Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Feb 1.
Published in final edited form as: Biol Psychiatry. 2016 Feb 13;81(3):220–230. doi: 10.1016/j.biopsych.2016.02.008

Sumoylation of FOXP2 regulates motor function and vocal communication through Purkinje cell development

Noriyoshi Usui 1, Marissa Co 1, Matthew Harper 1, Michael A Rieger 2, Joseph D Dougherty 2, Genevieve Konopka 1
PMCID: PMC4983264  NIHMSID: NIHMS760041  PMID: 27009683

Abstract

Background

Mutations in the gene encoding the transcription factor forkhead box P2, FOXP2, result in brain developmental abnormalities including reduced gray matter in both human patients and rodent models, and speech and language deficits. However, neither the region-specific function of FOXP2 in the brain, in particular the cerebellum, nor the effects of any post-translational modifications of FOXP2 in the brain and disorders have been explored.

Methods

We characterized sumoylation of FOXP2 biochemically, and analyzed the region-specific function and sumoylation of FOXP2 in the developing mouse cerebellum. Using in utero electroporation to manipulate the sumoylation-state of Foxp2 as well as Foxp2 expression levels in Purkinje cells (PCs) of the cerebellum in vivo, we reduced Foxp2 expression approximately 40% in the mouse cerebellum. Such a reduction approximates the haploinsufficiency observed in human patients who demonstrate speech and language impairments.

Results

We identified sumoylation of FOXP2 at K674 (K673 in mouse) in the cerebellum of neonates. In vitro co-immunoprecipitation and in vivo colocalization experiments suggest that PIAS3 acts as the SUMO E3 ligase for FOXP2 sumoylation. This sumoylation modifies transcriptional regulation by FOXP2. We demonstrate that Foxp2 sumoylation is required for regulation of cerebellar motor function and vocal communication, likely through dendritic outgrowth and arborization of PCs in the mouse cerebellum.

Conclusions

Sumoylation of Foxp2 in neonatal mouse cerebellum regulates PC development as well as motor functions and vocal communication, demonstrating evidence for sumoylation in regulating mammalian behaviors.

Keywords: FOXP2, Purkinje cells, cerebellum, sumoylation, vocal communication, motor function

Introduction

The transcription factor FOXP2 has been implicated in human brain evolution, language, cognition, vocal-motor integration, and neural development in the central nervous system (CNS) through orchestration of transcriptional cascades that also tend to be at risk in several neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia (13). Previous work using humanized Foxp2 mouse models has suggested that humanized Foxp2 alters cortico-striatal function (46), but the cerebellum appears to be a key brain region for FOXP2 function as patients with mutations in FOXP2 demonstrate significant grey matter reduction in the cerebellum as evidenced by MRI (7), and genetic disruption of Foxp2 in mice results in decreased cerebellar size (811). Recent studies have uncovered important roles for the cerebellum in higher cognitive functions such as language, cognition, emotion, and memory (1219). In particular, function of PCs in the mouse cerebellum is critical for ASD-relevant behaviors (20). However, the cerebellar-specific function of FOXP2 has not been explored.

Sumoylation, a highly conserved post-translational modification, regulates protein function in numerous ways including subcellular localization, stability, and transcriptional activity (21, 22). In the CNS, sumoylation regulates transcription, ion channel activity, synapse formation and regulation, mRNA transport in axons, and mitochondrial function (2224). During sumoylation, the SUMO proteins are conjugated to lysine residues of the target proteins by SUMO enzymes (E1 activating, E2 conjugating, and E3 ligase enzymes), and are subsequently removed by SUMO-specific proteases, SENPs (25). Disruption of sumoylation can affect pathology in brain disorders such as Huntington’s disease (Htt), spinal and bulbar muscular atrophy (SUMO-1 positive intranuclear inclusions), spinocerebellar ataxias (Ataxin-1, 3, 7), Alzheimer’s disease (APP, Tau), Parkinson’s disease (α-Synuclein, Parkin, DJ-1) and ischemia (increase of SUMO-2/3, Drp1) (22, 24). A recent report has shown that FOXP2 is a substrate for sumoylation in transformed cell lines (26), however the role of sumoylation and potentially other post-translational modifications of FOXP2 in the CNS is completely unknown.

In this study, we identified sumoylation of FOXP2 in the cerebellum of neonates, a critical time for neural circuit formation and the emergence of vocal communication in mammals. Therefore, we explored the role of FOXP2 sumoylation in neuronal development and mammalian behavior related to the cerebellum. Here, we provide in vivo evidence demonstrating the requirement for sumoylation and cerebellar-specific expression of FOXP2 in directing complex motor behaviors and vocal communication. We found sumoylation of FOXP2 regulates dendritic outgrowth and arborization in PCs of the cerebellum, resulting in altered mammalian behavior and transcriptional regulation of FOXP2 respectively. These data demonstrate a critical role for FOXP2 in the cerebellum to regulate PC development, motor function and vocal communication that might be relevant to neurodevelopmental disorders.

Methods and Materials

Detailed Methods and Materials are described in the Supplemental Methods and Materials.

Animal experiments

Wild type C57BL/6J mice were used for all in vivo experiments. For in utero electroporation (IUE), plasmid DNA (1–2 μg/μl) was microinjected into the 4th ventricles of E12.5 embryos to target PCs. The embryo was electroporated (five 50-millisecond pulses of 33 V with an interval of 950 milliseconds; CUY21SC, NEPA GENE, Ichikawa-City, Chiba, Japan) using platinum plate electrode tweezers (CUY650P5; Protech International Inc., Boerne, TX) (2730). All procedures were approved by the Institutional Animal Care and Use Committee of UT Southwestern Medical Center.

Biochemical experiments

293T cells were transfected using FuGENE6 (#E2691, Promega, Madison, WI) and harvested 48 hrs later. 50 mM N-ethylmaleimide (NEM) (#E3876, Sigma-Aldrich, St. Louis, MO) was used as a SUMO protease inhibitor. 1 mM hydrogen peroxide (H2O2) (31) and 100 μM ginkgolic acid (#75741, Sigma-Aldrich, St. Louis, MO) (26, 32) were used as sumoylation inhibitors.

Results

FOXP2 is sumoylated in the neonatal cerebellum

In the course of examining Foxp2 expression in the developing cerebellum, we observed that while unmodified Foxp2 peaks in expression embryonically, a higher molecular weight band corresponding to Foxp2 peaks in expression in neonatal mouse cerebellum (Figure 1A, B). When we expressed wild type FOXP2 (FOXP2 WT) in 293T cells, we also observed a higher molecular weight band recognized by a FOXP2 antibody that is decreased by H2O2 treatment, a mechanism for reversible inhibition of SUMO conjugating enzymes (31) (Figure 1C) and ginkgolic acid (26, 32), a sumoylation-specific inhibitor (Figure 1D). Furthermore, in both 293T cells and mouse cerebellum, this high molecular weight band is recognized by either FOXP2 or SUMO-1 antibody in lysates that have undergone immunoprecipitation with an antibody recognizing SUMO-1 or FLAG (to capture FLAG-tagged FOXP2) respectively (Figure 1B–D). Based on these observations, we identified a conserved consensus sumoylation site (ψKXE) at K674 (K673 in mouse) that is outside of the annotated functional domains of FOXP2 (26) (Figure 1E, F). Upon mutation of this lysine to arginine (the non-sumoylated form of FOXP2K674R, FOXP2 KR), the high molecular weight band was not observed (Figure 1G), however this point mutation does not affect the protein expression of FOXP2 in 293T cells, nor the amount immunoprecipitated by an antibody (Figure S1 in Supplement 1). Together, these data demonstrate that the high molecular weight modification of Foxp2 in mouse cerebellum is a sumoylated form of Foxp2.

