Next Article in Journal
Comparing between Different Sets of Preprocessing, Classifiers, and Channels Selection Techniques to Optimise Motor Imagery Pattern Classification System from EEG Pattern Recognition
Next Article in Special Issue
Correction: Liu et al. Responsible Genes for Neuronal Migration in the Chromosome 17p13.3: Beyond Pafah1b1(Lis1), Crk and Ywhae(14-3-3ε). Brain Sci. 2022, 12, 56
Previous Article in Journal
Mapping Actuarial Criteria for Parkinson’s Disease-Mild Cognitive Impairment onto Data-Driven Cognitive Phenotypes
 
 
Correction published on 25 February 2022, see Brain Sci. 2022, 12(3), 311.
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Responsible Genes for Neuronal Migration in the Chromosome 17p13.3: Beyond Pafah1b1(Lis1), Crk and Ywhae(14-3-3ε)

1
Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
2
Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
*
Author to whom correspondence should be addressed.
Brain Sci. 2022, 12(1), 56; https://doi.org/10.3390/brainsci12010056
Submission received: 13 November 2021 / Revised: 13 December 2021 / Accepted: 23 December 2021 / Published: 30 December 2021 / Corrected: 25 February 2022

Abstract

:
The 17p13.3 chromosome region is often deleted or duplicated in humans, resulting in severe neurodevelopmental disorders such as Miller–Dieker syndrome (MDS) and 17p13.3 duplication syndrome. Lissencephaly can also be caused by gene mutations or deletions of a small piece of the 17p13.3 region, including a single gene or a few genes. PAFAH1B1 gene, coding for LIS1 protein, is a responsible gene for lissencephaly and MDS and regulates neuronal migration by controlling microtubules (MTs) and cargo transport along MTs via dynein. CRK is a downstream regulator of the reelin signaling pathways and regulates neuronal migration. YWHAE, coding for 14-3-3ε, is also responsible for MDS and regulates neuronal migration by binding to LIS1-interacting protein, NDEL1. Although these three proteins are known to be responsible for neuronal migration defects in MDS, there are 23 other genes in the MDS critical region on chromosome 17p13.3, and little is known about their functions in neurodevelopment, especially in neuronal migration. This review will summarize the recent progress on the functions of LIS1, CRK, and 14-3-3ε and describe the recent findings of other molecules in the MDS critical regions in neuronal migration.

Graphical Abstract

1. Introduction

The formation of the human brain requires the accurate completion of neurogenesis, neuronal migration, and the formation of appropriate connections of more than 100 billion neuronal cells during brain development. Neuronal migration in the developing cortex is composed of multiple steps, including multipolar migration, locomotion, and terminal translocation, and all steps are essential for creating a functional brain. If neuronal migration is disrupted, it results in a wide range of diseases, including brain morphological disorders such as Miller–Dieker syndrome (MDS), epilepsy, and neuropsychiatric disorders such as schizophrenia, attention-deficit hyperactivity disorder (ADHD), and autism spectrum disorder (ASD) [1,2,3,4,5,6,7,8,9]. Lissencephaly is a human brain malformation characterized by a smooth cerebral surface where the characteristic gyral folding pattern of the cerebral cortex is reduced or absent. Isolated lissencephaly sequence (ILS) is a heterogeneous disorder consisting of lissencephaly with no other major malformations. Causative mutations and deletions in PAFAH1B1 (also known as LIS1) as well as centromeric chromosome deletions in 17p13.3 account for most cases of ILS [10]. Mutations in other chromosome regions have also been associated with ILS, such as RELN on human chromosome 7q22.1, DMRTA2 on 1p32.3, and TUBA1A (tubulin alpha 1a) on 12q13.2 [11,12,13,14]. X-linked lissencephaly results from mutations in X chromosome-residing genes—the Aristaless-related homeobox gene (ARX), and doublecortin (DCX)—which are the two most notable X-linked genes causing lissencephaly [15]. By contrast, MDS consists of severe classical lissencephaly associated with other symptoms, such as facial anomalies, and MDS patients have larger heterozygous deletions that include the PAFAH1B1 (LIS1) and YWHAE (14-3-3ε) genes, compared with patients with ILS. The 17p13.3 region is well conserved, and genes within the human 17p13.3 chromosome region are syntenic to mouse chromosome 11 [16,17]. A previous study clarified that an approximate 1.4-M deletion region (MDS critical region) from the PAFAH1B1 gene to the YWHAE gene in the 17p13.3 region is critical for the development of MDS [18].
Mechanistically, regulation of MTs and F-actin organization, as well as regulation in cell adhesion and neurotrophic stimulation, is essential for neuronal migration, and the coordination between these mechanisms makes it possible for the neurons to migrate. Cortical neurons typically migrate by two modes of migration: radial migration and tangential migration. Some neurons also display more complicated migration called switching migration [19]. In lissencephaly, radial migration defects account for the majority of the cause, although defects in tangential migration are also implicated [15]. During migration, neurons polarize to form a leading process and a trailing process. The tip of the leading process is enriched in F-actin and actin motor proteins (i.e., myosins), whereas the stem region of the leading process is enriched with MTs. In locomotion, post-mitotic neuron migration occurs along the radial glial cells, and adhesion molecules help neurons to attach to the radial glial cells, creating traction force for the movement. MTs form a cage-like structure surrounding the nucleus, which helps the nucleus move during migration [20]. The retraction of the trailing process in migrating neurons is mainly mediated by actomyosin but is also mediated by MTs [21].
There are 26 genes from PAFAH1B1 to YWHAE in the human chromosome 17p13.3 region that are deleted in MDS patients (Figure 1). So far, 3 out of the 26 genes (PAFAH1B1, CRK, and YWHAE) in the MDS critical region have been widely known to be involved in cortical development, including neuronal migration [18,22,23]. Pafah1b1, Ywhae, and Crk knockout mice display cortical dysplasia. In patients, multiple genes in the 17p13.3 region are disrupted. Therefore, it is difficult to prove which genes are involved in neuronal migration defects in addition to these three genes. Patients with either ILS or MDS have a heterozygous loss of the PAFAH1B1 gene in the 17p13.3 locus and display combinatorial defects, including neuronal migration defects, epilepsy, and craniofacial defects. However, brain malformation is the most characteristic and prominent phenotype in ILS and MDS patients. PAFAH1B1, YWHAE, and CRK genes are involved in brain malformation caused by the neuronal migration defects observed in MDS patients. However, it is unclear if the other 23 genes are also important for proper cortical formation and involved in brain malformation in MDS patients. Therefore, it is important to fill gaps in our knowledge about the functions of the remaining 23 genes in cortical development. The purpose of this review paper is to summarize the knowns of the three most notable genes—PAFAH1B1, YWHAE, and CRK—and to collect the pieces related to neuronal migration in the other genes in the 17p13.3 region since these have not gained much attention previously. This will shed light upon the etiology of MDS as well as the fundamental mechanisms of cortical development.

2. The Roles of YWHAE, PAFAH1B1, and CRK in Neuronal Migration

2.1. YWHAE/14-3-3ε

14-3-3 proteins are conserved and ubiquitously-expressed [24]. The 14-3-3 family is composed of seven isoforms in mammalian cells (denoted by Greek letters β, γ, ε, η, σ, τ, and ζ). These 14-3-3 proteins bind to more than 100 targets and are involved in multiple cellular functions, including cell cycle control, apoptosis, and cancer [24]. The protein 14-3-3ε is coded by a gene, YWHAE (Tyrosine 3-Monooxygenase/Tryptophan 5-Monooxygenase Activation Protein Epsilon), located in the 17p13.3 chromosome region. Mechanistically, 14-3-3ε regulates MT organization via protection of the nuclear distribution protein nude-like 1 (NDEL1, formerly known as NUDEL) from postmodification (Figure 2A). The 14-3-3ε protein interacts with the NDEL1, and this binding prevents phospho-NDEL1 from dephosphorylating by protein phosphatase 2 (PP2A). Thus, NDEL1 stays active and positively regulates the interaction between LIS1 and dynein. We have previously demonstrated that 14-3-3ε is a critical factor in neuronal migration, indicating that the YWHAE gene is a strong candidate as one of the major genes responsible for the more severe lissencephaly phenotype displayed by MDS patients [25]. Using human patient samples, we also demonstrated a strong correlation between deficiency of the YWHAE gene and the severity of cerebral cortical malformations in MDS patients [18]. In addition to the 14-3-3ε functions in locomotion, 14-3-3ε regulates earlier cortical development, including neurogenesis, neuronal differentiation, and the distribution of intermediate progenitor cells (IPCs), suggesting the importance of 14-3-3ε in multipolar migration [26], and 14-3-3ε KO resulted in an increased number of proliferating progenitor cells and a broader distribution of the cells in the cortex, as well as an altered progenitor cell differentiation into neurons in the mouse model.
YWHAE is also implicated in the 17p13.3 microduplication syndrome [27]. It has been reported that microduplications in the 17p13.3 region result in a new genetic syndrome termed “the 17p13.3 microduplication syndrome”, which is associated with neurological disorders [28,29,30,31,32,33,34,35,36]. Specifically, 17p13.3 microduplication syndrome is characterized by various-sized duplications in the 17p13.3 chromosome locus, resulting in severe developmental defects, including autism spectrum disorder (ASD), epilepsy, intellectual disabilities, and malformation in the hands and feet. Importantly, the microduplication minimal region, a 72kb region within the 17p13.3 locus, has been defined, and this region exclusively contains the YWHAE gene encoding the 14-3-3ε protein and is strongly associated with ASD [30,33]. Although no pathological analyses have been performed in patients with 17p13.3 microduplication syndrome before, ASD patients show defects in neuronal morphogenesis, including neurite formation and spine formation [37,38,39]. Therefore, the defects in neuronal morphogenesis and spine formation and synaptogenesis caused by YWHAE overexpression may be an associated cause of ASD seen in patients with this syndrome and provide the first pathological findings. These studies strongly implicate YWHAE as a causative gene for ASD. We confirmed that Ywhae overexpression resulted in defects in neurite initiation during cortical development in vivo, mechanistically involving the aforementioned X-linked lissencephaly-associated gene DCX [40]. In addition, an association of YWHAE with schizophrenia has been recently recognized [41]. Interestingly, the 14-3-3 protein family, which consists of 7 isoforms, has been implicated in schizophrenia, ADHD, and general brain development, either when disrupted individually or together. This poses a possibility of cross-talk between the 14-3-3 isoforms. For instance, expression alteration in 5 out of 7 14-3-3 family members—YWHAB (14-3-3β), YWHAE, YWHAH (14-3-3η), YWHAZ (14-3-3ζ), and SFN (14-3-3σ)—is seen in cases of ASD and schizophrenia [41]. Moreover, the Ywhae/Ywhaz double KO mice show more defects in neuronal migration and additional disruptions in other neurodevelopmental stages, such as proliferation in neuronal progenitor cells compared with single KO Ywhae or Ywhaz in mice, suggesting a strong genetic interaction between YWHAE and YWHAZ [25,26,42].
Hence, 14-3-3ε is a gene responsible for MDS by regulating neuronal migration but is also implicated in multiple steps of cortical development, including neurogenesis, neuronal differentiation, and neuronal morphogenesis.

