The covalent modification of histones is critical for many biological functions in mammals, including gene regulation and chromatin structure. Posttranslational histone modifications are added and removed by specialised ‘writer’ and ‘eraser’ enzymes, respectively. One such writer protein implicated in a wide range of cellular processes is SET domain-containing 2 (SETD2), a histone methyltransferase that catalyses the trimethylation of lysine 36 on histone H3 (H3K36me3). Recently, SETD2 has also been found to modify proteins other than histones, including actin and tubulin. The emerging roles of SETD2 in the development and function of the mammalian central nervous system (CNS) are of particular interest as several SETD2 variants have been implicated in neurodevelopmental disorders, such as autism spectrum disorder and the overgrowth disorder Luscan–Lumish syndrome. Here, we summarise the numerous roles of SETD2 in mammalian cellular functions and development, with a focus on the CNS. We also provide an overview of the consequences of SETD2 variants in human disease and discuss future directions for understanding essential cellular functions of SETD2.

In eukaryotes, genomic DNA is compacted into chromatin by a group of proteins known as histones. These proteins assemble into octamers consisting of pairs of four core canonical histones: H2A, H2B, H3 and H4. There are several variants of each of these histones. For example, mammals have three main H3 variants, the canonical H3.1 and H3.2, and the variant H3.3, which is strongly associated with active genes (for a comprehensive review see Talbert and Henikoff, 2021). DNA wraps ∼1.65 turns around core histone octamers to form nucleosomes, the basic repeating units of chromatin (Luger et al., 1997). Enzymes known as ‘writers’ and ‘erasers’ catalyse the addition and removal, respectively, of posttranslational modifications (PTMs; e.g. methylation, acetylation and phosphorylation) to specific residues on histones, particularly those located in the N-terminal histone tail (Berger, 2002; Biswas and Rao, 2018). Histone modification plays a key role in the regulation of gene expression in eukaryotes. Modifications can influence physical interactions between histones, modifying chromatin structure and therefore accessibility of DNA to the transcriptional machinery. They can also indirectly regulate gene expression through the recruitment of gene-regulatory ‘reader’ proteins. These proteins contain specialised domains that can bind to histone modifications to facilitate a wide range of downstream cellular effects. For a more in-depth overview of histone modifications, we refer readers to a recent review by Ramazi et al. (2020).

Histone modifications are critical for proper central nervous system (CNS) function and development (Park et al., 2022). These include trimethylation of lysine 27 on histone H3 (H3K27me3), which is associated with transcriptional repression, and the acetylation of lysine 14 on histone H3 (H3K14ac), which is associated with active transcription (for an in-depth review see Park et al., 2022). Here, we focus on the trimethylation of lysine 36 on histone H3 (H3K36me3), as recent reports have revealed that this mark plays a key role in regulating diverse cellular processes and is dysregulated in some developmental disorders and a range of cancers. H3K36me3 influences gene expression via several mechanisms, including transcriptional regulation and mRNA processing; for an in-depth review see Sharda and Humphrey (2022). The writer protein responsible for this mark was first identified in Saccharomyces cerevisiae as the Su(var)3-9, Enhancer of Zeste and Trithorax (SET) domain-containing protein Set2 (Strahl et al., 2002). Set2 catalyses the mono-, di- and tri-methylation of lysine 36 on histone H3 (H3K36) (Venkatesh and Workman, 2013). The mammalian paralogue of Set2, SET domain-containing 2 (SETD2), also has methyltransferase activity (Sun et al., 2005). However, SETD2 is primarily responsible for only the trimethylation of H3K36 in vivo, as deletion of the protein in mammalian cells leads to a dramatic reduction in H3K36me3, but no changes in H3K36 monomethylation (H3K36me1) or dimethylation (H3K36me2) levels (Edmunds et al., 2008).

The function of SETD2 extends beyond H3K36me3, however, with studies identifying methylation of additional histone residues and a range of non-histone substrates for the protein (Fig. 1). Through H3K36me3 and these non-histone interactions, SETD2 is implicated in several different cellular processes. Many of these processes are critical for the development and function of the CNS. This is particularly evident in humans, where pathogenic SETD2 variants lead to neurodevelopmental disorders such as Luscan–Lumish syndrome (OMIM 616831). Here, we provide an overview of the cellular functions of SETD2, highlighting recently identified substrates, with a particular focus on its role in CNS. We will also discuss the involvement of SETD2 dysregulation in neurodevelopmental disorders.

Fig. 1.

Overview of SETD2 cellular functions. SETD2 has a wide range of cellular functions, which are carried out through numerous pathways. The protein methylates several histone and non-histone substrates. SETD2 ‘writes’ the histone modifications H3K36me3, H3K14me3 and possibly H3K37me1, which have various effects on chromatin organisation, transcription, replication and mRNA processing. It also interacts with proteins such as p53, hnRNP L, EZH2 and STAT1 to influence gene expression. SETD2 has recently been found to be capable of methylating structural and cytoskeletal proteins including tubulin, actin and fibrillin-1, revealing potential new roles in cell migration and cytoskeletal dynamics. Single-headed arrows indicate methyltransferase activity. Double-headed arrows indicate direct interactions and/or binding. Dashed arrows indicate potential unconfirmed SETD2 functions. Created with BioRender.com.

Fig. 1.

Overview of SETD2 cellular functions. SETD2 has a wide range of cellular functions, which are carried out through numerous pathways. The protein methylates several histone and non-histone substrates. SETD2 ‘writes’ the histone modifications H3K36me3, H3K14me3 and possibly H3K37me1, which have various effects on chromatin organisation, transcription, replication and mRNA processing. It also interacts with proteins such as p53, hnRNP L, EZH2 and STAT1 to influence gene expression. SETD2 has recently been found to be capable of methylating structural and cytoskeletal proteins including tubulin, actin and fibrillin-1, revealing potential new roles in cell migration and cytoskeletal dynamics. Single-headed arrows indicate methyltransferase activity. Double-headed arrows indicate direct interactions and/or binding. Dashed arrows indicate potential unconfirmed SETD2 functions. Created with BioRender.com.

Human SETD2 is a 2564-amino-acid protein that contains several functional domains (Fig. 2). These include a SET domain flanked by an ‘associated with SET’ (AWS) domain and a post-SET domain, which together provide methyltransferase activity (Sun et al., 2005). SETD2 also contains a Set2–Rpb1 interaction (SRI) domain, which interacts with RNA polymerase II (RNAPII) (Li et al., 2005), and a SETD2–hnRNP interaction (SHI) domain and a WW domain, which mediate interactions with heterogeneous nuclear ribonucleoprotein L (hnRNP L) and huntingtin (HTT), respectively (Bhattacharya et al., 2021; Faber et al., 1998). Within the SHI domain is a coiled-coil (CC) domain, which likely participates in interactions with hnRNP L (Bhattacharya et al., 2021). All these domains reside in the C-terminal half of the protein. The N-terminal half of the protein is largely unstructured but might regulate protein stability (Bhattacharya and Workman, 2020).

Fig. 2.

SETD2 structure and documented variants. Schematic showing SETD2 functional domains. Sites corresponding to SETD2 variants identified in individuals with suspected Luscan–Lumish syndrome (blue), Rabin–Pappas syndrome (red) and intellectual development disorder, autosomal dominant 70 (yellow) are indicated. SETD2 variant data were obtained from several published articles (Chen et al., 2021; Lelieveld et al., 2016; Lumish et al., 2015; Luscan et al., 2014; Marzin et al., 2019; O'Roak et al., 2012; Parra et al., 2023; Rabin et al., 2020; Suda et al., 2021; Tlemsani et al., 2016; van Rij et al., 2018; Wu et al., 2023; Zhang et al., 2023). Truncating (square), missense (triangle), splice site (star) and in-frame deletion (circle) SETD2 variants have been identified in these neurodevelopmental disorders. Luscan–Lumish syndrome SETD2 variants are thought to be associated with loss of function and haploinsufficiency, whereas Rabin–Pappas syndrome variants are considered to be gain-of-function variants.

Fig. 2.

SETD2 structure and documented variants. Schematic showing SETD2 functional domains. Sites corresponding to SETD2 variants identified in individuals with suspected Luscan–Lumish syndrome (blue), Rabin–Pappas syndrome (red) and intellectual development disorder, autosomal dominant 70 (yellow) are indicated. SETD2 variant data were obtained from several published articles (Chen et al., 2021; Lelieveld et al., 2016; Lumish et al., 2015; Luscan et al., 2014; Marzin et al., 2019; O'Roak et al., 2012; Parra et al., 2023; Rabin et al., 2020; Suda et al., 2021; Tlemsani et al., 2016; van Rij et al., 2018; Wu et al., 2023; Zhang et al., 2023). Truncating (square), missense (triangle), splice site (star) and in-frame deletion (circle) SETD2 variants have been identified in these neurodevelopmental disorders. Luscan–Lumish syndrome SETD2 variants are thought to be associated with loss of function and haploinsufficiency, whereas Rabin–Pappas syndrome variants are considered to be gain-of-function variants.

Regulation of transcription and splicing by H3K36me3 and SETD2

The most well-characterised function of SETD2 is trimethylation of H3K36, and this modification is chiefly added during transcription. The largest subunit of RNAPII contains a C-terminal repeat domain (CTD) that is hyperphosphorylated during active transcription. SETD2 interacts specifically with this phosphorylated form of the RNAPII CTD through its SRI domain (Li et al., 2005; Sun et al., 2005). This allows SETD2 to selectively associate with actively transcribed regions of the genome. As a result, H3K36me3 is generally deposited at the 3′ end of actively transcribed gene bodies and is associated with euchromatin (Bannister et al., 2005). However, one study on mice has shown that H3K36me3 is also present in heterochromatin, although the mechanisms behind this association are largely unknown (Chantalat et al., 2011). H3K36me3 levels are regulated in several ways (highlighted in Box 1).

Box 1. Regulation of H3K36me3

SETD2 is a major writer protein of H3K36me3 in mammals, depositing the modification primarily at the gene bodies of actively transcribed genes. SETD2 was initially thought to be the sole protein responsible for this process, but a recent study has indicated that SETD5 can also deposit H3K36me3 at active gene bodies in vivo (Sessa et al., 2019). The functional overlap between these two methyltransferases is unclear. It is possible that SETD5 targets different regions of the genome or is more prevalent in specific cell types. Furthermore, the methyltransferase SET and MYND domain-containing 5 (SMYD5) and the zinc finger protein PR/SET domain 9 (PRDM9) have been shown to deposit H3K36me3 at promoter regions and during meiosis, respectively (Powers et al., 2016; Zhang et al., 2022).

