Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Dec 23.
Published in final edited form as: FEBS J. 2021 Sep 29;289(24):7776–7787. doi: 10.1111/febs.16204

Molecular and cellular events linking variants in the histone demethylase KDM5C to the intellectual disability disorder Claes-Jensen syndrome

Hayden A M Hatch 1, Julie Secombe 1,2
PMCID: PMC8930784  NIHMSID: NIHMS1772302  PMID: 34536985

Abstract

The widespread availability of genetic testing for those with neurodevelopmental disorders has highlighted the importance of many genes necessary for the proper development and function of the nervous system. One gene found to be genetically altered in the X-linked intellectual disability disorder Claes-Jensen syndrome is KDM5C, which encodes a histone demethylase that regulates transcription by altering chromatin. While the genetic link between KDM5C and cognitive (dys)function is clear, how KDM5C functions to control transcriptional programs within neurons to impact their growth and activity remains the subject of ongoing research. Here, we review our current knowledge of Claes-Jensen syndrome and discuss important new data using model organisms that have revealed the importance of KDM5C in regulating aspects of neuronal development and function. Continued research into the molecular and cellular activities regulated by KDM5C is expected to provide critical etiological insights into Claes-Jensen syndrome and highlight potential targets for developing therapies to improve the quality of life of those affected.

Keywords: animal models of disease, autism spectrum disorders, CJ-XLID, Claes-Jensen syndrome, histone demethylase, intellectual disability, KDM5C, KDM5C-RD, MRXSCJ, neurodevelopmental disorders

Genetic variants in the KDM5C gene lead to the intellectual disability disorder Claes-Jensen syndrome

Neurodevelopmental disorders (NDDs) are a group of related conditions that alter the functioning of the nervous system of affected individuals and include intellectual disability (ID), autism spectrum disorders (ASD), communication disorders, and developmental delay (DD). Environmental factors such as maternal stress during pregnancy or preterm birth can increase the risk of NDDs [1-3]. In addition, many genetic variants have been etiologically linked to NDDs using genome-wide approaches such as comparative genomic hybridization and whole-exome sequencing [4]. These NDD-associated changes to DNA can range from single base pair changes to large deletions and can either display a familial inheritance profile or occur de novo in affected individuals. While genes with roles in a range of cellular processes have been associated with NDDs, many variants affect transcriptional regulators, clearly demonstrating the importance of regulated gene expression to the proper development and functioning of the brain [5,6]. This review focuses on one transcriptional regulator, KDM5C, which is found to be genetically altered in individuals with the NDD intellectual disability, X-linked, syndromic, Claes-Jensen type (OMIM#300534) (Fig. 1A; Table 1). We will refer to this disorder as Claes-Jensen syndrome, although it should be noted that it has also been referred to as CJ-XLID, MRXSCJ, and KDM5C-RD [7-10].

Fig. 1.

Fig. 1.

The KDM5C gene that is genetically altered in individuals with Claes-Jensen syndrome encodes a conserved protein. (A) Genetic variants observed in individuals with Claes-Jensen syndrome. Types of genetic change are indicated by colored circles, with missense in black, frameshift in green, splice site in yellow, and nonsense variants in gray. Details of each variant can be found in Table 1. (B) Phylogenetic relationship between the four paralogous KDM5 family proteins in humans and the single orthologs in flies and worms. Domains are shown by colored boxes. Animal images generated using Biorender.com.

Table 1.

KDM5C variants observed in individuals with Claes-Jensen syndrome. Type of variants and corresponding change to the encoded protein observed in males and/or females with Claes-Jensen syndrome. For missense variants, predicted domain(s) affected by the change in amino acid are indicated. Effects to in vitro demethylase activity are also indicated, if applicable. The "–" symbol indicates unknown or not determined.

Missense variants NDD KDM5C domain Enzymatic activity Frameshift variants NDD
M1T [28] ID - - A50Rfs*23 [26] ID
W52C [26] ID JmjN - R68fs*7 [10] ID
A77T [22] ID ARID - G170Efs*64 [88] ID/DD
Y85F [89] ID/DD ARID - L197fs*23 [26] ID/DD
D87G [26,90] (2 families) ID ARID No defect [91] R211fs*23 [88] ID/DD
Y164N [92] ID ARID - R211fs*22 [88] ID/DD
A388P [10] ID PHD/JmjC Reduced [14] L257Afs*4 [93] ID
D402Y [10] ID PHD/JmjC Reduced [91,94] T270fs*2 [95] ID
S451R [96] ID PHD/JmjC - W534Gfs*15 [92] ID
P480L [97] ID PHD/JmjC Reduced [94] A683fs*81 [25] ID
Y503C [89] ID/DD JmjC - R795fs*5 [26] ID
V504 M [22] ID JmjC - E810Cfs*5 [98] NDD
S522F [23] ID JmjC - V1075fs*2 [94] ID
K532N [99] ID JmjC - K1087fs*43 [24] ID
P554T [24] ID JmjC Reduced [24] A1292Qfs*7 [27] ID
R599C [26,89] ID/DD JmjC - L1336Pfs*11 [100] ID
E613K [26] ID JmjC - R1481Gfs*9 [22] ID
W622C [26] ID JmjC - Nonsense variants NDD
C640Y [101] ID JmjC - Q237* [102] ID
F642L [90] ID JmjC/C5HC2 Reduced [14] R322* [90] ID
E698K [10] ID C5HC2 - E424* [103] ID
T713M [104] ID C5HC2 - E433* [105] ID
A718P [105] ID C5HC2 - E467* [89] ID/DD
L731F [10,106] ID C5HC2 Reduced [14] R694* [10] ID
R750W [90] ID C5HC2 - C724* [36] ID
Y751C [90] ID C5HC2 Reduced [14] R828* [107] ID
R766W [108] ID/ASD C5HC2/PLU-1 - Q970* [92] ID
E1024D [109] ID PLU-1 - C1095* [90] ID
R1115H [35] ID/ASD - No defect [35] W1288* [10] ID
A1277T [89] ID/DD - - E1299* [110] ID
D1300V [80,111] ASD - - E1468* [99] ID
Splice site variants NDD
c.160G > T [112] ID
c.1243-2A > G [26] ID
c.658-1G > T [113] ID/DD
c.1583 + 5G > A [22] ID
c.2243 + 2T > C [92] ID
c.2622 + 2dupT [26] ID

KDM5C is one of four paralogous genes, KDM5A-D, that encode structurally similar proteins that function to regulate transcription (Fig. 1B). KDM5 genes are expressed in a broad range of tissues, although it is notable that KDM5C is expressed at high levels within the brain, consistent with it playing a critical role in cognitive function [11]. The most characterized means by which KDM5C regulates gene expression is via its enzymatic demethylase activity. This function is mediated through its Jumonji N (JmjN) and Jumonji C (JmjC) domains, which enzymatically removes, di- and trimethyl marks from lysine 4 of histone H3 (H3K4me2/3) (Fig. 1) [12-15]. The target of KDM5C protein demethylation, H3K4me2/3, is found primarily surrounding promoter regions of genes and correlates with transcriptional activation [16]. Consistent with its ability to regulate the activity of promoters, KDM5C binds to these regulatory elements to alter transcription [7,8,17,18].