2.2. PAFAH1B1 (LIS1)

The PAFAH1B1 (LIS1) gene localizes at the telomeric region of chromosome 17p13.3 [30,43]. The implication of PAFAH1B1 in neuronal migration and Lissencephaly/MDS has been extensively analyzed and reported [44,45,46,47,48,49,50]. PAFAH1B1 was first identified as a subunit of platelet-activating factor acetylhydrolase (PAF-AH) [51], and functions as a regulator of microtubule (MT) motor proteins have been reported [52]. Point mutations and intragenic deletions in PAFAH1B1 were identified in ILS patients who showed no gross structural chromosomal rearrangements [53,54]. Although the more severe neuronal migration phenotype displayed by MDS patients suggests that genes other than PAFAH1B1 are responsible for this phenotype, PAFAH1B1 is a major responsible gene for lissencephaly phenotypes seen in MDS patients. In addition, PAFAH1B1 mutations in humans and mice result in seizures, and PAFAH1B1-deficient mice show sociability defects related to ASD but not the repetitive behavior, implicating PAFAH1B1 in the proper establishment of neural circuits in addition to neuronal migration [55,56,57,58,59].
Studies about PAFAH1B1 functions in dynein regulation not only in neurons but also in other cell types have been extensively performed, and numerous pieces of evidence have been accumulated [47,50,60]. Previous studies suggest multiple functions of LIS1 in dynein regulation (Figure 2B). For example, PAFAH1B1 is important for binding to MTs and dissociating dynein from MTs [61,62]. The structural analysis of PAFAH1B1 functions in dynein also revealed that PAFAH1B1 induces a persistent microtubule-bound state in dynein by acting on its linker domain [63]. Furthermore, PAFAH1B1 proteins seem to be dissociated from dynein after the dynein–dynactin complexes are composed [64]. In addition to the PAFAH1B1 functions in MTs and dynein, recent studies have reported PAFAH1B1 functions in actin regulation [65,66]. In migrating NIH3T3 cells, Pafah1b1 knockdown resulted in a reduction in traction force and disorganization of the microtubules and actin filaments [67]. This suggests that PAFAH1B1 might regulate actin in addition to MTs, and PAF1H1B1-driven traction force might be essential for neuronal migration.

2.3. CRK

The CRK gene is also localized in the 17p13.3 region, encodes an adapter protein that binds to tyrosine-phosphorylated proteins, and is involved in the Reelin signaling pathway [68]. Reelin is a secreted extracellular glycoprotein encoded by one of the ILS-associated genes, RELN, as we briefly mentioned in the introduction [11]. The reeler mice were first reported in 1951 [69], and their brain defects were extensively characterized [70,71]. Reelin binds to its two receptors: very-low-density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (apoER2). The two receptors then activate the adaptor protein Disabled-1 (Dab1), recruiting and activating the Src family tyrosine kinases (SFK), such as Src and Fyn, as well as interacting with various adaptor proteins, including Crk and Crk-like (CrkL) [72,73,74,75] (Figure 2C). Since homozygous Crk and CrkL knockout mice are lethal in early development, the functions of Crk and CrkL in neuronal migration were not clear [76,77]. By producing and analyzing the double Crk and CrkL conditional deficient mice, it has been found that Crk and CrkL are essential for neuronal migration [77]. After Reelin binds to its receptor, α5β1 integrin is activated through the intracellular pathway involving Dab1, Crk/CrkL, C3G, and Rap1, and this signaling pathway is vital for terminal translocation, the final step of neuronal migration [78,79]. Patients with chromosome 17p13.3 deletions involving CRK and YWHAE but not PAFAH1B1 were reported, and they showed generalized epilepsy, growth retardation, and macrocephaly, implicating Crk deficiency in epilepsy [80]. Another patient who had 730Kb deletion containing 11 genes in chromosome 17p13.3, including YWHAE and CRK but not PAFAH1B1, showed infantile spasms syndrome [81]. These data suggest that the CRK gene is one of the causative genes for more severe neuronal migration defects and may be partially responsible for the seizure phenotypes in MDS patients.
Moreover, in ASD patients, Reelin protein expression is reduced, predicting a downregulation of CRK signaling in ASD [82,83]. Interestingly, RELN mRNA is a target of the fragile X mental retardation protein (FMRP), coded by ASD risk gene FMR1 [84]. Therefore, the reduced RELN may be due to alteration of FMRP expression. FMR1 mutations result in fragile X syndrome, one of the most common heritable forms of ASD [85]. Neuronal migration and neurite formation defects, as well as altered synaptic plasticity, have been demonstrated in FMRP deficiency models [38,86,87,88,89]. The Fmr1-deficient mice display repetitive behavior and sociability defects [90]. Social behavior phenotypes are also observed in different loss-of-function models for Reelin functional study. However, no repetitive behavioral abnormality has been described so far [91,92].
In conclusion, CRK might be also one of the causes of the autistic phenotype in MDS patients and might be functionally associated with FMRP.

