Skip to main content

Epigenetics of cognition and behavior: insights from Mendelian disorders of epigenetic machinery

Abstract

Epigenetics, one mechanism by which gene expression can change without any changes to the DNA sequence, was described nearly a century ago. However, the importance of epigenetic processes to neurodevelopment and higher order neurological functions like cognition and behavior is only now being realized. A group of disorders known as the Mendelian disorders of the epigenetic machinery are caused by the altered function of epigenetic machinery proteins, which consequently affects downstream expression of many genes. These disorders almost universally have cognitive dysfunction and behavioral issues as core features. Here, we review what is known about the neurodevelopmental phenotypes of some key examples of these disorders divided into categories based on the underlying function of the affected protein. Understanding these Mendelian disorders of the epigenetic machinery can illuminate the role of epigenetic regulation in typical brain function and can lead to future therapies and better management for a host of neurodevelopmental and neuropsychological disorders.

Background

In 1942, Conrad Waddington coined the term “epigenetics” to describe biological events that occur in response to external factors, therefore providing a relationship between genotype and phenotype [1]. Today, it is known that epigenetic changes are heritable modifications that alter gene expression patterns without changing the DNA sequence. This is done through DNA methylation, histone modifications, chromatin remodeling, and RNA-based mechanisms [2]. Together, these mechanisms are crucial for a wide range of processes, including early development, X-inactivation, imprinting, and tissue-specific gene regulation, which, if disturbed, can lead to detrimental consequences. Here, we will focus on the role epigenetics plays in neurodevelopment, specifically utilizing Mendelian disorders of the epigenetic machinery (MDEMs) as examples to explain the underpinnings of the consequences of dysregulation.

Epigenetics in neurodevelopment

When examining the genes that have been found to be causative of neurodevelopmental disabilities (NDDs), several patterns emerge. Many of the genes share common pathways including neurogenesis—developmental and adult, proliferation and differentiation of neural progenitors, neural migration, axonal guidance, disrupted synaptogenesis, impaired synaptic function, and imbalance of excitatory and inhibitory signaling [3, 4]. Epigenetic regulation and chromatin modification play a crucial role in all aspects of these processes.

The most common type of DNA methylation occurs at the fifth position of cytosine at CpG dinucleotides and is traditionally associated with repression of gene expression at promoters, although several studies have suggested it can also be involved in upregulation of expression [5]. The regulation of this process is vital for neurodevelopment as suggested by the fact that certain brain regions and cell types have specific methylation signatures and that these methylation profiles start in flux and become fixed over the course of development [6, 7]. Additionally, non-CpG methylation is specifically enriched in brain tissues and accumulates throughout development into adulthood and has been shown to be crucial for both neurogenesis and maturation of neural progenitor cells [8]. DNA methylation is also the mechanism by which genes are imprinted. Imprinting is the process by which only one allele of certain autosomal genes is expressed in the body or in a certain tissue depending on the allele’s parent of origin. Imprinting disorders occur when the expression of imprinted genes are disrupted by a sequence change including deletion or duplication or epigenetic regulation. While imprinting disorders are outside the scope of this review, the existing literature on NDDs with disrupted genomic imprinting—like Prader-Willi syndrome and Angelman syndrome—highlight the vital role of imprinting on the regulation of gene expression, and subsequent, downstream effects on cognitive and behavioral functioning [9,10,11].

In addition to DNA methylation, 3D chromatin structure and the dynamic process of chromatin remodeling is a crucial piece underlying the transition of cells from neural progenitor cells (NPCs) to their terminally differentiated cell types. Along this differentiation process, certain regions of chromatin become increasingly more compressed causing certain genes—presumably those involved in dictating NPC fate—to become permanently repressed [12, 13]. Specific chromatin remodeler proteins have been shown to be essential for proper brain development and neuronal differentiation. For example, heterozygous knockout of the chromatin remodeler Chd2 in mice leads to a distinct deficiency in production of GABAergic neurons specifically, significantly altering the cellular makeup of the cortex [14]. Knockout of Ctcf in mice, one of the main proteins responsible for 3D chromatin organization, led to increased apoptosis and disorganization of the forebrain and telencephalon [15]. Importantly, in humans, heterozygous loss of function of CTCF and CHD2 are both associated with neurodevelopmental disorders resulting in intellectual disability, seizures, and behavioral issues [16].

Histone modifications also play an essential role in the development and functioning of the nervous system. The idea of histone modifications leading to either “open” or “closed” chromatin has long been established [17]. “Open” chromatin, or euchromatin, allows for transcription, resulting in gene expression, whereas “closed” chromatin, or heterochromatin, is more compact, making it difficult for factors to bind, resulting in silenced gene expression. As such, disruption of methylation or demethylation and/or acetylation or deacetylation at a variety of spots leads to abnormalities in neurodevelopment. For example, deletion of Dpy30—a common protein subunit among many histone methyltransferases—in mouse brains leads to both neurogenic and gliogenic deficits [18]. The polycomb repressive complex (PRC), one of the best studied mechanisms of gene expression regulation, contains both histone methyltransferase and histone demethylase components. Ezh2, a histone methyltransferase component of PRC2, has been shown to stimulate neurogenesis and stimulate proper differentiation of NPCs into neurons and glia [19, 20]. Maintaining the appropriate balance of the state of histone acetylation also seems to be particularly important for differentiation of NPCs into terminal cell types. Several studies have shown that an orchestrated and tightly regulated decrease and then increase of H3K9 acetylation is necessary for early neural differentiation, and if this process is disrupted, differentiation is either inhibited altogether or mistimed [21, 22].

While we continue to learn more about the role of epigenetics in neurodevelopment, some of the best evidence available comes from examining the resulting cognitive and behavioral phenotype when one specific component of the epigenetic machinery is disrupted. These disorders are collectively referred to as Mendelian disorders of the epigenetic machinery [23].

Mendelian disorders of the epigenetic machinery (MDEMs)

The majority of MDEMs are caused by heterozygous loss of function variants in components of the machinery that perform the writing, erasing, reading, and remodeling of epigenetic marks [23]. Writers place the appropriate chemical groups, with the most widely studied alterations being methylation and acetylation. Methylation, or the addition of methyl group(s), can occur on both DNA, catalyzed by DNA methyltransferases (DNMTs) and typically associated with silencing of gene expression, and on histone proteins, catalyzed by histone methyltransferases (i.e., protein arginine methyltransferases (PRMTs) and histone lysine methyltransferases (KMTs)) which can result in open or closed chromatin. Acetylation, which occurs only on histones, is the addition of acetyl groups by histone acetyltransferases (HATs) and is typically associated with transcriptional activation [24]. Erasers, on the other hand, remove these modifications added to DNA or histones. Erasers include ten eleven translocation (TET) enzymes, which oxidize 5-methylcytosine to 5-hydroxymethylcytosine resulting in subsequent DNA demethylation, histone demethylases, which remove methyl groups from lysine residues in histones, and histone deacetylases (HDACs), which are enzymes that remove the acetyl group from histones [25].

These modifications then need to be recognized and complexes recruited, which is done by readers. Readers contain specialized docking domains which bind to specific covalent modifications placed by writers. For example, methyl CpG binding proteins (MBPs) bind to methylated DNA and recruit chromatin remodeling complexes to repress transcription [24]. The fourth type of epigenetic proteins are the remodelers. There are four structural families of chromatin remodelers including switch/sucrose non-fermenting (SWI/SNF), imitation switch (ISWI), inositol requiring 80-like (IN080), and chromodomain helicase DNA binding (CHD) [26]. These remodelers utilize adenosine triphosphate (ATP) to alter chromatin structures to either activate or repress gene expression.

It has become increasingly evident that variants causing alterations in the normal biallelic state of genes that encode proteins with direct roles in epigenetic regulation (www.epigeneticmachinery.org) often lead to disease. Many of these proteins that do cause disease when there is loss of function have dual epigenetic functions; for example, both an enzymatic and a reader domain [27]. Additionally, it has also been found that these genes that control epigenetic regulation have significantly higher probability of being loss of function intolerant (pLI) scores when compared to all other genes, indicating that these epigenetic machinery genes are highly intolerant to loss of function variation, with chromatin remodelers being the most highly intolerant group [27, 28]. This dosage sensitivity is an unusual feature of these genes, as about 80% of all Mendelian diseases caused by enzyme deficiency display recessive inheritance, whereas 86% of MDEMs display dominant inheritance [29, 30]. Interestingly, the vast majority of these MDEMs are associated with neurological dysfunction and more specifically, NDDs (Table 1) [23, 31]. This, along with the known role of epigenetics in neurodevelopment and ongoing neural circuitry functioning outlined above, underscores the importance of these mechanisms to the development of cognitive and behavioral phenotypes in humans in both health and disease. Several of the disease-causing histone modifiers have overlapping molecular mechanisms (e.g., writers at the same lysine residue, regulation of transcription of the same genes, etc.) [32]. Additionally, many of these proteins work together in complexes or are codependent [5]. If it is determined from deep phenotyping that MDEMs resulting from proteins with overlapping functions or codependent functions have similar cognitive or neurobehavioral endophenotypes, it gives us invaluable information about how these proteins and their epigenetic mechanisms function in both the developing and adult brains, not only in pathologic states but in health as well. Here, we will outline what is known about the cognitive and behavioral profiles of certain MDEMs divided into broad mechanistic categories to begin to examine these associations. Although literature involving cognitive and behavioral phenotypes of MDEMs are generally limited, the following MDEMs represent those with a relatively larger body of research or clinical case reviews that are also molecularly and/or phenotypically representative of the group.

Table 1 MDEM genes associated with neurodevelopmental disabilities and reported neurobehavioral phenotypes

DNA methylationTatton-Brown-Rahman syndrome

Tatton-Brown-syndrome (TBRS) is an overgrowth disorder caused by loss of function variants in DNMT3A which is involved in encoding an epigenetic regulator that mediates DNA methylation [33]. TBRS is characterized by intellectual impairment with the majority in the moderate range (IQ 39 to 76) [34, 35]. Recent literature suggests that those with TBRS may present with stronger verbal reasoning skills than non-verbal or spatial processing skills [35]. TBRS is associated with a high incidence of autism spectrum disorder (36–44%) [34, 35], and neuropsychiatric concerns, which includes psychotic disorders or schizophrenia largely varying from 5 to 42% of published clinical samples [34, 36]. Aggressive behaviors, stereotypic behaviors, obsessive compulsive behaviors, anxiety, features of attention-deficit/hyperactivity disorder, and neurodevelopmental regression have also been observed, although these may be associated with the broader clinical presentation of autism spectrum disorder or psychosis [34, 37]. Consistent with human subject research, mouse models with a heterozygous mutation in Dnmt3a similarly present with increased anxiety-related behaviors including reduced exploration and increased freezing response, as well as abnormal social behaviors (e.g., reduced exploration and communication with other mice) and reduced social drive, albeit, cognition was less affected [38].

DNA demethylationBeck-Fahrner syndrome

Beck-Fahrner syndrome (BEFAHRS) is the first identified Mendelian disorder of DNA demethylation caused by TET3 deficiency [39]. Given the recent discovery of this syndrome, literature on the associated cognitive and behavioral phenotype is extremely limited. Of 11 cases of those with BEFAHRS or TET3 deficiency, all had global developmental delay or intellectual disability, most with hypotonia or hypermobility (9/11) and a little over half presented with autistic features (6/11) [39]. At present, the majority of the literature on TET3 deficiency has largely been conducted using animal models. Tet3 deficiency, deletion, or ablation in neurons have been linked to increased anxiety behaviors [40,41,42] and fear generalization in mice [42], in addition to impaired spatial orientation [41] and short-term memory [40], implicating its role in the neurogenesis of the hippocampus and prefrontal cortex. Moreover, expression of genes involved in memory formation are regulated by Tet3 levels [43]. Taken together, these findings may indicate hippocampal functions including spatial processing and semantic memory formation may be particularly affected among those with BEFAHRS, although this remains to be seen in human subject research. Anxiety and autistic traits, which may be bidirectionally related, may be more common features of the syndrome.

Chromatin remodeling—CHARGE syndrome

The majority of those diagnosed with CHARGE syndrome have heterozygous mutations in CHD7 [44]. CHARGE syndrome is characterized by growth retardation in addition to multiple organ anomalies that includes the heart, choanae, genital/urinary systems, ear, and ocular coloboma, and, less commonly, vertebral/limb, renal, and tracheal abnormalities [44,45,46]. Cognitive functioning associated with CHARGE syndrome vary significantly with some studies reporting from impaired to broadly average intellect (IQ 54–92 in a case series of 7 patients) [47] albeit nearly half of those with the syndrome present with intellectual impairment (IQ < 70) in studies with larger samples (study with 50 participants) [48]. Hearing loss was represented in about a third of the sample which may contribute to speech/language delay [48]. Clinical studies involving patients with CHARGE syndrome report primary weaknesses in visuospatial construction, sequential processing, and selective attention juxtaposed with relative strengths in semantic skills, logical reasoning, and planning [47]. Notably, brain malformations, microcephaly, and low vision were prognostic predictors of intellectual functioning [49]. The neurobehavioral profile associated with CHARGE syndrome includes increased incidence of psychiatric illnesses including obsessive compulsive disorder (43–49%) and anxiety disorder (37–53%) [50, 51] with potential high comorbidity of the two psychopathologies [52]. Depression (8–24%) [51, 53, 54] and attention deficit hyperactivity disorder (ADHD) (26–34%) [52, 53], self-injurious behaviors (40–54%) [51, 53,54,55], and aggressive behaviors (38–53%) [51, 53, 54, 56] are also commonly found in those with this syndrome. Those with dual sensory impairment (deaf-blind) generally present with more challenging behaviors [53]. Autistic behaviors are commonly reported (26–43%) [50, 51, 53], although some research suggest these features may be due to sensory deficits, such as hearing loss and visual impairment, and are qualitatively different from the social impairment seen in idiopathic autism spectrum disorder [57].

Histone acetylation dysregulationRubinstein-Taybi syndrome and KAT6A syndrome

Rubinstein-Taybi syndrome (RTS) and KAT6A syndrome are both neurogenetic disorders with variants in genes that regulate gene expression via histone acetylation. Heterozygous mutations in CREBBP or EP300 genes—both involved in encoding transcription cofactors necessary for histone acetylation—cause RTS. KAT6A syndrome results from pathogenic variants in KAT6A which typically encodes lysine acetyltransferase and serves as a transcriptional coactivator. Despite shared disruptions in epigenetic machinery, emergent research suggests distinct cognitive and behavioral phenotypes across syndromes. While RTS is generally characterized by intellectual impairment and developmental delay, those with mutations in EP300, which represents a little less than 10% of cases [58], present with a milder form of intellectual disability than patients with variants in CREBBP [59]. Those with RTS have been described to show relative strengths in strong behavior regulation [60], social communication and affinity for interacting with others [61], regardless of severe intellectual impairment [62]. While limited studies have characterized cognitive functioning utilizing performance-based measures, the few extant investigations suggest verbal skills are relatively stronger than non-verbal and spatial skills among those with RTS [60, 63]. Behaviorally, attention problems, hyperactivity, and motor stereotypies are prevalent among those with RTS [61, 63]. Two studies report 37–43% of their samples with RTS met clinical cut-off for autism spectrum disorder utilizing screening questionnaires [64, 65]; however, given standardized measures were not used, these results may not reflect true prevalence of the developmental disorder. Externalizing problems are less consistently documented in research with RTS, whereas elevated rates of anxiety, obsessive compulsive disorder, and depression have been reported, particularly with older age [66].

In contrast, those with KAT6A syndrome uniformly show intellectual disability and developmental delay—with receptive/expressive language and communication deficits as the most well-documented feature [67]. Nearly 70% of affected individuals are minimally verbal [68]. Comprehension skills are reported to be more preserved than expressive language [67]. To date, with the exception of St. John et al. [68], descriptions of the syndrome have largely relied on retrospective review of medical history or case studies, and thus most non-verbal cognitive functions (e.g., non-verbal reasoning, spatial processing, executive functions, etc.) remains poorly characterized. Studies have documented 25–33% of those with KAT6A syndrome have a diagnosis of autism spectrum disorder although the rates may vary based on diagnostic methods applied [68]. Emotional disturbances appear to be less of a concern among affected individuals [69]. Heterogeneity in measurement tools combined with the absence of standardized neuropsychological assessments leave cross-MDEM comparisons challenging. Subsequent investigations may consider alternate methodologies (e.g., eye tracking) that can capture cognitive and behavioral processes across syndromes.

Histone methylation dysregulationKabuki syndrome and Wiedemann-Steiner syndrome

Kabuki syndrome (KS) and Wiedemann-Steiner syndrome (WSS) are two MDEMS caused by pathogenic variants of a gene from the same KMT family of proteins. Most cases of KS result from heterozygous variants in KMT2D (80%) or KMD6A (5–10%) [70], and WSS is due to haploinsufficiency of KMT2A [71]. The KMT genes encode histone methyltransferases, thus, these disorders disrupt histone methylation and chromatin remodeling, with both KMT2D and KMT2A also having reader domains. Emergent evidence on the cognitive and neurobehavioral phenotypes of KS and WSS suggest some shared features. Intellectual functioning associated with KS ranges from severe impairment to average functioning although mild to moderate intellectual disability constitutes the majority of those affected [72,73,74]. Likewise, intellectual functioning estimates for those with WSS vary from moderate intellectual disability to average intelligence [75], with most in the mild to moderate impairment range [75, 76]. Interestingly, emergent evidence suggests some common characteristics in cognition across KS and WSS, with both sharing relative weaknesses in nonverbal reasoning, visuoconstruction, and visuospatial skills [74, 77,78,79], which implicate abnormal development of hippocampal formation as the potential underpinning of the shared pathogenesis of both MDEMs. Executive functioning and working memory are generally less affected in those with KS [77, 79], whereas recent case series examining cognitive functions in WSS suggest relative challenges in these areas [78]. KS is generally associated with few behavioral problems with anxiety, obsessive behaviors and attention difficulties most often observed by caregivers [80,81,82]. Social skills are considered a relative strength among those with KS, as individuals have been described as sweet and affectionate with relatively more preserved social adaptive skills [80, 83] paired with challenges in pragmatic language [83, 84]. Those with WSS present with elevated rates of anxiety [75, 78, 85] and behaviors concerning for ADHD [78, 85]. In contrast to the low levels of aggression seen in those with KS [80,81,82], relatively high rates of aggression [75] and conduct problems have been reported among those with WSS [85,86,87] which may be due to poorer executive functioning skills [85,86,87]. Rates of autism spectrum disorder in WSS vary widely across case studies given different clinical sample sizes and types of assessments used to determine diagnostic classification [75, 85, 88], although new evidence highlights similar high sociability and strong prosocial skills in those affected regardless of diagnosis of intellectual disability or autism spectrum disorder [88, 89]. In brief, KS and WSS are neurogenetic disorders with some common neuropsychological characteristics that may be reflective of shared disease-causing pathways which can be considered for future development of clinical trials.

Conclusions

Mendelian disorders of the epigenetic machinery are a rapidly expanding group of disorders that almost universally have cognitive and behavioral issues as core features, and together account for a large proportion of genetic intellectual disability. We can start to understand the mechanisms of epigenetic control of cognition and behavior by not only examining similarities and differences in the cognitive and behavioral endophenotypes of MDEMs as an entire group, but also by dividing them into the epigenetic function of the protein translated from the causative gene, e.g., histone methyltransferases, chromatin remodelers, DNA methylation erasers, etc. (Fig. 1).

Fig. 1
figure 1

Epigenetic modifier proteins control cognition and behavior. An illustration summarizing the broad categories of epigenetic mechanisms that are disrupted in select MDEMs. Specifically highlighted are the mechanisms and conditions described in detail in this review. Mutations in epigenetic protein modifiers—DNA methylation writers (green marker), erasers (pink eraser), readers (glasses), and chromatin remodelers (truck)—drive atypical cognitive and behavioral development in MDEMs. Specifically, loss of function in different components of epigenetic machinery results in unique neurodevelopmental disorders. Variants in DNMT3A (a writer) affect DNA methylation causing Tatton Brown Rahman syndrome. Variants in TET3 (an eraser) affect DNA demethylation causing Beck Fahrner syndrome. Variants in CHD7 (a remodeler) impact chromatin remodeling causing CHARGE syndrome. Variants in CREBBP/EP300 and KAT6A (primary writers with a reader component) result in histone acetylation dysregulation causing Rubinstein Taybi syndrome and KAT6A syndrome, respectively. Variants in KMT2D (primary writer with reader component)/KDM6A (eraser) and KMT2A (primary writer with reader component) dysregulate histone methylation causing Kabuki syndrome and Wiedemann-Steiner syndrome, respectively

As science further elucidates the role of epigenetics in neurodevelopment, and more and more MDEMs are discovered that cause cognitive and behavioral issues, this area of study holds a great deal of promise not only for understanding how typical cognition and behavior is controlled but also for therapies for neurodevelopmental disabilities. One such area of promise is in the use of genome-wide DNA methylation “signatures". Most MDEMs as well as a number of other genetic NDD syndromes such as fragile X, have a very specific pattern of DNA methylation at certain identified regions of the genome that is extremely differentiated from unaffected individuals and from those with other genetic syndromes that it can be used to diagnose these disorders [90, 91]. While this alone is very exciting, even more intriguing is the idea that subtle variations within these syndrome-specific signatures may correlate with aspects of the cognitive and behavioral phenotype and can be used not only for diagnosis but also for prognosis and therapeutic monitoring [92]. In the future, larger sample sizes of individuals with MDEMs with known episignatures need to undergo systematic deep phenotyping combined with genome-wide DNA methylation analysis in order to identify correlations both within and between syndromes. This can also help to identify common signature loci and attributes for certain neurodevelopmental endophenotypes. Moreover, these findings and methodology may be applied to much more broad categories of neurodevelopmental and neuropsychiatric disorders.

Finally, this area of research involving MDEMs is likely to lead to novel and effective therapies for neurodevelopmental and neuropsychiatric disorders more broadly. While MDEMs are the example of the most direct and detrimental dysregulations of the epigenetic machinery, numerous other disorders from autism to epilepsy to schizophrenia have been linked to epigenetic machinery dysfunction [93,94,95]. Therapies used in these MDEMs to restore epigenetic balance may also prove very beneficial for a host of more common conditions. In effect, these rare genetic NDDs may be an important window to the understanding of the developmental processes of cognition and behavior in the human brain as well as possible treatment mechanisms to address disruptions in these pathways.

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Abbreviations

ADHD:

Attention deficit hyperactivity disorder

ATP:

Adenosine triphosphate

BEFAHRS:

Beck-Fahrner syndrome

CHD:

Chromatin helicase domain

DNMT:

DNA methyltransferase

HAT:

Histone acetyltransferase

HDAC:

Histone deacetylase

KMT:

Lysine methyltransferase

KS:

Kabuki syndrome

MBP:

Methyl-binding protein

MDEM:

Mendelian disorder of the epigenetic machinery

NDD:

Neurodevelopmental disability

NPC:

Neural progenitor cells

pLI:

Probability of being loss of function intolerant

PRC:

Polycomb repressor complex

PRMT:

Protein arginine methyltransferase

RTS:

Rubinstein-Taybi syndrome

TBRS:

Tatton-Brown-Rahman syndrome

TET:

Ten eleven translocation

WSS:

Wiedemann-Steiner syndrome

IN080:

Inositol requiring 80-like

ISWI:

Imitation switch

SWI/SNF:

Switch/sucrose non-fermenting

References

  1. Waddington CH. The epigenotype. Endeavour. 1942;1:18–20.

  2. Gibney ER, Nolan CM. Epigenetics and gene expression. Heredity. 2010;105(1):4–13.

    Article  CAS  PubMed  Google Scholar 

  3. Iwase S, Bérubé NG, Zhou Z, Kasri NN, Battaglioli E, Scandaglia M, Barco A. Epigenetic etiology of intellectual disability. J Neurosci. 2017;37(45):10773–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Parenti I, Rabaneda LG, Schoen H, Novarino G. Neurodevelopmental disorders: from genetics to functional pathways. Trends Neurosci. 2020;43(8):608–21.

    Article  CAS  PubMed  Google Scholar 

  5. Salinas RD, Connolly DR, Song H. Invited review: epigenetics in neurodevelopment. Neuropathol Appl Neurobiol. 2020;46(1):6–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jang HS, Shin WJ, Lee JE, Do JT. CpG and non-CpG methylation in epigenetic gene regulation and brain function. Genes. 2017;8(6):148.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Ladd-Acosta C, Pevsner J, Sabunciyan S, Yolken RH, Webster MJ, Dinkins T, Callinan PA, Fan J, Potash JB, Feinberg AP. DNA methylation signatures within the human brain. Am J Hum Genet. 2007;81(6):1304–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lister R, Mukamel EA, Nery JR, Urich M, Puddifoot CA, Johnson ND, Lucero J, Huang Y, Dwork AJ, Schultz MD, Yu M, Tonti-Filippini J, Heyn H, Hu S, Wu JC, Rao A, Esteller M, He C, Haghighi FG, Sejnowski TJ, Behrens MM, Ecker JR. Global epigenomic reconfiguration during mammalian brain development. Science. 2013;341(6146):1237905.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Juriaans AF, Kerkhof GF, Mahabier EF, Sas TCJ, Zwaveling-Soonawala N, Touwslager RNH, Rotteveel J, Hokken-Koelega ACS. Temple syndrome: clinical findings, body composition and cognition in 15 patients. Clin Chem (Baltimore, Md). 2022;11(21):6289.

    Google Scholar 

  10. Heald M, Adams D, Walls E, Oliver C. Refining the behavioral phenotype of Angelman syndrome: examining differences in motivation for social contact between genetic subgroups. Front Behav Neurosci. 2021;15: 618271.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Baker EK, Butler MG, Hartin SN, Ling L, Bui M, Francis D, Rogers C, Field MJ, Slee J, Gamage D, Amor DJ, Godler DE. Relationships between UBE3A and SNORD116 expression and features of autism in chromosome 15 imprinting disorders. Transl Psychiatry. 2020;10(1):362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kishi Y, Fujii Y, Hirabayashi Y, Gotoh Y. HMGA regulates the global chromatin state and neurogenic potential in neocortical precursor cells. Nat Neurosci. 2012;15(8):1127–33.

    Article  CAS  PubMed  Google Scholar 

  13. Meshorer E, Yellajoshula D, George E, Scambler P, Brown D, Misteli T. Hyperdynamic plasticity of chromatin proteins in pluripotent embryonic stem cells. Dev Cell. 2012;22(1):233–4.

    Article  CAS  Google Scholar 

  14. Kim YJ, Khoshkhoo S, Frankowski JC, Zhu B, Abbasi S, Lee S, Wu YE, Hunt RF. Chd2 is necessary for neural circuit development and long-term memory. Neuron. 2018;100(5):1180-1193.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Watson LA, Wang X, Elbert A, Kernohan KD, Galjart N, Berube NG. dual effect of CTCF loss on neuroprogenitor differentiation and survival. J Neurosci. 2014;34(8):2860–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mossink B, Negwer M, Schubert D, Nadif Kasri N. The emerging role of chromatin remodelers in neurodevelopmental disorders: a developmental perspective. Cell Mol Life Sci. 2021;78(6):2517–63.

    Article  CAS  PubMed  Google Scholar 

  17. Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, Plath K, Jaenisch R, Wagschal A, Feil R, Schreiber SL, Lander ES. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell. 2006;125(2):315–26.

    Article  CAS  PubMed  Google Scholar 

  18. Shah K, King GD, Jiang H. A chromatin modulator sustains self-renewal and enables differentiation of postnatal neural stem and progenitor cells. J Mol Cell Biol. 2020;12(1):4–16.

    Article  CAS  PubMed  Google Scholar 

  19. Pereira JD, Sansom SN, Smith J, Dobenecker M, Tarakhovsky A, Livesey FJ, Crabtree GR. Ezh2, the histone methyltransferase of PRC2, regulates the balance between self-renewal and differentiation in the cerebral cortex. Proc Natl Acad Sci. 2010;107(36):15957–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hwang WW, Salinas RD, Siu JJ, Kelley KW, Delgado RN, Paredes MF, Alvarez-Buylla A, Oldham MC, Lim DA. Distinct and separable roles for EZH2 in neurogenic astroglia. eLife. 2014;3:02439.

    Article  Google Scholar 

  21. Qiao Y, Wang R, Yang X, Tang K, Jing N. Dual roles of histone H3 lysine 9 acetylation in human embryonic stem cell pluripotency and neural differentiation. J Biol Chem. 2015;290(4):2508–20.

    Article  CAS  PubMed  Google Scholar 

  22. Nothof SA, Magdinier F, Van-Gils J. Chromatin structure and dynamics: focus on neuronal differentiation and pathological implication. Genes (Basel). 2022;13(4):639. https://doi.org/10.3390/genes13040639.

    Article  CAS  PubMed  Google Scholar 

  23. Fahrner JA, Bjornsson HT. Mendelian disorders of the epigenetic machinery: postnatal malleability and therapeutic prospects. Hum Mol Genet. 2019;28(R2):R254–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Biswas S, Rao CM. Epigenetic tools (The Writers, The Readers and The Erasers) and their implications in cancer therapy. Eur J Pharmacol. 2018;837:8–24.

    Article  CAS  PubMed  Google Scholar 

  25. Yang AY, Kim H, Li W, Kong AT. Natural compound-derived epigenetic regulators targeting epigenetic readers, writers and erasers. Curr Top Med Chem. 2016;16(7):697–713.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Giles KA, Gould CM, Du Q, Skvortsova K, Song JZ, Maddugoda MP, Achinger-Kawecka J, Stirzaker C, Clark SJ, Taberlay PC. Integrated epigenomic analysis stratifies chromatin remodellers into distinct functional groups. Epigenetics Chromatin. 2019;12(1):12.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Boukas L, Havrilla JM, Hickey PF, Quinlan AR, Bjornsson HT, Hansen KD. Coexpression patterns define epigenetic regulators associated with neurological dysfunction. Genome Res. 2019;29(4):532–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lek M, Karczewski KJ, Minikel EV, Samocha KE, Posthuma D. Exome aggregation consortium [: analysis of protein-coding genetic variation in 60,706 humans. Nature (London). 2016;536(7616):285–91.

    Article  CAS  PubMed  Google Scholar 

  29. Bjornsson HT. The Mendelian disorders of the epigenetic machinery. Genome Res. 2015;25(10):1473–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Jimenez-Sanchez G, Childs B, Valle D. Human disease genes. Nature. 2001;409(6822):853–5.

    Article  CAS  PubMed  Google Scholar 

  31. Larizza L, Finelli P. Developmental disorders with intellectual disability driven by chromatin dysregulation: clinical overlaps and molecular mechanisms. Clin Genet. 2019;95(2):231–40.

    Article  CAS  PubMed  Google Scholar 

  32. Luperchio TR, Boukas L, Zhang L, Pilarowski G, Jiang J, Kalinousky A, Hansen KD, Bjornsson HT. Leveraging the Mendelian disorders of the epigenetic machinery to systematically map functional epigenetic variation. Elife. 2021;10:e65884. https://doi.org/10.7554/eLife.65884.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tatton-Brown K, Seal S, Ruark E, Harmer J, Ramsay E, Del Vecchio Duarte S, Zachariou A, Hanks S, O’Brien E, Aksglaede L, Baralle D, Dabir T, Gener B, Goudie D, Homfray T, Kumar A, Pilz DT, Selicorni A, Temple IK, Van Maldergem L, Yachelevich N, van Montfort R, Rahman N. Mutations in the DNA methyltransferase gene DNMT3A cause an overgrowth syndrome with intellectual disability. Nat Genet. 2014;46(4):385–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Tatton-Brown K, Zachariou A, Loveday C, Renwick A, Mahamdallie S, Aksglaede L, Baralle D, Barge-Schaapveld D, Blyth M, Bouma M, Breckpot J, Crabb B, Dabir T, Cormier-Daire V, Fauth C, Fisher R, Gener B, Goudie D, Homfray T, Hunter M, Jorgensen A, Kant SG, Kirally-Borri C, Koolen D, Kumar A, Labilloy A, Lees M, Marcelis C, Mercer C, Mignot C, Miller K, Neas K, Newbury-Ecob R, Pilz DT, Posmyk R, Prada C, Ramsey K, Randolph LM, Selicorni A, Shears D, Suri M, Temple IK, Turnpenny P, Val Maldergem L, Varghese V, Veenstra-Knol HE, Yachelevich N, Yates L, Rahman N. The Tatton-Brown-Rahman syndrome: a clinical study of 55 individuals with de novo constitutive DNMT3A variants. Wellcome Open Res. 2018;3:46.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Lane C, Tatton-Brown K, Freeth M. Tatton-Brown-Rahman syndrome: cognitive and behavioural phenotypes. Dev Med Child Neurol. 2020;62(8):993–8.

    Article  PubMed  Google Scholar 

  36. Tenorio J, Alarcón P, Arias P, Dapía I, García-Miñaur S, Palomares Bralo M, Campistol J, Climent S, Valenzuela I, Ramos S, Monseny AM, Grondona FL, Botet J, Serrano M, Solís M, Santos-Simarro F, Álvarez S, Teixidó-Tura G, Fernández Jaén A, Gordo G, Bardón Rivera MB, Nevado J, Hernández A, Cigudosa JC, Ruiz-Pérez VL, Tizzano EF, Lapunzina P. Further delineation of neuropsychiatric findings in Tatton-Brown-Rahman syndrome due to disease-causing variants in DNMT3A: seven new patients. Eur J Human Genet. 2020;28(4):469–79.

    Article  CAS  Google Scholar 

  37. Balci TB, Strong A, Kalish JM, Zackai E, Maris JM, Reilly A, Surrey LF, Wertheim GB, Marcadier JL, Graham GE, Carter MT. Tatton-Brown-Rahman syndrome: six individuals with novel features. Am J Med Genet A. 2020;182(4):673–80.

    Article  CAS  PubMed  Google Scholar 

  38. Christian DL, Wu DY, Martin JR, Moore JR, Liu YR, Clemens AW, Nettles SA, Kirkland NM, Papouin T, Hill CA, Wozniak DF, Dougherty JD, Gabel HW. DNMT3A haploinsufficiency results in behavioral deficits and global epigenomic dysregulation shared across neurodevelopmental disorders. Cell Rep (Cambridge). 2020;33(8):108416.

    Article  CAS  Google Scholar 

  39. Beck DB, Petracovici A, He C, Moore HW, Louie RJ, Ansar M, Douzgou S, Sithambaram S, Cottrell T, Santos-Cortez RLP, Prijoles EJ, Bend R, Keren B, Mignot C, Nougues M, Õunap K, Reimand T, Pajusalu S, Zahid M, Saqib MAN, Buratti J, Seaby EG, McWalter K, Telegrafi A, Baldridge D, Shinawi M, Leal SM, Schaefer GB, Stevenson RE, Banka S, Bonasio R, Fahrner JA. Delineation of a human Mendelian disorder of the DNA demethylation machinery: TET3 deficiency. Am J Hum Genet. 2020;106(2):234–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Antunes C, Da Silva JD, Guerra-Gomes S, Alves ND, Ferreira F, Loureiro-Campos E, Branco MR, Sousa N, Reik W, Pinto L, Marques CJ. Tet3 ablation in adult brain neurons increases anxiety-like behavior and regulates cognitive function in mice. Mol Psychiatry. 2021;26(5):1445–57.

    Article  CAS  PubMed  Google Scholar 

  41. Antunes C, Da Silva JD, Guerra-Gomes S, Alves ND, Loureiro-Campos E, Pinto L, Marques CJ. Tet3 deletion in adult brain neurons of female mice results in anxiety-like behavior and cognitive impairments. Mol Neurobiol. 2022;59(8):4892–901.

    Article  CAS  PubMed  Google Scholar 

  42. Fan B, Hao B, Dai Y, Xue L, Shi Y, Liu L, Xuan S, Yang N, Wang X, Zhao H. Deficiency of Tet3 in nucleus accumbens enhances fear generalization and anxiety-like behaviors in mice. Brain Pathol (Zurich, Switzerland). 2022;32(6):e13080-n/a.

    Article  CAS  Google Scholar 

  43. Kremer EA, Gaur N, Lee MA, Engmann O, Bohacek J, Mansuy IM. Interplay between TETs and microRNAs in the adult brain for memory formation. Sci Rep. 2018;8(1):1678–710.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Janssen N, Bergman JEH, Swertz MA, Tranebjaerg L, Lodahl M, Schoots J, Hofstra RMW, van Ravenswaaij-Arts CMA, Hoefsloot LH. Mutation update on the CHD7 gene involved in CHARGE syndrome. Hum Mutat. 2012;33(8):1149–60.

    Article  CAS  PubMed  Google Scholar 

  45. Vissers LELM, Ravenswaaij-Arts CMAv, Admiraal RJC, Hurst JA, Vries LBAd, Janssen IM, Vliet WAvd, Huys E, Jong PJd, Hamel BCJ, Schoenmakers EFPM, Brunner HG, Veltman JA, Geurts van Kessel A. H. M. Mutations in a new member of the chromodomain gene family cause CHARGE syndrome. Nat Genet. 2004;36(9):955–7.

    Article  CAS  PubMed  Google Scholar 

  46. Pagon RA, Graham JM, Zonana J, Yong S. Coloboma, congenital heart disease, and choanal atresia with multiple anomalies: CHARGE association. J Pediatr. 1981;99(2):223–7.

    Article  CAS  PubMed  Google Scholar 

  47. Lasserre E, Vaivre-Douret L, Abadie V. Psychomotor and cognitive impairments of children with CHARGE syndrome: common and variable features. Child Neuropsychol. 2013;19(5):449–65.

    Article  PubMed  Google Scholar 

  48. Vesseur A, Langereis M, Free R, Snik A, van Ravenswaaij-Arts C, Mylanus E. Influence of hearing loss and cognitive abilities on language development in CHARGE Syndrome. Am J Med Genet A. 2016;170(8):2022–30.

    Article  PubMed  Google Scholar 

  49. Raqbi F, Le Bihan C, Morisseau-Durand M, Dureau P, Lyonnet S, Abadie V. Early prognostic factors for intellectual outcome in CHARGE syndrome. Dev Med Child Neurol. 2003;45(7):483–8.

    Article  CAS  PubMed  Google Scholar 

  50. Wachtel LE, Hartshorne TS, Dailor AN. Psychiatric diagnoses and psychotropic medications in CHARGE syndrome: a pediatric survey. J Dev Phys Disabil. 2007;19(5):471–83.

    Article  Google Scholar 

  51. Blake KD, Salem-Hartshorne N, Daoud MA, Gradstein J. Adolescent and adult issues in charge syndrome. Clin Pediatr. 2005;44(2):151–9.

    Article  Google Scholar 

  52. Hartshorne N, Hudson A, MacCuspie J, Kennert B, Nacarato T, Hartshorne T, Blake K. Quality of life in adolescents and adults with CHARGE syndrome. Am J Med Genet A. 2016;170A(8):2012–21.

    Article  Google Scholar 

  53. Hartshorne TS, Grialou TL, Parker KR. Autistic-like behavior in CHARGE syndrome. Am J Med Genet A. 2005;133A(3):257–61.

    Article  PubMed  Google Scholar 

  54. Souriau J, Gimenes M, Blouin C, Benbrik I, Benbrik E, Churakowskyi A, Churakowskyi B. CHARGE syndrome: developmental and behavioral data. Am J Med Genet A. 2005;133A(3):278–81.

    Article  PubMed  Google Scholar 

  55. van Dijk JPM, de Kort A. Reducing challenging behaviors and fostering efficient learning of children with CHARGE syndrome. Am J Med Genet A. 2005;133A(3):273–7.

    Article  PubMed  Google Scholar 

  56. Thelin JW, Fussner JC. Factors related to the development of communication in CHARGE syndrome. Am J Med Genet A. 2005;133A(3):282–90.

    Article  PubMed  Google Scholar 

  57. Graham JM Jr, Rosner B, Dykens E, Visootsak J. Behavioral features of CHARGE syndrome (Hall-Hittner syndrome) comparison with Down syndrome, Prader-Willi syndrome, and Williams syndrome. Am J Med Genet A. 2005;133A(3):240–7.

    Article  PubMed  Google Scholar 

  58. Fergelot P, Van Belzen M, Van Gils J, Afenjar A, Armour CM, Arveiler B, Beets L, Burglen L, Busa T, Collet M, Deforges J, de Vries BBA, Dominguez Garrido E, Dorison N, Dupont J, Francannet C, Garciá-Minaúr S, Gabau Vila E, Gebre-Medhin S, Gener Querol B, Geneviève D, Gérard M, Gervasini CG, Goldenberg A, Josifova D, Lachlan K, Maas S, Maranda B, Moilanen JS, Nordgren A, Parent P, Rankin J, Reardon W, Rio M, Roume J, Shaw A, Smigiel R, Sojo A, Solomon B, Stembalska A, Stumpel C, Suarez F, Terhal P, Thomas S, Touraine R, Verloes A, Vincent-Delorme C, Wincent J, Peters DJM, Bartsch O, Larizza L, Lacombe D, Hennekam RC. Phenotype and genotype in 52 patients with Rubinstein-Taybi syndrome caused by EP300 mutations. Am J Med Genet A. 2016;170(12):3069–82.

    Article  CAS  PubMed  Google Scholar 

  59. López M, García-Oguiza A, Armstrong J, García-Cobaleda I, García-Miñaur S, Santos-Simarro F, Seidel V, Domínguez-Garrido E. Rubinstein-Taybi 2 associated to novel EP300 mutations: deepening the clinical and genetic spectrum. BMC Med Genet. 2018;19(1):36.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Adrien J, Taupiac E, Thiébaut E, Paulais M, Van-Gils J, Kaye K, Blanc R, Gattegno MP, Contejean Y, Michel G, Dean A, Barthélémy C, Lacombe D. A comparative study of cognitive and socio-emotional development in children with Rubinstein-Taybi syndrome and children with autism spectrum disorder associated with a severe intellectual disability, and in young typically developing children with matched developmental ages. Res Dev Disabil. 2021;116:104029.

    Article  PubMed  Google Scholar 

  61. Galéra C, Taupiac E, Fraisse S, Naudion S, Toussaint E, Rooryck-Thambo C, Delrue M, Arveiler B, Lacombe D, Bouvard M. Socio-behavioral characteristics of children with Rubinstein-Taybi syndrome. J Autism Dev Disord. 2009;39(9):1252–60.

    Article  PubMed  Google Scholar 

  62. Taupiac E, Lacombe D, Thiébaut E, Van-Gils J, Michel G, Fergelot P, Adrien J. Psychomotor, cognitive, and socio-emotional developmental profiles of children with Rubinstein-Taybi syndrome and a severe intellectual disability. J Intellect Dev Disabil. 2021;46(1):80–9.

    Article  Google Scholar 

  63. Waite J, Beck SR, Heald M, Powis L, Oliver C. Dissociation of cross-sectional trajectories for verbal and visuo-spatial working memory development in Rubinstein-Taybi syndrome. J Autism Dev Disord. 2016;46(6):2064–71.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Ajmone PF, Avignone S, Gervasini C, Giacobbe A, Monti F, Costantino A, Esposito S, Marchisio P, Triulzi F, Milani D. Rubinstein-Taybi syndrome: new neuroradiological and neuropsychiatric insights from a multidisciplinary approach. Am J Med Genet Part B Neuropsychiatr Genet. 2018;177(4):406–15.

    Article  Google Scholar 

  65. Crawford H, Waite J, Oliver C. Diverse profiles of anxiety related disorders in fragile X, Cornelia de Lange and Rubinstein-Taybi syndromes. J Autism Dev Disord. 2017;47(12):3728–40.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Awan N, Pearson E, Shelley L, Greenhill C, Tarver J, Waite J. The behavioral phenotype of Rubinstein-Taybi syndrome: a scoping review of the literature. Am J Med Genet A. 2022;188(9):2536–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kennedy J, Goudie D, Blair E, Chandler K, Joss S, McKay V, Green A, Armstrong R, Lees M, Kamien B, Hopper B, Tan TY, Yap P, Stark Z, Okamoto N, Miyake N, Matsumoto N, Macnamara E, Murphy JL, McCormick E, Hakonarson H, Falk MJ, Li D, Blackburn P, Klee E, Babovic-Vuksanovic D, Schelley S, Hudgins L, Kant S, Isidor B, Cogne B, Bradbury K, Williams M, Patel C, Heussler H, Duff-Farrier C, Lakeman P, Scurr I, Kini U, Elting M, Reijnders M, Schuurs-Hoeijmakers J, Wafik M, Blomhoff A, Ruivenkamp CAL, Nibbeling E, Dingemans AJM, Douine ED, Nelson SF, Arboleda VA, Newbury-Ecob R. KAT6A syndrome: genotype–phenotype correlation in 76 patients with pathogenic KAT6A variants. Genet Med. 2019;21(4):850–60.

    Article  CAS  PubMed  Google Scholar 

  68. St John M, Amor DJ, Morgan AT. Speech and language development and genotype–phenotype correlation in 49 individuals with KAT6A syndrome. Am J Med Genet A. 2022;188(12):3389–400.

    Article  CAS  PubMed  Google Scholar 

  69. Smith C, Harris J. Sleep, behavior, and adaptive function in KAT6A syndrome. Brain Sci. 2021;11(8):966.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Boniel S, Szymańska K, Śmigiel R, Szczałuba K. Kabuki syndrome-clinical review with molecular aspects. Genes. 2021;12(4):468.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Jones W, Dafou D, McEntagart M, Woollard W, Elmslie F, Holder-Espinasse M, Irving M, Saggar A, Smithson S, Trembath R, Deshpande C, Simpson M. De novo mutations in MLL cause Wiedemann-Steiner syndrome. Am J Hum Genet. 2012;91(2):358–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Bogershausen N, Wollnik B. Unmasking Kabuki syndrome. Clin Genet. 2013;83(3):201–11.

    Article  CAS  PubMed  Google Scholar 

  73. Niikawa N, Matsuura N, Fukushima Y, Ohsawa T, Kajii T. Kabuki make-up syndrome: a syndrome of mental retardation, unusual facies, large and protruding ears, and postnatal growth deficiency. J Pediatr. 1981;99(4):565–9.

    Article  CAS  PubMed  Google Scholar 

  74. Lehman N, Mazery AC, Visier A, Baumann C, Lachesnais D, Capri Y, Toutain A, Odent S, Mikaty M, Goizet C, Taupiac E, Jacquemont ML, Sanchez E, Schaefer E, Gatinois V, Faivre L, Minot D, Kayirangwa H, Sang K-LQ, Boddaert N, Bayard S, Lacombe D, Moutton S, Touitou I, Rio M, Amiel J, Lyonnet S, Sanlaville D, Picot MC, Geneviève D. Molecular, clinical and neuropsychological study in 31 patients with Kabuki syndrome and KMT2D mutations. Clin Genet. 2017;92(3):298–305.

    Article  CAS  PubMed  Google Scholar 

  75. Sheppard SE, Campbell IM, Harr MH, Gold N, Li D, Bjornsson HT, Cohen JS, Fahrner JA, Fatemi A, Harris JR, Nowak C, Stevens CA, Grand K, Au M, Graham JM, Sanchez-Lara PA, Campo MD, Jones MC, Abdul-Rahman O, Bassetti JA, Bergstrom K, Bhoj E, Dugan S, Kaplan JD, Derar N, Gripp KW, Hauser N, Innes AM, Keena B, Kodra N, Miller R, Nelson B, Nowaczyk MJ, Rahbeeni Z, Ben-Shachar S, Shieh JT, Slavotinek A, Sobering AK, Abbott M, Allain DC, Amlie-Wolf L, Au PYB, Bedoukian E, Beek G, Barry J, Berg J, Bernstein JA, Cytrynbaum C, Chung BH, Donoghue S, Dorrani N, Eaton A, Flores-Daboub JA, Dubbs H, Felix CA, Fong C, Fung JLF, Gangaram B, Goldstein A, Greenberg R, Ha TK, Hersh J, Izumi K, Kravets E, Kwok P, Jobling RK, Knight Johnson AE, Kushner J, Lee BH, Levin B, Lindstrom K, Manickam K, Mardach R, McCormick E, McLeod DR, Mentch FD, Minks K, Muraresku C, Nelson SF, Porazzi P, Pichurin PN, Powell-Hamilton NN, Powis Z, Ritter A, Rogers C, Rohena L, Ronspies C, Schroeder A, Stark Z, Starr L, Stoler J, Suwannarat P, Velinov M, Weksberg R, Wilnai Y, Zadeh N, Falk MJ, Hakonarson H, Zackai EH, Quintero-Rivera F. Expanding the genotypic and phenotypic spectrum in a diverse cohort of 104 individuals with Wiedemann-Steiner syndrome. Am J Med Genet A. 2021;185(6):1649–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Baer S, Afenjar A, Smol T, Piton A, Gérard B, Alembik Y, Bienvenu T, Boursier G, Boute O, Colson C, Cordier M, Cormier-Daire V, Delobel B, Doco-Fenzy M, Duban-Bedu B, Fradin M, Geneviève D, Goldenberg A, Grelet M, Haye D, Heron D, Isidor B, Keren B, Lacombe D, Lèbre A, Lesca G, Masurel A, Mathieu-Dramard M, Nava C, Pasquier L, Petit A, Philip N, Piard J, Rondeau S, Saugier-Veber P, Sukno S, Thevenon J, Van-Gils J, Vincent-Delorme C, Willems M, Schaefer E, Morin G. Wiedemann-Steiner syndrome as a major cause of syndromic intellectual disability: a study of 33 French cases. Clin Genet. 2018;94(1):141–52.

    Article  CAS  PubMed  Google Scholar 

  77. Harris J, Mahone EM, Bjornsson HT. Molecularly confirmed Kabuki (Niikawa-Kuroki) syndrome patients demonstrate a specific cognitive profile with extensive visuospatial abnormalities. J Intellect Disabil Res. 2019;63(6):489–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Ng R, Harris J, Fahrner JA, Bjornsson HT. Individuals with Wiedemann-Steiner syndrome show nonverbal reasoning and visuospatial defects with relative verbal skill sparing. J Int Neuropsychol Soc. 2022;29(5):1–7.

  79. Dongen LCMv, Wingbermühle PAM, Veld WMvd, Stumpel CTRM, Kleefstra T, Egger JIM. Exploring the cognitive phenotype of Kabuki (NiikawaKuroki) syndrome. J Intellect Disabil Res. 2019;63(6):498–506.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Mervis CB, Becerra AM, Rowe ML, Hersh JH, Morris CA. Intellectual abilities and adaptive behavior of children and adolescents with Kabuki syndrome: a preliminary study. Am J Med Genet A. 2005;132A(3):248–55.

    Article  PubMed  Google Scholar 

  81. Kalinousky AJ, Rapp T, Hijazi H, Johnson J, Bjornsson HT, Harris JR. Neurobehavioral phenotype of Kabuki syndrome: anxiety is a common feature. Front Genet. 2022;13:1007046.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Theodore-Oklota C, Hartman DS, Hoffman DL, Björnsson HT. A qualitative study to characterize the humanistic burden of Kabuki syndrome in the United States and Canada. Adv Ther. 2021;39(1):619–31.

    Article  PubMed  PubMed Central  Google Scholar 

  83. White SM, Thompson EM, Kidd A, Savarirayan R, Turner A, Amor D, Delatycki MB, Fahey M, Baxendale A, White S, Haan E, Gibson K, Halliday JL, Bankier A. Growth, behavior, and clinical findings in 27 patients with Kabuki (Niikawa-Kuroki) syndrome. Am J Med Genet A. 2004;127A(2):118–27.

    Article  CAS  PubMed  Google Scholar 

  84. Morgan AT, Mei C, Da Costa A, Fifer J, Lederer D, Benoit V, McMillin MJ, Buckingham KJ, Bamshad MJ, Pope K, White SM. Speech and language in a genotyped cohort of individuals with Kabuki syndrome. Am J Med Genet A. 2015;167(7):1483–92.

    Article  CAS  PubMed  Google Scholar 

  85. Durand B, Schaefer E, Burger P, Baer S, Schroder C, Mandel J, Piton A, Coutelle R. Neurocognitive and neurobehavioral characterization of two frequent forms of neurodevelopmental disorders: the DYRK1A and the Wiedemann-Steiner syndromes. Clin Genet. 2022;102(4):296–304.

    Article  CAS  PubMed  Google Scholar 

  86. Ng R, Bjornsson HT, Fahrner JA, Harris J. Sleep disturbances correlate with behavioral problems among individuals with Wiedemann-Steiner syndrome. Front Genet. 2022;13:950082.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Ng R, Bjornsson HT, Fahrner JA, Harris J. Anxiety in wiedemann-steiner syndrome. Am J Med Genet A. 2022;191(2):437–44.

  88. Chan AJS, Cytrynbaum C, Hoang N, Ambrozewicz PM, Weksberg R, Drmic I, Ritzema A, Schachar R, Walker S, Uddin M, Zarrei M, Yuen RKC, Scherer SW. Expanding the neurodevelopmental phenotypes of individuals with de novo KMT2A variants. NPJ Genom Med. 2019;4(1):9.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Ng R, Bjornsson HT, Fahrner JA, Harris J. High prosociality and social drive among individuals with Wiedemann-Steiner Syndrome. Minneapolis: American Academy of Clinical Neuropsychology Annual Conference; 2022.

  90. Aref-Eshghi E, Kerkhof J, Pedro VP, Groupe DI France, Barat-Houari M, Ruiz-Pallares N, Andrau J, Lacombe D, Van-Gils J, Fergelot P, Dubourg C, Cormier-Daire V, Rondeau S, Lecoquierre F, Saugier-Veber P, Nicolas G, Lesca G, Chatron N, Sanlaville D, Vitobello A, Faivre L, Thauvin-Robinet C, Laumonnier F, Raynaud M, Alders M, Mannens M, Henneman P, Hennekam RC, Velasco G, Francastel C, Ulveling D, Ciolfi A, Pizzi S, Tartaglia M, Heide S, Héron D, Mignot C, Keren B, Whalen S, Afenjar A, Bienvenu T, Campeau PM, Rousseau J, Levy MA, Brick L, Kozenko M, Balci TB, Siu VM, Stuart A, Kadour M, Masters J, Takano K, Kleefstra T, de Leeuw N, Field M, Shaw M, Gecz J, Ainsworth PJ, Lin H, Rodenhiser DI, Friez MJ, Tedder M, Lee JA, DuPont BR, Stevenson RE, Skinner SA, Schwartz CE, Genevieve D, Sadikovic B. Evaluation of DNA methylation episignatures for diagnosis and phenotype correlations in 42 Mendelian neurodevelopmental disorders. Am J Hum Genet. 2020;106(3):356–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Montano C, Britton JF, Harris JR, Kerkhof J, Barnes BT, Lee JA, Sadikovic B, Sobreira N, Fahrner JA. Genome-wide DNA methylation profiling confirms a case of low-level mosaic Kabuki syndrome 1. Am J Med Genet Part A. 2022;188(7):2217–25.

    Article  CAS  PubMed  Google Scholar 

  92. Levy MA, Relator R, McConkey H, Pranckeviciene E, Kerkhof J, Barat-Houari M, Bargiacchi S, Biamino E, Palomares Bralo M, Cappuccio G, Ciolfi A, Clarke A, DuPont BR, Elting MW, Faivre L, Fee T, Ferilli M, Fletcher RS, Cherick F, Foroutan A, Friez MJ, Gervasini C, Haghshenas S, Hilton BA, Jenkins Z, Kaur S, Lewis S, Louie RJ, Maitz S, Milani D, Morgan AT, Oegema R, Østergaard E, Pallares NR, Piccione M, Plomp AS, Poulton C, Reilly J, Rius R, Robertson S, Rooney K, Rousseau J, Santen GWE, Santos-Simarro F, Schijns J, Squeo GM, John MS, Thauvin-Robinet C, Traficante G, van der Sluijs PJ, Vergano SA, Vos N, Walden KK, Azmanov D, Balci TB, Banka S, Gecz J, Henneman P, Lee JA, Mannens MMAM, Roscioli T, Siu V, Amor DJ, Baynam G, Bend EG, Boycott K, Brunetti-Pierri N, Campeau PM, Campion D, Christodoulou J, Dyment D, Esber N, Fahrner JA, Fleming MD, Genevieve D, Heron D, Husson T, Kernohan KD, McNeill A, Menke LA, Merla G, Prontera P, Rockman-Greenberg C, Schwartz C, Skinner SA, Stevenson RE, Vincent M, Vitobello A, Tartaglia M, Alders M, Tedder ML, Sadikovic B. Functional correlation of genome-wide DNA methylation profiles in genetic neurodevelopmental disorders. Hum Mutat. 2022;43(11):1609–28.

    Article  CAS  PubMed  Google Scholar 

  93. Conboy K, Henshall DC, Brennan GP. Epigenetic principles underlying epileptogenesis and epilepsy syndromes. Neurobiol Dis. 2021;148:105179.

    Article  CAS  PubMed  Google Scholar 

  94. Rajarajan P, Jiang Y, Kassim BS, Akbarian S. Chromosomal conformations and epigenomic regulation in schizophrenia. Prog Mol Biol Transl Sci. 2018;157:21–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Siu MT, Weksberg R. Epigenetics of autism spectrum disorder. Adv Exp Med Biol. 2017;978:63–90.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the patient groups for their continued support and involvement with all aspects of our work. The authors would like to acknowledge the support of the entire Epigenetics research and clinical teams at Johns Hopkins and Kennedy Krieger Institute, without whom none of our work would be possible.

Funding

No funding was used specifically for the writing of this article and no funding body had any role in it. RN receives funding from the Wiedemann-Steiner Syndrome Foundation and KAT6 Foundation. JH receives funding from NIH/NICHD 1K23HD101646, the Kabuki Syndrome Foundation, the Rubinstein-Taybi Syndrome Children’s Foundation, and the Sekel-Breidenstein Family Fund. RN and JH receive support from the Kennedy Krieger/Johns Hopkins IDDRC NIH P50HD103538 and ICTR (UL1 TR003098).

Author information

Authors and Affiliations

Authors

Contributions

RN, AK, and JH all conceived of the manuscript, wrote, and edited the manuscript. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Jacqueline Harris.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

JH receives research funding for Kabuki syndrome from Oryzon Genomics.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ng, R., Kalinousky, A. & Harris, J. Epigenetics of cognition and behavior: insights from Mendelian disorders of epigenetic machinery. J Neurodevelop Disord 15, 16 (2023). https://doi.org/10.1186/s11689-023-09482-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s11689-023-09482-0

Keywords