Abstract

MeCP2 (Methyl CpG binding protein 2) is an intrinsically disordered protein that binds to methylated genome regions. The protein is a critical transcriptional regulator of the brain, and its mutations account for 95% of Rett syndrome (RTT) cases. Early studies of this neurodevelopmental disorder revealed a close connection with dysregulations of the ubiquitin system (UbS), notably as related to UBE3A, a ubiquitin ligase involved in the proteasome-mediated degradation of proteins. MeCP2 undergoes numerous post-translational modifications (PTMs), including ubiquitination and sumoylation, which, in addition to the potential functional outcomes of their monomeric forms in gene regulation and synaptic plasticity, in their polymeric organization, these modifications play a critical role in proteasomal degradation. UbS-mediated proteasomal degradation is crucial in maintaining MeCP2 homeostasis for proper function and is involved in decreasing MeCP2 in some RTT-causing mutations. However, regardless of all these connections to UbS, the molecular details involved in the signaling of MeCP2 for its targeting by the ubiquitin-proteasome system (UPS) and the functional roles of monomeric MeCP2 ubiquitination and sumoylation remain largely unexplored and are the focus of this review.

Introduction

In eukaryote organisms, DNA is the repository of genetic information and is present in cells as a nucleoprotein complex known as chromatin [1]. In addition to DNA, histones are the main protein component of such a complex. Both DNA and histones are subject to post-synthetic chemical modifications that play a critical role in the epigenetic information of the complex. Methylation is one such modification, which has been studied extensively in DNA and histones [2, 3]. The chemical modifications of chromatin are regulatory and constitute one of the main contributors to the epigenetic chromatin component [4]. Their downstream outcome is mediated by the dynamics with which the interacting proteins specifically recognize and bind to them. Hence, these proteins are generically referred to as “readers”. Ubiquitination and sumoylation can target these protein effectors and, in so doing can contribute to the modulation of their function. One such reader is MeCP2 (Methyl CpG binding protein 2) [5], which binds to DNA preferentially methylated at CpG and CpH (H = A or T or C [6–8]). It also binds to histone (H3 K27me3) [9, 10]. MeCP2 is a target of ubiquitination and sumoylation, which can contribute to its functional complexity. Nevertheless, little is known about the molecular mechanisms involved, and their role on MeCP2 function. In this article, we summarize our current knowledge of this important yet relatively unexplored topic.

MeCP2 is an intrinsically disordered protein with two isoforms

From structural and functional perspectives [11], the complexity of MeCP2 is enhanced by the presence of two isoforms (E1 and E2), the product of alternative splicing [12]. The two proteins differ in their first N-terminal amino acids at the N-terminal domain (NTD), with human MeCP2-E1 extending 12 amino acids longer than E2 (Fig. 1A), but they are identical otherwise. MeCP2-E2 seems to have diversified from a vertebrate precursor E1-isoform in mammals [13]. Despite the minor differences in size and amino acid sequence, the two variants bind to chromatin with different affinities [11], and although they can share some functional roles and have controversial functional redundancy [14, 15], they possess distinct functional attributes [11] and different half-lives [13].

Two structural views of MeCP2. (A) Schematic representation of the two human MeCP2 E1 and E2 isoforms indicating their structural/functional domains: NTD, N-terminal domain; MBD, methyl-binding domain; ID, intervening domain; TRD, transcription repression domain and CTD, C-terminal domain. (B) An AlphaFold [131] view of MeCP2 and (C) structure obtained using a combination of all-atom and coarse-grained simulations [19]. The blue margin square highlights the predicted condensed organization of the molecule around the TRD. The red indicates the MBD [132].
Figure 1

Two structural views of MeCP2. (A) Schematic representation of the two human MeCP2 E1 and E2 isoforms indicating their structural/functional domains: NTD, N-terminal domain; MBD, methyl-binding domain; ID, intervening domain; TRD, transcription repression domain and CTD, C-terminal domain. (B) An AlphaFold [131] view of MeCP2 and (C) structure obtained using a combination of all-atom and coarse-grained simulations [19]. The blue margin square highlights the predicted condensed organization of the molecule around the TRD. The red indicates the MBD [132].

With about 60% unstructured regions, MeCP2 represents a prototypical intrinsically disordered protein (IDP) [16–18]. Thus, while the methyl binding domain (MBD) targets the protein to methylated regions of the genome, its overall IDP nature accounts for the large variety of interactors [17], which further impart the protein with manifold downstream functions. Of interest, while the MBD has a well-defined tertiary structure organization, all-atom and coarse-grained simulations [19] predict a less constrained form, but a significant 3D folded organization encompasses the transcriptional repression domain (TRD) (Fig. 1B); the folded domain overlaps with the short trypsin-resistant peptide around the TRD [18].

MeCP2 in health and disease

Because of its potential for many partner interactions, a process that, in the case of protein interactors, is usually mediated by PTMs [20], it is not surprising that MeCP2 can exhibit contrasting functions as a repressor and an activator of gene expression depending on the partner’s nature [21, 22]. As an essential transcriptional regulator [23] in vertebrates, the protein is involved in a plethora of cellular events with diverse functions [24]. MeCP2 phosphorylation at S433 (421 in E2) has been shown to release it from the promoter of Bdnf (brain-derived neurotrophic factor) to induce its transcription, which is critical for neuronal survival and growth. In its phosphorylated form, MeCP2 contributes to regulating spine maturation and dendritic growth [25], which are essential for developing neuron circuits. Moreover, the protein has been involved in ciliogenesis by associating with the centrosome [26]. In the adult brain, the protein is involved in long-term memory [27] and in the circadian cycle-dependent regulation of gene expression [11, 28] in mice. An essential role in maintaining the blood-brain barrier has been recently reported [29]. Moreover, MeCP2 has been shown to link DNA methylation to brain metabolism, where it affects glucose and cholesterol metabolism [30]. It has also been shown to participate in the differential sensitivity of the protein translation initiation machinery and mTOR signaling [31, 32]. Importantly, MeCP2 is a global chromatin organizer of the brain both during neurodevelopment and in the mature brain [11, 33]. Hence, its involvement in neurodevelopmental and neurodegenerative diseases and many adult brain disorders and conditions is no wonder [34].

While the functionality of MeCP2 is reasonably understood in the brain because of its significant presence in this tissue, it is crucial to recognize that the protein is also expressed in numerous non-neural tissues, including the lung. Although the levels of MeCP2 expression in the lung are still controversial [35–37], MeCP2 has been shown to associate with alpha-smooth muscle actin and promote fibroblast differentiation in this tissue [38]. Furthermore, Vashi et al. [39] found that MeCP2 regulates many lipid metabolism genes in the lung and is required to maintain a normal lipid composition of pulmonary surfactant, which reduces the surface tension in the lungs’ alveoli. The intrinsic ability in transcriptional regulation of many genes and indirectly participating in their downstream effects expands the spectrum of its non-neural functionality. A recent publication describing its regulation of Homeobox A9 (HOXA9) that affects vascular smooth muscle cells differentiation [40] provides an excellent example.

Although MeCP2 was first described in 1992 [5], studies on the relevance of the protein did not become fully embraced by the scientific community until 1999 [41] when Ruth Amir and Huda Zoghbi established its connection with Rett syndrome (RTT), showing that mutations of this protein are the genetic cause of 95% of RTT cases [42]. RTT is a monogenic X chromosome-linked rare neurodevelopmental disease mainly affecting girls [43]. All these mutations affect brain functions to different extents described with phenotypes whose symptoms’ severity depends on the particular mutation involved [44] and in cellular mosaicism, which is prevalent in females [45] and somatic mosaicism in males [46, 47].

Another MeCP2-driven rare neurodevelopmental disease with symptoms overlapping with RTT [48] is MeCP2 duplication syndrome (MDS) [49–51] which is caused by an extra copy of MeCP2, predominantly affecting males. Under-expression of MeCP2 in RTT and over-expression in MDS brought awareness to the critical tightly regulated homeostasis for the proper function of this protein in the brain [52, 53]. Moreover, MeCP2 gain- and loss-of-function involves its isoform-specific homeostasis in this tissue [31]. Such controlled protein balance is widespread in neurons and involves proteostasis regulation [54], which requires tight coordination of protein synthesis, ubiquitination and proteasomal degradation [55].

Given the functional relevance and abundance of MeCP2 in the brain [9, 56], it is not surprising that alterations of this protein not only affect RTT but are pleiotropic, involved in many psychological disorders such as depression, autism [50], schizophrenia [57], and epilepsy [58]. All the conditions involve MeCP2 destabilization. Outside the brain, the regulation of HOXA9 described earlier was observed in MeCP2’s role in atherosclerosis [40], and MeCP2 has been described to be a bona fide oncogene [59], a role not yet particularly attributed to any of its two different isoforms. The oncogene role has been described in many cancer types (see the recent review [60]). Nevertheless, in cases such as breast cancer where MeCP2 has been described to as an oncogene [61], it exhibits a role to suppress cell proliferation [62]. Thus, the involvement of MeCP2 in cancer seems complex and its role may vary in a tissue-specific context.

