Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Polymicrogyria (PMG) is a relatively common complex malformation with cortical development, characterized by an exorbitant number of abnormally tiny gyri separated by shallow sulci. It is a neuronal migration disorder. Familial cases of PMG and the manifestation of PMG in patients with chromosomal aberrations and mutations indicate their important role of genetics in this disorder. The highly stereotyped and well-conserved nature of the cortical folding pattern in humans is suggestive of the genetic regulation of the process. The chromosomal abnormalities observed in PMG include deletions, duplications, chromosomal rearrangements, and aneuploidies. Two of the most common deletions in PMG are 22q11.2 deletion and 1p36 deletion. Further, mutations in several genes such as GPR56, TUBB2B, SRPX2, PAX6, EOMES, WDR62, TUBA8, KIAA1279, and COL18A1 are known to be associated with PMG. Intriguingly, these genes are responsible only for a small number of cases of PMG. The protein products of these genes are implicated in diverse molecular and cellular functions. Taken together, PMG could be the result of the disruption of several biological pathways. Different modes of Mendelian inheritance and non-Mendelian inheritance are seen in PMG. We have suggested a gene panel that can be used for the detection of malformations of cortical development.

Free full text 


Logo of aianeurolHomeCurrent issueInstructionsSubmit article
Ann Indian Acad Neurol. 2022 Jul-Aug; 25(4): 616–626.
Published online 2022 May 5. https://doi.org/10.4103/aian.aian_97_22
PMCID: PMC9540929
PMID: 36211152

The Genetic Landscape of Polymicrogyria

Abstract

Polymicrogyria (PMG) is a relatively common complex malformation with cortical development, characterized by an exorbitant number of abnormally tiny gyri separated by shallow sulci. It is a neuronal migration disorder. Familial cases of PMG and the manifestation of PMG in patients with chromosomal aberrations and mutations indicate their important role of genetics in this disorder. The highly stereotyped and well-conserved nature of the cortical folding pattern in humans is suggestive of the genetic regulation of the process. The chromosomal abnormalities observed in PMG include deletions, duplications, chromosomal rearrangements, and aneuploidies. Two of the most common deletions in PMG are 22q11.2 deletion and 1p36 deletion. Further, mutations in several genes such as GPR56, TUBB2B, SRPX2, PAX6, EOMES, WDR62, TUBA8, KIAA1279, and COL18A1 are known to be associated with PMG. Intriguingly, these genes are responsible only for a small number of cases of PMG. The protein products of these genes are implicated in diverse molecular and cellular functions. Taken together, PMG could be the result of the disruption of several biological pathways. Different modes of Mendelian inheritance and non-Mendelian inheritance are seen in PMG. We have suggested a gene panel that can be used for the detection of malformations of cortical development.

Keywords: 22q11.2 deletion, cortical folding, GPR56, polymicrogyria, tubulins

Polymicrogyria (PMG) is a relatively common complex malformation of cortical development, characterized by an exorbitant number of abnormally tiny gyri separated by shallow sulci, leading to an irregular and lumpy cortical surface. PMG may be unilateral or bilateral, symmetrical or asymmetrical, focal, multifocal or diffuse, and may affect the whole brain or part of the brain [Table 1; Figure 1]. The most frequently affected brain region in PMG is the perisylvian cortex.

Table 1

Common polymicrogyria syndromes

TypeBrain region affectedSymmetrical or notClinical featuresIncidence of seizuresReference
Bilateral frontal polymicrogyria (BFP)Frontal regionSymmetricalDelayed motor and language milestones, mild to moderate mental retardation, seizures, spastic hemiparesis or quadriparesis,38%[1]
Bilateral frontoparietal polymicrogyria (BFPP)Frontal and parietal lobesSymmetricalGlobal developmental delay, moderate to severe intellectual disability, dysconjugate gaze, ataxia, finger dysmetria, seizures94%[1]
Bilateral perisylvian polymicrogyria (BPP)Perisylvian cortexUsually, symmetricalPseudobulbar palsy with diplegia of the facial, pharyngeal, and masticatory muscles, dysphagia, dysarthria, mild to severe intellectual disability, seizures90%[1]
Bilateral parasagittal parieto-occipital polymicrogyria (BPPOP)Parasagittal and mesial aspects of the parieto-occipital cortexSymmetricalIQ scores ranging from average to mild mental retardation, cognitive slowing, seizuresNot available[1]
Bilateral generalized polymicrogyria (BGP)Generalized distribution; most severe in the perisylvian regionsSymmetricalCognitive and motor delay, spastic hemiparesis or quadriparesis, seizuresNot available[1]
Unilateral polymicrogyriaDifferent cortical areasAssymetricalVariable degree of mental retardation, spastic hemiparesis with primary involvement of the upper extremity, seizuresNot available[1]
An external file that holds a picture, illustration, etc.
Object name is AIAN-25-616-g001.jpg

MRI images of various patterns of polymicrogyria. Source: http://www.genereviews.org/. Copyright © 1993-2022 University of Washington

PMG is a neuronal migration disorder wherein the normal process of cerebral cortical development is perturbed in the late stage of neuronal migration or in the early stage of cortical organization.[2] The signs and symptoms of PMG vary, based on the brain region and how much surface of the brain is involved [Table 1]. PMG previously remained underdiagnosed owing to the limitations in imaging techniques. However, the recent use of high-resolution radioimaging approach has paved the way to improved diagnosis and classification. PMG can occur in association with other malformations of the brain such as microcephaly, megalencephaly, ventriculomegaly, and grey matter heterotopia.[3] In children, the clinical manifestations of PMG include delayed motor and language milestones, mild/moderate mental retardation, spastic hemiparesis/quadriparesis, and epilepsy.[1] It has been suggested that PMG is the culmination of a range of aberrations of cortical development.[4]

The incidence and prevalence of PMG are not accurately known due to the etiological and clinical heterogeneity of PMG. There is very little information on the factors leading to the development of PMG. Some of the factors that have been suggested to be associated with PMG include intrauterine cytomegalovirus infection,[5] maternal drug ingestion (Barkovich et al.,[6] 1995), fetal cerebral ischemia from placental perfusion failure, a twin-twin transfusion,[7] loss of a twin in utero,[8] mutations,[3] and chromosomal aberrations.[9]

The occurrence of familial cases of PMG,[10,11] the manifestation of PMG in patients with chromosomal abnormalities and mutations,[12,13,14,15] and the association of PMG with genetically determined syndromes such as Zellweger,[16] Aicardi,[17] and Walker-Warburg syndrome[18]- all indicate an important role of genetic factors in the pathogenesis of PMG. Efforts to elucidate the molecular genetic underpinnings of PMG are underway.

GENETIC UNDERPINNINGS OF CORTICAL FOLDING

Cortical folding is the process by which the massive cortex is fitted into a limited cranial volume. This occurs during embryonic development and is essential for the optimization of brain wiring and functional organization of the brain.[19] The intricate mechanisms underlying the gyrification of the human cerebral cortex have been a topic of immense interest to neurobiologists. Understanding this mechanism is key to recognizing the functional organization of the brain and how it is affected in neurological disorders. The highly stereotyped and well-conserved nature of cortical folding patterns in humans is suggestive of strong genetic regulation of the process.[19,20]

In mice and humans, the fibroblast growth factor (FGF) signaling has been implicated in cortical development, gyrification, and patterning. Fgf2 has been demonstrated to induce cortical gyrification in mice brains.[21] In humans, strong activating mutations in FGFR3 caused expansion of the surface area of the occipitotemporal region leading to excessive gyrification.[22] The WNT–β-catenin pathway (also referred to as canonical WNT signaling) is also shown to be important in gyrification.[23,24] It was shown to regulate rostral-caudal and medial-lateral patterning in the developing cortex, and in establishing the radial inside to outside organization of the cerebral cortex.[25] The hominoid-specific gene TBC1D3 was shown to promote the generation of basal neural progenitor cells and induce cortical folding.[26] The gene GPR56, which encodes a member of the G protein-coupled receptor family, is known to regulate cortical progenitor proliferation and cortical patterning.[27] A 15-base pair deletion in the regulatory region of GPR56, which results in dysregulation of GPR56 expression, disrupts human cortical folding around the Sylvian fissure.[28] Heterozygous missense mutations in TUBB3, which encodes beta-tubulin isotype 3, a neuron-specific component of microtubules, are reported to cause dysmorphic cortical folding patterns.[29] Mutations in the regulatory region of human T-box transcription factor TBR2 have adverse effects on cortical size and folding.[30] In gyrencephalic mammals, the expression of Cdk5 in the upper-layer neurons was found to be essential for the efficient folding of the cerebral cortex.[31]

ROLE OF GENETICS IN PMG

Familial recurrence has been reported for several types of PMG, especially, bilateral frontoparietal PMG[32] and bilateral perisylvian PMG.[33] Chromosomal abnormalities (deletions and duplications) and gene mutations have been reported in PMG patients.

CHROMOSOMAL ABNORMALITIES

Cytogenetic and molecular genetic studies in 29 patients with PMG identified several candidate loci in 1p36.3, 2p16.1-p23, 4q21.21-q22.1, 6q26-q27, and 21q21.3-q22.1, 1q44 and 18p. However, most of these loci demonstrated incomplete penetrance and variable expressivity.[9] Deletions, duplications, chromosomal rearrangements, and chromosomal aneuploidies have been observed in PMG.

