Skip to main content

Main menu

  • HOME
  • CONTENT
    • Early Release
    • Featured
    • Current Issue
    • Issue Archive
    • Collections
    • Podcast
  • ALERTS
  • FOR AUTHORS
    • Information for Authors
    • Fees
    • Journal Clubs
    • eLetters
    • Submit
  • EDITORIAL BOARD
  • ABOUT
    • Overview
    • Advertise
    • For the Media
    • Rights and Permissions
    • Privacy Policy
    • Feedback
  • SUBSCRIBE

User menu

  • Log in
  • My Cart

Search

  • Advanced search
Journal of Neuroscience
  • Log in
  • My Cart
Journal of Neuroscience

Advanced Search

Submit a Manuscript
  • HOME
  • CONTENT
    • Early Release
    • Featured
    • Current Issue
    • Issue Archive
    • Collections
    • Podcast
  • ALERTS
  • FOR AUTHORS
    • Information for Authors
    • Fees
    • Journal Clubs
    • eLetters
    • Submit
  • EDITORIAL BOARD
  • ABOUT
    • Overview
    • Advertise
    • For the Media
    • Rights and Permissions
    • Privacy Policy
    • Feedback
  • SUBSCRIBE
PreviousNext
Featured ArticleResearch Articles, Development/Plasticity/Repair

Migrating Pyramidal Neurons Require DSCAM to Bypass the Border of the Developing Cortical Plate

Tao Yang, Macy W. Veling, Xiao-Feng Zhao, Nicholas P. Prin, Limei Zhu, Ty Hergenreder, Hao Liu, Lu Liu, Zachary S. Rane, Masha G. Savelieff, Peter G. Fuerst, Qing Li, Kenneth Y. Kwan, Roman J. Giger, Yu Wang and Bing Ye
Journal of Neuroscience 13 July 2022, 42 (28) 5510-5521; DOI: https://doi.org/10.1523/JNEUROSCI.0997-21.2022
Tao Yang
1Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
6Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Macy W. Veling
1Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xiao-Feng Zhao
2Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Xiao-Feng Zhao
Nicholas P. Prin
1Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Limei Zhu
1Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ty Hergenreder
1Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hao Liu
1Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
2Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lu Liu
3Internal Medicine, Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Zachary S. Rane
1Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Masha G. Savelieff
1Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Peter G. Fuerst
4Department of Biological Sciences, University of Idaho, Moscow, Idaho 83844
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Peter G. Fuerst
Qing Li
3Internal Medicine, Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kenneth Y. Kwan
5Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Kenneth Y. Kwan
Roman J. Giger
2Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yu Wang
6Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Bing Ye
1Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109
2Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Bing Ye
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

During mammalian neocortex development, nascent pyramidal neurons migrate along radial glial cells and overtake earlier-born neurons to terminate at the front of the developing cortical plate (CP), leading to the outward expansion of the CP border. While much has been learned about the cellular and molecular mechanisms that underlie the migration of pyramidal neurons, how migrating neurons bypass the preceding neurons at the end of migration to reach their final positions remains poorly understood. Here, we report that Down syndrome cell adhesion molecule (DSCAM) is required for migrating neurons to bypass their postmigratory predecessors during the expansion of the upper cortical layers. DSCAM is a type I transmembrane cell adhesion molecule. It has been linked to Down syndrome through its location on Chromosome 21 trisomy and to autism spectrum disorders through loss-of-function mutations. Ex vivo time-lapse imaging demonstrates that DSCAM is required for migrating neurons to bypass their postmigratory predecessors, crossing the CP border to expand the upper cortical layers. In DSCAM-deficient cortices, migrating neurons stop prematurely under the CP border, leading to thinner upper cortical layers with higher neuronal density. We further show that DSCAM weakens cell adhesion mediated by N-cadherin in the upper cortical plate, allowing migrating neurons to traverse the CP border and expand the CP. These findings suggest that DSCAM is required for proper migratory termination and final positioning of nascent pyramidal neurons, which may provide insight into brain disorders that exhibit thinner upper layers of the cerebral cortex without neuronal loss.

SIGNIFICANCE STATEMENT Newly born neurons in the developing mammalian neocortex migrate outward toward the cortical surface, bypassing earlier born neurons to expand the developing cortex. How migrating neurons bypass the preceding neurons and terminate at the front of the expanding cortex remains poorly understood. We demonstrate that Down syndrome cell adhesion molecule (DSCAM), linked to Down syndrome and autism spectrum disorder, is required by migrating neurons to bypass their postmigratory predecessors and terminate migration in the outwardly expanding cortical layer. Migrating neurons deficient in DSCAM stop prematurely, failing to expand the cortex. We further show that DSCAM likely mediates migratory termination by weakening cell adhesion mediated by N-cadherin.

  • cell adhesion
  • cortex development
  • DSCAM
  • migration
  • pyramidal neuron
  • termination

Introduction

During mammalian neocortical development, nascent pyramidal neurons, born in the ventricular or subventricular zone (SVZ), migrate along radial glial cells to generate cortical layers. As they migrate, the later-born pyramidal neurons overtake earlier-born neurons to terminate at the border of the cortical plate (CP), leading to an “inside-out” pattern of cortical layer formation. Despite intense interest in this important process of cortical development, how migrating neurons bypass the preceding neurons at the end of migration to locate their final positions remains poorly understood.

Neuronal migration defects in mutant mice have provided insights into neuronal positioning during cortical genesis. In Reeler (reelin) and Scrambler [Disabled 1 (Dab1)] mice, migrating neurons do not follow the inside-out layer formation, resulting in an inverted cerebral cortex (Sheldon et al., 1997; D'Arcangelo, 2006). In reelin-deficient or Dab1-deficient mice, postmigratory neurons remain attached to the radial glia. Cell adhesion molecules regulate the neuronal detachment from radial glia (Anton et al., 1996). For example, α3 integrin expression in the upper CP differs in wild-type and Scrambler mice, and Reelin blocks α3 integrin expression at the top of the CP to facilitate neuron detachment (Sanada et al., 2004). SPARC-like 1 is also specifically expressed in the upper CP and may modulate neuronal adhesion to promote detachment (Gongidi et al., 2004). While these studies demonstrate that proper exit from radial migration is critical for the inside-out formation of cortical layers (Pinto-Lord et al., 1982; Sanada et al., 2004), how later-born neurons bypass the postmigratory preceding neurons at the CP border has not been characterized.

Down syndrome cell adhesion molecule (DSCAM) is a type I transmembrane cell adhesion molecule of the Ig superfamily (Yamakawa et al., 1998; Agarwala et al., 2000). Loss-of-function DSCAM mutations in human have been linked to neurobehavioral conditions, such as autism spectrum disorder (Turner et al., 2016; Wang et al., 2016; Narita et al., 2020), highlighting the importance of DSCAM in neurodevelopment. The Drosophila homolog of DSCAM, Dscam1, mediates dendrite or axon self-avoidance (Hughes et al., 2007; Matthews et al., 2007; Soba et al., 2007), a process that requires remarkable molecular diversity. Alternative splicing of the extracellular ectodomain of Drosophila Dscam1 can, in theory, generate >19,000 isoforms (Schmucker et al., 2000; Hattori et al., 2009). Furthermore, Drosophila Dscam1 expression levels dictate neuronal axon terminal growth independent of its ectodomain diversity (Kim et al., 2013).

Mammalian DSCAM does not possess ectodomain diversity like its Drosophila homolog (Schmucker and Chen, 2009). Rather, in mammals, neurite self-avoidance is mediated through protocadherin diversity (Zipursky and Sanes, 2010; Lefebvre et al., 2012). DSCAM also contributes to neurite self-avoidance in mammals in a fashion that does not require large molecular diversity. This is achieved by masking the cell adhesion mediated by cadherins (Garrett et al., 2018). For instance, several DSCAM−/− mutants exhibit retinal mosaic defects in mice, with excessive soma adhesion and process fasciculation (Fuerst et al., 2008, 2009, 2010). Intercellular DSCAM interaction decreases cell adhesion through masking cadherins and consequently promotes self-avoidance. DSCAM also regulates migrating neuron detachment from the ventricular surface in the midbrain (Arimura et al., 2020). Whether DSCAM is involved in cortical development remains poorly unknown.

In this study, we used ex vivo time-lapse imaging to demonstrate at the single-neuron resolution that DSCAM is required for migrating nascent neurons destined to upper cortical layers to bypass their postmigratory predecessor and migrate across the CP to expand the cortical layer. Migrating neurons deficient in DSCAM prematurely stops at the CP border together with their postmigratory predecessors. Thus, DSCAM-deficient neurons fail to expand the upper cortical layers, leading to a thinner and denser cortical layer. Our results show that DSCAM masks cell adhesion mediated by N-cadherin in the upper cortical plate, facilitating radial migrating neurons to bypass the border of the developing CP. These findings suggest that DSCAM is required for proper migratory termination and final positioning of nascent pyramidal neurons, which may provide insight into brain disorders that exhibit thinner upper layers of cerebral cortex without neuronal loss (Arnold and Trojanowski, 1996).

Materials and Methods

Mice.

In this study, we used DSCAM2j/2j mutant mice for DSCAM loss-of-function studies because it does not produce a protein product and mice survive postnatally on the inbred background that it arose on (Fuerst et al., 2010). The DSCAM2J allele harbors a spontaneous 4 bp duplication in exon 19, leading to a frameshift and loss of function. The 8-week-old DSCAM2j/+ mice (hereafter referred to as DSCAM+/−) were crossed to generate DSCAM+/+, DSCAM+/−, and DSCAM−/− mice. The nestin-CreERT2 mouse [C57BL/6-Tg(Nes-creERT2)KEisc/J; stock #016261, The Jackson Laboratory; Lagace et al., 2007], under the nestin promoter, expresses tamoxifen-induced Cre recombinase in adult and developing mouse brains, including neural progenitor cells (NPCs). The Ai14 mouse [GeneTrap B6.Cg-Gt(ROSA)26Sortm14(CAG-tdTomato)Hze/J; stock #007914, The Jackson Laboratory; Madisen et al., 2010] expresses the red fluorescent protein (RFP) tdTomato in a Cre-dependent fashion. DSCAM+/− animals were bred into nestin-CreERT2 and Ai14 mice to generate DSCAM−/−/nestin-CreERT2/CAG-tdTomato progeny with tamoxifen-inducible nestin-promoter-restricted tdTomato expression in NPCs. tdTomato was induced by tamoxifen injection into dams bearing embryonic day 14.5 (E14.5) embryos and was expressed by all NPC progeny cells.

All mice were handled in accordance with a protocol approved by the Institutional Animal Care and Use Committee at the University of Michigan. The mice were housed in a dedicated temperature-controlled (20°C) animal facility with a 12 h light/dark cycle maintained by the Unit for Laboratory Animal Medicine at the University of Michigan. The mice were housed in groups of no more than five per cage and were provided regular chow and water ad libitum. Their health was monitored onsite by a dedicated veterinarian staff.

In utero electroporation of mouse embryos.

In utero electroporation (IUE) was performed as previously described with some modifications (Sun et al., 2010; Yang et al., 2012). Pregnant female mice (E14.5) were anesthetized with isoflurane, and an incision was made to expose the embryos while still within the uterus. A plasmid DNA solution (∼2–3 µg/µl) containing 0.01% (w/v) Fast Green dye was injected (∼1–2 µl) into the right side of the lateral ventricle of each embryo brain using a glass micropipette. Each embryo head, still within the uterus, was placed between the leads of a Tweezertrode Electrode system (diameter, 5 mm; BTX, Harvard Bioscience) and administered five electrical pulses (50 V, 50 ms duration, 950 ms intervals) using an electroporator (ECM830, Harvard Bioscience). Embryos were returned to the pregnant female, and the muscle and skin were sutured, respectively. The pregnant female was monitored until she revived and was administered carprofen (5 mg/kg, i.p.) for pain management.

Genotyping embryonic tissues.

Embryo genomic DNA was purified for genotyping by digesting tail tissue in 300 µl of TES buffer (50 mm Tris, pH 8.0, 0.5% SDS, 0.1 m EDTA, in 1× PBS) with proteinase K overnight. Then, 5 m NaCl (160–200 µl) was added before centrifugation to remove precipitates. Next, isopropanol (180 µl) was added to the 300 µl of supernatant to precipitate the DNA. The DNA pellets were washed with 500 µl of 70% ethanol before dissolving in 20 µl of water. DNA flanking the DSCAM2j mutation were amplified by PCR (forward primer: 5′-gccctgtggtatttgctggtgtg-3′; reverse primer: 5′-gatgggcaaatgtcaaaggtcaaa-3′). The PCR product was sequenced with either primer to verify the presence of the mutation.

Live-cell time-lapse confocal microscopy of brain sections.

Brain slice culture and time-lapse confocal imaging were performed as previously described (Yang et al., 2012). Brains of E19.5 embryos were coronally sectioned in ice-cold artificial cortical spinal cord fluid on a vibratome (Leica Microsystems) into 300 µm slices. Brain slices were then immersed in DMEM and continuously perfused with DMEM bubbled with 95% O2 and 5% CO2 at 37°C on the microscope stage. Time-lapse imaging was performed using confocal microscopy (model TCS SP5, Leica) with an HC Fluotar L 25×/0.95 W VISIR immersion objective lens at 5 min intervals for a 15 h duration. The embryos were genotyped as described above.

Fluorescence immunohistochemistry of brain sections.

Brains were fixed by 4% paraformaldehyde (PFA; in PBS) overnight. Then, brains were immersed in 30% sucrose overnight, followed by embedding in optimal cutting temperature (O.C.T.) media (Tissue-Tek, Sakura), and freezing at −80°C. Brains were sectioned coronally at 100 µm thickness, permeabilized with 0.1% Triton X-100 in PBS, blocked with 2% donkey serum in PBS, and incubated overnight at 4°C with primary antibodies. The antibodies used are as follows: (1) mouse anti-NeuN (1:1000; catalog #ab104224, Abcam); (2) rabbit anti-Satb2 (1:1000; catalog #ab92446, Abcam); (3) rabbit anti-Tbr1 (1:1000; catalog #ab31940, Abcam); (4) rabbit anti-Ki67 (1:1000; catalog #ab15580, Abcam); (5) rabbit anti-SOX2 (1:1000; catalog #ab5603, Chemicon); or (6) rabbit anti-Cleaved Caspase-9 (1:500; catalog #PA5-17 913, Thermo Fisher Scientific). Sections were rinsed and probed with secondary antibodies against mouse or rabbit (1:500; Jackson ImmunoResearch). The brain sections were then counterstained with DAPI, rinsed, and mounted in 50% glycerol before visualization. Images were captured using an SP5 confocal microscope (Leica).

RNA in situ hybridization of brain sections.

DSCAM mRNA in situ hybridization (ISH) was conducted using previously published probe sequences (Fuerst et al., 2009) and from the Genepaint website. Probe sequences were amplified from DSCAM cDNA (catalog #18737, Addgene; Yamagata and Sanes, 2008) and cloned into a pTOPO vector. DIG-labeled cRNA probes were generated using a DIG RNA labeling kit (Roche), as previously described (Lin et al., 2017). Briefly, DIG-labeled cRNA probes were generated by in vitro transcription from the DSCAM cDNA template. ISH was performed on fresh-frozen mouse brain sections mounted on microscope slides, which were fixed, permeabilized by proteinase K, incubated with DIG-labeled cRNA probes, rinsed, and incubated with anti-DIG-AP (alkaline phosphatase) antibody (Roche). The signal was visualized by an AP substrate NBT (nitro-blue tetrazolium chloride)/BCIP (5-bromo-4-chloro-3′-indolyphosphate p-toluidine salt; Roche).

Cell brain slice adhesion test.

HEK293NC (no N-cadherin expressed) and TA (with N-cadherin expressed) cells (Yamagata and Sanes, 2018) were cultured in DMEM with 10% FBS in a humidified cell culture hood at 37°C and 5% CO2. N-cadherin (1.5 µg plasmid; catalog #18870, Addgene; Nechiporuk et al., 2007) or N-cadherin (1.5 µg plasmid)/DSCAM (0.5 µg plasmid) were transfected respectively with Lipofectamine (Thermo Fisher Scientific) into HEK293NC and TA cells (Yamagata and Sanes, 2018). Transfected HEK293 cells were incubated in a humidified cell culture hood at 37°C and 5% CO2 for 2 d. E19.5/postnatal day 0 (P0) brains were dissected and sectioned into 300 µm brain slices. To neutralize N-cadherin-mediated cell adhesion in HEK293NC cells expressing N-cadherin, 50 µl mouse anti-N-cadherin antibody (GC-4 clone; C3865, Sigma-Aldrich; RRID:AB_262097; Wallerand et al., 2010) was added to the isolated HEK293 cells for 10 min before they were added to cortical slices. The cortical slices were attached to the bottom of wells of a 24-well plate. Isolated HEK293 cells were incubated with the cortical slices for 2 h. After that, the brain slices were rinsed three times with 1× PBS to remove unattached cells and debris. Then, the brain slices and attached HEK293 cells were fixed with 4% PFA. Cell numbers were counted within each layer, the marginal zone (MZ), upper CP, and lower CP. The percentage of cells in each layer of the total cell number was calculated to represent the attaching ability introduced by N-cadherin.

To test the adhesion of primary neurons to the cortical plate, neurons were labeled with RFP expressed by PBCAG-mRFP (catalog #40996, Addgene) by IUE at E14.5. Then, at E19.5 the transfected cortices were dissected and triturated to dissociate the neurons. The neurons were incubated on wild-type cortical slices for 2 h. The remaining steps are the same as those for the HEK293 cortical slice adhesion assay.

Western blotting.

HEK293 cells were lysed in 2× sample buffer, sonicated, and boiled at 95°C for 5 min. Samples were then loaded onto 4–20% SDS-PAGE gels (catalog #4561093, BIO-RAD) for electrophoresis and transferred to nitrocellulose membrane (Novex). The membranes were blocked in 10% milk in 1× PBS (with 0.1% Triton X-100), then incubated overnight at 4°C with the following primary antibodies: (1) goat anti-DSCAM antibody (1:100; catalog #AF3666, R&D Systems); (2) rabbit anti-N-cadherin (1:2000; catalog #ab18203, Abcam); or (3) mouse anti-β-actin (1:10,000; catalog #AM4302, Thermo Fisher Scientific). After washes, the membranes were incubated with blocking buffer containing HRP-conjugated secondary antibodies (BIO-RAD) for 2 h at room temperature. The signal was developed in ChemiDoc MP imager and quantified using ImageJ software. The signal of N-cadherin bands, β-actin bands, and background were measured with ImageJ.

Statistical analysis.

Data were analyzed using GraphPad Prism 8. The types of statistical tests are shown in figure legends. All experiments were quantified in double-blind fashion so that the person who quantified the images did not know the genotypes or transfection plasmids.

Results

DSCAM is required for expanding upper cortical layers

To examine the contribution of DSCAM to cortical development, we compared the cortical histology of homozygous DSCAM2J/2J mice (referred to as DSCAM−/− here on) and their wild-type and heterozygous littermates. Previous studies have shown that DSCAM2j is a protein-null allele (Schramm et al., 2012; de Andrade et al., 2014). Fluorescence immunohistochemistry was performed on coronal brain sections from young adult mice at P21 using an antibody against NeuN, a pan-neuronal marker. The upper cortical layers (II–IV) were 21% thinner in DSCAM−/− cortices compared with wild-type or heterozygous cortex (Fig. 1A,B, top panels). Surprisingly, the deeper cortical layers (V–VI) were unaffected (Fig. 1A,B, bottom panels). These results suggest that DSCAM is required for expanding the thickness of upper cortical layers. Formation of the upper cortical layers begins after E18.5 through radial migration of pyramidal neurons (Molyneaux et al., 2007) and approaches completion at approximately P5 (Caviness, 1982; Supèr et al., 2000). Therefore, we determined whether DSCAM was required for proper cortical thickness during this developmental window. At E17.5, the CP thickness in DSCAM−/− cortex was comparable to wild-type or DSCAM+/− cortex (Fig. 1C,D), whereas at P5, DSCAM−/− upper cortex was 25% thinner than wild-type or heterozygous cortex (Fig. 1E,F). These findings were further confirmed by immunostaining with an antibody against Satb2, a marker for neurons in cortical layers II–IV (Britanova et al., 2008). The density of Satb2+ neurons in DSCAM−/− cortex was 26% higher than in wild-type cortex (Fig. 1G). However, the total number of Satb2+ neurons were similar between DSCAM−/− and wild-type cortices (Fig. 1H). Thus, examining a wider developmental window further confirms that DSCAM is required for expanding the upper cortical layers.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

DSCAM is required for expanding the developing upper cortical layers. A, Immunostaining of NeuN (green channel) in wild-type (WT) and DSCAM−/− cortex at P21 counterstained with DAPI (blue channel). B, Quantification of the thickness of cortical layers in WT, DSCAM+/−, and DSCAM−/− at P21. Top, Upper cortical layers. Bottom, Deeper cortical layers. One-way ANOVA followed by post hoc Student's t test. NS, Not significant. p > 0.05; **p < 0.01. C, E, Immunostaining of Satb2 (red channel) in WT and DSCAM−/− cortices counterstained with DAPI (blue) at E17.5 (C) and P5 (E). D, F, Quantification of the thickness of developing cortices in WT, DSCAM+/−, and DSCAM−/− at E17.5 (D) and P5 (F). One-way ANOVA followed by post hoc Student's t test. NSp > 0.05; ***p < 0.001. G, H, Quantification of Satb2+ cell densities (G) and total number (H) in upper cortical layers (II–IV) at P5. Mann–Whitney U test. NSp > 0.05; *p < 0.05. I, tdTomato+ pyramidal neurons (red) labeled by in utero electroporation at E14.5 locate in the upper cortical layers (labeled by DAPI staining, blue) at P5. J, Quantification of the percentage of tdTomato+ neurons in upper cortical layers in WT and DSCAM−/− at P5. Fisher's exact test. NSp > 0.05. K–N, Quantification of cell densities in upper cortical layers (II–IV), layer V, layer VI at P21, and upper cortical layers at P5. One-way ANOVA followed by post hoc Student's t test. NSp > 0.05; **p < 0.01; ***p < 0.0001. Scale bar, 50 µm.

DSCAM is required for neuronal dispersion in developing upper cortical layers

Defective radial migration distributes neurons to the incorrect layer, which could cause a thinner cortex (Rakic, 1988; Dobyns and Truwit, 1995). The thinner cortical layers that we observed in DSCAM−/− mice could similarly arise from defective radial migration that mistakenly redirects neurons destined to the upper cortical layers to the deep cortical layers. To test this possibility, we used IUE in E14.5 embryos to introduce a tdTomato expression vector into dorsal NPCs, which give rise to pyramidal neurons (Ayala et al., 2007). The progeny neurons of these NPCs radially migrate and differentiate into pyramidal neurons in the upper cortical layers during E17.5 to P5 (Molyneaux et al., 2007). We examined the cortices at P5, after radial migration had occurred, and only detected tdTomato neurons in the upper cortical layers in both wild-type and DSCAM−/− cortex (Fig. 1I,J). This result demonstrates that lack of DSCAM does not prevent pyramidal neurons from migrating to the upper cortical layers through radial migration.

Alternatively, a thinner neocortex may also arise from the birth of fewer neurons. To assess whether this occurs in DSCAM−/− cortex, we quantified the neuronal density from immunoreactivity to NeuN in the upper cortical layers at P5 and P21. The density of neurons in layers II–IV was >30% higher in DSCAM−/− cortex compared with wild-type and heterozygous cortex (Fig. 1K,N; also see Fig. 1G). On the other hand, the neuronal density in deeper layers in DSCAM−/− cortex was comparable to wild-type and heterozygotes cortex (Fig. 1L,M). The number of neural progenitors in the ventricular zone did not differ between DSCAM−/− and wild-type at E17.5, as suggested by immunostaining with the neural progenitor cell marker SOX2 and cell proliferation marker Ki67 (Graham et al., 2003; Tsai et al., 2005; Fig. 2A–D). Moreover, immunostaining with the marker for active caspase signaling suggests that programmed cell death was not altered at E19.5/P0 in the DSCAM−/− cortex (Zou et al., 1999; Fig. 2E,F). These results demonstrate that the thinner upper cortical layers II–IV in DSCAM−/− cortex does not result from fewer neurons. Rather, a similar number of pyramidal neurons are generated in DSCAM−/− versus wild-type cortex, but these DSCAM−/− neurons fail to disperse, leading to a thinner but denser upper cortical layer.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Loss of DSCAM does not affect the number of neural progenitors and that of cells undergoing programmed cell death in the neocortex. A, B, Immunostaining of the proliferation marker Ki67 (A) and the marker Sox2 of the neural progenitor cell (B, red channel) in wild-type (WT) and DSCAM−/− cortices at E17.5. The cortical slices were counterstained with DAPI (blue channel). Scale bar, 50 µm. C, D, Quantification of the total number of neural progenitors in the ventricular zone in WT and DSCAM−/− at E17.5. Student's t test. NSp > 0.05. E, Immunostaining of cleaved Caspase-9 (red channel)—which indicates activated Caspase-9—in WT and DSCAM−/− cortical slices counterstained with DAPI (blue) at E19.5. Upper cortical neurons were labeled by transfecting an EGFP-expressing plasmid (pCAGGS-IRES-EGFP) via IUE at E14.5. The programmed cell death in the positive controls was caused by MUNC18-1 knockout (Verhage et al., 2000). F, Quantification of the cells positive for cleaved Caspase-9 in upper cortical layers of WT, DSCAM−/−, and the positive control in 100-µm-wide cerebral cortices at E19.5. Shown are the percentage of GFP+ neurons that were also positive for cleaved Caspase-9 in the total number of GFP+ neurons. One-way ANOVA followed by post hoc Mann–Whitney U test. NSp > 0.05, ***p < 0.0001.

DSCAM is expressed in radially migrating neurons targeting the upper cortical layers

Since DSCAM is required for neuronal dispersion to the upper cortical layer, we sought to determine whether DSCAM was expressed in neurons that were destined to cortical layers II–IV. Since there are no reliable antibodies against mouse DSCAM for immunohistochemistry, we used RNA ISH to investigate DSCAM mRNA expression in the developing cortex. The upper cortical layer begins to develop at approximately E18.5; therefore, we performed RNA ISH on wild-type cortex at earlier time points, E14.5 and E16.5, and at a later time point, E19.5/P0. We found that DSCAM mRNA levels were low in the CP at E14.5 and E16.5 (Fig. 3A) but increased significantly in the upper CP at E19.5/P0 (Fig. 3A). These results indicate that increased DSCAM expression coincides with the formation of the upper cortical layers.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

DSCAM is preferentially expressed in radial migrating neurons in the upper cortical plate. A, DSCAM mRNA in situ hybridization in developing WT cortices at E14.5 (left), E16.5 (middle), and E19.5 (right). IZ, Intermediate zone; LCP, lower cortical plate; UCP, upper cortical plate; MZ, marginal zone. Scale bar, 100 µm. B, DSCAM mRNA is expressed in all tdTomato+ neurons, which are destined to the upper cortical layers in WT cortices at E19.5. Scale bar, 50 µm. B′, Enlarged image of the boxed area in B; green arrowheads point to individual neurons. Left, DSCAM mRNA ISH. Middle, tdTomato. Right, Merge.

A salient difference between the later (E19.5/P0; high DSCAM expression) and earlier (E14.5 and E16.5; low DSCAM expression) time points that we examined is that nascent pyramidal neurons of upper cortical layers will have radially migrated and accumulated in the upper CP by the E19.5/P0 but not E14.5 and E 16.5 time points, since the upper cortical layer forms at approximately E18 onward. Therefore, we further examined whether DSCAM mRNA was expressed in these nascent neurons. We achieved this by genetically labeling NPCs at E14.5, which gives rise to the pyramidal neurons of the upper cortical layers, with tdTomato, and performing DSCAM mRNA ISH. We observed that DSCAM mRNA was expressed in all tdTomato+ neurons at E19.5/P0 (Fig. 3B,B′), which suggests that DSCAM is expressed in those nascent pyramidal neurons destined to the upper cortical layers.

DSCAM is required for newly incoming neurons to bypass their postmigratory predecessors at the CP border

Since DSCAM is expressed by radially migrating neurons but its deficiency does not cause defective radial migration, we sought to determine whether DSCAM was instead involved in the termination of migration. We used time-lapse live imaging to investigate this possibility. We fluorescently labeled radially migrating neurons in wild-type and DSCAM−/− cortex by transfecting NPCs with a tdTomato expression vector by IUE at E14.5. In mice, neurons born at E14.5 establish the cortical layer IV at E19/P0 (Kwan et al., 2012); therefore, we performed time-lapse imaging around the CP border at E19.5/P0.

In wild-type cortex, neurons migrated across the CP with a bipolar morphology and a leading process oriented radially (Fig. 4A, 1:00–4:00 h, A′, 1:00–8:00 h). After crossing the original CP boundary, which is delineated by the lower cell density in MZ, the neurons ceased migrating radially. At this stage, neurons either continued migrating nonradially a short distance to their final stopping position (Fig. 4A) or immediately halted without further migration (Fig. 4A′). Once terminated, the neurons sent out a new prominent process to replace the leading process used during radial migration (Fig. 4A, 3:00–7:00 h), and then extended and retracted several highly dynamic processes (Figs. 4A, 7:00–15:00 h, A′, 9:00–15:00 h, B, 10:00–15:00 h, and B′, 5:00–9:00 h, 5). Tracking several neurons in wild-type cortices revealed that most neurons stopped their radial migration after crossing the original CP border. The neurons transitioned from a bipolar to multipolar shape and terminated at 36.8 ± 22.3 µm above the original CP border (Fig. 4D,E).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

DSCAM is required for migrating neurons to bypass the CP border. A, A′, Cellular behaviors at the termination phase of radially migrating neurons. Representative time-lapse images of tdTomato+ neurons in WT cortices at E19.5. The images show several stages of migration. Neurons with a bipolar morphology and a thick leading process migrate radially to the CP border. They then extend multiple dynamic processes and, as migration terminates, retract the thick leading process. A, A representative neuron migrates laterally after radial migration. A′, A representative neuron stops immediately after radial migration. One hour per frame. The temporary CP border at the beginning of time-lapse imaging represented by a green horizontal line. Green arrowheads point to the soma of migrating neurons. Scale bar, 50 µm. B, B′, Representative time-lapse images of tdTomato+ neurons in DSCAM−/− cortices at E19.5. B, A representative neuron migrates laterally after radial migration. One hour per frame. B′, A representative neuron stops immediately after radial migration; 0.5 h/frame. C, D, The wild-type, but not DSCAM−/−, cortices (P0) expanded during the course of the time-lapse imaging. C, Representative images. Left panels, The start point of the time lapse (E19.5/P0). Right panels, The same cortical regions 15 h later. The CP expanded in WT (top) but not in DSCAM−/− (bottom). The original cortical border is indicated by a green line, and the new CP border 15 h later is indicated by a magenta line. Scale bar, 100 µm. D, Quantification of the final positions of the migrating neurons. “0” is the CP border at the beginning of the time-lapse imaging. Student's t test, ***p < 0.0001. E, Schematic of the termination stage of neuronal migration.

By contrast, in DSCAM−/− cortices, radially migrating neurons had a bipolar morphology and came to a halt within the CP (Fig. 4B, 1:00–5:00 h, B′, 1:00–5:00 h). After radial migration, the neurons in DSCAM−/− cortices might also start a nonradial migration (Fig. 4B, 6:00–9:00 h) while they transitioned from a bipolar to a multipolar shape and came to a full rest at 3.5 ± 14.8 µm below the original CP border, almost at the original CP–MZ interface (Fig. 4B, 10:00–15:00 h, B′, 5:30–9:00 h, D, E). During the course of the time-lapse imaging, the wild-type cortices expanded significantly, while the DSCAM−/− cortices did not (Fig. 4C). Loss of DSCAM did not significantly alter the morphology of neurons that were terminating their radial migration (Fig. 5). These observations suggest that the positioning of nascent pyramidal neurons relative to the original CP border directly impacts the expansion of the cerebral cortex.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

The morphology of neurons during the termination of radial migration. A, B, Time-lapse imaging shows the morphology of neurons that are terminating their radial migration in wild-type (A) and DSCAM−/− (B) cortices at E19.5/P0. The neuron morphology was traced in black. C, D, Quantifications of the percentage of pyramidal neurons extending two or more highly dynamic neurites (C) and the average number of highly dynamic neurites (D) during the termination of radial migration in wild-type and DSCAM−/− cortices at E19.5/P0. Student's t test. NSp > 0.05.

The developing CP border constantly moves outward during the formation of the upper cortical layer, rendering it imprecise to estimate the relative position of the expanding CP border to individually migrating neurons. Therefore, we applied a more precise method of evaluating CP outward expansion at the single-neuron resolution. This technique takes advantage of neuron pairs that migrate along the same radial axis, which enables precise measurement of the relative positions of the trailing migrating neurons to the leading migrating neurons. We fluorescently labeled pyramidal neurons using a tamoxifen-induced Nestin-Cre to drive tdTomato expression in NPCs in DSCAM+/− and DSCAM−/− mice. tdTomato expression was induced by a tamoxifen injection at E14.5, labeling all the nascent neurons born after E14.5, which were imaged by time-lapse microscopy.

We evaluated CP outward expansion using this system at single-neuron resolution by monitoring neuron pairs migrating along the same radial path. Under this experimental paradigm, the termination position of the leading neuron defines the outermost position of the expanding CP. The position of the trailing neuron was then measured relative to the termination position of the leading neuron to track the progress of the trailing neuron to its destination. This setup allowed us to determine at single-neuron resolution whether the trailing neuron terminates at a position before (i.e., still within the CP) or past the leading neuron (i.e., reaches the new CP to expand the upper cortical layers).

In DSCAM+/− cortex, 67% of trailing neurons bypassed their paired leading neurons to occupy their final position beyond the original CP border (Fig. 6A,A′,C,E). In contrast, in DSCAM−/− cortex, only 11% of trailing neurons bypassed their paired leading predecessors, which instead terminated their migration adjacent to their paired leading neurons or deeper within the cortical layer (Fig. 6B,D,E). Furthermore, trailing neurons in DSCAM+/− cortex terminated migration at a position further past their paired leading neurons (14.7 ± 13.6 µm) compared with trailing neurons in DSCAM−/− cortex (−1.5 ± 7.1 µm; Fig. 6F). These results suggest that DSCAM is required by migrating pyramidal neurons to bypass their postmigratory predecessors to expand upper cortical layers.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

DSCAM is required for the trailing neurons to bypass their postmigratory predecessor neurons at the CP border. A–B, Representative time-lapse imaging of a pair of tdTomato+ migrating neurons (1 h/frame) in DSCAM+/− (A, A′) and DSCAM−/− (B) cortices in which the trailing neuron (green dots) bypasses the leading neuron (magenta dots) and attains the CP border. The green and magenta dots label the cell body positions and show the relative positions of leading and trailing neurons during the termination phase of neuronal migration. C, D, Schematics of the relative position of individual neurons of in the migrating pairs. E, Quantification of neuron pairs in which the trailing neuron bypasses the leading neuron in DSCAM+/− (n = 3 animals) and DSCAM−/− (n = 4 animals) cortex. Mann–Whitney U test, **p < 0.01. F, Quantification of the distance between the trailing and leading neurons in DSCAM+/− (n = 30 neurons pairs) and DSCAM−/− (n = 32 neuron pairs). Student's t test, ***p < 0.0001.

DSCAM decreases N-cadherin-mediated adhesion in developing upper CP

How might DSCAM, as a homophilic cell adhesion molecule, facilitate the migration of pyramidal neurons to bypass their postmigratory predecessors? A previous study showed that in the mouse retina DSCAM masks cadherin-mediated cell adhesion (Garrett et al., 2018). We thus tested the possibility that DSCAM may mask or weaken adhesion from other cell adhesion molecules and therefore decrease the total cell adhesion strength in the upper CP. In this manner, DSCAM may facilitate the entry of migrating neurons into the MZ. N-cadherin is expressed throughout the CP and MZ and plays key roles in radial migration (Lai et al., 2015). To investigate the function of DSCAM in cell adhesion in the CP, we developed a HEK293T cortical slice adhesion assay. HEK293T cells express a low level of N-cadherin (Yamagata et al., 2018), which is not sufficient for mediating robust cell adhesion to brain slices (Fig. 7A,C,D). Nevertheless, to completely avoid a potential complication from the low level of endogenous N-cadherin in HEK293T cells, we used the HEK293NC cell line with N-cadherin knocked out, which was developed from a cloned HEK293T cell line called HEK293TA (Yamagata et al., 2018; Fig. 7A). Overexpressing N-cadherin in HEK293NC cells significantly increased their attachment to cortical slices (Fig. 7C,D). Furthermore, the adhesion of HEK293NC cells overexpressing N-cadherin to cortical slices was blocked when the cells were preincubated with a monoclonal anti-N-cadherin antibody (clone GC-4), which is known to neutralize N-cadherin function (Wallerand et al., 2010). These results suggest that the N-cadherin in HEK293NC cells and cortical slice specifically mediates the cell–cortical plate attachment.

Figure 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 7.

DSCAM decreases N-cadherin-mediated adhesion in developing upper CP. A, Expression of N-cadherin in HEK293TA and NC cells. B, Levels of N-cadherin in HEK293TA and NC cells with or without N-cadherin-GFP overexpression. C, D, While neither HEK293TA nor NC cells attach to cortical slices efficiently, overexpressing N-cadherin significantly enhances the attachment of HEK293NC to cortical slices. The attachment depends on N-cadherin because it is diminished by incubation of the HEK293 cells with an anti-N-cadherin antibody (C, right panel). Quantification of cells attached to E19.5 cortical slices (width, 1.5 mm) is show in D. One-way ANOVA followed by post hoc Mann–Whitney U test: NSp > 0.05; *p < 0.05. Scale bar, 100 µm. E, The distributions of HEK293NC cells overexpressing N-cadherin (N-Cad OE; red) on wild-type cortical slices (blue). Left, without DSCAM coexpression; right, with DSCAM coexpression. IZ, Intermediate zone; LCP, lower cortical plate; UCP, upper cortical plate; MZ, marginal zone. F, Western blots (left) and quantification (right) showing that overexpressing (OE) DSCAM does not affect the level of cotransfected N-cadherin. β-Actin is used as an internal control. The y-axis shows the ratio of N-cadherin signals to β-actin signals after background subtraction of both. Mann–Whitney U test. NSp > 0.05. G–I, Quantification of the percentage of HEK293NC cells adhered to the UCP (G), the LCP (H), and the MZ (I). Mann–Whitney U test: NSp > 0.05; **p < 0.01. J, The distributions of primary neurons (red) on wild-type cortical slices (blue). Left, The distribution of wild-type primary neurons, which expresses DSCAM. Right, The distribution of primary neurons from DSCAM−/− mice. K–M, Quantification of the percentage of primary neurons adhered to the UCP (K), LCP (L), and the MZ (M). Mann–Whitney U test: NSp > 0.05; ***p < 0.001.

With this test, we examined the adhesion of N-cadherin-expressing HEK293NC cells, with or without DSCAM coexpression, to cortical slices (Fig. 7E,G–I). HEK293NC cells expressed comparable levels of N-cadherin with or without DSCAM coexpression (Fig. 7F). After incubating these HEK293NC cells on wild-type brain slices, we quantified the percentages of cells attached to the MZ, upper CP, and lower CP. We found a dramatic decrease in the percentage of HEK293NC with N-cadherin and DSCAM coexpression in the upper CP (Figs. 7E,G), whereas the percentage of HEK293NC cells in the lower CP and MZ were similar with or without DSCAM coexpression (Fig. 7H,I). These results suggest that DSCAM weakens N-cadherin-mediated cell adhesion in the developing upper CP.

We further tested this notion by replacing HEK293NC cells with pyramidal neurons dissociated from developing cortical plates. Pyramidal neurons destined to the upper cortical layers were labeled with red fluorescent protein by IUE in either DSCAM−/− embryos or their wild-type littermates at E14.5. Then, at E19.5, dissociated neurons from transfected cortical plates were incubated with wild-type cortical slices for 2 h before fixation. DSCAM−/− neurons attached to the upper CP significantly more than wild-type neurons (Fig. 7J,K). By contrast, there was no difference between DSCAM−/− and wild-type neurons in their attachment to the lower CP and MZ (Fig. 7L,M). Overall, these results suggest that DSCAM weakens the attachment of migrating pyramidal neurons to the upper CP.

Discussion

The termination of neuronal radial migration is critical for cortex formation (Ohtaka-Maruyama and Okado, 2015). In this study, we found that DSCAM is involved in the termination of neuronal migration in upper cortical layers. DSCAM allows incoming nascent pyramidal neurons to bypass their predecessors at the CP border to expand the developing cerebral cortex. Therefore, the loss of DSCAM reduces the thickness of the upper cortical layers while increasing their neuronal density. Consistent with the selective effect on upper cortical layers in Dscam−/− mutants, Dscam mRNA expression is preferentially elevated in the upper CP during upper cortical layer formation. We used the time-lapse imaging technique to demonstrate that DSCAM is required for the termination of neuronal radial migration and the determination of their final position at the CP border of the developing cortex. Through a cell adhesion assay on cortical slices, we found that DSCAM masks or weakens N-cadherin-mediated cell adhesion in the upper CP, therefore decreasing the affinity between neurons. These findings suggest that DSCAM allows the migrating neurons to bypass the CP border by masking the high-affinity cell adhesion of N-cadherin.

Radial migration of nascent pyramidal neurons in the developing cortex involves several steps. The process begins with nascent pyramidal neurons arising from NPCs in the subventricular zones, transitioning from a multipolar to a bipolar morphology before initiating the migration along radial glia. Migrating pyramidal neurons cross the SVZ, intermediate zone, and CP to terminate at the front of the developing CP, where they lose their leading process as they come to their final position (Gongidi et al., 2004; Sanada et al., 2004; Ohtaka-Maruyama and Okado, 2015). Of all these steps, migration termination is the least understood process (Rakic, 1988; Rakic and Caviness, 1995; Gongidi et al., 2004; Ohtaka-Maruyama and Okado, 2015), despite its critical importance in cortical layer formation. The cellular and molecular underpinning of neuronal migration termination remain unknown.

We noted upper cortical layer thickness defects in the DSCAM−/− mice. To investigate more closely, we tagged migrating pyramidal neurons with tdTomato by IUE and used ex vivo time-lapse imaging to monitor migration in real time. Through a quantitative analysis that we developed (Fig. 4), we found that the ability of migrating pyramidal neurons to overtake their postmigratory predecessors was impaired in DSCAM−/− cortex. Instead, we found that DSCAM-deficient migrating neurons abruptly halted just at or beneath the CP border. We refined our analysis by examining neuron pairs (Fig. 6) to track relative positions more accurately. This experiment confirmed that migrating DSCAM−/− neurons are defective in their ability to overtake the postmigratory predecessors. Importantly, our time-lapse imaging approach resolved the question of how new incoming pyramidal neurons bypass their postmigratory predecessors. Although it has been previously assumed that new incoming pyramidal neurons bypass postmigratory neurons to traverse the developing CP border, this study provides evidence demonstrating the role of DSCAM in this process.

Several of our observations suggest that DSCAM is required for the termination phase of radial migration but not earlier phases. First, the loss of Dscam reduces the thickness of upper cortical layers (layers II/III/IV) without affecting deeper cortical layer thickness (layers V/VI). Importantly, the reduction in thickness coincides with an increase in neuron density in the same layers, suggesting a defect in neuron positioning on completing migration. Second, defects in thickness occur during upper cortical layer formation, when neurons terminate migration. Third, DSCAM expression increases at E19.5/P0, when upper cortical histogenesis begins, and especially in the nascent migrating neurons destined to form the upper cortical layer over the existing CP. On the other hand, radial migration itself was not compromised in DSCAM−/− neurons, leaving a termination defect the likeliest explanation. This possibility motivated us to study the defect in migratory termination by ex vivo time-lapse imaging, which showed significantly fewer new incoming nascent neurons bypassing their predecessors to expand the CP in DSCAM−/− mutant.

Previous studies have shown that extracellular matrix components, such as α3 integrins, guide nascent pyramidal neurons as they migrate along radial glia across the entire developing cortex to the CP–MZ interface. Reelin from Cajal-Retzius cells inhibits α3 integrin expression to facilitate the detachment of migrating pyramidal neurons from radial glial cells (Sanada et al., 2004). Plexin A2/A4 (PlxnA2/A4) or Semaphorin 6A (Sema6A) has been proposed to play a similar role as Reelin signaling in the detachment of migrating neurons from radial glia. Sema6A and PlxnA2/A4 are hypothesized to mediate a repulsive interaction between migrating neurons and radial glial cells (Hatanaka et al., 2019). Loss of PlxnA2/A4 or Sema6A leads to a phenotype that resembles the Cobblestone lissencephaly (Olson and Walsh, 2002; Hatanaka et al., 2019). We found that the loss of DSCAM causes migrating neurons to fail to traverse the CP border, suggesting that Reelin–Dab1 and PlxnA2/A4–Sema6A signaling probably function before neurons reach the CP border. However, it is also possible that DSCAM functionally interacts with these signaling pathways. Clarifying these is an interesting future research direction.