Rett-causing mutations impair the overall ubiquitination state of the cell. MeCP2 deficiency has been shown to dysregulate genes encoding E3 ubiquitin ligases [63–67]. However, the direct effects on MeCP2 exerted by ubiquitination and sumoylation are only beginning to be elucidated.

Rett syndrome, ubiquitination and MeCP2 degradation

The RTT pathology has provided us with much information relevant to MeCP2 functionality. The early studies established a relationship between RTT and UBE3A, implicating MeCP2 in UBE3A regulation [63]. UBE3A, also known as E6AP, is a ubiquitin ligase that affects proteasome function and is also associated with the neurodevelopmental diseases Prader-Willi syndrome, Angelman syndrome, and Dup15q syndrome [68]. UBE3A is involved in neural activity-regulated synaptic processes mediated by the protein degradation pathway [69]. It was initially believed that the MeCP2 deficiency associated with RTT triggered histone-PTM epigenetic aberrations that ultimately reduced UBE3A production [70]; the observation was challenged in MeCP2 mutant mice, which did not exhibit reduced UBE3A [70, 71]. Subsequently, UBE3A was identified as an important MeCP2 cofactor, and co-immunoprecipitation analyses demonstrated its physical association with MeCP2 [72]. A study of gene expression profiles modulated in RTT using peripheral blood lymphocytes from patients followed by functional clustering identified 146 specific genes involved in pathways, some related to mitochondrial function, protein ubiquitination and proteasome degradation [73]. Also, fibroblasts from RTT patients with non-sense MeCP2 mutations have impaired and reduced proteasome subunits due to the downregulation of chaperone proteins PAC1 and PAC2 [74].

Although much evidence has been accumulated on the relationship between RTT and the proteasome-mediated protein degradation system, less is known about the effects of MeCP2 mutations on protein stability. An early seminal paper by the late Alan Wolffe’s lab expressing the RNAs of different mutants in Xenopus oocytes followed by cycloheximide pulse-chase of the protein provided evidence for differential stability of the RTT-related MeCP2 mutants [75]. Using a RTT mouse model developed in Rudolph Jaeinsch’s lab expressing MeCP2 missing an essential part of its MBD [76], the amount of the truncated protein expressed in the brain decreased to <50% [77]. While this represents an extreme situation, it underscores the relevance of the whole MeCP2 protein, particularly that of its MBD, to retain its stability. Table 1 summarizes the information to date on the stability of several of the RTT-MeCP2 mutants; mutations affecting the MBD consistently appear to result in lower expression of the protein in agreement with the results observed with Jaenisch mouse model [76]. However, to date, only the T158M mutant decrease in MeCP2 expression has been studied in detail and shown to involve proteasomal degradation [78]. Interestingly, this RTT phenotype was ameliorated, and the average life span in male mice reportedly increased from a median of 90 days to 150 days in transgenic mice overexpressing the mutated T158M. It was further shown that these improvements resulted from the increased binding of MeCP2 T158M to DNA. These observations suggest that the instability of the protein introduced by MBD mutations and their altered binding affinity for methylated cytosine [79, 80] are responsible for their propensity to degrade. Another example of the relevance of the ubiquitin-mediated degradation on MeCP2 with Rett-causing mutations is the missense A2V mutation [13] affecting the MeCP2-E1 isoform. This mutation causes a higher proteasomal degradation rate than the wild type. This degradation involves the removal of N-terminal methionine residues followed by acetylation of the penultimate alanine or valine [81], a process that requires ubiquitination [82].

Table 1

Classification of Rett-causing mutations based on their position in MeCP2 domains.

Domain of MeCP2MutationMolecular phenotypeProtein levels compare to WT% of RTT casesMedian life span in ♂ miceReference
N-terminal Domain (NTD)MeCP2-E1- A2VMethionine at the P1 position and the penultimate valine (P’1) were acetylated in vitro. N-methionine excision (NME) was absent when compared with MeCP2-E1. MeCP2 colocalization to chromocenters was unaffected.~1/40.2%Unknown[13, 106, 136]
Methyl Binding Domain (MBD)T158M (T170M)Impaired stability of Asx-ST motif. Impaired binding to chromocenters due to reduced affinity for mCpG.~1/38.79%12 weeks[12, 78, 136, 137]
P152R/A (P164R/A)Impaired clustering of MeCP2 to chromocenters.Unknown1.5%Unknown[136, 138]
R133C (R145C)Impaired stability of 4th β-sheet and α-helix. Reduced binding to hmC (retained binding to mC). Impaired interaction with TCF20 complex. Reduced MeCP2-driven LLPS.~1/2 in the hippocampus; indistinguishable in cortex and cerebellum4.24%42 weeks[136, 137, 139–142]
R111G (R123G)Abolished binding to mC; disrupted nuclear localization; disrupted binding to TCF20 complex. Reduced MeCP2-driven LLPS.unknown>1%6 weeks[80, 136, 141–143]
R106W (R118W)Impaired stability of 2nd beta-sheet and reduced affinity to mCpG. Decreased MeCP2-driven LLPS.~1/32.76%10 weeks[136, 137, 142, 144–146]
Transcription Repression Domain (TRD)R294X (R306X)Increased binding to DNA. Clustering to chromocenters was unaffected.Indistinguishable6.1%35 weeks[106, 147–149]
NCoR/SMRT Interaction Domain (NID)R306C (R318C)Abolished interaction between NCoR/SMRT by disrupting TBLR1 binding. Disrupted binding to ATRX complex. Reduced HDAC activity in the forebrain of mice.Indistinguishable6.8%18 weeks[150–152]
Domain of MeCP2MutationMolecular phenotypeProtein levels compare to WT% of RTT casesMedian life span in ♂ miceReference
N-terminal Domain (NTD)MeCP2-E1- A2VMethionine at the P1 position and the penultimate valine (P’1) were acetylated in vitro. N-methionine excision (NME) was absent when compared with MeCP2-E1. MeCP2 colocalization to chromocenters was unaffected.~1/40.2%Unknown[13, 106, 136]
Methyl Binding Domain (MBD)T158M (T170M)Impaired stability of Asx-ST motif. Impaired binding to chromocenters due to reduced affinity for mCpG.~1/38.79%12 weeks[12, 78, 136, 137]
P152R/A (P164R/A)Impaired clustering of MeCP2 to chromocenters.Unknown1.5%Unknown[136, 138]
R133C (R145C)Impaired stability of 4th β-sheet and α-helix. Reduced binding to hmC (retained binding to mC). Impaired interaction with TCF20 complex. Reduced MeCP2-driven LLPS.~1/2 in the hippocampus; indistinguishable in cortex and cerebellum4.24%42 weeks[136, 137, 139–142]
R111G (R123G)Abolished binding to mC; disrupted nuclear localization; disrupted binding to TCF20 complex. Reduced MeCP2-driven LLPS.unknown>1%6 weeks[80, 136, 141–143]
R106W (R118W)Impaired stability of 2nd beta-sheet and reduced affinity to mCpG. Decreased MeCP2-driven LLPS.~1/32.76%10 weeks[136, 137, 142, 144–146]
Transcription Repression Domain (TRD)R294X (R306X)Increased binding to DNA. Clustering to chromocenters was unaffected.Indistinguishable6.1%35 weeks[106, 147–149]
NCoR/SMRT Interaction Domain (NID)R306C (R318C)Abolished interaction between NCoR/SMRT by disrupting TBLR1 binding. Disrupted binding to ATRX complex. Reduced HDAC activity in the forebrain of mice.Indistinguishable6.8%18 weeks[150–152]
Table 1

Classification of Rett-causing mutations based on their position in MeCP2 domains.