3. The Functions of Other Genes in the 17p13.3 Region and Their Potential Roles in Neuronal Migration

There are 23 genes in the critical region of chromosome 17p13.3 MDS, aside from the most notable ones (i.e., PAFAH1B1, CRK, and YWHAE). Some of them are involved in non-neuronal cell migration and microtubule and actin regulation (Table 1). Therefore, it could be worthwhile to study them in neuronal migration and dissect their functions in the onset and development of lissencephaly. Here, we focus on four genes that are involved in neuronal development, cytoskeletal organization, or engaging in protein interactions that have been implicated in cell migration.
SERPHINF1 is a member of the superfamily of serine protease inhibitors (Serpin) encoding the 50-kDa secreted protein pigment epithelium-derived factor (PEDF) [93], which was originally purified from the culture media of human retinal pigment epithelial cells [94]. PEDF is a non-inhibitory serpin, and PEDF acts as a multifunctional factor, such as neurotrophic, anti-tumorigenic, and anti-angiogenic factor [95,96,97]. Serpinf1 KO mice show multiple defects in the retina, particularly vessel formation [98]. Although the Serpinf1 gene is localized in the 17p13.3 chromosome region and often deleted or duplicated, resulting in neurodevelopmental disorders, few studies have taken into account the roles of Serpinf1 (PEDF) in brain development [43]. Our group recently analyzed Pedf’s function in corticogenesis and found that Pedf deficiency by specific shRNA resulted in multiple defects in neuronal development [43]. These include radial glial cell morphogenesis, neuronal migration, neurite formation, and spine formation [43]. We performed in utero electroporation (IUE) at the E15.5 embryos with Pedf-specific shRNA to target the upper layer neurons. Pedf-deficient post-mitotic neurons could migrate and reach the cortical plate (CP), suggesting no effects on multipolar migration from the ventricular zone to the subventricular zone and locomotion from the intermediate zone to the CP. However, Pedf -deficient neurons are sparsely distributed within the CP, suggesting defects in terminal translocation. PEDF is a secreted protein, and four receptors have been identified. They are PEDF-R, ribosomal protein SA (RPSA), plexin domain containing 1 (PLXDC1), and plexin domain containing 2 (PLXDC2) [99,100,101]. PEDF-R, also known as ATGL, is responsible for mediating PEDF’s effect on vascular hyperpermeability and triglyceride degradation, as well as protection against apoptosis induced by glucocorticoid [102,103]. RPSA, previously termed the 37-kDa laminin receptor precursor/67-kDa laminin receptor, is a laminin receptor that contributes to cell adhesion, migration, neurite outgrowth, and a variety of other events. PLXDC1, also known as tumor endothelial marker 7 (TEM7), and PLXDC2, also known as a mitogen for neuroprogenitors, are homologous membrane proteins [104,105]. Rpsa knockdown (KD) by shRNA in combination with IUE at E15.5 resulted in similar results to Pedf KD in neuronal migration and neurite morphogenesis [43]. Rpsa-deficient neurons in the upper cortical layers could migrate the CP but were sparsely distributed in the CP, suggesting defects in the terminal translocation. The lower layer neurons were also analyzed by performing IUE at E13.5 and showed more severe defects in terminal translocation. Thus, the Pedf-Rpsa signaling pathway is important for the proper completion of neuronal migration by regulating terminal translocation during cortical development. This is a new observation that implicates another gene in neuronal migration in addition to three genes: PAFAH1B1, CRK, and YWHAE.
Myosin 1c (MYO1C) is an unconventional myosin belonging to the class one myosin family. MYO1C is composed of three major domains: the N-terminal actin motor domain, regulatory neck domain that can bound to regulatory molecules, and the C-terminal tail domain that contains a pleckstrin homology domain. MYO1C has been indicated to regulate actin organization and lipid transportation, which are two aspects that are important for cell migration. As for actin regulation, MYO1C has been shown to be important for the regulation of cell cortical actin networks. The motor and tail regions of MYO1C cooperate to tether F-actin to the plasma membrane, thereby facilitating the establishment and dynamics of the cell shape. MYO1C not only works as a hook but also as a support point to power the F-actin to slide. Moreover, MYO1C promotes the formation of filamentous actin by transporting the globular actin [106]. On the other hand, the transportation of lipids to the cell membrane provides material for membrane extension. It has been found that MYO1C is involved in lipid recycling by promoting lipids to recycle to the cell membrane in HeLa cells [107]. In addition, the MYO1C PH domain can bind to phosphatidylinositol (4,5)-bisphosphate, which is intensively involved in molecular signals that associate with cell proliferation, polarity, and migration. Although, to the best of our knowledge, no previous studies showed MYO1C being involved in cortical neuronal migration, a selection of papers reported MYO1C regulating migration in tumor cells, such as glioblastoma cells (1321 N1 cell) and endometrial carcinoma cells [108,109].
Scavenger receptor class F member 1 (SCARF1), also known as scavenger receptor expressed by endothelial cells (SREC), encodes a transmembrane protein of the scavenger receptor family. Although its name implies its expression in endothelial cells, it is present in various cell types, including neuronal cells and epithelial cells [110]. In addition, SCARF1 is widely expressed throughout different tissues, including the brain [111]. The fundamental function of SCARF1 is to mediate the uptake of chemically modified low-density lipoproteins (LDLs) into cells via the extracellular domain. Other functions that have been revealed, although very limited, indicate SCARF1 might be important in neuronal migration. Shibata et al. have reported that the cytoplasmic domain of SCARF1 is important for actin organization via interacting with the actin regulatory protein Advillin [111]. Patten et al. found that SCARF1 acts as a critical adhesion regulator in the endothelial cells and leukocyte adhesive interaction. In this way, SCARF1 regulates leukocyte migration. However, the SCARF1 ligand in leukocytes that mediates this adhesive cascade is still unknown [110]. To the best of our knowledge, SCARF1 has not yet been studied in neuronal migration. However, SCARF1 could be a strong candidate gene for regulating neuronal migration due to its known functions in actin organization, cell adhesion, and non-neuronal cell migration.
Serine racemase (SRR) is an enzyme that was first discovered in the rodent brain, and it is enriched in glial cultures of the rodent cerebral cortex [112]. SRR is responsible for synthesizing D-serine from L-serine. The former is an agonist for the N-methyl-D-aspartate (NMDA) receptor and is essential in the NMDA signaling. SRR also helps to produce pyruvate and ammonia in the process of D-serine or L-serine alpha through beta elimination, which is a way to dehydrate serine [113]. While generating D-serine, SRR promotes N-methyl-D-aspartate (NMDA) receptor signaling, one of the most important machinery-regulating neuronal activities and neurodevelopmental events. NMDA receptors mediate pathways essential for neuronal migration by activating NMDAR-mediated calcium signaling and activating extracellular signal-regulated kinase (ERK) signaling cascades [114,115]. Aside from the catalytic functions, SRR is also involved in direct interactions with other proteins. One in particular is Disrupted-in-Schizophrenia-1 (DISC1), whose name alludes to its correlation with the neurodevelopmental disease schizophrenia [116]. Indeed, DISC1 was found to be involved in multiple neurodevelopmental events, including neuronal migration [117]. DISC1 plays a determinative role in the radial migration of the hippocampal pyramidal neurons as well as tangential migration of the cerebral cortical interneurons [118,119]. Furthermore, a DISC1 interacting protein, CAMDI, has been shown to regulate radial migration in the cortical neurons [120]. Although the interaction between DISC1 and SRR is important for D-serine synthesis, SRR can also generate agglomerates with DISC1 in cortical neurons, which also promotes NMDAR activity [121]. According to these findings, it is possible that SRR might also be implicated in neuronal migration. However, direct analysis of SRR in neuronal migration has not been performed, leaving a niche in the neuronal migration research field.

4. Conclusions

Abnormal neuronal migration is associated with neurodevelopmental diseases, most notably lissencephaly and more severely MDS. In this review, we have discussed the most well-studied three MDS critical genes—PAFAH1B1, CRK, and YWHAE—in the human chromosome 17p13.3, as well as those that have been unexplored in the brain development area, such as PEDF, SCARF1, SRR, and MYO1C. It is interesting to find that a lot of the genes have been studied in the migration of many cell types other than neurons. Additionally, some of these genes have been demonstrated in the regulation of cytoskeletal components, namely microtubules and F-actin. Therefore, they pose some level of potential to be involved in neuronal migration. We think this is extremely important, because MDS is not caused by a single gene but rather by a combination of PAFAH1B1, CRK, and YWHAE deletion. In fact, a large portion of the patients have a piece of gene deletion or mutation spanning from PAFAH1B1 to YWHAE, including the under-examined genes. We hope this review can curtain up for studying the other 23 genes in neurodevelopment, especially in the neuronal migration research field.
Current animal models for lissencephaly and MDS are Pafah1b1, Ywhae, and Crk single-KO models and the Pafah1b1/Ywhae double-KO model [25,49,77]. Models for other genes have been generated, such as Srr KO mice, Serpinf1 KO mice, and Scarf1 KO mice. Neuronal migration analysis using those single-gene KO mice may help us understand the functions of the other 23 genes in neuronal migration. Human-induced pluripotent stem cells (hiPSCs) derived from MDS patients are currently available, and cerebral organoids derived from the hiPSCs have become a useful model for neuronal migration research [122,123]. Comparisons between hiPSCs derived from patients carrying different MDS critical gene deletions or mutations could become useful for dissecting the functions of each MDS critical gene.
Table 1. Genes’ functions and involvements in cell migration.
Table 1. Genes’ functions and involvements in cell migration.
GeneFunctions of the ProteinInvolvementReferences
MNTRegulator of the MYC/MAX/MAD network
  • Affects migration in the human hepatocellular carcinoma (HCC) cells
  • Mnt KO mice have craniofacial defects
[124] Wu et al., 2012
[125] Toyo-oka et al., 2004
SGSM2GTPase-activating protein involving in the modulation of the GTPases RAP and RAB
  • Interacts with E-cadherin and enhances migratory cell adhesion in the human epithelial T47D cells
[126] Lin et al., 2019
SRRProduction of D-serine from L-serineInteracts with Disrupted-in-Schizophrenia-1 (DISC1), and DISC1 KD causes a defect in cortical neuron radial migration[121] Jacobi et al., 2019
HIC1Transcription repressor and
tumor suppressor
  • Important for cranial neural crest migration via regulating cadherin protein expression pattern and canonical Wnt signaling
  • Hic1 KO mice show craniofacial defects
[127] Ray et al., 2020
[128] Valenta et al., 2006
[129] Carter et al., 2000
DPH1 (OVCA1)Responsible for diphthamide biosynthesisDph1 KO causes craniofacial abnormalities in mice, but no observations indicate a defect in neuronal migration[130] Yu et al., 2014
RTN4RL1 (NGR3)
  • Cell surface receptor
  • Regulate the phosphorylation of SRC and FAK
Regulates epithelial cell migration[131] He et al., 2018
RPA1Replication protein AOverexpression causes 17p13.3 instability[36] Outwin et al., 2011
RILP
  • Endocytosis regulator
  • Induces the recruitment of dynein–dynactin to Rab7-containing late endosomes and lysosomes
  • Promote the transport of endosomes and lysosomes along MTs
  • Interact with Ral guanine nucleotide dissociation stimulator
RILP inhibits cell migration in cancer cells[132] Margiotta et al., 2017
[133] Wang et al., 2015
SCARF1A member of the Scavenger receptor. Regulates endocytosis
  • Regulates cell adhesion in human endothelial cells.
  • Expresses in the embryonic brain.
  • Interacts with actin-regulatory protein, Advillin in mouse neuroblastoma cell (N2a)
[110] Patten et al., 2017
[111] Shibata et al. 2004
SLC43A2Amino acid transporter for methionine uptakeEssential for mouse embryonic development[134] Guetg et al., 2015
MYO1CUnconventional actin motor
  • Regulates cell cortex tension
  • Regulates F-actin polymerization by transporting G-actin at the leading edge of migrating endothelial cells
  • Promotes migration of 1321 N1 glioblastoma cell
[106] Fan et al., 2012
[108] Edimo et al., 2016

