB correlates with the neuronal Sosa, B.A., Demircioglu, F.E., Chen, J.Z., Ingram, J., Ploegh, H.L., and specific importance of DYT1 dystonia-associated torsinA. Hum. Mol. Genet. Schwartz, T.U. (2014). How lamina-associated polypeptide 1 (LAP1) activates 19, 888–900. Torsin. eLife 3, e03239. Jungwirth, M.T., Kumar, D., Jeong, D.Y., and Goodchild, R.E. (2011). The nu- Speese, S.D., Ashley, J., Jokhi, V., Nunnari, J., Barria, R., Li, Y., Ataman, B., clear envelope localization of DYT1 dystonia torsinA-DE requires the SUN1 Koon, A., Chang, Y.-T., Li, Q., et al. (2012). Nuclear envelope budding enables LINC complex component. BMC Cell Biol. 12,24. large ribonucleoprotein particle export during synaptic Wnt signaling. Cell 149, Kim, C.E., Perez, A., Perkins, G., Ellisman, M.H., and Dauer, W.T. (2010). A mo- 832–846. lecular mechanism underlying the neural-specific defect in torsinA mutant Strambio-De-Castilla, C. (2013). Jumping over the fence: RNA nuclear export mice. Proc. Natl. Acad. Sci. USA 107, 9861–9866. revisited. Nucleus 4, 95–99. Liang, C.-C., Tanabe, L.M., Jou, S., Chi, F., and Dauer, W.T. (2014). TorsinA Szo¨ llo¨ si, D. (1965). Extrusion of nucleoli from pronuclei of the rat. J. Cell Biol. hypofunction causes abnormal twisting movements and sensorimotor circuit 25, 545–562. neurodegeneration. J. Clin. Invest. 124, 3080–3092. Szo¨ llo¨ si, M.S., and Szo¨ llo¨ si, D. (1988). ‘Blebbing’ of the nuclear envelope of Lodin, Z., Blumajer, J., and Mares, V. (1978). Nuclear pore complexes in cells mouse zygotes, early embryos and hybrid cells. J. Cell Sci. 91, 257–267. of the developing mouse cerebral cortex. Acta Histochem. 63, 74–79. Tanabe, L.M., Martin, C., and Dauer, W.T. (2012). Genetic background modu- Machold, R., and Fishell, G. (2005). Math1 is expressed in temporally discrete lates the phenotype of a mouse model of DYT1 dystonia. PLoS ONE 7, e32245. pools of cerebellar rhombic-lip neural progenitors. Neuron 48, 17–24. Turner, E.M., Brown, R.S.H., Laudermilch, E., Tsai, P.-L., and Schlieker, C. Maric, M., Haugo, A.C., Dauer, W., Johnson, D., and Roller, R.J. (2014). (2015). The Torsin activator LULL1 is required for efficient growth of herpes Nuclear envelope breakdown induced by herpes simplex virus type 1 involves simplex virus 1. J. Virol. 89, 8444–8452. the activity of viral fusion proteins. Virology 460-461, 128–137. Maul, G.G., Deaven, L.L., Freed, J.J., Campbell, G.L., and Bec¸ ak, W. (1980). Uryu, K., Butler, A.K., and Chesselet, M.F. (1999). Synaptogenesis and ultra- Investigation of the determinants of nuclear pore number. Cytogenet. Cell structural localization of the polysialylated neural cell adhesion molecule in Genet. 26, 175–190. the developing striatum. J. Comp. Neurol. 