Domain of MeCP2MutationMolecular phenotypeProtein levels compare to WT% of RTT casesMedian life span in ♂ miceReference
N-terminal Domain (NTD)MeCP2-E1- A2VMethionine at the P1 position and the penultimate valine (P’1) were acetylated in vitro. N-methionine excision (NME) was absent when compared with MeCP2-E1. MeCP2 colocalization to chromocenters was unaffected.~1/40.2%Unknown[13, 106, 136]
Methyl Binding Domain (MBD)T158M (T170M)Impaired stability of Asx-ST motif. Impaired binding to chromocenters due to reduced affinity for mCpG.~1/38.79%12 weeks[12, 78, 136, 137]
P152R/A (P164R/A)Impaired clustering of MeCP2 to chromocenters.Unknown1.5%Unknown[136, 138]
R133C (R145C)Impaired stability of 4th β-sheet and α-helix. Reduced binding to hmC (retained binding to mC). Impaired interaction with TCF20 complex. Reduced MeCP2-driven LLPS.~1/2 in the hippocampus; indistinguishable in cortex and cerebellum4.24%42 weeks[136, 137, 139–142]
R111G (R123G)Abolished binding to mC; disrupted nuclear localization; disrupted binding to TCF20 complex. Reduced MeCP2-driven LLPS.unknown>1%6 weeks[80, 136, 141–143]
R106W (R118W)Impaired stability of 2nd beta-sheet and reduced affinity to mCpG. Decreased MeCP2-driven LLPS.~1/32.76%10 weeks[136, 137, 142, 144–146]
Transcription Repression Domain (TRD)R294X (R306X)Increased binding to DNA. Clustering to chromocenters was unaffected.Indistinguishable6.1%35 weeks[106, 147–149]
NCoR/SMRT Interaction Domain (NID)R306C (R318C)Abolished interaction between NCoR/SMRT by disrupting TBLR1 binding. Disrupted binding to ATRX complex. Reduced HDAC activity in the forebrain of mice.Indistinguishable6.8%18 weeks[150–152]
Domain of MeCP2MutationMolecular phenotypeProtein levels compare to WT% of RTT casesMedian life span in ♂ miceReference
N-terminal Domain (NTD)MeCP2-E1- A2VMethionine at the P1 position and the penultimate valine (P’1) were acetylated in vitro. N-methionine excision (NME) was absent when compared with MeCP2-E1. MeCP2 colocalization to chromocenters was unaffected.~1/40.2%Unknown[13, 106, 136]
Methyl Binding Domain (MBD)T158M (T170M)Impaired stability of Asx-ST motif. Impaired binding to chromocenters due to reduced affinity for mCpG.~1/38.79%12 weeks[12, 78, 136, 137]
P152R/A (P164R/A)Impaired clustering of MeCP2 to chromocenters.Unknown1.5%Unknown[136, 138]
R133C (R145C)Impaired stability of 4th β-sheet and α-helix. Reduced binding to hmC (retained binding to mC). Impaired interaction with TCF20 complex. Reduced MeCP2-driven LLPS.~1/2 in the hippocampus; indistinguishable in cortex and cerebellum4.24%42 weeks[136, 137, 139–142]
R111G (R123G)Abolished binding to mC; disrupted nuclear localization; disrupted binding to TCF20 complex. Reduced MeCP2-driven LLPS.unknown>1%6 weeks[80, 136, 141–143]
R106W (R118W)Impaired stability of 2nd beta-sheet and reduced affinity to mCpG. Decreased MeCP2-driven LLPS.~1/32.76%10 weeks[136, 137, 142, 144–146]
Transcription Repression Domain (TRD)R294X (R306X)Increased binding to DNA. Clustering to chromocenters was unaffected.Indistinguishable6.1%35 weeks[106, 147–149]
NCoR/SMRT Interaction Domain (NID)R306C (R318C)Abolished interaction between NCoR/SMRT by disrupting TBLR1 binding. Disrupted binding to ATRX complex. Reduced HDAC activity in the forebrain of mice.Indistinguishable6.8%18 weeks[150–152]

Regardless of the MeCP2-UBE3A-RTT connection and the critical role played by ubiquitin and ubiquitin-like proteins in the equilibrium between synapse physiology and neurodevelopment disorders [83], the role of MeCP2 ubiquitination and sumoylation has been less explored, and its involvement in the mutation phenotypes is less clear. In the last two sections, we will describe how these PTMs impact MeCP2.

MeCP2 ubiquitination

MeCP2 is subject to many PTMs, including phosphorylation, acetylation, methylation, O-glycosylation, ubiquitination, sumoylation [84, 85], lactylation [86] and N-methionine excision (NME) [13]. Among them, phosphorylation and ubiquitination are prominent, as many sites for both have been experimentally identified [87]. Yet, while the phosphorylation of MeCP2 has already been extensively investigated [25, 85, 87], the ubiquitination profile of MeCP2 (Fig. 2) has remained unexplored, and its role remains largely unknown.

MeCP2 ubiquitination and sumoylation. (A) Tertiary structure of human ubiquitin (1) and SUMO-1 (2). The red circles indicate the lysine sites forming K11 or K48 homotypic/heterotypic linkage polyubiquitin chains [133] that contribute to the signaling for proteasomal degradation. (B) Amino acid sequences of (1) human ubiquitin and (2) inactive human SUMO-1. The red circles are as in (A); the blue squared box is the di-glycine motif involved in the formation of an iso-peptide bond between the alpha-amino group of the C-terminal Gly and the epsilon amino group of the Lys that has been ubiquitinated in the target protein. The orange box in SUMO-1 is the sequence removed by SENP (sentrin/SUMO-specific protease) enzymes during the activation of SUMO previously to its conjugation with the target protein [134]. (C) Schematic representation of human MeCP2-E1 indicating the sites of ubiquitination (light blue pentagons). Empty filled correspond to predicted sites [96], solid/lightly shaded are those experimentally determined [87] the lightly shaded refer to the ones falling within a PEST domain. Sites of sumoylation are fuchsia hexagons. Sumoylation at mouse MeCP2-E1 K429 (mK429)/(mouse MeCP2-E2 K412) [114] corresponds to site K426 in human MeCP2-E1.The intensity of their filling colors is as in ubiquitination [96, 111]. The regions in red correspond to the two PEST domains, and the white numbers within each region correspond to their respective PEST find scores [96].
Figure 2

MeCP2 ubiquitination and sumoylation. (A) Tertiary structure of human ubiquitin (1) and SUMO-1 (2). The red circles indicate the lysine sites forming K11 or K48 homotypic/heterotypic linkage polyubiquitin chains [133] that contribute to the signaling for proteasomal degradation. (B) Amino acid sequences of (1) human ubiquitin and (2) inactive human SUMO-1. The red circles are as in (A); the blue squared box is the di-glycine motif involved in the formation of an iso-peptide bond between the alpha-amino group of the C-terminal Gly and the epsilon amino group of the Lys that has been ubiquitinated in the target protein. The orange box in SUMO-1 is the sequence removed by SENP (sentrin/SUMO-specific protease) enzymes during the activation of SUMO previously to its conjugation with the target protein [134]. (C) Schematic representation of human MeCP2-E1 indicating the sites of ubiquitination (light blue pentagons). Empty filled correspond to predicted sites [96], solid/lightly shaded are those experimentally determined [87] the lightly shaded refer to the ones falling within a PEST domain. Sites of sumoylation are fuchsia hexagons. Sumoylation at mouse MeCP2-E1 K429 (mK429)/(mouse MeCP2-E2 K412) [114] corresponds to site K426 in human MeCP2-E1.The intensity of their filling colors is as in ubiquitination [96, 111]. The regions in red correspond to the two PEST domains, and the white numbers within each region correspond to their respective PEST find scores [96].

Ubiquitin is a 76 amino acid long protein expressed in all eukaryotes. It is covalently attached to substrate proteins as a PTM, providing a signaling mechanism involved in versatile functions such as control of stability, cell-signaling, cell proliferation, and immune response, among others [88–90]. The process of attachment involves three enzymes: E1, ubiquitin-activating enzyme; E2, ubiquitin-conjugating enzyme; and E3, ubiquitin ligase [91], resulting in the formation of an isopeptide bond between the C-terminal glycine alpha carboxyl group of ubiquitin (Fig. 2B) and the epsilon amino of lysine in the target protein. Depending on the signaling pathway, a polyubiquitin chain can follow the mono ubiquitin [91]. The diverse functional outcomes of such PTMs are mediated by the nature of their structural diversity, spanning from mono-ubiquitination to a highly complex variety of polyubiquitinylated molecules, which additionally depends on the linkage type which can involve the internal lysine residues (K6, K11, K27, K29, K33, K48, and K63) to be of a homotypic and heterotypic linkage [92]. For example, a polyubiquitin chain attached by internal K11 and K48 heterotypic linkages binds and stimulates proteasomal degradation (Fig. 3) while the homotypic K11 linked polyubiquitin weakly associates with the proteasome machinery [93–95]. The 26S ubiquitin-proteasome system (26S UPS) is responsible for the breakdown of MeCP2 [78, 96]. The ubiquitin linkage type involved in protein degradation is likely the K48-linked homotypic or K48/K11 branched polymer (Figs 2A and3) as these linkages precede the recruitment of the 26S UPS [78, 97].