Author Contributions

X.L. and S.A.B. wrote the manuscript; X.L. created the illustrations and tables; L.R. organized and checked the citations; K.T.-o. edited the manuscript and finalized it. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Drexel University Students Tackling Advanced Research (STAR) Scholars Program for L.R., the National Institute of Child Health and Human Development, grant number F31HD103405-01A1 for S.A.B. and the National Institute of Neurological Disorders and Stroke, grant number NS096098-01A1 for K.T.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank the Department of Neurobiology and Anatomy at the Drexel University College of Medicine for their support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Muraki, K.; Tanigaki, K. Neuronal migration abnormalities and its possible implications for schizophrenia. Front. Neurosci. 2015, 9, 74. [Google Scholar] [CrossRef] [Green Version]
  2. Catts, V.S.; Fung, S.J.; Long, L.E.; Joshi, D.; Vercammen, A.; Allen, K.M.; Fillman, S.G.; Rothmond, D.A.; Sinclair, D.; Tiwari, Y.; et al. Rethinking schizophrenia in the context of normal neurodevelopment. Front. Cell Neurosci. 2013, 7, 60. [Google Scholar] [CrossRef] [Green Version]
  3. Kato, M.; Dobyns, W.B. Lissencephaly and the molecular basis of neuronal migration. Hum. Mol. Genet. 2003, 12, R89–R96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Liu, J.S. Molecular genetics of neuronal migration disorders. Curr. Neurol. Neurosci. Rep. 2011, 11, 171–178. [Google Scholar] [CrossRef] [PubMed]
  5. Kuzniecky, R. Epilepsy and malformations of cortical development: New developments. Curr. Opin. Neurol. 2015, 28, 151–157. [Google Scholar] [CrossRef] [PubMed]
  6. Barkovich, A.J.; Dobyns, W.B.; Guerrini, R. Malformations of cortical development and epilepsy. Cold Spring Harb. Perspect. Med. 2015, 5, a022392. [Google Scholar] [CrossRef] [PubMed]
  7. Telias, M. Molecular Mechanisms of Synaptic Dysregulation in Fragile X Syndrome and Autism Spectrum Disorders. Front. Mol. Neurosci. 2019, 12, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Almeida Montes, L.G.; Prado Alcántara, H.; Martínez García, R.B.; De La Torre, L.B.; Avila Acosta, D.; Duarte, M.G. Brain cortical thickness in ADHD: Age, sex, and clinical correlations. J. Atten. Disord. 2013, 17, 641–654. [Google Scholar] [CrossRef] [PubMed]
  9. Reiner, O.; Karzbrun, E.; Kshirsagar, A.; Kaibuchi, K. Regulation of neuronal migration, an emerging topic in autism spectrum disorders. J. Neurochem. 2016, 136, 440–456. [Google Scholar] [CrossRef] [PubMed]
  10. Haverfield, E.V.; Whited, A.J.; Petras, K.S.; Dobyns, W.B.; Das, S. Intragenic deletions and duplications of the LIS1 and DCX genes: A major disease-causing mechanism in lissencephaly and subcortical band heterotopia. Eur. J. Hum. Genet. 2009, 17, 911–918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Chang, B.S.; Duzcan, F.; Kim, S.; Cinbis, M.; Aggarwal, A.; Apse, K.A.; Ozdel, O.; Atmaca, M.; Zencir, S.; Bagci, H.; et al. The role of RELN in lissencephaly and neuropsychiatric disease. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2007, 144, 58–63. [Google Scholar] [CrossRef]
  12. Urquhart, J.E.; Beaman, G.; Byers, H.; Roberts, N.A.; Chervinsky, E.; O’Sullivan, J.; Pilz, D.; Fry, A.; Williams, S.G.; Bhaskar, S.S.; et al. DMRTA2 (DMRT5) is mutated in a novel cortical brain malformation. Clin. Genet. 2016, 89, 724–727. [Google Scholar] [CrossRef]
  13. Hebebrand, M.; Hüffmeier, U.; Trollmann, R.; Hehr, U.; Uebe, S.; Ekici, A.B.; Kraus, C.; Krumbiegel, M.; Reis, A.; Thiel, C.T.; et al. The mutational and phenotypic spectrum of TUBA1A-associated tubulinopathy. Orphanet J. Rare Dis. 2019, 14, 38. [Google Scholar] [CrossRef] [Green Version]
  14. Hikita, N.; Hattori, H.; Kato, M.; Sakuma, S.; Morotomi, Y.; Ishida, H.; Seto, T.; Tanaka, K.; Shimono, T.; Shintaku, H.; et al. A case of TUBA1A mutation presenting with lissencephaly and Hirschsprung disease. Brain Dev. 2014, 36, 159–162. [Google Scholar] [CrossRef]
  15. Marcorelles, P.; Laquerrière, A.; Adde-Michel, C.; Marret, S.; Saugier-Veber, P.; Beldjord, C.; Friocourt, G. Evidence for tangential migration disturbances in human lissencephaly resulting from a defect in LIS1, DCX and ARX genes. Acta Neuropathol. 2010, 120, 503–515. [Google Scholar] [CrossRef]
  16. Buchberg, A.M.; Brownell, E.; Nagata, S.; Jenkins, N.A.; Copeland, N.G. A comprehensive genetic map of murine chromosome 11 reveals extensive linkage conservation between mouse and human. Genetics 1989, 122, 153–161. [Google Scholar] [CrossRef]
  17. Hirotsune, S.; Pack, S.D.; Chong, S.S.; Robbins, C.M.; Pavan, W.J.; Ledbetter, D.H.; Wynshaw-Boris, A. Genomic organization of the murine Miller-Dieker/lissencephaly region: Conservation of linkage with the human region. Genome Res. 1997, 7, 625–634. [Google Scholar] [CrossRef] [Green Version]
  18. Cardoso, C.; Leventer, R.J.; Ward, H.L.; Toyo-Oka, K.; Chung, J.; Gross, A.; Martin, C.L.; Allanson, J.; Pilz, D.T.; Olney, A.H.; et al. Refinement of a 400-kb critical region allows genotypic differentiation between isolated lissencephaly, Miller-Dieker syndrome, and other phenotypes secondary to deletions of 17p13.3. Am. J. Hum. Genet. 2003, 72, 918–930. [Google Scholar] [CrossRef] [Green Version]
  19. Hatanaka, Y.; Zhu, Y.; Torigoe, M.; Kita, Y.; Murakami, F. From migration to settlement: The pathways, migration modes and dynamics of neurons in the developing brain. Proc. Jpn. Acad. Ser. B 2016, 92, 1–19. [Google Scholar] [CrossRef] [Green Version]
  20. Rivas, R.J.; Hatten, M.E. Motility and cytoskeletal organization of migrating cerebellar granule neurons. J. Neurosci. 1995, 15, 981–989. [Google Scholar] [CrossRef] [Green Version]
  21. Kopf, A.; Renkawitz, J.; Hauschild, R.; Girkontaite, I.; Tedford, K.; Merrin, J.; Thorn-Seshold, O.; Trauner, D.; Häcker, H.; Fischer, K.-D.; et al. Microtubules control cellular shape and coherence in amoeboid migrating cells. J. Cell Biol. 2020, 219. [Google Scholar] [CrossRef]
  22. Park, T.J.; Curran, T. Crk and Crk-like play essential overlapping roles downstream of disabled-1 in the Reelin pathway. J. Neurosci. 2008, 28, 13551–13562. [Google Scholar] [CrossRef] [Green Version]
  23. Dobyns, W.B.; Reiner, O.; Carrozzo, R.; Ledbetter, D.H. Lissencephaly. A human brain malformation associated with deletion of the LIS1 gene located at chromosome 17p13. JAMA 1993, 270, 2838–2842. [Google Scholar] [CrossRef]
  24. Bridges, D.; Moorhead, G.B.G. 14-3-3 Proteins: A Number of Functions for a Numbered Protein. Sci. STKE 2005, 2005, re10. [Google Scholar] [CrossRef]
  25. Toyo-oka, K.; Shionoya, A.; Gambello, M.J.; Cardoso, C.; Leventer, R.; Ward, H.L.; Ayala, R.; Tsai, L.H.; Dobyns, W.; Ledbetter, D.; et al. 14-3-3epsilon is important for neuronal migration by binding to NUDEL: A molecular explanation for Miller-Dieker syndrome. Nat. Genet. 2003, 34, 274–285. [Google Scholar] [CrossRef]
  26. Toyo-oka, K.; Wachi, T.; Hunt, R.F.; Baraban, S.C.; Taya, S.; Ramshaw, H.; Kaibuchi, K.; Schwarz, Q.P.; Lopez, A.F.; Wynshaw-Boris, A. 14-3-3ε and ζ regulate neurogenesis and differentiation of neuronal progenitor cells in the developing brain. J. Neurosci. 2014, 34, 12168–12181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Blazejewski, S.M.; Bennison, S.A.; Smith, T.H.; Toyo-Oka, K. Neurodevelopmental Genetic Diseases Associated With Microdeletions and Microduplications of Chromosome 17p13.3. Front. Genet. 2018, 9, 80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Tucker, M.E.; Escobar, L.F. Cleft lip/palate associated with 17p13.3 duplication involving a single candidate gene (YWHAE). Clin. Genet. 2014, 85, 600–601. [Google Scholar] [CrossRef] [PubMed]
  29. Al Kaissi, A.; Ganger, R.; Rötzer, K.M.; Klaushofer, K.; Grill, F. A child with split-hand/foot associated with tibial hemimelia (SHFLD syndrome) and thrombocytopenia maps to chromosome region 17p13.3. Am. J. Med. Genet. 2014, 164, 2338–2343. [Google Scholar] [CrossRef] [PubMed]