Figure 1. Sumoylation of FOXP2 in the cerebellum.

Figure 1

(A) Endogenous Foxp2 immunoblot of mouse cerebellum using an anti-Foxp2 antibody over time demonstrates an increase in sumoylated Foxp2 protein in early postnatal life. Right panel is quantification of left panel (Sumo-Foxp2 protein). Immunoblot results were normalized to Gapdh and Foxp2 at each time point and then subsequently normalized to non-sumoylated Foxp2 levels at P0. Data are represented as means (±sem), n=4/condition. (B) Endogenous co-immunoprecipitation of Foxp2 and Sumo-1 in the mouse cerebellum at P0, P10 and P21 using an anti-Sumo-1 antibody. Right panel is quantification of left panels (Sumo-Foxp2 protein). Immunoblot results were normalized to Foxp2 at each time point and then subsequently normalized to non-sumoylated Foxp2 levels at P0. Data are represented as means (±sem), n=3/condition. (C, D) A high molecular weight band is observed blotting for SUMO-1 and FOXP2 in 293T cells. 293T cells were transfected with FLAG-tagged FOXP2 WT or KR construct, and then immunoprecipitation was performed with an anti-FLAG antibody. The intensity of this band is reduced by 1 mM hydrogen peroxide (H2O2) for 1 hour (C) or 100 μM ginkgolic acid for 6 hours (D), and increased by N-ethylmaleimide (NEM). (E) Schematic of FOXP2 protein showing K674 location. PolyQ: polyglutamine motif, ZF: zinc finger, LZ: leucine zipper. (F) K674 is conserved across species. (G) Immunoblot of immunoprecipitated FOXP2 showing that mutation of K674 to an arginine results in disappearance of a sumoylated high molecular weight band recognized by an anti-SUMO-1 antibody. 293T cells were transfected with FLAG-tagged FOXP2 WT or KR construct except the 293T control, and then immunoprecipitation was performed with an anti-FLAG antibody. FOXP2 WT: wild type FOXP2, FOXP2 KR: non-sumoylated form of FOXP2K674. Asterisk indicates non-specific band of SUMO-1 antibody.

In support of Foxp2 sumoylation in the cerebellum, a previous yeast two-hybrid screen demonstrated interaction of FOXP2 with PIAS3 (33), an E3 ligase that attaches SUMO proteins to their substrates (34). Hence, we investigated the physiological interaction of FOXP2 with SUMO-1, SUMO-2/3, or PIAS3 in 293T cells (Figure 2A–C). SUMO-2 and SUMO-3 are 97% homologous and therefore are indistinguishable (35, 36). We also examined mRNA expression of Sumo proteins; Sumo-1, Sumo-2, Sumo-3 and Sumo-4 (Figure 2D) and Pias family proteins; Pias1, Pias2, Pias3 and Pias4 (Figure 2E) and observed mostly unchanged expression throughout development of the neonatal cerebellum. We also examined expression of these sumoylation proteins in the mouse cerebellum, and observed co-localization of Foxp2, Sumo-1, Sumo-2/3 and Pias3 in PCs of mouse cerebellum at postnatal day 7 (P7) when Foxp2 is highly sumoylated (Figure 2F, Figure S2 in Supplement 1). Together, these data suggest FOXP2 is sumoylated by SUMO-1/2 and PIAS family proteins, most likely PIAS3. In particular, FOXP2 sumoylation is increased during the time points corresponding to neuronal differentiation in the cerebellum (Figure 1A, B), suggesting sumoylation of FOXP2 plays a role in neuronal development.

Figure 2. Sumoylation of FOXP2 by SUMO-1/2 and PIAS3.

Figure 2

(A, B) Co-immunoprecipitations of FOXP2 WT, KR and SUMO-1, SUMO-2 proteins in 293T cells. 293T cells were transfected with FLAG-tagged FOXP2 WT or KR construct together with GFP-tagged SUMO-1 (A) or SUMO-2 (B) construct, and then immunoprecipitation was performed by an anti-FLAG antibody. FOXP2 is sumoylated by both SUMO-1 (A) and SUMO-2 (B), however these interactions are not observed with FOXP2 KR. (C) Co-immunoprecipitations of FLAG-tagged FOXP2 and V5-tagged PIAS3 in 293T cells. 293T cells were transfected with FLAG-tagged FOXP2 WT or KR construct together with V5-tagged PIAS3, and then immunoprecipitation was performed by an anti-FLAG antibody. FOXP2 physically interacts with SUMO E3 ligase PIAS3. (D, E) qRT-PCR of key sumoylation genes during cerebellar development. Expression of Sumo-1, Sumo-2 and Sumo-3 mRNAs (D), Pias1, Pias2, Pias3 and Pias4 mRNAs (E) remain relatively stable across postnatal mouse cerebellar development. qRT-PCR results were normalized to expression of each gene at P0. Data are represented as means (±sem), n=3/condition. FOXP2 WT: wild type FOXP2, FOXP2 KR: non-sumoylated form of FOXP2K674. (F) Fluorescent images of in sagittal sections of mouse cerebellum at P7. Co-expression of Foxp2, Sumo-1, Sumo-2/3 and Pias3 are observed in Purkinje cells (PCs) of mouse cerebellum (see also Figure S2 in Supplement 1). Insets show a higher magnification of the boxed area depicted in each fluorescent image. CST: Pias3 antibody from Cell Signaling Technology, Pias3 SC: Pias3 antibody from Santa Cruz Biotechnology. Scale bars: 50 μm.

Sumoylation of FOXP2 promotes neuronal differentiation through regulation of dendritic growth

To investigate whether sumoylation of FOXP2 affects its regulation of neuronal function, we assessed whether neurite outgrowth is dependent upon FOXP2 sumoylation in general in the brain using a system of mouse neural progenitors (mNPs) as previous work has demonstrated a role for FOXP2 in promoting dendrite formation (3739). We forced expression of FOXP2 in mNPs, and found that FOXP2 WT significantly promoted the length of neurites expressing either an immature neuronal maker Tuj1 or a mature neuronal marker MAP2 (Figure S3 in Supplement 1). In contrast, FOXP2 KR was unable to promote the length of Tuj1-positive and MAP2-positive neurites as effectively (Figure S3 in Supplement 1). These data indicate that sumoylation of FOXP2 plays a role in promoting neuronal maturation potentially in any neuron expressing FOXP2.

Next, we assessed whether sumoylation of FOXP2 affects neuronal maturation in vivo. Extensive characterization of Foxp2 expression in the cerebellum has shown that Foxp2 expression is limited to PCs (Figures S2 and S4A in Supplement 1) (4042). PCs send projections to the deep cerebellar nuclei and vestibular nuclei, the sole motor output of the cerebellum, indicating this neuronal pathway plays an important role in known cerebellar functions such as motor coordination and speech. The dendritic arbors of PCs are severely diminished in Foxp2 mutant mice (8, 10), suggesting a role for Foxp2 in PC development.