Structure of the PEST domains in MeCP2 in human and mice. The phosphorylation, ubiquitination and sumoylation of MeCP2 PEST domains are shown by red, green and blue circles, respectively. The yellow boxes display the N and C-terminus PEST amino acid sequences. Predicted (broken circles) [96] and experimentally determined [87] phosphorylation, ubiquitination and sumoylation sites are indicated. The AlphaFold [131] prediction of the helical polyproline type II (PPII) secondary structure arising from sequences containing repeated proline residues [135], which is characteristic of some PEST sequences [100], is displayed underneath the PEST-II domain.
Figure 3

Structure of the PEST domains in MeCP2 in human and mice. The phosphorylation, ubiquitination and sumoylation of MeCP2 PEST domains are shown by red, green and blue circles, respectively. The yellow boxes display the N and C-terminus PEST amino acid sequences. Predicted (broken circles) [96] and experimentally determined [87] phosphorylation, ubiquitination and sumoylation sites are indicated. The AlphaFold [131] prediction of the helical polyproline type II (PPII) secondary structure arising from sequences containing repeated proline residues [135], which is characteristic of some PEST sequences [100], is displayed underneath the PEST-II domain.

Initial attempts to determine the PTM profile of MeCP2 were made by stable transfection of human neuroblastoma cells SH-SY5Y with mouse MeCP2-E1 isoform. Tandem mass spectrometry (MS/MS) analysis revealed nine putative ubiquitylation sites at K24, K131, K142, K147, K245, K262, K256, K283 and K333 [87] of the human MeCP2-E1 isoform (Fig. 2C). The E3 ubiquitin ligase that ubiquitinates MeCP2 is the RING finger protein 4 (RNF4) (Fig. 4A) [98]. It is yet to be determined via which linkage type, and on which lysine residues within MeCP2, that RNF4 ubiquitinates [98]. However, RNF4 is a known recruiter for protein degradation [99]. It also triggers the removal of MeCP2 from methylated promoters and acts as a transcriptional activator [98] (Fig. 4A).

MeCP2 ubiquitination and PEST-mediated degradation. (A) RNF4 is the ubiquitin ligase of MeCP2 that acts as a transcriptional co-activator in a DNA methylation-dependent way, likely regulating its turnover [98]. (B) MeCP2 (red) contains two PEST domains (orange) [96]. Mutations in MeCP2 MBD (T158M [78]) impair the binding of the protein to methylated DNA, favoring its ubiquitination at the PEST domains and resulting in a decrease of the overall amount of protein as a result of degradation by the 26S UPS. The light brown/blue lollipop structures correspond to methylated CpG/CH (H = A, C or T) [8], respectively.
Figure 4

MeCP2 ubiquitination and PEST-mediated degradation. (A) RNF4 is the ubiquitin ligase of MeCP2 that acts as a transcriptional co-activator in a DNA methylation-dependent way, likely regulating its turnover [98]. (B) MeCP2 (red) contains two PEST domains (orange) [96]. Mutations in MeCP2 MBD (T158M [78]) impair the binding of the protein to methylated DNA, favoring its ubiquitination at the PEST domains and resulting in a decrease of the overall amount of protein as a result of degradation by the 26S UPS. The light brown/blue lollipop structures correspond to methylated CpG/CH (H = A, C or T) [8], respectively.

Indeed, most of what we know about MeCP2 ubiquitination has to do with its potential role in degradation by the 26S UPS [13, 78]. In 2009, we proposed a hypothesis that the MeCP2 turnover in the cell is partially regulated by PEST-mediated degradation. We identified two bona fide PEST (enriched in proline, glutamic acid, serine, and threonine) domains [96] (Fig. 2C) which are highly conserved across vertebrate evolution (Fig. 3 and Supplementary Fig. 1). These motifs are common in proteins with a short half-life. PEST domains have marginal stability due to their secondary structures, such as polyproline type II (PPII) and alpha helices (Fig. 3). The PEST hypothesis of protein degradation itself proposes that upon phosphorylation at a serine or threonine, the conformational stability of PEST domains becomes compromised [78, 100] and this instability is recognized by the E3 ubiquitin ligase, which ubiquitinates the flanking lysine residues via K48 or K11/48 linked polyubiquitination [100, 101] acting as a signal for the 26S UPS degradation (Fig. 4B). A phosphorylation site for each of the PEST domains at S92 [87, 102] and S413 [87] have been experimentally identified (Fig. 3), supporting the PEST mediated degradation hypothesis.

The ubiquitin-proteasome system is involved in recognizing the destabilizing N-terminal residues (N-degrons) following the N-terminal removal of methionine (NME) and N-terminal acetylation of the following alanine or valine as mentioned earlier. However, there is still a need to experimentally confirm if the E3 ubiquitin ligase that binds to N-degrons following NME and N-acetylation is RNF4 or a different E3 ubiquitin ligase.

Besides the putative role of ubiquitination in MeCP2 degradation, very little is known about the experimentally determined ubiquitination sites which fall outside the realm of PEST domains (Fig. 2C). One such residue is K283 within the TRD, which was heavily represented in the number of fragments of the MS/MS analysis [87]. In 2015, Pandey et al. reported that when K283 and a few other ubiquitin-targeted lysines are acetylated in cancerous cells by p300 acetyltransferase upon pharmacological inhibition of the histone deacetylase SIRT1 [103], it decreases the binding of ATP-dependant helicase (ATRX) and histone deacetylase 1 (HDAC1) to methylated gene promoters. Beyond cancer, MeCP2 interacts with ATRX [104], and there is a Rett frameshift mutation at K283 [105, 106]. Indeed, some of the lysines acetylated upon histone deacetylase treatment overlap with previously reported ubiquitinated sites [87]. Given the suggested competition between acetylation and ubiquitination of the same lysine residues and its role in regulating protein stability, such ubiquitination might, in this instance, play a stabilizing role [107]. With the observed overlap, it is possible that ubiquitination operates as a functional switch when presented as a monomer [108]. Altogether, there has been a paucity of investigation of MeCP2 ubiquitination and the importance of this PTM. Despite its high relevance in the critical homeostasis for MeCP2’s proper function, it remains largely unknown.

MeCP2 SUMOylation

As mentioned above, experimental evidence already exists for the existence of SUMOylation in MeCP2 [85, 109] (Fig. 2C). Yet, the functional relevance and consequences of this MeCP2 PTM are just starting to come to light. SUMO (Small Ubiquitin-related Modifier) proteins are a family of relatively small proteins of about 100 amino acids, and in humans they comprise four members (SUMO-1/SUMO-4). Except for SUMO-4, after their translation as inactive precursors, several C-terminal amino acids are removed by highly SUMO-specific proteases (SENPs) to expose conserved C-terminal Gly-Gly residues (Fig. 2B-2) that participate in the formation of the iso-peptide bond during their conjugation to the target protein. The resulting PTM usually occurs as a co-modification with phosphorylation [109]. The duality of sumoylation and phosphorylation plays an essential role in the cytoplasmic-nuclear translocation of MeCP2 during neuronal differentiation/maturation, as MeCP2 is also present in the neuronal cytosol before differentiation occurs [110].

The first MeCP2 sumoylation site reported was at K235 or K223 in E2 isoform, and it was found to be required for the recruitment of histone deacetylase complexes 1/2 [111], which together with Sin3a, was one of the first described MeCP2 interacting co-repressor complexes [112]. Moreover, such sumoylation is essential in MeCP2 repressor activity, as mutation of K235 to an arginine abolishes its mediated gene silencing in mouse primary cortical neurons. In addition, sumoylation at K235 is crucial in synaptic development in the central nervous system [111, 113].

Another sumoylation site is that of mouse MeCP2-E2 K412 [114] that corresponds to human MeCP2 K426 (Fig. 3), which falls within one of the PEST regions (Fig. 2C), and is mediated by E3 ligase PIAS. Interestingly, as mentioned earlier, this sumoylation takes place as a co-modification that involves the phosphorylation of MeCP2-E2 mouse S421 and T308 [114]. The E3 SUMO ligase protein inhibitor of activated STAT1 (PIAS1) is an essential component of sumoylation pathways. A model was proposed in which induction of PIASI K412 sumoylation by NMDA, IGF-1 and corticotropic releasing factor (CRF) stimuli in the brain enhances MeCP2 binding to methylated DNA. This, in turn, results in the release of CREB from the Sin3a-HDAC1 co-repressor complex, allowing it, in association with CBP, to interact with the Bdnf CRE promoter element and increase the expression of the gene [114] (Fig. 5). The model fits well with the notion that one of the roles of sumoylation is to regulate transcription factor activity by changing the interaction with DNA and chromatin [115]. Of interest to this review, the authors of the paper found that MeCP2 mutants (residues refer to E2 isoform) R106W, R133C, P152A, T158M, R306C, and P376R (Table 1) significantly decreased MeCP2 SUMOylation [116]. Moreover, transduction to the basolateral amygdala (BLA) of a conditional MeCP2 knockout (cKO) in mice with lenti-mRFP-MeCP2WT-SUMO1 fusion vector rescued the deficits of social interaction, and it improved their memory performance when compared to the cKO. All in all, the paper’s results by Tai et al. revealed an essential functional role of MeCP2 sumoylation in synaptic plasticity that is altered in RTT [114].