Deletions and duplications: Two of the most common deletions in PMG, 22q11.2 deletion and 1p36 deletion, are discussed in detail below. Other deletions and duplications identified in PMG are listed in Table 2.

Table 2

Deletions and duplications associated with polymicrogyria

Type of PMGChromosomal abnormalityNumber of patientsReference
Symmetric perisylvian PMGDeletion of 1q441[9]
Bilateral centro-temporo-parietal PMG, periventricular nodular heterotopia, arachnoid cyst, hypoplasia of falx, and agenesis of the corpus callosumDeletion of 2q31-2q331[34]
Symmetric perisylvian PMGDeletion of 4q21.21-q22.12[9]
Extensive PMGDeletion of 6q26-q277[9]
Bilateral frontotemporal PMG and left parietooccipital PMGDeletion of 13q14.1-13q31.21[35]
Symmetric perisylvian PMGDeletion of 18p1[9]
Symmetric perisylvian PMGDuplication of 2p16.1-p23.12[9]
Bilateral perisylvian PMGDuplication of 17p13.3-p13.21[36]

PMG: Polymicrogyria

22q11.2 deletion: Deletion 22q11.2 (DEL22q11.2), with a reported incidence as high as 1:4000 live births, is one of the most common microdeletions in humans.[37] Among the various brain malformations described with DEL22q11.2, PMG is the most frequently reported condition.[35] DEL22q11.2 is the most common chromosomal abnormality associated with PMG. However, owing to the low penetrance and variable expressivity of this microdeletion, the underlying molecular mechanisms are unknown.[38]

In DEL22q11.2, PMG is observed mostly in the perisylvian areas and most often is asymmetrical with a predisposition for the right hemisphere of the brain.[39] This asymmetric presentation has led to the hypothesis that the candidate genes of PMG may be asymmetrically expressed between the two hemispheres of the brain.[38] In a fetal brain, the haploinsufficiency of a gene, that could be responsible for cortical development, in this deleted region could lead to PMG.[38] However, there is no strong functional candidate gene, within or around the critical deleted region, currently known to be associated with PMG. Another hypothesis suggests asymmetric hypoperfusion of the developing brain, resulting from the haploinsufficiency of a gene expressed in the fetal vascular brain tissue.[38] Bingham[40] postulated that DEL22q11.2 may influence one or more of the predisposing factors of PMG such as fetal cerebral vasculature, a tendency toward thrombosis, regulation of energy metabolism, and post-migrational cortical maturation. In children with DEL22q11.2 syndrome, brain magnetic resonance imaging (MRI) should be performed to look for PMG. The prognosis might depend on the presence of PMG.[39]

1p36 deletion: PMG has been reported in patients with 1p36 monosomy.[41] However, this shows only incomplete penetrance.[42] Even though 1p36 harbors several genes involved in brain development or function, the gene whose insufficiency leads to PMG has not yet been identified. Some of the potential candidate genes in this region are TP73, PEX10, SKI, GABRD, GNB1, PRKCZ, MMP23B, MMP23A, and PRDM16.[42,43] 1p36 duplications are rare. The smallest 1p36 microduplication was reported in a patient with bilateral perisylvian PMG.[44] Functional validation studies of the genes in this duplicated region revealed that ENO1 could be the most likely causative gene for the brain malformation.[44]

Chromosomal rearrangements: The following unbalanced rearrangements have been observed in PMG:

  • Deletion of segment 1q44-1qter and duplication of segment 12p13.3-12pter in two patients with unilateral perisylvian PMG[45]

  • Deletion of segments 18p11.2-18pter and 21pter-21q22.1 in a patient with localized PMG[46]

  • Duplication of segments 9pter-9q22.2 and 7q35-7qter in a patient with bilateral parietooccipital PMG and caudal hypoplasia of cerebellar vermis[47]

A balanced translocation by breakpoints at 1p12 and 6p12.2 that leads to locus disruption was observed in a patient with bilateral perisylvian PMG, bilateral periventricular nodular heterotopia, and hypoplasia of corpus callosum.[48]

Chromosomal aneuploidies: Trisomy 13 in bilateral perisylvian PMG,[49] duplication of the short arm of the X chromosome in unilateral hemispheric PMG,[50] and turner mosaicism in bilateral frontal PMG[51] have been reported.

GENE MUTATIONS ASSOCIATED WITH PMG

Different modes of Mendelian inheritance such as autosomal dominant,[10] autosomal recessive,[52,53] and X-linked[54,55] are seen in PMG. Several genes including GPR56, TUBB2B, SRPX2, PAX6, EOMES, WDR62, TUBA8, COL18A1, and KIAA1279 are known to be associated with PMG [Table 3]. The proteins products of these genes are involved in diverse molecular and cellular functions. Thus, PMG could be the result of the disruption of several biological pathways. Some of the well-studied genes are discussed below.

Table 3

Genes associated with polymicrogyria

Type of PMGGeneMode of inheritanceReference
Bilateral frontoparietal PMG ADGRG1, GPR56 AR[27,56]
Bilateral perisylvian PMG PIK3R2 De novo [57]
SRPX2 Maternal[58]
Bilateral temporooccipital PMG FIG4 AR[59]
Bilateral asymmetric PMG TUBB2B AD[13]
Bilateral asymmetric PMG and lissencephaly TUBA1A, TUBB2B De novo [61,62,63]
Bilateral PMG MECP2 De novo [63]
Unilateral PMG PAX6 Maternal[64]
PMG and lissencephaly TUBA1A AD[65]
PMG and agenesis of corpus callosum, microcephaly EOMES AD[30]
PMG and microcephaly WDR62, RTTN AR[66,67]
PMG and optic nerve hypoplasia TUBA8 AR[68]
PMG and band-like calcification OCLN AR[14]
PMG and Warburg Micro syndrome RAB18 AR[69]
PMG and Goldberg-Shprintzen syndrome KIAA1279 AR[70]
PMG and CK syndrome NSDHL X-linked[71]
PMG and Knobloch syndrome COL18A1 AR[72]
Warburg Micro syndrome (characterized by ocular and neurodevelopmental abnormalities, including PMG) RAB3GAP AR[73]

PMG: Polymicrogyria; AD: Autosomal dominant; AR: autosomal recessive

GPR56: GPR56 is an orphan G protein-coupled receptor that regulates the migration of neural progenitors via a G alpha 12/13 and Rho pathway, suggesting its importance in the development of the central nervous system (CNS).[74] Autosomal recessive mutations in the GPR56 gene located on chromosome 16q13 are shown to be associated with bilateral frontoparietal PMG.[27,75] GPR56 expression levels are suggested to regulate cortical progenitor proliferation by affecting neuronal migration and cell fate.[27] GPR56 mutations tend to disrupt human cortex folding around the Sylvian fissure.[28] A homozygous germline nonsense mutation (p.R271*) in GPR56 contributes to phenotypic variability in bilateral frontoparietal PMG.[76] Intriguingly, GPR56 knockout mice suggest that GPR56-related bilateral frontoparietal PMG shares certain features of the cobblestone brain malformation.[75] Detailed studies are needed to elucidate the range of phenotypic features associated with GPR56 mutations.

Tubulins: Tubulins, present in the postmitotic cells of the human CNS, are involved in the formation of microtubules that are vital for neuronal proliferation, migration, differentiation, and axonal guidance during cortical development.[77] Mutations of tubulin genes (TUBA1A, TUBB2B, TUBA8) are seen in PMG.[13,60,65,68] Neuropathological examination of the pattern of PMG in a 27-gestational-week fetus with TUBB2B mutation revealed ruptures in the pial basement membrane, cerebellar nodular heterotopias, over-migration of neurons, and disorganization of radial glial fibers.[13] The cortical malformations associated with mutations of tubulin genes encompass a broad spectrum of cerebral anomalies ranging from lissencephalic to polymicrogyric cortical dysplasias, suggesting common underlying pathogenic mechanisms that involve microtubular function, reinforcing the role of microtubule-related proteins in cortical development.[78,79]

PMG is strongly associated with epilepsy, the incidence being 33% to 87%.[80] Mutations in the tubulin genes TUBAA1, TUBB2B, TUBB3 are known to be associated with epilepsy. Mutations of TUBA1A and TUBB2B genes are most frequently associated with epilepsy compared with TUBB3 mutations. When the distribution of epilepsy-associated mutations in these genes was examined, the mutations in TUBB2B were equally distributed, while the N-terminal domain harbored most of the mutations in TUBA1A.[81] Structural abnormalities of the cortex, fractured pyramidal layer of the hippocampus, and defects due to impairment of neuronal migration were revealed in histological examinations of Tuba1a-mutant mice brains.[82,83] TUBB2B mutations are known to affect tubulin heterodimer folding and incorporation into microtubules, leading to impairments in neuronal migration, radial glia dysfunction, and impaired cortical development.[13] These malformations of cortical development are recognized as a cause of early-onset epilepsy.[84] Epileptogenicity associated with tubulin gene mutations is a complex phenomenon, depending on the mechanism by which the mutations alter the dynamic properties and functions of microtubules. Epilepsy frequently occurs when the mutations lead to malformations of cortical development, while it is less frequent when mutations are associated with axonal guidance disorders. Early-onset epilepsy is often generalized and considered as an expression of diffuse cortical malformation.[85] A role of TUBB3 in regulating epileptic seizures via GABA-A receptor-mediated synaptic transmission has been reported, explaining the rare epileptic cases in patients with TUBB3 mutations.[86]

SRPX2: SRPX2, expressed in the neurons of the adult human brain, is a secreted protein that modulates synapse density.[87] SRPX2 serves as a target of the FOXP2 transcription factor and is associated with epilepsy and language development[58,88]. Mutations in SRPX2 have been linked to bilateral perisylvian PMG.[58] In the developing rat cerebral cortex, Srpx2 downregulation led to the altered position of projection neurons.[89] Detailed studies into the role of SRPX2 in neuronal migration and brain development are warranted.

