- Review
- Open access
- Published:
Focal cortical dysplasia type II: review of neuropathological manifestations and pathogenetic mechanisms
Acta Epileptologica volume 7, Article number: 12 (2025)
Abstract
Focal cortical dysplasia (FCD) is an important cause of intractable epilepsy, with FCD type II (FCD II) being the most common subtype. FCD II is characterized by cortical dyslamination accompanied by dysmorphic neurons (DNs). Identifying the molecular alterations and targetable biomarkers is pivotal for developing therapies. Here, we provide a detailed description of the neuropathological manifestations of FCD II, including morphological alterations and immunophenotypic profiles, indicating that abnormal cells exhibit a diverse spectrum of mixed differentiation states. Furthermore, we summarize current research on the pathogenetic mechanisms, indicating that gene mutations, epigenetic alterations, cortical developmental protein disturbances, inflammatory processes, and extrinsic damages may lead to abnormal neuronal proliferation and migration, thereby contributing to the emergence and progression of FCD II. These findings not only enhance our understanding of the pathogenesis of FCD II but also offer new directions for clinical diagnosis and treatment. Future research should further explore the interactions among these factors and employ multidisciplinary approaches to advance our understanding of FCD II.
Background
Focal cortical dysplasia (FCD), a subtype of malformations of cortical development (MCD) [1], is characterized by congenital structural and cytoarchitectural abnormalities of the six layers of the cerebral cortex [2]. It is a predominant cause of intractable epilepsy in the pediatric population [3], and the third leading etiology of refractory epilepsy in adults [4]. According to the latest International League Against Epilepsy (ILAE) classification, FCD is divided into five subtypes, with FCD II being the most common, accounting for 45.3% [5, 6]. FCD II is characterized by cortical dyslamination, dysmorphic neurons (DNs), reduced myelination in the white matter, and indistinct gray matter to white matter boundaries [7, 8]. And FCD IIb is distinguished from FCD IIa by the additional presence of balloon cells (BCs) and disrupted clusters of oligodendroglial cells [9].
Extensive research has been conducted on FCD II, including its histomorphology and pathology, neuroimaging, gene mutations, transcriptomics, proteomics, changes in metabolomics expression, EEG alterations, and abnormal cellular electrophysiological characteristics [10,11,12,13,14,15]. However, FCD II still presents challenges in diagnosis and treatment.
In this review, we summarize the neuropathological features of FCD II, including morphological alterations and immunophenotypic profiles. Furthermore, we explore the pathogenetic mechanisms associated with FCD II, including gene mutations, epigenetic alterations, cortical developmental protein disturbances, inflammatory processes, and extrinsic damages. Summarizing these neuropathological manifestations and pathogenetic mechanisms can help clinical diagnosis and provide direction for future treatment.
Clinical challenges of FCD II
As the most common type of cortical dysplasia in epilepsy surgery, FCD II is typically with an average onset age of 5 years for seizures. Among confirmed cases, 51% are localized to the frontal lobe [5]. Compared to patients with FCD IIa, those with FCD IIb exhibit a shorter duration of symptoms, higher seizure frequency, and younger mean age at surgery, indicating greater severity of the seizure syndrome [16]. Combining radiological and histopathological findings with genetic information has introduced a new classification for FCD II called bottom-of-sulcus dysplasia (BOSD), which refers to lesions anatomically confined to the bottom of the sulcus [6].
It has been indicated that both ictal discharges and interictal rhythmic spiking originate from the dysplastic areas characterized by DNs [17, 18]. In addition, researches have shown that phase-amplitude coupling, spikes, fast gamma and ripples are related to the density of DNs and help localize the seizure onset zone [19,20,21]. Possible epileptogenic mechanisms include abnormal N-methyl-D-aspartate (NMDA) receptors, alteration in GABAergic neurons and abnormality of NKCC1/KCC2, pacemaker GABA synaptic activity, and the reorganization of the perisomatic inhibitory system [22,23,24,25,26]. In addition, aberrant adenosine signaling, endoplasmic reticulum stress, unfolded protein accumulation, and neuroinflammation can also contribute to the imbalance in the central nervous system’s excitation-inhibition system [27,28,29].
While the complex relationship between areas of developmental abnormality and the occurrence of epileptic seizures is progressively being elucidated, and resection epileptogenic tissue is essential for significantly improving seizure outcomes, determining clear margins remains a challenge [30]. Firstly, despite some FCD II cases showing significant changes on MRI, such as the transmantle sign and black line sign, MRI remains negative in 1/3 to 1/2 of FCD II patients, leading to unsatisfactory postoperative seizure-free rates and a poor prognosis [31,32,33,34,35,36]. Secondly, the region exhibiting the greatest cortical dysplasia does not necessarily align with the area most prone to epileptic activity [37]. Dysplastic tissue can also extend microscopically beyond the visible lesion or alter the epileptogenic threshold of adjacent cortical tissue [38, 39]. In addition to surgery, drugs for the treatment of FCD II also in constant developments. MTOR (mechanistic target of rapamycin) inhibitors have emerged as a novel therapeutic option for targeting FCD-related seizures, as 60% of FCD II cases, particularly FCD IIb, display gene mutations in the mTOR signaling pathway [40]. However, the efficacy of treatment with the mTOR inhibitor remains controversial [41, 42].
Therefore, identifying the molecular alterations and targetable biomarkers of DNs and BCs is crucial to understanding the pathogenesis of FCD II and developing targeted therapies [43].
Neuropathological manifestations of FCD II
Morphological alterations in FCD II
In FCD II, the six-layered homotypic structure typically disintegrates when mixed with normal pyramidal cells, while cortical layer I often remains well-defined but broadened [44]. And the impacted neocortex also experiences a decrease in cell density, which is more pronounced in FCD IIb compared to FCD IIa [9]. However, concurrent research indicates the presence of hidden cortical lamination involving normal-looking neurons, which retain their ability to migrate correctly within the cortex, whereas DNs exhibit alterations in their migratory patterns [45]. In addition to observing a reduction in myelin staining in the underlying white matter, studies at the ultrastructural level have found that the myelin sheaths of layer V axons are thinner in dysplastic specimens compared to controls (Fig. 1). This indicates that myelination is also compromised in the gray matter of the dysplastic area [46]. Furthermore, DNs and BCs predominantly accumulate at the base of the sulcus, with their numbers gradually diminishing along the ascending cortex [47].
The schematic depicts the architectural and cellular abnormalities in FCD II. In FCD II, lamination is virtually absent, except for an expanded layer I. Dysmorphic pyramidal neurons and cytomegalic interneurons are dispersed throughout layers II to VI, frequently extending into the white matter. In FCD IIb, BCs are predominantly observed in the upper layers and within the white matter
DNs, first described by Crome [48] and Taylor [49], can be disseminated across the full extent of the cortical architecture or embedded within the white matter [7]. They are distinctly delineated by a constellation of marked cytological aberrations: both the neuronal cell diameters and the cell nucleus diameter are significantly enlarged (the shortest diameter exceeds 25 μm, which is significantly larger than that of typical pyramidal cell in age- and location-matched postmortem controls [50]). Beyond the augmented somatic volume, DNs manifest morphological deviations characterized by abnormal thickness of the initial portion of apical dendrite and axon [51]. Despite morphological similarities in cell shape among distinct DNs, they may encompass a diversity of cellular phenotypes, as DNs primarily represent altered pyramidal neurons, but some may also exhibit features consistent with interneurons [52]. Utilizing a series of morphological parameters for the morphological reconstruction of brain tissue excised from FCD II patients, it suggests that DNs located primarily in the upper layers (layers II–IV) of the grey matter might contribute to the seizure genesis [53].
BCs exhibit a substantial cellular body with peripheral nuclei and enlarged, glassy cytoplasm, resembling the giant cells observed in tuberous sclerosis complex (TSC) [54]. These cells can be found in any cortical location, including layer I, and are frequently detected in the underlying white matter [55]. However, they always circumvent areas where dysmorphic neurons accumulate [9], and it has been confirmed that they are not the source of abnormal electrical activity [56].
Immunophenotypic profile of FCD II
The immunophenotypic profiles of DNs and BCs have been extensively investigated, predominantly to elucidate their lineage and maturation processes [57].
Nissl substance is aggregated and displaced towards the cell membrane, while phosphorylated (antibody 2F11) and non-phosphorylated neurofilament isoforms (SMI-32) accumulate in the cytoplasm of DNs [7]. In contrast, BCs exhibit an opalescent, glassy, eosinophilic cytoplasm devoid of Nissl substance. Furthermore, BCs commonly accumulate intermediate filaments such as vimentin and nestin [58]. These markers signify the presence of cytoskeletal abnormalities within DNs and BCs [57].