  30. Curry, C.J.; Rosenfeld, J.A.; Grant, E.; Gripp, K.W.; Anderson, C.; Aylsworth, A.S.; Saad, T.B.; Chizhikov, V.V.; Dybose, G.; Fagerberg, C.; et al. The duplication 17p13.3 phenotype: Analysis of 21 families delineates developmental, behavioral and brain abnormalities, and rare variant phenotypes. Am. J. Med. Genet. A 2013, 161, 1833–1852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Capra, V.; Mirabelli-Badenier, M.; Stagnaro, M.; Rossi, A.; Tassano, E.; Gimelli, S.; Gimelli, G. Identification of a rare 17p13.3 duplication including the BHLHA9 and YWHAE genes in a family with developmental delay and behavioural problems. BMC Med. Genet. 2012, 13, 93. [Google Scholar] [CrossRef] [Green Version]
  32. Avela, K.; Aktan-Collan, K.; Horelli-Kuitunen, N.; Knuutila, S.; Somer, M. A microduplication on chromosome 17p13.1p13.3 including the PAFAH1B1 (LIS1) gene. Am. J. Med. Genet. 2011, 155, 875–879. [Google Scholar] [CrossRef]
  33. Bruno, D.L.; Anderlid, B.M.; Lindstrand, A.; van Ravenswaaij-Arts, C.; Ganesamoorthy, D.; Lundin, J.; Martin, C.L.; Douglas, J.; Nowak, C.; Adam, M.P.; et al. Further molecular and clinical delineation of co-locating 17p13.3 microdeletions and microduplications that show distinctive phenotypes. J. Med. Genet. 2010, 47, 299–311. [Google Scholar] [CrossRef]
  34. Hyon, C.; Marlin, S.; Chantot-Bastaraud, S.; Mabboux, P.; Beaujard, M.P.; Al Ageeli, E.; Vazquez, M.P.; Picard, A.; Siffroi, J.P.; Portnoi, M.F. A new 17p13.3 microduplication including the PAFAH1B1 and YWHAE genes resulting from an unbalanced X;17 translocation. Eur. J. Med. Genet. 2011, 54, 287–291. [Google Scholar] [CrossRef]
  35. Roos, L.; Jonch, A.E.; Kjaergaard, S.; Taudorf, K.; Simonsen, H.; Hamborg-Petersen, B.; Brondum-Nielsen, K.; Kirchhoff, M. A new microduplication syndrome encompassing the region of the Miller-Dieker (17p13 deletion) syndrome. J. Med. Genet. 2009, 46, 703–710. [Google Scholar] [CrossRef]
  36. Outwin, E.; Carpenter, G.; Bi, W.; Withers, M.A.; Lupski, J.R.; O’Driscoll, M. Increased RPA1 Gene Dosage Affects Genomic Stability Potentially Contributing to 17p13.3 Duplication Syndrome. PLoS Genet. 2011, 7, e1002247. [Google Scholar] [CrossRef]
  37. Hashimoto, R.; Nakazawa, T.; Tsurusaki, Y.; Yasuda, Y.; Nagayasu, K.; Matsumura, K.; Kawashima, H.; Yamamori, H.; Fujimoto, M.; Ohi, K.; et al. Whole-exome sequencing and neurite outgrowth analysis in autism spectrum disorder. J. Hum. Genet. 2015, 61, 199–206. [Google Scholar] [CrossRef] [Green Version]
  38. Doers, M.E.; Musser, M.T.; Nichol, R.; Berndt, E.R.; Baker, M.; Gomez, T.M.; Zhang, S.-C.; Abbeduto, L.; Bhattacharyya, A. iPSC-derived forebrain neurons from FXS individuals show defects in initial neurite outgrowth. Stem Cells Dev. 2014, 23, 1777–1787. [Google Scholar] [CrossRef] [PubMed]
  39. Phillips, M.; Pozzo-Miller, L. Dendritic spine dysgenesis in autism related disorders. Neurosci. Lett. 2015, 601, 30–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Cornell, B.; Wachi, T.; Zhukarev, V.; Toyo-Oka, K. Regulation of neuronal morphogenesis by 14-3-3epsilon (Ywhae) via the microtubule binding protein, doublecortin. Hum. Mol. Genet. 2016, 25, 4405–4418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Torrico, B.; Antón-Galindo, E.; Fernàndez-Castillo, N.; Rojo-Francàs, E.; Ghorbani, S.; Pineda-Cirera, L.; Hervás, A.; Rueda, I.; Moreno, E.; Fullerton, J.M.; et al. Involvement of the 14-3-3 Gene Family in Autism Spectrum Disorder and Schizophrenia: Genetics, Transcriptomics and Functional Analyses. J. Clin. Med. 2020, 9, 1851. [Google Scholar] [CrossRef]
  42. Cheah, P.S.; Ramshaw, H.S.; Thomas, P.Q.; Toyo-Oka, K.; Xu, X.; Martin, S.; Coyle, P.; Guthridge, M.A.; Stomski, F.; van den Buuse, M.; et al. Neurodevelopmental and neuropsychiatric behaviour defects arise from 14-3-3ζ deficiency. Mol. Psychiatry 2012, 17, 451–466. [Google Scholar] [CrossRef] [Green Version]
  43. Blazejewski, S.M.; Bennison, S.A.; Ha, N.T.; Liu, X.; Smith, T.H.; Dougherty, K.J.; Toyo-Oka, K. Rpsa Signaling Regulates Cortical Neuronal Morphogenesis via Its Ligand, PEDF, and Plasma Membrane Interaction Partner, Itga6. Cereb. Cortex 2021. accepted. [Google Scholar] [CrossRef]
  44. Wynshaw-Boris, A. Lissencephaly and LIS1: Insights into the molecular mechanisms of neuronal migration and development. Clin. Genet. 2007, 72, 296–304. [Google Scholar] [CrossRef] [PubMed]
  45. Gupta, A.; Tsai, L.H.; Wynshaw-Boris, A. Life is a journey: A genetic look at neocortical development. Nat. Rev. Genet. 2002, 3, 342–355. [Google Scholar] [CrossRef] [PubMed]
  46. Vallee, R.B.; Tsai, J.W. The cellular roles of the lissencephaly gene LIS1, and what they tell us about brain development. Genes Dev. 2006, 20, 1384–1393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Markus, S.M.; Marzo, M.G.; McKenney, R.J. New insights into the mechanism of dynein motor regulation by lissencephaly-1. ELife. 2020, 9, e59737. [Google Scholar] [CrossRef] [PubMed]
  48. Reiner, O.; Carrozzo, R.; Shen, Y.; Wehnert, M.; Faustinella, F.; Dobyns, W.B.; Caskey, C.T.; Ledbetter, D.H. Isolation of a Miller-Dieker lissencephaly gene containing G protein beta-subunit-like repeats. Nature 1993, 364, 717–721. [Google Scholar] [CrossRef] [PubMed]
  49. Hirotsune, S.; Fleck, M.W.; Gambello, M.J.; Bix, G.J.; Chen, A.; Clark, G.D.; Ledbetter, D.H.; McBain, C.J.; Wynshaw-Boris, A. Graded reduction of Pafah1b1 (Lis1) activity results in neuronal migration defects and early embryonic lethality. Nat. Genet. 1998, 19, 333–339. [Google Scholar] [CrossRef] [PubMed]
  50. Wynshaw-Boris, A.; Pramparo, T.; Youn, Y.H.; Hirotsune, S. Lissencephaly: Mechanistic insights from animal models and potential therapeutic strategies. Semin. Cell Dev. Biol. 2010, 21, 823–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Hattori, M.; Adachi, H.; Tsujimoto, M.; Arai, H.; Inoue, K. Miller-Dieker lissencephaly gene encodes a subunit of brain platelet-activating factor acetylhydrolase. Nature 1994, 370, 216–218. [Google Scholar] [CrossRef]
  52. Sapir, T.; Elbaum, M.; Reiner, O. Reduction of microtubule catastrophe events by LIS1, platelet-activating factor acetylhydrolase subunit. EMBO J. 1997, 16, 6977–6984. [Google Scholar] [CrossRef] [Green Version]
  53. Lo Nigro, C.; Chong, C.S.; Smith, A.C.; Dobyns, W.B.; Carrozzo, R.; Ledbetter, D.H. Point mutations and an intragenic deletion in LIS1, the lissencephaly causative gene in isolated lissencephaly sequence and Miller-Dieker syndrome. Hum. Mol. Genet. 1997, 6, 157–164. [Google Scholar] [CrossRef] [Green Version]
  54. Pilz, D.T.; Macha, M.E.; Precht, K.S.; Smith, A.C.; Dobyns, W.B.; Ledbetter, D.H. Fluorescence in situ hybridization analysis with LIS1 specific probes reveals a high deletion mutation rate in isolated lissencephaly sequence. Genet. Med. 1998, 1, 29–33. [Google Scholar] [CrossRef] [Green Version]
  55. Hunt, R.F.; Dinday, M.T.; Hindle-Katel, W.; Baraban, S.C. LIS1 deficiency promotes dysfunctional synaptic integration of granule cells generated in the developing and adult dentate gyrus. J. Neurosci. 2012, 32, 12862–12875. [Google Scholar] [CrossRef] [Green Version]
  56. Shimojima, K.; Sugiura, C.; Takahashi, H.; Ikegami, M.; Takahashi, Y.; Ohno, K.; Matsuo, M.; Saito, K.; Yamamoto, T. Genomic copy number variations at 17p13.3 and epileptogenesis. Epilepsy Res. 2010, 89, 303–309. [Google Scholar] [CrossRef]
  57. Greenwood, J.S.; Wang, Y.; Estrada, R.C.; Ackerman, L.; Ohara, P.T.; Baraban, S.C. Seizures, enhanced excitation, and increased vesicle number in Lis1 mutant mice. Ann. Neurol. 2009, 66, 644–653. [Google Scholar] [CrossRef]
  58. Jones, D.L.; Baraban, S.C. Characterization of inhibitory circuits in the malformed hippocampus of Lis1 mutant mice. J. Neurophysiol. 2007, 98, 2737–2746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Sudarov, A.; Gooden, F.; Tseng, D.; Gan, W.B.; Ross, M.E. Lis1 controls dynamics of neuronal filopodia and spines to impact synaptogenesis and social behaviour. EMBO Mol. Med. 2013, 5, 591–607. [Google Scholar] [CrossRef] [PubMed]
  60. Canty, J.T.; Yildiz, A. Activation and Regulation of Cytoplasmic Dynein. Trends Biochem. Sci. 2020, 45, 440–453. [Google Scholar] [CrossRef] [PubMed]
  61. Faulkner, N.E.; Dujardin, D.L.; Tai, C.Y.; Vaughan, K.T.; O’Connell, C.B.; Wang, Y.; Vallee, R.B. A role for the lissencephaly gene LIS1 in mitosis and cytoplasmic dynein function. Nat. Cell Biol. 2000, 2, 784–791. [Google Scholar] [CrossRef]
  62. Smith, D.S.; Niethammer, M.; Ayala, R.; Zhou, Y.; Gambello, M.J.; Wynshaw-Boris, A.; Tsai, L.H. Regulation of cytoplasmic dynein behaviour and microtubule organization by mammalian Lis1. Nat. Cell. Biol. 2000, 2, 767–775. [Google Scholar] [CrossRef]
  63. Toropova, K.; Zou, S.; Roberts, A.J.; Redwine, W.B.; Goodman, B.S.; Reck-Peterson, S.L.; Leschziner, A.E. Lis1 regulates dynein by sterically blocking its mechanochemical cycle. Elife 2014, 3, e03372. [Google Scholar] [CrossRef]
  64. Htet, Z.M.; Gillies, J.P.; Baker, R.W.; Leschziner, A.E.; DeSantis, M.E.; Reck-Peterson, S.L. LIS1 promotes the formation of activated cytoplasmic dynein-1 complexes. Nat. Cell Biol. 2020, 22, 518–525. [Google Scholar] [CrossRef]
  65. Chhatre, A.; Sanghavi, P.; Mallik, R. Lis1 co-localizes with actin in the phagocytic cup and regulates phagocytosis. Cytoskeleton 2020, 77, 249–260. [Google Scholar] [CrossRef]
  66. Chen, J.; Cai, Z.; Zhang, L.; Yin, Y.; Chen, X.; Chen, C.; Zhang, Y.; Zhai, S.; Long, X.; Liu, X.; et al. Lis1 Regulates Germinal Center B Cell Antigen Acquisition and Affinity Maturation. J. Immunol. 2021, 198, 4304–4311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Jheng, G.-W.; Hur, S.S.; Chang, C.-M.; Wu, C.-C.; Cheng, J.-S.; Lee, H.-H.; Chung, B.-C.; Wang, Y.-K.; Lin, K.-H.; Del Álamo, J.C.; et al. Lis1 dysfunction leads to traction force reduction and cytoskeletal disorganization during cell migration. Biochem. Biophys. Res. Commun. 2018, 497, 869–875. [Google Scholar] [CrossRef]
  68. Chen, K.; Ochalski, P.G.; Tran, T.S.; Sahir, N.; Schubert, M.; Pramatarova, A.; Howell, B.W. Interaction between Dab1 and CrkII is promoted by Reelin signaling. J. Cell Sci. 2004, 117, 4527–4536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Falconer, D.S. Two new mutants, ‘trembler’ and ‘reeler’, with neurological actions in the house mouse (Mus musculus L.). J. Genet. 1951, 50, 192–201. [Google Scholar] [CrossRef] [PubMed]
  70. Miao, G.G.; Smeyne, R.J.; D’Arcangelo, G.; Copeland, N.G.; Jenkins, N.A.; Morgan, J.I.; Curran, T. Isolation of an allele of reeler by insertional mutagenesis. Proc. Natl. Acad. Sci. USA 1994, 91, 11050–11054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. D’Arcangelo, G.; Miao, G.G.; Chen, S.C.; Soares, H.D.; Morgan, J.I.; Curran, T. A protein related to extracellular matrix proteins deleted in the mouse mutant reeler. Nature 1995, 374, 719–723. [Google Scholar] [CrossRef]
  72. Keshvara, L.; Benhayon, D.; Magdaleno, S.; Curran, T. Identification of reelin-induced sites of tyrosyl phosphorylation on disabled 1. J. Biol. Chem. 2001, 276, 16008–16014. [Google Scholar] [CrossRef] [Green Version]
  73. Kuo, G.; Arnaud, L.; Kronstad-O’Brien, P.; Cooper, J.A. Absence of Fyn and Src causes a reeler-like phenotype. J. Neurosci. 2005, 25, 8578–8586. [Google Scholar] [CrossRef]
  74. Bock, H.H.; Herz, J. Reelin activates SRC family tyrosine kinases in neurons. Curr. Biol. 2003, 13, 18–26. [Google Scholar] [CrossRef] [Green Version]
  75. Ballif, B.A.; Arnaud, L.; Arthur, W.T.; Guris, D.; Imamoto, A.; Cooper, J.A. Activation of a Dab1/CrkL/C3G/Rap1 pathway in Reelin-stimulated neurons. Curr. Biol. 2004, 14, 606–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Guris, D.L.; Fantes, J.; Tara, D.; Druker, B.J.; Imamoto, A. Mice lacking the homologue of the human 22q11.2 gene CRKL phenocopy neurocristopathies of DiGeorge syndrome. Nat. Genet. 2001, 27, 293–298. [Google Scholar] [CrossRef]
  77. Park, T.-J.; Boyd, K.; Curran, T. Cardiovascular and craniofacial defects in Crk-null mice. Mol. Cell. Biol. 2006, 26, 6272–6282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Sekine, K.; Kawauchi, T.; Kubo, K.-I.; Honda, T.; Herz, J.; Hattori, M.; Kinashi, T.; Nakajima, K. Reelin controls neuronal positioning by promoting cell-matrix adhesion via inside-out activation of integrin α5β1. Neuron 2012, 76, 353–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. O’Dell, R.S.; Cameron, D.A.; Zipfel, W.R.; Olson, E.C. Reelin Prevents Apical Neurite Retraction during Terminal Translocation and Dendrite Initiation. J. Neurosci. 2015, 35, 10659. [Google Scholar] [CrossRef] [Green Version]
  80. Tenney, J.R.; Hopkin, R.J.; Schapiro, M.B. Deletion of 14-3-3ε and CRK: A clinical syndrome with macrocephaly, developmental delay, and generalized epilepsy. J. Child Neurol. 2011, 26, 223–227. [Google Scholar] [CrossRef]
  81. Romano, C.; Ferranti, S.; Mencarelli, M.A.; Longo, I.; Renieri, A.; Grosso, S. 17p13.3 microdeletion including YWHAE and CRK genes: Towards a clinical characterization. Neurol. Sci. 2020, 41, 2259–2262. [Google Scholar] [CrossRef]
  82. Fatemi, S.H.; Stary, J.M.; Halt, A.R.; Realmuto, G.R. Dysregulation of Reelin and Bcl-2 proteins in autistic cerebellum. J. Autism Dev. Disord. 2001, 31, 529–535. [Google Scholar] [CrossRef] [PubMed]
  83. Fatemi, S.H.; Stary, J.M.; Egan, E.A. Reduced blood levels of reelin as a vulnerability factor in pathophysiology of autistic disorder. Cell Mol. Neurobiol. 2002, 22, 139–152. [Google Scholar] [CrossRef] [PubMed]
  84. Darnell, J.C.; Van Driesche, S.J.; Zhang, C.; Hung, K.Y.; Mele, A.; Fraser, C.E.; Stone, E.F.; Chen, C.; Fak, J.J.; Chi, S.W.; et al. FMRP stalls ribosomal translocation on mRNAs linked to synaptic function and autism. Cell 2011, 146, 247–261. [Google Scholar] [CrossRef] [Green Version]
  85. Budimirovic, D.B.; Subramanian, M. Neurobiology of Disease. In Neurobiology of Autism and Intellectual Disability Fragile X Syndrome: Fragile X Syndrome; Oxford University Press: Oxford, UK, 2017. [Google Scholar]
  86. Pan, L.; Zhang, Y.Q.; Woodruff, E.; Broadie, K. The Drosophila fragile X gene negatively regulates neuronal elaboration and synaptic differentiation. Curr. Biol. 2004, 14, 1863–1870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Budimirovic, D.B.; Schlageter, A.; Filipovic-Sadic, S.; Protic, D.D.; Bram, E.; Mahone, E.M.; Nicholson, K.; Culp, K.; Javanmardi, K.; Kemppainen, J.; et al. A Genotype-Phenotype Study of High-Resolution FMR1 Nucleic Acid and Protein Analyses in Fragile X Patients with Neurobehavioral Assessments. Brain Sci. 2020, 10, 694. [Google Scholar] [CrossRef]
  88. Wu, C.; Zhang, X.; Chen, P.; Ruan, X.; Liu, W.; Li, Y.; Sun, C.; Hou, L.; Yin, B.; Qiang, B.; et al. MicroRNA-129 modulates neuronal migration by targeting Fmr1 in the developing mouse cortex. Cell Death Dis. 2019, 10, 287. [Google Scholar] [CrossRef] [PubMed]
  89. Sidorov, M.S.; Auerbach, B.D.; Bear, M.F. Fragile X mental retardation protein and synaptic plasticity. Mol. Brain 2013, 6, 15. [Google Scholar] [CrossRef] [Green Version]
  90. Kazdoba, T.M.; Leach, P.T.; Silverman, J.L.; Crawley, J.N. Modeling fragile X syndrome in the Fmr1 knockout mouse. Intractable Rare Dis. Res. 2014, 3, 118–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Podhorna, J.; Didriksen, M. The heterozygous reeler mouse: Behavioural phenotype. Behav. Brain Res. 2004, 153, 43–54. [Google Scholar] [CrossRef]
  92. Sakai, K.; Shoji, H.; Kohno, T.; Miyakawa, T.; Hattori, M. Mice that lack the C-terminal region of Reelin exhibit behavioral abnormalities related to neuropsychiatric disorders. Sci. Rep. 2016, 6, 28636. [Google Scholar] [CrossRef]
  93. Kawaguchi, T.; Yamagishi, S.I.; Sata, M. Structure-function relationships of PEDF. Curr. Mol. Med. 2010, 10, 302–311. [Google Scholar] [CrossRef] [PubMed]