PAX6: PAX6 is a developmentally regulated highly conserved gene. Located on 11p13, this gene encodes a transcription factor. In an MRI study involving 24 subjects heterozygous for PAX6 mutations, Mitchell et al.[64] observed widespread structural abnormalities including unilateral PMG and absence of the pineal gland. Extensive PMG was reported in an infant who was a compound heterozygote for two of PAX6.[90] Being a transcription factor, PAX6 could be involved in regulating gene expression during corticogenesis.[91,92] Murine models have demonstrated a significant role of PAX6 in brain development. Pax6 plays a key role in the dorsoventral patterning of telencephalon,[93] modulating the proliferation and differentiation of neural progenitor cells,[94,95,96] specification of neuronal identity,[97] radial glia differentiation,[94] and neuronal migration.[98] PAX6 mutations could lead to disruption of these functions leading to PMG.

TBR2 (EOMES): TBR2 is a transcriptional factor that serves an important role during development. It is a crucial modulator of neurogenesis in the subventricular zone.[99] It is involved in neuronal proliferation and migration,[30] and in implementing regional identity in the cortex.[100] In humans, a homozygous chromosomal translocation that disrupts the expression of TBR2 was found to be associated with microcephaly, PMG, agenesis of the corpus callosum, cognitive deficits, and severe motor delay.[30] Tbr2 is known to regulate the transcriptome by upregulating or downregulating the expression of numerous target genes.[101] Interestingly, the TBR2 protein acts immediately downstream of PAX6 and regulates the division of intermediate progenitor cells. Pax6, Tbr2, and Tbr1 are expressed sequentially by the radial glia, intermediate progenitor cells, and postmitotic neurons in the developing neocortex.[102] This sequential expression suggests that each of these transcription factors regulates specific steps in neuron differentiation. The altered fate of neurons due to mutations of TBR2 can be one of the causes of PMG.[9]

WDR62: The protein encoded by WDR62 localizes to the centrosome and the nucleus, depending on the cell type and cell phase.[103] Recessive mutations of WDR62 are identified as one of the causes of a broad range of severe cerebral cortical malformations including PMG, pachygyria, lissencephaly, microcephaly, schizencephaly, and cerebellar hypoplasia.[104] Downregulation of Wdr62 in mice has revealed neuronal migration abnormalities.[105] WDR62 transcripts and protein are abundant in the neural progenitors in ventricular and subventricular zones.[104] Thus, progenitor abnormalities have been suggested as a cause of PMG.

RAB3GAP: RAB3GAP is a heterodimeric complex that serves as a guanine-nucleotide exchange factor for RAB18. Mutations of RAB18 cause Warburg Micro syndrome, which is characterized by several neurodevelopmental abnormalities, including PMG.[73,106] The Rab proteins are key regulators of vesicular membrane transport in both the exocytic and endocytic pathways.[107] Members of the Rab3 subfamily have been implicated in the regulated exocytosis of neurotransmitters and hormones.[108] But how this contributes to PMG is yet to be elucidated.

KIAA1279: The KIAA1279 gene encodes a ubiquitously expressed protein that interacts with the microtubules. A homozygous nonsense mutation in KIAA1279 causes Goldberg-Shprintzen syndrome, which is characterized by an enteric nervous disorder, mental retardation, microcephaly, and bilateral generalized PMG.[109,110] KIAA1279 is found to be involved in neurite outgrowth.[109]

DEEP SEQUENCING APPROACH

Recently, to identify the monogenic causes of PMG, Stutterd et al.[111] recruited 123 patients with PMG for a deep sequencing study using gene panels containing 377 known candidate genes for malformations of cortical development. Pathogenic or likely pathogenic variants were identified in 20.3% of the patients. TUBA1A and PIK3R2 were the most common causal genes. The other causal genes were NEDD4L, PIK3CA, COL4A1, COL4A2, GRIN2B, GPSM2, TUBB3, WDR62, TUBB2B, FH, and ACTG1.

Among the causative/candidate variants, 30.7% were in the tubulin-related genes (TUBA1A, TUBB2B, TUBB3 causative variants identified in the), and the patients harboring a tubulin gene variant were diagnosed with tubulinopathies. The mTOR-PI3K-AKT pathway was the second most frequently implicated molecular pathway, with causative variants identified in the PIK3R2 or PIK3CA genes in 26.9% of the patients who were diagnosed with mTORopathies. The PIK3R2 variant p.Gly373Arg itself constituted 20% of all the identified variants. Collagenopathies were diagnosed in 2.4% of the patients harboring causative variants in COL4A1 and COL4A2.

Between the male and female patients, there was no significant difference in the genetic causes identified. The possibilities of identifying a genetic cause were high in patients with abnormal head growth. A genetic cause could be identified in 58.3% of the patients with macrocephaly and 30.8% of the patients with microcephaly. The diagnostic yield of 20.3% shows that PMG may be associated with non-genetic factors, somatic mutations that are brain-specific, or other genes that were not included in the gene panel.

DE NOVO MUTATIONS

De novo variations have been identified in PMG. In a whole-exome sequencing (WES) study of 124 patients with PMG, de novo variations were observed in the ATP1A3 gene of eight patients. These patients had a severe form of PMG with developmental delay and epilepsy.[112] In mice, Atp1a3 variants were associated with defects in brain architecture.[112] In another WES study of 57 PMG patients, de novo mutations in GRIN1 were observed in 11 patients.[113] GRIN1 encodes an essential subunit of the N-methyl-D-aspartate (NMDA) receptor. The PMG-associated GRIN1 mutations significantly altered the in vitro activity of the receptor, denoting an important role of NMDA signaling in the pathogenesis of PMG.[113] A de novo PIK3R2 variation has been reported in a patient with asymmetrical bilateral PMG.[114] De novo variants of PIK3R2, which is an upstream component of the mTOR pathway, have also been identified in patients with megalencephaly–polymicrogyria–polydactyly–hydrocephalus syndrome (MPPH).[115] In mice, Pik3r2 mutations were associated with brain overgrowth.[116] Interestingly, de novo variants of CCND2 have been identified as another pathogenic cause of brain overgrowth and severe cortical malformations including MPPH. The variants can lead to pathological stabilization of cyclin D2, which in turn lead to malformations in the developing cerebral cortex.[117] It has been suggested that mutations of PIK3R2 and CCND2 result in a common functional endpoint- increased cyclin D2 activity in neural precursors leading to cortical malformations.[117] De novo variations in MAPK8IP3, which is a crucial component of the retrograde axonal transport system, are associated with intellectual disability and brain anomalies such as perisylvian PMG, cerebral/cerebellar atrophy, and hypoplasia of the corpus callosum.[118] A recurrent de novo BICD2 missense mutation is associated with PMG and severe arthrogryposis.[119] A novel de novo DDX3X missense variant was reported in a female with delayed psychomotor development, delayed myelination, brachycephaly, and PMG.[120] Ddx3x regulates neuron generation and cortical development. Pathogenic DDX3X mutations lead to impaired RNA helicase activity, stimulate ectopic RNA-protein granules in neurons, and disrupt translation.[121] In the aforementioned deep sequencing study, de novo variations were observed in ACTG1, COL4A1, GRIN2B, NEDD4L, PIK3R2, TUBA1A, TUBB2B, and TUBB3.[111]

SOMATIC MOSAICISM

Somatic mosaicism has been reported in PMG. In the deep sequencing study of 123 PMG patients, five variants (c.1117G>A variant of PIK3R2 in three patients, and two PIK3CA variants) were mosaic with allele fractions less than 0.33, the lowest allele fraction being 0.09.[111] Mosaic mutations in the PIK3R2 gene are associated with bilateral perisylvian PMG.[57] Megalencephaly-capillary malformation-polymicrogyria syndrome (MCAP) is caused by somatic mosaicism of the PIK3CA gene.[122] Mosaic trisomy of chromosome 1q in the human brain is associated with unilateral PMG, very early-onset focal epilepsy, and severe developmental delay.[123]