To directly test the cell autonomous role of Foxp2 expression in PCs in the absence of alteration of Foxp2 in other cell types or brain regions, we, carried out directed in utero electroporation (IUE) experiments. We knocked down Foxp2 expression specifically in PCs of the mouse cerebellum by Foxp2 shRNA, and concurrently rescued Foxp2 expression with either a wild type FOXP2 (WT rescue) or non-sumoylated FOXP2 (KR rescue) shRNA-resistant construct (Figure 3A–D). As a control, we used a non-silencing shRNA (Control shRNA). All shRNA sequences are in the microRNA context (43). To provide IUE-specificity in targeting only PCs, we performed IUE at embryonic day 12.5 (E12.5), when PCs arise from the ventricular zone in the mouse cerebellum (Figure 3C). The absence of Foxp2 expression in other cerebellar cell types such as the deep cerebellar nuclei at this time point (42) further supports our targeting specificity. Moreover, the uniform expression of Foxp2 in PCs throughout the entire cerebellum at this developmental time point (42) permits targeting of all cerebellar subdivisions. We confirmed IUE specificity at P7 by GFP immunostaining when FOXP2 is highly sumoylated in vivo, and observed GFP expression limited to only PCs and the dendrites and output fibers of the PCs, which project to the deep cerebellar nuclei and vestibular nucleus (Figure 3E, Figure S4 in Supplement 1). In line with this, approximately 30.7±2.6% (19–48%) of PCs were transfected by IUE without affecting other cerebellar cell types (Figure. 3E, Figure S4B–E in Supplement 1) (27, 44). Using this in vivo manipulation, we reduced Foxp2 expression approximately 40.1±2.8% in vivo in the mouse cerebellum at P7 (Figure 3D). Such a reduction approximates the haploinsufficiency observed in human patients who demonstrate speech and language impairments (1, 7, 45).

Figure 3. Foxp2 sumoylation regulates Purkinje cell development in vivo.

Figure 3

(A) Representative immunoblot for Foxp2 demonstrating knockdown of mouse (m) Foxp2 protein with a specific shRNA. Co-expression with either FOXP2 WT or KR rescue constructs that are modified to resist the hairpin rescues the knockdown using 293T cells. 293T cells were transfected with control shRNA or Foxp2 shRNA construct together with empty vector for control with mFoxp2 construct, FOXP2 WT or KR rescue construct. (B) Quantification of knockdown and rescue of Foxp2/FOXP2 protein in 293T cells. Data are represented as means (±sem). Asterisks indicate P<0.01, one-way ANOVA with a Tukey’s multiple comparison test (P<0.0018), n=3/condition. (C) Schematic and time line of in utero electroporation (IUE) of Foxp2 manipulation into the mouse cerebellum followed by neonatal morphological assessment of PCs. (D) Immunoblot and quantification of Foxp2 protein knockdown in electroporated mouse cerebellum at P7. Approximately 40.1±2.8% knockdown in cerebellum by IUE is observed. Data are represented as means (±sem), Asterisks indicate P=0.0007, t-test, n=3/condition. (E) An example of IUE specificity is confirmed by GFP expression limited to only PCs in sagittal section of electroporated mouse cerebellum at P7. Calb1 is a marker for PCs. (F) Fluorescent images of PCs in sagittal sections of mouse cerebellum at P7 for each condition. (G, H) Quantification of dendritic length (G) and branching (H) in PCs for each condition. Decreased dendritic outgrowth and arborization of PCs are observed after Foxp2 knockdown, and FOXP2 WT and KR forms rescue dendritic outgrowth, but KR is unable to fully rescue the arborization. Data are represented as means (±sem). Asterisks indicate ***P<0.001, **P<0.01, *P<0.05, one-way ANOVA with a Tukey’s multiple comparison test (P<0.0001 for dendritic length, P<0.0001 for dendritic branching), n=70–155 cells for dendritic length, 50–77 cells for branching/condition from 3–4 animals. (I) Fluorescent images of mouse cortex at P7, which are electroporated into cerebellum. No differences in Foxp2 expression by IUE of Foxp2 knockdown are observed in both cortex and striatum. Ctx: cortex, Str: striatum. Scale bars: 500 μm in E, 20 μm in F, 500 μm in I.

Using IUE to manipulate Foxp2 expression and its sumoylation, we observed a reduction in dendritic outgrowth and arborization of PCs at P7 in pups receiving Foxp2 shRNA (Figure 3F–H), consistent with published reports of Foxp2 genetically modified mice (8). This reduction of dendritic outgrowth and arborization in PCs was restored by WT rescue but not by KR rescue (Figure 3F–H). These data indicate that sumoylation of FOXP2 plays a role in promoting neuronal differentiation through neurite/dendritic outgrowth and arborization without affecting Foxp2 expression in cortex and striatum (Figure 3F–I) and PCs viability (Figure S5 in Supplement 1).

Sumoylation of FOXP2 regulates cerebellar motor functions

Since PC development appears to depend upon FOXP2 expression and sumoylation, we assessed whether Foxp2 sumoylation impacts cerebellar motor function. Previous studies have demonstrated a deficit in cerebellar-based motor behaviors such as righting reflex and negative geotaxis in Foxp2 KO mice during neonatal stages (8). We found that reduction of Foxp2 specifically in the cerebellum significantly alters neonatal righting reflex and negative geotaxis at P4 and P7 (Figure 4A–C). These phenotypes were not due to a global developmental delay as has been observed in Foxp2 KO mice (8, 10) as weights were not significantly different except for a slight decrease in weight with KR rescue at P7 that was not correlated with behavior (Figure 4D and Table S1 in Supplement 1). We also did not observe any sex differences in these behaviors (Table S2 in Supplement 1). Strikingly, both motor phenotypes were rescued by WT complementation, but not by KR complementation (Figure 4A–C). These data support a role for FOXP2 sumoylation in motor functions.

Figure 4. Foxp2 sumoylation regulates motor functions.

Figure 4

(A–C) Knockdown of endogenous Foxp2 results in deficiencies of righting reflex (A) and negative geotaxis at 30 (B) and 45 degrees (C) at P4 and P7. These Foxp2 knockdown phenotypes can be rescued with FOXP2 WT, but not KR. The 45 degree angle of our system is difficult for P4 pups, but not for P7 pups. Asterisks indicate ***P<0.001, **P<0.01, *P<0.05, two-way ANOVA with a Tukey’s multiple comparison test (righting reflex: interaction, P=0.69; age, P=0.89, genotype, P<0.0001; negative geotaxis at 30 degrees: interaction, P=0.0011; age, P<0.0001, genotype, P<0.0001; negative geotaxis at 45 degrees: interaction, P=0.0089; age, P<0.0001, genotype, P<0.0001; n=9–15/condition). (D) Mouse weight was measured at P4 and P7 after behavioral testing. There is no statistical difference among conditions at both stages. Data are represented as means (±sem). Asterisk indicates P<0.05 two-way ANOVA with a Tukey’s multiple comparison test (interaction, P=0.71; age, P<0.0001, genotype, P=0.023), n=9–15/condition.

Sumoylation regulates subcellular localization of Foxp2

We next explored the molecular mechanism underlying sumoylation of FOXP2 in neuronal development. As sumoylation often affects protein-protein interactions (21), we determined whether FOXP2K674 affects interactions with known binding partners. FOXP2 homodimerizes and also heterodimerizes with other FOXP family members, FOXP1 and FOXP4 (46). We found that FOXP2 KR does not affect homo- or heterodimerization (Figure S6A–C in Supplement 1). Foxp2 has also been shown to interact with a co-repressor, c-terminal binding protein, CTBP (46). However, this interaction was also not changed with FOXP2 KR (Figure S6D in Supplement 1).

As sumoylation can alter the subcellular localization of transcription factors (21), we next assessed the effects of sumoylation on FOXP2 localization. By inhibiting sumoylation with H2O2 treatment, we observed an increased cytoplasmic localization of FOXP2 and its co-repressor CTBP (Figure S7A in Supplement 1). Furthermore, sumoylation of CTBP has been reported to profoundly affect its subcellular localization and increase co-repressor activity (47). We therefore measured whether FOXP2 KR mutation also affects subcellular localization, and found a significant 100% increase in cytoplasmic and a significant 20% decrease in nuclear localization of FOXP2 KR compared with WT in 293T cells in vitro (Figure S7B–D in Supplement 1). We further investigated this shift in subcellular localization in vivo in PCs and cortical layer 6 neurons of mouse brain, a region where Foxp2 is also highly expressed. Consistent with our in vitro data, we observed increased cytoplasmic and decreased nuclear localization of FOXP2 KR in vivo (Figure S8 in Supplement 1). These data indicate that sumoylation modulates the subcellular localization of FOXP2 and CTBP (Figure S7 in Supplement 1), and suggest that the transcriptional function of FOXP2 is regulated in a sumoylation-dependent manner.

Sumoylation of FOXP2 regulates vocal communication

To directly determine the potential impact of sumoylation on Foxp2 transcriptional function, we first investigated whether FOXP2K674 is required for DNA transrepression through luciferase assays, and found that both FOXP2 WT and KR can repress a luciferase reporter equally well when presented with a canonical FOXP2 motif (AATTTG) in triplicate (48) (Figure S9 in Supplement 1). As the luciferase experiments do not utilize the endogenous chromatin state, we examined the in vivo regulation of a well-characterized target of FOXP2, contactin associated protein-like 2, Cntnap2. CNTNAP2 polymorphisms are associated with specific language impairment (45, 49, 50), CNTNAP2 promoter variants have been identified as potential ASD risk factors (50, 51). and rodent models lacking Cntnap2 exhibit abnormal neuronal migration as well as ASD-relevant behaviors including altered ultrasonic vocalizations (USVs) (52). We observed relatively low expression of Cntnap2 when sumoylated Foxp2 is highest in the developing cerebellum (Figures 1A, B and 5A). We therefore directly assessed the requirement of FOXP2 sumoylation on transcriptional regulation of CNTNAP2 expression using human neural progenitors (hNPs). As previously reported (45), we confirmed repression of CNTNAP2 by FOXP2 WT (Figure 5B). In contrast, FOXP2 KR was unable to significantly repress CNTNAP2 (Figure 5B). These data demonstrate that transcriptional regulation of at least one ASD-associated gene, CNTNAP2, is dependent on sumoylation of FOXP2.

Figure 5. Foxp2 sumoylation regulates vocal communication.

Figure 5

(A) qRT-PCR of Cntnap2 during mouse cerebellar development. Cntnap2 mRNA expression is decreased at the time point when sumoylation of Foxp2 is strongly observed in developing mouse cerebellum at P7. qRT-PCR results were normalized to Cntnap2 expression at P0 (Figure 1A). Data are represented as means (±sem), n=3/condition. (B) Quantification of qRT-PCR in human neural progenitors (hNPs) expressing either FOXP2 WT or FOXP2 KR. FOXP2 WT but not KR can repress expression of CNTNAP2. Data are represented as means (±sem). Asterisks indicate ***P<0.001, *P<0.05, one-way ANOVA with a Tukey’s multiple comparison test (P<0.0001 for FOXP2, P<0.0001 for CNTNAP2), n=12/condition. (C–H) USVs were analyzed in detail. Foxp2 knockdown results in a decrease in USVs at P4 and P7. This USV deficiency can be rescued with FOXP2 WT construct, but not KR construct. (C), Total number of whistle calls (USVs: interaction, P=0.70; age, P=0.0001, genotype, P<0.0001); (D), percentage of calls with frequency jumps (interaction, P=0.63; age, P=0.61, genotype, P=0.048); (E), call duration (interaction, P=0.11; age, P=0.12, genotype, P=0.08); (F), mean frequency (interaction, P=0.86; age, P<0.0001, genotype, P=0.89); (G), frequency range (interaction, P=0.74; age, P=0.37, genotype, P=0.14); and (H), mean call slope (interaction, P=0.44; age, P=0.14, genotype, P=0.98). Data are represented as means (±sem). Asterisks indicate ***P<0.001, **P<0.01, *P<0.05, two-way ANOVA with a Tukey’s multiple comparison test, n=9–15/condition.

This altered regulation of CNTNAP2 was striking as both FOXP2 and CNTNAP2 have been implicated in vocal motor behaviors in both human patients as well as mouse models (1, 7, 45, 49, 52). Foxp2 KO mice have few, if any, neonatal USVs (8). It is unknown whether reduction of Foxp2 specifically in the cerebellum contributes to the diminished USVs in this mouse model; however, recent work has shown that mice containing a patient-relevant Foxp2 mutation exhibit decreased number of USVs (10) that can be rescued with forced cerebellar expression of FOXP2 (53). Together, these data support a role for region-specific FOXP2 function in the cerebellum related to vocal behaviors. Therefore, we examined USVs at the same developmental stages at which we examined righting reflex and negative geotaxis. A significant decrease in the number of USVs was observed in animals receiving cerebellar-directed Foxp2 shRNA at P4 and P7 (Figure 5C and Tables S1 and S2 in Supplement 1). This result suggests that normal Foxp2 expression in the cerebellum is required for neonatal USVs. We observed no difference with Foxp2 shRNA in the number of calls with frequency jumps, call duration, mean frequency or frequency range of USVs (Figure 5D–H), suggesting the calls that were present were relatively normal. As we observed with other motor behaviors, the decrease in number of USVs was rescued by the WT construct at P4 and P7, but not by the KR construct (Figure 5C). These data demonstrate sumoylation of Foxp2 in the cerebellum is also required for vocal communication in mouse.

Discussion

In this study, we identify in vivo sumoylation of FOXP2 at K674 (K673 in mouse) in mouse cerebellum during neonatal stages. We demonstrate that sumoylation of FOXP2 plays a critical role in transcription regulation, neuronal development, motor functions and USVs, specifically in the cerebellum. The timing of FOXP2 sumoylation in the cerebellum occurs during a critical period in the formation of the cerebellar neuronal network when dendritic arborization, synaptogenesis, and clustering of potassium channels take place (44, 54). This network is essential for normal cerebellar functions that are at risk in neurodevelopmental disorders such as ASD (15, 16, 18, 19).

To elucidate molecular mechanisms of FOXP2 sumoylation, we demonstrate FOXP2 physically interacts with SUMO-1, SUMO-2 and PIAS3 in 293T cells. To demonstrate in vivo evidence that Pias3 sumoylates Foxp2, we performed endogenous co-immunoprecipitation using mouse cerebellum at P10 but we were unable to detect such an interaction (data not shown). In order to investigate the possibility that Foxp2 is sumoylated by other Pias family proteins, we also examined Pias1, Pias2, and Pias4, but again could not detect an interaction (data not shown). Therefore, we hypothesize that the interaction of Foxp2 and the Pias family proteins is a dynamic interaction during the catalytic reaction of an enzyme and a substrate, and therefore it would be challenging to detect endogenous interaction of Foxp2 and Pias family proteins by co-immunoprecipitation. By immunostaining, we observed in vivo co-localization of Foxp2 and Pias3 in PCs of mouse cerebellum at P7 (Figure 2F and Figure S2 in Supplement 1), suggesting an opportunity for Foxp2 to be sumoylated by Pias3. However, we cannot rule out possible sumoylation of Foxp2 by other factors in vivo. Future studies using knockdown or knockout of Pias3 in vivo should determine the requirement of Pias3 in Foxp2 sumoylation. Moreover, loss of the lysine residue of FOXP2 at K647 could have additional detrimental effects in addition to loss of sumoylation, such as conformational changes of FOXP2 leading to altered protein-protein interaction or DNA binding, alterations of other post-transcriptional modifications like ubiquitination, or protein stability. We investigated a number of these possibilities, but there were no differences between FOXP2 WT and KR in our experiments (Figures S6 and S9 in Supplement 1, data not shown), consistent with a recent study showing that FOXP2 sumoylation does not affect protein stability (26).

This is the first examination of the role of FOXP2 post-translational modification in brain development and function. We observe Foxp2 sumoylation occurs specifically during neonatal stages of mouse cerebellar development. We demonstrate FOXP2 sumoylation plays a role in promoting neuronal development through neurite/dendritic outgrowth in both mNPs in vitro and PCs in vivo. In support of our results, previous studies have shown that FOXP2 transcriptional targets are enriched for genes involved in neuronal differentiation and axon guidance (37, 39, 48). Our data suggest that sumoylation of FOXP2 is spatiotemporally regulated for promoting maturation of neural networks. We further investigated the molecular mechanisms whereby FOXP2 sumoylation may affect transcriptional regulation. We found that sumoylation of FOXP2 does not affect DNA transrepression of a canonical FOXP2 motif. However, we did find that a FOXP2 target gene, CNTNAP2, was derepressed by FOXP2 KR. In addition, a recent study has shown that FOXP2 sumoylation modulates transcriptional activity of FOXP2 in regulating target genes such as DISC1, SRPX2 and MIR200C (26). In support of these results, sumoylation of L3MBTL2, a protein implicated in transcriptional repression and chromatin compaction, affects only a subset of its target genes, due to derepression by a non-sumoylated form of L3MBTL2, but does not affect chromatin binding as evidenced by ChIP-seq (55). Finally, we found sumoylation of FOXP2 altered the subcellular localization of FOXP2, suggesting that non-nuclear FOXP2 is unable to act as a transcriptional repressor. In contrast, another FOXP2 sumoylation study has reported non-quantitative evidence that sumoylation does not affect subcellular localization of FOXP2 in vitro in MCF7 cells (a breast cancer cell line) (26). It is possible that different phenotypes will be observed depending on cell type. In addition, we confirmed that sumoylation modulates the subcellular localization of FOXP2 using quantification of high-resolution confocal imaging both in vitro and in vivo. Together, these findings suggest sumoylation of FOXP2 provides specificity and selectivity in transcriptional regulation. However several unanswered questions remain: 1) is sumoylation of other transcription factors important for PC function? And 2) is sumoylation of Foxp2 important outside of the cerebellum? We believe this study is just the first of many future studies delineating these distinctions

The cerebellum has been typically thought to be involved in motor functions in the CNS, however recent studies have uncovered important roles for the cerebellum in higher cognitive functions such as language, cognition, emotion, and memory (1219). We demonstrate that Foxp2 expression is required for normal cerebellar development through dendritic outgrowth and arborization of PCs. Consequently, abnormal PC development in the mouse cerebellum leads to motor and vocal impairments, consistent with previous whole body Foxp2 KO and mutant mice studies (8, 10). Foxp2 KO mice also reportedly exhibit a reduction in cerebellar size (811). This is consistent with a reduction in both cerebellar size and PCs in patients with ataxia (14, 1619, 56), language disorders (7, 1619), ASD (1416, 18, 19, 57, 58) and schizophrenia (16, 5961). In this study, we could not ascertain a significant reduction in cerebellar size in our mouse models due to the transfection efficiency of IUE. However, given this inherent variability of IUE it is even more remarkable that we demonstrate consistent and robust developmental disruption to PCs, and impairments of motor functions and USVs with FOXP2 knockdown. PCs play a critical role in modulating and integrating all cerebellar inputs into a unified output: a single PC receives synaptic inputs from up to 200,000 parallel fibers (62). Therefore, developmental disruptions of even a limited number of PCs can significantly alter cerebellar function in this study. Future studies should investigate the physiological consequences of altered PC development with loss of Foxp2 function. In addition, the other region-specific roles of FOXP2 in brain areas such as cortex and striatum, where FOXP2 is also highly expressed, are still unknown and need to be investigated. However, our in vitro data using mNPs and previous in vitro studies (38, 39, 45) suggest that FOXP2 and its modification may play a role in neuronal differentiation, which could universally affect neural circuit formation in regions with FOXP2 expression.

Our data using mouse models show that sumoylation can regulate motor behaviors such as USVs. Measuring USVs is widely carried out in genetic ASD model mice (63), with phenotypes observed in models of Foxp1 (64), Cntnap2 (52), Tsc1 (20), Tsc2 (65), Nlgn3 (66), Nlgn4 (67), Shank1 (68), Shank2 (69), Shank3 (70) and Mecp2 (71). In support of our behavioral results, two recent studies have demonstrated that sumoylation in hippocampus can modify mammalian cognitive behaviors by altering hippocampal-dependent learning and memory by Ubc9, a E2 conjugating enzyme (72), and spatial memory by sumoylation of CREB (73). In addition, neuron-specific knockdown of all SUMO-1/2/3 in RNAi transgenic mice leads to anxiety-like behavior, and impairs episodic and fear memories (74). Many genes implicated in ASD encode synaptic proteins important for regulating synaptic homeostasis involved in cognition (3, 75, 76). Sumoylation of ASD genes (e.g. MEF2A (77), CASK (78), MeCP2 (79)) has been reported to play roles in regulating synaptic development and function (22, 24, 80), but direct evidence for the requirement for sumoylation in regulating ASD-relevant behaviors has not been shown. In contrast, hyper-sumoylation has been shown to cause functional abnormalities in the brain including seizures by affecting potassium channel function (81). As there is high co-morbidity of seizure disorders and ASD, these results further support a potential role for sumoylation in ASD pathophysiology.

In conclusion, our data demonstrate three novel findings: 1) sumoylation of FOXP2 regulates PC development, 2) sumoylation can direct cerebellar-specific motor behaviors, in particular vocal communication, and 3) cerebellar-specific expression of FOXP2 is required for rodent vocalizations. These findings support a critical role for FOXP2 in the cerebellum. This is compelling given the mounting evidence for cerebellar dysfunction in ASD (15, 16, 1820, 57, 58) and the identification of numerous ASD-relevant genes regulated by FOXP2 (2). Our data suggest that FOXP2 sumoylation at a single amino acid orchestrates a switch in FOXP2 function in the brain. These findings provide insight into understanding the mechanisms underlying functional diversification of FOXP2 across circuits mediating distinct behaviors in the brain. Further understanding of sumoylation in the CNS should give rise to novel insights and targets for understanding the molecular mechanisms underlying neurodevelopmental disorders.

Supplementary Material

supplement

Acknowledgments

We thank Drs. Joseph S. Takahashi, Carla B. Green, Todd Roberts and Peter Tsai for discussions and comments, Drs. Shin Yamazaki, Jay Gibson and Kris Loerwald for technical advice, Mr. Jayant Madugula for technical support, and Dr. Pier Paolo Scaglioni for SUMO-1-GFP and SUMO-2-GFP cDNA. N.U. is a Research Fellow of the Uehara Memorial Foundation. M.C. is a Predoctoral Fellow of the Autism Science Foundation. G. K. is a Jon Heighten Scholar in Autism Research at UT Southwestern. This work was supported by the HHMI Med into Grad Initiative to M.C.; an NIH training grant (T32GM007067) to M.R.; the McDonnell Center for Systems Neuroscience to J.D.; the NIMH (R00MH090238, R01DC014702, R01MH102603), a March of Dimes Basil O’Connor Starter Scholar Research Award (Grant No. 5-FY13-19), Once Upon a Time Foundation, Friends of the Alzheimer’s Disease Center at UT Southwestern, and CREW Dallas to G. K.

Footnotes

Financial Disclosures

The authors report no biomedical financial interests or potential conflicts of interest.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Lai CS, Fisher SE, Hurst JA, Vargha-Khadem F, Monaco AP. A forkhead-domain gene is mutated in a severe speech and language disorder. Nature. 2001;413:519–523. doi: 10.1038/35097076. [DOI] [PubMed] [Google Scholar]
  • 2.Lepp S, Anderson A, Konopka G. Connecting signaling pathways underlying communication to ASD vulnerability. International review of neurobiology. 2013;113:97–133. doi: 10.1016/B978-0-12-418700-9.00004-6. [DOI] [PubMed] [Google Scholar]
  • 3.Usui N, Co M, Konopka G. Decoding the molecular evolution of human cognition using comparative genomics. Brain, behavior and evolution. 2014;84:103–116. doi: 10.1159/000365182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Enard W, Gehre S, Hammerschmidt K, Holter SM, Blass T, Somel M, et al. A humanized version of Foxp2 affects cortico-basal ganglia circuits in mice. Cell. 2009;137:961–971. doi: 10.1016/j.cell.2009.03.041. [DOI] [PubMed] [Google Scholar]
  • 5.Reimers-Kipping S, Hevers W, Paabo S, Enard W. Humanized Foxp2 specifically affects cortico-basal ganglia circuits. Neuroscience. 2011;175:75–84. doi: 10.1016/j.neuroscience.2010.11.042. [DOI] [PubMed] [Google Scholar]
  • 6.Schreiweis C, Bornschein U, Burguiere E, Kerimoglu C, Schreiter S, Dannemann M, et al. Humanized Foxp2 accelerates learning by enhancing transitions from declarative to procedural performance. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:14253–14258. doi: 10.1073/pnas.1414542111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Vargha-Khadem F, Gadian DG, Copp A, Mishkin M. FOXP2 and the neuroanatomy of speech and language. Nature reviews Neuroscience. 2005;6:131–138. doi: 10.1038/nrn1605. [DOI] [PubMed] [Google Scholar]
  • 8.Shu W, Cho JY, Jiang Y, Zhang M, Weisz D, Elder GA, et al. Altered ultrasonic vocalization in mice with a disruption in the Foxp2 gene. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:9643–9648. doi: 10.1073/pnas.0503739102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.French CA, Groszer M, Preece C, Coupe AM, Rajewsky K, Fisher SE. Generation of mice with a conditional Foxp2 null allele. Genesis (New York, NY : 2000) 2007;45:440–446. doi: 10.1002/dvg.20305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Fujita E, Tanabe Y, Shiota A, Ueda M, Suwa K, Momoi MY, et al. Ultrasonic vocalization impairment of Foxp2 (R552H) knockin mice related to speech-language disorder and abnormality of Purkinje cells. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:3117–3122. doi: 10.1073/pnas.0712298105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Groszer M, Keays DA, Deacon RM, de Bono JP, Prasad-Mulcare S, Gaub S, et al. Impaired synaptic plasticity and motor learning in mice with a point mutation implicated in human speech deficits. Current biology : CB. 2008;18:354–362. doi: 10.1016/j.cub.2008.01.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wolf U, Rapoport MJ, Schweizer TA. Evaluating the affective component of the cerebellar cognitive affective syndrome. J Neuropsychiatry Clin Neurosci. 2009;21:245–253. doi: 10.1176/jnp.2009.21.3.245. [DOI] [PubMed] [Google Scholar]
  • 13.D’Angelo E, Casali S. Seeking a unified framework for cerebellar function and dysfunction: from circuit operations to cognition. Frontiers in neural circuits. 2012;6:116. doi: 10.3389/fncir.2012.00116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Reeber SL, Otis TS, Sillitoe RV. New roles for the cerebellum in health and disease. Front Syst Neurosci. 2013;7:83. doi: 10.3389/fnsys.2013.00083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wang SS, Kloth AD, Badura A. The cerebellum, sensitive periods, and autism. Neuron. 2014;83:518–532. doi: 10.1016/j.neuron.2014.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Koziol LF, Budding D, Andreasen N, D’Arrigo S, Bulgheroni S, Imamizu H, et al. Consensus paper: the cerebellum’s role in movement and cognition. Cerebellum (London, England) 2014;13:151–177. doi: 10.1007/s12311-013-0511-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Marien P, Ackermann H, Adamaszek M, Barwood CH, Beaton A, Desmond J, et al. Consensus paper: Language and the cerebellum: an ongoing enigma. Cerebellum (London, England) 2014;13:386–410. doi: 10.1007/s12311-013-0540-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hampson DR, Blatt GJ. Autism spectrum disorders and neuropathology of the cerebellum. Frontiers in neuroscience. 2015;9:420. doi: 10.3389/fnins.2015.00420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Mosconi MW, Wang Z, Schmitt LM, Tsai P, Sweeney JA. The role of cerebellar circuitry alterations in the pathophysiology of autism spectrum disorders. Frontiers in neuroscience. 2015;9:296. doi: 10.3389/fnins.2015.00296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tsai PT, Hull C, Chu Y, Greene-Colozzi E, Sadowski AR, Leech JM, et al. Autistic-like behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant mice. Nature. 2012;488:647–651. doi: 10.1038/nature11310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gill G. Post-translational modification by the small ubiquitin-related modifier SUMO has big effects on transcription factor activity. Current opinion in genetics & development. 2003;13:108–113. doi: 10.1016/s0959-437x(03)00021-2. [DOI] [PubMed] [Google Scholar]
  • 22.Henley JM, Craig TJ, Wilkinson KA. Neuronal SUMOylation: mechanisms, physiology, and roles in neuronal dysfunction. Physiological reviews. 2014;94:1249–1285. doi: 10.1152/physrev.00008.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Shalizi A, Gaudilliere B, Yuan Z, Stegmuller J, Shirogane T, Ge Q, et al. A calcium-regulated MEF2 sumoylation switch controls postsynaptic differentiation. Science. 2006;311:1012–1017. doi: 10.1126/science.1122513. [DOI] [PubMed] [Google Scholar]
  • 24.Wilkinson KA, Nakamura Y, Henley JM. Targets and consequences of protein SUMOylation in neurons. Brain research reviews. 2010;64:195–212. doi: 10.1016/j.brainresrev.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Martin S, Wilkinson KA, Nishimune A, Henley JM. Emerging extranuclear roles of protein SUMOylation in neuronal function and dysfunction. Nature reviews Neuroscience. 2007;8:948–959. doi: 10.1038/nrn2276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Meredith LJ, Wang CM, Nascimento L, Liu R, Wang L, Yang WH. The Key Regulator for Language and Speech Development, FOXP2, is a Novel Substrate for SUMOylation. J Cell Biochem. 2015;117:426–438. doi: 10.1002/jcb.25288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Nishiyama J, Hayashi Y, Nomura T, Miura E, Kakegawa W, Yuzaki M. Selective and regulated gene expression in murine Purkinje cells by in utero electroporation. The European journal of neuroscience. 2012;36:2867–2876. doi: 10.1111/j.1460-9568.2012.08203.x. [DOI] [PubMed] [Google Scholar]
  • 28.dal Maschio M, Ghezzi D, Bony G, Alabastri A, Deidda G, Brondi M, et al. High-performance and site-directed in utero electroporation by a triple-electrode probe. Nature communications. 2012;3:960. doi: 10.1038/ncomms1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Franco SJ, Gil-Sanz C, Martinez-Garay I, Espinosa A, Harkins-Perry SR, Ramos C, et al. Fate-restricted neural progenitors in the mammalian cerebral cortex. Science. 2012;337:746–749. doi: 10.1126/science.1223616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sia GM, Clem RL, Huganir RL. The human language-associated gene SRPX2 regulates synapse formation and vocalization in mice. Science. 2013;342:987–991. doi: 10.1126/science.1245079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bossis G, Melchior F. Regulation of SUMOylation by reversible oxidation of SUMO conjugating enzymes. Molecular cell. 2006;21:349–357. doi: 10.1016/j.molcel.2005.12.019. [DOI] [PubMed] [Google Scholar]
  • 32.Fukuda I, Ito A, Hirai G, Nishimura S, Kawasaki H, Saitoh H, et al. Ginkgolic acid inhibits protein SUMOylation by blocking formation of the E1-SUMO intermediate. Chemistry & biology. 2009;16:133–140. doi: 10.1016/j.chembiol.2009.01.009. [DOI] [PubMed] [Google Scholar]
  • 33.Sakai Y, Shaw CA, Dawson BC, Dugas DV, Al-Mohtaseb Z, Hill DE, et al. Protein interactome reveals converging molecular pathways among autism disorders. Science translational medicine. 2011;3:86ra49. doi: 10.1126/scitranslmed.3002166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sharrocks AD. PIAS proteins and transcriptional regulation--more than just SUMO E3 ligases? Genes & development. 2006;20:754–758. doi: 10.1101/gad.1421006. [DOI] [PubMed] [Google Scholar]
  • 35.Geiss-Friedlander R, Melchior F. Concepts in sumoylation: a decade on. Nature reviews Molecular cell biology. 2007;8:947–956. doi: 10.1038/nrm2293. [DOI] [PubMed] [Google Scholar]
  • 36.Hasegawa Y, Yoshida D, Nakamura Y, Sakakibara S. Spatiotemporal distribution of SUMOylation components during mouse brain development. The Journal of comparative neurology. 2014;522:3020–3036. doi: 10.1002/cne.23563. [DOI] [PubMed] [Google Scholar]
  • 37.Vernes SC, Oliver PL, Spiteri E, Lockstone HE, Puliyadi R, Taylor JM, et al. Foxp2 regulates gene networks implicated in neurite outgrowth in the developing brain. PLoS genetics. 2011;7:e1002145. doi: 10.1371/journal.pgen.1002145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chiu YC, Li MY, Liu YH, Ding JY, Yu JY, Wang TW. Foxp2 regulates neuronal differentiation and neuronal subtype specification. Developmental neurobiology. 2014;74:723–738. doi: 10.1002/dneu.22166. [DOI] [PubMed] [Google Scholar]
  • 39.Devanna P, Middelbeek J, Vernes SC. FOXP2 drives neuronal differentiation by interacting with retinoic acid signaling pathways. Frontiers in cellular neuroscience. 2014;8:305. doi: 10.3389/fncel.2014.00305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ferland RJ, Cherry TJ, Preware PO, Morrisey EE, Walsh CA. Characterization of Foxp2 and Foxp1 mRNA and protein in the developing and mature brain. The Journal of comparative neurology. 2003;460:266–279. doi: 10.1002/cne.10654. [DOI] [PubMed] [Google Scholar]
  • 41.Takahashi K, Liu FC, Hirokawa K, Takahashi H. Expression of Foxp2, a gene involved in speech and language, in the developing and adult striatum. J Neurosci Res. 2003;73:61–72. doi: 10.1002/jnr.10638. [DOI] [PubMed] [Google Scholar]
  • 42.Fujita H, Sugihara I. FoxP2 expression in the cerebellum and inferior olive: development of the transverse stripe-shaped expression pattern in the mouse cerebellar cortex. The Journal of comparative neurology. 2012;520:656–677. doi: 10.1002/cne.22760. [DOI] [PubMed] [Google Scholar]
  • 43.Silva JM, Li MZ, Chang K, Ge W, Golding MC, Rickles RJ, et al. Second-generation shRNA libraries covering the mouse and human genomes. Nature genetics. 2005;37:1281–1288. doi: 10.1038/ng1650. [DOI] [PubMed] [Google Scholar]
  • 44.Dastjerdi FV, Consalez GG, Hawkes R. Pattern formation during development of the embryonic cerebellum. Frontiers in neuroanatomy. 2012;6:10. doi: 10.3389/fnana.2012.00010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Vernes SC, Newbury DF, Abrahams BS, Winchester L, Nicod J, Groszer M, et al. A functional genetic link between distinct developmental language disorders. The New England journal of medicine. 2008;359:2337–2345. doi: 10.1056/NEJMoa0802828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Li S, Weidenfeld J, Morrisey EE. Transcriptional and DNA binding activity of the Foxp1/2/4 family is modulated by heterotypic and homotypic protein interactions. Molecular and cellular biology. 2004;24:809–822. doi: 10.1128/MCB.24.2.809-822.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Lin X, Sun B, Liang M, Liang YY, Gast A, Hildebrand J, et al. Opposed regulation of corepressor CtBP by SUMOylation and PDZ binding. Molecular cell. 2003;11:1389–1396. doi: 10.1016/s1097-2765(03)00175-8. [DOI] [PubMed] [Google Scholar]
  • 48.Konopka G, Bomar JM, Winden K, Coppola G, Jonsson ZO, Gao F, et al. Human-specific transcriptional regulation of CNS development genes by FOXP2. Nature. 2009;462:213–217. doi: 10.1038/nature08549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Penagarikano O, Geschwind DH. What does CNTNAP2 reveal about autism spectrum disorder? Trends in molecular medicine. 2012;18:156–163. doi: 10.1016/j.molmed.2012.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chiocchetti AG, Kopp M, Waltes R, Haslinger D, Duketis E, Jarczok TA, et al. Variants of the CNTNAP2 5′ promoter as risk factors for autism spectrum disorders: a genetic and functional approach. Molecular psychiatry. 2015;20:839–849. doi: 10.1038/mp.2014.103. [DOI] [PubMed] [Google Scholar]
  • 51.Anderson GR, Galfin T, Xu W, Aoto J, Malenka RC, Sudhof TC. Candidate autism gene screen identifies critical role for cell-adhesion molecule CASPR2 in dendritic arborization and spine development. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:18120–18125. doi: 10.1073/pnas.1216398109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Penagarikano O, Abrahams BS, Herman EI, Winden KD, Gdalyahu A, Dong H, et al. Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities, and core autism-related deficits. Cell. 2011;147:235–246. doi: 10.1016/j.cell.2011.08.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Fujita-Jimbo E, Momoi T. Specific expression of FOXP2 in cerebellum improves ultrasonic vocalization in heterozygous but not in homozygous Foxp2 (R552H) knock-in pups. Neuroscience letters. 2014;566:162–166. doi: 10.1016/j.neulet.2014.02.062. [DOI] [PubMed] [Google Scholar]
  • 54.Buttermore ED, Piochon C, Wallace ML, Philpot BD, Hansel C, Bhat MA. Pinceau organization in the cerebellum requires distinct functions of neurofascin in Purkinje and basket neurons during postnatal development. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2012;32:4724–4742. doi: 10.1523/JNEUROSCI.5602-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Stielow C, Stielow B, Finkernagel F, Scharfe M, Jarek M, Suske G. SUMOylation of the polycomb group protein L3MBTL2 facilitates repression of its target genes. Nucleic acids research. 2014;42:3044–3058. doi: 10.1093/nar/gkt1317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Klockgether T. Sporadic ataxia with adult onset: classification and diagnostic criteria. Lancet Neurol. 2010;9:94–104. doi: 10.1016/S1474-4422(09)70305-9. [DOI] [PubMed] [Google Scholar]
  • 57.Becker EB, Stoodley CJ. Autism spectrum disorder and the cerebellum. International review of neurobiology. 2013;113:1–34. doi: 10.1016/B978-0-12-418700-9.00001-0. [DOI] [PubMed] [Google Scholar]
  • 58.Palmen SJ, van Engeland H, Hof PR, Schmitz C. Neuropathological findings in autism. Brain. 2004;127:2572–2583. doi: 10.1093/brain/awh287. [DOI] [PubMed] [Google Scholar]
  • 59.Reyes MG, Gordon A. Cerebellar vermis in schizophrenia. Lancet. 1981;2:700–701. doi: 10.1016/s0140-6736(81)91039-4. [DOI] [PubMed] [Google Scholar]
  • 60.Tran KD, Smutzer GS, Doty RL, Arnold SE. Reduced Purkinje cell size in the cerebellar vermis of elderly patients with schizophrenia. Am J Psychiatry. 1998;155:1288–1290. doi: 10.1176/ajp.155.9.1288. [DOI] [PubMed] [Google Scholar]
  • 61.Ichimiya T, Okubo Y, Suhara T, Sudo Y. Reduced volume of the cerebellar vermis in neuroleptic-naive schizophrenia. Biol Psychiatry. 2001;49:20–27. doi: 10.1016/s0006-3223(00)01081-7. [DOI] [PubMed] [Google Scholar]
  • 62.Ramnani N. The primate cortico-cerebellar system: anatomy and function. Nature reviews Neuroscience. 2006;7:511–522. doi: 10.1038/nrn1953. [DOI] [PubMed] [Google Scholar]
  • 63.Scattoni ML, Crawley J, Ricceri L. Ultrasonic vocalizations: a tool for behavioural phenotyping of mouse models of neurodevelopmental disorders. Neuroscience and biobehavioral reviews. 2009;33:508–515. doi: 10.1016/j.neubiorev.2008.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Araujo DJ, Anderson AG, Berto S, Runnels W, Harper M, Ammanuel S, et al. FoxP1 orchestration of ASD-relevant signaling pathways in the striatum. Genes & development. 2015;29:2081–2096. doi: 10.1101/gad.267989.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Young DM, Schenk AK, Yang SB, Jan YN, Jan LY. Altered ultrasonic vocalizations in a tuberous sclerosis mouse model of autism. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:11074–11079. doi: 10.1073/pnas.1005620107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Radyushkin K, Hammerschmidt K, Boretius S, Varoqueaux F, El-Kordi A, Ronnenberg A, et al. Neuroligin-3-deficient mice: model of a monogenic heritable form of autism with an olfactory deficit. Genes, brain, and behavior. 2009;8:416–425. doi: 10.1111/j.1601-183X.2009.00487.x. [DOI] [PubMed] [Google Scholar]
  • 67.Jamain S, Radyushkin K, Hammerschmidt K, Granon S, Boretius S, Varoqueaux F, et al. Reduced social interaction and ultrasonic communication in a mouse model of monogenic heritable autism. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:1710–1715. doi: 10.1073/pnas.0711555105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wohr M, Roullet FI, Hung AY, Sheng M, Crawley JN. Communication impairments in mice lacking Shank1: reduced levels of ultrasonic vocalizations and scent marking behavior. PloS one. 2011;6:e20631. doi: 10.1371/journal.pone.0020631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Won H, Lee HR, Gee HY, Mah W, Kim JI, Lee J, et al. Autistic-like social behaviour in Shank2-mutant mice improved by restoring NMDA receptor function. Nature. 2012;486:261–265. doi: 10.1038/nature11208. [DOI] [PubMed] [Google Scholar]
  • 70.Yang M, Bozdagi O, Scattoni ML, Wohr M, Roullet FI, Katz AM, et al. Reduced excitatory neurotransmission and mild autism-relevant phenotypes in adolescent Shank3 null mutant mice. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2012;32:6525–6541. doi: 10.1523/JNEUROSCI.6107-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Picker JD, Yang R, Ricceri L, Berger-Sweeney J. An altered neonatal behavioral phenotype in Mecp2 mutant mice. Neuroreport. 2006;17:541–544. doi: 10.1097/01.wnr.0000208995.38695.2f. [DOI] [PubMed] [Google Scholar]
  • 72.Lee L, Dale E, Staniszewski A, Zhang H, Saeed F, Sakurai M, et al. Regulation of synaptic plasticity and cognition by SUMO in normal physiology and Alzheimer’s disease. Sci Rep. 2014;4:7190. doi: 10.1038/srep07190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Chen YC, Hsu WL, Ma YL, Tai DJ, Lee EH. CREB SUMOylation by the E3 ligase PIAS1 enhances spatial memory. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2014;34:9574–9589. doi: 10.1523/JNEUROSCI.4302-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wang L, Rodriguiz RM, Wetsel WC, Sheng H, Zhao S, Liu X, et al. Neuron-specific Sumo1-3 knockdown in mice impairs episodic and fear memories. J Psychiatry Neurosci. 2014;39:259–266. doi: 10.1503/jpn.130148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Toro R, Konyukh M, Delorme R, Leblond C, Chaste P, Fauchereau F, et al. Key role for gene dosage and synaptic homeostasis in autism spectrum disorders. Trends Genet. 2010;26:363–372. doi: 10.1016/j.tig.2010.05.007. [DOI] [PubMed] [Google Scholar]
  • 76.Delorme R, Ey E, Toro R, Leboyer M, Gillberg C, Bourgeron T. Progress toward treatments for synaptic defects in autism. Nat Med. 2013;19:685–694. doi: 10.1038/nm.3193. [DOI] [PubMed] [Google Scholar]
  • 77.Flavell SW, Cowan CW, Kim TK, Greer PL, Lin Y, Paradis S, et al. Activity-dependent regulation of MEF2 transcription factors suppresses excitatory synapse number. Science. 2006;311:1008–1012. doi: 10.1126/science.1122511. [DOI] [PubMed] [Google Scholar]
  • 78.Chao HW, Hong CJ, Huang TN, Lin YL, Hsueh YP. SUMOylation of the MAGUK protein CASK regulates dendritic spinogenesis. J Cell Biol. 2008;182:141–155. doi: 10.1083/jcb.200712094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cheng J, Huang M, Zhu Y, Xin YJ, Zhao YK, Huang J, et al. SUMOylation of MeCP2 is essential for transcriptional repression and hippocampal synapse development. Journal of neurochemistry. 2014;128:798–806. doi: 10.1111/jnc.12523. [DOI] [PubMed] [Google Scholar]
  • 80.Craig TJ, Henley JM. Protein SUMOylation in spine structure and function. Current opinion in neurobiology. 2012;22:480–487. doi: 10.1016/j.conb.2011.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Qi Y, Wang J, Bomben VC, Li DP, Chen SR, Sun H, et al. Hyper-SUMOylation of the Kv7 potassium channel diminishes the M-current leading to seizures and sudden death. Neuron. 2014;83:1159–1171. doi: 10.1016/j.neuron.2014.07.042. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

supplement

RESOURCES