MeCP2 sumoylation. (A) MeCP2 binds to methylated DNA under normal physiological conditions. It is associated with the Sin3a-HDAC1 co-repressor complex and represses gene expression [112], thereby suppressing the interaction of CREB with the corticotropic releasing factor (CRE) of Bdnf in neurons and inhibiting its expression. (B) Upon stimulation by different factors and phosphorylation of MeCP2 (sites T308 and S421 of mouse MeCP2-E2), the protein becomes sumoylated at lysine 412 (mouse MeCP2-E2) by protein inhibitor of activated STAT 1 (PIAS1). As a result, the MeCP2 interaction with methylated DNA tightens (red halo), releasing CREB from the repressor complex and dissociation of Sin3a-HDAC1, allowing CREB in association with CBP to bind to the CRE in the promoter region of Bdnf and increasing its expression. [Figure adapted and modified from Tai et al. [114]].
Figure 5

MeCP2 sumoylation. (A) MeCP2 binds to methylated DNA under normal physiological conditions. It is associated with the Sin3a-HDAC1 co-repressor complex and represses gene expression [112], thereby suppressing the interaction of CREB with the corticotropic releasing factor (CRE) of Bdnf in neurons and inhibiting its expression. (B) Upon stimulation by different factors and phosphorylation of MeCP2 (sites T308 and S421 of mouse MeCP2-E2), the protein becomes sumoylated at lysine 412 (mouse MeCP2-E2) by protein inhibitor of activated STAT 1 (PIAS1). As a result, the MeCP2 interaction with methylated DNA tightens (red halo), releasing CREB from the repressor complex and dissociation of Sin3a-HDAC1, allowing CREB in association with CBP to bind to the CRE in the promoter region of Bdnf and increasing its expression. [Figure adapted and modified from Tai et al. [114]].

Of note, the mouse MeCP2-E2 K412 (MeCP2-E1 429) sumoylation site happens within one of the predicted PEST sequences (Figs 2C and3). As indicated by Heaton and Wilson, the ubiquitination and SUMOylation systems are likely to have many points of intersection and that their coordination contributes to regulated fundamental cellular events [117]. They can work in concert to lead to proteasomal degradation [118]. The mono and poly-SUMOylated forms of the centromeric proteins have been shown to play a critical role in centromere homeostasis [119]. It would be interesting to determine whether these PTMs, in conjunction with their ubiquitinated counterparts or independently, play a similar role in the critical homeostasis required for proper MeCP2 function [119].

Recently, HDAC4 has been shown to form a complex with MeCP2. Compared to HDAC1/2, which are members of class 1 of HDACs, HDAC4 is a member of class IIa [120], and its increased nuclear abundance impairs neuronal development and long-term memory [121]. In addition to regulating protein acetylation, HDAC4 functions as a cytoplasmic SUMO E3 ligase [122]. However, the molecular mechanism relating to HDAC4-mediated cytoplasmic MeCP2 sumoylation and the relevance of sumoylated MeCP2 HDAC in the nucleus needs additional investigation.

Future perspectives

As with intellectual disability and autism spectrum disorder, where the ubiquitin system (UbS) is one of the relevant cellular processes affected [123, 124], through this review it should also be evident that dysregulation of this system plays an equally critical role in RTT. To better understand the molecular mechanisms involved, in-depth studies are required. An important reason for the difficulty in addressing impaired UbS and SUMO pathways is that intermediate states of the ubiquitin and sumo are short-lived [125]. In this regard, recently developed powerful techniques, such as the chemical trapping strategies [126], enable site-selective targeting of E2-ubiquitin-conjugates to full-length protein substrates. These techniques are expedient for the study of E3 ligases, as they can trap the transient intermediates of ubiquitin ligase, and thus could prove to be practical for studying the short-lived intermediates of ubiquitin and SUMO pathways.

We have also described how some of the RTT-relevant MeCP2 mutations (Table 1) alter and, in several instances, decrease the levels of MeCP2 in a proteasome-dependent way involving ubiquitination. In this regard, the possibility of using therapeutics hijacking E3 ligases using the PROteolysis Targeting Chimera (PROTAC) approach [127, 128] should provide good therapeutic strategies for treatment in such conditions. Moreover, this technology might also provide essential insights into fundamental biological aspects of MeCP2 ubiquitination and sumoylation.

Despite the functional relevance of MeCP2 ubiquitination and sumoylation described above, a key challenge in advancing our knowledge on this topic resides in the low abundance and highly dynamic nature of these PTMs [129], and the lack of antibodies with enough specificity, particularly for ubiquitin. In this regard, approaches such as that of sequential ChIPseq for sumoylated MeCP2 in neurons [130] and modifications thereof prove to be an informative and invaluable tool to gain information on the occupancy of SUMOylated transcription and chromatin factors at specific genomic loci.

Acknowledgements

We are very thankful to Mikko Karttunen and Cecilia Chavez Garcia at the University of Western Ontario for kindly providing the image shown in Fig.1B. This paper was supported by the Center for Addition and Mental Health Foundation Discovery Fund grant to J.B.V. and J.A.

Conflict of interest statement: None declared.

Footnotes

The amino acid positions referred to in this paper are those of human MeCP2-E1 isoform which is the most abundant isoform in brain.

References

1.

Holde
 
KEV
.
Chromatin
.
NY
:
Springer
,
1988
.

2.

Singal
 
R
,
Ginder
 
GD
.
DNA methylation
.
Blood
 
1999
;
93
:
4059
70
.

3.

Allfrey
 
VG
,
Faulkner
 
R
,
Mirsky
 
AE
.
Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis
.
Proc Natl Acad Sci
 
1964
;
51
:
786
94
.

4.

Goldberg
 
AD
,
Allis
 
CD
,
Bernstein
 
E
.
Epigenetics: a landscape takes shape
.
Cell
 
2007
;
128
:
635
8
.

5.

Lewis
 
JD
,
Meehan
 
RR
,
Henzel
 
WJ
. et al.  
Purification, sequence, and cellular localization of a novel chromosomal protein that binds to methylated DNA
.
Cell
 
1992
;
69
:
905
14
.

6.

Guo
 
JU
,
Su
 
Y
,
Shin
 
JH
. et al.  
Distribution, recognition and regulation of non-CpG methylation in the adult mammalian brain
.
Nat Neurosci
 
2014
;
17
:
215
22
.

7.

Gabel
 
HW
,
Kinde
 
B
,
Stroud
 
H
. et al.  
Disruption of DNA-methylation-dependent long gene repression in Rett syndrome
.
Nature
 
2015
;
522
:
89
93
.

8.

He
 
Y
,
Ecker
 
JR
.
Non-CG methylation in the human genome
.
Annu Rev Genomics Hum Genet
 
2015
;
16
:
55
77
.

9.

Thambirajah
 
AA
,
Ng
 
MK
,
Frehlick
 
LJ
. et al.  
MeCP2 binds to nucleosome free (linker DNA) regions and to H3K9/H3K27 methylated nucleosomes in the brain
.
Nucleic Acids Res
 
2012
;
40
:
2884
97
.

10.

Lee
 
W
,
Kim
 
J
,
Yun
 
JM
. et al.  
MeCP2 regulates gene expression through recognition of H3K27me3
.
Nat Commun
 
2020
;
11
:
3140
.

11.

Martínez de Paz
 
A
,
Khajavi
 
L
,
Martin
 
H
. et al.  
MeCP2-E1 isoform is a dynamically expressed, weakly DNA-bound protein with different protein and DNA interactions compared to MeCP2-E2
.
Epigenetics Chromatin
 
2019
;
12
:
63
.

12.

Good
 
KV
,
Vincent
 
JB
,
Ausio
 
J
.
MeCP2: the genetic driver of Rett syndrome epigenetics
.
Front Genet
 
2021
;
12
:
620859
.

13.

Sheikh
 
TI
,
de
 
Paz
 
AM
,
Akhtar
 
S
. et al.  
MeCP2_E1 N-terminal modifications affect its degradation rate and are disrupted by the Ala2Val Rett mutation
.
Hum Mol Genet
 
2017
;
26
:
4132
41
.

14.

Itoh
 
M
,
Tahimic
 
CG
,
Ide
 
S
. et al.  
Methyl CpG-binding protein isoform MeCP2_e2 is dispensable for Rett syndrome phenotypes but essential for embryo viability and placenta development
.
J Biol Chem
 
2012
;
287
:
13859
67
.

15.

Kerr
 
B
,
Soto
 
CJ
,
Saez
 
M
. et al.  
Transgenic complementation of MeCP2 deficiency: phenotypic rescue of Mecp2-null mice by isoform-specific transgenes
.
Eur J Hum Genet
 
2012
;
20
:
69
76
.