Identification of somatic mosaicism is extremely challenging. Recent advances in high-depth next-generation sequencing (NGS) have made possible the detection of disease-associated somatic mosaicism.[124] The detection requires high depth sequence coverage, preferably 500× (giving alternate-allele coverage of >20×, assuming the allele is present in ~ 10% of cells), as against the 40 to 80× coverage of clinical exome sequencing or WES. Deep sequencing using targeted gene panels are a good option in this regard as they facilitate a higher depth of coverage and cost-efficient detection of somatic mosaicism.[125]

COPY NUMBER VARIATIONS

PMG has been observed in genetic syndromes associated with copy number variations (CNVs), such as the 22q11.2 deletion syndrome.[38] Dobyns et al.[9] identified six PMG loci associated with pathogenic CNVs (1p36.3, 2p16.1-p23.1 4q21.21-q22.1, 6q26-q27, 21q21.3-q22.1, 22q11.2 and another five possible loci (1q44, 2p15-p16.1, 11q12-q13, 13q14.1-q31.2, 18p). Two patients were included in the deep sequencing study of Stutterd et al.[111] had chromosome copy number variants of uncertain significance that could be relevant to their PMG. Kobow et al.[123] identified a somatic duplication of the entire long arm of chromosome 1 in the surgical brain tissue of 7 out of 26 PMG patients studied. Duplication of 2p16 is associated with perisylvian PMG.[126] Gene pathway analysis suggested several developmentally relevant genes and gene clusters in this region. Further, a rare locus for PMG was narrowed to a region of 2p16.1-p16.3, which contained 23 genes that expressed in the cerebral cortex during fetal development. Several of the duplicated genes contributed to neurodevelopmental pathways including cytokine, growth factor, and hormonal signaling, modulation of cell cycle progression, neuronal migration, and axonal guidance.[126] Microduplication of 22q11.2 and inverted 9p duplication/deletion was observed in a child with asymmetrical PMG predominantly affecting the right occipital lobe.[127]

WHAT DO THE GENES TELL US ABOUT THE PATHOGENESIS OF PMG?

PMG is a disorder of neuronal migration in which normal cortical development is disrupted during the later stages of neuronal migration or the earlier stages of cortical organization.[2] Precise tuning of neuronal proliferation, differentiation, migration, and connectivity are pivotal in the normal development of the cerebral cortex.[128] PMG can be considered as the culmination of multiple diverse pathological processes, occurring at different time points during cortical development. It can result from mutations in multiple genes or different mutations in the same gene with pleiotropic effects. The low/incomplete penetrance of mutations and variable expressivity pose problems in understanding the underlying molecular mechanisms. Further, it needs to be noted that these genes still account for only a small percentage of the cases of PMG. The potential influence of epigenetic factors (e.g., DNA methylation, histone acetylation, microRNAs) in the pathogenesis of PMG has not yet been studied. Since epigenetic processes play a crucial role during cortical development by regulating neurogenesis, proliferation, and cell specification,[129,130,131] a study into the role of epigenetic factors in PMG could constitute a topic for future research.

TREATMENT AND MANAGEMENT OF PMG

PMG is diagnosed by brain MRI, which will reveal the abnormalities in brain structure, and the location and severity of the abnormality. In individuals with seizures, an electroencephalogram will help to confirm the type of seizure. The possibilities of genetic testing should also be explored by a detailed study of the medical history of the patient and his/her family members. Knowing the cause of the disease might help to formulate efficient therapeutic strategies.

PMG is a lifelong disorder. The brain malformation cannot be reversed, but the symptoms may be treated in children and adults. Thus, only symptomatic treatment is possible for PMG at present. The first-line treatment for those with seizures is anti-seizure medications. Individuals with unilateral PMG affecting only a small region could be considered for resective surgery. Advanced neuroradiologic and neurophysiologic techniques are required to provide an effective and safe resection of the epileptogenic cortex.[132] Patients with bilateral PMG are not good candidates for resective surgery. Occupational therapy, physiotherapy, and speech therapy may help some of the affected individuals.

GENETIC TESTING AND COUNSELING FOR PMG

A flow chart that provides the guidelines for genetic testing and counseling of families with brain malformations is provided in Figure 2. Familial PMG has shown an autosomal dominant, autosomal recessive, or X-linked mode of inheritance. In such familial cases, risk assessment and genetic counseling depend on the genetic cause of PMG in the affected individual. The phenotypic heterogeneity of PMG, low/incomplete penetrance of mutations, and the occurrence of de novo variants, somatic mosaicism, CNVs challenge the conventional genetic testing strategies and have important implications for genetic counseling. If a typical Mendelian inheritance pattern is observed in the family of the proband, a WES approach will help to identify the inherited monogenic causative variant. On the other hand, if a Mendelian pattern of inheritance is not observed in the family, the possibilities of de novo variants, CNVs, or somatic mosaicism should be looked into. CNVs can be tested using chromosomal microarrays, while mosaicism can be tested by deep sequencing. Table 4 shows a suggested gene panel for the NGS testing of malformations of cortical development.

An external file that holds a picture, illustration, etc.
Object name is AIAN-25-616-g002.jpg

A flow chart for genetic testing and counseling of families with brain malformations

Table 4

A gene panel for the genetic testing of malformations of cortical development

ACTBACTG1ADGRG1AKT3ARHGAP31ARFGEF2ARXASPM
ATP6V0A2B3GALNT2B4GAT1BMPERCCDC88ACCND2CITCOL18A1
CPT2CUL4BDAG1DCHS1DCXDOCK6DYNC1H1EML1
ERCC1FAT4FHFIG4FKRPFKTNFLNAGMPPB
GPHNGPSM2HSD17B41BA57ISPDKATNB1KIAA0586KIAA1279
KIF2AKIF5CKLHL15LAGE3LAMB1LAMC3LARGE1MBOAT7
MECP2MTORNANSNDE1NEDD4LNSDHLOCLNPAFAH1B1
PAX6PEX1PEX13PEX2PEX26PI4KAPIK3CAPIK3R2
POMGNT1POMGNT2POMKPOMT1POMT2PQBP1RAB18RAB3GAP1
RAB3GAP2RAC1RELNRTTNSF3B4SMOSNAP29SRD5A3
SRPX2TBC1D20TCTN2TMEM5TMTC3TP53RKTUBA1ATUBA8
TUBBTUBB2ATUBB2BTUBB3TUBB4ATUBBG1USP18VLDLR
WDR62

CONCLUSIONS

Despite several clinical and pathological studies, there is very limited understanding of the etiological mechanisms of PMG. It has not been possible to identify the primary brain insult that leads to each type of PMG. Studies indicate that disruption of neuronal migration and differentiation could lead to abnormalities of cortical gyration. In-depth molecular genetic studies will further shed light on the molecular basis of normal cortical development, contribute to genotype-phenotype correlations in PMG, and determine whether the different types of PMG are genetically heterogeneous or share a common molecular mechanism. We have suggested a panel of 96 genes that can be used for targeted NGS to detect the malformations of cortical development. Some of the important genes included in the panel are ADGRG1, AKT3, GPSM2, KIAA1279, LAMC3, OCLN, RAB18, TBC1D20, tubulins, and WDR62. This will help to improve the diagnosis and treatment of PMG and can be made use of in early detection and genetic counseling. The integration of advanced neuroimaging, genetics, and animal model studies will assist in finding answers to the many unanswered questions.

Financial support and sponsorship

This work was funded by the Indian Council of Medical Research (ICMR) Task Force on Rare Diseases (33/11/2019-TF/Rare/BMS).

Conflicts of interest

There are no conflicts of interest.

REFERENCES

1. Jansen A, Andermann E. Genetics of the polymicrogyria syndromes. J Med Genet. 2005;42:369–78. [Europe PMC free article] [Abstract] [Google Scholar]
2. Barkovich AJ, Guerrini R, Kuzniecky RI, Jackson GD, Dobyns WB. A developmental and genetic classification for malformations of cortical development: Update 2012. Brain J Neurol. 2012;135:1348–69. [Europe PMC free article] [Abstract] [Google Scholar]
3. Parrini E, Conti V, Dobyns WB, Guerrini R. Genetic basis of brain malformations. Mol Syndromol. 2016;7:220–33. [Europe PMC free article] [Abstract] [Google Scholar]
4. Squier W, Jansen A. Polymicrogyria: Pathology, fetal origins and mechanisms. Acta Neuropathol Commun. 2014:2. 10.1186/s40478-014-0080-3. [Europe PMC free article] [Abstract] [Google Scholar]
5. Bosnjak VM, Daković I, Duranović V, Lujić L, Krakar G, Marn B. Malformations of cortical development.t in children with congenital cytomegalovirus infection—A study of nine children with proven congenital cytomegalovirus infection. Coll Anthropol. 2011;35(Suppl 1):229–34. [Abstract] [Google Scholar]
6. Barkovich AJ, Rowley H, Bollen A. Correlation of prenatal events with the development of polymicrogyria. Am J Neuroradiol. 1995;16(Suppl 4):822–7. [Europe PMC free article] [Abstract] [Google Scholar]
7. Sugama S, Kusano K. Monozygous twin with polymicrogyria and normal co-twin. Pediatr Neurol. 1994;11:62–3. [Abstract] [Google Scholar]
8. Bogaert PV, Donner C, David P, Rodesch F, Avni EF, Szliwowski HB. Congenital bilateral perisylvian syndrome in a monozygotic twin with intra-uterine death of the co-twin. Dev Med Child Neurol. 1996;38:166–70. [Abstract] [Google Scholar]
9. Dobyns WB, Mirzaa G, Christian SL, Petras K, Roseberry J, Clark GD, et al. Consistent chromosome abnormalities identify novel polymicrogyria loci in 1p36.3, 2p16.1-p23.1, 4q21.21-q22.1, 6q26-q27, and 21q2. Am J Med Genet A. 2008;146A:1637–54. [Europe PMC free article] [Abstract] [Google Scholar]
10. Guerreiro MM, Andermann E, Guerrini R, Dobyns WB, Kuzniecky R, Silver K, et al. Familial perisylvian polymicrogyria: A new familial syndrome of cortical maldevelopment. Annals Neurol. 2000;48:39–48. [Abstract] [Google Scholar]
11. Chang BS, Apse KA, Caraballo R, Cross JH, Mclellan A, Jacobson RD, et al. A familial syndrome of unilateral polymicrogyria affecting the right hemisphere. Neurology. 2006;66:133–5. [Abstract] [Google Scholar]
12. Sztriha L, Guerrini R, Harding B, Stewart F, Chelloug N, Johansen JG. Clinical, MRI, and pathological features of polymicrogyria in chromosome 22q11 deletion syndrome. Am J Med Genet A. 2004;127A:313–7. [Abstract] [Google Scholar]
13. Jaglin XH, Poirier K, Saillour Y, Buhler E, Tian G, Bahi-Buisson N, et al. Mutations in the beta-tubulin gene TUBB2B result in asymmetrical polymicrogyria. Nat Genet. 2009;41:746–52. [Europe PMC free article] [Abstract] [Google Scholar]
14. O'Driscoll MC, Daly SB, Urquhart JE, Black GC, Pilz DT, Brockmann K, et al. Recessive mutations in the gene encoding the tight junction protein occludin cause band-like calcification with simplified gyration and polymicrogyria. Am J Hum Genet. 2010;87:354–64. [Europe PMC free article] [Abstract] [Google Scholar]
15. Valence S, Poirier K, Lebrun N, Saillour Y, Sonigo P, Bessières B, et al. Homozygous truncating mutation of the KBP gene, encoding a KIF1B-binding protein, in a familial case of fetal polymicrogyria. Neurogenetics. 2013;14:215–24. [Abstract] [Google Scholar]
16. Mohebbi MR, Rush ET, Rizzo WB, Banagale RC. Zellweger syndrome and associated brain malformations: Report of a novel peroxin1 (pe×1) mutation in a native American infant. J Child Neurol. 2012;27:1589–92. [Abstract] [Google Scholar]
17. Hopkins B, Sutton VR, Lewis RA, Van den Veyver I, Clark G. Neuroimaging aspects of Aicardi syndrome. Am J Med Genet A. 2008;146A:2871–8. [Europe PMC free article] [Abstract] [Google Scholar]
18. Barkovich AJ. Neuroimaging manifestations and classification of congenital muscular dystrophies. Am J Neuroradiol. 1998;19:1389–96. [Europe PMC free article] [Abstract] [Google Scholar]
19. Fernández V, Llinares-Benadero C, Borrell V. Cerebral cortex expansion and folding: What have we learned? EMBO J. 2016;35:1021–44. [Europe PMC free article] [Abstract] [Google Scholar]
20. Mallela AN, Deng H, Bush A, Goldschmidt E. Different principles govern different scales of brain folding. Cereb Cortex. 2020;30:4938–48. [Abstract] [Google Scholar]
21. Rash BG, Tomasi S, Lim HD, Suh CY, Vaccarino FM. Cortical gyrification induced by fibroblast growth factor 2 in the mouse brain. J Neurosci. 2013;33:10802–14. [Europe PMC free article] [Abstract] [Google Scholar]
22. Hevner RF. The cerebral cortex malformation in thanatophoric dysplasia: Neuropathology and pathogenesis. Acta Neuropathol. 2005;110:208–21. [Abstract] [Google Scholar]
23. Chinnappa K, Márquez-Galera Á, Prieto-Colomina A, Nomura Y, Cárdenas A, López-Atalaya JP, et al. MIR3607 regulates cerebral cortex development via activation of Wnt/ßCat signaling. BioRxiv. 2019 10.1101/729939. [Google Scholar]
24. Chizhikov VV, Iskusnykh IY, Steshina EY, Fattakhov N, Lindgren AG, Shetty AS, et al. Early dorsomedial tissue interactions regulate gyrification of distal neocortex. Nat Commun. 2019:10. 10.1038/s41467-019-12913-z. [Europe PMC free article] [Abstract] [Google Scholar]
25. Chenn A. Wnt/β-catenin signaling in cerebral cortical development. Organogenesis. 2008;4:76–80. [Europe PMC free article] [Abstract] [Google Scholar]
26. Ju XC, Hou QQ, Sheng AL, Wu KY, Zhou Y, Jin Y, et al. The hominoid-specific gene TBC1D3 promotes generation of basal neural progenitors and induces cortical folding in mice. ELife. 2016:5. 10.7554/eLife. 18197. [Europe PMC free article] [Abstract] [Google Scholar]
27. Piao X, Hill RS, Bodell A, Chang BS, Basel-Vanagaite L, Straussberg R, et al. G protein-coupled receptor-dependent development of human frontal cortex. Science. 2004;303:2033–6. [Abstract] [Google Scholar]
28. Bae BI, Tietjen I, Atabay KD, Evrony GD, Johnson MB, Asare E, et al. Evolutionarily dynamic alternative splicing of GPR56 regulates regional cerebral cortical patterning. Science. 2014;343:764–8. [Europe PMC free article] [Abstract] [Google Scholar]
29. Whitman MC, Andrews C, Chan WM, Tischfield MA, Stasheff SF, Brancati F, et al. Two unique TUBB3 mutations cause both CFEOM3 and malformations of cortical development. Am J Med Genet A. 2016;170:297–305. [Europe PMC free article] [Abstract] [Google Scholar]
30. Baala L, Briault S, Etchevers HC, Laumonnier F, Natiq A, Amiel J, et al. Homozygous silencing of T-box transcription factor EOMES leads to microcephaly with polymicrogyria and corpus callosum agenesis. Nat Genet. 2007;39:454–6. [Abstract] [Google Scholar]
31. Shinmyo Y, Terashita Y, Dinh, Duong TA, Horiike T, Kawasumi M, Hosomichi K, et al. Folding of the cerebral cortex requires Cdk5 in upper-layer neurons in gyrencephalic mammals. Cell Rep. 2017;20:2131–43. [Abstract] [Google Scholar]
32. Chang BS, Piao X, Bodell A, Basel-Vanagaite L, Straussberg R, Dobyns WB, et al. Bilateral frontoparietal polymicrogyria: Clinical and radiological features in 10 families with linkage to chromosome 16. Annals Neurol. 2003;53:596–606. [Abstract] [Google Scholar]
33. Kuzniecky R, Andermann F, Guerrini R. Congenital bilateral perisylvian syndrome: Study of 31 patients. The CBPS multicenter collaborative study. Lancet. 1993;341:608–12. [Abstract] [Google Scholar]
34. Ramer JC, Mowrey PN, Robins DB, Ligato S, Towfighi J, Ladda RL. Five children with del (2)(q31q33) and one individual with dup (2)(q31q33) from a single family: Review of brain, cardiac, and limb malformations. Am J Med Genet. 1990;37:392–400. [Abstract] [Google Scholar]
35. Kogan JM, Egelhoff JC, Saal HM. Interstitial deletion of 13q associated with polymicrogyria. Am J Med Genet A. 2008;146A:910–6. [Abstract] [Google Scholar]
36. Stutterd CA, Francis D, McGillivray G, Lockhart PJ, Leventer RJ. Polymicrogyria associated with 17p13.3p13.2 duplication: Case report and review of the literature. Eur J Med Genet. 2020;63:103774. 10.1016/j.ejmg.2019.103774. [Abstract] [Google Scholar]
37. McDonald-McGinn DM. 22q11.2 deletion syndrome: A tiny piece leading to a big picture. Am J Med Genet A. 2018;176:2055–7. [Europe PMC free article] [Abstract] [Google Scholar]
38. Robin NH, Taylor CJ, McDonald-McGinn DM, Zackai EH, Bingham P, Collins KJ, et al. Polymicrogyria and deletion 22q11.2 syndrome: Window to the etiology of a common cortical malformation. Am J Med Genet A. 2006;140:2416–25. [Abstract] [Google Scholar]
39. Gerkes EH, Hordijk R, Dijkhuizen T, Sival DA, Meiners LC, Sikkema-Raddatz B, et al. Bilateral polymicrogyria as the indicative feature in a child with a 22q11.2 deletion. Eur J Med Genet. 2010;53:344–6. [Abstract] [Google Scholar]
40. Bingham PM, Lynch D, McDonald-McGinn D, Zackai E. Polymicrogyria in chromosome 22 deletion syndrome. Neurology. 1998;51:1500–2. [Abstract] [Google Scholar]
41. Saito Y, Kubota M, Kurosawa K, Ichihashi I, Kaneko Y, Hattori A, et al. Polymicrogyria and infantile spasms in a patient with 1p36 deletion syndrome. Brain Dev. 2011;33:437–41. [Abstract] [Google Scholar]
42. Shiba N, Daza RA, Shaffer LG, Barkovich AJ, Dobyns WB, Hevner RF. Neuropathology of brain and spinal malformations in a case of monosomy 1p36? Acta Neuropathol Commun. 2013;1:45. 10.1186/2051-5960-1-45. [Europe PMC free article] [Abstract] [Google Scholar]
43. Hussen DF, Kamel AK, Mekkawy MK, Ashaat EA, El Ruby MO, et al. Phenotypic and molecular cytogenetic analysis of a case of monosomy 1p36 syndrome due to unbalanced translocation. Mol Syndromol. 2020;11:284–95. [Europe PMC free article] [Abstract] [Google Scholar]
44. El Waly B, Mignon-Ravix C, Cacciagli P, Buhler E, Ben Zeev B, Villard L. Molecular characterization of a 1p36 chromosomal duplication and in utero interference define ENO1 as a candidate gene for polymicrogyria. Eur J Hum Genet. 2020;28:1703–13. [Europe PMC free article] [Abstract] [Google Scholar]
45. Zollino M, Colosimo C, Zuffardi O, Rossi E, Tosolini A, Walsh CA, et al. Cryptic t (1;12)(q44;p13.3) translocation in a previously described syndrome with polymicrogyria, segregating as an apparently X-linked trait. Am J Med Genet A. 2003;117A:65–71. [Abstract] [Google Scholar]
46. Alkan M, Ramelli GP, Hirsiger H, Keser I, Remonda L, Bühler EM, et al. Presumptive monosomy 21 with neuronal migration disorder re-diagnosed as de novo unbalanced translocation t (18p; 21q) by fluorescence in situ hybridization. Genet Couns. 2002;13:151–6. [Abstract] [Google Scholar]
47. Metzke-Heidemann S, Kuhling-von Kaisenberg H, Caliebe A, Janssen D, Jonat W, Grote W, et al. Phenotypical variation in cousins with the identical partial trisomy 9 (pter-q22.2) and 7 (q35-qter) at 16 and 23 weeks gestation. Am J Med Genet A. 2004;126A:197–203. [Abstract] [Google Scholar]
48. Leeflang EP, Marsh SE, Parrini E, Moro F, Pilz D, Dobyns WB, et al. Patient with bilateral periventricular nodular heterotopia and polymicrogyria with apparently balanced reciprocal translocation t (1;6)(p12;p12.2) that interrupts the mannosidase alpha, class 1A, and glutathione S-transferase A2 genes. J Med Genet. 2003;40:e128. [Europe PMC free article] [Abstract] [Google Scholar]
49. Sener RN. Bilateral, perisylvian and rolandic cortical dysplasia in trisomy 13 syndrome. J Neuroradiol. 1996;23:231–3. [Abstract] [Google Scholar]
50. Pascual-Castroviejo I, Pascual-Pascual SI, Viaño J, Martinez V, Palencia R. Unilateral polymicrogyria: A common cause of hemiplegia of prenatal origin. Brain Dev. 2001;23:216–22. [Abstract] [Google Scholar]
51. Tombini M, Marciani MG, Romigi A, Izzi F, Sperli F, Bozzao A, et al. Bilateral frontal polymicrogyria and epilepsy in a patient with Turner mosaicism: A case report. J Neurol Sci. 2003;213:83–86. [Abstract] [Google Scholar]
52. Piao X, Basel-Vanagaite L, Straussberg R, Grant PE, Pugh EW, Doheny K, et al. An autosomal recessive form of bilateral frontoparietal polymicrogyria maps to chromosome 16q12.2-21. Am J Hum Genet. 2002;70:1028–33. [Europe PMC free article] [Abstract] [Google Scholar]
53. Nooraine J, Vasudha K, Natesh S, Iyer RB, Raghavendra S. Autosomal recessive bilateral frontal polymicrogyria with ectopia lentis and chorioretinal dystrophy. Annals Indian Acad Neurol. 2013;16:678–80. [Europe PMC free article] [Abstract] [Google Scholar]
54. Villard L, Nguyen K, Cardoso C, Martin CL, Weiss AM, Sifry-Platt M, et al. A locus for bilateral perisylvian polymicrogyria maps to Xq28. Am J Hum Genet. 2002;70:1003–8. [Europe PMC free article] [Abstract] [Google Scholar]
55. Santos NF, Secolin R, Brandão-Almeida IL, Silva MS, Torres FR, Tsuneda SS, et al. A new candidate locus for bilateral perisylvian polymicrogyria mapped on chromosome Xq27. Am J Med Genet A. 2008;146A:1151–7. [Abstract] [Google Scholar]
56. Piao X, Chang BS, Bodell A, Woods K, Benzeev B, Topcu M, et al. Genotype-phenotype analysis of human frontoparietal polymicrogyria syndromes. Annals Neurol. 2005;58:680–7. [Abstract] [Google Scholar]
57. Mirzaa GM, Conti V, Timms AE, Smyser CD, Ahmed S, Carter M, et al. Characterisation of mutations of the phosphoinositide-3-kinase regulatory subunit, PIK3R2, in perisylvian polymicrogyria: A next-generation sequencing study. Lancet Neurol. 2015;14:1182–95. [Europe PMC free article] [Abstract] [Google Scholar]
58. Roll P, Rudolf G, Pereira S, Royer B, Scheffer IE, Massacrier A, et al. SRPX2 mutations in disorders of language cortex and cognition. Hum Mol Genet. 2006;15:1195–207. [Abstract] [Google Scholar]
59. Baulac S, Lenk GM, Dufresnois B, Ouled Amar Bencheikh B, Couarch P, et al. Role of the phosphoinositide phosphatase FIG4 gene in familial epilepsy with polymicrogyria. Neurology. 2014;82:1068–75. [Europe PMC free article] [Abstract] [Google Scholar]
60. Jaglin XH, Chelly J. Tubulin-related cortical dysgeneses: Microtubule dysfunction underlying neuronal migration defects. Trends Genet. 2009;25:555–66. [Abstract] [Google Scholar]
61. Guerrini R, Mei D, Cordelli DM, Pucatti D, Franzoni E, Parrini E. Symmetric polymicrogyria and pachygyria associated with TUBB2B gene mutations. Eur J Hum Genet. 2012;20:995–8. [Europe PMC free article] [Abstract] [Google Scholar]
62. Fallet-Bianco C, Laquerrière A, Poirier K, Razavi F, Guimiot F, Dias P, et al. Mutations in tubulin genes are frequent causes of various foetal malformations of cortical development including microlissencephaly? Acta Neuropathol Commun. 2014;2:1–22. 10.1186/2051-5960-2-69. [Europe PMC free article] [Abstract] [Google Scholar]
63. Geerdink N, Rotteveel JJ, Lammens M, Sistermans EA, Heikens GT, Gabreëls FJ, et al. MECP2 mutation in a boy with severe neonatal encephalopathy: Clinical, neuropathological and molecular findings. Neuropediatrics. 2002;33:33–6. [Abstract] [Google Scholar]
64. Mitchell TN, Free SL, Williamson KA, Stevens JM, Churchill AJ, Hanson IM, et al. Polymicrogyria and absence of pineal gland due to PAX6 mutation. Annals Neurol. 2003;53:658–63. [Abstract] [Google Scholar]
65. Jansen AC, Oostra A, Desprechins B, De Vlaeminck Y, Verhelst H, Régal L, et al. TUBA1A mutations: From isolated lissencephaly to familial polymicrogyria. Neurology. 2011;76:988–92. [Abstract] [Google Scholar]
66. Yu TW, Mochida GH, Tischfield DJ, Sgaier SK, Flores-Sarnat L, Sergi CM, et al. Mutations in WDR62, encoding a centrosome-associated protein, cause microcephaly with simplified gyri and abnormal cortical architecture. Nat Genet. 2010;42:1015–20. [Europe PMC free article] [Abstract] [Google Scholar]
67. Kheradmand Kia S, Verbeek E, Engelen E, Schot R, Poot RA, de Coo IF, et al. RTTN mutations link primary cilia function to organization of the human cerebral cortex. Am J Hum Genet. 2012;91:533–40. [Europe PMC free article] [Abstract] [Google Scholar]
68. Abdollahi MR, Morrison E, Sirey T, Molnar Z, Hayward BE, Carr IM, et al. Mutation of the variant alpha-tubulin TUBA8 results in polymicrogyria with optic nerve hypoplasia. Am J Hum Genet. 2009;85:737–44. [Europe PMC free article] [Abstract] [Google Scholar]
69. Bem D, Yoshimura SI, Nunes-Bastos R, Bond FC, Kurian MA, Rahman F, et al. Loss-of-function mutations in RAB18 cause Warburg micro syndrome. Am J Hum Genet. 2011;88:499–507. [Europe PMC free article] [Abstract] [Google Scholar]
70. Brooks AS, Bertoli-Avella AM, Burzynski GM, Breedveld GJ, Osinga J, Boven LG, et al. Homozygous nonsense mutations in KIAA1279 are associated with malformations of the central and enteric nervous systems. Am J Hum Genet. 2005;77:120–6. [Europe PMC free article] [Abstract] [Google Scholar]
71. McLarren KW, Severson TM, du Souich C, Stockton DW, Kratz LE, Cunningham D, et al. Hypomorphic temperature-sensitive alleles of NSDHL cause CK syndrome. Am J Hum Genet. 2010;87:905–14. [Europe PMC free article] [Abstract] [Google Scholar]
72. Paisán-Ruiz C, Scopes G, Lee P, Houlden H. Homozygosity mapping through whole genome analysis identifies a COL18A1 mutation in an Indian family presenting with an autosomal recessive neurological disorder. Am J Med Genet B. 2009;150B:993–7. [Abstract] [Google Scholar]
73. Aligianis IA, Johnson CA, Gissen P, Chen D, Hampshire D, Hoffmann K, et al. Mutations of the catalytic subunit of RAB3GAP cause Warburg micro syndrome. Nat Genet. 2005;37:221–23. [Abstract] [Google Scholar]
74. Iguchi T, Sakata K, Yoshizaki K, Tago K, Mizuno N, Itoh H. Orphan G protein-coupled receptor GPR56 regulates neural progenitor cell migration via a Gα12/13 and Rho pathway. J Biol Chem. 2008;283:14469–78. [Abstract] [Google Scholar]
75. Bahi-Buisson N, Poirier K, Boddaert N, Fallet-Bianco C, Specchio N, Bertini E, et al. GPR56-related bilateral frontoparietal polymicrogyria: Further evidence for an overlap with the cobblestone complex. Brain. 2010;133:3194–209. [Abstract] [Google Scholar]
76. Öncü-Öner T, Ünalp A, Porsuk-Doru İ, Ağılkaya S, Güleryüz H, Saraç A, et al. GPR56 homozygous nonsense mutation p.R271* associated with phenotypic variability in bilateral frontoparietal polymicrogyria. Turk J Pediatr. 2018;60:229–37. [Abstract] [Google Scholar]
77. Breuss MW, Leca I, Gstrein T, Hansen AH, Keays DA. Tubulins and brain development-The origins of functional specification. Mol Cell Neurosci. 2017;84:58–67. [Abstract] [Google Scholar]
78. Cushion TD, Dobyns WB, Mullins JGL, Stoodley N, Chung SK, Fry AE, et al. Overlapping cortical malformations and mutations in TUBB2B and TUBA1A. Brain J Neurol. 2013;136:536–48. [Abstract] [Google Scholar]
79. Poirier K, Saillour Y, Fourniol F, Francis F, Souville I, Valence S, et al. Expanding the spectrum of TUBA1A-related cortical dysgenesis to polymicrogyria. Eur J Hum Genet. 2013;21:381–5. [Europe PMC free article] [Abstract] [Google Scholar]
80. Shain C, Ramgopal S, Fallil Z, Parulkar I, Alongi R, Knowlton R, et al. Polymicrogyria-associated epilepsy: A multi-center phenotypic study from the Epilepsy Phenome/Genome Project. Epilepsia. 2013;54:1368–75. [Europe PMC free article] [Abstract] [Google Scholar]
81. Romaniello R, Zucca C, Arrigoni F, Bonanni P, Panzeri E, Bassi MT, et al. Epilepsy in tubulinopathy: Personal series and literature review? Cells. 2019;8:669. 10.3390/cells8070669. [Europe PMC free article] [Abstract] [Google Scholar]
82. Keays DA, Tian G, Poirier K, Huang G-J, Siebold C, Cleak J, et al. Mutations in alpha-tubulin cause abnormal neuronal migration in mice and lissencephaly in humans. Cell. 2007;128:45–57. [Europe PMC free article] [Abstract] [Google Scholar]
83. Belvindrah R, Natarajan K, Shabajee P, Bruel-Jungerman E, Bernard J, Goutierre M, et al. Mutation of the α-tubulin Tuba1a leads to straighter microtubules and perturbs neuronal migration. J Cell Biol. 2017;216:2443–61. [Europe PMC free article] [Abstract] [Google Scholar]
84. Schröter J, Popp B, Brennenstuhl H, Döring JH, Donze SH, Bijlsma EK, et al. Complementing the phenotypical spectrum of TUBA1A tubulinopathy and its role in early-onset epilepsies. Eur J Hum Genet. 2022:1–9. 10.1038/s41431-021-01027-0. [Europe PMC free article] [Abstract] [Google Scholar]
85. Romaniello R, Borgatti R. Epilepsy in multigene tubulin family mutations. J Neurol Neurophysiol. 2015;6:6. [Google Scholar]
86. Xu X, Shangguan Y, Lu S, Wang W, Du C, Xiao F, et al. Tubulin β-III modulates seizure activity in epilepsy. J Pathol. 2017;242:297–308. [Abstract] [Google Scholar]
87. Sia GM, Clem RL, Huganir RL. The human language and epilepsy associated gene SRPX2 regulates synapse formation and vocalization in mice. Science. 2013;342:987–91. [Europe PMC free article] [Abstract] [Google Scholar]
88. Roll P, Vernes SC, Bruneau N, Cillario J, Ponsole-Lenfant M, Massacrier A, et al. Molecular networks implicated in speech-related disorders: FOXP2 regulates the SRPX2/uPAR complex. Hum Mol Genet. 2010;19:4848–60. [Europe PMC free article] [Abstract] [Google Scholar]
89. Salmi M, Bruneau N, Cillario J, Lozovaya N, Massacrier A, Buhler E, et al. Tubacin prevents neuronal migration defects and epileptic activity caused by rat Srpx2 silencing in utero. Brain J Neurol. 2013;136:2457–73. [Abstract] [Google Scholar]
90. Glaser T, Jepeal L, Edwards JG, Young SR, Favor J, Maas RL, et al. PAX6 gene dosage effect in a family with congenital cataracts, aniridia, anophthalmia and central nervous system defects. Nat Genet. 1994;7:463–71. [Abstract] [Google Scholar]
91. Kikkawa T, Obayashi T, Takahashi M, Fukuzaki-Dohi U, Numayama-Tsuruta K, Osumi N, et al. Dmrta1 regulates proneural gene expression downstream of Pax6 in the mammalian telencephalon. Genes Cells. 2013;18:636–49. [Abstract] [Google Scholar]
92. Xie Q, Yang Y, Huang J, Ninkovic J, Walcher T, Wolf L, et al. Pax6 interactions with chromatin and identification of its novel direct target genes in lens and forebrain? PLoS One. 2013;8:e54507. 10.1371/journal.pone.0054507. [Europe PMC free article] [Abstract] [Google Scholar]
93. Stoykova A, Treichel D, Hallonet M, Gruss P , Pax6 modulates the dorsoventral patterning of the mammalian telencephalon. J Neurosci. 2000;20:8042–50. [Europe PMC free article] [Abstract] [Google Scholar]
94. Götz M, Stoykova A, Gruss P. Pax6 controls radial glia differentiation in the cerebral cortex. Neuron. 1998;21:1031–44. [Abstract] [Google Scholar]
95. Warren N, Caric D, Pratt T, Clausen JA, Asavaritikrai P, Mason JO, et al. The transcription factor, Pax6, is required for cell proliferation and differentiation in the developing cerebral cortex. Cereb Cortex. 1999;9:627–35. [Abstract] [Google Scholar]
96. Estivill-Torrus G, Pearson H, van Heyningen V, Price DJ, Rashbass P. Pax6 is required to regulate the cell cycle and the rate of progression from symmetrical to asymmetrical division in mammalian cortical progenitors. Development. 2002;129:455–66. [Abstract] [Google Scholar]
97. Mastick GS, Andrews GL. Pax6 regulates the identity of embryonic diencephalic neurons. Mol Cell Neurosci. 2001;17:190–207. [Abstract] [Google Scholar]
98. Talamillo A, Quinn JC, Collinson JM, Caric D, Price DJ, West JD, et al. Pax6 regulates regional development and neuronal migration in the cerebral cortex. Dev Biol. 2003;255:151–63. [Abstract] [Google Scholar]
99. Arnold SJ, Huang GJ, Cheung AFP, Era T, Nishikawa S, Bikoff EK, et al. The T-box transcription factor Eomes/Tbr2 regulates neurogenesis in the cortical subventricular zone. Genes Dev. 2008;22:2479–84. [Europe PMC free article] [Abstract] [Google Scholar]
100. Elsen GE, Hodge RD, Bedogni F, Daza RA, Nelson BR, Shiba N, et al. The protomap is propagated to cortical plate neurons through an Eomes-dependent intermediate map. Proc Natl Acad Sci. 2013;110:4081–6. [Europe PMC free article] [Abstract] [Google Scholar]
101. Hevner RF. Intermediate progenitors and Tbr2 in cortical development. J Anat. 2019;235:616–25. [Europe PMC free article] [Abstract] [Google Scholar]
102. Englund C, Fink A, Lau C, Pham D, Daza RA, Bulfone A, Kowalczyk T, et al. Pax6, Tbr2, and Tbr1 are expressed sequentially by radial glia, intermediate progenitor cells, and postmitotic neurons in developing neocortex. J Neurosci. 2005;25:247–51. [Europe PMC free article] [Abstract] [Google Scholar]
103. Bhat V, Girimaji SC, Mohan G, Arvinda HR, Singhmar P, Duvvari MR, et al. Mutations in WDR62, encoding a centrosomal and nuclear protein, in Indian primary microcephaly families with cortical malformations. Clin Genet. 2011;80:532–40. [Abstract] [Google Scholar]
104. Bilgüvar K, Oztürk AK, Louvi A, Kwan KY, Choi M, Tatli B, et al. Whole-exome sequencing identifies recessive WDR62 mutations in severe brain malformations. Nature. 2010;467:207–10. [Europe PMC free article] [Abstract] [Google Scholar]
105. Xu D, Zhang F, Wang Y, Sun Y, Xu Z. Microcephaly-associated protein WDR62 regulates neurogenesis through JNK1 in the developing neocortex. Cell Rep. 2014;6:104–16. [Abstract] [Google Scholar]
106. Handley MT, Carpanini SM, Mali GR, Sidjanin DJ, Aligianis IA, Jackson IJ, et al. Warburg micro syndrome is caused by RAB18 deficiency or dysregulation? Open Biol. 2015;5:150047. 10.1098/rsob. 150047. [Europe PMC free article] [Abstract] [Google Scholar]
107. Hutagalung AH, Novick PJ. Role of Rab GTPases in membrane traffic and cell physiology. Physiol Rev. 2011;91:119–49. [Europe PMC free article] [Abstract] [Google Scholar]
108. Aligianis IA, Morgan NV, Mione M, Johnson CA, Rosser E, Hennekam RC, et al. Mutation in Rab3 GTPase-activating protein (RAB3GAP) noncatalytic subunit in a kindred with Martsolf syndrome. Am J Hum Genet. 2006;78:702–7. [Europe PMC free article] [Abstract] [Google Scholar]
109. Drévillon L, Megarbane A, Demeer B, Matar C, Benit P, Briand-Suleau A, et al. KBP-cytoskeleton interactions underlie developmental anomalies in Goldberg-Shprintzen syndrome. Hum Mol Genet. 2013;22:2387–99. [Abstract] [Google Scholar]
110. Salehpour S, Hashemi-Gorji F, Soltani Z, Ghafouri-Fard S, Miryounesi M. Association of a novel nonsense mutation in KIAA1279 with Goldberg-Shprintzen syndrome. Iran J Child Neurol. 2017;11:70–4. [Europe PMC free article] [Abstract] [Google Scholar]
111. Stutterd CA, Brock S, Stouffs K, Fanjul-Fernandez M, Lockhart PJ, McGillivray G, et al. Genetic heterogeneity of polymicrogyria: Study of 123 patients using deep sequencing. Brain Commun. 2021;3:fcaa221. [Europe PMC free article] [Abstract] [Google Scholar]
112. Miyatake S, Kato M, Kumamoto T, Hirose T, Koshimizu E, Matsui T, et al. De novo ATP1A3 variants cause polymicrogyria. Sci Adv. 2021;7:eabd2368. [Europe PMC free article] [Abstract] [Google Scholar]
113. Fry AE, Fawcett KA, Zelnik N, Yuan H, Thompson BAN, Shemer-Meiri L, et al. De novo mutations in GRIN1 cause extensive bilateral polymicrogyria. Brain. 2018;141:698–712. [Europe PMC free article] [Abstract] [Google Scholar]
114. Terrone G, Voisin N, Abdullah Alfaiz A, Cappuccio G, Vitiello G, Guex N, et al. De novo PIK3R2 variant causes polymicrogyria, corpus callosum hyperplasia and focal cortical dysplasia. Eur J Hum Genet. 2016;24:1359–62. [Europe PMC free article] [Abstract] [Google Scholar]
115. Rivière J-B, Mirzaa GM, O'Roak BJ, Beddaoui M, Alcantara D, Conway RL, et al. De novo germline and postzygotic mutations in AKT3, PIK3R2 and PIK3CA cause a spectrum of related megalencephaly syndromes. Nat Genet. 2012;44:934–40. [Europe PMC free article] [Abstract] [Google Scholar]
116. Shi X, Lim Y, Myers AK, Stallings BL, Mccoy A, Zeiger J, et al. PIK3R2/Pik3r2 activating mutations result in brain overgrowth and EEG changes. Ann Neurol. 2020;88:1077–94. [Europe PMC free article] [Abstract] [Google Scholar]
117. Mirzaa G, Parry DA, Fry AE, Giamanco KA, Schwartzentruber J, Vanstone M, et al. De novo CCND2 mutations leading to stabilization of cyclin D2 cause megalencephaly-polymicrogyria-polydactyly-hydrocephalus syndrome. Nat Genet. 2014;46:510–5. [Europe PMC free article] [Abstract] [Google Scholar]
118. Platzer K, Sticht H, Edwards SL, Allen W, Angione KM, Bonati MT, et al. De novo variants in MAPK8IP3 cause intellectual disability with variable brain anomalies. Am J Hum Genet. 2019;104:203–12. [Europe PMC free article] [Abstract] [Google Scholar]
119. Ravenscroft G, Donato ND, Davis M, Craven P, Poke G, Neas K, et al. Recurrent de novo BICD2 mutation associated with severe arthrogryposis and polymicrogyria: Expanding the phenotype. Neuromuscular Disorders. 2016;26:S106–7. [Abstract] [Google Scholar]
120. Moresco G, Costanza J, Santaniello C, Rondinone O, Grilli F, Prada E, et al. A novel de novo DDX3X missense variant in a female with brachycephaly and intellectual disability: A case report. Italian J Pediatr. 2021;47:81. [Europe PMC free article] [Abstract] [Google Scholar]
121. Lennox AL, Hoye ML, Jiang R, Johnson-Kerner BL, Suit LA, Venkataramanan S, et al. Pathogenic DDX3X mutations impair RNA metabolism and neurogenesis during fetal cortical development. Neuron. 2020;106:404–20.e8. [Europe PMC free article] [Abstract] [Google Scholar]
122. Park HJ, Shin CH, Yoo WJ, Cho T-J, Kim MJ, Seong M-W, et al. Detailed analysis of phenotypes and genotypes in megalencephaly-capillary malformation-polymicrogyria syndrome caused by somatic mosaicism of PIK3CA mutations. Orphanet J Rare Dis. 2020;15:205. [Europe PMC free article] [Abstract] [Google Scholar]
123. Kobow K, Jabari S, Pieper T, Kudernatsch M, Polster T, Woermann FG, et al. Mosaic trisomy of chromosome 1q in human brain tissue associates with unilateral polymicrogyria, very early-onset focal epilepsy, and severe developmental delay. Acta Neuropathol. 2020;140:881–91. [Europe PMC free article] [Abstract] [Google Scholar]
124. Cohen AS, Wilson SL, Trinh J, Ye XC. Detecting somatic mosaicism: Considerations and clinical implications. Clin Genet. 2015;87:554–62. [Abstract] [Google Scholar]
125. Jamuar SS, Lam A-TN, Kircher M, D'Gama AM, Wang J, Barry BJ, et al. Somatic mutations in cerebral cortical malformations. N Engl J Med. 2014;371:733–43. [Europe PMC free article] [Abstract] [Google Scholar]
126. Amrom D, Poduri A, Goldman JS, Dan B, Deconinck N, Pichon B, et al. Duplication 2p16 is associated with perisylvian polymicrogyria. Am J Med Genet A. 2019;179:2343–56. [Abstract] [Google Scholar]
127. Mosca AL, Callier P, Faivre L, Marle N, Mejean N, Thauvin-Robinet C, et al. Polymicrogyria in a child with inv dup del (9p) and 22q11.2 microduplication. Am J Med Genet A. 2009;149A:475–81. [Abstract] [Google Scholar]
128. Yamada M, Hirotsune S, Wynshaw-Boris A. A novel strategy for therapeutic intervention for the genetic disease: Preventing proteolytic cleavage using small chemical compound. Int J Biochem Cell Biol. 2010;42:1401–7. [Europe PMC free article] [Abstract] [Google Scholar]
129. Hsieh J, Gage FH. Epigenetic control of neural stem cell fate. Curr Opin Genes Dev. 2004;14:461–9. [Abstract] [Google Scholar]
130. Mehler MF. Epigenetic principles and mechanisms underlying nervous system functions in health and disease. Prog Neurobiol. 2008;86:305–41. [Europe PMC free article] [Abstract] [Google Scholar]
131. MuhChyi C, Juliandi B, Matsuda T, Nakashima K. Epigenetic regulation of neural stem cell fate during corticogenesis. Int J Dev Neurosci. 2013;31:424–33. [Abstract] [Google Scholar]
132. Işık U, Dinçer A, Özek MM. Surgical treatment of polymicrogyria with advanced radiologic and neurophysiologic techniques. Child Nerv Sys. 2007;23:443–8. [Abstract] [Google Scholar]

Articles from Annals of Indian Academy of Neurology are provided here courtesy of Wolters Kluwer -- Medknow Publications

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.