DSCAM is a cell adhesion molecule primarily involved in homophilic cell adhesion (Yamakawa et al., 1998; Agarwala et al., 2000). We found that DSCAM is highly expressed in the developing upper CP and, surprisingly, is required by nascent pyramidal neurons to bypass the preceding neurons. This is in apparent conflict with the homophilic cell adhesion function of DSCAM. However, in mammals, DSCAM may function in concert with protocadherins (Zipursky and Sanes, 2010; Lefebvre et al., 2012) or cadherins (Garrett et al., 2018). In the mouse retina, DSCAM−/− mutants exhibit neurite fasciculation mediated by classical cadherins (Garrett et al., 2018), suggesting a function in masking cadherin-mediated cell adhesion. DSCAM prevents fasciculation in a homotypic fashion, being present on both neurons. Here, we found that coexpressing DSCAM with N-cadherin rendered HEK293NC cells less adhesive to the upper CP of brain slices, but not the lower CP or MZ regions. These results suggest that DSCAM masks N-cadherin, specifically in the upper CP, allowing migrating neurons to enter the MZ. Therefore, our findings support the notion that DSCAM modulates adhesion strength by blocking strong cell adhesion mediated by cadherins (Garrett et al., 2018).

Previous studies have shown that the Drosophila homolog of DSCAM, Dscam1, mediates neurite avoidance. Whether DSCAM plays a similar role in migrating neurons is unknown. In the live-imaging experiments, we found that the trailing neuron bypasses the leading neuron right along each other—in some cases even contacting each other (Fig. 6A′, arrowheads). This finding indicates the absence of repulsion between the migrating neurons. Moreover, although DSCAM/Dscam1 regulates the dendrite and axon morphology of neurons, it does not seem to be required for the proper morphology of migrating neurons (Fig. 5).

In summary, we report that DSCAM is required for migrating neurons to bypass their postmigratory predecessors during the expansion of the upper cortical layers. This study provides insights into the proper termination of neuronal migration at the expanding border of the cortical plate. The knowledge might also help us to understand the brain disorders, which exhibit thinner cortical layers of the cerebral cortex without neuronal loss, as well as Down syndrome and autism spectrum disorders, to which DSCAM has been linked.

Footnotes

  • This study was supported by National Institutes of Health Grants R01-EB-028159 and R21-NS-094091; a Seed Grant from the Brain Research Foundation; the Protein Folding Disease Initiative of the University of Michigan to B.Y.; and a University of Michigan Mcubed pilot grant to B.Y., Y.W., and K.Y.K. The content of this work is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. We thank Dr. Andrew Garrett for critical feedback on the work, Dr. Joshua Sanes for sharing HEK293NC and TA cell lines, and Drs. Robert Reiter and Joyce Yamshiro for sharing anti-N-cadherin antibodies and related information.

  • The authors declare no competing financial interests.

  • Correspondence should be addressed to Bing Ye at bingye{at}umich.edu

SfN exclusive license.

References

  1. ↵
    1. Agarwala KL,
    2. Nakamura S,
    3. Tsutsumi Y,
    4. Yamakawa K
    (2000) Down syndrome cell adhesion molecule DSCAM mediates homophilic intercellular adhesion. Brain Res Mol Brain Res 79:118–126. doi:10.1016/s0169-328x(00)00108-x pmid:10925149
    OpenUrlCrossRefPubMed
  2. ↵
    1. Anton ES,
    2. Cameron RS,
    3. Rakic P
    (1996) Role of neuron-glial junctional domain proteins in the maintenance and termination of neuronal migration across the embryonic cerebral wall. J Neurosci 16:2283–2293. doi:10.1523/JNEUROSCI.16-07-02283.1996 pmid:8601808
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. Arimura N,
    2. Okada M,
    3. Taya S,
    4. Dewa KI,
    5. Tsuzuki A,
    6. Uetake H,
    7. Miyashita S,
    8. Hashizume K,
    9. Shimaoka K,
    10. Egusa S,
    11. Nishioka T,
    12. Yanagawa Y,
    13. Yamakawa K,
    14. Inoue YU,
    15. Inoue T,
    16. Kaibuchi K,
    17. Hoshino M
    (2020) DSCAM regulates delamination of neurons in the developing midbrain. Sci Adv 6:eaba1693.
    OpenUrlFREE Full Text
  4. ↵
    1. Arnold SE,
    2. Trojanowski JQ
    (1996) Recent advances in defining the neuropathology of schizophrenia. Acta Neuropathol 92:217–231. doi:10.1007/s004010050512 pmid:8870823
    OpenUrlCrossRefPubMed
  5. ↵
    1. Ayala R,
    2. Shu T,
    3. Tsai LH
    (2007) Trekking across the brain: the journey of neuronal migration. Cell 128:29–43. doi:10.1016/j.cell.2006.12.021 pmid:17218253
    OpenUrlCrossRefPubMed
  6. ↵
    1. Britanova O,
    2. de Juan Romero C,
    3. Cheung A,
    4. Kwan KY,
    5. Schwark M,
    6. Gyorgy A,
    7. Vogel T,
    8. Akopov S,
    9. Mitkovski M,
    10. Agoston D,
    11. Sestan N,
    12. Molnár Z,
    13. Tarabykin V
    (2008) Satb2 is a postmitotic determinant for upper-layer neuron specification in the neocortex. Neuron 57:378–392. doi:10.1016/j.neuron.2007.12.028 pmid:18255031
    OpenUrlCrossRefPubMed
  7. ↵
    1. Caviness VS Jr.
    (1982) Neocortical histogenesis in normal and reeler mice: a developmental study based upon [3H]thymidine autoradiography. Brain Res 4:293–302. doi:10.1016/0165-3806(82)90141-9
    OpenUrlCrossRef
  8. ↵
    1. D'Arcangelo G
    (2006) Reelin mouse mutants as models of cortical development disorders. Epilepsy Behav 8:81–90. pmid:16266828
    OpenUrlCrossRefPubMed
  9. ↵
    1. de Andrade GB,
    2. Long SS,
    3. Fleming H,
    4. Li W,
    5. Fuerst PG
    (2014) DSCAM localization and function at the mouse cone synapse. J Comp Neurol 522:2609–2633. doi:10.1002/cne.23552 pmid:24477985
    OpenUrlCrossRefPubMed
  10. ↵
    1. Dobyns WB,
    2. Truwit CL
    (1995) Lissencephaly and other malformations of cortical development: 1995 update. Neuropediatrics 26:132–147. doi:10.1055/s-2007-979744 pmid:7477752
    OpenUrlCrossRefPubMed
  11. ↵
    1. Fuerst PG,
    2. Koizumi A,
    3. Masland RH,
    4. Burgess RW
    (2008) Neurite arborization and mosaic spacing in the mouse retina require DSCAM. Nature 451:470–U478. doi:10.1038/nature06514 pmid:18216855
    OpenUrlCrossRefPubMed
  12. ↵
    1. Fuerst PG,
    2. Bruce F,
    3. Tian M,
    4. Wei W,
    5. Elstrott J,
    6. Feller MB,
    7. Erskine L,
    8. Singer JH,
    9. Burgess RW
    (2009) DSCAM and DSCAML1 function in self-avoidance in multiple cell types in the developing mouse retina. Neuron 64:484–497. doi:10.1016/j.neuron.2009.09.027 pmid:19945391
    OpenUrlCrossRefPubMed
  13. ↵
    1. Fuerst PG,
    2. Harris BS,
    3. Johnson KR,
    4. Burgess RW
    (2010) A novel null allele of mouse DSCAM survives to adulthood on an inbred C3H background with reduced phenotypic variability. Genesis 48:578–584. doi:10.1002/dvg.20662 pmid:20715164
    OpenUrlCrossRefPubMed
  14. ↵
    1. Garrett AM,
    2. Khalil A,
    3. Walton DO,
    4. Burgess RW
    (2018) DSCAM promotes self-avoidance in the developing mouse retina by masking the functions of cadherin superfamily members. Proc Natl Acad Sci U|S|A 115:E10216–E10224. doi:10.1073/pnas.1809430115 pmid:30297418
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Gongidi V,
    2. Ring C,
    3. Moody M,
    4. Brekken R,
    5. Sage EH,
    6. Rakic P,
    7. Anton ES
    (2004) SPARC-like 1 regulates the terminal phase of radial glia-guided migration in the cerebral cortex. Neuron 41:57–69. doi:10.1016/s0896-6273(03)00818-3 pmid:14715135
    OpenUrlCrossRefPubMed
  16. ↵
    1. Graham V,
    2. Khudyakov J,
    3. Ellis P,
    4. Pevny L
    (2003) SOX2 functions to maintain neural progenitor identity. Neuron 39:749–765. doi:10.1016/s0896-6273(03)00497-5 pmid:12948443
    OpenUrlCrossRefPubMed
  17. ↵
    1. Hatanaka Y,
    2. Kawasaki T,
    3. Abe T,
    4. Shioi G,
    5. Kohno T,
    6. Hattori M,
    7. Sakakibara A,
    8. Kawaguchi Y,
    9. Hirata T
    (2019) Semaphorin 6A-plexin A2/A4 interactions with radial glia regulate migration termination of superficial layer cortical neurons. iScience 21:359–374. doi:10.1016/j.isci.2019.10.034 pmid:31698249
    OpenUrlCrossRefPubMed
  18. ↵
    1. Hattori D,
    2. Chen Y,
    3. Matthews BJ,
    4. Salwinski L,
    5. Sabatti C,
    6. Grueber WB,
    7. Zipursky SL
    (2009) Robust discrimination between self and non-self neurites requires thousands of Dscam1 isoforms. Nature 461:644–648. doi:10.1038/nature08431 pmid:19794492
    OpenUrlCrossRefPubMed
  19. ↵
    1. Hughes ME,
    2. Bortnick R,
    3. Tsubouchi A,
    4. Bäumer P,
    5. Kondo M,
    6. Uemura T,
    7. Schmucker D
    (2007) Homophilic Dscam interactions control complex dendrite morphogenesis. Neuron 54:417–427. doi:10.1016/j.neuron.2007.04.013 pmid:17481395
    OpenUrlCrossRefPubMed
  20. ↵
    1. Kim JH,
    2. Wang X,
    3. Coolon R,
    4. Ye B
    (2013) Dscam expression levels determine presynaptic arbor sizes in Drosophila sensory neurons. Neuron 78:827–838. doi:10.1016/j.neuron.2013.05.020 pmid:23764288
    OpenUrlCrossRefPubMed
  21. ↵
    1. Kwan KY,
    2. Sestan N,
    3. Anton ES
    (2012) Transcriptional co-regulation of neuronal migration and laminar identity in the neocortex. Development 139:1535–1546. doi:10.1242/dev.069963 pmid:22492350
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Lagace DC,
    2. Whitman MC,
    3. Noonan MA,
    4. Ables JL,
    5. DeCarolis NA,
    6. Arguello AA,
    7. Donovan MH,
    8. Fischer SJ,
    9. Farnbauch LA,
    10. Beech RD,
    11. DiLeone RJ,
    12. Greer CA,
    13. Mandyam CD,
    14. Eisch AJ
    (2007) Dynamic contribution of nestin-expressing stem cells to adult neurogenesis. J Neurosci 27:12623–12629. doi:10.1523/JNEUROSCI.3812-07.2007 pmid:18003841
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Lai M,
    2. Guo Y,
    3. Ma J,
    4. Yu H,
    5. Zhao D,
    6. Fan W,
    7. Ju X,
    8. Sheikh MA,
    9. Malik YS,
    10. Xiong W,
    11. Guo W,
    12. Zhu X
    (2015) Myosin X regulates neuronal radial migration through interacting with N-cadherin. Front Cell Neurosci 9:326. doi:10.3389/fncel.2015.00326 pmid:26347613
    OpenUrlCrossRefPubMed
  24. ↵
    1. Lefebvre JL,
    2. Kostadinov D,
    3. Chen WV,
    4. Maniatis T,
    5. Sanes JR
    (2012) Protocadherins mediate dendritic self-avoidance in the mammalian nervous system. Nature 488:517–521. doi:10.1038/nature11305 pmid:22842903
    OpenUrlCrossRefPubMed
  25. ↵
    1. Lin JP,
    2. Mironova YA,
    3. Shrager P,
    4. Giger RJ
    (2017) LRP1 regulates peroxisome biogenesis and cholesterol homeostasis in oligodendrocytes and is required for proper CNS myelin development and repair. eLife 6:e30498. doi:10.7554/eLife.30498
    OpenUrlCrossRef
  26. ↵
    1. Madisen L,
    2. Zwingman TA,
    3. Sunkin SM,
    4. Oh SW,
    5. Zariwala HA,
    6. Gu H,
    7. Ng LL,
    8. Palmiter RD,
    9. Hawrylycz MJ,
    10. Jones AR,
    11. Lein ES,
    12. Zeng H
    (2010) A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat Neurosci 13:133–140. doi:10.1038/nn.2467 pmid:20023653
    OpenUrlCrossRefPubMed
  27. ↵
    1. Matthews BJ,
    2. Kim ME,
    3. Flanagan JJ,
    4. Hattori D,
    5. Clemens JC,
    6. Zipursky SL,
    7. Grueber WB
    (2007) Dendrite self-avoidance is controlled by Dscam. Cell 129:593–604. doi:10.1016/j.cell.2007.04.013 pmid:17482551
    OpenUrlCrossRefPubMed
  28. ↵
    1. Molyneaux BJ,
    2. Arlotta P,
    3. Menezes JR,
    4. Macklis JD
    (2007) Neuronal subtype specification in the cerebral cortex. Nat Rev Neurosci 8:427–437. doi:10.1038/nrn2151 pmid:17514196
    OpenUrlCrossRefPubMed
  29. ↵
    1. Narita A, et al
    . (2020) Clustering by phenotype and genome-wide association study in autism. Transl Psychiatry 10:290. doi:10.1038/s41398-020-00951-x pmid:32807774
    OpenUrlCrossRefPubMed
  30. ↵
    1. Nechiporuk T,
    2. Fernandez TE,
    3. Vasioukhin V
    (2007) Failure of epithelial tube maintenance causes hydrocephalus and renal cysts in Dlg5-/- mice. Dev Cell 13:338–350. doi:10.1016/j.devcel.2007.07.017 pmid:17765678
    OpenUrlCrossRefPubMed
  31. ↵
    1. Ohtaka-Maruyama C,
    2. Okado H
    (2015) Molecular pathways underlying projection neuron production and migration during cerebral cortical development. Front Neurosci 9:447. doi:10.3389/fnins.2015.00447 pmid:26733777
    OpenUrlCrossRefPubMed
  32. ↵
    1. Olson EC,
    2. Walsh CA
    (2002) Smooth, rough and upside-down neocortical development. Curr Opin Genet Dev 12:320–327. doi:10.1016/s0959-437x(02)00305-2 pmid:12076676
    OpenUrlCrossRefPubMed
  33. ↵
    1. Pinto-Lord MC,
    2. Evrard P,
    3. Caviness VS Jr.
    (1982) Obstructed neuronal migration along radial glial fibers in the neocortex of the reeler mouse: a Golgi-EM analysis. Brain Res 256:379–393. doi:10.1016/0165-3806(82)90181-X pmid:7127145
    OpenUrlCrossRefPubMed
  34. ↵
    1. Rakic P
    (1988) Defects of neuronal migration and the pathogenesis of cortical malformations. Prog Brain Res 73:15–37. doi:10.1016/s0079-6123(08)60494-x pmid:3047794
    OpenUrlCrossRefPubMed
  35. ↵
    1. Rakic P,
    2. Caviness VS Jr.
    (1995) Cortical development: view from neurological mutants two decades later. Neuron 14:1101–1104. doi:10.1016/0896-6273(95)90258-9 pmid:7605626
    OpenUrlCrossRefPubMed
  36. ↵
    1. Sanada K,
    2. Gupta A,
    3. Tsai LH
    (2004) Disabled-1-regulated adhesion of migrating neurons to radial glial fiber contributes to neuronal positioning during early corticogenesis. Neuron 42:197–211. doi:10.1016/s0896-6273(04)00222-3 pmid:15091337
    OpenUrlCrossRefPubMed
  37. ↵
    1. Schmucker D,
    2. Chen B
    (2009) Dscam and DSCAM: complex genes in simple animals, complex animals yet simple genes. Genes Dev 23:147–156. doi:10.1101/gad.1752909 pmid:19171779
    OpenUrlAbstract/FREE Full Text
  38. ↵
    1. Schmucker D,
    2. Clemens JC,
    3. Shu H,
    4. Worby CA,
    5. Xiao J,
    6. Muda M,
    7. Dixon JE,
    8. Zipursky SL
    (2000) Drosophila Dscam is an axon guidance receptor exhibiting extraordinary molecular diversity. Cell 101:671–684. doi:10.1016/s0092-8674(00)80878-8 pmid:10892653
    OpenUrlCrossRefPubMed
  39. ↵
    1. Schramm RD,
    2. Li S,
    3. Harris BS,
    4. Rounds RP,
    5. Burgess RW,
    6. Ytreberg FM,
    7. Fuerst PG
    (2012) A novel mouse Dscam mutation inhibits localization and shedding of DSCAM. PLoS One 7:e52652. doi:10.1371/journal.pone.0052652 pmid:23300735
    OpenUrlCrossRefPubMed
  40. ↵
    1. Sheldon M,
    2. Rice DS,
    3. D'Arcangelo G,
    4. Yoneshima H,
    5. Nakajima K,
    6. Mikoshiba K,
    7. Howell BW,
    8. Cooper JA,
    9. Goldowitz D,
    10. Curran T
    (1997) Scrambler and yotari disrupt the disabled gene and produce a reeler-like phenotype in mice. Nature 389:730–733. doi:10.1038/39601 pmid:9338784
    OpenUrlCrossRefPubMed
  41. ↵
    1. Soba P,
    2. Zhu S,
    3. Emoto K,
    4. Younger S,
    5. Yang SJ,
    6. Yu HH,
    7. Lee T,
    8. Jan LY,
    9. Jan YN
    (2007) Drosophila sensory neurons require Dscam for dendritic self-avoidance and proper dendritic field organization. Neuron 54:403–416. doi:10.1016/j.neuron.2007.03.029 pmid:17481394
    OpenUrlCrossRefPubMed
  42. ↵
    1. Sun Y,
    2. Fei T,
    3. Yang T,
    4. Zhang F,
    5. Chen YG,
    6. Li H,
    7. Xu Z
    (2010) The suppression of CRMP2 expression by bone morphogenetic protein (BMP)-SMAD gradient signaling controls multiple stages of neuronal development. J Biol Chem 285:39039–39050. doi:10.1074/jbc.M110.168351 pmid:20926379
    OpenUrlAbstract/FREE Full Text
  43. ↵
    1. Supèr H,
    2. Del Río JA,
    3. Martínez A,
    4. Pérez-Sust P,
    5. Soriano E
    (2000) Disruption of neuronal migration and radial glia in the developing cerebral cortex following ablation of Cajal-Retzius cells. Cereb Cortex 10:602–613. doi:10.1093/cercor/10.6.602 pmid:10859138
    OpenUrlCrossRefPubMed
  44. ↵
    1. Tsai JW,
    2. Chen Y,
    3. Kriegstein AR,
    4. Vallee RB
    (2005) LIS1 RNA interference blocks neural stem cell division, morphogenesis, and motility at multiple stages. J Cell Biol 170:935–945. doi:10.1083/jcb.200505166 pmid:16144905
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Turner TN, et al
    . (2016) Genome sequencing of autism-affected families reveals disruption of putative noncoding regulatory DNA. Am J Hum Genet 98:58–74. doi:10.1016/j.ajhg.2015.11.023 pmid:26749308
    OpenUrlCrossRefPubMed
  46. ↵
    1. Verhage M,
    2. Maia AS,
    3. Plomp JJ,
    4. Brussaard AB,
    5. Heeroma JH,
    6. Vermeer H,
    7. Toonen RF,
    8. Hammer RE,
    9. van den Berg TK,
    10. Missler M,
    11. Geuze HJ,
    12. Südhof TC
    (2000) Synaptic assembly of the brain in the absence of neurotransmitter secretion. Science 287:864–869. doi:10.1126/science.287.5454.864 pmid:10657302
    OpenUrlAbstract/FREE Full Text
  47. ↵
    1. Wallerand H,
    2. Cai Y,
    3. Wainberg ZA,
    4. Garraway I,
    5. Lascombe I,
    6. Nicolle G,
    7. Thiery JP,
    8. Bittard H,
    9. Radvanyi F,
    10. Reiter RR
    (2010) Phospho-Akt pathway activation and inhibition depends on N-cadherin or phospho-EGFR expression in invasive human bladder cancer cell lines. Urol Oncol 28:180–188. doi:10.1016/j.urolonc.2008.09.041 pmid:19070520
    OpenUrlCrossRefPubMed
  48. ↵
    1. Wang T, et al
    . (2016) De novo genic mutations among a Chinese autism spectrum disorder cohort. Nat Commun 7:13316. doi:10.1038/ncomms13316 pmid:27824329
    OpenUrlCrossRefPubMed
  49. ↵
    1. Yamagata M,
    2. Sanes JR
    (2008) Dscam and Sidekick proteins direct lamina-specific synaptic connections in vertebrate retina. Nature 451:465–469. doi:10.1038/nature06469 pmid:18216854
    OpenUrlCrossRefPubMed
  50. ↵
    1. Yamagata M,
    2. Sanes JR
    (2018) Expression and roles of the immunoglobulin superfamily recognition molecule sidekick1 in mouse retina. Front Mol Neurosci 11:485. doi:10.3389/fnmol.2018.00485 pmid:30687002
    OpenUrlCrossRefPubMed
  51. ↵
    1. Yamagata M,
    2. Duan X,
    3. Sanes JR
    (2018) Cadherins interact with synaptic organizers to promote synaptic differentiation. Front Mol Neurosci 11:142. doi:10.3389/fnmol.2018.00142 pmid:29760652
    OpenUrlCrossRefPubMed
  52. ↵
    1. Yamakawa K,
    2. Huot YK,
    3. Haendelt MA,
    4. Hubert R,
    5. Chen XN,
    6. Lyons GE,
    7. Korenberg JR
    (1998) DSCAM: a novel member of the immunoglobulin superfamily maps in a Down syndrome region and is involved in the development of the nervous system. Hum Mol Genet 7:227–237. doi:10.1093/hmg/7.2.227 pmid:9426258
    OpenUrlCrossRefPubMed
  53. ↵
    1. Yang T,
    2. Sun Y,
    3. Zhang F,
    4. Zhu Y,
    5. Shi L,
    6. Li H,
    7. Xu Z
    (2012) POSH localizes activated Rac1 to control the formation of cytoplasmic dilation of the leading process and neuronal migration. Cell Rep 2:640–651. doi:10.1016/j.celrep.2012.08.007 pmid:22959435
    OpenUrlCrossRefPubMed
  54. ↵
    1. Zipursky SL,
    2. Sanes JR
    (2010) Chemoaffinity revisited: dscams, protocadherins, and neural circuit assembly. Cell 143:343–353. doi:10.1016/j.cell.2010.10.009 pmid:21029858
    OpenUrlCrossRefPubMed
  55. ↵
    1. Zou H,
    2. Li Y,
    3. Liu X,
    4. Wang X
    (1999) An APAF-1.cytochrome c multimeric complex is a functional apoptosome that activates procaspase-9. J Biol Chem 274:11549–11556. doi:10.1074/jbc.274.17.11549 pmid:10206961
    OpenUrlAbstract/FREE Full Text
Back to top

In this issue

The Journal of Neuroscience: 42 (28)
Journal of Neuroscience
Vol. 42, Issue 28
13 Jul 2022
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Masthead (PDF)
Email

Thank you for sharing this Journal of Neuroscience article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Migrating Pyramidal Neurons Require DSCAM to Bypass the Border of the Developing Cortical Plate
(Your Name) has forwarded a page to you from Journal of Neuroscience
(Your Name) thought you would be interested in this article in Journal of Neuroscience.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
View Full Page PDF
Citation Tools
Migrating Pyramidal Neurons Require DSCAM to Bypass the Border of the Developing Cortical Plate
Tao Yang, Macy W. Veling, Xiao-Feng Zhao, Nicholas P. Prin, Limei Zhu, Ty Hergenreder, Hao Liu, Lu Liu, Zachary S. Rane, Masha G. Savelieff, Peter G. Fuerst, Qing Li, Kenneth Y. Kwan, Roman J. Giger, Yu Wang, Bing Ye
Journal of Neuroscience 13 July 2022, 42 (28) 5510-5521; DOI: 10.1523/JNEUROSCI.0997-21.2022

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Respond to this article
Request Permissions
Share
Migrating Pyramidal Neurons Require DSCAM to Bypass the Border of the Developing Cortical Plate
Tao Yang, Macy W. Veling, Xiao-Feng Zhao, Nicholas P. Prin, Limei Zhu, Ty Hergenreder, Hao Liu, Lu Liu, Zachary S. Rane, Masha G. Savelieff, Peter G. Fuerst, Qing Li, Kenneth Y. Kwan, Roman J. Giger, Yu Wang, Bing Ye
Journal of Neuroscience 13 July 2022, 42 (28) 5510-5521; DOI: 10.1523/JNEUROSCI.0997-21.2022
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Keywords

  • cell adhesion
  • cortex development
  • Dscam
  • migration
  • pyramidal neuron
  • termination

Responses to this article

Respond to this article

Jump to comment:

No eLetters have been published for this article.

Related Articles

Cited By...

More in this TOC Section

Research Articles

  • Macrophages Promote Repair of Inner Hair Cell Ribbon Synapses following Noise-Induced Cochlear Synaptopathy
  • The Basolateral Amygdala Sends a Mixed (GABAergic and Glutamatergic) Projection to the Mediodorsal Thalamic Nucleus
  • Total Sleep Deprivation Increases Brain Age Prediction Reversibly in Multisite Samples of Young Healthy Adults
Show more Research Articles

Development/Plasticity/Repair

  • Macrophages Promote Repair of Inner Hair Cell Ribbon Synapses following Noise-Induced Cochlear Synaptopathy
  • Pairing with enriched sound exposure restores auditory processing degraded by an antidepressant
  • Cbln1 Directs Axon Targeting by Corticospinal Neurons Specifically toward Thoraco-Lumbar Spinal Cord
Show more Development/Plasticity/Repair
  • Home
  • Alerts
  • Visit Society for Neuroscience on Facebook
  • Follow Society for Neuroscience on Twitter
  • Follow Society for Neuroscience on LinkedIn
  • Visit Society for Neuroscience on Youtube
  • Follow our RSS feeds

Content

  • Early Release
  • Current Issue
  • Issue Archive
  • Collections

Information

  • For Authors
  • For Advertisers
  • For the Media
  • For Subscribers

About

  • About the Journal
  • Editorial Board
  • Privacy Policy
  • Contact
(JNeurosci logo)
(SfN logo)

Copyright © 2023 by the Society for Neuroscience.
JNeurosci Online ISSN: 1529-2401

The ideas and opinions expressed in JNeurosci do not necessarily reflect those of SfN or the JNeurosci Editorial Board. Publication of an advertisement or other product mention in JNeurosci should not be construed as an endorsement of the manufacturer’s claims. SfN does not assume any responsibility for any injury and/or damage to persons or property arising from or related to any use of any material contained in JNeurosci.