DNs and BCs have been demonstrated to express certain stem cell markers. Elevated levels of nuclear receptor subfamily 2 group F member 1 (NR2F1) are observed in DNs, while both NR2F1 and sex-determining region Y-box 2 (SOX2) are expressed in BCs [59]. Furthermore, BCs also express a range of stem cell markers such as GFAP-delta [60], CD133 [61], beta-1 integrins [62], and the onco-fetal antigen CD34 [63]. Both BCs and pyramidal neurons in DNs express Otx1, phospho-vimentin, Pax6, and BLBP, but lack Dlx1/Dlx2 expression, suggesting that they are derived from radial glial cells (RGCs) in the telencephalic ventricular zone (VZ) [64]. Additionally, interneurons in DNs demonstrate calbindin and parvalbumin expression but lack calretinin, suggesting an origin from the medial ganglionic eminence [52].
The high expression of NKCC1 in DNs and the altered subcellular distribution of KCC2 support the hypothesis of impaired developmental maturation in FCD II [65]. Within the normal cerebral cortex, the progression of neuronal differentiation is characterized by the successive expression of proteins, starting with MAP1B, then followed by NF-H, culminating in the expression of NeuN. MAP1B, an immature neuronal marker, frequently co-localized with GFAP (a glial marker), whereas the mature neuronal marker NeuN rarely did [66,67,68]. In DNs and BCs, co-expression of MAP1B and GFAP can be observed. However, NeuN and GFAP are rarely co-localized in DNs, and only slightly more frequently in BCs [68]. Studies have found that immunoreactivity with NF-H is not observed in any BCs [69]. Additionally, BCs also express β-tubulin III, which is the earliest marker of postmitotic neurons [61].
In summary, the mixed glio-neuronal features imply that DNs and BCs exhibit a diverse spectrum of mixed differentiation states [68].
The pathogenetic mechanisms of FCD II
Cellular proliferation within the mammalian cortex predominantly takes place in the germinal regions known as the VZ and subventricular zone (SVZ) [70]. In the initial stages, neuroepithelial progenitor cells within the VZ engage in mitotic activity to sustain the progenitor pool while concurrently giving rise to radial glial cells (RGCs), which are not only proliferative but also serve as migratory guides for neurons [71]. Following the formation of the first neuronal layer (the primordial plexiform layer or pre-plate), the SVZ forms between the VZ and pre-plate [72]. RGCs facilitate neurogenesis through successive self-renewal and asymmetric division, and that new-born neurons often use the parent cell’s radial fibre to migrate to the cortical plate [45, 73].
The immunophenotypic profile of BCs and pyramidal neurons in DNs indicates their derivation from RGCs in the telencephalic VZ [64].Together with immunophenotypic profile, the intrinsic membrane properties and synaptic features of DNs and BCs reveal developmental arrest and immaturity at both cellular and synaptic levels [74]. The co-expression of glial and neuronal antigens in different combinations indicates that the generation of aberrant cells in FCD II is associated with aberrant neuronal-glial differentiation [68, 75]. Concurrent studies have also suggested DNs exhibit alterations in their migratory patterns [45]. Collectively, these data suggest that the appearance of aberrant cells may be related to disruptions in the processes of neuronal-glial proliferation and migration, potentially occurring during the first half of gestation [45, 68]. The somatic mutation hypothesis posits that a somatic gene mutation occurs in a single progenitor cell during a critical phase of cortical development. This mutant cell continues to proliferate, producing progeny that carry the mutation. Consequently, DNs and BCs exhibit distinct morphological characteristics and are likely derived from different differentiations pathways of the same mutant neuroglial progenitor cell [76, 77]. This hypothesis would be consistent with the characteristic funnel-shaped morphological distribution of FCD II lesions, since clonally related neurons show the same distribution pattern [68, 78]. Furthermore, abnormalities in cellular migration may be the primary cause of pronounced MRI anomalies predominantly observed at the bottom of the sulcus. The tendency for BOSD to localize in the frontal sulci may be attributed to the shorter neurogenesis time required in rostral cortices compared to caudal areas [47].
However, the X-androgen receptor (XAR) inactivation assay has detected varying lengths of XAR-CAG repeats in individual DNs and BCs. This suggests that DNs and BCs may originate from a mixed cell population derived from multiple progenitor cells [79]. Simultaneously, the pyramidal neurons and interneurons in DNs exhibit distinct origins [52]. These findings suggest DNs and BCs in FCD lesions are heterogeneous, likely arising from multiple progenitor cells or post-mitotic neurons that were extrinsically damaged during cortical development [68, 79]. Besides, some studies have suggested that the etiology of FCD may be partially attributed to the abnormal retention of pre-plate cells in the subplate and marginal zones. Cytomegalic neurons could be postnatally retained subplate cells. This phenomenon likely occurs during the advanced stages of cortical development, as it does not align with the expected pattern if severe cortical dysplasia were the result of developmental processes significantly altering early to mid-corticoneurogenesis [74, 80]. Thus, the pathological mechanisms underlying FCD II remain controversial (Fig. 2).
Gene mutations
MTOR serves as an intricately conserved signaling nexus, integrating neuronal activity and a variety of synaptic inputs. MTOR is found in two functionally distinct complexes: mTORC1 and mTORC2, which are pivotal in regulating long-term synaptic efficacy and memory consolidation [81]. In addition, erturbations in mTOR signaling pathways are correlated with an array of neurodevelopmental and neuropsychiatric pathologies [82]. The cell count analysis confirmed that phospho-S6 (pS6), a downstream target of the mTOR pathway, has been detected expressing in the majority of DNs, which indicates that the activation of the mTOR pathway plays a critical role in the formation of FCD II [45].
Studies have reported the existence of a “mutation gradient”, indicating that the mutational burden is higher at the bottom of the sulcus compared to the gyral crown [83]. Over the past decades, sequencing studies have identified both single-hit somatic mosaicism and two-hit germline plus somatic mutations in genes regulating the mammalian target of the mTOR pathway in FCD II (Fig. 3) [55, 84]. These genes include PIK3CA [85], PTEN [86], TSC1/TSC2 [87], GATOR1-encoding complex (DEPDC5, NPRL2, NPRL3) [88,89,90], RHEB [91] and MTOR itself [92]. Somatic gain-of-function variants in MTOR and its activators, as well as germline, somatic and two-hit loss-of-function variants in its repressors, can culminate in the hyperactivation of the mTOR-signaling cascade [40]. Notably, the genotype-phenotype correlation is increasingly being investigated. Patients with brain somatic MTOR variants typically exhibit larger lesions on MRI, including involvement of the white matter. In contrast, patients with GATOR1 complex variants often display subtler MRI visibility and relatively poorer postoperative outcomes. A subset of these patients with GATOR1 complex variants also show a unique and predominantly vacuolizing phenotype, characterized by p62 immunofluorescent aggregates within autophagosomes [93].
The mTOR pathway and its regulators. mTOR is a protein kinase present in the cell in two complexes: mTORC1 and mTORC2. mTORC1 is related to cell size and proliferation, whereas mTORC2 influences cytoskeleton dynamics. Activation of the PI3K-Akt pathway leads to the inactivation of the TSC1/TSC2 complex, resulting in the indirect activation of mTORC1. PTEN inhibits the PI3K-Akt pathway. Furthermore, PI3K signaling also activates mTORC2. The GATOR1 complex, composed of DEP domain-containing protein 5 (DEPDC5), nitrogen permease regulator-like 2 (Nprl2), and nitrogen permease regulator-like 3 (Nprl3), inhibits mTORC1 signaling by repressing Ras-related GTP-binding protein A/B (RagA/B). Rapamycin binds to FK506-binding protein 12 (FKBP12) and inhibits mTORC1. Red diamonds indicate the proteins whose corresponding genes are mutated in FCD II
DNA sequencing of cerebral samples obtained from subjects with hemimegalencephaly and FCD II has demonstrated that pathogenic somatic variants may arise in both neuronal and non-neuronal lineage. But in minor FCD II instances, these aberrations manifest exclusively within the neuronal lineage [87]. Subsequent to laser capture microdissection of aberrant tissue formations with confirmed gene mutations in the mTOR pathway, a pronounced enrichment of variant allele frequency, reaching up to 45%, was observed in both pools of DNs and BCs compared to morphologically normal neurons (down to 1.4%), and glial cells (down to 1.4%), or bulk tissue [40]. Collectively, these findings indicate that DNs and BCs serve as the primary vectors for mTOR pathway hyperactivating variants in FCD II. Some identified gene mutations in the mTOR pathway recurrently found in patients were engineered into the developing mouse cortex utilizing in utero electroporation techniques, and successfully generated the mouse model recapitulating all neuropathological features of FCD II patients, such as migration defect, DNs, and spontaneous seizures [94,95,96]. Additionally, it has been substantiated that the inhibition of hyperactivated mTOR kinase can mitigate epilepsy and impede the development of cytomegalic neurons [94]. These genetic discoveries confirmed that de novo gene mutations in mTOR pathway are associated with focal cortical malformation [84].
Currently pathogenic variants in mTOR pathway-related genes can only be detected in 60% of FCD II cases, but the panel-negative FCD II cases also display pS6-positive DNs and BCs [84]. Besides, there is no significant difference in pS6 expression between mutated and non-mutated FCD IIb cases, nor are there significant differences in the clinical manifestations among these patients [97].
The primary reason is the difficulty in detection. These somatic mutations are typically confined to a subset of cells within the brain, making detection through blood or cerebrospinal fluid (CSF) samples less effective; therefore, access to brain tissue is required [98]. Since the mutations arise later in development, they are present in only a smaller fraction of cells. In fact, in 79% of reported cases of mutated FCD II, the brain mosaicism rate is less than 5%, which makes conventional sequencing methods insufficient for detecting low-level mosaic mutations [40]. To address this limitation, numerous novel techniques have now emerged, including fluorescence-activated cell sorting, droplet digital PCR, and novel depth electrode harvesting technique [99,100,101]. The application of these advanced technologies aids in the detection of ultra-low levels of cellular mutations, holding promise for improving the genetic diagnostic rate within FCD II cohorts.
Besides, there may be undiscovered gene mutations related to FCD II. By integrating whole exome sequencing with amplicon sequencing, researchers identified some new variants not previously linked to FCD. For example, a somatic missense variant in MED12, a gene associated with neurodevelopmental syndrome and an upstream regulator of the mTOR pathway, was identified in a patient with FCD IIa [102]. Variants in IRS1, RAB6B, ZNF337, RALA, and HTR6 were also detected. Subsequent in vitro functional studies demonstrated that the IRS1 variant led to hyperactivation of mTOR [103] and the mutant IRS1 variant (IRS-1 c.1791dupG) elicited hyperactivity in phosphorylated extracellular signal-regulated kinase (p-ERK) and augmented cellular volume through the mitogen-activated protein kinase (MAPK) signaling pathway [104]. These findings suggest that a wide range of undiscovered genetic mutations may contribute to the development of FCD II, highlighting the need for further exploration.
Epigenetic alterations
Epigenetic mechanisms, which represent heritable modifications in gene expression that do not result from an alteration in the DNA sequence, may modulate gene expression and be accountable for some mutation-negative patients [105]. DNA methylation and microRNAs are two epigenetic mechanisms that have been identified in FCD II.
In the mammalian genome, DNA methylation is an epigenetic mechanism achieved by the enzymatic addition of a methyl group (-CH3) from S-adenosyl methionine to the fifth carbon position of cytosine in a CpG dinucleotide [106]. The processes of DNA methylation and hydroxy-methylation impact the proliferation and differentiation of neural stem cells, thereby playing a critical role in the normal development and function of the brain [107]. A comprehensive analysis of DNA methylation and RNA sequencing in FCD II tissues has elucidated the relationship between promoter methylation and gene expression in a subset of genes implicated in receptor tyrosine kinase (RTK) and mTOR signaling, synaptic transmission, and neuronal development and cell-cell interactions. Furthermore, it has been observed that the expression levels of DNA methyltransferase 3α (DNMT3α) are significantly upregulated in patients with FCD II, which may epigenetically regulate the expression of genes involved in cortical development and neuronal plasticity [108]. However, Kobow et al. discovered no significant enrichment of differential DNA methylation or gene expression in mTOR pathway-related genes across different subtypes of FCD, whereas DNs were present in FCD II only [109]. The generation of FCD II may involve complex DNA methylation processes.
MicroRNAs (miRNAs) are a class of single-stranded, noncoding RNAs that function as endogenous posttranscriptional regulators of gene expression by targeting selected protein-coding mRNAs [110]. Recent studies have uncovered a strong association between miRNAs and FCD II. Lee et al. performed miRNA microarray analysis on surgical specimens from cortical dysplasia patients and normal control children who underwent surgical treatment, identifying differentially expressed miRNAs. When the putative target mRNAs were annotated and analyzed by cell signaling pathway, numerous upstream and downstream genes within the mTOR signaling cascade were identified as prospective target entities [111]. Beyond the miRNAs connected to target genes within the mTOR pathway, those correlated with the LIS1 and Hippo pathways have also been pinpointed [111, 112]. LIS1 is crucial for neuronal migration [113], and the Hippo signaling pathway is a regulator of cell proliferation, apoptosis, and differentiation [114]. As a member of the basic helix-loop-helix transcription factor family, NEUROG2 plays a crucial role in determining cell fate and promoting neuronal differentiation [115], exhibiting pronounced nuclear expression in BCs and DNs, related to miRNA regulation. Disruption of the interaction between miR-34a and NEUROG2 precipitated an upregulation in the expression of NEUROG2, thereby compromising the inhibition of neurogenesis and culminating in aberrant neuronal migration and differentiation, ultimately resulting in the formation of FCD II [105, 116]. In summary, epigenetic alterations will also be a key focus in researching the causes of FCD II.
Cortical developmental protein disturbances
The deregulation in the expression levels and alterations in the cellular distribution of numerous proteins linked to cortical development have been observed in FCD II specimens [54].
Extensive studies have documented elevated levels of numerous growth factors in DNs, encompassing members of the vascular endothelial growth factor (VEGF) family and their receptors [117, 118], the fibroblast growth factor (FGF) family and their receptors [119, 120], along with epidermal growth factor receptor (EGFR) and high-affinity neurotrophin receptors [121, 122]. As pivotal components of the VEGF signaling pathway, the aberrant expression of VEGFA, VEGFB, VEGFC, and their receptors in DNs may potentially impact neuronal differentiation and aberrant NMDA receptor-mediated currents, thereby contributing to the pathogenesis of cortical lesions in patients. Furthermore, the aberrant expression of the FGF family and their receptors not only reflects the putative perturbations of developmental events during the early fetal period that underlie the pathogenesis of these dysplastic lesions [123], but also may lead to the activation of the mTOR pathway [124] and the aberrant expression of bone morphogenic protein-4 (BMP-4) [125].
Bone morphogenetic proteins (BMPs), a significant subfamily within the transforming growth factor-β (TGF-β) superfamily, are multifunctional ligands crucial for regulating intricate cellular processes, such as cell proliferation, differentiation, chemotactic migration, and apoptosis [126]. In comparison to the control group, the FCD II samples showed a reduction in the overall immunoreactivity of BMP-4 staining in their dysplastic cortices. However, moderate to strong BMP-4 immunoreactivity was still observed in malformed neurons, particularly DNs and BCs [127].
Doublecortin-like kinase (DCLK) and its splice variant DCL, play pivotal roles in the orchestration of neurogenesis, neuronal migration, and axonal outgrowth [128]. As a microtubule-associated protein, DCL is prominently expressed in neuronal progenitor cells within the fetal brain and has been demonstrated to be crucial for the microtubule-guided transport of signaling proteins to the nucleus [129]. The pronounced postnatal manifestation of DCL in DNs and BCs underscores the potential relevance to the pathogenesis of FCD II [130].
Nogo-A, a member of the reticulon protein family, is expressed in oligodendrocytes of the adult central nervous system and in developing neurons, serving as a potent inhibitor of neurite outgrowth [131]. In comparison to autopsy control samples, the messenger RNA and protein levels of the Nogo-A receptor (NgR) and its downstream targets, leucine-rich repeat and immunoglobulin-like domain-containing protein 1 (LINGO-1), tumor necrosis factor receptor superfamily member 19 (TROY), and Ras homolog family member A (RhoA), are upregulated in the cortices of FCD II patients. Immunohistochemical analyses have revealed robust expression of Nogo-A and NgR in aberrantly shaped cells, with a particular emphasis on DNs [132]. Additionally, the human leukocyte immunoglobulin-like receptor B2 (LILRB2) signaling pathway, regulated by Nogo-A, is highly expressed in DNs and BCs [133], thereby inhibiting neurite regeneration and prolongation [134]. Collectively, these findings suggest that the Nogo-A system may play a pivotal role in the development and/or seizure activity associated with cortical lesions in FCD II.
Within BCs, the heightened cytoplasmic presence of the dishevelled (Dvl-1) protein was concomitant with the absence of Notch-1, augmented levels of adenomatous polyposis coli (APC), modified cytoplasmic β-catenin, and reduced nuclear β-catenin expression [135]. These findings align with the concept that Wnt/Notch signaling pathways play a pivotal role in neurogenesis, neuroglial cell fate determination, neuronal migration and neural tube development. This suggests disruptions in Wnt/Notch signaling may contribute to the neuropathology of FCD II [54].
Cortical tissue from patients with FCDII exhibited elevated levels of filamin A (FLNA), a consequence of increased transcriptional activity downstream of hyperactive MEK/MAPK signaling, independent of mTOR pathways [136]. Furthermore, reducing FLNA expression alleviated misplacement and dysmorphogenesis [137]. Therefore, targeting FLNA may offer a therapeutic approach that circumvents mTOR dependency.
The aberrant expression of these proteins involved in cortical development regulation may disrupt neuronal-glial proliferation and migration, resulting in the formation of abnormal cortex. However, the causes and mechanisms underlying the abnormal expression of these proteins remain to be explored. Therefore, gaining a more profound insight into these aberrant proteins could offer novel perspectives for therapeutic intervention.
Inflammatory processes
DNs are recognized as hallmark features of FCD II; however, similar cells are also occasionally observed in certain acquired epilepsy-associated pathologies, such as Rasmussen’s encephalitis (RE)—a rare, chronic inflammatory disorder characterized by intractable seizures during childhood [138]. In RE specimens, the pyroptosis pathway in neurons was mediated by nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 1 (NLRP1) and NLRP3 inflammasomes, involving activated caspase-1 and increased gasdermin D (GSDMD) expression, which can promote the release of the inflammatory cytokine such as interleukin-1β (IL-1β) [139]. In FCD II cases, phosphorylated phosphoinositide-dependent kinase-1 (pPDK1) and phosphorylated Akt (pAkt), markers of mTOR pathway activation, were expressed only in a subset of pS6-positive DNs, but not in the abnormal cells of RE specimens. The pS6 and IL-1β were co-expressed in DNs that did not exhibit upstream activation of the mTOR pathway [140]. Recent research indicates an upregulation of the expression of the purinergic ionotropic P2X7 receptor (P2X7R) [141] and nuclear factor-κB (NF-κB) p65 accumulates in the nuclei of selected subpopulations of DNs and BCs in FCD IIb and TSC [142]. IL-1β, a downstream factor of the P2X7R signaling pathway, and the canonical NF-κB pathway could lead to the excessive production of inflammatory mediators [143]. Additionally, high-level expression of the inflammatory cytokine interleukin 2 (IL-2) and its receptors were also localized in the malformed cells in FCD II [144]. Furthermore, FCD II tissue shows upregulation of the high-mobility group box 1-Toll-like receptor 4 (HMGB1-TLR4) pathway, resulting in increased downstream pro-inflammatory cytokines, and downregulation of formyl peptide receptor 2, which impairs the neuroinflammation resolution [145, 146]. Activation of the classical complement pathway and microglial reactivity have also been found to be associated with abnormalities in FCD II tissue [147]. Notably, BCs may be pivotal drivers of inflammation, which could partially elucidate why the alterations in FCD IIa are less severe [8].
Inflammatory mediators produced during inflammatory processes, such as pro-inflammatory cytokines, can phosphorylate TSC1, leading to the inhibition of the TSC1/2 complex and subsequently activation of mTORC1 [148]. Due to the inflammatory processes may cause the emergence of pS6-positive DNs bypassing the upstream mTOR pathway, the interplay between inflammatory environment and neuroplasticity could be responsible for the etiology of FCD II (Fig. 4) [140].
Extrinsic damages
A clinical study has found that 32% of patients with cortical malformations have identifiable antenatal risk factors [149]. Thus, a hypothesis posits that extrinsic damages to the developing brain may lead to a focal, field-like defect in cortical maturation [68].
Experimental evidence suggests that animals exposed to physical or chemical factors during early developmental stages exhibit a convergence of pathological phenotypes observed in FCD. For instance, in the rat model of freeze lesioning performed at embryonic day 18, the laminar structure was found to be completely disrupted, with neurons of varying sizes exhibiting disorganized orientation [150]. Addtionally utero X-ray radiated rats showed some neurons losing cortical polarity in morphology at 30 days after birth [118]. Besides, prenatal administration of a double methylazoxymethanol intraperitoneal injection to pregnant rats leads to cellular dysmorphology and laminar disorganization [151]. Similarly, in utero exposure to 1-3-bis-chloroethyl-nitrosurea results in cortical heterotopias and laminar disorganization, accompanied by cytomegalic neurons [152]. This indicates that prenatal exposure to physical or chemical factors can also disrupt the development of the fetal cortex, leading to the formation of cellular abnormalities. However, these cytomegalic neurons did not exhibit clear abnormal expression of pS6, suggesting that the connection between extrinsic damages and FCD II needs additional research [153].
Conclusions
In this review, we summarize the neuropathological manifestations and pathogenetic mechanisms of FCD II. The neuropathological characteristics, including morphological alterations and specific immunophenotypes, suggest that DNs and BCs exhibit a diverse spectrum of mixed differentiation states. Furthermore, the formation of FCD II is associated with abnormal neuronal proliferation and migration during cortical development. Previously, mutations in the mTOR signaling pathway were considered the primary cause of FCD II. However, mutations in other pathway genes, epigenetic changes, cortical developmental protein disturbances, inflammatory processes, and extrinsic damages have also been linked to the formation of FCD II abnormal tissue. These factors may not be isolated but interconnected. Here, we summarize current research on the pathogenetic mechanisms of FCD II, providing directions for future treatments. Going forward, further research on FCD II abnormal tissue at the single-cell level is needed. A combination of various methods including neuropathology, molecular genetics, cell biology, and computational biology can be utilized to delve deeper into FCD II.
Data availability
Availability of data and materials is not applicable in this study.
Abbreviations
- BCs:
-
Balloon cells
- BMP:
-
Bone morphogenetic protein
- DCLK:
-
Doublecortin-like kinase
- DNs:
-
Dysmorphic neurons
- FCD:
-
Focal cortical dysplasia
- FCD II:
-
Focal cortical dysplasia type II
- ILAE:
-
International League Against Epilepsy
- MCD:
-
Malformations of cortical development
- MTOR:
-
Mechanistic target of rapamycin
- pS6:
-
Phospho-S6
- RE:
-
Rasmussen’s encephalitis
- RGCs:
-
Radial glial cells
- SVZ:
-
Subventricular zone
- TSC:
-
Tuberous sclerosis complex
- VZ:
-
Ventricular zone
- XAR:
-
X-androgen receptor
References
Barkovich AJ, Dobyns WB, Guerrini R. Malformations of cortical development and epilepsy. CCold Spring Harb Perspect Med. 2015;5(5):a022392.
Mao C, Jin L, Dou W, Lu Q, Zhou L, Ren H, et al. Type IIB focal cortical dysplasia with balloon cells in medial temporal lobe epilepsy: clinical, neuroimaging, and histopathological findings. Epilepsy Res. 2019;157:106189.
Kun Y, Zejun D, Jian Z, Feng Z, Changqing L, Xueling Q. Surgical histopathologic findings of 232 Chinese children cases with drug-resistant seizures. Brain Behav. 2020;10(4):e01565.
Cloppenborg T, May TW, Blümcke I, Fauser S, Grewe P, Hopf JL, et al. Differences in pediatric and adult epilepsy surgery: a comparison at one center from 1990 to 2014. Epilepsia. 2019;60(2):233–45.
Blumcke I, Spreafico R, Haaker G, Coras R, Kobow K, Bien CG, et al. Histopathological findings in brain tissue obtained during epilepsy surgery. N Engl J Med. 2017;377(17):1648–56.
Agarwal A, Bathla G, Soni N, Desai A, Middlebrooks E, Patel V, et al. Updates from the international league against epilepsy classification of epilepsy (2017) and focal cortical dysplasias (2022): imaging phenotype and genetic characterization. AJNR American Neuroradiol. 2024;45(8):991–9.
Blümcke I, Thom M, Aronica E, Armstrong DD, Vinters HV, Palmini A, et al. The clinicopathologic spectrum of focal cortical dysplasias: a consensus classification proposed by an ad hoc task force of the ILAE diagnostic methods commission. Epilepsia. 2011;52(1):158–74.
Zimmer TS, Broekaart DWM, Luinenburg M, Mijnsbergen C, Anink JJ, Sim NS, et al. Balloon cells promote immune system activation in focal cortical dysplasia type 2b. Neuropath Appl Neuro. 2021;47(6):826–39.
Najm I, Lal D, Alonso Vanegas M, Cendes F, Lopes-Cendes I, Palmini A, et al. The ILAE consensus classification of focal cortical dysplasia: an update proposed by an ad hoc task force of the ILAE diagnostic methods commission. Epilepsia. 2022;63(8):1899–919.
Balestrini S, Barba C, Thom M, Guerrini R. Focal cortical dysplasia: a practical guide for neurologists. Pract Neurol. 2023;23(4):293–302.
Genç B, Aksoy A, Aslan K. Cortical and subcortical morphometric changes in patients with frontal focal cortical dysplasia type II. Neuroradiol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00234-024-03471-3.
Ferri L, Menghi V, Licchetta L, Dimartino P, Minardi R, Davì C, et al. Detection of somatic and germline pathogenic variants in adult cohort of drug-resistant focal epilepsies. Epilepsy and behavior: EandB. 2024;153:109716.
Vermeulen I, Rodriguez-Alvarez N, François L, Viot D, Poosti F, Aronica E, et al. Spatial omics reveals molecular changes in focal cortical dysplasia type II. Neurobiol DIS. 2024;195:106491.
Srivastava A, Kumar K, Banerjee J, Tripathi M, Dubey V, Sharma D, et al. Transcriptomic profiling of high- and low-spiking regions reveals novel epileptogenic mechanisms in focal cortical dysplasia type II patients. Mol Brain. 2021;14(1):120.
Boonyapisit K, Najm I, Klem G, Ying Z, Burrier C, LaPresto E, et al. Epileptogenicity of focal malformations due to abnormal cortical development: direct electrocorticographic-histopathologic correlations. Epilepsia. 2003;44(1):69–76.
Kumari K, Sharma MC, Kakkar A, Malgulwar PB, Pathak P, Suri V, et al. mTOR pathway activation in focal cortical dysplasia. Ann Diagn Pathol. 2020;46:151523.
Chassoux F, Devaux B, Landré E, Turak B, Nataf F, Varlet P, et al. Stereoelectroencephalography in focal cortical dysplasia: a 3D approach to delineating the dysplastic cortex. Brain. 2000;123(Pt 8):1733–51.
Kao H, Hu S, Mihaylova T, Ziobro J, Ahn E, Fine C, et al. Defining the latent period of epileptogenesis and epileptogenic zone in a focal cortical dysplasia type II (FCDII) rat model. Epilepsia. 2021;62(5):1268–79.
Garbelli R, Spreafico R, Barbaglia A, Rossini L, Milesi G, Zucca I, et al. Stereo-EEG, radiofrequency thermocoagulation and neuropathological correlations in a patient with MRI-negative type IIb focal cortical dysplasia. Seizure. 2016;41:1–3.
Zucca I, Milesi G, Padelli F, Rossini L, Gozzo F, Figini M, et al. An image registration protocol to integrate electrophysiology, MRI and neuropathology data in epileptic patients explored with intracerebral electrodes. J Neurosci Meth. 2018;303:159–68.
Rampp S, Rössler K, Hamer H, Illek M, Buchfelder M, Doerfler A, et al. Dysmorphic neurons as cellular source for phase-amplitude coupling in focal cortical dysplasia type II. Clin Neurophysiology: Official J Int Federation Clin Neurophysiol. 2021;132(3):782–92.
Pineau L, Buhler E, Tarhini S, Bauer S, Crepel V, Watrin F, et al. Pathogenic MTOR somatic variant causing focal cortical dysplasia drives hyperexcitability via overactivation of neuronal GluN2C N-methyl-D-aspartate receptors. Epilepsia. 2024;65(7):2111–26.
Yan L, Yun-Lin L, Yong-Ling L, Wei-Wei Z, Yue-Shan P. Alteration of GABAergic neurons and abnormality of NKCC1/KCC2 in focal cortical dysplasia (FCD) type II lesions. Epilepsy Res. 2023;194:107180.
Cepeda C, Chen JY, Wu JY, Fisher RS, Vinters HV, Mathern GW, et al. Pacemaker GABA synaptic activity may contribute to network synchronization in pediatric cortical dysplasia. Neurobiol Dis. 2014;62:208–17.
Szekeres-Paraczky C, Szocsics P, Erőss L, Fabó D, Mód L, Maglóczky Z. Reorganization of parvalbumin immunopositive perisomatic innervation of principal cells in focal cortical dysplasia type IIB in human epileptic patients. Int J Mol Sci. 2022;23(9):4746.
Zhang L, Huang J, Dai L, Zhu G, Yang X, He Z, et al. Expression profiles of α-synuclein in cortical lesions of patients with FCD IIb and TSC, and FCD rats. Front NEUROL. 2023;14:1255097.
Guo M, Zhang J, Wang J, Wang X, Gao Q, Tang C, et al. Aberrant adenosine signaling in patients with focal cortical dysplasia. Mol Neurobiol. 2023;60(8):4396–417.
Madhamanchi K, Madhamanchi P, Jayalakshmi S, Panigrahi M, Patil A, Phanithi PB. Endoplasmic reticulum stress and unfolded protein accumulation correlate to seizure recurrence in focal cortical dysplasia patients. Cell Stress Chaperon. 2022;27(6):633–43.
Alfano V, Romagnolo A, Mills JD, Cifelli P, Gaeta A, Morano A, et al. Unexpected effect of IL-1β on the function of GABA(A) receptors in pediatric focal cortical dysplasia. Brain Sci. 2022;12(6):807.
Cohen-Gadol AA, Ozduman K, Bronen RA, Kim JH, Spencer DD. Long-term outcome after epilepsy surgery for focal cortical dysplasia. J Neurosurg. 2004;101(1):55–65.
Kimura Y, Shioya A, Saito Y, Oitani Y, Shigemoto Y, Morimoto E, et al. Radiologic and pathologic features of the transmantle sign in focal cortical dysplasia: the T1 signal is useful for differentiating subtypes. AJNR Am J Neuroradiol. 2019;40(6):1060–6.
Tang Y, Blümcke I, Su T, Choi JY, Krishnan B, Murakami H, et al. Black line sign in focal cortical dysplasia IIB: a 7T MRI and electroclinicopathologic study. Neurology. 2022;99(6):e616-26.
Krsek P, Maton B, Korman B, Pacheco-Jacome E, Jayakar P, Dunoyer C, et al. Different features of histopathological subtypes of pediatric focal cortical dysplasia. Ann NEUROL. 2008;63(6):758–69.
Liu Z, Hu W, Sun Z, Wang X, Liu L, Shao X, et al. MRI abnormalities predominate in the bottom part of the sulcus with type II focal cortical dysplasia: a quantitative study. AJNR Am J Neuroradiol. 2019;40(1):184–90.
Zhang S, Luo Y, Zhao Y, Zhu F, Jiang X, Wang X, et al. Prognostic analysis in children with focal cortical dysplasia II undergoing epilepsy surgery: clinical and radiological factors. Front Neurol. 2023;14:1123429.
Xiao L, Yang J, Zhu H, Zhou M, Li J, Liu D, et al. [(18)F]SynVesT-1 and [(18)F]FDG quantitative PET imaging in the presurgical evaluation of MRI-negative children with focal cortical dysplasia type II. Eur J Nucl Med Mol I. 2024;51(6):1651–61.
Akeret K, Bellut D, Huppertz H, Ramantani G, König K, Serra C, et al. Ultrasonographic features of focal cortical dysplasia and their relevance for epilepsy surgery. Neurosurg Focus. 2018;45(3):E5.
Tamula G, Katyal R, Beniczky S, Nascimento FA. Focal cortical dysplasias: new advances for curing epilepsy. Epileptic Disorders: Int Epilepsy J Videotape. 2023;25(2):284.
Marinowic DR, Zanirati GG, Xavier FAC, Varella FJ, Azevedo SPDC, Ghilardi IM, et al. WNT pathway in focal cortical dysplasia compared to perilesional nonlesional tissue in refractory epilepsies. BMC Neurol. 2023;23(1):338.
Baldassari S, Ribierre T, Marsan E, Adle-Biassette H, Ferrand-Sorbets S, Bulteau C, et al. Dissecting the genetic basis of focal cortical dysplasia: a large cohort study. Acta Neuropathol. 2019;138(6):885–900.
Kato M, Kada A, Shiraishi H, Tohyama J, Nakagawa E, Takahashi Y, et al. Sirolimus for epileptic seizures associated with focal cortical dysplasia type II. Ann Clin Transl Neur. 2022;9(2):181–92.
Shiraishi H, Teramoto T, Yokoshiki S, Tohyama J, Ueda Y, Egawa K, et al. Efficacy of sirolimus for epileptic seizures in childhood associated with focal cortical dysplasia type II. Brain Dev. 2023;45(6):343–7.
Ribierre T, Bacq A, Donneger F, Doladilhe M, Maletic M, Roussel D, et al. Targeting pathological cells with senolytic drugs reduces seizures in neurodevelopmental mTOR-related epilepsy. Nat Neurosci. 2024;27(6):1125–36.
Abdijadid S, Mathern GW, Levine MS, Cepeda C. Basic mechanisms of epileptogenesis in pediatric cortical dysplasia. CNS Neurosci Ther. 2015;21(2):92–103.
Rossini L, Medici V, Tassi L, Cardinale F, Tringali G, Bramerio M, et al. Layer-specific gene expression in epileptogenic type II focal cortical dysplasia: normal-looking neurons reveal the presence of a hidden laminar organization. Acta Neuropathol Com. 2014;2:45.
Donkels C, Peters M, Fariña Núñez MT, Nakagawa JM, Kirsch M, Vlachos A, et al. Oligodendrocyte lineage and myelination are compromised in the gray matter of focal cortical dysplasia type IIa. Epilepsia. 2020;61(1):171–84.
Studer M, Rossini L, Spreafico R, Pelliccia V, Tassi L, de Curtis M, et al. Why are type II focal cortical dysplasias frequently located at the bottom of sulcus? A neurodevelopmental hypothesis. Epilepsia. 2022;63(10):2716–21.