Clinically, males with pathogenic genetic variants in KDM5C are almost universally diagnosed with ID (Table 1). According to the most recent DSM-5 release, a diagnosis of ID is defined by an intelligence quotient (IQ) of less than 70 along with deficits in two or more adaptive behaviors that significantly affect daily functioning by the age of 18 [19]. Adaptive behaviors include conceptual skills related to language and problem solving, in addition to social proficiencies in interpersonal communication, social judgment, and empathy [19-21]. Also considered in a diagnosis of ID is the ability of affected individuals to independently carry out tasks required for self-care, to maintain employment, and be fiscally responsible. The degree of ID observed in males with Claes-Jensen syndrome varies from mild to severe, with children often also displaying and an increased incidence of epilepsy, aggression, and motor delays. Individuals may also present with physical characteristics such as short stature and craniofacial features [22-25]. Typically, those with mild ID have an IQ of 50–70 and have difficulty with speech, reading and writing, straightforward arithmetic, and/or adapting to societal norms. Those with moderate and severe ID have IQs of 35–50 and 20–40, respectively, and show greater deficits to adaptive behaviors. They may additionally require daily assistance with tasks involving self-care and social interaction.

Unlike males hemizygous for pathogenic KDM5C variants, the clinical presentation of heterozygous females varies widely and is only now beginning to be characterized in detail. While up to 50% of females have no overt deficits, others show intellectual disability, developmental delay, learning and speech difficulties, hormonal imbalance, and anxiety [9,23,26-29]. The basis for the incomplete penetrance of symptoms is not clear, though for other genetic causes of X-linked cognitive disorders such as Fragile X syndrome, skewing of X-chromosome inactivation can impact the severity of symptoms in females [30,31]. Despite initially being thought to escape X-inactivation [32], the extent to which KDM5C is expressed from the inactive X-chromosome appears to vary widely [33,34]. It is therefore likely that variability in KDM5C inactivation contributes to disease severity in females with Claes-Jensen syndrome [26,28,35].

Pathogenic KDM5C variants alter neuronal structure and function

Very little published literature exists detailing the anatomical and functional changes to the brain that occur in those with Claes-Jensen syndrome. A subset of individuals has been documented to have microcephaly, and, in one case, an MRI revealed a disproportionately small cerebellum [27,36]; however, overall changes in brain size and structure do not appear to be common features of this disorder. To better understand the links between KDM5C function and brain development, several powerful genetic model systems have been employed. These include the mouse Mus musculus, the vinegar fly Drosophila melanogaster, and the nematode worm Caenorhabditis elegans. Studies using these animal models suggest that KDM5C plays vital roles in several different aspects of neuronal development and function, all of which could contribute to the clinical manifestations seen in those with Claes-Jensen syndrome.

The first in vivo model developed to study the molecular and cellular mechanisms underlying Claes-Jensen syndrome utilized mice. Like humans, mice encode four paralogous Kdm5 genes and genetic knockout of the X-linked Kdm5c (Kdm5cKO) results in features that resemble those observed in patients. For example, hemizygous male Kdm5cKO mice are smaller than their wild-type littermates and show deficits in learning and memory and motor control while displaying increased aggression and seizure susceptibility [7,8]. Heterozygous female Kdm5cKO mice have milder phenotypes than hemizygous males, being only slightly smaller than expected and exhibiting mild learning deficits [8]. While the brains from Kdm5cKO adult mice did not show any overall cytoarchitectural defects, cellular studies revealed that pyramidal neurons from the basolateral amygdala and the ventral hippocampus showed dendritic spine defects [7,18]. Dendritic spines receive synaptic signals from the axons of adjacent neurons and can change based on synaptic strength [37]. Notably, individuals with a range of different NDDs have been shown to have alterations in dendritic spine number and morphology [38-40]. Whether the changes to dendritic structure seen in Kdm5c knockout mice are the result of altered synaptic transmission or whether such morphological defects occur in other neuronal subtypes remains important and open questions.

The subject of whether KDM5C regulates gene expression programs necessary for synaptic activity has been investigated using another animal model, Drosophila. In contrast to mice and humans, Drosophila has a smaller genome that encodes a single KDM5 protein-containing conserved domains from all four mammalian paralogs (Fig. 1). Because ~70% of human disease-causing genes are conserved in Drosophila, it is widely used to provide fundamental insights into many disorders, including NDDs [41-43]. Drosophila has recently been developed as a model for Claes-Jensen syndrome, with KDM5 being shown to be necessary for associative learning and memory in adult flies [44,45]. The Drosophila larval neuromuscular junction (NMJ) is a glutamatergic synapse that is functionally similar to an excitatory synaptic connection in the human brain [46]. Analyses of genetic null animals have shown that KDM5 is essential in motor neurons to regulate the size and number of synaptic boutons at the NMJ, as well as for proper synaptic transmission [44]. Because altered glutamatergic signaling has been implicated in a range of NDDs [5,47], KDM5C-mediated regulation of synaptic signaling could contribute to the cognitive changes seen in Claes-Jensen syndrome.

Studies from Drosophila and C. elegans suggest that KDM5C is also likely to alter neuronal connectivity through its regulation of axonal growth and guidance. Whereas the Drosophila larval NMJ is an excellent system to examine synaptic morphology and function, the mushroom body, a key learning and memory structure within the adult brain, is a well-established model for studying axonal growth and guidance [46]. Animals lacking the kdm5 gene show significant structural defects that are caused by failure of the neurons that comprise the mushroom body (Kenyon cells) to properly project their axons [48]. A similar phenotype has been described in worms with mutations in the single kdm5 gene (rbr-2), where the axons of inter- and motoneurons show altered trajectory [49]. The repertoire of neurons affected by these growth and guidance defects in the fly and worm systems remains to be determined, as does the extent to which KDM5C regulates this process within mammalian brains. Combined, these data do provide compelling evidence that KDM5C controls more than one aspect of neuronal development across multiple cell types and developmental stages (Fig. 2).

Fig. 2.

Fig. 2.

Neuronal functions of KDM5C that could contribute to Claes-Jensen syndrome. KDM5C is a transcriptional regulator required for several distinct aspects of neuronal development and function based on studies in animal models (mice, flies, and worms). See text for details. Model created using Biorender.com.