405, 216–232. Mizuhara, E., Minaki, Y., Nakatani, T., Kumai, M., Inoue, T., Muguruma, K., Sa- VanGompel, M.J.W., Nguyen, K.C.Q., Hall, D.H., Dauer, W.T., and Rose, L.S. sai, Y., and Ono, Y. (2010). Purkinje cells originate from cerebellar ventricular (2015). A novel function for the Caenorhabditis elegans torsin OOC-5 in nucle- zone progenitors positive for Neph3 and E-cadherin. Dev. Biol. 338, 202–214. oporin localization and nuclear import. Mol. Biol. Cell 26, 1752–1763. Monory, K., Massa, F., Egertova´ , M., Eder, M., Blaudzun, H., Westenbroek, R., Vasudevan, A., Breakefield, X.O., and Bhide, P.G. (2006). Developmental pat- Kelsch, W., Jacob, W., Marsch, R., Ekker, M., et al. (2006). The endocannabi- terns of torsinA and torsinB expression. Brain Res. 1073-1074, 139–145. noid system controls key epileptogenic circuits in the hippocampus. Neuron Weisheit, C.E., and Dauer, W.T. (2015). A novel conditional knock-in approach 51, 455–466. defines molecular and circuit effects of the DYT1 dystonia mutation. Hum. Mol. Naismith, T.V., Heuser, J.E., Breakefield, X.O., and Hanson, P.I. (2004). Genet. 24, 6459–6472. TorsinA in the nuclear envelope. Proc. Natl. Acad. Sci. USA 101, 7612–7617. Wente, S.R., and Blobel, G. (1994). NUP145 encodes a novel yeast glycine- Naismith, T.V., Dalal, S., and Hanson, P.I. (2009). Interaction of torsinA with leucine-phenylalanine-glycine (GLFG) nucleoporin required for nuclear enve- its major binding partners is impaired by the dystonia-associated DeltaGAG lope structure. J. Cell Biol. 125, 955–969. deletion. J. Biol. Chem. 284, 27866–27874. Wittmann, M., Queisser, G., Eder, A., Wiegert, J.S., Bengtson, C.P., Hellwig, Nery, F.C., Zeng, J., Niland, B.P., Hewett, J., Farley, J., Irimia, D., Li, Y., Wiche, A., Wittum, G., and Bading, H. (2009). Synaptic activity induces dramatic G., Sonnenberg, A., and Breakefield, X.O. (2008). TorsinA binds the KASH changes in the geometry of the cell nucleus: interplay between nuclear struc- domain of nesprins and participates in linkage between nuclear envelope ture, histone H3 phosphorylation, and nuclear calcium signaling. J Neurosci. and cytoskeleton. J. Cell Sci. 121, 3476–3486. 29, 14687–14700. Ozelius, L.J., Hewett, J.W., Page, C.E., Bressman, S.B., Kramer, P.L., Shalish, Zabel, U., Doye, V., Tekotte, H., Wepf, R., Grandi, P., and Hurt, E.C. (1996). C., de Leon, D., Brin, M.F., Raymond, D., Corey, D.P., et al. (1997). The early- Nic96p is required for nuclear pore formation and functionally interacts with onset torsion dystonia gene (DYT1) encodes an ATP-binding protein. Nat. a novel nucleoporin, Nup188p. J. Cell Biol. 133, 1141–1152. Genet. 17, 40–48. Zhao, C., Brown, R.S.H., Chase, A.R., Eisele, M.R., and Schlieker, C. (2013). Pappas, S.S., Darr, K., Holley, S.M., Cepeda, C., Mabrouk, O.S., Wong, Regulation of Torsin ATPases by LAP1 and LULL1. Proc. Natl. Acad. Sci. J.-M.T., LeWitt, T.M., Paudel, R., Houlden, H., Kennedy, R.T., et al. (2015). USA 110, E1545–E1554. Forebrain deletion of the dystonia protein torsinA causes dystonic-like move- Zhu, Y., Romero, M.I., Ghosh, P., Ye, Z., Charnay, P., Rushing, E.J., Marth, ments and loss of striatal cholinergic neurons. eLife 4, e08352. J.D., and Parada, L.F. (2001). Ablation of NF1 function in neurons induces Schneiter, R., Hitomi, M., Ivessa, A.S., Fasch, E.V., Kohlwein, S.D., and Tartak- abnormal development of cerebral cortex and reactive gliosis in the brain. off, A.M. (1996). A yeast acetyl coenzyme A carboxylase mutant links very- Genes Dev. 15, 859–876.

Cell Reports 16, 3322–3333, September 20, 2016 3333