H3K36me3 levels in the genome are regulated in several ways, such as demethylation of the mark by the eraser protein KDM4A (Klose et al., 2006). Another method is through the regulation of SETD2 stability. Homeostatic SETD2 protein expression in mammals is low, as it is readily degraded by the ubiquitin–proteasome system (Zhu et al., 2016). The Cullin 3 (CUL3) ubiquitin E3 ligase complex subunit, speckle-type BTB/POZ protein (SPOP) interacts with SETD2, polyubiquitylating it and marking the protein for proteasomal degradation (Zhu et al., 2016). This interaction likely occurs at the N-terminal region of SETD2. Removal of either the N-terminal SETD2 region or SPOP causes an accumulation of SETD2, resulting in an increase in H3K36me3 (Bhattacharya and Workman, 2020; Zhu et al., 2016). SETD2 is also negatively regulated at the transcriptional level by the microRNA miR-106b-5p. Overexpression of miR-106b-5p reduces SETD2 expression (Xiang et al., 2015).

During transcription, DNA partially unwraps itself from nucleosomes, exposing SETD2 target residues on histone H3. Subsequently, the SETD2 AWS domain binds to the N-terminal region of histone H3 (Liu et al., 2021). A regulatory LIN loop between the SET and post-SET domains positions H3K36 in the active site of the SET domain, where methyl groups from the enzyme cofactor S-adenosylmethionine (SAM) are attached to H3K36 (Liu et al., 2021; Yang et al., 2016).

H3K36me3 deposited by SETD2 interacts with a large suite of H3K36me3-binding proteins to participate in numerous cellular processes (Table 1). One such process is de novo DNA methylation at genomic sites enriched with H3K36me3. This is performed by the reader protein DNA methyltransferase 3 beta (DNMT3B) (Baubec et al., 2015; Okano et al., 1999). DNA methylation plays an important role in regulating gene expression, predominantly through transcriptional repression. It also aids in repressing cryptic transcription, which is the inappropriate initiation of transcription from intragenic sites of protein-coding genes (Neri et al., 2017). Cryptic transcription can have deleterious effects on a range of cellular processes (for a review see McCauley and Dang, 2021).

Table 1.

H3K36me3-binding proteins and their cellular functions

H3K36me3-binding proteins and their cellular functions
H3K36me3-binding proteins and their cellular functions

The alternative splicing of pre-mRNA is also regulated by SETD2-mediated H3K36me3. The H3K36me3 reader protein MORF-related gene on chromosome 15 (MRG15, also known as MORF4L1) recruits the splice factor polypyrimidine tract-binding protein (PTB) to regulate the alternative splicing of various genes in mammalian cells (Luco et al., 2010). Another splicing protein, zinc finger MYND domain-containing 11 (ZMYND11), facilitates intron retention in human cells by interacting with U5 small nuclear ribonucleoprotein (snRNP) splice factors, and loss of SETD2 disrupts this process (Guo et al., 2014). ZMYND11 specifically interacts with trimethylated lysine 36 on the histone variant H3.3 (H3.3K36me3), a modification also catalysed by SETD2 (Guo et al., 2014). Notably, SETD2-mediated H3K36me3 might also regulate alternative splicing indirectly by regulating the expression of a splice factor. Indeed, H3K36me3 has been demonstrated in mice to directly regulate alternative splicing of the splice factor gene Srsf11, thus influencing downstream splicing (Xu et al., 2021b). Furthermore, there also appears to be a reciprocal effect of splicing on H3K36me3 deposition. The inhibition of pre-mRNA splicing in mammalian cells causes a reduction in SETD2 recruitment and subsequent H3K36me3 deposition (de Almeida et al., 2011; Kim et al., 2011). Indeed, SETD2 itself directly interacts with splice factors, such as hnRNP L, through its SHI domain, facilitating co-transcriptional splicing and influencing several alternative splicing events (Bhattacharya et al., 2021). The interaction with hnRNP L also regulates methyltransferase activity of SETD2. Deleting the SETD2 SHI domain or hnRNP L leads to a reduction in H3K36me3 (Bhattacharya et al., 2021; Yuan et al., 2009). Further mechanistic studies are required to elucidate how exactly hnRNP L and SETD2 influence each other's activity. Several biological functions are regulated by SETD2-dependent alternative splicing, including autophagosome–lysosome fusion (González-Rodríguez et al., 2022) and the inhibition of intestinal tumorigenesis (Yuan et al., 2017).

Other cellular processes regulated by SETD2-catalysed H3K36me3 include N6-methyladenosine (m6A) mRNA modification (Huang et al., 2019), DNA damage signalling and repair (Carvalho et al., 2014; Chu et al., 2020; Li et al., 2013; Pfister et al., 2014), and prevention of replication stress (Bleuyard et al., 2017; Pfister et al., 2015). These processes are mediated by several H3K36me3-binding proteins (Table 1).

Regulation of cell size

SETD2 is additionally involved in the regulation of cell size. MicroRNA-mediated SETD2 knockdown in human cells causes an increase in cell size and total protein content accompanied by an increased protein synthesis rate in vitro (Molenaar et al., 2022). The mechanisms behind the observed cell size increases are unclear. SETD2 might indirectly regulate cell size by influencing cell cycle dynamics or by directly controlling protein synthesis rates (Molenaar et al., 2022). Further mechanistic studies are necessary to investigate this. It is also unclear whether this process is dependent on the ability of SETD2 to trimethylate H3K36. Overexpression of the H3K36 demethylase lysine demethylase 4A (KDM4A) or of the oncohistones H3.3K36M and H3.3K36A, which deplete H3K36me3, causes an increase in cell volume; however, despite H3.3K36M overexpression almost completely abolishing H3K36me3 expression, it results in a much smaller increase in cell size than that observed following SETD2 knockdown (Molenaar et al., 2022). This suggests that SETD2-mediated regulation of cell size is not entirely dependent on H3K36me3 and highlights that the function of SETD2 is not limited to H3K36me3 deposition.

Regulation of H3K14me3

Intriguingly, SETD2 has recently been identified to catalyse the trimethylation of lysine 14 on histone H3 (H3K14me3). Unlike the well-characterised H3K36me3 modification, which is highly enriched at sites of active transcription, the cellular functions of H3K14me3 and its association with transcription are largely unclear (Zhao et al., 2018). SETD2-deficient HeLa cells have reduced overall H3K14me3 levels, and in vitro methylation assays have demonstrated that SETD2, as well as the histone methyltransferase SUV39H1, is capable of trimethylating lysine 14 on histone H3 (Zhu et al., 2021). SETD2-mediated H3K14me3 levels have been observed to increase in HeLa cells under replication stress. In this context, the H3K14me3 modification has been shown to recruit a group of replication proteins known as the replication protein A (RPA) complex, which in turn activates ataxia telangiectasia related (ATR) protein, facilitating the restart of stalled replication forks and increasing cell survival compared to that of SETD2-deficient cells (Zhu et al., 2021). It remains to be seen whether SETD2 deposits H3K14me3 in vivo. The discovery that SETD2 is capable of methylating more than one histone residue raises key questions. Are there even more histone residues that SETD2 is capable of methylating? Indeed, a recent study has suggested that SETD2 might also monomethylate lysine 37 on histone H3 (H3K37me1). Deletion of SETD2 in human cells causes a reduction in H3K37me1 levels (Santos-Rosa et al., 2021). The histone modification is associated with the prevention of inappropriate replication in yeast, but it is currently unclear if this function is conserved in mammals (Santos-Rosa et al., 2021). What mechanisms govern the choice of histone substrate by SETD2? It is possible that cell signalling pathways activated by replication stress could alter SETD2 activity, potentially through the addition of posttranslational modifications (Zhu et al., 2021). Further mechanistic studies on SETD2-mediated H3K14 trimethylation might address these questions.

Non-histone SETD2 substrates

The methyltransferase activity of SETD2 is not limited to histones, as demonstrated by recent studies that have identified several non-histone substrates that are methylated by SETD2. One such substrate is α-tubulin. Dimers of α- and β-tubulin polymerise to produce microtubules, a critical component of the cellular cytoskeleton. SETD2 trimethylates α-tubulin at lysine 40 (α-TubK40me3), both in vitro (Park et al., 2016) and in vivo (Koenning et al., 2021). Currently, SETD2 is the only protein known to catalyse this modification. Distinct residues within the SRI domain of SETD2 are proposed to bind with the negatively charged C-terminal tail of α-tubulin. SETD2 then catalyses the methylation of α-TubK40 with its SET domain (Kearns et al., 2021). This mark is present on mitotic spindle microtubules during mitosis and cytokinesis, implicating SETD2 in regulation of these processes (Park et al., 2016). Loss of SETD2-mediated α-TubK40me3 in mouse embryonic fibroblasts results in mitotic defects, including failure of chromosome congression during prometaphase, formation of multipolar mitotic spindles, lagging of chromosomes at anaphase, and formation of chromosomal bridges and micronuclei at cytokinesis (Park et al., 2016).

SETD2 can methylate another cytoskeletal component: actin. Specifically, SETD2 performs trimethylation of lysine 68 on actin (ActK68me3) in mammalian cells (Seervai et al., 2020). The interaction between SETD2 and actin is mediated by the WW domain of SETD2, which binds the scaffolding protein HTT. HTT binds the adapter protein huntingtin-interacting protein 1 related (HIP1R), which binds to actin. This axis allows SETD2 to associate with actin and methylate it with its catalytic SET domain (Seervai et al., 2020). ActK68me3 has been observed to localise primarily to insoluble polymerised filamentous (F)-actin, and deletion of Setd2 in mouse embryonic fibroblasts results in a reduction in the ratio of F-actin to monomeric globular (G)-actin. These results suggest that SETD2-mediated ActK68me3 plays a role in actin polymerisation (Seervai et al., 2020). SETD2-deficient cells are observed to have cell migration defects, which could be attributed to the loss of ActK68me3 disrupting actin polymerisation (Seervai et al., 2020). However, the mechanisms underlying how ActK68me3 regulates actin dynamics remain unclear.

SETD2 also directly monomethylates lysine 525 on the transcription factor signal transducer and activator of transcription 1 (STAT1), via its catalytic SET domain (Chen et al., 2017). This is in contrast to other SETD2 substrates, which are primarily trimethylated, indicating that there is some level of flexibility in SETD2 methyltransferase activity. SETD2-mediated STAT1 methylation enhances the interferon α (IFNα)-mediated antiviral immune response in mammalian cells by promoting STAT1 phosphorylation by Janus kinase 1 (JAK1), a protein activated by IFNα signalling (Chen et al., 2017). Phosphorylated STAT1 then translocates to the nucleus to induce the transcription of genes involved in the IFNα-mediated antiviral immune response (Chen et al., 2017).

Another non-histone substrate of SETD2 is enhancer of zeste 2 (EZH2), a subunit of Polycomb repressive complex 2 (PRC2) (Yuan et al., 2020). PRC2 catalyses the deposition of H3K27me3, a modification primarily associated with transcriptional silencing. SETD2 monomethylates lysine 735 on EZH2. Methylated EZH2 is targeted for degradation through E3 ubiquitin ligase signalling, which is crucial for suppressing prostate cancer progression in mice (Yuan et al., 2020). SETD2 can thereby indirectly influence levels of further histone modifications.

Additionally, SETD2 appears to play a role in DNA damage signalling by directly interacting with the tumour suppressor protein p53 (also known as TP53). p53 inhibits the replication of cells containing damaged DNA and directly interacts with the C-terminal region of SETD2 in human cells (Xie et al., 2008). This interaction appears to enhance the transcriptional activity of p53, with overexpression of SETD2 causing a subsequent increase in the expression of various p53 target genes. SETD2 also indirectly enhances p53 stability by downregulating the transcription of HDM2 (also known as MDM2), which is involved in p53 degradation (Xie et al., 2008). Human cells lacking SETD2 have decreased p53 activation and therefore a lower proportion of cells entering cell cycle arrest at the G1-S DNA damage checkpoint (Carvalho et al., 2014). The mechanisms behind this interaction remain unclear. SETD2 might directly methylate p53 to enhance its activity. Indeed, another lysine methyltransferase, SET8 (also known as KMT5A), has been shown to methylate p53 and modulate its activity (Shi et al., 2007). Alternatively, SETD2 might direct the transcription factor to target genes marked with H3K36me3 (Xie et al., 2008).

The capacity of SETD2 to methylate other non-histone substrates remains largely unexplored. As such, it is possible that SETD2 has significant effects on a range of different cellular processes via non-histone substrates yet to be discovered. Indeed, substrate sequence specificity analysis has identified nine potential non-histone peptide substrates that could be methylated by SETD2; in vitro testing has confirmed that one of these substrates, lysine 666 of the extracellular matrix protein fibrillin-1, is methylated by SETD2 (Schuhmacher et al., 2020). It remains to be seen whether this process occurs in vivo and how it might influence biological function.

Given that SETD2 has a diverse range of cellular functions, it is perhaps unsurprising that it plays a critical role in the development and functioning of the mammalian CNS. Data from the Human Protein Atlas indicates that SETD2 RNA is ubiquitously expressed throughout the body of adult humans, including the CNS (Karlsson et al., 2021). SETD2 protein expression is predominantly localised to the nucleus and has been detected in multiple different cell and tissue types (Uhlén et al., 2015). SETD2 is expressed in all adult human brain regions, although it is particularly enriched in the cerebellum (Sjöstedt et al., 2020). Human Developmental Biology Resource expression data has also revealed SETD2 mRNA expression in foetal human brains (Lindsay et al., 2016). Setd2 expression patterns in adult mice largely reflect patterns observed in humans, although no enrichment in the cerebellum is observed in mice (Sjöstedt et al., 2020). Setd2 is also expressed widely across the developing mouse brain at embryonic day (E)13.5 and E15.5 (Xu et al., 2021a). Notably, western blot experiments show much higher SETD2 expression in the prenatal and neonatal mouse brain when compared to the adult brain (Koenning et al., 2021). This result has been corroborated with qPCR data from the developing mouse cortex, which shows that Setd2 mRNA expression is at its highest at around E14–E18 and then decreases postnatally (Xie et al., 2021). Despite this, immunohistochemistry has confirmed that SETD2 protein is still expressed across several brain regions in the adult mouse (Koenning et al., 2021).

Several recent studies have explored the role of SETD2 in the mammalian CNS. One such study reported the presence of the SETD2-mediated tubulin methylation mark, α-TubK40me3, in the developing and adult mouse brain using western blotting (Koenning et al., 2021). Further immunohistochemistry on adult mouse brains revealed that the mark is present in postmitotic cortical neurons and Purkinje cells, as well as mossy fibres in the hippocampus. In addition, immunostaining of primary neurons cultured from the cortex and hippocampus of postnatal day (P)0 mice revealed that the mark is particularly prevalent in the dendrites. The same study also probed the biological relevance of α-TubK40me3 by generating a mouse line containing a pathogenic variant of SETD2 with a mutant SRI domain. This variant was found to lack the ability to deposit α-TubK40me3, whilst critically maintaining the ability to deposit H3K36me3 in vivo, enabling the divergent roles of SETD2 in tubulin and histone methylation to be parsed. Homozygous mice showed embryonic lethality, but heterozygous mice were viable and displayed an anxiety-like phenotype. Primary neurons cultured from the cortex and hippocampus of the heterozygous mice at P0 showed reduced dendritic arborisation and axon length. Notably, RNA-seq and ChIP-seq using brains of heterozygous mice revealed minimal changes in gene expression and H3K36me3 deposition, further emphasising that the phenotypes observed likely resulted from reduced α-TubK40me3 deposition and not modification of chromatin (Koenning et al., 2021). Loss of α-TubK40me3 might disrupt microtubule formation in neurons, a process critical for dendritic and axonal organisation (Koenning et al., 2021). These abnormal neurons might contribute to the observed anxiety-like phenotype through the disruption of important neural circuits. Indeed, disruptions in dendritic arborisation have been observed in several other mouse models that display anxiety-like behaviours (Novais et al., 2013; Pla et al., 2013). Potential links between structural and behavioural abnormalities in the mice could be investigated further by performing electrophysiological experiments to probe neuronal function.

The role of α-TubK40me3 in microtubule formation has been explored further in another study (Xie et al., 2021). Setd2 knockdown in the developing cortex of mice led to neuronal migration deficits that were rescued with the expression of a truncated SETD2 construct containing only the AWS, SET and post-SET functional domains, and nuclear export signals for cytoplasmic localisation. This construct was found to retain the ability to trimethylate α-TubK40 but not H3K36. The deficits could also be rescued with the expression of an α-tubulinK40F mutant that mimics K40 trimethylation. Furthermore, cultured neurons lacking SETD2 showed polarisation deficits and reduced microtubule numbers. This implicates the tubulin methyltransferase activity of SETD2 in the regulation of neuronal migration and polarity. α-TubK40me3 is primarily found on tubulin polymerised into microtubules, as opposed to soluble tubulin, suggesting that the mark plays a role in the formation of stable microtubules. The deficit in microtubule formation is proposed to impede the dendritic outgrowth required for neuronal polarisation. These unpolarised neurons lack the ability to efficiently migrate through the cortex, leading to lamination deficits and potentially abnormal cortical function (Xie et al., 2021).

SETD2 has also been implicated in the patterning of the cerebral cortex during development. Using a conditional Setd2 allele crossed to Emx1-Cre, Setd2 was conditionally ablated from the developing dorsal telencephalon of mice (Xu et al., 2021a). These mutants presented with a smaller cortex at P7. Whole-mount in situ hybridisation for several cortical area markers, such as Lmo4, Cad8 (also known as Cdh8) and Rorβ (also known as Rorb), revealed disrupted organisation of cortical areas, independent of any dysregulation of major patterning transcription factors such as Emx2, Pax6, Coup-TF1 (also known as Nr2f1) and Sp8. Furthermore, anterograde tracing experiments on adult Setd2 conditional knockout mice revealed disorganised corticothalamic and thalamocortical circuits. Behavioural tests revealed potential deficits in motor learning, spatial memory and social interaction in the conditional knockout mice. At a cellular level, disruptions to cortical arealisation were attributed to an H3K36me3-mediated disruption of DNMT3B activity, resulting in the downregulation of clustered protocadherin (cPcdh) gene expression during development (Xu et al., 2021a). It is possible that the expression of several other genes critical for brain function is also regulated by SETD2-dependent DNMT3B activity, although this has not yet been explored. Notably, the reason underlying the observed microcephaly in the Setd2 conditional knockout mice was not identified in this study. A reduction in brain size could be caused by reduced neurogenesis or increased apoptosis during development. Intriguingly, 5′-bromo-2′-deoxyuridine (BrdU) pulse chasing and cleaved caspase-3 (CC3) staining of the Setd2 conditional knockout mice revealed no observable changes in neurogenesis or apoptosis, respectively (Xu et al., 2021a). Further investigation is required to identify the cause of the brain size reduction in these mice. Other studies have identified roles for SETD2 in both neurogenesis and neuronal apoptosis, however. One such study revealed that SETD2 directly interacts with H2A.Z, a variant of histone H2A, to regulate neurogenesis in the developing mouse cortex (Shen et al., 2017). This interaction was found to promote the deposition of SETD2-mediated H3K36me3 at the promoter region of the transcription factor Nkx2.4. This enhances Nkx2.4 transcription, leading to the downstream transcription of genes involved in regulating neurogenesis. Disruption of the interaction by shRNA-mediated H2A.z knockdown in embryonic mouse brains was observed to increase the proliferation of neural progenitors at the expense of differentiation (Shen et al., 2017). It is worth noting, however, that although SETD2 dysfunction contributes to the neurogenesis deficits in the H2A.z knockdown mice, it might not be the sole cause. H2A.z knockdown might produce additional effects independently of SETD2, which could explain why neurogenesis deficits were not observed in the Setd2 conditional knockout mice from the aforementioned study (Xu et al., 2021a).

SETD2 has been implicated in the apoptosis of postmitotic neurons by using Nestin-Cre to selectively delete Setd2 from the developing nervous system of mice (Chu et al., 2020). CC3 staining and terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assays revealed that these mice have increased levels of apoptosis throughout the brain at E18.5 and P0 (Chu et al., 2020). This directly contrasts with the lack of changes in apoptosis observed in P0 Emx1-Cre Setd2 conditional knockout mice (Xu et al., 2021a). The discrepancy might be due to differences in the mouse model. Emx1-Cre produces a dorsal-telencephalon-specific knockout, whereas Nestin-Cre produces a nervous-system-wide knockout. It is possible that Setd2 knockout cells derived from the dorsal telencephalon do not have apoptotic deficits, but cells derived from elsewhere in the CNS do. The increased apoptosis in the Nestin-Cre Setd2 conditional knockout mice has been attributed to SETD2 loss disrupting non-homologous end joining (NHEJ), which is the primary means of DNA double-stranded break repair in postmitotic neurons (Frappart and McKinnon, 2008). Loss of NHEJ machinery has been demonstrated to increase neuronal apoptosis (Barnes et al., 1998; Gao et al., 1998). It is unclear whether the observed apoptosis in the Nestin-Cre Setd2 conditional knockout mice was sufficient to significantly reduce brain size, however, as the study did not quantify brain size (Chu et al., 2020).

The role of SETD2 in alternative splicing of pre-mRNA might also influence brain function and development. By using ChIP-seq and RNA-seq, one study in mice has observed that H3K36me3 is strongly associated with different alternatively spliced exons in the nucleus accumbens, a brain region involved in the reward system (Hu et al., 2017). A related study has indicated that H3K36me3-mediated alternative splicing might play a role in cocaine-reward behaviour. Here, administration of cocaine to mice was found to cause alternative splicing in the nucleus accumbens, along with a concomitant enrichment of H3K36me3 at alternatively spliced exons (Xu et al., 2021b). One H3K36me3-enriched and alternatively spliced gene identified in the study was Srsf11, which encodes a splice factor. Targeted enrichment of H3K36me3 at the Srsf11 gene site in the nucleus accumbens was found to modulate alternative splicing of the gene, resulting in increased cocaine-reward behaviour in mice. These results suggest that H3K36me3 mediates the alternative splicing of the splice factor SRSF11, which then regulates downstream targets to control reward behaviour (Xu et al., 2021b).

Srsf11 is likely just one of many genes regulated by H3K36me3-mediated alternative splicing in the brain. Indeed, SETD2-mediated H3K36me3 has been implicated in the alternative splicing of several genes expressed in the hippocampus that are involved with neural function and linked with autism spectrum disorder (ASD) (Shah et al., 2020). Setd2 transcription has been observed to be regulated by fragile X messenger ribonucleoprotein 1 (FMRP). Loss of FMRP, which causes fragile X syndrome, leads to decreased stalling of ribosomes during transcription, increased SETD2 expression and, therefore, abnormal H3K36me3 deposition. In mice lacking FMRP, multiple genes implicated in neural function and development show changes in H3K36me3 deposition, as do genes linked to ASD. Several of these genes also display aberrant splicing, suggesting that H3K36me3 might play a role in regulating the alternative splicing of genes involved in neural development and function (Shah et al., 2020). Further study into how the alternative splicing of these genes affects neural function is necessary to better delineate the role of SETD2 in the brain. Nonetheless, this link between ASD-related genes and SETD2-dependent alternative splicing strongly implicates SETD2 in human pathology.

The important role of SETD2 in the CNS is exemplified by the effects of pathogenic SETD2 variants on humans (Table 2). Variants have been identified in individuals diagnosed with neurodevelopmental disorders such as ASD, intellectual disability and Luscan–Lumish syndrome (Fig. 2) (Chen et al., 2021). Pathogenic heterozygous SETD2 variants are the primary cause of the autosomal dominant Luscan–Lumish syndrome, an overgrowth disorder characterised by speech and motor delay, macrocephaly, autism-related symptoms, and intellectual disability (Chen et al., 2021). Magnetic resonance imaging (MRI) has revealed enlarged cerebral ventricles and structural abnormalities in the corpus callosum, hippocampus and cerebellum of some people with suspected Luscan–Lumish syndrome (Marzin et al., 2019; Zhang et al., 2023). The Genome Aggregation Database (gnoMAD) classifies SETD2 as intolerant to loss of function in humans, indicating haploinsufficiency (Karczewski et al., 2020). Many SETD2 variants, including missense and truncating variants, occur across the gene in people with Luscan–Lumish syndrome, although they are slightly more common around the SET domain (Fig. 2). All SETD2 variants associated with Luscan–Lumish syndrome are suspected to result in loss of function, inhibiting H3K36me3 deposition or other SETD2 functions (Marzin et al., 2019). However, comprehensive functional analysis of many of these variants is lacking, so definitive conclusions on their effects cannot be drawn.

Table 2.

Neurodevelopmental disorders linked to SETD2 variants

Neurodevelopmental disorders linked to SETD2 variants
Neurodevelopmental disorders linked to SETD2 variants

Pathogenic variants of other genes result in overgrowth disorders similar to Luscan–Lumish syndrome, providing some insight into to its pathophysiology. Sotos syndrome (OMIM 117550), which is caused by pathogenic NSD1 variants, and Tatton-Brown–Rahman syndrome (OMIM 615879), which is caused by pathogenic DNMT3A variants, both have considerable overlap with Luscan–Lumish syndrome, suggesting that they might have mutual aetiologies (Baujat and Cormier-Daire, 2007; Tatton-Brown et al., 2014; Tlemsani et al., 2016). NSD1 has H3K36 methyltransferase activity responsible for depositing H3K36me1 and H3K36me2 (Li et al., 2009). The DNA methyltransferase DNMT3A also interacts with H3K36me2 and H3K36me3 to methylate DNA (Dhayalan et al., 2010). Both Sotos syndrome and Tatton-Brown–Rahman syndrome are associated with aberrant DNA methylation (Weinberg et al., 2019). Against this backdrop, it is possible that symptoms of Luscan–Lumish syndrome are caused by a loss of H3K36 methylation, resulting in abnormal DNA methylation and downstream dysregulation of gene expression. Recently, genome-wide profiling of several individuals with Luscan–Lumish syndrome has revealed widespread DNA hypomethylation, providing further evidence for this theory (Lee et al., 2023). Some of these hypomethylated genes, such as NRP2 and IGF2BP1, are linked to growth regulation, providing a possible mechanism for the observed overgrowth in Luscan–Lumish syndrome (Lee et al., 2023). Further investigation into the effects of these genes on growth is warranted.

The exact mechanisms by which SETD2 variants produce Luscan–Lumish syndrome remain largely unclear. Deficits in neuronal migration, cortical arealisation, neural circuit formation and alternative splicing like those observed in mouse models might also be present in humans and contribute to disorders such as ASD and intellectual disability. Several deficits observed in mouse models with irregular SETD2 function are associated with ASD, including abnormal cortical patterning (Lombardo et al., 2021), cPcdh expression (Anitha et al., 2013), neural circuit formation (Benkarim et al., 2021), neuronal migration (Reiner et al., 2016), microtubule structure (Gąssowska-Dobrowolska et al., 2022), as well as alternative splicing (Stamova et al., 2013).

Many questions also surround the development of macrocephaly in people with Luscan–Lumish syndrome. Limited data suggest that the symptom appears to mostly arise postnatally, with head circumference at birth often being normal (Marzin et al., 2019). It remains to be seen, however, whether prenatal brain abnormalities are present that later give rise to this postnatal macrocephaly. It is also unclear whether the macrocephaly is a result of enlarged brain size (megalencephaly), dysregulation of the ventricular system (hydrocephaly) or overgrowth of the skull (cranial hyperostosis). Observed ventriculomegaly in some individuals with SETD2 variants suggests that hydrocephaly might be a contributing factor (Luscan et al., 2014; Marzin et al., 2019), but more comprehensive MRI analysis would be necessary to discern the exact cause of the macrocephaly. Intriguingly, mice with cortex-specific homozygous Setd2 knockout show a reduction in mature cortex size, a seemingly opposing phenotype to humans with SETD2 loss of function (Xu et al., 2021a). Hence, it is possible that mouse and human SETD2 have different functions, or that macrocephaly arises from SETD2 dysregulation in areas outside the cortex, such as the ventricular system or the skull. However, there are two important differences between the mouse and human studies. First, the conditional deletion of Setd2 by Emx1-Cre in the mouse model will leave most, if not all, cortical interneurons and many oligodendrocytes with Setd2 intact. Second, the mice with reduced brain size in the conditional knockout study were homozygous for the Setd2 deletion, whereas human SETD2 disorders are all heterozygous. Heterozygosity might produce a completely different effect. Unlike in homozygous mice, no apparent changes in cortical organisation were identified in conditional heterozygote mice compared to controls in the aforementioned study (Xu et al., 2021a). Further study of heterozygous mouse models will be necessary to better understand the role of SETD2 in human disease.

Interestingly, a separate neurodevelopmental disorder associated with SETD2 variants has been recently identified, known as Rabin–Pappas syndrome (OMIM 620155) (Rabin et al., 2020). Individuals with this syndrome typically present with intellectual disability, microcephaly, and hypoplasia of the corpus callosum, pons and cerebellum (Rabin et al., 2020). Many of these features do not overlap with Luscan–Lumish syndrome. Of particular interest is the observed microcephaly, which is in direct contrast to the macrocephaly observed in Luscan–Lumish syndrome, but which might correlate with mouse Setd2 mutants. Rabin–Pappas syndrome was initially thought to be caused by a single de novo heterozygous SETD2 missense variant, p.R1740W (Rabin et al., 2020). However, a recent study has identified two other missense variants, p.E1718K and p.D2251E, that lead to a similar phenotype in humans (Parra et al., 2023). These variants are suspected to result in gain of function, although functional studies are lacking (Parra et al., 2023; Rabin et al., 2020). Recent genome-wide profiling has revealed that p.R1740W is associated with hypermethylation of DNA, which contrasts with the hypomethylation observed in Luscan–Lumish syndrome. Indeed, all the overlapping differentially methylated genes between the two syndromes show opposing methylation profiles, which might explain the opposing phenotypes (Lee et al., 2023). Furthermore, gain-of-function variants of DNMT3A are associated with hypermethylation and microcephalic dwarfism, further implicating Rabin–Pappas syndrome as a gain-of-function disorder (Heyn et al., 2019). Another reported missense variant, p.R1740Q, is associated with a less severe phenotype characterised by intellectual disability but no microcephaly (Lee et al., 2023). This disorder has been named intellectual development disorder, autosomal dominant 70 (OMIM 620157). Notably, codon 1740 does not lie in a known functional domain of SETD2. No individuals with Luscan–Lumish syndrome have been observed with a missense variant at or near this codon. It is possible that it resides in a functional domain that is yet to be described, and that dysregulation of functions mediated by this domain produces disease (Lee et al., 2023).

SETD2 has a wide range of cellular functions, which are mediated by its ability to deposit H3K36me3 and to interact with a variety of non-histone substrates. Many of these functions are critical to the development and function of the CNS, highlighted by the fact that pathogenic SETD2 variants in humans lead to neurodevelopmental disorders. Despite recent advances, it is still largely unclear what specific aspects of SETD2 function, when abrogated, contribute to disorders observed in humans. As mentioned previously, mice heterozygous for mutations that impair SETD2 tubulin methyltransferase activity, but not histone methyltransferase activity, present with neuronal and behavioural defects (Koenning et al., 2021). These results indicate that the tubulin methyltransferase activity of SETD2 is haploinsufficient in heterozygous mice, but evidence of haploinsufficiency for histone methyltransferase activity is lacking. In mouse embryonic fibroblast models, loss of one Setd2 allele does not significantly abrogate H3K36me3 deposition (Chiang et al., 2018). This suggests that human disorders like Luscan–Lumish syndrome might primarily be a result of defects in the ability of SETD2 to methylate non-histone substrates. However, this does not explain the functional overlap of Luscan–Lumish syndrome with Sotos syndrome and Tatton-Brown–Rahman syndrome, or the observed alterations in DNA methylation in individuals with pathogenic SETD2 variants, which could be attributed to H3K36me3-mediated DNA methylation deficits. Further study on mouse models and humans will be necessary to precisely distinguish the role of SETD2 in human disease.

Recent advances in mouse models and experimental techniques might provide insight into the role of SETD2 in the CNS in the future. Originally, one of the major roadblocks for studying SETD2 in the CNS of mice was the fact that total knockouts of Setd2 cause embryonic lethality at E10.5–E11.5 (Hu et al., 2010). Now, Setd2-floxed mice are readily available. These mice can be crossed with a range of Cre deleter strains to produce conditional knockouts that can survive into adulthood, providing more suitable models for interrogating the role of SETD2 in CNS function (Xu et al., 2021a). The identification of mutant forms of Setd2 that specifically abrogate certain aspects of SETD2 activity, such as its ability to methylate α-tubulin or H3K36, has allowed for the further delineation of the mechanisms underlying the role of this protein in the CNS (Koenning et al., 2021; Park et al., 2016; Xie et al., 2021). Understanding which specific SETD2 substrates contribute to CNS dysfunction might provide insight into possible treatments for disorders such as Luscan–Lumish syndrome. Emerging techniques such as single-cell RNA sequencing, ChIP sequencing and ATAC sequencing might provide further insight into the function of SETD2 in the brain by identifying how it regulates gene expression, epigenetic interactions with genes and chromatin architecture, respectively. These approaches could provide more answers on how observed deficits arise in current mouse models and might provide a better understanding of how SETD2 dysfunction causes disease, potentially leading to the development of treatments for Luscan–Lumish syndrome and other SETD2-associated pathologies.

Funding

Our work in this area is supported by the Australian Research Council (DP230101750).

Anitha
,
A.
,
Thanseem
,
I.
,
Nakamura
,
K.
,
Yamada
,
K.
,
Iwayama
,
Y.
,
Toyota
,
T.
,
Iwata
,
Y.
,
Suzuki
,
K.
,
Sugiyama
,
T.
,
Tsujii
,
M.
et al. 
(
2013
).
Protocadherin α (PCDHA) as a novel susceptibility gene for autism
.
J. Psychiatry Neurosci.
38
,
192
-
198
.
Bannister
,
A. J.
,
Schneider
,
R.
,
Myers
,
F. A.
,
Thorne
,
A. W.
,
Crane-Robinson
,
C.
and
Kouzarides
,
T.
(
2005
).
Spatial distribution of di- and tri-methyl lysine 36 of histone H3 at active genes
.
J. Biol. Chem.
280
,
17732
-
17736
.
Barnes
,
D. E.
,
Stamp
,
G.
,
Rosewell
,
I.
,
Denzel
,
A.
and
Lindahl
,
T.
(
1998
).
Targeted disruption of the gene encoding DNA ligase IV leads to lethality in embryonic mice
.
Curr. Biol.
8
,
1395
-
1398
.
Baubec
,
T.
,
Colombo
,
D. F.
,
Wirbelauer
,
C.
,
Schmidt
,
J.
,
Burger
,
L.
,
Krebs
,
A. R.
,
Akalin
,
A.
and
Schübeler
,
D.
(
2015
).
Genomic profiling of DNA methyltransferases reveals a role for DNMT3B in genic methylation
.
Nature
520
,
243
-
247
.
Baujat
,
G.
and
Cormier-Daire
,
V.
(
2007
).
Sotos syndrome
.
Orphanet J. Rare Dis.
2
,
36
.
Benkarim
,
O.
,
Paquola
,
C.
,
Park
,
B.-y.
,
Hong
,
S.-J.
,
Royer
,
J.
,
Vos De Wael
,
R.
,
Lariviere
,
S.
,
Valk
,
S.
,
Bzdok
,
D.
,
Mottron
,
L.
et al. 
(
2021
).
Connectivity alterations in autism reflect functional idiosyncrasy
.
Communications Biology
4
,
1078
.
Berger
,
S. L.
(
2002
).
Histone modifications in transcriptional regulation
.
Curr. Opin. Genet. Dev.
12
,
142
-
148
.
Bhattacharya
,
S.
and
Workman
,
J. L.
(
2020
).
Regulation of SETD2 stability is important for the fidelity of H3K36me3 deposition
.
Epigenetics & Chromatin
13
,
40
.
Bhattacharya
,
S.
,
Levy
,
M. J.
,
Zhang
,
N.
,
Li
,
H.
,
Florens
,
L.
,
Washburn
,
M. P.
and
Workman
,
J. L.
(
2021
).
The methyltransferase SETD2 couples transcription and splicing by engaging mRNA processing factors through its SHI domain
.
Nat. Commun.
12
,
1443
.
Biswas
,
S.
and
Rao
,
C. M.
(
2018
).
Epigenetic tools (the writers, the readers and the erasers) and their implications in cancer therapy
.
Eur. J. Pharmacol.
837
,
8
-
24
.
Bleuyard
,
J.-Y.
,
Fournier
,
M.
,
Nakato
,
R.
,
Couturier
,
A. M.
,
Katou
,
Y.
,
Ralf
,
C.
,
Hester
,
S. S.
,
Dominguez
,
D.
,
Rhodes
,
D.
,
Humphrey
,
T. C.
et al. 
(
2017
).
MRG15-mediated tethering of PALB2 to unperturbed chromatin protects active genes from genotoxic stress
.
Proc. Natl Acad. Sci. USA
114
,
7671
-
7676
.
Brien
,
G. L.
,
Gambero
,
G.
,
O'connell
,
D. J.
,
Jerman
,
E.
,
Turner
,
S. A.
,
Egan
,
C. M.
,
Dunne
,
E. J.
,
Jurgens
,
M. C.
,
Wynne
,
K.
,
Piao
,
L.
et al. 
(
2012
).
Polycomb PHF19 binds H3K36me3 and recruits PRC2 and demethylase NO66 to embryonic stem cell genes during differentiation
.
Nat. Struct. Mol. Biol.
19
,
1273
-
1281
.
Cai
,
L.
,
Rothbart
,
S. B.
,
Lu
,
R.
,
Xu
,
B.
,
Chen
,
W. Y.
,
Tripathy
,
A.
,
Rockowitz
,
S.
,
Zheng
,
D.
,
Patel
,
D. J.
,
Allis
,
C. D.
et al. 
(
2013
).
An H3K36 methylation-engaging Tudor motif of polycomb-like proteins mediates PRC2 complex targeting
.
Mol. Cell
49
,
571
-
582
.
Carvalho
,
S.
,
Raposo
,
A. C.
,
Martins
,
F. B.
,
Grosso
,
A. R.
,
Sridhara
,
S. C.
,
Rino
,
J.
,
Carmo-Fonseca
,
M.
and
De Almeida
,
S. F.
(
2013
).
Histone methyltransferase SETD2 coordinates FACT recruitment with nucleosome dynamics during transcription
.
Nucleic Acids Res.
41
,
2881
-
2893
.
Carvalho
,
S.
,
Vítor
,
A. C.
,
Sridhara
,
S. C.
,
Martins
,
F. B.
,
Raposo
,
A. C.
,
Desterro
,
J. M.
,
Ferreira
,
J.
and
De Almeida
,
S. F.
(
2014
).
SETD2 is required for DNA double-strand break repair and activation of the p53-mediated checkpoint
.
Elife
3
,
e02482
.
Chantalat
,
S.
,
Depaux
,
A.
,
Héry
,
P.
,
Barral
,
S.
,
Thuret
,
J. Y.
,
Dimitrov
,
S.
and
Gérard
,
M.
(
2011
).
Histone H3 trimethylation at lysine 36 is associated with constitutive and facultative heterochromatin
.
Genome Res.
21
,
1426
-
1437
.
Chen
,
K.
,
Liu
,
J.
,
Liu
,
S.
,
Xia
,
M.
,
Zhang
,
X.
,
Han
,
D.
,
Jiang
,
Y.
,
Wang
,
C.
and
Cao
,
X.
(
2017
).
Methyltransferase SETD2-mediated methylation of STAT1 is critical for interferon antiviral activity
.
Cell
170
,
492
-
506.e14
.
Chen
,
M.
,
Quan
,
Y.
,
Duan
,
G.
,
Wu
,
H.
,
Bai
,
T.
,
Wang
,
Y.
,
Zhou
,
S.
,
Ou
,
J.
,
Shen
,
Y.
,
Hu
,
Z.
et al. 
(
2021
).
Mutation pattern and genotype-phenotype correlations of SETD2 in neurodevelopmental disorders
.
Eur. J. Med. Genet.
64
,
104200
.
Chiang
,
Y.-C.
,
Park
,
I.-Y.
,
Terzo
,
E. A.
,
Tripathi
,
D. N.
,
Mason
,
F. M.
,
Fahey
,
C. C.
,
Karki
,
M.
,
Shuster
,
C. B.
,
Sohn
,
B.-H.
,
Chowdhury
,
P.
et al. 
(
2018
).
SETD2 haploinsufficiency for microtubule methylation is an early driver of genomic instability in renal cell carcinoma
.
Cancer Res.
78
,
3135
-
3146
.
Chu
,
S. H.
,
Chabon
,
J. R.
,
Matovina
,
C. N.
,
Minehart
,
J. C.
,
Chen
,
B. R.
,
Zhang
,
J.
,
Kumar
,
V.
,
Xiong
,
Y.
,
Callen
,
E.
,
Hung
,
P. J.
et al. 
(
2020
).
Loss of H3K36 methyltransferase SETD2 impairs V(D)J recombination during lymphoid development
.
iScience
23
,
100941
.
De Almeida
,
S. F.
,
Grosso
,
A. R.
,
Koch
,
F.
,
Fenouil
,
R.
,
Carvalho
,
S.
,
Andrade
,
J.
,
Levezinho
,
H.
,
Gut
,
M.
,
Eick
,
D.
,
Gut
,
I.
et al. 
(
2011
).
Splicing enhances recruitment of methyltransferase HYPB/Setd2 and methylation of histone H3 Lys36
.
Nat. Struct. Mol. Biol.
18
,
977
-
983
.
Dhayalan
,
A.
,
Rajavelu
,
A.
,
Rathert
,
P.
,
Tamas
,
R.
,
Jurkowska
,
R. Z.
,
Ragozin
,
S.
and
Jeltsch
,
A.
(
2010
).
The Dnmt3a PWWP domain reads histone 3 lysine 36 trimethylation and guides DNA methylation
.
J. Biol. Chem.
285
,
26114
-
26120
.
Edmunds
,
J. W.
,
Mahadevan
,
L. C.
and
Clayton
,
A. L.
(
2008
).
Dynamic histone H3 methylation during gene induction: HYPB/Setd2 mediates all H3K36 trimethylation
.
EMBO J.
27
,
406
-
420
.
Faber
,
P. W.
,
Barnes
,
G. T.
,
Srinidhi
,
J.
,
Chen
,
J.
,
Gusella
,
J. F.
and
Macdonald
,
M. E.
(
1998
).
Huntingtin Interacts with a Family of WW Domain Proteins
.
Hum. Mol. Genet.
7
,
1463
-
1474
.
Frappart
,
P. O.
and
Mckinnon
,
P. J.
(
2008
).
Mouse models of DNA double-strand break repair and neurological disease
.
DNA Repair (Amst)
7
,
1051
-
1060
.
Gao
,
Y.
,
Sun
,
Y.
,
Frank
,
K. M.
,
Dikkes
,
P.
,
Fujiwara
,
Y.
,
Seidl
,
K. J.
,
Sekiguchi
,
J. M.
,
Rathbun
,
G. A.
,
Swat
,
W.
,
Wang
,
J.
et al. 
(
1998
).
A Critical Role for DNA End-Joining Proteins in Both Lymphogenesis and Neurogenesis
.
Cell
95
,
891
-
902
.
Gąssowska-Dobrowolska
,
M.
,
Kolasa
,
A.
,
Beversdorf
,
D. Q.
and
Adamczyk
,
A.
(
2022
).
Alterations in Cerebellar Microtubule Cytoskeletal Network in a ValproicAcid-Induced Rat Model of Autism Spectrum Disorders
.
Biomedicines
10
,
3031
.
González-Rodríguez
,
P.
,
Delorme-Axford
,
E.
,
Bernard
,
A.
,
Keane
,
L.
,
Stratoulias
,
V.
,
Grabert
,
K.
,
Engskog-Vlachos
,
P.
,
Füllgrabe
,
J.
,
Klionsky
,
D. J.
and
Joseph
,
B.
(
2022
).
SETD2 transcriptional control of ATG14L/S isoforms regulates autophagosome–lysosome fusion
.
Cell Death & Disease
13
,
953
.
Guo
,
R.
,
Zheng
,
L.
,
Park
,
J. W.
,
Lv
,
R.
,
Chen
,
H.
,
Jiao
,
F.
,
Xu
,
W.
,
Mu
,
S.
,
Wen
,
H.
,
Qiu
,
J.
et al. 
(
2014
).
BS69/ZMYND11 Reads and Connects Histone H3.3 Lysine 36 Trimethylation-Decorated Chromatin to Regulated Pre-mRNA Processing
.
Mol. Cell
56
,
298
-
310
.
Heyn
,
P.
,
Logan
,
C. V.
,
Fluteau
,
A.
,
Challis
,
R. C.
,
Auchynnikava
,
T.
,
Martin
,
C.-A.
,
Marsh
,
J. A.
,
Taglini
,
F.
,
Kilanowski
,
F.
,
Parry
,
D. A.
et al. 
(
2019
).
Gain-of-function DNMT3A mutations cause microcephalic dwarfism and hypermethylation of Polycomb-regulated regions
.
Nat. Genet.
51
,
96
-
105
.
Hu
,
M.
,
Sun
,
X.-J.
,
Zhang
,
Y.-L.
,
Kuang
,
Y.
,
Hu
,
C.-Q.
,
Wu
,
W.-L.
,
Shen
,
S.-H.
,
Du
,
T.-T.
,
Li
,
H.
,
He
,
F.
et al. 
(
2010
).
Histone H3 lysine 36 methyltransferase Hypb/Setd2 is required for embryonic vascular remodeling
.
Proc. Natl Acad. Sci. USA
107
,
2956
-
2961
.
Hu
,
Q.
,
Kim
,
E. J.
,
Feng
,
J.
,
Grant
,
G. R.
and
Heller
,
E. A.
(
2017
).
Histone posttranslational modifications predict specific alternative exon subtypes in mammalian brain
.
PLoS Comput. Biol.
13
,
e1005602
.
Huang
,
H.
,
Weng
,
H.
,
Zhou
,
K.
,
Wu
,
T.
,
Zhao
,
B. S.
,
Sun
,
M.
,
Chen
,
Z.
,
Deng
,
X.
,
Xiao
,
G.
,
Auer
,
F.
et al. 
(
2019
).
Histone H3 trimethylation at lysine 36 guides m(6)A RNA modification co-transcriptionally
.
Nature
567
,
414
-
419
.
Karczewski
,
K. J.
,
Francioli
,
L. C.
,
Tiao
,
G.
,
Cummings
,
B. B.
,
Alföldi
,
J.
,
Wang
,
Q.
,
Collins
,
R. L.
,
Laricchia
,
K. M.
,
Ganna
,
A.
,
Birnbaum
,
D. P.
et al. 
(
2020
).
The mutational constraint spectrum quantified from variation in 141,456 humans
.
Nature
581
,
434
-
443
.
Karlsson
,
M.
,
Zhang
,
C.
,
Méar
,
L.
,
Zhong
,
W.
,
Digre
,
A.
,
Katona
,
B.
,
Sjöstedt
,
E.
,
Butler
,
L.
,
Odeberg
,
J.
,
Dusart
,
P.
et al. 
(
2021
).
A single-cell type transcriptomics map of human tissues
.
Sci. Adv.
7
,
eabh2169
.
Kearns
,
S.
,
Mason
,
F. M.
,
Rathmell
,
W. K.
,
Park
,
I. Y.
,
Walker
,
C.
,
Verhey
,
K. J.
and
Cianfrocco
,
M. A.
(
2021
).
Molecular determinants for α-tubulin methylation by SETD2
.
J. Biol. Chem.
297
,
100898
.
Kim
,
S.
,
Kim
,
H.
,
Fong
,
N.
,
Erickson
,
B.
and
Bentley
,
D. L.
(
2011
).
Pre-mRNA splicing is a determinant of histone H3K36 methylation
.
Proc. Natl. Acad. Sci. USA
108
,
13564
-
13569
.
Klose
,
R. J.
,
Yamane
,
K.
,
Bae
,
Y.
,
Zhang
,
D.
,
Erdjument-Bromage
,
H.
,
Tempst
,
P.
,
Wong
,
J.
and
Zhang
,
Y.
(
2006
).
The transcriptional repressor JHDM3A demethylates trimethyl histone H3 lysine9 and lysine36
.
Nature
442
,
312
-
316
.
Koenning
,
M.
,
Wang
,
X.
,
Karki
,
M.
,
Jangid
,
R. K.
,
Kearns
,
S.
,
Tripathi
,
D. N.
,
Cianfrocco
,
M.
,
Verhey
,
K. J.
,
Jung
,
S. Y.
,
Coarfa
,
C.
et al. 
(
2021
).
Neuronal SETD2 activity links microtubule methylation to an anxiety-like phenotype in mice
.
Brain
144
,
2527
-
2540
.
Lee
,
S.
,
Menzies
,
L.
,
Hay
,
E.
,
Ochoa
,
E.
,
Docquier
,
F.
,
Rodger
,
F.
,
Deshpande
,
C.
,
Foulds
,
N. C.
,
Jacquemont
,
S.
,
Jizi
,
K.
et al. 
(
2023
).
Epigenotype-genotype-phenotype correlations in SETD1A and SETD2 chromatin disorders
.
Hum. Mol. Genet.
. [Epub]
ddad079
.
Lelieveld
,
S. H.
,
Reijnders
,
M. R.
,
Pfundt
,
R.
,
Yntema
,
H. G.
,
Kamsteeg
,
E. J.
,
De Vries
,
P.
,
De Vries
,
B. B.
,
Willemsen
,
M. H.
,
Kleefstra
,
T.
,
Löhner
,
K.
et al. 
(
2016
).
Meta-analysis of 2,104 trios provides support for 10 new genes for intellectual disability
.
Nat. Neurosci.
19
,
1194
-
1196
.
Li
,
M.
,
Phatnani
,
H. P.
,
Guan
,
Z.
,
Sage
,
H.
,
Greenleaf
,
A. L.
and
Zhou
,
P.
(
2005
).
Solution structure of the Set2–Rpb1 interacting domain of human Set2 and its interaction with the hyperphosphorylated C-terminal domain of Rpb1
.
Proc. Natl Acad. Sci. USA
102
,
17636
-
17641
.
Li
,
Y.
,
Trojer
,
P.
,
Xu
,
C.-F.
,
Cheung
,
P.
,
Kuo
,
A.
,
Drury
,
W. J.
,
Qiao
,
Q.
,
Neubert
,
T. A.
,
Xu
,
R.-M.
and
Gozani
,
O.
et al. 
(
2009
).
The target of the NSD family of histone lysine methyltransferases depends on the nature of the substrate
.
J. Biol. Chem.
284
,
34283
-
34295
.
Li
,
F.
,
Mao
,
G.
,
Tong
,
D.
,
Huang
,
J.
,
Gu
,
L.
,
Yang
,
W.
and
Li
,
G. M.
(
2013
).
The histone mark H3K36me3 regulates human DNA mismatch repair through its interaction with MutSα
.
Cell
153
,
590
-
600
.
Lindsay
,
S. J.
,
Xu
,
Y.
,
Lisgo
,
S. N.
,
Harkin
,
L. F.
,
Copp
,
A. J.
,
Gerrelli
,
D.
,
Clowry
,
G. J.
,
Talbot
,
A.
,
Keogh
,
M. J.
,
Coxhead
,
J.
et al. 
(
2016
).
HDBR expression: a unique resource for global and individual gene expression studies during early human brain development
.
Front Neuroanat
10
,
86
.
Liu
,
Y.
,
Zhang
,
Y.
,
Xue
,
H.
,
Cao
,
M.
,
Bai
,
G.
,
Mu
,
Z.
,
Yao
,
Y.
,
Sun
,
S.
,
Fang
,
D.
and
Huang
,
J.
(
2021
).
Cryo-EM structure of SETD2/Set2 methyltransferase bound to a nucleosome containing oncohistone mutations
.
Cell Discovery
7
,
32
.
Lombardo
,
M. V.
,
Eyler
,
L.
,
Pramparo
,
T.
,
Gazestani
,
V. H.
,
Hagler
,
D. J.
, Jr.
,
Chen
,
C. H.
,
Dale
,
A. M.
,
Seidlitz
,
J.
,
Bethlehem
,
R. A. I.
,
Bertelsen
,
N.
et al. 
(
2021
).
Atypical genomic cortical patterning in autism with poor early language outcome
.
Sci. Adv.
7
,
eabh1663
.
Luco
,
R. F.
,
Pan
,
Q.
,
Tominaga
,
K.
,
Blencowe
,
B. J.
,
Pereira-Smith
,
O. M.
and
Misteli
,
T.
(
2010
).
Regulation of Alternative Splicing by Histone Modifications
.
Science
327
,
996
-
1000
.
Luger
K.
,
Mäder
A. W.
,
Richmond
R. K.
,
Sargent
D. F.
and
Richmond
T. J.
(
1997
).
Crystal structure of the nucleosome core particle at 2.8 Å resolution
.
Nature
389
,
251
-
260
.
Lumish
,
H. S.
,
Wynn
,
J.
,
Devinsky
,
O.
and
Chung
,
W. K.
(
2015
).
Brief report: SETD2 mutation in a child with autism, intellectual disabilities and epilepsy
.
J. Autism Dev. Disord.
45
,
3764
-
3770
.
Luscan
,
A.
,
Laurendeau
,
I.
,
Malan
,
V.
,
Francannet
,
C.
,
Odent
,
S.
,
Giuliano
,
F.
,
Lacombe
,
D.
,
Touraine
,
R.
,
Vidaud
,
M.
,
Pasmant
,
E.
et al. 
(
2014
).
Mutations in SETD2 cause a novel overgrowth condition
.
J. Med. Genet.
51
,
512
-
517
.
Marzin
,
P.
,
Rondeau
,
S.
,
Aldinger
,
K. A.
,
Alessandri
,
J.-L.
,
Isidor
,
B.
,
Heron
,
D.
,
Keren
,
B.
,
Dobyns
,
W. B.
and
Cormier-Daire
,
V.
(
2019
).
SETD2 related overgrowth syndrome: Presentation of four new patients and review of the literature
.
American Journal of Medical Genetics Part C: Seminars in Medical Genetics
181
,
509
-
518
.
Mccauley
,
B. S.
and
Dang
,
W.
(
2021
).
Loosening chromatin and dysregulated transcription: a perspective on cryptic transcription during mammalian aging
.
Briefings in Functional Genomics
21
,
56
-
61
.
Molenaar
,
T. M.
,
Malik
,
M.
,
Silva
,
J.
,
Liu
,
N. Q.
,
Haarhuis
,
J. H. I.
,
Ambrosi
,
C.
,
Kwesi-Maliepaard
,
E. M.
,
Van Welsem
,
T.
,
Baubec
,
T.
,
Faller
,
W. J.
et al. 
(
2022
).
The histone methyltransferase SETD2 negatively regulates cell size
.
J. Cell Sci.
135
,
jcs259856
.
Musselman
,
C. A.
,
Avvakumov
,
N.
,
Watanabe
,
R.
,
Abraham
,
C. G.
,
Lalonde
,
M. E.
,
Hong
,
Z.
,
Allen
,
C.
,
Roy
,
S.
,
Nuñez
,
J. K.
,
Nickoloff
,
J.
et al. 
(
2012
).
Molecular basis for H3K36me3 recognition by the Tudor domain of PHF1
.
Nat. Struct. Mol. Biol.
19
,
1266
-
1272
.
Neri
,
F.
,
Rapelli
,
S.
,
Krepelova
,
A.
,
Incarnato
,
D.
,
Parlato
,
C.
,
Basile
,
G.
,
Maldotti
,
M.
,
Anselmi
,
F.
and
Oliviero
,
S.
(
2017
).
Intragenic DNA methylation prevents spurious transcription initiation
.
Nature
543
,
72
-
77
.
Novais
,
A.
,
Ferreira
,
A.
,
Marques
,
F.
,
Pêgo
,
J.
,
Cerqueira
,
J.
,
David-Pereira
,
A.
,
Campos
,
F.
,
Dalla
,
C.
,
Kokras
,
N.
,
Sousa
,
N.
et al. 
(
2013
).
Neudesin is involved in anxiety behavior: structural and neurochemical correlates
.
Frontiers in Behavioral Neuroscience
7
,
119
.
Okano
,
M.
,
Bell
,
D. W.
,
Haber
,
D. A.
and
Li
,
E.
(
1999
).
DNA methyltransferases dnmt3a and dnmt3b are essential for De Novo methylation and mammalian development
.
Cell
99
,
247
-
257
.
O'Roak
,
B. J.
,
Vives
,
L.
,
Fu
,
W.
,
Egertson
,
J. D.
,
Stanaway
,
I. B.
,
Phelps
,
I. G.
,
Carvill
,
G.
,
Kumar
,
A.
,
Lee
,
C.
,
Ankenman
,
K.
et al. 
(
2012
).
Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum disorders
.
Science
338
,
1619
-
1622
.
Park
,
I. Y.
,
Powell
,
R. T.
,
Tripathi
,
D. N.
,
Dere
,
R.
,
Ho
,
T. H.
,
Blasius
,
T. L.
,
Chiang
,
Y. C.
,
Davis
,
I. J.
,
Fahey
,
C. C.
,
Hacker
,
K. E.
et al. 
(
2016
).
Dual Chromatin and Cytoskeletal Remodeling by SETD2
.
Cell
166
,
950
-
962
.
Park
,
J.
,
Lee
,
K.
,
Kim
,
K.
and
Yi
,
S.-J.
(
2022
).
The role of histone modifications: from neurodevelopment to neurodiseases
.
Signal Transduction and Targeted Therapy
7
,
217
.
Parra
,
A.
,
Rabin
,
R.
,
Pappas
,
J.
,
Pascual
,
P.
,
Cazalla
,
M.
,
Arias
,
P.
,
Gallego-Zazo
,
N.
,
Santana
,
A.
,
Arroyo
,
I.
,
Artigas
,
M.
et al. 
(
2023
).
Clinical heterogeneity and different phenotypes in patients with setd2 variants: 18 new patients and review of the literature
.
Genes
14
,
1179
.
Pfister
,
S. X.
,
Ahrabi
,
S.
,
Zalmas
,
L. P.
,
Sarkar
,
S.
,
Aymard
,
F.
,
Bachrati
,
C. Z.
,
Helleday
,
T.
,
Legube
,
G.
,
La Thangue
,
N. B.
,
Porter
,
A. C.
et al. 
(
2014
).
SETD2-dependent histone H3K36 trimethylation is required for homologous recombination repair and genome stability
.
Cell Rep
7
,
2006
-
2018
.
Pfister
,
S. X.
,
Markkanen
,
E.
,
Jiang
,
Y.
,
Sarkar
,
S.
,
Woodcock
,
M.
,
Orlando
,
G.
,
Mavrommati
,
I.
,
Pai
,
C.-C.
,
Zalmas
,
L.-P.
,
Drobnitzky
,
N.
et al. 
(
2015
).
Inhibiting WEE1 Selectively Kills Histone H3K36me3-Deficient Cancers by dNTP Starvation
.
Cancer Cell
28
,
557
-
568
.
Pla
,
P.
,
Orvoen
,
S.
,
Benstaali
,
C.
,
Dodier
,
S.
,
Gardier
,
A. M.
,
David
,
D. J.
,
Humbert
,
S.
and
Saudou
,
F.
(
2013
).
Huntingtin acts non cell-autonomously on hippocampal neurogenesis and controls anxiety-related behaviors in adult mouse
.
PLOS ONE
8
,
e73902
.
Powers
,
N. R.
,
Parvanov
,
E. D.
,
Baker
,
C. L.
,
Walker
,
M.
,
Petkov
,
P. M.
and
Paigen
,
K.
(
2016
).
The meiotic recombination activator PRDM9 trimethylates both H3K36 and H3K4 at recombination hotspots in vivo
.
PLoS Genet.
12
,
e1006146
.
Pradeepa
,
M. M.
,
Sutherland
,
H. G.
,
Ule
,
J.
,
Grimes
,
G. R.
and
Bickmore
,
W. A.
(
2012
).
Psip1/Ledgf p52 binds methylated histone H3K36 and splicing factors and contributes to the regulation of alternative splicing
.
PLoS Genet.
8
,
e1002717
.
Rabin
,
R.
,
Radmanesh
,
A.
,
Glass
,
I. A.
,
Dobyns
,
W. B.
,
Aldinger
,
K. A.
,
Shieh
,
J. T.
,
Romoser
,
S.
,
Bombei
,
H.
,
Dowsett
,
L.
,
Trapane
,
P.
et al. 
(
2020
).
Genotype–phenotype correlation at codon 1740 of SETD2
.
American Journal of Medical Genetics Part A
182
,
2037
-
2048
.
Ramazi
,
S.
,
Allahverdi
,
A.
and
Zahiri
,
J.
(
2020
).
Evaluation of post-translational modifications in histone proteins: A review on histone modification defects in developmental and neurological disorders
.
J. Biosci.
45
,
135
.
Reiner
,
O.
,
Karzbrun
,
E.
,
Kshirsagar
,
A.
and
Kaibuchi
,
K.
(
2016
).
Regulation of neuronal migration, an emerging topic in autism spectrum disorders
.
J. Neurochem.
136
,
440
-
456
.
Santos-Rosa
,
H.
,
Millán-Zambrano
,
G.
,
Han
,
N.
,
Leonardi
,
T.
,
Klimontova
,
M.
,
Nasiscionyte
,
S.
,
Pandolfini
,
L.
,
Tzelepis
,
K.
,
Bartke
,
T.
and
Kouzarides
,
T.
(
2021
).
Methylation of histone H3 at lysine 37 by Set1 and Set2 prevents spurious DNA replication
.
Mol. Cell
81
,
2793
-
2807.e8
.
Schuhmacher
,
M. K.
,
Beldar
,
S.
,
Khella
,
M. S.
,
Bröhm
,
A.
,
Ludwig
,
J.
,
Tempel
,
W.
,
Weirich
,
S.
,
Min
,
J.
and
Jeltsch
,
A.
(
2020
).
Sequence specificity analysis of the SETD2 protein lysine methyltransferase and discovery of a SETD2 super-substrate
.
Communications Biology
3
,
511
.
Seervai
,
R. N. H.
,
Jangid
,
R. K.
,
Karki
,
M.
,
Tripathi
,
D. N.
,
Jung
,
S. Y.
,
Kearns
,
S. E.
,
Verhey
,
K. J.
,
Cianfrocco
,
M. A.
,
Millis
,
B. A.
,
Tyska
,
M. J.
et al. 
(
2020
).
The Huntingtin-interacting protein SETD2/HYPB is an actin lysine methyltransferase
.
Sci. Adv.
6
,
eabb7854
.
Sessa
,
A.
,
Fagnocchi
,
L.
,
Mastrototaro
,
G.
,
Massimino
,
L.
,
Zaghi
,
M.
,
Indrigo
,
M.
,
Cattaneo
,
S.
,
Martini
,
D.
,
Gabellini
,
C.
,
Pucci
,
C.
et al. 
(
2019
).
SETD5 regulates chromatin methylation state and preserves global transcriptional fidelity during brain development and neuronal wiring
.
Neuron
104
,
271
-
289.e13
.
Shah
,
S.
,
Molinaro
,
G.
,
Liu
,
B.
,
Wang
,
R.
,
Huber
,
K. M.
and
Richter
,
J. D.
(
2020
).
FMRP control of ribosome translocation promotes chromatin modifications and alternative splicing of neuronal genes linked to autism
.
Cell Reports
30
,
4459
-
4472
.e6.
Sharda
,
A.
and
Humphrey
,
T. C.
(
2022
).
The role of histone H3K36me3 writers, readers and erasers in maintaining genome stability
.
DNA Repair
119
,
103407
.
Shen
,
T.
,
Ji
,
F.
,
Wang
,
Y.
,
Lei
,
X.
,
Zhang
,
D.
and
Jiao
,
J.
(
2017
).
Brain-specific deletion of histone variant H2A.z results in cortical neurogenesis defects and neurodevelopmental disorder
.
Nucleic Acids Res.
46
,
2290
-
2307
.
Shi
,
X.
,
Kachirskaia
,
I.
,
Yamaguchi
,
H.
,
West
,
L. E.
,
Wen
,
H.
,
Wang
,
E. W.
,
Dutta
,
S.
,
Appella
,
E.
and
Gozani
,
O.
(
2007
).
Modulation of p53 Function by SET8-Mediated Methylation at Lysine 382
.
Mol. Cell
27
,
636
-
646
.
Sjöstedt
,
E.
,
Zhong
,
W.
,
Fagerberg
,
L.
,
Karlsson
,
M.
,
Mitsios
,
N.
,
Adori
,
C.
,
Oksvold
,
P.
,
Edfors
,
F.
,
Limiszewska
,
A.
,
Hikmet
,
F.
et al. 
(
2020
).
An atlas of the protein-coding genes in the human, pig, and mouse brain
.
Science
367
,
eaay5947
.
Stamova
,
B. S.
,
Tian
,
Y.
,
Nordahl
,
C. W.
,
Shen
,
M. D.
,
Rogers
,
S.
,
Amaral
,
D. G.
and
Sharp
,
F. R.
(
2013
).
Evidence for differential alternative splicing in blood of young boys with autism spectrum disorders
.
Molecular Autism
4
,
30
.
Strahl
,
B. D.
,
Grant
,
P. A.
,
Briggs
,
S. D.
,
Sun
,
Z. W.
,
Bone
,
J. R.
,
Caldwell
,
J. A.
,
Mollah
,
S.
,
Cook
,
R. G.
,
Shabanowitz
,
J.
,
Hunt
,
D. F.
et al. 
(
2002
).
Set2 is a nucleosomal histone H3-selective methyltransferase that mediates transcriptional repression
.
Mol. Cell. Biol.
22
,
1298
-
1306
.
Suda
,
K.
,
Fukuoka
,
H.
,
Iguchi
,
G.
,
Kanie
,
K.
,
Fujita
,
Y.
,
Odake
,
Y.
,
Matsumoto
,
R.
,
Bando
,
H.
,
Ito
,
H.
,
Takahashi
,
M.
et al. 
(
2021
).
A case of Luscan-Lumish syndrome: possible involvement of enhanced GH signaling
.
The Journal of Clinical Endocrinology & Metabolism
106
,
718
-
723
.
Sun
,
X.-J.
,
Wei
,
J.
,
Wu
,
X.-Y.
,
Hu
,
M.
,
Wang
,
L.
,
Wang
,
H.-H.
,
Zhang
,
Q.-H.
,
Chen
,
S.-J.
,
Huang
,
Q.-H.
and
Chen
,
Z.
(
2005
).
Identification and characterization of a novel human histone H3 lysine 36-specific methyltransferase
.
J. Biol. Chem.
280
,
35261
-
35271
.
Talbert
,
P. B.
and
Henikoff
,
S.
(
2021
).
Histone variants at a glance
.
J. Cell Sci.
134
,
jcs244749
.
Tatton-Brown
,
K.
,
Seal
,
S.
,
Ruark
,
E.
,
Harmer
,
J.
,
Ramsay
,
E.
,
del Vecchio Duarte
,
S.
,
Zachariou
,
A.
,
Hanks
,
S.
,
O'brien
,
E.
,
Aksglaede
,
L.
et al. 
(
2014
).
Mutations in the DNA methyltransferase gene DNMT3A cause an overgrowth syndrome with intellectual disability
.
Nat. Genet.
46
,
385
-
388
.
Tlemsani
,
C.
,
Luscan
,
A.
,
Leulliot
,
N.
,
Bieth
,
E.
,
Afenjar
,
A.
,
Baujat
,
G.
,
Doco-Fenzy
,
M.
,
Goldenberg
,
A.
,
Lacombe
,
D.
,
Lambert
,
L.
et al. 
(
2016
).
SETD2 and DNMT3A screen in the Sotos-like syndrome French cohort
.
J. Med. Genet.
53
,
743
-
751
.
Uhlén
,
M.
,
Fagerberg
,
L.
,
Hallström
,
B. M.
,
Lindskog
,
C.
,
Oksvold
,
P.
,
Mardinoglu
,
A.
,
Sivertsson
,
Å.
,
Kampf
,
C.
,
Sjöstedt
,
E.
,
Asplund
,
A.
et al. 
(
2015
).
Tissue-based map of the human proteome
.
Science
347
,
1260419
.
Van Rij
,
M. C.
,
Hollink
,
I. H.
,
Terhal
,
P. A.
,
Kant
,
S. G.
,
Ruivenkamp
,
C.
,
Van Haeringen
,
A.
,
Kievit
,
J. A.
and
Van Belzen
,
M. J.
(
2018
).
Two novel cases expanding the phenotype of SETD2–related overgrowth syndrome
.
American Journal of Medical Genetics Part A
176
,
1212
-
1215
.
Venkatesh
,
S.
and
Workman
,
J. L.
(
2013
).
Set2 mediated H3 lysine 36 methylation: regulation of transcription elongation and implications in organismal development
.
WIREs Developmental Biology
2
,
685
-
700
.
Weinberg
,
D. N.
,
Papillon-Cavanagh
,
S.
,
Chen
,
H.
,
Yue
,
Y.
,
Chen
,
X.
,
Rajagopalan
,
K. N.
,
Horth
,
C.
,
Mcguire
,
J. T.
,
Xu
,
X.
,
Nikbakht
,
H.
et al. 
(
2019
).
The histone mark H3K36me2 recruits DNMT3A and shapes the intergenic DNA methylation landscape
.
Nature
573
,
281
-
286
.
Wu
,
Y.
,
Liu
,
F.
,
Wan
,
R.
and
Jiao
,
B.
(
2023
).
A novel SETD2 variant causing global development delay without overgrowth in a Chinese 3-year-old boy
.
Frontiers in Genetics
14
,
1153284
.
Xiang
,
W.
,
He
,
J.
,
Huang
,
C.
,
Chen
,
L.
,
Tao
,
D.
,
Wu
,
X.
,
Wang
,
M.
,
Luo
,
G.
,
Xiao
,
X.
,
Zeng
,
F.
et al. 
(
2015
).
miR-106b-5p targets tumor suppressor gene SETD2 to inactive its function in clear cell renal cell carcinoma
.
Oncotarget
6
,
4066
-
4079
.
Xie
,
P.
,
Tian
,
C.
,
An
,
L.
,
Nie
,
J.
,
Lu
,
K.
,
Xing
,
G.
,
Zhang
,
L.
and
He
,
F.
(
2008
).
Histone methyltransferase protein SETD2 interacts with p53 and selectively regulates its downstream genes
.
Cell. Signal.
20
,
1671
-
1678
.
Xie
,
X.
,
Wang
,
S.
,
Li
,
M.
,
Diao
,
L.
,
Pan
,
X.
,
Chen
,
J.
,
Zou
,
W.
,
Zhang
,
X.
,
Feng
,
W.
and
Bao
,
L.
(
2021
).
α-TubK40me3 is required for neuronal polarization and migration by promoting microtubule formation
.
Nat. Commun.
12
,
4113
.
Xu
,
L.
,
Zheng
,
Y.
,
Li
,
X.
,
Wang
,
A.
,
Huo
,
D.
,
Li
,
Q.
,
Wang
,
S.
,
Luo
,
Z.
,
Liu
,
Y.
,
Xu
,
F.
et al. 
(
2021a
).
Abnormal neocortex arealization and Sotos-like syndrome-associated behavior in Setd2 mutant mice
.
Sci. Adv.
7
,
eaba1180
.
Xu
,
S.-J.
,
Lombroso
,
S. I.
,
Fischer
,
D. K.
,
Carpenter
,
M. D.
,
Marchione
,
D. M.
,
Hamilton
,
P. J.
,
Lim
,
C. J.
,
Neve
,
R. L.
,
Garcia
,
B. A.
,
Wimmer
,
M. E.
et al. 
(
2021b
).
Chromatin-mediated alternative splicing regulates cocaine-reward behavior
.
Neuron
109
,
2943
-
2966.e8
.
Yang
,
S.
,
Zheng
,
X.
,
Lu
,
C.
,
Li
,
G. M.
,
Allis
,
C. D.
and
Li
,
H.
(
2016
).
Molecular basis for oncohistone H3 recognition by SETD2 methyltransferase
.
Genes Dev.
30
,
1611
-
1616
.
Yuan
,
W.
,
Xie
,
J.
,
Long
,
C.
,
Erdjument-Bromage
,
H.
,
Ding
,
X.
,
Zheng
,
Y.
,
Tempst
,
P.
,
Chen
,
S.
,
Zhu
,
B.
and
Reinberg
,
D.
(
2009
).
Heterogeneous nuclear ribonucleoprotein L Is a subunit of human KMT3a/Set2 complex required for H3 Lys-36 trimethylation activity in vivo
.
J. Biol. Chem.
284
,
15701
-
15707
.
Yuan
,
H.
,
Li
,
N.
,
Fu
,
D.
,
Ren
,
J.
,
Hui
,
J.
,
Peng
,
J.
,
Liu
,
Y.
,
Qiu
,
T.
,
Jiang
,
M.
,
Pan
,
Q.
et al. 
(
2017
).
Histone methyltransferase SETD2 modulates alternative splicing to inhibit intestinal tumorigenesis
.
J. Clin. Invest.
127
,
3375
-
3391
.
Yuan
,
H.
,
Han
,
Y.
,
Wang
,
X.
,
Li
,
N.
,
Liu
,
Q.
,
Yin
,
Y.
,
Wang
,
H.
,
Pan
,
L.
,
Li
,
L.
,
Song
,
K.
et al. 
(
2020
).
SETD2 restricts prostate cancer metastasis by integrating EZH2 and AMPK signaling pathways
.
Cancer Cell
38
,
350
-
365.e7
.
Zhang
,
Y.
,
Fang
,
Y.
,
Tang
,
Y.
,
Han
,
S.
,
Jia
,
J.
,
Wan
,
X.
,
Chen
,
J.
,
Yuan
,
Y.
,
Zhao
,
B.
and
Fang
,
D.
(
2022
).
SMYD5 catalyzes histone H3 lysine 36 trimethylation at promoters
.
Nat. Commun.
13
,
3190
.
Zhang
,
Y.
,
Zhang
,
H.
,
Wu
,
W.
,
Wang
,
D.
,
Lv
,
Y.
,
Zhao
,
D.
,
Wang
,
L.
,
Liu
,
Y.
and
Zhang
,
K.
(
2023
).
Clinical and genetic features of luscan-lumish syndrome associated with a novel de novo variant of SETD2 gene: Case report and literature review
.
Front Genet
14
,
1081391
.
Zhao
,
B.
,
Xu
,
W.
,
Rong
,
B.
,
Chen
,
G.
,
Ye
,
X.
,
Dai
,
R.
,
Li
,
W.
,
Chen
,
J.
,
Cai
,
J.
,
Song
,
L.
et al. 
(
2018
).
H3K14me3 genomic distributions and its regulation by KDM4 family demethylases
.
Cell Res.
28
,
1118
-
1120
.
Zhu
,
K.
,
Lei
,
P.-J.
,
Ju
,
L.-G.
,
Wang
,
X.
,
Huang
,
K.
,
Yang
,
B.
,
Shao
,
C.
,
Zhu
,
Y.
,
Wei
,
G.
,
Fu
,
X.-D.
et al. 
(
2016
).
SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing
.
Nucleic Acids Res.
45
,
92
-
105
.
Zhu
,
Q.
,
Yang
,
Q.
,
Lu
,
X.
,
Wang
,
H.
,
Tong
,
L.
,
Li
,
Z.
,
Liu
,
G.
,
Bao
,
Y.
,
Xu
,
X.
,
Gu
,
L.
et al. 
(
2021
).
SETD2-mediated H3K14 trimethylation promotes ATR activation and stalled replication fork restart in response to DNA replication stress
.
Proc. Natl. Acad. Sci. USA
118
,
e2011278118
.

Competing interests

The authors declare no competing or financial interests.