  94. Tombran-Tink, J.; Johnson, L.V. Neuronal differentiation of retinoblastoma cells induced by medium conditioned by human RPE cells. Investig. Ophthalmol. Vis. Sci. 1989, 30, 1700–1707. [Google Scholar]
  95. Sugita, Y.; Becerra, S.P.; Chader, G.J.; Schwartz, J.P. Pigment epithelium-derived factor (PEDF) has direct effects on the metabolism and proliferation of microglia and indirect effects on astrocytes. J. Neurosci. Res. 1997, 49, 710–718. [Google Scholar] [CrossRef]
  96. Cao, W.; Tombran-Tink, J.; Chen, W.; Mrazek, D.; Elias, R.; McGinnis, J.F. Pigment epithelium-derived factor protects cultured retinal neurons against hydrogen peroxide-induced cell death. J. Neurosci. Res. 1999, 57, 789–800. [Google Scholar] [CrossRef]
  97. Dawson, D.W.; Volpert, O.V.; Gillis, P.; Crawford, S.E.; Xu, H.; Benedict, W.; Bouck, N.P. Pigment epithelium-derived factor: A potent inhibitor of angiogenesis. Science 1999, 285, 245–248. [Google Scholar] [CrossRef] [PubMed]
  98. Steele, F.R.; Chader, G.J.; Johnson, L.V.; Tombran-Tink, J. Pigment epithelium-derived factor: Neurotrophic activity and identification as a member of the serine protease inhibitor gene family. Proc. Natl. Acad. Sci. USA 1993, 90, 1526–1530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Subramanian, P.; Notario, P.M.; Becerra, S.P. Pigment epithelium-derived factor receptor (PEDF-R): A plasma membrane-linked phospholipase with PEDF binding affinity. Adv. Exp. Med. Biol. 2010, 664, 29–37. [Google Scholar] [CrossRef] [Green Version]
  100. Bernard, A.; Gao-Li, J.; Franco, C.-A.; Bouceba, T.; Huet, A.; Li, Z. Laminin receptor involvement in the anti-angiogenic activity of pigment epithelium-derived factor. J. Biol. Chem. 2009, 284, 10480–10490. [Google Scholar] [CrossRef] [Green Version]
  101. Cheng, G.; Zhong, M.; Kawaguchi, R.; Kassai, M.; Al-Ubaidi, M.; Deng, J.; Ter-Stepanian, M.; Sun, H. Identification of PLXDC1 and PLXDC2 as the transmembrane receptors for the multifunctional factor PEDF. Elife 2014, 3, e05401. [Google Scholar] [CrossRef]
  102. He, T.; Hu, J.; Yan, G.; Li, L.; Zhang, D.; Zhang, Q.; Chen, B.; Huang, Y. Pigment epithelium-derived factor regulates microvascular permeability through adipose triglyceride lipase in sepsis. Clin. Sci. 2015, 129, 49–61. [Google Scholar] [CrossRef]
  103. Yao, S.; Zhang, Y.; Wang, X.; Zhao, F.; Sun, M.; Zheng, X.; Dong, H.; Guo, K. Pigment Epithelium-Derived Factor (PEDF) Protects Osteoblastic Cell Line from Glucocorticoid-Induced Apoptosis via PEDF-R. Int. J. Mol. Sci. 2016, 17, 730. [Google Scholar] [CrossRef]
  104. Beaty, R.M.; Edwards, J.B.; Boon, K.; Siu, I.M.; Conway, J.E.; Riggins, G.J. PLXDC1 (TEM7) is identified in a genome-wide expression screen of glioblastoma endothelium. J. Neurooncol. 2007, 81, 241–248. [Google Scholar] [CrossRef]
  105. Miller-Delaney, S.F.C.; Lieberam, I.; Murphy, P.; Mitchell, K.J. Plxdc2 Is a Mitogen for Neural Progenitors. PLoS ONE 2011, 6, e14565. [Google Scholar] [CrossRef] [Green Version]
  106. Fan, Y.; Eswarappa, S.M.; Hitomi, M.; Fox, P.L. Myo1c facilitates G-actin transport to the leading edge of migrating endothelial cells. J. Cell Biol. 2012, 198, 47–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Brandstaetter, H.; Kendrick-Jones, J.; Buss, F. Myo1c regulates lipid raft recycling to control cell spreading, migration and Salmonella invasion. J. Cell Sci. 2012, 125, 1991–2003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Edimo, W.E.; Ramos, A.R.; Ghosh, S.; Vanderwinden, J.M.; Erneux, C. The SHIP2 interactor Myo1c is required for cell migration in 1321 N1 glioblastoma cells. Biochem. Biophys. Res. Commun. 2016, 476, 508–514. [Google Scholar] [CrossRef] [PubMed]
  109. Visuttijai, K.; Pettersson, J.; Mehrbani Azar, Y.; van den Bout, I.; Örndal, C.; Marcickiewicz, J.; Nilsson, S.; Hörnquist, M.; Olsson, B.; Ejeskär, K.; et al. Lowered Expression of Tumor Suppressor Candidate MYO1C Stimulates Cell Proliferation, Suppresses Cell Adhesion and Activates AKT. PLoS ONE 2016, 11, e0164063. [Google Scholar] [CrossRef] [PubMed]
  110. Patten, D.A.; Kamarajah, S.K.; Rose, J.M.; Tickle, J.; Shepherd, E.L.; Adams, D.H.; Weston, C.J.; Shetty, S. SCARF-1 promotes adhesion of CD4(+) T cells to human hepatic sinusoidal endothelium under conditions of shear stress. Sci. Rep. 2017, 7, 17600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Shibata, M.; Ishii, J.; Koizumi, H.; Shibata, N.; Dohmae, N.; Takio, K.; Adachi, H.; Tsujimoto, M.; Arai, H. Type F scavenger receptor SREC-I interacts with advillin, a member of the gelsolin/villin family, and induces neurite-like outgrowth. J. Biol. Chem. 2004, 279, 40084–40090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Wolosker, H.; Sheth, K.N.; Takahashi, M.; Mothet, J.-P.; Brady, R.O.; Ferris, C.D.; Snyder, S.H. Purification of serine racemase: Biosynthesis of the neuromodulator D-serine. Proc. Natl. Acad. Sci. USA 1999, 96, 721. [Google Scholar] [CrossRef] [Green Version]
  113. Foltyn, V.N.; Bendikov, I.; De Miranda, J.; Panizzutti, R.; Dumin, E.; Shleper, M.; Li, P.; Toney, M.D.; Kartvelishvily, E.; Wolosker, H. Serine racemase modulates intracellular D-serine levels through an alpha,beta-elimination activity. J. Biol. Chem. 2005, 280, 1754–1763. [Google Scholar] [CrossRef] [Green Version]
  114. Lau, C.G.; Takeuchi, K.; Rodenas-Ruano, A.; Takayasu, Y.; Murphy, J.; Bennett, M.V.L.; Zukin, R.S. Regulation of NMDA receptor Ca2+ signalling and synaptic plasticity. Biochem. Soc. Trans. 2009, 37, 1369–1374. [Google Scholar] [CrossRef] [Green Version]
  115. Xing, L.; Larsen, R.S.; Bjorklund, G.R.; Li, X.; Wu, Y.; Philpot, B.D.; Snider, W.D.; Newbern, J.M. Layer specific and general requirements for ERK/MAPK signaling in the developing neocortex. Elife 2016, 5, e11123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Millar, J.K.; Christie, S.; Semple, C.A.M.; Porteous, D.J. Chromosomal Location and Genomic Structure of the Human Translin-Associated Factor X Gene (TRAX; TSNAX) Revealed by Intergenic Splicing to DISC1, a Gene Disrupted by a Translocation Segregating with Schizophrenia. Genomics 2000, 67, 69–77. [Google Scholar] [CrossRef] [PubMed]
  117. Brandon, N.J.; Sawa, A. Linking neurodevelopmental and synaptic theories of mental illness through DISC1. Nat. Rev. Neurosci. 2011, 12, 707–722. [Google Scholar] [CrossRef] [PubMed]
  118. Tomita, K.; Kubo, K.-I.; Ishii, K.; Nakajima, K. Disrupted-in-Schizophrenia-1 (Disc1) is necessary for migration of the pyramidal neurons during mouse hippocampal development. Hum. Mol. Genet. 2011, 20, 2834–2845. [Google Scholar] [CrossRef] [PubMed]
  119. Steinecke, A.; Gampe, C.; Nitzsche, F.; Bolz, J. DISC1 knockdown impairs the tangential migration of cortical interneurons by affecting the actin cytoskeleton. Front. Cell. Neurosci. 2014, 8, 190. [Google Scholar] [CrossRef] [Green Version]
  120. Fukuda, T.; Sugita, S.; Inatome, R.; Yanagi, S. CAMDI, a novel disrupted in schizophrenia 1 (DISC1)-binding protein, is required for radial migration. J. Biol. Chem. 2010, 285, 40554–40561. [Google Scholar] [CrossRef] [Green Version]
  121. Jacobi, A.A.; Halawani, S.; Lynch, D.R.; Lin, H. Neuronal serine racemase associates with Disrupted-In-Schizophrenia-1 and DISC1 agglomerates: Implications for schizophrenia. Neurosci. Lett. 2019, 692, 107–114. [Google Scholar] [CrossRef]
  122. Bershteyn, M.; Nowakowski, T.J.; Pollen, A.A.; Di Lullo, E.; Nene, A.; Wynshaw-Boris, A.; Kriegstein, A.R. Human iPSC-Derived Cerebral Organoids Model Cellular Features of Lissencephaly and Reveal Prolonged Mitosis of Outer Radial Glia. Cell Stem Cell 2017, 20, 435–449.e434. [Google Scholar] [CrossRef] [Green Version]
  123. Bershteyn, M.; Hayashi, Y.; Desachy, G.; Hsiao, E.C.; Sami, S.; Tsang, K.M.; Weiss, L.A.; Kriegstein, A.R.; Yamanaka, S.; Wynshaw-Boris, A. Cell-autonomous correction of ring chromosomes in human induced pluripotent stem cells. Nature 2014, 507, 99–103. [Google Scholar] [CrossRef] [Green Version]
  124. Wu, J.; Zhou, Q.; Wang, Y.; Zhou, X.; Li, J. MNT inhibits the migration of human hepatocellular carcinoma SMMC7721 cells. Biochem. Biophys. Res. Commun. 2012, 418, 93–97. [Google Scholar] [CrossRef]
  125. Toyo-oka, K.; Hirotsune, S.; Gambello, M.J.; Zhou, Z.-Q.; Olson, L.; Rosenfeld, M.G.; Eisenman, R.; Hurlin, P.; Wynshaw-Boris, A. Loss of the Max-interacting protein Mnt in mice results in decreased viability, defective embryonic growth and craniofacial defects: Relevance to Miller–Dieker syndrome. Hum. Mol. Genet. 2004, 13, 1057–1067. [Google Scholar] [CrossRef] [Green Version]
  126. Lin, J.-H.; Lee, W.-J.; Wu, H.-C.; Wu, C.-H.; Chen, L.-C.; Huang, C.-C.; Chang, H.-L.; Cheng, T.-C.; Chang, H.-W.; Ho, C.-T.; et al. Small G protein signalling modulator 2 (SGSM2) is involved in oestrogen receptor-positive breast cancer metastasis through enhancement of migratory cell adhesion via interaction with E-cadherin. Cell Adhes. Migr. 2019, 13, 120–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Ray, H.; Chang, C. The transcription factor Hypermethylated in Cancer 1 (Hic1) regulates neural crest migration via interaction with Wnt signaling. Dev. Biol. 2020, 463, 169–181. [Google Scholar] [CrossRef]
  128. Valenta, T.; Lukas, J.; Doubravska, L.; Fafilek, B.; Korinek, V. HIC1 attenuates Wnt signaling by recruitment of TCF-4 and beta-catenin to the nuclear bodies. EMBO J. 2006, 25, 2326–2337. [Google Scholar] [CrossRef] [Green Version]
  129. Carter, M.G.; Johns, M.A.; Zeng, X.; Zhou, L.; Zink, M.C.; Mankowski, J.L.; Donovan, D.M.; Baylin, S.B. Mice deficient in the candidate tumor suppressor gene Hic1 exhibit developmental defects of structures affected in the Miller–Dieker syndrome. Hum. Mol. Genet. 2000, 9, 413–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Yu, Y.-R.; You, L.-R.; Yan, Y.-T.; Chen, C.-M. Role of OVCA1/DPH1 in craniofacial abnormalities of Miller–Dieker syndrome. Hum. Mol. Genet. 2014, 23, 5579–5596. [Google Scholar] [CrossRef] [Green Version]
  131. He, J.Y.; Han, P.; Zhang, Y.; Liu, Y.D.; Song, S.J.; Feng, G.K.; An, Y.; Zhou, A.J.; Wang, H.B.; Yuan, L.; et al. Overexpression of Nogo receptor 3 (NgR3) correlates with poor prognosis and contributes to the migration of epithelial cells of nasopharyngeal carcinoma patients. J. Mol. Med. 2018, 96, 265–279. [Google Scholar] [CrossRef] [PubMed]
  132. Margiotta, A.; Progida, C.; Bakke, O.; Bucci, C. Characterization of the role of RILP in cell migration. EJH 2017, 61, 2783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Wang, Z.; Zhou, Y.; Hu, X.; Chen, W.; Lin, X.; Sun, L.; Xu, X.; Hong, W.; Wang, T. RILP suppresses invasion of breast cancer cells by modulating the activity of RalA through interaction with RalGDS. Cell Death Dis. 2015, 6, e1923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Guetg, A.; Mariotta, L.; Bock, L.; Herzog, B.; Fingerhut, R.; Camargo, S.M.; Verrey, F. Essential amino acid transporter Lat4 (Slc43a2) is required for mouse development. J. Physiol. 2015, 593, 1273–1289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic illustration of the 26 genes in the human chromosome 17p13.3 Miller–Dieker syndrome critical region.
Figure 1. Schematic illustration of the 26 genes in the human chromosome 17p13.3 Miller–Dieker syndrome critical region.
Brainsci 12 00056 g001
Figure 2. Schematic diagrams illustrating the molecular mechanisms of PAFAH1B1 (LIS1), CRK, and 14-3-3ε in regulating neuronal migration. (A) Phosphorylated NDEL1 (NUDEL) protected by 14-3-3ε from dephosphorylation. NDEL1 is phosphorylated by CDK5. Once phosphorylated, 14-3-3ε interacts with NDEL1 and protects it from the protein phosphatase 2 (PP2A)-mediated dephosphorylation. Therefore, NDEL1 could stay activated. Active NDEL1 transiently interacts with PAFAH1B1 (LIS1) and helps PAFAH1B1 (LIS1) bind to dynein, therefore promoting the movement of dynein along microtubules (MTs). However, it is unclear whether 14-3-3ε stays with dynein in the MTs. (B). PAFAH1B1 (LIS1) promotes dynein “open” conformation. When dynein is in an autoinhibited state, it has a low microtubule on rate and a reduced ability to bind to dynactin. PAFAH1B1 (LIS1) binds to “open” state dynein and stabilizes dynein at the “open” state. This allows dynein to stay at a high microtubule-binding status. Afterward, PAFAH1B1 (LIS1) dissociates from the dynein–dynactin complex. In this way, PAFAH1B1 (LIS1) promotes microtubule mediate migration by regulating dynein function. (C) CRK plays an essential role in the Reelin signaling cascade. Reelin pathways are activated by Reelin interacting with the transmembrane Reelin receptors, ApoRE2, and VLDLR. This triggers the phosphorylation of Dab1 by SFK at the inner leaflet of the plasma membrane. DAB1 then recruits CRK and activates CRK by phosphorylation. Activated CRK promotes the activation of the Crk SH3-binding guanine nucleotide-releasing/exchange factor (C3G)/Ras-proximate-1 (RAP1) pathway, which activates the cell matrix adhesion molecule α5β1 integrin, thus promoting cell migration. The illustrations were created with BioRenders.com (7 December 2021).
Figure 2. Schematic diagrams illustrating the molecular mechanisms of PAFAH1B1 (LIS1), CRK, and 14-3-3ε in regulating neuronal migration. (A) Phosphorylated NDEL1 (NUDEL) protected by 14-3-3ε from dephosphorylation. NDEL1 is phosphorylated by CDK5. Once phosphorylated, 14-3-3ε interacts with NDEL1 and protects it from the protein phosphatase 2 (PP2A)-mediated dephosphorylation. Therefore, NDEL1 could stay activated. Active NDEL1 transiently interacts with PAFAH1B1 (LIS1) and helps PAFAH1B1 (LIS1) bind to dynein, therefore promoting the movement of dynein along microtubules (MTs). However, it is unclear whether 14-3-3ε stays with dynein in the MTs. (B). PAFAH1B1 (LIS1) promotes dynein “open” conformation. When dynein is in an autoinhibited state, it has a low microtubule on rate and a reduced ability to bind to dynactin. PAFAH1B1 (LIS1) binds to “open” state dynein and stabilizes dynein at the “open” state. This allows dynein to stay at a high microtubule-binding status. Afterward, PAFAH1B1 (LIS1) dissociates from the dynein–dynactin complex. In this way, PAFAH1B1 (LIS1) promotes microtubule mediate migration by regulating dynein function. (C) CRK plays an essential role in the Reelin signaling cascade. Reelin pathways are activated by Reelin interacting with the transmembrane Reelin receptors, ApoRE2, and VLDLR. This triggers the phosphorylation of Dab1 by SFK at the inner leaflet of the plasma membrane. DAB1 then recruits CRK and activates CRK by phosphorylation. Activated CRK promotes the activation of the Crk SH3-binding guanine nucleotide-releasing/exchange factor (C3G)/Ras-proximate-1 (RAP1) pathway, which activates the cell matrix adhesion molecule α5β1 integrin, thus promoting cell migration. The illustrations were created with BioRenders.com (7 December 2021).
Brainsci 12 00056 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, X.; Bennison, S.A.; Robinson, L.; Toyo-oka, K. Responsible Genes for Neuronal Migration in the Chromosome 17p13.3: Beyond Pafah1b1(Lis1), Crk and Ywhae(14-3-3ε). Brain Sci. 2022, 12, 56. https://doi.org/10.3390/brainsci12010056

AMA Style

Liu X, Bennison SA, Robinson L, Toyo-oka K. Responsible Genes for Neuronal Migration in the Chromosome 17p13.3: Beyond Pafah1b1(Lis1), Crk and Ywhae(14-3-3ε). Brain Sciences. 2022; 12(1):56. https://doi.org/10.3390/brainsci12010056

Chicago/Turabian Style

Liu, Xiaonan, Sarah A. Bennison, Lozen Robinson, and Kazuhito Toyo-oka. 2022. "Responsible Genes for Neuronal Migration in the Chromosome 17p13.3: Beyond Pafah1b1(Lis1), Crk and Ywhae(14-3-3ε)" Brain Sciences 12, no. 1: 56. https://doi.org/10.3390/brainsci12010056

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop