Abstract
Bergmann glia facilitate granule neuron migration during development and maintain the cerebellar organization and functional integrity. At present, molecular control of Bergmann glia specification from cerebellar radial glia is not fully understood. In this report, we show that ZEB2 (aka, SIP1 or ZFHX1B), a Mowat–Wilson syndrome-associated transcriptional regulator, is highly expressed in Bergmann glia, but hardly detectable in astrocytes in the cerebellum. The mice lacking Zeb2 in cerebellar radial glia exhibit severe deficits in Bergmann glia specification, and develop cerebellar cortical lamination dysgenesis and locomotion defects. In developing Zeb2-mutant cerebella, inward migration of granule neuron progenitors is compromised, the proliferation of glial precursors is reduced, and radial glia fail to differentiate into Bergmann glia in the Purkinje cell layer. In contrast, Zeb2 ablation in granule neuron precursors or oligodendrocyte progenitors does not affect Bergmann glia formation, despite myelination deficits caused by Zeb2 mutation in the oligodendrocyte lineage. Transcriptome profiling identified that ZEB2 regulates a set of Bergmann glia-related genes and FGF, NOTCH, and TGFβ/BMP signaling pathway components. Our data reveal that ZEB2 acts as an integral regulator of Bergmann glia formation ensuring maintenance of cerebellar integrity, suggesting that ZEB2 dysfunction in Bergmann gliogenesis might contribute to motor deficits in Mowat–Wilson syndrome.
SIGNIFICANCE STATEMENT Bergmann glia are essential for proper cerebellar organization and functional circuitry, however, the molecular mechanisms that control the specification of Bergmann glia remain elusive. Here, we show that transcriptional factor ZEB2 is highly expressed in mature Bergmann glia, but not in cerebellar astrocytes. The mice lacking Zeb2 in cerebellar radial glia, but not oligodendrocyte progenitors or granular neuron progenitors, exhibit severe defects in Bergmann glia formation. The orderly radial scaffolding formed by Bergmann glial fibers critical for cerebellar lamination was not established in Zeb2 mutants, displaying motor behavior deficits. This finding demonstrates a previously unrecognized critical role for ZEB2 in Bergmann glia specification, and points to an important contribution of ZEB2 dysfunction to cerebellar motor disorders in Mowat–Wilson syndrome.
- Bergmann glia
- cerebellar gliogenesis and neurogenesis
- cerebellar lamination
- granular neuron migration
- motor deficits
- ZEB2
Introduction
The cerebellum is essential for smoothly coordinated vestibular, motor and cognitive function. Cerebellar malformation disrupts not only balance and locomotion (Ito, 2006), but also sensory-motor learning, speech, and spatial memory (Boyden et al., 2004; Buckner, 2013). The cerebellar cortex consists of three layers: an outer molecular layer (ML), the middle Purkinje cell layer (PCL), and an inner granular layer (IGL). The IGL neurons, the most prevalent cerebellar neurons, are derived from granule neuron precursors (GNPs), which are peripherally located in the external germinal layer (EGL) during early postnatal stages. The PCL contains the soma of Purkinje neurons and Bergmann glia.
Bergmann glia are “specialized” radial astrocytes in the cerebellar cortex, where their cell bodies are located in the PCL and processes extend into the ML layer, terminating at the pial surface (Hatten, 1999; Wang and Zoghbi, 2001). During early postnatal stages, GNPs proliferate in the EGL and migrate radially along the Bergmann glial process in an outside-in manner to form mature IGL neurons in the cerebellum (Sillitoe and Joyner, 2007; Millen and Gleeson, 2008). Bergmann glia also interact with Purkinje neurons and facilitate synaptic transmission and maintenance (Bellamy, 2006; López-Bayghen et al., 2007). Impairment of Bergmann-glia-mediated GNP migration disrupts the laminar structure of the cerebellum and synaptic connections and thereby disrupts cerebellar functions (Roussel and Hatten, 2011; Buckner, 2013).
Bergmann glia are derived from radial glia in the cerebellar ventricular zone through retraction of apical processes (Yuasa, 1996; Yamada and Watanabe, 2002). During the radial-to-Bergmann glial transition, which occurs between mouse embryonic days (E)14.5 and E18.5, Bergmann glia precursors maintain the radial basal processes and relocate their soma from the ventricular zone to the future PCL, and subsequently differentiate into mature Bergmann glia at ∼P6 (Yuasa, 1996; Yamada and Watanabe, 2002). Bergmann glial radial fibers facilitate inward migration of differentiating GNPs from the EGL to form the IGL (Buckner, 2013).
Coordinated spatial and temporal gene expression drives the production and patterning of Bergmann glia, which are required for proper cerebellum lamination (Sudarov and Joyner, 2007). A number of signaling pathways have been implicated in regulating these processes. FGFR signaling in Bergmann glia is required for normal cerebellar architecture formation (Müller Smith et al., 2012). In addition, Notch signaling ligand DNER and NOTCH1/RBP-J are critical for monolayer formation and morphogenesis of Bergmann glia (Lütolf et al., 2002; Eiraku et al., 2005; Komine et al., 2007). Integrin-linked kinase signaling is also required for Bergmann glial differentiation (Belvindrah et al., 2006). Recently, a role of Ptpn11/Shp2 and ErbB3 signaling in Bergmann glia maturation was identified during the initiation of cerebellar foliation (Li et al., 2014; Sathyamurthy et al., 2015). In contrast to the multiple signaling pathways identified for Bergmann glia development, the cell-intrinsic transcriptional regulators that control Bergmann glia formation have not been fully defined.
Patients with Mowat–Wilson syndrome (MOWS), an autosomal dominant disorder, exhibit congenital developmental abnormalities such as mental retardation, growth delay, epilepsy, and motor deficits (Mowat et al., 2003). MOWS is caused by mutations in ZEB2 (Cacheux et al., 2001; Wakamatsu et al., 2001). Recent studies indicate that Zeb2-mediated gene programs control neurogenesis, the fate switch between cortical and striatal interneurons, and cortical interneuron migration in the developing brain (Seuntjens et al., 2009; McKinsey et al., 2013; van den Berghe et al., 2013; Parthasarathy et al., 2014). ZEB2, also known as SIP1 (Smad-interacting protein 1), can interact with Smads and modulate TGFβ/BMP signaling to regulate oligodendroglial differentiation (Verschueren et al., 1999; Weng et al., 2012).
In this study, we find that ZEB2 is expressed in Bergmann glia and is required for Bergmann glial specification from radial glia. Mice lacking Zeb2 in radial glia failed to form Bergmann glia, and the orderly radial scaffolding formed by Bergmann glial fibers critical for cerebellar lamination was not established. Thus, our data reveal that ZEB2 is a key regulator for Bergmann glial formation during cerebellar development.
Materials and Methods
Animals.
Zeb2lox/lox mice (Wu et al., 2016) were crossed with hGFAP-Cre or Atoh1/Math1-Cre (He et al., 2014), or Olig1-Cre (Xin et al., 2005) mice to produce control (Zeb2lox/+:Cre+/−) and Zeb2 conditional knock-out offspring (Zeb2lox/lox:Cre+/−). Littermates Zeb2lox/lox or Zeb2lox/+:Cre+/− mice were used as controls. Transgenic hGFAP-Cre and GFAP-GFP reporter mice were obtained from The Jackson Laboratory, FVB-Tg(GFAP-cre)25Mes/J and FVB/N-Tg(GFAPGFP)14Mes/J, respectively. Animals of either sex were used in the study and littermates were used as controls unless otherwise indicated. The GFAP-GFP line was intercrossed with the hGFAP-Cre line to obtain Zeb2-conditional knock-out (cKO) mice carrying the GFAP-GFP transgene. The mouse strains used in this study were generated and maintained on a mixed C57BL/6;129Sv background and housed in a vivarium with a 12 h light/dark cycle. All animal experiments were conducted in mice of both genders. All animal use and studies were approved by the Institutional Animal Care and Use Committee at the Sichuan University and the Cincinnati Children's Hospital Medical Center.
Immunostaining and in situ hybridization.
The brains of mice at defined ages were dissected and fixed for 45 min in 4% PFA, embedded in 4% agarose, and sectioned at 50 μm (for postnatal samples) and 60 μm (for embryonic samples) as longitudinal vibratome-sections. For BrdU pulse labeling, animals were injected subcutaneously with 100 mg BrdU/kg body weight for appropriate times before collection. For immunostaining, we used antibodies to ZEB2 (rabbit; Santa Cruz Biotechnology, SC-48789), BLBP (rabbit; Abcam, ab32423), ZIC1 (rabbit; Rockland, 200-401-159), Calbindin (rabbit; Immunostar, 24427), NeuN (mouse; Millipore, MAB377), GFAP (mouse; Sigma-Aldrich, G3893), MBP (goat; Santa Cruz Biotechnology, SC-13914), Ki67 (rabbit; Thermo Scientific, RM-9106), and BrdU (rat; Abcam, ab6326). Secondary antibodies conjugated to Cy2, Cy3, or Cy5 were from Jackson ImmunoResearch Laboratories. All images were acquired using an Olympus Fluoview FV1000 confocal microscope and quantified in a double-blinded manner by ImageJ (https://imagej.nih.gov/ij/). Images from at least five sections per animal were collected for analysis. RNA in situ hybridization was performed using digoxigenin-labeled riboprobes as described previously. The probes used were as follows: murine Glast/Slc1a3, Fgfr2, Ntng2, Gdf10, Notch2, and Hes5. Detailed protocols are available upon request. Images were taken using a Nikon Eclipse 80i microscope.
Cell number quantification in the cerebellum.
Cerebella were sectioned sagittally at 50 μm. Cerebellar hemispheric sections were immunostained with cell-type-specific markers and counted by visualizing labeling positive cells in the parasagittal sections using ImageJ software. Three individual mice from each group were used for the experiment, and at least four sagittal sections from each animal were used to count the total number of labeled cells in a genotyping blinded manner. Data were analyzed with unpaired Student's t tests.
RNA isolation and quantitative real-time PCR.
RNAs from control and Zeb2-cKO mouse cerebella were extracted using TRIZOL (Life Technologies). cDNA was synthesized from 1 μg RNA using PrimeScript RT reagent Kit (Takara) according to the manufacturer's instructions. QRT-PCR was performed using the CFX96 Touch Real-Time PCR Detection System (Bio-Rad). qRT-PCR was performed using FAST qPCR Kit Master Mix (KAPA Biosystems). Primers used for qRT-PCR analysis are as follows: Zeb2: Forward, accttacgaatgcccaaact; Reverse, gggaagaacccgtcttgatatt; Glast/Slc1a3: Forward, aagttcagagcctcaccaag; Reverse, ctcattttatacggtcggaggg; Fgfr2: Forward, tatggaag aggaccagggatt; Reverse, tggttctaaagtggtatcctcaac; Notch2: Forward atgaagacgaagatgctgagg; Reverse, catcagctctcgaatagcgg Hes1: Forward, ggcgaagggcaagaataaatg; Reverse, gtgcttcacagtcatttccag; Hes5: Forward, gaaacacagcaaagccttcg Reverse, agcttcatctgcgtgtcg; Gdf10: Forward, gtcctcattgccctcgg; Reverse, cggttgtacttctcatagagcc; Ntng2: Forward, gctctcccaatgcctgtg; Reverse, gttgtgtttacagctgacgc.
Transient transfections and luciferase assays.
For reporter assays, HEK293 cells were transfected with pGL3-luciferase reporters driven by indicated promoters carrying ZEB2 consensus binding sites to the transcription start site (Fgfr1: −1.9 to −0.1 kb; Fgfr2: +1.3 to +2.8 kb; Hes1: −5.0 to −2.3 kb; Hes5: −1.5 to +0.1 kb; erbB3: −0.3 to +1.2 kb; Gdf10: −2.8 to −1.6 kb) using PolyJet per the manufacturer's protocol and assayed 48 h post-transfection for luciferase activity by using a Promega luciferase assay kit according to the manufacturer's instructions. The pSV-β-galactosidase control vector was included to control for variable transfection efficiencies between independent experiments.
RNA sequencing and data analysis.
RNA from control and Zeb2 mutant cerebella were extracted using TRIZOL (Life Technologies) followed by purification using an RNeasy Mini Kit (Qiagen). RNA-seq libraries were prepared using Illumina RNA-Seq Preparation Kit and sequenced by HiSeq 2000 Sequencer. RNA-seq reads were mapped using TopHat with default settings (http://tophat.cbcb.umd.edu). TopHat output data were then analyzed by Cufflinks to (1) calculate FPKM values for known transcripts in mouse genome reference, and (2) test the changes of gene expression between mutant and control. GO-analysis of gene expression changes was performed using Gene Set Enrichment (GSEA; http://www.broadinstitute.org/gsea/index.jsp). Normalized enrichment score (NES) reflects the degree to which the gene-set is overrepresented at the top or bottom of a ranked list of genes. The GSEA summary plots showing upregulated and downregulated pathways were plotted according to (https://www.biostars.org/p/168044/). Genes categorized with negative or positive NES are downregulated or upregulated, respectively. Circle size is proportional to the number of significant genes defined here as the number of genes represented in the leading-edge subset, i.e., the subset of members within a gene set that shows statistically significant, concordant differences between two biological states and contribute most to the NES. Circle colors represent FDR q values. The heat map was generated based on log2 [FPKM] (fragments per kilobase of transcript per million mapped reads) by AltAnalyze (http://www.altanalyze.org/) with normalization of rows relative to row mean.
Statistical analysis.
All analyses were done using Microsoft Excel or GraphPad Prism 6.00 (www.graphpad.com). Data are shown in histograms as mean ± SEM. p < 0.05 is deemed statistically significant. Data distribution was assumed to be normal, but this was not formally tested. Count data were assumed to be nonparametric, and appropriate statistical tests were used. Statistical analysis was performed by two-tailed unpaired Student's t tests. Quantifications were performed from at least three experimental groups in a blinded fashion. No statistical methods were used to predetermine sample sizes, but our sample sizes are similar to those generally used in the field. No animals or data points were excluded from analyses.
Accession code.
All the RNA-seq data have been deposited in the NCBI Gene Expression Omnibus under accession number GSE84058.
Results
Bergmann glia express ZEB2 in the developing cerebellar cortex
To determine the identity of Zeb2-expressing cells in the developing cerebellar cortex, we assessed ZEB2 expression by immunohistochemistry in GFAP-promoter driven GFP (GFAP-GFP) transgenic mice. In these mice, the GFAP-GFP reporter is expressed in Bergmann glia in the PCL and in astrocytes in the white matter in the postnatal stages (Zhuo et al., 1997; Koirala and Corfas, 2010). ZEB2 expression in the PCL overlapped with GFAP-GFP (Fig. 1A), a surrogate marker for Bergmann glia within the PCL (Koirala and Corfas, 2010). Approximately 98% and 90% ZEB2+ cells were colabeled with GFP in the PCL at P0 and P6, respectively (Fig. 1B). GFAP-GFP colabeling with BLBP (brain lipid-binding protein), a marker for Bergmann glia, confirmed that the GFP reporter was expressed in Bergmann glia (Fig. 1C). Consistently, at P6, ZEB2 was detected in GFAP-expressing Bergmann glia in the PCL (Fig. 1D). In contrast, ZEB2 was hardly detectable in the GFAP-GFP+ (Fig. 1E,F) or GFAP+ (Fig. 1G) astrocytes of the cerebellar white matter, suggesting that ZEB2 is mainly expressed in Bergmann glia but not astrocytes in the cerebellum. We did not detect ZEB2 expression in IGL neurons marked NeuN at P6 (Fig. 1H). Similarly, ZEB2 was essentially absent in the granule neuron progenitors in the EGL at P0 (Fig. 1I). In the developing cerebellum, a population of ZEB2 was detected in SOX2+ radial glia or neural stem/progenitors in the ventricular zone (VZ) of the cerebellum at E14.5 (Fig. 1J). These observations suggest that ZEB2 expression is detected in the radial glia and Bergmann glia in the developing cerebellum.
Deletion of Zeb2 in radial glia/neural progenitors disrupts cerebellar development
To assess the function of Zeb2 in Bergmann glia formation from radial glia/neural progenitors during cerebellar development, we bred Zeb2-floxed mice with a human GFAP promoter driven-Cre (hGFAP-Cre) deleter line (Zhuo et al., 2001). hGFAP-Cre is expressed in cerebellar radial glia or neural stem/progenitors, which can give rise to the majority of the cell types including Bergmann glia, astrocytes, oligodendrocytes, and granule neurons in the cerebellum (Zhuo et al., 2001; Malatesta et al., 2003; Yue et al., 2006). The resulting Zeb2-ablated mice (hGFAP-Cre-Zeb2fl/fl, referred to here as Zeb2-cKO mice) were born at a normal Mendelian ratio; however, Zeb2-cKO mice developed severe tremors and defects in balance control beginning ∼2 weeks after birth. The phenotypes of Zeb2 heterozygous control animals (Zeb2fl/+:hGFAP-Cre) are essentially the same as those in wild-type or Zeb2fl/fl mice. In contrast to control mice, which stride forward with a regular gait in a footprint test, the Zeb2-cKO mice exhibited irregular gaits and had difficulty in maintaining equilibrium (Fig. 2A), indicating that locomotor function is severely impaired in mice that lack Zeb2.
At E18.5, multiple lobular foliations were detected in the cerebella of heterozygous control animals that were similar to that in wild-type animals. In contrast, cerebellar foliation and fissures between adjacent lobules were barely detectable in Zeb2-cKO mutants at this stage (Fig. 2B). At P28, the cerebella of control mice exhibited well organized cerebellar lobules with formation of distinct molecular layers and granular layers, whereas Zeb2-cKO mice had smaller cerebella with disorganized lobular structures (Fig. 2C,D). Consistently, the cerebellum in the Zeb2-cKO mice exhibited hypoplasia in the cerebellar vermis and hemisphere (Fig. 2C), and appeared smaller and does not reach the size of the control cerebellum. In contrast to the disappearance of GNPs in EGL due to their inward migration in control mice at P28, clusters of GNPs were retained at the surface of the cerebellum in the Zeb2 mutant mice (Fig. 2E, arrows).
To determine whether ectopic cells were GNPs that had not successfully migrated, we stained control and Zeb2-cKO cerebella for ZIC1, which marks both GNPs in the EGL and mature neurons in the granular layer (Wang and Zoghbi, 2001; He et al., 2014), and for NeuN, which marks mature neurons in the IGL. In control mice at P18, the majority of GNPs had migrated to the IGL (Fig. 2F). Only a few ZIC1+ cells were present in the ML; these cells are likely completing the migration process from EGL to the IGL. In contrast, clusters of ZIC1+ and NeuN+ neurons were detected in the EGL and ML in Zeb2-cKO cerebella (Fig. 2E, inset). The ectopic cell clusters stayed on the surface of ML in the cerebellar cortex, however, they were not Ki67+ proliferative GNPs (Fig. 2G), suggesting that they are postmitotic GNPs or immature granule neurons, and fail to migrate into the molecular layer to become mature granule neurons in the IGL of the Zeb2-cKO mice.
Impaired radial migration of granule neurons in Zeb2-cKO mutants
A characteristic feature of Bergmann glia is that they exhibit unipolar morphology and form parallel processes that provide scaffolding for GNP migration during cerebellar development. Beginning at P6, GNPs begin to migrate inward along Bergmann glia processes and eventually cross the PCL to form the IGL. In control mice at P10, GFAP+ Bergmann glial processes were parallel to each other and projected into the pial surface along the ML (Fig. 3A). These migrating GNPs were visible in the ML as were GNPs distributed along the EGL in wild-type cerebella (Fig. 3A). Postmitotic granule neurons, which have successfully migrated across the PCL, form the IGL. Cerebellar cortical lamination was distinctly organized into the EGL, ML, and IGL at P10 (Fig. 3A).