KDM5C variants alter transcriptional programs in neurons

Because KDM5 family proteins regulate gene expression, changes to critical transcriptional programs are likely to lie at the heart of the clinical features seen in individuals with Claes-Jensen syndrome. Across species, KDM5 family proteins act primarily as modulators of gene expression, with their loss leading to modest (mostly < 2-fold) changes to the expression of downstream target genes [17,44,45,48-56]. This observation suggests that the phenotypes caused by alleles of KDM5C are due to the combined impact of many, relatively small, changes to gene expression. Pathogenic variants in KDM5C are thus likely to contribute to the neurodevelopmental features seen in Claes-Jensen syndrome by affecting multiple key transcriptional programs. This can make it challenging to define the in vivo transcriptional targets of KDM5C, particularly when comparing human samples that can be genetically heterogeneous. This challenge is highlighted by a study that used cells from patients with Claes-Jensen syndrome [11]. Because the human brain is not amenable to direct assays to define KDM5C functions, transformed lymphoblastoid cells from patients hemizygous for KDM5C alleles were used for genome-wide and targeted transcriptional analyses. While this led to the identification of a handful of genes that were dysregulated across all patient-derived cell lines, it did not lead to testable models of how variants in KDM5C could affect cognition and behavior. Although easy to access and culture, the use of lymphoid cells may complicate the interpretation of these data, since they may only partially recapitulate all the gene regulatory activities of KDM5C in the brain. In addition, differences in genetic background between individuals with Claes-Jensen syndrome and controls could make it difficult to detect small changes in gene expression. It is also notable that many key transcriptional changes are likely to occur during development, and thus may be missed by studies using mature cell types collected from patients.

Model organisms provide genetically controlled systems that are amenable to studies aimed at understanding how KDM5C regulates gene expression in neuronal cell types. Indeed, transcriptomic analyses from mice and flies have revealed interesting insights into possible mechanisms contributing to Claes-Jensen syndrome. In-keeping with the range of neuronal functions found to be phenotypically altered by loss of Kdm5c in mice or its orthologs in Drosophila and C. elegans, KDM5C can regulate different distinct transcriptional programs. In some contexts, the gene expression changes seen upon loss of KDM5C appear to fit expectations based on the observed nervous system deficits. For example, consistent with its role in synaptic structure and function in mice and flies, KDM5C regulates the expression of genes involved in synaptic plasticity and neurotransmitter release [7,44,57]. Similarly, known regulators of axonal growth, such as the actin cytoskeleton binding protein Wasp-1 and the transcriptional regulator Prospero, have been found to be key mediators of the neuronal guidance defects observed in worms and flies [48,49].

Other changes to gene expression programs regulated by KDM5C have been more surprising. For example, gene expression changes in the hippocampus of Kdm5cKO mice revealed the derepression of a significant number of genes whose expression is normally limited to the germline [8]. This gene expression signature has the potential to be significant for the neuropathology of Claes-Jensen syndrome, as germline-enriched genes are found to be derepressed in mouse models of other NDDs such as Kleefstra syndrome and Rett syndrome [58-60]. An additional cellular process that was uncovered through analyses of a Drosophila strain harboring a patient-associated variant in the fly ortholog of KDM5C was the regulation of ribosomal protein genes [55]. Proper control of translation is critical to neuronal function, with deficits in this process being observed in individuals with other inherited forms of cognitive impairment, including Fragile X syndrome, ASD, and Alzheimer’s disease [61-70]. This suggests that altered translation may be a common pathogenic mechanism of a subset of cognitive disorders that includes Claes-Jensen syndrome. Consistent with the possibility that the regulation of translation may be conserved in vertebrate animals, ChIP data from cultured embryonic mouse cortical neurons show that KDM5C binds to the promoter region of most ribosomal protein genes [7]. Both the inappropriate expression of germline genes and altered expression of ribosomal protein genes have the potential to interfere with neuronal structure and function, thereby contributing to the cognitive changes seen in those with Claes-Jensen syndrome.

Leveraging model organisms to uncover disrupted KDM5C regulatory mechanisms in Claes-Jensen syndrome

It is generally assumed that the histone demethylase activity of KDM5C is the primary means by which it regulates gene expression and that loss of this activity leads to cognitive impairment. This hypothesis is appealing since it points toward a potential means for targeted therapies for individuals with Claes-Jensen syndrome. The most compelling data in support of this model come from a study showing that the learning and memory phenotypes of hemizygous Kdm5cKO mice are attenuated by genetically reducing the levels of one of the enzymes that deposits the H3K4me3 mark, KMT2A [18]. Corroborating evidence comes from studies using a Drosophila model of Claes-Jensen syndrome. Analyses of a fly strain specifically lacking KDM5 histone demethylase activity have revealed that this enzymatic function is essential both for proper synaptic function at the larval NMJ and for learning and memory in adults [44,45,55]. Similarly, the axonal guidance defects observed in C. elegans are caused by the loss of the catalytic activity of KDM5 [49]. Importantly, these data are consistent with the general observation that tight regulation of H3K4me3 levels appears to be critical in the brain, as mutations in other regulators of this chromatin mark are also observed in individuals with NDDs [71].

There is, however, accumulating evidence that KDM5C may affect transcriptional programs critical for normal neuronal development and function via nonenzymatic means. While some missense mutations in human KDM5C attenuate its in vitro enzymatic activity to some extent, this is not universally true, as two patient-associated mutations do not result in reduced demethylase function (Table 1) [14,15,35]. Interestingly, the missense variants that do not affect KDM5C’s in vitro demethylase activity occur in two different regions of the protein. The D87G variant is at the N-terminal extreme of the A/T-rich interaction domain (ARID) that can bind both A/T- and C/G-rich DNA sequences in vitro [72,73]. While this change could alter the ability of KDM5C to be recruited to some target genes, structural modeling studies suggest that this variant is unlikely to affect ARID-mediated DNA binding [74]. Instead, this change could affect protein–protein interactions necessary for KDM5C to regulate the expression of its target genes. The other variant, R1115H, occurs in a region of unknown function toward the C terminus of KDM5C. Like D87G, this change could alter critical protein–protein interactions. Additional evidence supporting nonenzymatic roles comes from Drosophila, where the regulation of axonal growth and guidance by KDM5 in Kenyon cells occurs independently of its demethylase activity [48]. Precisely how KDM5 family proteins regulate neuronal gene expression via nonenzymatic mechanisms is still not clear. However, the involvement of additional mechanisms of gene regulation by KDM5C in neuronal lineages is unsurprising given the multidomain nature of this family of proteins (Fig. 1). Indeed, there is now considerable evidence that all KDM5 family proteins can regulate gene expression by multiple mechanisms, such as by interacting with lysine deacetylases and chromatin remodelers [49,75,76]. These data highlight the complex nature of KDM5-regulated gene expression and suggest that there may be more than one way that mutations in KDM5 family genes can lead to cognitive phenotypes.

Conclusions and perspectives

Since the first molecular identification of KDM5C variants in patients with ID in 2005 [10], many additional pathogenic alleles have been identified in individuals with NDDs. More recently, it has also become clear that a more general feature of KDM5 paralogs may be to regulate critical neuronal functions. Most notably, variants in KDM5B have recently been observed in individuals with NDDs and can result in clinical features that overlap with, but are not identical to, those observed in individuals with Claes-Jensen syndrome [5,77-81]. Despite the clear link between KDM5 proteins and cognition, we still have much to learn about how these proteins function molecularly to orchestrate gene expression programs that are needed for brain development. For example, we still lack a basic understanding of how KDM5 proteins are recruited to their target promoters, in addition to which proteins they interact with that facilitate their transcriptional regulatory functions. It is also possible that KDM5C acts through nontranscriptional means to affect neuronal development and function. Key to these fundamental discoveries will be the use of animal model systems. In addition, there is a great deal of excitement about the development of organoid models generated from human induced pluripotent stem cells. Cerebral organoids recapitulate some of the structural and molecular aspects of brain development and are increasingly used to understand the basis of NDDs [82-87]. Analyses of organoids are therefore expected to complement studies in model organisms to provide a more complete understanding of the effects of specific patient alleles on neuronal development and function. This fundamental knowledge will, in turn, lead to the development of targeted therapies to help individuals with Claes-Jensen syndrome.

Acknowledgments

We thank members of the Secombe lab for helpful discussions and feedback on this review. JS is supported by National Institutes of Health (NIH) R01GM112783, R01AG053269, and P50HD105352 in addition to an Irma T. Hirschl career award, and HAMH is supported by NIH Ruth L. Kirschstein National Research Service Award F31NS110278.

Abbreviations

ARID

A/T-rich interaction domain

ASD

autism spectrum disorder

CJ-XLID

Claes-Jensen X-linked intellectual disability

DD

developmental delay

DSM

diagnostic and statistical manual of mental disorders

H3K4me3

histone H3 trimethylated at lysine residue at amino acid position 4

ID

intellectual disability

IQ

intelligence quotient

JmjC

jumonji C-terminal domain

JmjN

jumonji N-terminal domain

KDM5

lysine demethylase 5

KDM5C-RD

KDM5C-related disorder

KMT2A

lysine methyltransferase 2A

KO

knock out

MRI

magnetic resonance imaging

MRXSCJ

mental retardation, X-linked, syndromic, Claes-Jensen type

NDD

neurodevelopmental disorder

NMJ

neuromuscular junction

OMIM

online Mendelian inheritance in man

Footnotes

Conflict of interest

The authors declare no conflict of interest.

References

  • 1.Leppert B, Havdahl A, Riglin L, Jones HJ, Zheng J, Davey Smith G, Tilling K, Thapar A, Reichborn-Kjennerud T & Stergiakouli E (2019) Association of maternal neurodevelopmental risk alleles with early-life exposures. JAMA Psych 76, 834–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Han VX, Patel S, Jones HF, Nielsen TC, Mohammad SS, Hofer MJ, Gold W, Brilot F, Lain SJ, Nassar N et al. (2021) Maternal acute and chronic inflammation in pregnancy is associated with common neurodevelopmental disorders: a systematic review. Transl Psych 11, 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Joseph RM, Korzeniewski SJ, Allred EN, O’Shea TM, Heeren T, Frazier JA, Ware J, Hirtz D, Leviton A, Kuban K et al. (2017) Extremely low gestational age and very low birthweight for gestational age are risk factors for autism spectrum disorder in a large cohort study of 10-year-old children born at 23–27 weeks’ gestation. Am J Obstet Gynecol 216, 304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy MI, Brown MA & Yang J (2017) 10 Years of GWAS Discovery: Biology, Function, and Translation. Am J Hum Genet 101, 5–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.De Rubeis S, He X, Goldberg AP, Poultney CS, Samocha K, Cicek AE, Kou Y, Liu L, Fromer M et al. (2014) Synaptic, transcriptional and chromatin genes disrupted in autism. Nature 515, 209–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.van Bokhoven H (2011) Genetic and epigenetic networks in intellectual disabilities. Annu Rev Genet 45, 81–104. [DOI] [PubMed] [Google Scholar]
  • 7.Iwase S, Brookes E, Agarwal S, Badeaux AI, Ito H, Vallianatos CN, Tomassy GS, Kasza T, Lin G, Thompson A et al. (2016) A mouse model of X-linked intellectual disability associated with impaired removal of histone methylation. Cell Rep 14, 1000–1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Scandaglia M, Lopez-Atalaya JP, Medrano-Fernandez A, Lopez-Cascales MT, Del Blanco B, Lipinski M, Benito E, Olivares R, Iwase S, Shi Y et al. (2017) Loss of Kdm5c causes spurious transcription and prevents the fine-tuning of activity-regulated enhancers in neurons. Cell Rep 21, 47–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hatch HAM, O’Neil MH, Marion RW, Secombe J & Shulman LH. (2021) Caregiver-reported characteristics of children diagnosed with pathogenic variants in KDM5C. Am J Med Genet A 185, 75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Jensen LR, Amende M, Gurok U, Moser B, Gimmel V, Tzschach A, Janecke AR, Tariverdian G, Chelly J, Fryns JP et al. (2005) Mutations in the JARID1C gene, which is involved in transcriptional regulation and chromatin remodeling, cause X-linked mental retardation. Am J Hum Genet 76, 227–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Jensen LR, Bartenschlager H, Rujirabanjerd S, Tzschach A, Nümann A, Janecke AR, Spörle R, Stricker S, Raynaud M, Nelson J et al. (2010) A distinctive gene expression fingerprint in mentally retarded male patients reflects disease-causing defects in the histone demethylase KDM5C. PathoGenetics 3. 10.1186/1755-8417-3-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Secombe J, Li L, Carlos L & Eisenman RN (2007) The Trithorax group protein Lid is a trimethyl histone H3K4 demethylase required for dMyc-induced cell growth. Genes Dev 21, 537–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Christensen J, Agger K, Cloos PA, Pasini D, Rose S, Sennels L, Rappsilber J, Hansen KH, Salcini AE & Helin K (2007) RBP2 belongs to a family of demethylases, specific for tri-and dimethylated lysine 4 on histone 3. Cell 128, 1063–1076. [DOI] [PubMed] [Google Scholar]
  • 14.Iwase S, Lan F, Bayliss P, de la Torre-Ubieta L, Huarte M, Qi HH, Whetstine JR, Bonni A, Roberts TM & Shi Y (2007) The X-linked mental retardation gene SMCX/JARID1C defines a family of histone H3 lysine 4 demethylases. Cell 128, 1077–1088. [DOI] [PubMed] [Google Scholar]
  • 15.Tahiliani M, Mei P, Fang R, Leonor T, Rutenberg M, Shimizu F, Li J, Rao A & Shi Y (2007) The histone H3K4 demethylase SMCX links REST target genes to X-linked mental retardation. Nature 447, 601–605. [DOI] [PubMed] [Google Scholar]
  • 16.Howe FS, Fischl H, Murray SC & Mellor J (2017) Is H3K4me3 instructive for transcription activation? BioEssays 39, e201600095. 10.1002/bies.201600095 [DOI] [PubMed] [Google Scholar]
  • 17.Ram O, Goren A, Amit I, Shoresh N, Yosef N, Ernst J, Kellis M, Gymrek M, Issner R, Coyne M et al. (2011) Combinatorial patterning of chromatin regulators uncovered by genome-wide location analysis in human cells. Cell 147, 1628–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Vallianatos CN, Raines B, Porter RS, Bonefas KM, Wu MC, Garay PM, Collette KM, Seo YA, Dou Y, Keegan CE et al. (2020) Mutually suppressive roles of KMT2A and KDM5C in behaviour, neuronal structure, and histone H3K4 methylation. Commun Biol 3, 278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Assocation AP (2013) Diagnostic and statistical manual of mental disorders, 5th edn. [Google Scholar]
  • 20.Patel DR, Cabral MD, Ho A & Merrick J (2020) A clinical primer on intellectual disability. Transl Pediatr 9, S23–S35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Obi O, Van Naarden Braun K, Baio J, Drews-Botsch C, Devine O & Yeargin-Allsopp M (2011) Effect of incorporating adaptive functioning scores on the prevalence of intellectual disability. Am J Intellect Dev Disabil 116, 360–370. [DOI] [PubMed] [Google Scholar]
  • 22.Abidi FE, Holloway L, Moore CA, Weaver DD, Simensen RJ, Stevenson RE, Rogers RC & Schwartz CE (2008) Mutations in JARID1C are associated with X-linked Mental retardation, short stature and hyperreflexia. J Med Genet 45, 787–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ding N, Zhang P, Mao Y, Feng S, Gao Z, Chen Q & Zhang X (2020) Clinical features and gene variant of a pedigree affected with X-linked recessive mental retardation Claes-Jensen type. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 37, 1352–1355. [DOI] [PubMed] [Google Scholar]
  • 24.Rujirabanjerd S, Nelson J, Tarpey PS, Hackett A, Edkins S, Raymond FL, Schwartz CE, Turner G, Iwase S, Shi Y et al. (2010) Identification and characterization of two novel JARID1C mutations: suggestion of an emerging genotype-phenotype correlation. Eur J Hum Genet 18, 330–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tzschach A, Grasshoff U, Beck-Woedl S, Dufke C, Bauer C, Kehrer M, Evers C, Moog U, Oehl-Jaschkowitz B, Di Donato N et al. (2015) Next-generation sequencing in X-linked intellectual disability. Eur J Hum Genet 23, 1513–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Carmignac V, Nambot S, Lehalle D, Callier P, Moortgat S, Benoit V, Ghoumid J, Delobel B, Smol T, Thuillier C et al. (2020) Further delineation of the female phenotype with KDM5C disease causing variants: 19 new individuals and review of the literature. Clin Genet 98, 43–45. [DOI] [PubMed] [Google Scholar]
  • 27.Kawano-Matsuda F, Maeda T, Kaname T, Yanagi K & Ihara K (2021) X-linked mental retardation and severe short stature with a novel mutation of the KDM5C gene. Clin Pediatr Endocrinol 30, 61–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ounap K, Puusepp-Benazzouz H, Peters M, Vaher U, Rein R, Proos A, Field M & Reimand T (2012) A novel c.2T > C mutation of the KDM5C/JARID1C gene in one large family with X-linked intellectual disability. Eur J Med Genet 55, 178–184. [DOI] [PubMed] [Google Scholar]
  • 29.Simensen RJ, Rogers RC, Collins JS, Abidi F, Schwartz CE & Stevenson RE (2012) Short-term memory deficits in carrier females with KDM5C mutations. Genet Couns 23, 31–40. [PubMed] [Google Scholar]
  • 30.Heine-Suñer D, Torres-Juan L, Morlà M, Busquets X, Barceló F, Picó G, Bonilla L, Govea N, Bernués M & Rosell J (2003) Fragile-X syndrome and skewed X-chromosome inactivation within a family: A female member with complete inactivation of the functional X chromosome. Am J Med Genet A 122A, 108–114. 10.1002/ajmg.a.20160 [DOI] [PubMed] [Google Scholar]
  • 31.Mila M, Alvarez-Mora MI, Madrigal I & Rodriguez-Revenga L (2018) Fragile X syndrome: An overview and update of the FMR1 gene. Clin Genet 93, 197–205. [DOI] [PubMed] [Google Scholar]
  • 32.Agulnik AI, Mitchell MJ, Mattei MG, Borsani G, Avner PA, Lerner JL & Bishop CE (1994) A novel X-gene with a widely transcribed Y-linked homolog escapes X-inactivation in mouse and human. Hum Mol Genet 3, 879–884. [DOI] [PubMed] [Google Scholar]
  • 33.Xu J, Deng X & Disteche CM (2008) Sex-Specific Expression of the X-Linked Histone Demethylase Gene Jarid1c in Brain. PLoS One 3, e2553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Li N & Carrel L (2008) Escape from X chromosome inactivation is an intrinsic property of the Jarid1c locus. Proc Natl Acad Sci USA 105, 17055–17060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Vallianatos CN, Farrehi C, Friez MJ, Burmeister M, Keegan CE & Iwase S (2018) Altered gene-regulatory function of KDM5C by a Novel Mutation associated with autism and intellectual disability. Front Mol Neurosci 11, 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Santos-Reboucas CB, Fintelman-Rodrigues N, Jensen LR, Kuss AW, Ribeiro MG, Campos M Jr, Santos JM & Pimentel MM (2011) A novel nonsense mutation in KDM5C/JARID1C gene causing intellectual disability, short stature and speech delay. Neurosci Lett 498, 67–71. [DOI] [PubMed] [Google Scholar]
  • 37.Tonnesen J & Nagerl UV (2016) Dendritic spines as tunable regulators of synaptic signals. Front Psychiatry 7, 101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Pchitskaya E & Bezprozvanny I (2020) dendritic spines shape analysis-classification or clusterization? perspective. Front Synaptic Neurosci 12, 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Huttenlocher PR (1991) Dendritic and synaptic pathology in mental retardation. Pediatr Neurol 7, 79–85. [DOI] [PubMed] [Google Scholar]
  • 40.Purpura DP (1974) Dendritic spine "dysgenesis" and mental retardation. Science 186, 1126–1128. [DOI] [PubMed] [Google Scholar]
  • 41.Gatto CL & Broadie K (2011) Drosophila modeling of heritable neurodevelopmental disorders. Curr Opin Neurobiol 21, 834–841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Deshpande M & Rodal AA (2016) The crossroads of synaptic growth signaling, membrane traffic and neurological disease: insights from Drosophila. Traffic 17, 87–101. [DOI] [PubMed] [Google Scholar]
  • 43.Bellosta P & Soldano A (2019) Dissecting the genetics of autism spectrum disorders: a drosophila perspective. Front Physiol 10, 987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Belalcazar HM, Hendricks EL, Zamurrad S, Liebl FLW & Secombe J (2021) The histone demethylase KDM5 is required for synaptic structure and function at the Drosophila neuromuscular junction. Cell Rep 34, 108753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Chen K, Luan X, Liu Q, Wang J, Chang X, Snijders AM, Mao JH, Secombe J, Dan Z, Chen JH et al. (2019) Drosophila histone demethylase KDM5 regulates social behavior through immune control and gut microbiota maintenance. Cell Host Microbe 25, 537–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Mariano V, Achsel T, Bagni C & Kanellopoulos AK (2020) Modelling learning and memory in drosophila to understand intellectual disabilities. Neuroscience 445, 12–30. [DOI] [PubMed] [Google Scholar]
  • 47.Volk L, Chiu SL, Sharma K & Huganir RL (2015) Glutamate synapses in human cognitive disorders. Annu Rev Neurosci 38, 127–149. [DOI] [PubMed] [Google Scholar]
  • 48.Hatch HAM, Belalcazar HM, Marshall OJ & Secombe J (2021) A KDM5-Prospero transcriptional axis functions during early neurodevelopment to regulate mushroom body formation. Elife 10, e63886. 10.7554/eLife.63886 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Mariani L, Lussi YC, Vandamme J, Riveiro A & Salcini AE (2016) The H3K4me3/2 histone demethylase RBR-2 controls axon guidance by repressing the actin-remodeling gene wsp-1. Development 143, 851–863. [DOI] [PubMed] [Google Scholar]
  • 50.Lopez-Bigas N, Kisiel TA, DeWaal DC, Holmes KB, Volkert TL, Gupta S, Love J, Murray HL, Young RA & Benevolenskaya EV (2008) Genome-wide analysis of the H3K4 histone demethylase RBP2 reveals a transcriptional program controlling differentiation. Mol Cell 31, 520–530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kidder BL, Hu G & Zhao K (2014) KDM5B focuses H3K4 methylation near promoters and enhancers during embryonic stem cell self-renewal and differentiation. Genome Biol 15, R32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Drelon C, Belalcazar HM & Secombe J (2018) The histone demethylase KDM5 is essential for larval growth in drosophila. Genetics 209, 773–787. 10.1534/genetics.118.301004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Liu X, Greer C & Secombe J (2014) KDM5 interacts with Foxo to modulate cellular levels of oxidative stress. PLoS Genet 10, e1004676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Liu X & Secombe J (2015) The histone demethylase KDM5 activates gene expression by recognizing chromatin context through its PHD reader motif. Cell Rep 13, 2219–2231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zamurrad S, Hatch HAM, Drelon C, Belalcazar HM & Secombe J (2018) A drosophila model of intellectual disability caused by mutations in the histone demethylase KDM5. Cell Rep 22, 2359–2369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lloret-Llinares M, Pérez-Lluch S, Rossell D, Morán T, Ponsa-Cobas J, Auer H, Corominas M & Azorín F (2012) dKDM5/LID regulates H3K4me3 dynamics at the transcription-start site (TSS) of actively transcribed developmental genes. Nucleic Acids Res 40, 9493–9505. 10.1093/nar/gks773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Iwase S, Berube NG, Zhou Z, Kasri NN, Battaglioli E, Scandaglia M & Barco A (2017) Epigenetic etiology of intellectual disability. J Neurosci 37, 10773–10782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Schaefer A, Sampath SC, Intrator A, Min A, Gertler TS, Surmeier DJ, Tarakhovsky A & Greengard P (2009) Control of cognition and adaptive behavior by the GLP/G9a epigenetic suppressor complex. Neuron 64, 678–691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ben-Shachar S, Chahrour M, Thaller C, Shaw CA & Zoghbi HY (2009) Mouse models of MeCP2 disorders share gene expression changes in the cerebellum and hypothalamus. Hum Mol Genet 18, 2431–2442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Samaco RC, Mandel-Brehm C, McGraw CM, Shaw CA, McGill BE & Zoghbi HY (2012) Crh and Oprm1 mediate anxiety-related behavior and social approach in a mouse model of MECP2 duplication syndrome. Nat Genet 44, 206–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Gal-Ben-Ari S, Kenney JW, Ounalla-Saad H, Taha E, David O, Levitan D, Gildish I, Panja D, Pai B, Wibrand K et al. (2012) Consolidation and translation regulation. Learn Mem 19, 410–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Jung H, Yoon BC & Holt CE (2012) Axonal mRNA localization and local protein synthesis in nervous system assembly, maintenance and repair. Nat Rev Neurosci 13, 308–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Slomnicki LP, Pietrzak M, Vashishta A, Jones J, Lynch N, Elliot S, Poulos E, Malicote D, Morris BE, Hallgren J et al. (2016) Requirement of neuronal ribosome synthesis for growth and maintenance of the dendritic tree. J Biol Chem 291, 5721–5739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Dockendorff TC & Labrador M. (2019) The Fragile X protein and genome function. Mol Neurobiol 56, 711–721. [DOI] [PubMed] [Google Scholar]
  • 65.Borrie SC, Brems H, Legius E & Bagni C (2017) Cognitive dysfunctions in intellectual disabilities: the contributions of the Ras-MAPK and PI3K-AKT-mTOR pathways. Annu Rev Genomics Hum Genet 18, 115–142. [DOI] [PubMed] [Google Scholar]
  • 66.Ding Q, Markesbery WR, Chen Q, Li F & Keller JN (2005) Ribosome dysfunction is an early event in Alzheimer’s disease. J Neurosci 25, 9171–9175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Fernandez E, Rajan N & Bagni C (2013) The FMRP regulon: from targets to disease convergence. Front Neurosci 7, 191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Halliday M, Radford H, Zents KAM, Molloy C, Moreno JA, Verity NC, Smith E, Ortori CA, Barrett DA, Bushell M et al. (2017) Repurposed drugs targeting eIF2&alpha;-P-mediated translational repression prevent neurodegeneration in mice. Brain 140, 1768–1783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Talos DM, Sun H, Zhou X, Fitzgerald EC, Jackson MC, Klein PM, Lan VJ, Joseph A & Jensen FE (2012) The interaction between early life epilepsy and autistic-like behavioral consequences: a role for the mammalian target of rapamycin (mTOR) pathway. PLoS One 7, e35885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Troca-Marin JA, Alves-Sampaio A & Montesinos ML (2012) Deregulated mTOR-mediated translation in intellectual disability. Prog Neurogibol 96, 268–282. [DOI] [PubMed] [Google Scholar]
  • 71.Vallianatos CN & Iwase S (2015) Disrupted intricacy of histone H3K4 methylation in neurodevelopmental disorders. Epigenomics 7, 503–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Tu S, Teng YC, Yuan C, Wu YT, Chan MY, Cheng AN, Lin PH, Juan LJ & Tsai MD (2008) The ARID domain of the H3K4 demethylase RBP2 binds to a DNA CCGCCC motif. Nat Struct Mol Biol 15, 419–421. [DOI] [PubMed] [Google Scholar]
  • 73.Peng Y, Suryadi J, Yang Y, Kucukkal TG, Cao W & Alexov E (2015) Mutations in the KDM5C ARID domain and their plausible association with syndromic claes-jensen-type disease. Int J Mol Sci 16, 27270–27287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Peng Y & Alexov E (2016) Cofactors-loaded quaternary structure of lysine-specific demethylase 5C (KDM5C) protein: Computational model. Proteins 84, 1797–1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lee N, Erdjument-Bromage H, Tempst P, Jones RS & Zhang Y (2009) The H3K4 demethylase lid associates with and inhibits histone deacetylase Rpd3. Mol Cell Biol 29, 1401–1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Nishibuchi G, Shibata Y, Hayakawa T, Hayakawa N, Ohtani Y, Sinmyozu K, Tagami H & Nakayama J (2014) Physical and functional interactions between the histone H3K4 demethylase KDM5A and the nucleosome remodeling and deacetylase (NuRD) complex. J Biol Chem 289, 28956–28970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Najmabadi H, Hu H, Garshasbi M, Zemojtel T, Abedini SS, Chen W, Hosseini M, Behjati F, Haas S, Jamali P et al. (2011) Deep sequencing reveals 50 novel genes for recessive cognitive disorders. Nature 478, 57–63. [DOI] [PubMed] [Google Scholar]
  • 78.Butler MG, Rafi SK, Hossain W, Stephan DA & Manzardo AM (2015) Whole exome sequencing in females with autism implicates novel and candidate genes. Int J Mol Sci 16, 1312–1335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.El Hayek L, Tuncay IO, Nijem N, Russell J, Ludwig S, Kaur K, Li X, Anderton P, Tang M et al. (2020) KDM5A mutations identified in autism spectrum disorder using forward genetics. Elife 9, 81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Iossifov I, O’Roak BJ, Sanders SJ, Ronemus M, Krumm N, Levy D, Stessman HA, Witherspoon KT, Vives L, Patterson KE et al. (2014) The contribution of de novo coding mutations to autism spectrum disorder. Nature 515, 216–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Martin HC, Jones WD, McIntyre R, Sanchez-Andrade G, Sanderson M, Stephenson JD, Jones CP, Handsaker J, Gallone G, Bruntraeger M et al. (2018) Quantifying the contribution of recessive coding variation to developmental disorders. Science 362, 1161–1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Ilieva M, Fex Svenningsen A, Thorsen M & Michel TM (2018) Psychiatry in a Dish: stem cells and brain organoids modeling autism spectrum disorders. Biol Psychiatry 83, 558–568. [DOI] [PubMed] [Google Scholar]
  • 83.Mariani J, Coppola G, Zhang P, Abyzov A, Provini L, Tomasini L, Amenduni M, Szekely A, Palejev D, Wilson M et al. (2015) FOXG1-Dependent Dysregulation of GABA/Glutamate neuron differentiation in autism spectrum disorders. Cell 162, 375–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lancaster MA, Corsini NS, Wolfinger S, Gustafson EH, Phillips AW, Burkard TR, Otani T, Livesey FJ & Knoblich JA (2017) Guided self-organization and cortical plate formation in human brain organoids. Nat Biotechnol 35, 659–666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Camp JG, Badsha F, Florio M, Kanton S, Gerber T, Wilsch-Brauninger M, Lewitus E, Sykes A, Hevers W, Lancaster M et al. (2015) Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc Natl Acad Sci USA 112, 15672–15677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Sloan SA, Andersen J, Pasca AM, Birey F & Pasca SP (2018) Generation and assembly of human brain region-specific three-dimensional cultures. Nat Protoc 13, 2062–2085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Sasai Y (2013) Next-generation regenerative medicine: organogenesis from stem cells in 3D culture. Cell Stem Cell 12, 520–530. [DOI] [PubMed] [Google Scholar]
  • 88.Fujita A, Waga C, Hachiya Y, Kurihara E, Kumada S, Takeshita E, Nakagawa E, Inoue K, Miyatake S, Tsurusaki Y et al. (2016) Different X-linked KDM5C mutations in affected male siblings: is maternal reversion error involved? Clin Genet 90, 276. [DOI] [PubMed] [Google Scholar]
  • 89.Faundes V, Newman WG, Bernardini L, Canham N, Clayton-Smith J, Dallapiccola B, Davies SJ, Demos MK, Goldman A et al. (2018) Histone lysine methylases and demethylases in the landscape of human developmental disorders. Am J Hum Genet 102, 175–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Tzschach A, Lenzner S, Moser B, Reinhardt R, Chelly J, Fryns JP, Kleefstra T, Raynaud M, Turner G, Ropers HH et al. (2006) Novel JARID1C/SMCX mutations in patients with X-linked mental retardation. Hum Mutat 27, 389. [DOI] [PubMed] [Google Scholar]
  • 91.Tahiliani M, Mei PC, Fang R, Leonor T, Rutenberg M, Shimizu F, Li J, Rao A & Shi YJ (2007) The histone H3K4 demethylase SMCX links REST target genes to X-linked mental retardation. Nature 447, 601. [DOI] [PubMed] [Google Scholar]
  • 92.Hu H, Haas SA, Chelly J, Van Esch H, Raynaud M, de Brouwer AP, Weinert S, Froyen G, Frints SG, Laumonnier F et al. (2016) X-exome sequencing of 405 unresolved families identifies seven novel intellectual disability genes. Mol Psychiatry 21, 133–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Fokstuen S, Makrythanasis P, Hammar E, Guipponi M, Ranza E, Varvagiannis K, Santoni FA, Albarca-Aguilera M, Poleggi ME, Couchepin F et al. (2016) Experience of a multidisciplinary task force with exome sequencing for Mendelian disorders. Hum Genomics 10, 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Brookes E, Laurent B, Ounap K, Carroll R, Moeschler JB, Field M, Schwartz CE, Gecz J & Shi Y (2015) Mutations in the intellectual disability gene KDM5C reduce protein stability and demethylase activity. Hum Mol Genet 24, 2861–2872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Guerra JVS, Oliveira-Santos J, Oliveira DF, Leal GF, Oliveira JRM, Costa SS, Krepischi ACV, Vianna-Morgante AM & Maschietto M (2020) DNA methylation fingerprint of monozygotic twins and their singleton sibling with intellectual disability carrying a novel KDM5C mutation. Eur J Med Genet 63, 103737. [DOI] [PubMed] [Google Scholar]
  • 96.Santos C, Rodriguez-Revenga L, Madrigal I, Badenas C, Pineda M & Mila M (2006) A novel mutation in JARID1C gene associated with mental retardation. Eur J Hum Genet 14, 583–586. [DOI] [PubMed] [Google Scholar]
  • 97.Grafodatskaya D, Chung BH, Butcher DT, Turinsky AL, Goodman SJ, Choufani S, Chen YA, Lou Y, Zhao C, Rajendram R et al. (2013) Multilocus loss of DNA methylation in individuals with mutations in the histone H3 lysine 4 demethylase KDM5C. BMC Med Genomics 6, 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kim SH, Kim B, Lee JS, Kim HD, Choi JR, Lee ST & Kang HC (2019) Proband-only clinical exome sequencing for neurodevelopmental disabilities. Pediatr Neurol 99, 47–54. [DOI] [PubMed] [Google Scholar]
  • 99.Wang YR, Miao LY, Xu YF, Chen Y, Ding Y, Li J, Wang J, Wang XM & Huang XD (2020) Two cases of X-linked mental retardation, Claes-Jensen syndrome caused by variation of KDM5C gene. Zhonghua Er Ke Za Zhi 58, 426–427. [DOI] [PubMed] [Google Scholar]
  • 100.Scocchia A, Wigby KM, Masser-Frye D, Del Campo M, Galarreta CI, Thorpe E, McEachern J, Robinson K, Gross A, Interpretation I et al. (2019) Clinical whole genome sequencing as a first-tier test at a resource-limited dysmorphology clinic in Mexico. NPJ Genom Med 4, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Vissers LE, de Ligt J, Gilissen C, Janssen I, Steehouwer M, de Vries P, van Lier B, Arts P, Wieskamp N, del Rosario M et al. (2010) A de novo paradigm for mental retardation. Nat Genet 42, 1109–1112. [DOI] [PubMed] [Google Scholar]
  • 102.Turro E, Astle WJ, Megy K, Graf S, Greene D, Shamardina O, Allen HL, Sanchis-Juan A, Frontini M, Thys C et al. (2020) Whole-genome sequencing of patients with rare diseases in a national health system. Nature 583, 96–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Grozeva D, Carss K, Spasic-Boskovic O, Tejada MI, Gecz J, Shaw M, Corbett M, Haan E, Thompson E et al. (2015) Targeted next-generation sequencing analysis of 1000 individuals with intellectual disability. Hum Mutat 36, 1197–1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Tarpey PS, Smith R, Pleasance E, Whibley A, Edkins S, Hardy C, O’Meara S, Latimer C, Dicks E, Menzies A et al. (2009) A systematic, large-scale resequencing screen of X-chromosome coding exons in mental retardation. Nat Genet 41, 535–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Redin C, Gerard B, Lauer J, Herenger Y, Muller J, Quartier A, Masurel-Paulet A, Willems M, Lesca G, El-Chehadeh S et al. (2014) Efficient strategy for the molecular diagnosis of intellectual disability using targeted high-throughput sequencing. J Med Genet 51, 724–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Claes S, Devriendt K, Van Goethem G, Roelen L, Meireleire J, Raeymaekers P, Cassiman JJ & Fryns JP (2000) Novel Syndromic Form of X-linked Complicated Spastic Paraplegia. Am J Med Genet Part A 94, 1–4. [DOI] [PubMed] [Google Scholar]
  • 107.Cherot E, Keren B, Dubourg C, Carre W, Fradin M, Lavillaureix A, Afenjar A, Burglen L, Whalen S, Charles P et al. (2018) Using medical exome sequencing to identify the causes of neurodevelopmental disorders: Experience of 2 clinical units and 216 patients. Clin Genet 93, 567–576. [DOI] [PubMed] [Google Scholar]
  • 108.Adegbola A, Gao HL, Sommer S & Browning M (2008) A novel mutation in JARID1C/SMCX in a patient with autism spectrum disorder (ASD). Am J Med Genet A 146A, 505–511. [DOI] [PubMed] [Google Scholar]
  • 109.Alfares A, Alfadhel M, Wani T, Alsahli S, Alluhaydan I, Al Mutairi F, Alothaim A, Albalwi M, Al Subaie L, Alturki S et al. (2017) A multicenter clinical exome study in unselected cohorts from a consanguineous population of Saudi Arabia demonstrated a high diagnostic yield. Mol Genet Metab 121, 91–95. [DOI] [PubMed] [Google Scholar]
  • 110.Miller CR, Lee K, Pfau RB, Reshmi SC, Corsmeier DJ, Hashimoto S, Dave-Wala A, Jayaraman V, Koboldt D, Matthews T et al. (2020) Disease-associated mosaic variation in clinical exome sequencing: a two-year pediatric tertiary care experience. Cold Spring Harb Mol Case Stud 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Satterstrom FK, Kosmicki JA, Wang J, Breen MS, De Rubeis S, An JY, Peng M, Collins R, Grove J, Klei L et al. (2020) Large-scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism. Cell 180, 568–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Gao M, Xing M, Zhang K, Lyu Y, Ma J, Gai Z & Liu Y (2020) Clinical phenotype and genetic analysis of a Chinese patient featuring X-linked Claes-Jensen type syndromic mental retardation. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 37, 736–738. [DOI] [PubMed] [Google Scholar]
  • 113.Deciphering Developmental Disorders (2015) Large-scale discovery of novel genetic causes of developmental disorders. Nature 519, 223–228. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES