U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Noebels JL, Avoli M, Rogawski MA, et al., editors. Jasper's Basic Mechanisms of the Epilepsies [Internet]. 4th edition. Bethesda (MD): National Center for Biotechnology Information (US); 2012.

  • This title is an author manuscript version first made accessible on the NCBI Bookshelf website July 2, 2012.

This title is an author manuscript version first made accessible on the NCBI Bookshelf website July 2, 2012.

Cover of Jasper's Basic Mechanisms of the Epilepsies

Jasper's Basic Mechanisms of the Epilepsies [Internet]. 4th edition.

Show details

Developing Models of Aristaless-related homeobox mutations

and .

Author Information and Affiliations

Mutations in the Aristaless-related homeobox gene (ARX) have been causally linked to a variety of neurological conditions, particularly, infantile spasms syndrome. ARX is a developmentally regulated homeobox transcription factor with expression both in the ganglionic eminence and the cortical ventricular zone early in development1. Postnatally, the expression pattern is restricted to GABAergic neurons in the cortex and basal ganglia. During development, ARX functions primarily as a transcriptional repressor2: modulating migration and fate specification of interneurons and controlling ventricular zone proliferation. How loss of function of ARX leads to an epilepsy phenotype is poorly understood. Three genetically modified mice lines have been generated3–5 to address this issue. These models each develop epilepsy and all have changes in interneuron subtype patterns strongly implicating alterations of interneuron development as a cause of epilepsy. Analysis of these models will both further the molecular understanding of the function of ARX and allow dissection of the pathophysiological properties of the ARX related epilepsies. This chapter will review the current knowledge of the function of Arx, the Arx mouse models, and discuss how these models can lead to a better understanding of the role of interneuron loss in the development of epilepsy during early childhood.

INTRODUCTION

During the first few years of life there exists a spectrum of malignant epileptic disorders. These early epileptic encephalopathies often proceed from one clinical syndrome to another. The named syndromes include Early Infantile Epileptic Encephalopathy (Otahara’s Syndrome), Severe Myoclonic Epilepsy of Infancy (Dravet’s Syndrome), Infantile Spasms (West-Syndrome), Lennox-Gastaut Syndrome and a few others (for review see6, as well as various chapters in this book). Up until the last few years, the etiology of these conditions was unknown for many patients. Recently, a number of genes have been linked to early epileptic encephalopathies and often a single gene has been found to be responsible for multiple conditions. The Aristeless-related Homeobox gene (ARX) is, to date, one of the genes most commonly associated with these conditions. A sizeable literature has accumulated describing mutations in ARX that have been reported to cause a variety of neurological disorders, all with epilepsy as a major symptom. ARX was first associated with human disease by three groups in 2002.7–9 The initial descriptions were of 3 different disorders: X-linked Lissencephaly with ambiguous genitalia (XLAG), X-linked mental retardation (XLMR) with epilepsy and spasticity, and X-linked Infantile Spasms Syndrome (ISSX). The finding of a single gene having significant clinical and genetic heterogeneity has lead to a considerable amount of research, both clinical and basic science, on this gene. In this chapter, we will introduce the clinical and molecular details of ARX, then discuss the molecular mechanisms of the gene, the rodent models of ARX related disease, and end with a discussion of the role of ARX in the pathophysiology of the early epileptic encephalopathies.

ARX IN CLINICAL NEUROLOGY

Mutations in the gene ARX, have been primarily described in boys, and generally result in a clinical phenotype with intractable epilepsy as a predominant feature. The mutations can be broadly classified as those resulting in brain malformations and those without brain malformations. The brain malformation syndromes include XLAG8,10, Proud syndrome (Agenesis of the Corpus Callosum and ambiguous genitalia)11 and hydranencephaly with abnormal genitalia12 (OMIM 300215). The ARX disorders without brain malformations include XLMR7,9, West Syndrome/ISSX8,13, Partington syndrome (mental retardation with hand dystonia)14–16 (OMIM 309510), X-linked myoclonic epilepsy with spasticity and intellectual development (XMESID)17,18, and Ohtahara/Early Infantile Epileptic Encephalopathy Syndrome (EIEE)19–21, and other cases with epilepsy, intellectual disability, and dystonia/spasticity as part of the syndrome.22,23 A number of patients with ARX mutations, both with and without clear brain malformations have also been observed to have cysts within the basal ganglia.4,12,13 Though not a clear malformation, this is a finding that may be typical in patients with ARX mutations.

In females, the only CNS malformation currently described is agenesis of the corpus callosum (ACC) with or without mental retardation (MR) and seizures.4,12 Females have also been reported to have infantile spasms, epilepsy, and varying degrees of cognitive dysfunction without clear brain malformation.4,12,24,25 All of these conditions have been linked to a large number of mutations in the gene. From this large group of mutations and conditions, a number of authors have debated the existence of a clear genotype-phenotype correlation.12,26,27

Ostensibly, a clear genotype-phenotype correlation exists between the malformation phenotypes and the non-malformation phenotypes. Mutations leading to loss of protein function (deletions, non-sense, frame-shift, or splice site mutations) or to changes in the homeodomain (for details see below) lead to malformation syndromes, while missense mutations or expansions of the polyalanine tracts lead to epilepsy phenotypes with normal brain MRIs.12 However, this genotype-phenotype correlation breaks down when one looks at severity of disease in the non-malformation phenotypes and females.26 For example, the most common mutation reported (45% of all mutations), the expansion of the second polyalanine tract from 12–20 alanines, has been associated with a spectrum of phenotypes from mild MR and seizures to the severe phenotype of Ohtahara Syndrome, suggesting greater genotype-phenotype heterogeneity. Another instance where the genotype-phenotype correlation is less clear is in females with ARX mutations. There are mother-daughter and mother-aunt pairs where one is normal and the other has cognitive disabilities and epilepsy4, though this may be due to differences in X-inactivation. Overall, ARX mutations result in a spectrum of disorders all with epilepsy as a major phenotype. A degree of genotype-phenotype correlation exists, but other factors, such as familial modifiers or environmental mediators, must exist to explain the variability that does occur.

MOLECULAR BIOLOGY OF ARX

What Is ARX?

ARX encodes a transcription factor with a role in cortical development. Designated ARX in humans and Arx in mice, the gene codes for a paired class homeodomain protein and is the vertebrate ortholog to the Drosophila aristaless (al) gene. ARX resides at Xp22.11 and produces a 2.8-kb mRNA with significant homology between human and mouse (89.3% sequence and 95% amino acid homology). The gene encodes a 562 amino acid protein, localized to the nucleus, that functions by binding DNA.7,28,29 Arx is expressed in the developing hypothalamus, thalamus, basal ganglia and cerebral cortex beginning at embryonic day 8 (E8). In the forebrain, Arx is expressed in the anterior neural plate at E8.7,30 By E10.5, expression is clearly observed in the ventricular zone (VZ) of the dorsal cortex and begins to be in the mantle zone of the emerging ganglionic eminences (GE).30 Ventral telencephalic Arx expression is clearly present in the mantle zone of the GE at E12.5.30 By E14.5 Arx protein can be observed in the dorsal VZ, intermediate zone (IZ) of the GE, and in scattered cells in the marginal zone (MZ) and dorsal IZ and subventricular zone (SVZ)7,30,31 (Figure 1A). The ventral expression, at E14.5, has settled into the SVZ of the GE, primarily in proliferating neuroblasts30 as determined by co-labeling with Bromodeoxyuridine (BrdU) staining. Arx remains expressed as the cells leave the SVZ to tangentially migrate into the cortex.8,30,32 The ventral and migratory expression of Arx has almost complete overlap with Gad1 (Glutamic acid decarboxylase 1) expression.30 This expression pattern remains until shortly after birth, when the dorsal VZ and GE is no longer present and Arx expression in the forebrain is observed only in scattered cells in the cortex and basal ganglia (Figure 1B,C). Co-labeling experiments with different interneuron markers has revealed that greater than 75% of all interneurons are Arx positive.29–31,33,34 Arx is also expressed in the developing pancreas, testis, and muscle. Data from the pancreas suggests an important role of Arx in fate determination.35–37 The role of ARX in the brain appears to be more complicated then in the pancreas.

Ontogeny of Arx protein staining in dorsal neocortex of a wild type mouse.

Figure

Ontogeny of Arx protein staining in dorsal neocortex of a wild type mouse. E14.5, E18.5, and P14 coronal sections are presented. A. E14.5 coronal section (4×) with Arx staining (dark brown) in the GE and in the dorsal VZ (asterisk). Two trails (more...)

The functional role of Arx in the telencephalon is an active area of research. Recent studies suggest that Arx is primarily a transcriptional repressor2,38, and through this mechanism, has important roles in pallial progenitor cell proliferation, non-radial cell migration of interneurons from the ganglionic eminence, and basal ganglia development.8,32

Arx as a Transcriptional Repressor

The Aristaless homeodomain protein in drosophila is a paired related homeobox protein, distinguished by containing a paired homeodomain region, a C-terminal aristaless domain, and an N-terminal octapeptide/engrailed domain.29,39 The aristaless domain is known to act primarily as a repressor40 and the octapeptide domain is believed to be a potent transcriptional repressor.41 Based on the findings in Arx homologues, the transcriptional activity of murine Arx has been investigated and also found to be primarily repressive in nature.2,38,42 Using a Gal4-reporter assay, the engrailed/octapeptide domain, a region near the C-terminus, and the 4th polyalanine tract were found to mediate transcriptional repression.38,42 The transcriptional repression of the engrailed domain was determined to act via the Transducin-like enhancer (TLE) proteins38, which are known co-factors for engrailed domain containing proteins. Indeed, in the same reporter system, introduction of mutations believed to cause XLMR and ISSX reduced repression of the reporter construct38,42 by decreasing the binding to the TLE elements. Arx also functions as a transcriptional promoter, though this is not believed to be as prominent an action of the transcription factor.2,38 As the data fairly well establishes Arx as primarily an inhibitor of transcriptional programs, two questions remain, which gene networks are affected, and how does this repression alter normal cortical development?

Attempting to answer these questions, two labs performed mouse whole genome transcriptional arrays followed by bio-informatic analyses to begin to elucidate the downstream targets of Arx.2,43 In these experiments, the ventral telenecephalon, including the GE, developing basal ganglion, and hypothalamic regions were dissected away from the cortex, and their RNA isolated and hybridized to Affymetrix whole genome transcriptional arrays. Both studies demonstrated that the majority of differentially expressed genes were up-regulated in the KO animals. Many of these were genes previously not (or minimally) expressed in the ventral forebrain, suggesting that Arx plays an important role in regionalization and fate determination by suppressing the expression of other transcription factors. The loss of Arx also resulted in the decreased expression of a subset of differentially expressed genes, presumably ones that are up-regulated by Arx. Some of these latter genes appear to be important in the control of migration and proliferation.2,43 Together, these experiments confirmed the function of Arx as primarily a repressor, and began to define the downstream targets of Arx -related transcriptional networks that are important in ventral forebrain development.

Role in Interneuron Development

The above studies suggest that Arx plays an important role in ventral forebrain development. This is likely for basal ganglia formation, interneuron production, migration, and specification, as well as cholinergic neuronal specification. There is much interest in the interneuron phenotype because of the intractable epilepsy present in most patients with ARX mutations. With one of the original three clinical descriptions, a germ line knockout of Arx was developed and experiments on these animals showed a clear alteration in interneuronal development.8 The KO mouse data and other subsequent experiments have confirmed the important role of Arx in this developmental process. The expression pattern data described above (for example see Figure 1) further corroborates that Arx is essential for normal interneuron development.8,9,30,31 How Arx guides interneuronal development is less well understood.

A number of different transcription factors have important roles during interneuron development (for review see.44,45). Early (E8–9) Mash1 expression sets ventral forebrain identity46, and subsequent expression of Dlx1/2 is vital for interneuronal fate determination.47,48 Subsequently, ventral regionalization occurs by the sequential expression of various transcription factors including Nkx2.1 and Lhx6.1. Accumulating evidence begins to place Arx within this transcriptional network, with Arx as a direct target of Dlx1/234 by acting through an enhancer element over 12kB downstream from the end of the coding region. Activation of Arx by Dlx1/2 is thought to mediate the Dlx dependent interneuron migration but not specification of GABAergic cell fate.34,49 Loss of Arx increases the expression of Gbx1, Lhx6, Dlx2, Nkx2., Magel2, Efb3, and Lmo3 into ventral regions with minimal previous expression2,32,34 but decreases the expression of Cxcr4 and Bmper.34 From the results of these experiments, the effect of proper Arx expression is the correct specification of ventral areas into regions producing interneurons and subsets of basal ganglia neurons and the appropriate expression of proteins necessary for interneuronal migration (Cxcr4 and Ebf3). Exact mechanisms need to be further elucidated, but a framework for understanding how Arx disrupts interneuronal development is now present.

Role in Cortical Ventricular Zone Development

In contrast to the emerging evidence defining a mechanism of Arx action in interneuronal development, much less is known about the role in dorsal cortex during development. Arx expression is first observed in the dorsal VZ at E12.5, but by birth (P0), most VZ progenitor cells have matured and have entered the cortex with resultant loss of Arx expression. As stated in the previous sections, all postnatal expression of Arx is in interneurons and not the excitatory cells that are derived from the dorsal VZ. Arx expression in the dorsal VZ is under a presumed regulatory element in an ultraconserved region in the 3.5kB downstream of the Arx coding region. The function of Arx in the VZ is believed to be in controlling proliferation, and a number of pieces of evidence support this role. First, humans with ARX mutations can develop a form of lissencephaly, with only 3 thin layers and thickened white matter, suggesting a proliferative defect. Second, the germline knockout mice have a thin cortex and a small size of the forebrain.5,8 Third, the expression in the VZ apparently terminates when the cells enter the SVZ or IZ. Finally, in-utero electroporation experiments altering Arx levels in the VZ (by shRNA or Arx over expressing vector) described changes in cell cycle time and number of cells proliferating in the VZ.49 These authors showed a lack of cell death, but could not differentiate between Arx being involved in cell cycle timing and proliferation or neuronal differentiation, but they concluded that the role is in cell proliferation and cell cycle timing.49 Though Arx is likely involved in cell proliferation via controlling cell cycle dynamics, the downstream targets for this role and its location within a gene regulatory network in the dorsal cortex is not known. Research into this important area of Arx function is on-going.

Role of Expansion Mutations

A unique feature of Arx is the presence of 4 alanine repeats within the gene. A number of genes are known to contain polyalanine tracts, with the majority acting as transcription factors or having nuclear localization signals in the protein.50 Polyalanine repeats, unlike the more common/well known polyglutamine repeats, are shorter, averaging less than 20 alanines, more stable (lacking genetic anticipation), and presumed to cause disease by either protein misfolding or altering nuclear localization.50,51 Nine diseases are associated with expansions in these alanine tracts, 8 of which are transcription factors and result in developmental disorders similar to those caused by Arx. These results led to a series of experiments to determine the effect of different expansions on Arx function.52,53 Expansions in the first polyalanine tract from 16 to 23 alanines resulted in protein aggregations, with two labs finding these protein inclusions in different cellular locations. Our lab reported the presence of intranuclear inclusions52 while another lab described cytoplasmic aggregations.53 Both labs used over-expression experiments in cell culture. It is not clear why the two studies showed a difference in location of the protein aggregates as both used similar cell lines and culture methods, suggesting that subtle differences in methodology can lead to alterations in protein processing. Importantly, to understand the functional implication of the inclusions, our lab showed increased cell death in HEK cells and both partial reduction of the number of cells with inclusions and partial rescue of the cell death response by over expressing the heat shock protein HSP-70.52 Mechanistically, the polyalanine expansion may induce inclusions and cellular aggregations, not by altering the binding of Arx with one potential nuclear import protein, IPO13, but rather due to changes in protein-protein interactions.53 Finally, the expansions were found to occur in-vivo after in-utero electroporation.52 Inclusions, however, were not observed in the two expansion mouse models that have been recently generated.3,5 One expanded mouse model, did report alterations in cellular localization of the Arx protein3, partially consistent with some of cell culture experiments described above.53

Recent, work has begun to address the issue of mislocalization of Arx, though not with the expanded mutation. A few studies have addressed this issue, by producing full length Arx cDNA constructs with mutations/disruptions in the nuclear localization sequence (NLS) of Arx.54,55 Arx contains at least 3 NLSs, which are found in the homeodomain or in the 3′ region and alterations in these sequences cause nuclear or perinuclear protein aggregations. The transport of Arx into the nucleus is protein dependent, particularly the importins, importin 13 (IPO13)55 and importin β.54 The possibility that many ARX mutations, both expansions and point mutations, alter ARX binding, inhibiting nuclear import was tested. Both groups found the change in cellular localization occurs without affecting binding to nuclear import proteins54,55, suggesting that they are purely mistargeted, not altered in protein binding. This work is limited as there could be alterations in the binding to other Arx co-factors, either for nuclear import or DNA binding, and these may lead to the cellular aggregations or nuclear inclusions. Of note, in a single patient with XLAG, not due to an expansion of the polyalanine tract, no inclusions or aggregations were noted on histopathology.10 This data implies that in the more severe mutations, there may be alterations in protein processing and, hence, no aggregations. Further research is needed to fully understand how the protein expansion leads to the Arx clinical phenotypes.

ARX MODELS

Since the first description of Arx related disorders a number of Arx mouse models have been developed.3–5,8,36 These models have led to different insights into the disorder. The first mouse was made by homologous recombination of a lacZ containing vector into exon 2, producing an Arx allele with a premature stop codon8 and resulting in a truncated protein. These mice died at P0-P2, but were found to have many of the features of XLAG in humans, including a poorly formed small cortex, absent corpus collosum, abnormal tangential migration and differentiation of interneurons, and abnormalities in the male genitalia.8 There was a severe alteration in interneuron migration, with a delay in onset and loss of all migrating interneurons except for those along the SVZ.8 These authors also uncovered changes in basal ganglia cell populations. Because of the early lethality of the mice, no behavioral or electrophysiological studies could be performed. Shortly afterwards, Collombat and colleagues created a mouse line with targeted knockout of exon 1 and 2 producing a mutant protein with the loss of the first 360 N-terminal amino acids.36 These mice are born but die by P2 and are smaller and appear dehydrated.36 The mice were first developed to study the role of Arx in pancreatic development, but have subsequently been used to study both the down stream targets of Arx and the alterations in interneurons, basal ganglia, and cholinergic neurons, as described in the preceding sections.30,32,34,43 As these mice were also perinatal lethal, attributable to loss of glucagon producing alpha cells, no phenotypic or physiological analysis could be performed. The functional shortcomings of these two mice lines were solved with a series of 5 Arx model mice lines that were published in 2009.

The latter Arx models took two approaches to developing the mice. The first approach, from our lab, used the Cre-Lox system to stop expression of Arx in cells expressing Dlx5/6, a transcription factor that is expressed in the majority of developing interneurons within the GE.4 The Dlx5/6 promoter was chosen to drive the Cre recombinase as loss of interneurons in patients with Arx mutations are believed to be a major contributor to the epilepsy and infantile spasms phenotype.56 We were, therefore, attempting to prove the hypothesis that interneuron dysfunction is central to the epilepsy phenotype in various brain malformations. Indeed, the conditional knockout (CKO) mice appear to give credence to this view. The adult CKO male mice had faster, higher voltage EEG tracings with multifocal spikes reminiscent of a hypsarrythmic pattern seen in infants.4 The male mice develop convulsive seizures at P14, the earliest time we were able to accurately assess for the presence of electrographic seizures. The male CKO mice, which live past P14, continue to have seizures and develop an infantile spasms like seizure type. The development of the spasm phenotype was found more in the adult animals, most likely due to difference in maturity and neuroanatomy between mice and humans. In addition, we found that approximately half of the female animals also developed seizures at P14 which persisted into adulthood. The loss (or partial loss) of Arx reduced the numbers of calbindin positive interneurons.4 Currently, we are determining if there are other interneuronal changes as well as the developmental time course of this loss. Our approach contrasts with the method of the two other laboratories that recently made Arx models.

The approach taken by these labs was to generate mice with known human mutations associated with disease.3,5 The Noebels’ lab, developed a knock-in Arx mouse with an expansion of the first polyalanine tract, increasing the repeat size from 16 to 23 amino acids.3 These authors found the animals to have possible behavioral spasms between 7–11 days of age, electroclinical spasms between 14–21 days of age, then a development of other seizure types (behavioral arrest and motoric seizures) as the animals mature.3 Behavioral tests on these animals revealed deficits in anxiety, fear conditioning, and social behaviors. Notably, the animals were less anxious and had diminished retention of a conditioned stimulus compared to wild-type mice, suggesting more of an anxiety phenotype, then diminished ability to learn.3 The pathological alteration of the Arx expansion was a regional loss of a population of Arx expressing interneurons. A 50% reduction of Arx was found in the hilus of the hippocampus and straitum and a 68% decrease was discovered in somatosensory and motor cortex, but no change in parietal cortex.3 The authors reported a change in protein localization from nuclear to cytoplasmic, but this contrasts with the other Arx expanded mouse, which found no inclusions or alteration of protein localization.5 The partial loss of Arx positive interneurons was due primarily from loss of Calbindin positive cells, similar to the changes found in our Dlx5/6 conditional knockout mouse line. There was no loss of neuropeptide Y or Calretinin positive cells in the cortex. Finally, the authors reported a loss of Calbindin, NPY and cholinergic neurons in the striatum.3 Overall, the features of this mouse model were similar to the Arx conditional knockout.

The third group in the Kitamura laboratory generated three mouse lines. Two were knock-ins of known pathogenic mutations, both disrupting the homeodomain, but one mutation was associated with the XLAG phenotype and the other with the XLMR phenotype.5 The third line derived by this group was an expansion of 7 alanines to the first polyalanine tract5, a line similar to the mouse made by the Noebels’ lab. The XLAG mutation mouse line was similar in phenotype to the germ line knockout. This mouse was perinatal lethal, had smaller brains, severely diminished numbers of interneurons in the cortex, and had a large loss of cholinergic neurons in the striatum.5 The other two lines were viable and recapitulated aspects of the XLMR or ISSX phenotypes. These two lines both develop spontaneous seizures but of differing severities. Only 10% of XLMR point mutation mice were observed to have a seizure and no seizures were captured on video EEG, in the 3 mice that were recorded. In contrast, 70% of the expanded mice developed behavior seizures, and all (3/3) mice with EEG recordings developed seizures, but no interictal EEG abnormalities.5 Like the Noebels’ lab expansion mice, the two lines were tested for behavioral deficits. Both lines were observed to have impaired motor coordination and increased anxiety. The two lines were also found to learn poorly in both a passive avoidance task and a radial arm task, which tests hippocampal spatial memory.5 The differences in anxiety and motor phenotypes between the Kitamura and Noebels’ expanded mice could be due to the different background strains, C57BL/6J x 129Sv/SvEvBrd mice3 versus pure C57BL/6J mice5, or to other differences with strain generation, suggesting that in humans, genetic background and environmental features may modify the phenotypic expression in patients with the expansion mutation. This is indeed what other authors have proposed for the variability of phenotypes observed in patients with expansions of the first and second polyalanine tracts.26

As with the other conditional lines, the two viable lines from the Kitamura lab also had deficiencies in interneuron and striatal neurons. In these animals, interneurons began to migrate at the appropriate time (E12.5) and migrated normally, but with reduced numbers of interneurons entering the cortex. Although the total numbers of interneurons were reduced in postnatal mice, these authors did not find an interneuron subtype-specific difference in either the XLMR or ISSX mutations.5 This contrasts with what was found in the basal ganglia of these mice. In the striatum, the XLAG mutation resulted in both a loss of radially and tangential migration, whereas the two less severe mutations only altered tangential migration of interneurons into the basal ganglia. Finally, there was also a loss of cholinergic neurons in the cortex and basal ganglia, but it was greater in the striatum.5 Overall, these five mouse lines have a number of similarities, but also a few differences. These lines can now be used to attempt to understand how Arx alters normal cortical circuitry and how these changed neuronal networks result in an epilepsy phenotype and varying degrees of cognitive dysfunction.

ARX as Disease Mediator

Mutations in Arx result in a variety of phenotypes. There are two aspects of Arx function described in the sections above that have both research and clinical implications to the study of ARX-related disorders. The first relates to ARX’s role in interneuron development. A number of genes can lead to brain malformations, which can be focal or diffuse. In many cases these malformations present later in life when the child is evaluated for learning issues or a first seizure. Even in the most severe malformations, such as lissencephaly, there is variability in the severity of the cognitive disability and the intractable nature of the epilepsy that occurs. In ARX mediated malformations, and in particular, XLAG, the patients’ disease is particularly severe. The children often make no developmental progress and have seizures that are not responsive to any treatment. The severity of this disorder, compared to those resulting from other genes that cause lissencephaly or other malformations has been attributed to the specific loss of interneurons.56 Kato and Dobyns have used ARX related disorders to designate a new terminology for a class of neurological diseases; called interneuronopathies.56 Other interneuronopathies could be uncovered by screening patients with similar clinical phenotypes for genes either upstream or downstream from Arx in the gene regulatory network.

The second aspect of ARX activity to have clinical implications is the finding that Arx has seemingly very different roles in two regions of the developing forebrain. The fact that Arx may control proliferation in the dorsal VZ, but control migration or fate specification in the ventral forebrain, means that altering Arx activity throughout the entire brain could have multiple unintended consequences. Therefore, developing a better understanding of how Arx functions, and where in the gene regulatory networks of telencephalon development Arx acts, is vital to be able to generate therapeutic interventions for patients with Arx mutations. In addition, a greater understanding of the temporal and spatial regulation of Arx expression and function is necessary if future therapeutics are to be designed.

FUTURE DIRECTIONS FOR THE STUDY OF ARX

Over the last decade Arx has been linked to over 10 overlapping clinical disorders and a great deal of information regarding the expression pattern, molecular function, and role in cortical development has been generated. This work has answered many questions but inevitably has raised many more. Further study is necessary to understand how different mutations lead to the spectrum of disorders and whether genetic background effect, environment, or changes in the protein due to the mutations are the cause of such variability. Next, a full categorization of the Arx involved gene networks is needed and an understanding of how the gene would have differential effects in the ventral and dorsal telecephalon. Another area for future investigation is the role of Arx in the postnatal brain. Lastly, consideration of how alteration in interneurons leads to a seizure or cognitive phenotype is warranted, and the 4 mouse lines are available to study the pathophysiological changes that occur. The next decade will likely yield the answers to many of these questions.

Disclosure statement: these authors have nothing to disclose.

REFERENCES

1.
Cobos I, Broccoli V, Rubenstein JL. The vertebrate ortholog of Aristaless is regulated by Dlx genes in the developing forebrain. J Comp Neurol. 2005;483(3):292–303. [PubMed: 15682394]
2.
Fulp CT, Cho G, Marsh ED, Nasrallah IM, Labowski PA, Golden JA. Identification of Arx transcriptional targets in the developing basal forebrain. Hum Mol Genet. 2008;17(23):3740–60. [PMC free article: PMC2581427] [PubMed: 18799476]
3.
Price MG, Yoo JW, Burgess DL, Deng F, Hrachovy RA, Frost JD Jr, Noebels JL. A triplet repeat expansion genetic mouse model of infantile spasms syndrome, Arx(GCG)10+7, with Interneuronopathy, spasms in infancy, persistent seizures, and adult cognitive and behavioral impairment. J Neurosci. 2009;29(27):8752–63. [PMC free article: PMC2782569] [PubMed: 19587282]
4.
Marsh E, Fulp C, Gomez E, Nasrallah I, Minarcik J, Sudi J, Christian SL, Mancini G, Labosky P, Dobyns W, Brooks-Kayal A, Golden JA. Targeted loss of Arx results in a developmental epilepsy mouse model and recapitulates the human phenotype in heterozygous females. Brain. 2009;132:1563–1576. [PMC free article: PMC2685924] [PubMed: 19439424]
5.
Kitamura K, Itou Y, Yanazawa M, Ohsawa M, Suzuki-Migishima R, Umeki Y, Hohjoh H, Yanagawa Y, Shinba T, Itoh M, Nakamura K, Goto Y. Three human ARX mutations cause the lissencephaly-like and mental retardation with epilepsy-like pleiotropic phenotypes in mice. Hum Mol Genet. 2009;18(19):3708–24. [PubMed: 19605412]
6.
Korff CM, Nordli DR Jr. Epilepsy syndromes in infancy. Pediatr Neurol. 2006;34(4):253–63. [PubMed: 16638498]
7.
Bienvenu T, Poirier K, Friocourt G, Bahi N, Beaumont D, Fauchereau F, Ben Jeema L, Zemni R, Vinet MC, Francis F, Couvert P, Gomot M, Moraine C, van Bokhoven H, Kalscheuer V, Frints S, Gecz J, Ohzaki K, Chaabouni H, Fryns JP, Desportes V, Beldjord C, Chelly J. ARX, a novel Prd-class-homeobox gene highly expressed in the telencephalon, is mutated in X-linked mental retardation. Hum Mol Genet. 2002;11(8):981–91. [PubMed: 11971879]
8.
Kitamura K, Yanazawa M, Sugiyama N, Miura H, Iizuka-Kogo A, Kusaka M, Omichi K, Suzuki R, Kato-Fukui Y, Kamiirisa K, Matsuo M, Kamijo S, Kasahara M, Yoshioka H, Ogata T, Fukuda T, Kondo I, Kato M, Dobyns WB, Yokoyama M, Morohashi K. Mutation of ARX causes abnormal development of forebrain and testes in mice and X-linked lissencephaly with abnormal genitalia in humans. Nat Genet. 2002;32(3):359–69. [PubMed: 12379852]
9.
Stromme P, Mangelsdorf ME, Shaw MA, Lower KM, Lewis SM, Bruyere H, Lutcherath V, Gedeon AK, Wallace RH, Scheffer IE, Turner G, Partington M, Frints SG, Fryns JP, Sutherland GR, Mulley JC, Gecz J. Mutations in the human ortholog of Aristaless cause X-linked mental retardation and epilepsy. Nat Genet. 2002;30(4):441–5. [PubMed: 11889467]
10.
Bonneau D, Toutain A, Laquerriere A, Marret S, Saugier-Veber P, Barthez MA, Radi S, Biran-Mucignat V, Rodriguez D, Gelot A. X-linked lissencephaly with absent corpus callosum and ambiguous genitalia (XLAG): clinical, magnetic resonance imaging, and neuropathological findings. Ann Neurol. 2002;51(3):340–9. [PubMed: 11891829]
11.
Proud VK, Levine C, Carpenter NJ. New X-linked syndrome with seizures, acquired micrencephaly, and agenesis of the corpus callosum. Am J Med Genet. 1992;43(1–2):458–66. [PubMed: 1605226]
12.
Kato M, Das S, Petras K, Kitamura K, Morohashi K, Abuelo DN, Barr M, Bonneau D, Brady AF, Carpenter NJ, Cipero KL, Frisone F, Fukuda T, Guerrini R, Iida E, Itoh M, Lewanda AF, Nanba Y, Oka A, Proud VK, Saugier-Veber P, Schelley SL, Selicorni A, Shaner R, Silengo M, Stewart F, Sugiyama N, Toyama J, Toutain A, Vargas AL, Yanazawa M, Zackai EH, Dobyns WB. Mutations of ARX are associated with striking pleiotropy and consistent genotype-phenotype correlation. Hum Mutat. 2004;23(2):147–59. [PubMed: 14722918]
13.
Guerrini R, Moro F, Kato M, Barkovich AJ, Shiihara T, McShane MA, Hurst J, Loi M, Tohyama J, Norci V, Hayasaka K, Kang UJ, Das S, Dobyns WB. Expansion of the first PolyA tract of ARX causes infantile spasms and status dystonicus. Neurology. 2007;69(5):427–33. [PubMed: 17664401]
14.
Demos MK, Fullston T, Partington MW, Gecz J, Gibson WT. Clinical study of two brothers with a novel 33 bp duplication in the ARX gene. Am J Med Genet A. 2009;149A(7):1482–6. [PubMed: 19507262]
15.
Partington MW, Mulley JC, Sutherland GR, Hockey A, Thode A, Turner G. X-linked mental retardation with dystonic movements of the hands. Am J Med Genet. 1988;30(1–2):251–62. [PubMed: 3177452]
16.
Turner G, Partington M, Kerr B, Mangelsdorf M, Gecz J. Variable expression of mental retardation, autism, seizures, and dystonic hand movements in two families with an identical ARX gene mutation. Am J Med Genet. 2002;112(4):405–11. [PubMed: 12376946]
17.
Scheffer IE, Wallace RH, Phillips FL, Hewson P, Reardon K, Parasivam G, Stromme P, Berkovic SF, Gecz J, Mulley JC. X-linked myoclonic epilepsy with spasticity and intellectual disability: mutation in the homeobox gene ARX. Neurology. 2002;59(3):348–56. [PubMed: 12177367]
18.
Stromme P, Mangelsdorf ME, Scheffer IE, Gecz J. Infantile spasms, dystonia, and other X-linked phenotypes caused by mutations in Aristaless related homeobox gene, ARX. Brain Dev. 2002;24(5):266–8. [PubMed: 12142061]
19.
Kato M, Saitoh S, Kamei A, Shiraishi H, Ueda Y, Akasaka M, Tohyama J, Akasaka N, Hayasaka K. A longer polyalanine expansion mutation in the ARX gene causes early infantile epileptic encephalopathy with suppression-burst pattern (Ohtahara syndrome). Am J Hum Genet. 2007;81(2):361–6. [PMC free article: PMC1950814] [PubMed: 17668384]
20.
Kato M, Koyama N, Ohta M, Miura K, Hayasaka K. Frameshift mutations of the ARX gene in familial Ohtahara syndrome. Epilepsia. 2010;51(9):1679–1684. [PubMed: 20384723]
21.
Absoud M, Parr JR, Halliday D, Pretorius P, Zaiwalla Z, Jayawant S. A novel ARX phenotype: rapid neurodegeneration with Ohtahara syndrome and a dyskinetic movement disorder. Dev Med Child Neurol. 2010;52(3):305–7. [PubMed: 19747203]
22.
Poirier K, Eisermann M, Caubel I, Kaminska A, Peudonnier S, Boddaert N, Saillour Y, Dulac O, Souville I, Beldjord C, Lascelles K, Plouin P, Chelly J, Bahi-Buisson N. Combination of infantile spasms, non-epileptic seizures and complex movement disorder: a new case of ARX-related epilepsy. Epilepsy Res. 2008;80(2–3):224–8. [PubMed: 18468866]
23.
Shinozaki Y, Osawa M, Sakuma H, Komaki H, Nakagawa E, Sugai K, Sasaki M, Goto Y. Expansion of the first polyalanine tract of the ARX gene in a boy presenting with generalized dystonia in the absence of infantile spasms. Brain Dev. 2009;31(6):469–72. [PubMed: 18823727]
24.
Okazaki S, Ohsawa M, Kuki I, Kawawaki H, Koriyama T, Ri S, Ichiba H, Hai E, Inoue T, Nakamura H, Goto Y, Tomiwa K, Yamano T, Kitamura K, Itoh M. Aristaless-related homeobox gene disruption leads to abnormal distribution of GABAergic interneurons in human neocortex: evidence based on a case of X-linked lissencephaly with abnormal genitalia (XLAG). Acta Neuropathol. 2008;116(4):453–62. [PubMed: 18458920]
25.
Wallerstein R, Sugalski R, Cohn L, Jawetz R, Friez M. Expansion of the ARX spectrum. Clin Neurol Neurosurg. 2008;110(6):631–4. [PubMed: 18462864]
26.
Friocourt G, Parnavelas JG. Mutations in ARX Result in Several Defects Involving GABAergic Neurons. Front Cell Neurosci. 2010:1–11. [PMC free article: PMC2841486] [PubMed: 20300201]
27.
Friocourt G, Poirier K, Rakic S, Parnavelas JG, Chelly J. The role of ARX in cortical development. Eur J Neurosci. 2006;23(4):869–76. [PubMed: 16519652]
28.
Galliot B, de Vargas C, Miller D. Evolution of homeobox genes: Q50 Paired-like genes founded the Paired class. Dev Genes Evol. 1999;209(3):186–97. [PubMed: 10079362]
29.
Miura H, Yanazawa M, Kato K, Kitamura K. Expression of a novel aristaless related homeobox gene ‘Arx’ in the vertebrate telencephalon, diencephalon and floor plate. Mech Dev. 1997;65(1–2):99–109. [PubMed: 9256348]
30.
Colombo E, Galli R, Cossu G, Gecz J, Broccoli V. Mouse orthologue of ARX, a gene mutated in several X-linked forms of mental retardation and epilepsy, is a marker of adult neural stem cells and forebrain GABAergic neurons. Dev Dyn. 2004;231(3):631–9. [PubMed: 15376319]
31.
Poirier K, Van Esch H, Friocourt G, Saillour Y, Bahi N, Backer S, Souil E, Castelnau-Ptakhine L, Beldjord C, Francis F, Bienvenu T, Chelly J. Neuroanatomical distribution of ARX in brain and its localisation in GABAergic neurons. Brain Res Mol Brain Res. 2004;122(1):35–46. [PubMed: 14992814]
32.
Colombo E, Collombat P, Colasante G, Bianchi M, Long J, Mansouri A, Rubenstein JL, Broccoli V. Inactivation of Arx, the murine ortholog of the X-linked lissencephaly with ambiguous genitalia gene, leads to severe disorganization of the ventral telencephalon with impaired neuronal migration and differentiation. J Neurosci. 2007;27(17):4786–98. [PMC free article: PMC4916654] [PubMed: 17460091]
33.
Kitamura K, Miura H, Yanazawa M, Miyashita T, Kato K. Expression patterns of Brx1 (Rieg gene), Sonic hedgehog, Nkx2.2, Dlx1 and Arx during zona limitans intrathalamica and embryonic ventral lateral geniculate nuclear formation. Mech Dev. 1997;67(1):83–96. [PubMed: 9347917]
34.
Colasante G, Collombat P, Raimondi V, Bonanomi D, Ferrai C, Maira M, Yoshikawa K, Mansouri A, Valtorta F, Rubenstein JL, Broccoli V. Arx is a direct target of Dlx2 and thereby contributes to the tangential migration of GABAergic interneurons. J Neurosci. 2008;28(42):10674–86. [PMC free article: PMC3844830] [PubMed: 18923043]
35.
Collombat P, Hecksher-Sorensen J, Broccoli V, Krull J, Ponte I, Mundiger T, Smith J, Gruss P, Serup P, Mansouri A. The simultaneous loss of Arx and Pax4 genes promotes a somatostatin-producing cell fate specification at the expense of the alpha- and beta-cell lineages in the mouse endocrine pancreas. Development. 2005;132(13):2969–80. [PubMed: 15930104]
36.
Collombat P, Mansouri A, Hecksher-Sorensen J, Serup P, Krull J, Gradwohl G, Gruss P. Opposing actions of Arx and Pax4 in endocrine pancreas development. Genes Dev. 2003;17(20):2591–603. [PMC free article: PMC218152] [PubMed: 14561778]
37.
Hancock AS, Du A, Liu J, Miller M, May CL. Glucagon deficiency reduces hepatic glucose production and improves glucose tolerance in adult mice. Mol Endocrinol. 2010;24(8):1605–14. [PMC free article: PMC2940466] [PubMed: 20592160]
38.
McKenzie O, Ponte I, Mangelsdorf M, Finnis M, Colasante G, Shoubridge C, Stifani S, Gecz J, Broccoli V. Aristaless-related homeobox gene, the gene responsible for West syndrome and related disorders, is a Groucho/transducin-like enhancer of split dependent transcriptional repressor. Neuroscience. 2007;146(1):236–47. [PubMed: 17331656]
39.
Meijlink F, Beverdam A, Brouwer A, Oosterveen TC, Berge DT. Vertebrate aristaless-related genes. Int J Dev Biol. 1999;43(7):651–63. [PubMed: 10668975]
40.
Hudson R, Taniguchi-Sidle A, Boras K, Wiggan O, Hamel PA. Alx-4, a transcriptional activator whose expression is restricted to sites of epithelial-mesenchymal interactions. Dev Dyn. 1998;213(2):159–69. [PubMed: 9786416]
41.
Smith ST, Jaynes JB. A conserved region of engrailed, shared among all en-, gsc-, Nk1-, Nk2- and msh-class homeoproteins, mediates active transcriptional repression in vivo. Development. 1996;122(10):3141–50. [PMC free article: PMC2729110] [PubMed: 8898227]
42.
Fullenkamp AN, El-Hodiri HM. The function of the Aristaless-related homeobox (Arx) gene product as a transcriptional repressor is diminished by mutations associated with X-linked mental retardation (XLMR). Biochem Biophys Res Commun. 2008;377(1):73–8. [PubMed: 18835247]
43.
Colasante G, Sessa A, Crispi S, Calogero R, Mansouri A, Collombat P, Broccoli V. Arx acts as a regional key selector gene in the ventral telencephalon mainly through its transcriptional repression activity. Dev Biol. 2009;334(1):59–71. [PubMed: 19627984]
44.
Batista-Brito R, Fishell G. The developmental integration of cortical interneurons into a functional network. Curr Top Dev Biol. 2009;87:81–118. [PMC free article: PMC4465088] [PubMed: 19427517]
45.
Marin O, Rubenstein JL. A long, remarkable journey: tangential migration in the telencephalon. Nat Rev Neurosci. 2001;2(11):780–90. [PubMed: 11715055]
46.
Rallu M, Corbin JG, Fishell G. Parsing the prosencephalon. Nat Rev Neurosci. 2002;3(12):943–51. [PubMed: 12461551]
47.
Anderson SA, Eisenstat DD, Shi L, Rubenstein JL. Interneuron migration from basal forebrain to neocortex: dependence on Dlx genes. Science. 1997;278(5337):474–6. [PubMed: 9334308]
48.
Anderson SA, Qiu M, Bulfone A, Eisenstat DD, Meneses J, Pedersen R, Rubenstein JL. Mutations of the homeobox genes Dlx-1 and Dlx-2 disrupt the striatal subventricular zone and differentiation of late born striatal neurons. Neuron. 1997;19(1):27–37. [PubMed: 9247261]
49.
Friocourt G, Kanatani S, Tabata H, Yozu M, Takahashi T, Antypa M, Raguenes O, Chelly J, Ferec C, Nakajima K, Parnavelas JG. Cell-autonomous roles of ARX in cell proliferation and neuronal migration during corticogenesis. J Neurosci. 2008;28(22):5794–805. [PMC free article: PMC6670801] [PubMed: 18509041]
50.
Albrecht A, Mundlos S. The other trinucleotide repeat: polyalanine expansion disorders. Curr Opin Genet Dev. 2005;15(3):285–93. [PubMed: 15917204]
51.
Messaed C, Rouleau GA. Molecular mechanisms underlying polyalanine diseases. Neurobiol Dis. 2009;34(3):397–405. [PubMed: 19269323]
52.
Nasrallah IM, Minarcik JC, Golden JA. A polyalanine tract expansion in Arx forms intranuclear inclusions and results in increased cell death. J Cell Biol. 2004;167(3):411–6. [PMC free article: PMC2172475] [PubMed: 15533998]
53.
Shoubridge C, Cloosterman D, Parkinson-Lawerence E, Brooks D, Gecz J. Molecular pathology of expanded polyalanine tract mutations in the Aristaless-related homeobox gene. Genomics. 2007;90(1):59–71. [PubMed: 17490853]
54.
Lin W, Ye W, Cai L, Meng X, Ke G, Huang C, Peng Z, Yu Y, Golden JA, Tartakoff AM, Tao T. The roles of multiple importins for nuclear import of murine aristaless-related homeobox protein. J Biol Chem. 2009;284(30):20428–39. [PMC free article: PMC2740467] [PubMed: 19494118]
55.
Shoubridge C, Tan MH, Fullston T, Cloosterman D, Coman D, McGillivray G, Mancini GM, Kleefstra T, Gecz J. Mutations in the nuclear localization sequence of the Aristaless related homeobox; sequestration of mutant ARX with IPO13 disrupts normal subcellular distribution of the transcription factor and retards cell division. Pathogenetics. 2010;3:1–11. [PMC free article: PMC2819251] [PubMed: 20148114]
56.
Kato M, Dobyns WB. X-linked lissencephaly with abnormal genitalia as a tangential migration disorder causing intractable epilepsy: proposal for a new term, “interneuronopathy” J Child Neurol. 2005;20(4):392–7. [PubMed: 15921244]
Copyright © 2012, Michael A Rogawski, Antonio V Delgado-Escueta, Jeffrey L Noebels, Massimo Avoli and Richard W Olsen.

All Jasper's Basic Mechanisms of the Epilepsies content, except where otherwise noted, is licensed under a Creative Commons Attribution-NonCommercial-NoDerivs 3.0 Unported license, which permits copying, distribution and transmission of the work, provided the original work is properly cited, not used for commercial purposes, nor is altered or transformed.

Bookshelf ID: NBK98176PMID: 22787632

Views

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...