CROME L. Abnormal brain structure in mental deficiency. Med World. 1957;86(3):217–23.
Taylor DC, Falconer MA, Bruton CJ, Corsellis JA. Focal dysplasia of the cerebral cortex in epilepsy. J Neurol Neurosurg Psychiatry. 1971;34(4):369–87.
Mühlebner A, Coras R, Kobow K, Feucht M, Czech T, Stefan H, et al. Neuropathologic measurements in focal cortical dysplasias: validation of the ILAE 2011 classification system and diagnostic implications for MRI. Acta Neuropathol. 2012;123(2):259–72.
Cepeda C, Hurst RS, Flores-Hernández J, Hernández-Echeagaray E, Klapstein GJ, Boylan MK, et al. Morphological and electrophysiological characterization of abnormal cell types in pediatric cortical dysplasia. J Neurosci Res. 2003;72(4):472–86.
André VM, Wu N, Yamazaki I, Nguyen ST, Fisher RS, Vinters HV, et al. Cytomegalic interneurons: a new abnormal cell type in severe pediatric cortical dysplasia. J Neuropath Exp Neur. 2007;66(6):491–504.
Shao Y, Ge Q, Yang J, Wang M, Zhou Y, Guo J, et al. Pathological networks involving dysmorphic neurons in type II focal cortical dysplasia. Neurosci Bull. 2022;38(9):1007–24.
Siedlecka M, Grajkowska W, Galus R, Dembowska-Baginska B, Jozwiak J. Focal cortical dysplasia: molecular disturbances and clinicopathological classification (review). Int J Mol MED. 2016;38(5):1327–37.
Wang Y, Yu T, Blümcke I, Cai Y, Sun K, Gao R, et al. The clinico-pathological characterisation of focal cortical dysplasia type IIb genetically defined by MTOR mosaicism. Neuropath Appl Neuro. 2023;49(1):e12874.
Cepeda C, André VM, Flores-Hernández J, Nguyen OK, Wu N, Klapstein GJ, et al. Pediatric cortical dysplasia: correlations between neuroimaging, electrophysiology and location of cytomegalic neurons and balloon cells and glutamate/GABA synaptic circuits. Dev Neurosci. 2005;27(1):59–76.
Sisodiya SM, Fauser S, Cross JH, Thom M. Focal cortical dysplasia type II: biological features and clinical perspectives. The Lancet Neurology. 2009;8(9):830–43.
Urbach H, Scheffler B, Heinrichsmeier T, von Oertzen J, Kral T, Wellmer J, et al. Focal cortical dysplasia of Taylor’s balloon cell type: a clinicopathological entity with characteristic neuroimaging and histopathological features, and favorable postsurgical outcome. Epilepsia. 2002;43(1):33–40.
Foglio B, Rossini L, Garbelli R, Regondi MC, Mercurio S, Bertacchi M, et al. Dynamic expression of NR2F1 and SOX2 in developing and adult human cortex: comparison with cortical malformations. Brain Struct Funct. 2021;226(4):1303–22.
Martinian L, Boer K, Middeldorp J, Hol EM, Sisodiya SM, Squier W, et al. Expression patterns of glial fibrillary acidic protein (GFAP)-delta in epilepsy-associated lesional pathologies. Neuropath Appl Neuro. 2009;35(4):394–405.
Ying Z, Gonzalez-Martinez J, Tilelli C, Bingaman W, Najm I. Expression of neural stem cell surface marker CD133 in balloon cells of human focal cortical dysplasia. Epilepsia. 2005;46(11):1716–23.
Yasin SA, Latak K, Becherini F, Ganapathi A, Miller K, Campos O, et al. Balloon cells in human cortical dysplasia and tuberous sclerosis: isolation of a pathological progenitor-like cell. Acta Neuropathol. 2010;120(1):85–96.
Fauser S, Becker A, Schulze-Bonhage A, Hildebrandt M, Tuxhorn I, Pannek HW, et al. CD34-immunoreactive balloon cells in cortical malformations. Acta Neuropathol. 2004;108(4):272–8.
Lamparello P, Baybis M, Pollard J, Hol EM, Eisenstat DD, Aronica E, et al. Developmental lineage of cell types in cortical dysplasia with balloon cells. Brain. 2007;130(Pt 9):2267–76.
Aronica E, Boer K, Redeker S, Spliet WGM, van Rijen PC, Troost D, et al. Differential expression patterns of chloride transporters, Na+-K+-2Cl–cotransporter and K+-Cl–cotransporter, in epilepsy-associated malformations of cortical development. Neuroscience. 2007;145(1):185–96.
Sánchez C, Díaz-Nido J, Avila J. Phosphorylation of microtubule-associated protein 2 (MAP2) and its relevance for the regulation of the neuronal cytoskeleton function. Prog Neurobiol. 2000;61(2):133–68.
Gonzalez-Billault C, Jimenez-Mateos EM, Caceres A, Diaz-Nido J, Wandosell F, Avila J. Microtubule-associated protein 1B function during normal development, regeneration, and pathological conditions in the nervous system. J Neurobiol. 2004;58(1):48–59.
Englund C, Folkerth RD, Born D, Lacy JM, Hevner RF. Aberrant neuronal-glial differentiation in Taylor-type focal cortical dysplasia (type IIA/B). Acta Neuropathol. 2005;109(5):519–33.
Kapar O, Gurkan ZM, Dolgun M, Sencer A, Gürses C, Bilgic B. Focal cortical dysplasia pathology: diagnostic difficulty, classification, and utility for pathogenesis. Neurosurg Focus. 2022;53(4):E6.
Abdel-Mannan O, Cheung AFP, Molnár Z. Evolution of cortical neurogenesis. Brain Res Bull. 2008;75(2–4):398–404.
Parnavelas JG, Nadarajah B. Radial glial cells. Are they really glia? Neuron. 2001;31(6):881–4.
Marin-Padilla M. Early prenatal ontogenesis of the cerebral cortex (neocortex) of the cat (Felis Domestica). A golgi study. I. The primordial neocortical organization. Z fur Anatomie und Entwicklungsgeschichte. 1971;134(2):117–45.
Fietz SA, Kelava I, Vogt J, Wilsch-Bräuninger M, Stenzel D, Fish JL, et al. OSVZ progenitors of human and ferret neocortex are epithelial-like and expand by integrin signaling. Nat Neurosci. 2010;13(6):690–9.
Cepeda C, Andre VM, Levine MS, Salamon N, Miyata H, Vinters HV, et al. Epileptogenesis in pediatric cortical dysplasia: the dysmature cerebral developmental hypothesis. Epilepsy Behav. 2006;9(2):219–35.
Crino PB, Trojanowski JQ, Eberwine J. Internexin, MAP1B, and nestin in cortical dysplasia as markers of developmental maturity. Acta Neuropathol. 1997;93(6):619–27.
Crino PB. Focal brain malformations: a spectrum of disorders along the mTOR cascade. Novartis Found Symp. 2007;288:260–72; discussion 272−81.
Lin Y, Lin K, Kang D, Liu X, Wang X, Zheng S, et al. Similar PDK1-AKT-mTOR pathway activation in balloon cells and dysmorphic neurons of type II focal cortical dysplasia with refractory epilepsy. Epilepsy Res. 2015;112:137–49.
Monuki ES, Walsh CA. Mechanisms of cerebral cortical patterning in mice and humans. Nat Neurosci. 2001;4 Suppl:1199 – 206.
Hua Y, Crino PB. Single cell lineage analysis in human focal cortical dysplasia. Cerebral cortex. 2003;13(6):693–9.
Andres M, Andre VM, Nguyen S, Salamon N, Cepeda C, Levine MS, et al. Human cortical dysplasia and epilepsy: an ontogenetic hypothesis based on volumetric MRI and NeuN neuronal density and size measurements. Cerebral cortex. 2005;15(2):194–210.
Battaglioni S, Benjamin D, Wälchli M, Maier T, Hall MN. mTOR substrate phosphorylation in growth control. Cell. 2022;185(11):1814–36.
Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21(4):183–203.
Lee WS, Stephenson SEM, Pope K, Gillies G, Maixner W, Macdonald-Laurs E, et al. Genetic characterization identifies bottom-of-sulcus dysplasia as an mTORopathy. Neurology. 2020;95(18):e2542-51.
Lee WS, Baldassari S, Stephenson SEM, Lockhart PJ, Baulac S, Leventer RJ. Cortical dysplasia and the mTOR pathway: how the study of human brain tissue has led to insights into epileptogenesis. Int J Mol Sci. 2022;23(3):1344.
Pirozzi F, Berkseth M, Shear R, Gonzalez L, Timms AE, Sulc J, et al. Profiling PI3K-AKT-MTOR variants in focal brain malformations reveals new insights for diagnostic care. Brain. 2022;145(3):925–38.
Schick V, Majores M, Engels G, Spitoni S, Koch A, Elger CE, et al. Activation of akt independent of PTEN and CTMP tumor-suppressor gene mutations in epilepsy-associated Taylor-type focal cortical dysplasias. Acta Neuropathol. 2006;112(6):715–25.
D’Gama AM, Woodworth MB, Hossain AA, Bizzotto S, Hatem NE, LaCoursiere CM, et al. Somatic mutations activating the mTOR pathway in dorsal telencephalic progenitors cause a continuum of cortical dysplasias. Cell Rep. 2017;21(13):3754–66.
Sim JC, Scerri T, Fanjul-Fernández M, Riseley JR, Gillies G, Pope K, et al. Familial cortical dysplasia caused by mutation in the mammalian target of rapamycin regulator NPRL3. Ann Neurol. 2016;79(1):132–7.
Weckhuysen S, Marsan E, Lambrecq V, Marchal C, Morin-Brureau M, An-Gourfinkel I, et al. Involvement of GATOR complex genes in familial focal epilepsies and focal cortical dysplasia. Epilepsia. 2016;57(6):994–1003.
Ribierre T, Deleuze C, Bacq A, Baldassari S, Marsan E, Chipaux M, et al. Second-hit mosaic mutation in mTORC1 repressor DEPDC5 causes focal cortical dysplasia-associated epilepsy. J Clin Investig. 2018;128(6):2452–8.
Lee WS, Baldassari S, Chipaux M, Adle-Biassette H, Stephenson SEM, Maixner W, et al. Gradient of brain mosaic RHEB variants causes a continuum of cortical dysplasia. Ann Clin Transl Neur. 2021;8(2):485–90.
Sim NS, Ko A, Kim WK, Kim SH, Kim JS, Shim K, et al. Precise detection of low-level somatic mutation in resected epilepsy brain tissue. Acta Neuropathol. 2019;138(6):901–12.
Honke J, Hoffmann L, Coras R, Kobow K, Leu C, Pieper T, et al. Deep histopathology genotype-phenotype analysis of focal cortical dysplasia type II differentiates between the GATOR1-altered autophagocytic subtype IIa and MTOR-altered migration deficient subtype IIb. Acta Neuropathol Com. 2023;11(1):179.
Lim JS, Kim WI, Kang HC, Kim SH, Park AH, Park EK, et al. Brain somatic mutations in MTOR cause focal cortical dysplasia type II leading to intractable epilepsy. Nat Med. 2015;21(4):395–400.
Lim JS, Lee JH. Brain somatic mutations in MTOR leading to focal cortical dysplasia. BMB Rep. 2016;49(2):71–2.
Hu S, Knowlton RC, Watson BO, Glanowska KM, Murphy GG, Parent JM, et al. Somatic Depdc5 deletion recapitulates electroclinical features of human focal cortical dysplasia type IIA. Ann Neurol. 2018;84(1):140–6.
Nakashima M, Saitsu H, Takei N, Tohyama J, Kato M, Kitaura H, et al. Somatic mutations in the MTOR gene cause focal cortical dysplasia type IIb. Ann Neurol. 2015;78(3):375–86.
Kim S, Baldassari S, Sim NS, Chipaux M, Dorfmüller G, Kim DS, et al. Detection of Brain Somatic Mutations in Cerebrospinal Fluid from Refractory Epilepsy Patients. Ann Neurol. 2021;89(6):1248-52.
Kim JH, Park J, Lee J, Park JW, Kim HJ, Chang WS, et al. Ultra-low level somatic mutations and structural variations in focal cortical dysplasia type II. Ann Neurol. 2023;93(6):1082–93.
Lee WS, Leventer RJ, Lockhart PJ. Droplet digital PCR as a first-tier molecular diagnostic tool for focal cortical dysplasia type II. Brain. 2022;145(12):e119-21.
Klein KM, Mascarenhas R, Merrikh D, Khanbabaei M, Maroilley T, Kaur N, et al. Identification of a mosaic MTOR variant in purified neuronal DNA in a patient with focal cortical dysplasia using a novel depth electrode harvesting technique. Epilepsia. 2024;65(6):1768–76.
Xu Y, Zhao R, Wang M, Wang X, Wang Y, Li H, et al. Identification of genetic characteristics in pediatric epilepsy with focal cortical dysplasia type 2 using deep whole-exome sequencing. Mol Genet Genom Med. 2022;10(12):e2086.
Zhang Z, Gao K, Liu Q, Zhou J, Li X, Lang N, et al. Somatic variants in new candidate genes identified in focal cortical dysplasia type II. Epilepsia. 2020;61(4):667–78.
Li X, Wang T, Liu N, Cai A, Zhang J, Zhang F, et al. Focal cortical dysplasia II caused by brain somatic mutation of IRS-1 is associated with ERK signaling pathway activation. Cerebral cortex. 2024;34(6):bhae227.
Jesus-Ribeiro J, Pires LM, Melo JD, Ribeiro IP, Rebelo O, Sales F, et al. Genomic and epigenetic advances in focal cortical dysplasia types I and II: a scoping review. Front Neurosci. 2021;14:580357.
Moore LD, Le T, Fan G. DNA methylation and its basic function. Neuropsychopharmacology: Official Publication Am Coll Neuropsychopharmacol. 2013;38(1):23–38.
Delgado-Morales R, Agís-Balboa RC, Esteller M, Berdasco M. Epigenetic mechanisms during ageing and neurogenesis as novel therapeutic avenues in human brain disorders. Clin Epigenetics. 2017;9:67.
Dixit AB, Sharma D, Tripathi M, Srivastava A, Paul D, Prakash D, et al. Genome-wide DNA methylation and RNAseq analyses identify aberrant signalling pathways in focal cortical dysplasia (FCD) type II. Sci Rep. 2018;8(1):17976.
Kobow K, Ziemann M, Kaipananickal H, Khurana I, Mühlebner A, Feucht M, et al. Genomic DNA methylation distinguishes subtypes of human focal cortical dysplasia. Epilepsia. 2019;60(6):1091–103.
Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.
Lee JY, Park A, Lee E, Park W, Park S, Choi JW, et al. miRNA expression analysis in cortical dysplasia: regulation of mTOR and LIS1 pathway. Epilepsy Res. 2014;108(3):433–41.
Li L, Liu C, Li T, Guan Y, Zhou J, Qi X, et al. Analysis of altered micro RNA expression profiles in focal cortical dysplasia IIB. J Child Neurol. 2016;31(5):613–20.
Jheng G, Hur SS, Chang C, Wu C, Cheng J, Lee H, et al. Lis1 dysfunction leads to traction force reduction and cytoskeletal disorganization during cell migration. Biochem Bioph Res Co. 2018;497(3):869–75.
Huang J, Wu S, Barrera J, Matthews K, Pan D. The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila homolog of YAP. Cell. 2005;122(3):421–34.
Heng JI, Nguyen L, Castro DS, Zimmer C, Wildner H, Armant O, et al. Neurogenin 2 controls cortical neuron migration through regulation of Rnd2. Nature. 2008;455(7209):114–8.
Avansini SH, Torres FR, Vieira AS, Dogini DB, Rogerio F, Coan AC, et al. Dysregulation of NEUROG2 plays a key role in focal cortical dysplasia. Ann Neurol. 2018;83(3):623–35.
Boer K, Troost D, Spliet WGM, van Rijen PC, Gorter JA, Aronica E. Cellular distribution of vascular endothelial growth factor A (VEGFA) and B (VEGFB) and VEGF receptors 1 and 2 in focal cortical dysplasia type IIB. Acta Neuropathol. 2008;115(6):683–96.
Shen K, Duan Q, Duan W, Xu S, An N, Ke Y, et al. Vascular endothelial growth factor-C modulates cortical NMDA receptor activity in cortical lesions of young patients and rat model with focal cortical dysplasia. Brain Pathol. 2022;32(5):e13065.
Ueda M, Sugiura C, Ohno K, Kakita A, Hori A, Ohama E, et al. Immunohistochemical expression of fibroblast growth factor-2 in developing human cerebrum and epilepsy-associated malformations of cortical development. Neuropathology. 2011;31(6):589–98.
Chung C, Yang X, Bae T, Vong KI, Mittal S, Donkels C, et al. Comprehensive multi-omic profiling of somatic mutations in malformations of cortical development. Nat Genet. 2023;55(2):209–20.
Aronica E, Ozbas-Gerçeker F, Redeker S, Ramkema M, Spliet WGM, van Rijen PC, et al. Expression and cellular distribution of high- and low-affinity neurotrophin receptors in malformations of cortical development. Acta Neuropathol. 2004;108(5):422–34.
Schachenhofer J, Gruber V, Fehrer SV, Haider C, Glatter S, Liszewska E, et al. Targeting the EGFR pathway: an alternative strategy for the treatment of tuberous sclerosis complex? Neuropath Appl Neuro. 2024;50(2):e12974.
Sugiura C, Miyata H, Ueda M, Ohama E, Vinters HV, Ohno K. Immunohistochemical expression of fibroblast growth factor (FGF)-2 in epilepsy-associated malformations of cortical development (MCDs). Neuropathology. 2008;28(4):372–81.
Huang T, Liu D, Wang Y, Li P, Sun L, Xiong H, et al. FGFR2 promotes gastric cancer progression by inhibiting the expression of thrombospondin4 via PI3K-Akt-mtor pathway. Cell Physiol Biochem. 2018;50(4):1332–45.
Quarto N, Wan DC, Longaker MT. Molecular mechanisms of FGF-2 inhibitory activity in the osteogenic context of mouse adipose-derived stem cells (mASCs). Bone. 2008;42(6):1040–52.
Gipson GR, Goebel EJ, Hart KN, Kappes EC, Kattamuri C, McCoy JC, et al. Structural perspective of BMP ligands and signaling. Bone. 2020;140:115549.
Guo W, Zhang CQ, Shu HF, Yang MH, Yin Q, Yang H. Expression of bone morphogenetic protein-4 in the cortical lesions of focal cortical dysplasia IIb and the tuberous sclerosis complex. J Mol Neurosci. 2013;50(1):7–13.
Nosten-Bertrand M, Kappeler C, Dinocourt C, Denis C, Germain J, Phan Dinh Tuy F, et al. Epilepsy in Dcx knockout mice associated with discrete lamination defects and enhanced excitability in the hippocampus. PLoS One. 2008;3(6):e2473.
Boekhoorn K, Sarabdjitsingh A, Kommerie H, de Punder K, Schouten T, Lucassen PJ, et al. Doublecortin (DCX) and doublecortin-like (DCL) are differentially expressed in the early but not late stages of murine neocortical development. J Comp Neurol. 2008;507(4):1639–52.
Boer K, Lucassen PJ, Spliet WG, Vreugdenhil E, van Rijen PC, Troost D, et al. Doublecortin-like (DCL) expression in focal cortical dysplasia and cortical tubers. Epilepsia. 2009;50(12):2629–37.
Huber AB, Weinmann O, Brösamle C, Oertle T, Schwab ME. Patterns of Nogo mRNA and protein expression in the developing and adult rat and after CNS lesions. J Neuroscience. 2002;22(9):3553–67.
Yu SX, Li S, Shu HF, Zhang CQ, Liu SY, Yang H. Expression of the Nogo-A system in cortical lesions of pediatric patients with tuberous sclerosis complex and focal cortical dysplasia type IIb. J Neuropath Exp Neurol. 2012;71(7):665–77.
Yue J, Zhang C, Shi X, Wei Y, Liu L, Liu S, et al. Activation of leukocyte immunoglobulin-like receptor B2 signaling pathway in cortical lesions of pediatric patients with focal cortical dysplasia type IIb and tuberous sclerosis complex. Brain Dev-JPN. 2019;41(10):829–38.
Filbin MT. PirB, a second receptor for the myelin inhibitors of axonal regeneration Nogo66, MAG, and OMgp: implications for regeneration in vivo. Neuron. 2008;60(5):740–2.
Cotter D, Honavar M, Lovestone S, Raymond L, Kerwin R, Anderton B, et al. Disturbance of Notch-1 and Wnt signalling proteins in neuroglial balloon cells and abnormal large neurons in focal cortical dysplasia in human cortex. Acta Neuropathol. 1999;98(5):465–72.
Zhang L, Bartley CM, Gong X, Hsieh LS, Lin TV, Feliciano DM, et al. MEK-ERK1/2-dependent FLNA overexpression promotes abnormal dendritic patterning in tuberous sclerosis independent of mTOR. Neuron. 2014;84(1):78–91.
Zhang L, Huang T, Teaw S, Nguyen LH, Hsieh LS, Gong X, et al. Filamin a inhibition reduces seizure activity in a mouse model of focal cortical malformations. Sci Transl Med. 2020;12:531.
Prayson RA. Dual pathology in Rasmussen’s encephalitis: a report of coexistent focal cortical dysplasia and review of the literature. Case Rep Pathol. 2012;2012:569170.
Tang C, Wang X, Deng J, Xiong Z, Guan Y, Zhou J, et al. Increased inflammasome-activated pyroptosis mediated by caspase-1 in Rasmussen’s encephalitis. Epilepsy Res. 2022;179:106843.
Rossini L, Villani F, Granata T, Tassi L, Tringali G, Cardinale F, et al. FCD type II and mTOR pathway: evidence for different mechanisms involved in the pathogenesis of dysmorphic neurons. Epilepsy Res. 2017;129:146–56.
Wei YJ, Guo W, Sun FJ, Fu WL, Zheng DH, Chen X, et al. Increased expression and cellular localization of P2X7R in cortical lesions of patients with focal cortical dysplasia. J Neuropathol Exp Neurol. 2016;75(1):61–8.
Arena A, Zimmer TS, van Scheppingen J, Korotkov A, Anink JJ, Mühlebner A, et al. Oxidative stress and inflammation in a spectrum of epileptogenic cortical malformations: molecular insights into their interdependence. Brain Pathol. 2019;29(3):351–65.
Karin M, Ben-Neriah Y. Phosphorylation meets ubiquitination: the control of NF-[kappa]B activity. Annu Rev Immunol. 2000;18:621–63.
Guo W, Zheng D, Sun F, Yang J, Zang Z, Liu S, et al. Expression and cellular distribution of the interleukin 2 signaling system in cortical lesions from patients with focal cortical dysplasia. J Neuropath Exp Neur. 2014;73(3):206–22.
Zhang Z, Liu Q, Liu M, Wang H, Dong Y, Ji T, et al. Upregulation of HMGB1-TLR4 inflammatory pathway in focal cortical dysplasia type II. J Neuroinflamm. 2018;15(1):27.
Huang K, Wang Z, He Z, Li Y, Li S, Shen K, et al. Downregulated formyl peptide receptor 2 expression in the epileptogenic foci of patients with focal cortical dysplasia type IIb and tuberous sclerosis complex. Immun Inflamm Dis. 2022;10(11):e706.
Rossini L, De Santis D, Cecchini E, Cagnoli C, Maderna E, Cartelli D, et al. Dendritic spine loss in epileptogenic type II focal cortical dysplasia: role of enhanced classical complement pathway activation. Brain Pathol. 2023;33(3): e13141.
Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149(2):274–93.
Raymond AA, Fish DR, Sisodiya SM, Alsanjari N, Stevens JM, Shorvon SD. Abnormalities of gyration, heterotopias, tuberous sclerosis, focal cortical dysplasia, microdysgenesis, dysembryoplastic neuroepithelial tumour and dysgenesis of the archicortex in epilepsy. Clinical, EEG and neuroimaging features in 100 adult patients. Brain. 1995;118(Pt 3):629–60.
Kamada T, Sun W, Takase K, Shigeto H, Suzuki SO, Ohyagi Y, et al. Spontaneous seizures in a rat model of multiple prenatal freeze lesioning. Epilepsy Res. 2013;105(3):280–91.
Colacitti C, Sancini G, DeBiasi S, Franceschetti S, Caputi A, Frassoni C, et al. Prenatal methylazoxymethanol treatment in rats produces brain abnormalities with morphological similarities to human developmental brain dysgeneses. J Neuropath Exp Neur. 1999;58(1):92–106.
Inverardi F, Chikhladze M, Donzelli A, Moroni RF, Regondi MC, Pennacchio P, et al. Cytoarchitectural, behavioural and neurophysiological dysfunctions in the BCNU-treated rat model of cortical dysplasia. Eur J Neurosci. 2013;37(1):150–62.
Nguyen LH, Bordey A. Current review in basic science: animal models of focal cortical dysplasia and epilepsy. Epilepsy Curr. 2022;22(4):234–40.
Acknowledgements
The authors have no acknowledgements to declare.
Funding
This review was supported by the Shenzhen Municipal Science and Technology key projects of the Basic Research Program (file no. LMZ JCYJ20220818102007015) and the National Natural Science Foundation of China (file no. LMZ 82071447, 82371456).
Author information
Authors and Affiliations
Contributions
All authors participated in the writing of this manuscript. YF and YZ conceptualized the content scope of the article and wrote the first manuscript. TH, SY and YL prepared the figures. LZ reviewed and edited the manuscript. The authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Ethics approval and consent to participate is not applicable in this study.
Consent for publication
Consent for publication is not applicable in this study.
Competing interests
Author Liemin Zhou is the member of the Editorial Board for Acta Epileptologica, who was not involved in the journal’s review of or decisions related to this manuscript.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Fang, Y., Zhang, Y., Huang, T. et al. Focal cortical dysplasia type II: review of neuropathological manifestations and pathogenetic mechanisms. Acta Epileptologica 7, 12 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s42494-024-00195-y
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s42494-024-00195-y