In contrast to control mice, no parallel radial processes were detected in Zeb2-cKO mutants due to the absence of Bergmann glia at P10 (Fig. 3A). In addition, GFAP-GFP+ cells were scattered across the ML/PCL of Zeb2-cKO mutants compared with those lining along the PCL in the control (Fig. 3B). Furthermore, the number of Bergmann glia-like cells (GFP+ cells with radial processes) were much reduced in Zeb2-cKO animals at P3 and P10 (Fig. 3C). The majority of GFAP-GFP+ glia detected in the Zeb2-cKO cerebellar cortex expressed astrocytic S100β (Fig. 3D) without unipolar processes, suggesting that these GFAP-GFP+/S100β+ cells represent astrocytes. In the mutant cerebella, ZIC1+ granule neurons or their progenitors were dispersed throughout the EGL, ML, and IGL without a clear ML boundary between EGL and IGL as in the control (Fig. 3A).
To verify GNP migration defects, we performed BrdU fate-tracing study of proliferating GNPs by injecting BrdU at P6, when the majority of GNPs are proliferative, into control and mutant mice. The cerebella were harvested 110 h after BrdU injection at P6 and immunostained for BrdU and calbindin. In control mice, we observed very few granule neurons with BrdU label in the ML. Most BrdU label-retaining GNPs had migrated through PCL, marked by calbindin labeling, into the IGL (Fig. 3E). In the Zeb2-cKO cerebella, however, the majority of the BrdU-labeled GNPs remained in the EGL and ML, and very few BrdU+ GNPs migrated into the IGL (Fig. 3E,F). These observations suggest that deletion of Zeb2 in radial glia blocks Bergmann glial formation, leading to defects in GNP migration into the IGL.
Specification of Bergmann glia is impaired in Zeb2-cKO mutants
In control embryos, at E15.5, GFAP-GFP-expressing precursors appear to migrate outward to the cerebellar cortex from the ventricular zone with long radial processes directed toward the pial surface (Fig. 4A). By E18.5, the GFAP-GFP+ Bergmann glia had successfully reached their destination and cell bodies were aligned with the PCL (Fig. 4B). In contrast, in Zeb2-cKO embryos, the GFAP-GFP+ cells were barely detectable in the cerebellar cortex at E15.5. At E18.5, we however did not detect these GFAP-GFP+ glial cells in the location along the PCL occupied principally by Bergmann glia (Fig. 4B). Most of the detectable GFAP-GFP-expressing glia were dispersed throughout the inner cerebellar region in Zeb2-cKO mutants. These observations suggest a deficit in the specification of Bergmann glia in the developing cerebellum of Zeb2-cKO mutants. There were significantly fewer GFP+ glia in the presumptive PCL in Zeb2-cKO animals than in controls at E18.5 (Fig. 4C). The number of activated cleaved-caspase 3-labeled cells in the cerebellum was similar between control and Zeb2 mutant animals (data not shown), suggesting that the reduction in the number of Bergmann glia is not due the compromised cell survival.
To evaluate the effects of ZEB2 on the proliferation of GFAP-GFP+ glia, we immunostained for a cell proliferation marker Ki67 in cerebella during embryogenesis, when Bergmann glia undergo extensive proliferation. At E15.5 and E17.5, the number of GFP+ cells stained for Ki67 in the presumptive PCL was reduced in Zeb2-cKO animals (Fig. 4D–F). Similarly, the proliferation rate of GFAP-GFP+ cells (the percentage of Ki67+GFP+ among GFP+ cells) was lower in Zeb2-cKO cerebella compared with controls (Fig. 4G), suggesting that the defects in Bergmann glia generation is in part attributed to the deficiency of GFAP-GFP+ glial precursor expansion in Zeb2-cKO mutants.
Granule neurons and Purkinje cells are generated in the Zeb2-cKO cerebellum
To determine whether the formation of GNPs in the EGL was affected due to the loss of Zeb2 in cerebellar neural progenitors, we quantified GNPs in the EGL in control and Zeb2-cKO mutant cerebella assayed by expression of ZIC1, which marks both outer EGL (mitotic GNPs) and the inner EGL (postmitotic GNPs; Wang and Zoghbi, 2001). At P3, the density of ZIC1+ GNPs in the EGL was comparable between control and Zeb2-mutants (Fig. 5A,B). In addition, we did not detect a significant difference in the percentage of BrdU+ GNPs in the EGL between control and Zeb2-cKO mice (Fig. 5C,D), suggesting that Zeb2 deletion in radial glia does not affect GNP generation.
To determine the effects of Zeb2 ablation on Purkinje cell development, we examined expression of calbindin, a Purkinje cell marker, at postnatal stages. Calbindin+ Purkinje cells aligned to form a monolayer and extended their dendrites toward the EGL in the control animals at P3 and P10 (Fig. 5E). In Zeb2-cKO mutants, Purkinje cells had extended dendrites, but their cell bodies aggregated into multicellular clusters (Fig. 5E). Nonetheless, overall Purkinje cell numbers were comparable between control and mutant cerebella at P10 (Fig. 5F). These observations suggest that ZEB2 is not required for the formation of granule neurons or Purkinje cells.
Zeb2 deletion in oligodendrocyte progenitors or GNPs does not affect Bergmann glia formation
Previous studies showed that ZEB2 is expressed in oligodendrocyte lineage cells and is critical for oligodendrocyte differentiation (Weng et al., 2012). To evaluate whether the defects in Bergmann glia development in Zeb2-cKO animals can be caused by myelination deficiency in a non-cell autonomous manner, we examined cerebellar development in Zeb2fl/fl:Olig1-Cre mice with Zeb2 deletion in the oligodendrocyte lineage. At P14, Purkinje cells and Bergmann glia process formation marked by calbindin and GFAP, respectively, appeared normal in the Zeb2fl/fl:Olig1-Cre mutant cerebellum. The cerebellar cortical lamination and foliation were essentially intact except for the loss of expression of MBP and ZEB2 expression in the cerebellar white matter in the Zeb2fl/fl:Olig1-Cre mutant mice (Fig. 6A,B), suggesting that myelination deficits in the Zeb2fl/fl:Olig1-Cre mutants do not affect Bergmann glia formation.
To examine the possibility that the Bergmann glia defects in Zeb2-cKO mutant mice may be caused indirectly by the loss of ZEB2 function in GNPs, which are derived from neural progenitors, we generated mice in which Zeb2 floxed alleles were deleted using a GNP-specific Cre line, Atoh1/Math1-Cre (Lumpkin et al., 2003; He et al., 2014). The cerebellar foliation, and the morphology and number of S100β+ Bergmann glia in the cerebellum of Zeb2fl/fl:Atoh1-Cre mice were comparable to the control (Zeb2fl/+:Atoh1-Cre) mice at P24 (Fig. 6C,D). The granule neuron marker ZIC1 expression was not altered in the cerebellum of Zeb2fl/fl;Atoh1-Cre mutant mice (Fig. 6E), consistent with the lack of ZEB2 expression in GNPs at the EGL (Fig. 1). In Zeb2fl/fl:Atoh1-Cre mice at P1, BrdU-pulse labeling showed that the percentage of BrdU+ cells among ZIC1+ GNPs in the EGL were indistinguishable from control animals (Fig. 6F,G), suggesting that deletion of Zeb2 in Atoh1+ GNPs does not affect the formation of granule neurons and Bergmann glia.
Differentially expressed gene profiling in Zeb2-cKO cerebella identifies new Bergmann glial markers
To determine the potential mechanisms underlying ZEB2 regulation of Bergmann glia development, we performed RNA-seq profiling using cerebella of control and Zeb2-cKO mice at P0, when Bergmann glial cells undergo proliferation and specification processes. The control and Zeb2-cKO groups exhibited distinct gene expression profiles (Fig. 7A). We identified a set of differentially expressed genes in the Zeb2-cKO cerebella relative to the control by interrogating gene expression signatures. Gene profiling analysis indicated that the genes upregulated in Zeb2-cKO cerebella, were associated with epithelial-to-mesenchymal transition (EMT), which is consistent with a role of ZEB2 in EMT through repressing epithelial cell–cell junction gene expression (Vandewalle et al., 2005), GPCR signaling, calcium, STAT3 signaling, and astrocytic signature genes (Fig. 7B). Consistently, GSEA revealed an upregulation of astrocytic signatures alongside the EMT pathway in Zeb2-cKO cerebella (Fig. 7C). In contract, the genes that were downregulated in Zeb2-cKO cerebella indicated an enrichment of gene categories that are related to mitotic cell cycle, Bergmann glia- and oligodendrocyte-expressing genes (Fig. 7B–D), consistent with the defects in Bergmann glia specification and proliferation as well as myelination in Zeb2-cKO mice.
Genes known to be enriched in Bergmann glial cells like Glast (Slc1α3) were downregulated in the Zeb2-cKO mutant (Fig. 7D). In addition, genes encoding several signaling components in the Notch pathway (e.g., Hes5 and Notch2), FGF pathway (e.g., Fgfr2, Fgf9, and Ptpn11), and the BMP pathway such as Gdf10, also known as Bmp3b, were downregulated in Zeb2-cKO cerebella. FGF/FGFR signaling (Lin et al., 2009; Müller Smith et al., 2012; Li et al., 2014), NOTCH/HES signaling (Eiraku et al., 2005; Komine et al., 2007) and BMP signaling (Mecklenburg et al., 2014) have been shown to regulate Bergmann glial development. Notably, we found that Ntng2, which encode Netrin G2 in the Netrin family functioning in axonal guidance and neuronal migration (Barallobre et al., 2005; Nishimura-Akiyoshi et al., 2007), exhibited an expression pattern that resembles that of Bergmann glia-expressing markers like Glast and Gdf10 (Fig. 7D).
Given the defects in Bergmann glia differentiation, we focused on the downregulated genes in Zeb2-cKO mutants, which likely identified the genes that are highly expressed in Bergmann glia. To verify dysregulation in FGF, NOTCH, BMP, and Netrin pathways, we performed in situ hybridization to examine the expression of components of these pathways in control and Zeb2-cKO mutants. In control cerebella, expression patterns of Fgfr2, Hes5, Notch2, Gdf10, and Ntng2 at E17.5 were similar to that of a Bergmann glial-expressing gene Glast (Fig. 7E), suggesting that expression of these genes is enriched in Bergmann glia. In contrast, in Zeb2-cKO cerebella, expression of Fgfr2, Hes5, Notch2, Gdf10 and Ntng2 did not extend to where Bergmann glia are located along the PCL (Fig. 7E). Quantification by qRT-PCR further indicated that these mRNAs were expressed at lower levels in the Zeb2-cKO cerebella than wild-type controls (Fig. 7F), consonant with reduced in situ signals in the Bergmann glia at the PCL layer in the mutants. Consistently, Zeb2 overexpression enhanced the activity of reporter genes driven by the promoters of FGF receptors Fgfr1 and Fgfr2, a NOTCH effector Hes5, as well as a TGFβ receptor ligand Gdf10 (Fig. 7G), suggesting that ZEB2 expression potentially promotes FGF receptor, NOTCH and TGFβ receptor signaling pathways for Bergmann gliogenesis.
Discussion
Our studies present a previously unrecognized, essential role of the transcription factor ZEB2 for Bergmann glia specification from radial glial cells in the cerebellum. In addition to present cerebellar radial glia/neural progenitor cells in the VZ and oligodendrocytes, ZEB2 is highly expressed in Bergmann glia, but hardly detectable in astrocytes in the cerebellar white matter tract or interneurons in the postnatal cerebellum, consistent with expression of Zeb2/Sip1-EGFP knock-in reporter detected in Bergmann glia (Nishizaki et al., 2014). Because there are no well established specific markers to distinguish astrocytes and Bergmann glia, except for their distinct anatomical locations and morphology, ZEB2 may represent a cell nucleus-expressing marker for mature Bergmann glia that are distinct from astrocytes in the cerebellar cortex.
During embryonic development, Bergmann glia precursors are derived from the radial glial cells in the VZ. The nascent Bergmann glia precursors continue to proliferate until P7, when they exit the cell cycle and differentiate into mature Bergmann glia (Parmigiani et al., 2015). The lack of radial process-bearing Bergmann glial formation, but not astrocytes, in the presumptive PCL in the Zeb2-cKO embryos, where ZEB2 is deleted in radial glia/neural progenitors mediated by hGFAP-Cre, suggests that ZEB2 is required for Bergmann glia specification from radial glia in the developing cerebellum.
It is worth noting that GFP-expressing cells from the hGFAP-GFP transgenic line label specifically astrocytes and Bergmann glia in the developing cerebellum as previously reported (Su et al., 2004; Koirala and Corfas, 2010), they are not equivalent to those labeled by hGFAP-Cre transgenic line, where Cre recombination occurs in radial glia or neural progenitors at embryonic stages, which give rise to both glia and neurons (Zhuo et al., 2001). hGFAP-Cre is also shown to mediate floxed allele recombination in astrocytes, Bergmann glia and their progenitors (Komine et al., 2007). In the developing forebrain, ZEB2 is critical for neurogenesis, neuronal migration, and oligodendrogenesis from neural progenitor cells (Seuntjens et al., 2009; McKinsey et al., 2013; van den Berghe et al., 2013; Parthasarathy et al., 2014). Although we cannot completely exclude the possibility that the defects in cortical development lead to impairment of cerebellar development in Zeb2-cKO mice, we show that ZEB2 is expressed in Bergmann glia, and observed a specific loss of Bergman glia, but not granule neurons despite their migration being impaired.
Although suitable specific markers for Bergmann glia precursors are not available at present, the number and the proliferation of glial precursors labeled by GFAP-GFP in the presumptive PCL at embryonic stages is reduced. We do not detect significant alteration of cell death between control and Zeb2 mutant cerebella assayed by activated caspase 3 expression (data not shown). This suggests that ZEB2 may potentially regulate the proliferation of presumptive Bergmann glia precursor. Consistently, transcriptome profiling of Zeb2-cKO cerebellum at P0 reveals a significant downregulation of the genes related to mitotic cell cycle (Fig. 7). The failure in Bergmann glial differentiation likely contributes to severe defects in inward migration of GNPs from EGL to IGL, causing disorganization of the laminar structure of the cerebellar cortex in Zeb2-deficient cerebella.
Myelination defects caused by Zeb2 deletion in oligodendrocyte lineage cells by Olig1-Cre did not cause a defect in Bergmann glia formation, despite the lack of myelinating oligodendrocytes in the white matter of mutant cerebella. Knock-out of Zeb2 in Atoh1+ GNPs did not affect granule neuron formation and migration during cerebellar development, which is consistent with the lack of ZEB2 expression in GNPs. These observations suggest that defects in Bergmann glia formation in Zeb2-cKO mice was not due to the non-cell autonomous function of ZEB2 in myelination or GNPs. At present, there is no Bergmann glia-specific Cre line; we cannot definitely define the cell-autonomous role of ZEB2 in Bergmann glia formation or maturation. Given that Bergmann glia are derived from the cerebellar radial glia, our data indicate that ZEB2 is expressed in both Bergmann glia and the radial glia/neural progenitors in the cerebellar ventricular zone, and that deletion of Zeb2 in radial glia by hGFAP-Cre leads to defects in Bergmann glia specification.
In Zeb2-cKO animals, cerebellar dysgenesis appears more severe in anterior lobules than posterior lobules. The incomplete deletion of ZEB2 due to potential regional specificity of Cre recombination in the hGFAP-Cre line or distinct requirements of ZEB2 in a specific region of the cerebellum might contribute to the varying severity of the phenotypes across the Zeb2-cKO cerebellum. Alternatively, it is possible that other unidentified factors may compensate for the loss of ZEB2 function in Bergmann glia formation; however, their identity remains to be determined.
In the Zeb2-cKO cerebellum, intriguingly, some migration events occur and the IGL is visible, this suggests that GNPs potentially could migrate to a certain extent without radial processes or use other cellular processes. The mechanisms underlying inward migration of a population of GNPs in the absence of mature Bergmann glia in the Zeb2-mutant cerebellum remain to be determined. Nonetheless, ZIC1+ GNPs were dispersed throughout the ML and IGL layers appears to be disorganized in Zeb2-cKO mutant cerebella, suggesting that Bergmann glia are critical for the organized migration of GNPs and proper cerebellar lamination.
Given the defects in Bergmann glia differentiation in Zeb2-cKO mutants, downregulated genes may reflect those genes or pathways highly expressed in Bergmann glia. Transcriptome profiling analysis indicated a downregulation of NOTCH signaling components, including HES5, and FGF signaling such as FGFR2 in the Zeb2-cKO cerebellum. NOTCH signaling components are expressed in Bergmann glia in addition to neural progenitors (Tanaka et al., 1999; Ohtsuka et al., 2001; Koirala and Corfas, 2010; de Oliveira-Carlos et al., 2013). Similarly, FGFR2 is expressed in a spatiotemporal pattern consistent with expression in Bergmann glia (Lin et al., 2009). Given that the mice with deletion of Fgfr1/2 or Shp2, which is essential for FGF/ERK activation, display a defect in the induction of Bergmann glia formation similar to the Zeb2-cKO mice (Müller Smith et al., 2012; Li et al., 2014), it remains to be determined whether ZEB2 is critical for FGF signaling activation during Bergmann glia development. We find that overexpression of ZEB2 enhanced the promoter activity of FGF receptors Fgfr1 and Fgfr2, a NOTCH effector Hes5, as well as a TGFβ receptor ligand Gdf10, suggesting that ZEB2 overexpression may enhance FGF, NOTCH and TGFβ receptor signaling for Bergmann gliogenesis.
Transcriptome analysis reveals a set of new Bergmann glia-expressing genes. Consistent with the reduction of Bergmann glia in the Zeb2-cKO cerebellum, we observed a downregulation of axonal guidance factors such as Netrin G2, which regulates attraction and repulsion of neuronal axons (Barallobre et al., 2005; Nishimura-Akiyoshi et al., 2007) and GDF10, which modulates axonal sprouting (Li et al., 2015). In contrast to predominant expression of Netrin family members in neurons of other brain regions (Barallobre et al., 2005; Nishimura-Akiyoshi et al., 2007), our data that Ntng2 encoding Netrin G2 and GDF10 are highly enriched in Bergmann glia in the developing cerebellum, it would be interesting to determine whether Bergmann glia-expressing axonal guidance cues are required for GNP migration and Bergmann glial morphogenesis.
Bergmann glia and astrocytes share many common gene expression profiles, intriguingly, we found that Bergmann glia-enriched gene expression was downregulated, whereas a set of astrocytic genes (Cahoy et al., 2008) was upregulated in the Zeb2-cKO cerebellum (Fig. 7). GFAP-GFP+/S100β+ astrocytes were present in the PCL, where Bergmann glia were lost. This raises a possibility that ZEB2 may regulate the fate switch between Bergmann glia and astrocytes. In the absence of ZEB2, a population of Bergmann glia precursors that fail to differentiate into mature radial process-bearing Bergmann glia might become astrocytes.
Previous efforts to discover the mechanisms that drive cerebellar foliation initiation and gyrification have mostly focused on the role of GNPs and Purkinje cells (Lewis et al., 2004; Swanson et al., 2005; Corrales et al., 2006; Sudarov and Joyner, 2007). Our data show that the Zeb2 loss causes cerebellar foliation defects through regulating Bergmann glia formation, suggesting a critical role of Zeb2-dependent Bergmann glia development for cerebellar foliation and laminar organization. MOWS is an autosomal dominant disorder characterized by a nonrandom association of multiple birth defects impairing normal development and motor deficits. The phenotypic variation between the ZEB2 haploinsufficient human mutants and heterozygous knock-out mice could be due to a sensitivity to different gene dosage between mouse and human or dominant-negative effects of human mutant proteins, which may produce effects similar to homozygous-null Zeb2 in mice. The critical role of ZEB2 in the control of Bergmann glia formation in cerebellar lamination points to a potential mechanism of motor control and coordination in MOWS patients with ZEB2 mutations.
Footnotes
This work was funded in part by Grants from the U.S. National Institutes of Health R01NS072427 and R01NS075243 to Q.R.L., and the National Multiple Sclerosis Society (NMSS-4727) to Q.R.L. and the National Natural Science Foundation of China (81630038) to DM. We thank Guojiao Huang and Lingli Xu for technical support, Dr. Edward Hurlock for comments, and Dr. Danny Huylebroeck for Zeb2 floxed mice.
The authors declare no competing financial interests.
- Correspondence should be addressed to Dr. Q. Richard Lu, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229. richard.lu{at}cchmc.org