16.

Ghosh
 
RP
,
Nikitina
 
T
,
Horowitz-Scherer
 
RA
. et al.  
Unique physical properties and interactions of the domains of methylated DNA binding protein 2
.
Biochemistry
 
2010
;
49
:
4395
410
.

17.

Dunker
 
AK
,
Lawson
 
JD
,
Brown
 
CJ
. et al.  
Intrinsically disordered protein
.
J Mol Graph Model
 
2001
;
19
:
26
59
.

18.

Adams
 
VH
,
McBryant
 
SJ
,
Wade
 
PA
. et al.  
Intrinsic disorder and autonomous domain function in the multifunctional nuclear protein, MeCP2
.
J Biol Chem
 
2007
;
282
:
15057
64
.

19.

Chavez-Garcia
 
C
,
Henin
 
J
,
Karttunen
 
M
.
Multiscale computational study of the conformation of the full-length intrinsically disordered protein MeCP2
.
J Chem Inf Model
 
2022
;
62
:
958
70
.

20.

Bah
 
A
,
Vernon
 
RM
,
Siddiqui
 
Z
. et al.  
Folding of an intrinsically disordered protein by phosphorylation as a regulatory switch
.
Nature
 
2015
;
519
:
106
9
.

21.

Chahrour
 
M
,
Jung
 
SY
,
Shaw
 
C
. et al.  
MeCP2, a key contributor to neurological disease, activates and represses transcription
.
Science
 
2008
;
320
:
1224
9
.

22.

Yasui
 
DH
,
Peddada
 
S
,
Bieda
 
MC
. et al.  
Integrated epigenomic analyses of neuronal MeCP2 reveal a role for long-range interaction with active genes
.
Proc Natl Acad Sci
 
2007
;
104
:
19416
21
.

23.

Bin Akhtar
 
G
,
Buist
 
M
,
Rastegar
 
M
.
MeCP2 and transcriptional control of eukaryotic gene expression
.
Eur J Cell Biol
 
2022
;
101
:
151237
.

24.

Claveria-Gimeno
 
R
,
Abian
 
O
,
Velazquez-Campoy
 
A
. et al.  
MeCP2… Nature’s wonder protein or Medicine’s most feared one?
 
Curr Gene Med Reports
 
2016
;
4
:
180
94
.

25.

Zhou
 
Z
,
Hong
 
EJ
,
Cohen
 
S
. et al.  
Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation
.
Neuron
 
2006
;
52
:
255
69
.

26.

Frasca
 
A
,
Spiombi
 
E
,
Palmieri
 
M
. et al.  
MECP2 mutations affect ciliogenesis: a novel perspective for Rett syndrome and related disorders
.
EMBO Mol Med
 
2020
;
12
:
e10270
.

27.

Gulmez Karaca
 
K
,
Brito
 
DVC
,
Zeuch
 
B
. et al.  
Adult hippocampal MeCP2 preserves the genomic responsiveness to learning required for long-term memory formation
.
Neurobiol Learn Mem
 
2018
;
149
:
84
97
.

28.

Martinez de Paz
 
A
,
Vicente Sanchez-Mut
 
J
,
Samitier-Marti
 
M
. et al.  
Circadian cycle-dependent MeCP2 and brain chromatin changes
.
PLoS One
 
2015
;
10
:
e0123693
.

29.

Pepe
 
G
,
Fioriniello
 
S
,
Marracino
 
F
. et al.  
Blood-brain barrier integrity is perturbed in a Mecp2-null mouse model of Rett syndrome
.
Biomol Ther
 
2023
;
13
:
606
.

30.

Vuu
 
YM
,
Roberts
 
CT
,
Rastegar
 
M
.
MeCP2 is an epigenetic factor that links DNA methylation with brain metabolism
.
Int J Mol Sci
 
2023
;
24
:
4218
.

31.

Buist
 
M
,
El Tobgy
 
N
,
Shevkoplyas
 
D
. et al.  
Differential sensitivity of the protein translation initiation machinery and mTOR Signaling to MECP2 gain- and loss-of-function involves MeCP2 isoform-specific homeostasis in the brain
.
Cell
 
2022
;
11
:
1422
.

32.

Rangasamy
 
S
,
Olfers
 
S
,
Gerald
 
B
. et al.  
Reduced neuronal size and mTOR pathway activity in the Mecp2 A140V Rett syndrome mouse model
.
F1000Res
 
2016
;
5
:
2269
.

33.

Gulmez Karaca
 
K
,
Brito
 
DVC
,
Oliveira
 
AMM
.
MeCP2: a critical regulator of chromatin in neurodevelopment and adult brain function
.
Int J Mol Sci
 
2019
;
20
:
4577
.

34.

Ausio
 
J
.
Role of MeCP2 in neurological disorders: current status and future perspectives
.
Epigenomic
 
2018
;
10
:
5
8
.

35.

LaSalle
 
JM
,
Goldstine
 
J
,
Balmer
 
D
. et al.  
Quantitative localization of heterogeneous methyl-CpG-binding protein 2 (MeCP2) expression phenotypes in normal and Rett syndrome brain by laser scanning cytometry
.
Hum Mol Genet
 
2001
;
10
:
1729
40
.

36.

Shahbazian
 
MD
,
Antalffy
 
B
,
Armstrong
 
DL
. et al.  
Insight into Rett syndrome: MeCP2 levels display tissue- and cell-specific differences and correlate with neuronal maturation
.
Hum Mol Genet
 
2002
;
11
:
115
24
.

37.

Ross
 
PD
,
Guy
 
J
,
Selfridge
 
J
. et al.  
Exclusive expression of MeCP2 in the nervous system distinguishes between brain and peripheral Rett syndrome-like phenotypes
.
Hum Mol Genet
 
2016
;
25
:
4389
404
.

38.

Xiang
 
Z
,
Zhou
 
Q
,
Hu
 
M
. et al.  
MeCP2 epigenetically regulates alpha-smooth muscle actin in human lung fibroblasts
.
J Cell Biochem
 
2020
;
121
:
3616
25
.

39.

Vashi
 
N
,
Ackerley
 
C
,
Post
 
M
. et al.  
Aberrant lung lipids cause respiratory impairment in a Mecp2-deficient mouse model of Rett syndrome
.
Hum Mol Genet
 
2021
;
30
:
2161
76
.

40.

Fu
 
X
,
Fu
 
P
,
Yang
 
T
. et al.  
Homeobox A9 is a novel mediator of vascular smooth muscle cell phenotypic switching and proliferation by regulating methyl-CpG binding protein 2
.
Cell Signal
 
2023
;
108
:
110695
.

41.

Ausio
 
J
.
MeCP2 and the enigmatic organization of brain chromatin. Implications for depression and cocaine addiction
.
Clin Epigenetics
 
2016
;
8
:
58
.

42.

Amir
 
RE
,
Van den Veyver
 
IB
,
Wan
 
M
. et al.  
Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2
.
Nat Genet
 
1999
;
23
:
185
8
.

43.

Rett
 
A
.
On a unusual brain atrophy syndrome in hyperammonemia in childhood
.
Wien Med Wochenschr
 
1966
;
116
:
723
6
.

44.

Cheadle
 
JP
,
Gill
 
H
,
Fleming
 
N
. et al.  
Long-read sequence analysis of the MECP2 gene in Rett syndrome patients: correlation of disease severity with mutation type and location
.
Hum Mol Genet
 
2000
;
9
:
1119
29
.

45.

Ribeiro
 
MC
,
MacDonald
 
JL
.
Sex differences in Mecp2-mutant Rett syndrome model mice and the impact of cellular mosaicism in phenotype development
.
Brain Res
 
2020
;
1729
:
146644
.

46.

Cooley Coleman
 
JA
,
Fee
 
T
,
Bend
 
R
. et al.  
Mosaicism of common pathogenic MECP2 variants identified in two males with a clinical diagnosis of Rett syndrome
.
Am J Med Genet A
 
2022
;
188
:
2988
98
.

47.

Zhang
 
Q
,
Yang
 
X
,
Wang
 
J
. et al.  
Genomic mosaicism in the pathogenesis and inheritance of a Rett syndrome cohort
.
Genet Med
 
2019
;
21
:
1330
8
.

48.

Van Esch
 
H
,
Bauters
 
M
,
Ignatius
 
J
. et al.  
Duplication of the MECP2 region is a frequent cause of severe mental retardation and progressive neurological symptoms in males
.
Am J Hum Genet
 
2005
;
77
:
442
53
.

49.

Kirk
 
EP
,
Malaty-Brevaud
 
V
,
Martini
 
N
. et al.  
The clinical variability of the MECP2 duplication syndrome: description of two families with duplications excluding L1CAM and FLNA
.
Clin Genet
 
2009
;
75
:
301
3
.

50.

Ramocki
 
MB
,
Peters
 
SU
,
Tavyev
 
YJ
. et al.  
Autism and other neuropsychiatric symptoms are prevalent in individuals with MeCP2 duplication syndrome
.
Ann Neurol
 
2009
;
66
:
771
82
.

51.

Ta
 
D
,
Downs
 
J
,
Baynam
 
G
. et al.  
A brief history of MECP2 duplication syndrome: 20-years of clinical understanding
.
Orphanet J Rare Dis
 
2022
;
17
:
131
.

52.

Klein
 
ME
,
Lioy
 
DT
,
Ma
 
L
. et al.  
Homeostatic regulation of MeCP2 expression by a CREB-induced microRNA
.
Nat Neurosci
 
2007
;
10
:
1513
4
.

53.

Ramocki
 
MB
,
Zoghbi
 
HY
.
Failure of neuronal homeostasis results in common neuropsychiatric phenotypes
.
Nature
 
2008
;
455
:
912
8
.

54.

Giandomenico
 
SL
,
Alvarez-Castelao
 
B
,
Schuman
 
EM
.
Proteostatic regulation in neuronal compartments
.
Trends Neurosci
 
2022
;
45
:
41
52
.

55.

Tai
 
HC
,
Schuman
 
EM
.
Ubiquitin, the proteasome and protein degradation in neuronal function and dysfunction
.
Nat Rev Neurosci
 
2008
;
9
:
826
38
.

56.

Skene
 
PJ
,
Illingworth
 
RS
,
Webb
 
S
. et al.  
Neuronal MeCP2 is expressed at near histone-octamer levels and globally alters the chromatin state
.
Mol Cell
 
2010
;
37
:
457
68
.

57.

Chen
 
CH
,
Cheng
 
MC
,
Huang
 
A
. et al.  
Detection of rare methyl-CpG binding protein 2 gene missense mutations in patients with schizophrenia
.
Front Genet
 
2020
;
11
:
476
.

58.

Glaze
 
DG
,
Percy
 
AK
,
Skinner
 
S
. et al.  
Epilepsy and the natural history of Rett syndrome
.
Neurology
 
2010
;
74
:
909
12
.

59.

Neupane
 
M
,
Clark
 
AP
,
Landini
 
S
. et al.  
MECP2 is a frequently amplified oncogene with a novel epigenetic mechanism that mimics the role of activated RAS in malignancy
.
Cancer Discov
 
2015
;
6
:
45
58
.

60.

Nejati-Koshki
 
K
,
Roberts
 
CT
,
Babaei
 
G
. et al.  
The epigenetic reader methyl-CpG-binding protein 2 (MeCP2) is an emerging oncogene in cancer biology
.
Cancer
 
2023
;
15
:
15102683
.

61.

Tong
 
D
,
Zhang
 
J
,
Wang
 
X
. et al.  
MeCP2 facilitates breast cancer growth via promoting ubiquitination-mediated P53 degradation by inhibiting RPL5/RPL11 transcription
.
Oncogene
 
2020
;
9
:
56
.

62.

Jiang
 
W
,
Liang
 
YL
,
Liu
 
Y
. et al.  
MeCP2 inhibits proliferation and migration of breast cancer via suppression of epithelial-mesenchymal transition
.
J Cell Mol Med
 
2020
;
24
:
7959
67
.

63.

Samaco
 
RC
,
Hogart
 
A
,
LaSalle
 
JM
.
Epigenetic overlap in autism-spectrum neurodevelopmental disorders: MECP2 deficiency causes reduced expression of UBE3A and GABRB3
.
Hum Mol Genet
 
2005
;
14
:
483
92
.

64.

Lehman
 
NL
.
The ubiquitin proteasome system in neuropathology
.
Acta Neuropathol
 
2009
;
118
:
329
47
.

65.

Rodrigues
 
DC
,
Mufteev
 
M
,
Weatheritt
 
RJ
. et al.  
Shifts in ribosome engagement impact key gene sets in neurodevelopment and ubiquitination in Rett syndrome
.
Cell Rep
 
2020
;
30
:
4179
4196.e11
.

66.

Haase
 
F
,
Gloss
 
BS
,
Tam
 
PPL
. et al.  
WGCNA identifies translational and proteasome-ubiquitin dysfunction in Rett syndrome
.
Int J Mol Sci
 
2021
;
22
:9954. https://doi.org/10.3390/ijms22189954.

67.

Eagleson
 
KL
,
Levitt
 
P
.
Alterations in the proteome of developing neocortical synaptosomes in the absence of MET signaling revealed by comparative proteomics
.
Dev Neurosci
 
2023
;
45
:
126
38
.

68.

Lopez
 
SJ
,
Segal
 
DJ
,
LaSalle
 
JM
.
UBE3A: an E3 ubiquitin ligase with genome-wide impact in neurodevelopmental disease
.
Front Mol Neurosci
 
2018
;
11
:
476
.

69.

Qiu
 
Z
,
Cheng
 
J
.
The role of calcium-dependent gene expression in autism spectrum disorders: lessons from MeCP2, Ube3a and beyond
.
Neurosignals
 
2010
;
18
:
72
81
.

70.

Makedonski
 
K
,
Abuhatzira
 
L
,
Kaufman
 
Y
. et al.  
MeCP2 deficiency in Rett syndrome causes epigenetic aberrations at the PWS/AS imprinting center that affects UBE3A expression
.
Hum Mol Genet
 
2005
;
14
:
1049
58
.

71.

Jordan
 
C
,
Francke
 
U
.
Ube3a expression is not altered in Mecp2 mutant mice
.
Hum Mol Genet
 
2006
;
15
:
2210
5
.

72.

Kim
 
S
,
Chahrour
 
M
,
Ben-Shachar
 
S
. et al.  
Ube3a/E6AP is involved in a subset of MeCP2 functions
.
Biochem Biophys Res Commun
 
2013
;
437
:
67
73
.

73.

Pecorelli
 
A
,
Leoni
 
G
,
Cervellati
 
F
. et al.  
Genes related to mitochondrial functions, protein degradation, and chromatin folding are differentially expressed in lymphomonocytes of Rett syndrome patients
.
Mediat Inflamm
 
2013
;
2013
:
137629
.

74.

Sbardella
 
D
,
Tundo
 
GR
,
Cunsolo
 
V
. et al.  
Defective proteasome biogenesis into skin fibroblasts isolated from Rett syndrome subjects with MeCP2 non-sense mutations
.
Biochim Biophys Acta Mol basis Dis
 
2020
;
1866
:
165793
.

75.

Yusufzai
 
TM
,
Wolffe
 
AP
.
Functional consequences of Rett syndrome mutations on human MeCP2
.
Nucleic Acids Res
 
2000
;
28
:
4172
9
.

76.

Chen
 
RZ
,
Akbarian
 
S
,
Tudor
 
M
. et al.  
Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice
.
Nat Genet
 
2001
;
27
:
327
31
.

77.

Stuss
 
DP
,
Cheema
 
M
,
Ng
 
MK
. et al.  
Impaired in vivo binding of MeCP2 to chromatin in the absence of its DNA methyl-binding domain
.
Nucleic Acids Res
 
2013
;
41
:
4888
900
.

78.

Lamonica
 
JM
,
Kwon
 
DY
,
Goffin
 
D
. et al.  
Elevating expression of MeCP2 T158M rescues DNA binding and Rett syndrome-like phenotypes
.
J Clin Invest
 
2017
;
127
:
1889
904
.

79.

Kucukkal
 
TG
,
Yang
 
Y
,
Uvarov
 
O
. et al.  
Impact of Rett syndrome mutations on MeCP2 MBD stability
.
Biochemistry
 
2015
;
54
:
6357
68
.

80.

Yang
 
Y
,
Kucukkal
 
TG
,
Li
 
J
. et al.  
Binding analysis of methyl-CpG binding domain of MeCP2 and Rett syndrome mutations
.
ACS Chem Biol
 
2016
;
11
:
2706
15
.

81.

Polevoda
 
B
,
Sherman
 
F
.
Nalpha -terminal acetylation of eukaryotic proteins
.
J Biol Chem
 
2000
;
275
:
36479
82
.

82.

Meinnel
 
T
,
Serero
 
A
,
Giglione
 
C
.
Impact of the N-terminal amino acid on targeted protein degradation
.
Biol Chem
 
2006
;
387
:
839
51
.

83.

Folci
 
A
,
Mirabella
 
F
,
Fossati
 
M
.
Ubiquitin and ubiquitin-like proteins in the critical equilibrium between synapse physiology and intellectual disability
.
eNeuro
 
2020
;
7
:
ENEURO.0137
20.2020
.

84.

Ausió
 
J
,
de
 
Paz
 
A
,
Esteller
 
M
.
MeCP2: the long trip from a chromatin protein to neurological disorders
.
Trends Mol Med
 
2014
;
20
:
487
98
.

85.

Bellini
 
E
,
Pavesi
 
G
,
Barbiero
 
I
. et al.  
MeCP2 post-translational modifications: a mechanism to control its involvement in synaptic plasticity and homeostasis?
 
Front Cell Neurosci
 
2014
;
8
:
236
.

86.

Wang
 
Y
,
Chen
 
L
,
Zhang
 
M
. et al.  
Exercise-induced endothelial Mecp2 lactylation suppresses atherosclerosis via the Ereg/MAPK signalling pathway
.
Atherosclerosis
 
2023
;
375
:
45
58
.

87.

Gonzales
 
ML
,
Adams
 
S
,
Dunaway
 
KW
. et al.  
Phosphorylation of distinct sites in MeCP2 modifies cofactor associations and the dynamics of transcriptional regulation
.
Mol Cell Biol
 
2012
;
32
:
2894
903
.

88.

Pickart
 
CM
.
Back to the future with ubiquitin
.
Cell
 
2004
;
116
:
181
90
.

89.

Pickart
 
CM
,
Eddins
 
MJ
.
Ubiquitin: structures, functions, mechanisms
.
Biochim Biophys Acta
 
2004
;
1695
:
55
72
.

90.

Pickart
 
CM
,
Fushman
 
D
.
Polyubiquitin chains: polymeric protein signals
.
Curr Opin Chem Biol
 
2004
;
8
:
610
6
.

91.

Hershko
 
A
.
Lessons from the discovery of the ubiquitin system
.
Trends Biochem Sci
 
1996
;
21
:
445
9
.

92.

Callis
 
J
.
The ubiquitination machinery of the ubiquitin system
.
Arabidop B
 
2014
;
12
:
e0174
.

93.

Grice
 
GL
,
Lobb
 
IT
,
Weekes
 
MP
. et al.  
The proteasome distinguishes between heterotypic and homotypic lysine-11-linked Polyubiquitin chains
.
Cell Rep
 
2015
;
12
:
545
53
.

94.

Nath
 
D
,
Shadan
 
S
.
The ubiquitin system
.
Nature
 
2009
;
458
:
421
.

95.

van
 
Wijk
 
SJ
,
Fulda
 
S
,
Dikic
 
I
. et al.  
Visualizing ubiquitination in mammalian cells
.
EMBO Rep
 
2019
;
20
:
e46520
.

96.

Thambirajah
 
AA
,
Eubanks
 
JH
,
Ausio
 
J
.
MeCP2 post-translational regulation through PEST domains: two novel hypotheses: potential relevance and implications for Rett syndrome
.
BioEssays
 
2009
;
31
:
561
9
.

97.

Yau
 
RG
,
Doerner
 
K
,
Castellanos
 
ER
. et al.  
Assembly and function of heterotypic ubiquitin chains in cell-cycle and protein quality control
.
Cell
 
2017
;
171
:
918
933.e20
.

98.

Wang
 
Y
.
RING finger protein 4 (RNF4) derepresses gene expression from DNA methylation
.
J Biol Chem
 
2014
;
289
:
33808
13
.

99.

Ward
 
CC
,
Kleinman
 
JI
,
Brittain
 
SM
. et al.  
Covalent ligand screening uncovers a RNF4 E3 ligase recruiter for targeted protein degradation applications
.
ACS Chem Biol
 
2019
;
14
:
2430
40
.

100.

Garcia-Alai
 
MM
,
Gallo
 
M
,
Salame
 
M
. et al.  
Molecular basis for phosphorylation-dependent, PEST-mediated protein turnover
.
Structure
 
2006
;
14
:
309
19
.

101.

Rogers
 
S
,
Wells
 
R
,
Rechsteiner
 
M
.
Amino acid sequences common to rapidly degraded proteins: the PEST hypothesis
.
Science
 
1986
;
234
:
364
8
.

102.

Tao
 
J
,
Hu
 
K
,
Chang
 
Q
. et al.  
Phosphorylation of MeCP2 at serine 80 regulates its chromatin association and neurological function
.
Proc Natl Acad Sci
 
2009
;
106
:
4882
7
.

103.

Pandey
 
S
,
Simmons
 
GE
 Jr
,
Malyarchuk
 
S
. et al.  
A novel MeCP2 acetylation site regulates interaction with ATRX and HDAC1
.
Genes Cancer
 
2015
;
6
:
408
21
.

104.

Nan
 
X
,
Hou
 
J
,
Maclean
 
A
. et al.  
Interaction between chromatin proteins MECP2 and ATRX is disrupted by mutations that cause inherited mental retardation
.
Proc Natl Acad Sci
 
2007
;
104
:
2709
14
.

105.

Krishnaraj
 
R
,
Ho
 
G
,
Christodoulou
 
J
.
RettBASE: Rett syndrome database update
.
Hum Mutat
 
2017
;
38
:
922
31
.

106.

Ehrhart
 
F
,
Jacobsen
 
A
,
Rigau
 
M
. et al.  
A catalogue of 863 Rett-syndrome-causing MECP2 mutations and lessons learned from data integration
.
Sci Data
 
2021
;
8
:
10
.

107.

Gronroos
 
E
,
Hellman
 
U
,
Heldin
 
CH
. et al.  
Control of Smad7 stability by competition between acetylation and ubiquitination
.
Mol Cell
 
2002
;
10
:
483
93
.

108.

Jason
 
LJ
,
Moore
 
SC
,
Lewis
 
JD
. et al.  
Histone ubiquitination: a tagging tail unfolds?
 
BioEssays
 
2002
;
24
:
166
74
.

109.

Hendriks
 
IA
,
Lyon
 
D
,
Young
 
C
. et al.  
Site-specific mapping of the human SUMO proteome reveals co-modification with phosphorylation
.
Nat Struct Mol Biol
 
2017
;
24
:
325
36
.

110.

Miyake
 
K
,
Nagai
 
K
.
Phosphorylation of methyl-CpG binding protein 2 (MeCP2) regulates the intracellular localization during neuronal cell differentiation
.
Neurochem Int
 
2007
;
50
:
264
70
.

111.

Cheng
 
J
,
Huang
 
M
,
Zhu
 
Y
. et al.  
SUMOylation of MeCP2 is essential for transcriptional repression and hippocampal synapse development
.
J Neurochem
 
2013
;
128
:
798
806
.

112.

Nan
 
X
,
Ng
 
HH
,
Johnson
 
CA
. et al.  
Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex
.
Nature
 
1998
;
393
:
386
9
.

113.

Cheng
 
TL
,
Qiu
 
Z
.
MeCP2: multifaceted roles in gene regulation and neural development
.
Neurosci Bull
 
2014
;
30
:
601
9
.

114.

Tai
 
DJ
,
Liu
 
YC
,
Hsu
 
WL
. et al.  
MeCP2 SUMOylation rescues Mecp2-mutant-induced behavioural deficits in a mouse model of Rett syndrome
.
Nat Commun
 
2016
;
7
:
10552
.

115.

Zhao
 
Q
,
Ma
 
Y
,
Li
 
Z
. et al.  
The function of SUMOylation and its role in the development of cancer cells under stress conditions: a systematic review
.
Stem Cells Int
 
2020
;
2020
:
1
16
.

116.

Hsu
 
WL
,
Ma
 
YL
,
Liu
 
YC
. et al.  
Restoring Wnt6 signaling ameliorates behavioral deficits in MeCP2 T158A mouse model of Rett syndrome
.
Sci Rep
 
2020
;
10
:
1074
.

117.

Wilson
 
VG
,
Heaton
 
PR
.
Ubiquitin proteolytic system: focus on SUMO
.
Expert Rev Proteomics
 
2008
;
5
:
121
35
.

118.

Seeler
 
JS
,
Dejean
 
A
.
SUMO and the robustness of cancer
.
Nat Rev Cancer
 
2017
;
17
:
184
97
.

119.

van den
 
Berg
 
SJW
,
Jansen
 
LET
.
SUMO control of centromere homeostasis
.
Front Cell Dev Biol
 
2023
;
11
:
1193192
.

120.

Seto
 
E
,
Yoshida
 
M
.
Erasers of histone acetylation: the histone deacetylase enzymes
.
Cold Spring Harb Perspect Biol
 
2014
;
6
:
a018713
.

121.

Main
 
P
,
Tan
 
WJ
,
Wheeler
 
D
. et al.  
Increased abundance of nuclear HDAC4 impairs neuronal development and long-term memory
.
Front Mol Neurosci
 
2021
;
14
:
616642
.

122.

Grozinger
 
CM
,
Schreiber
 
SL
.
Regulation of histone deacetylase 4 and 5 and transcriptional activity by 14-3-3-dependent cellular localization
.
Proc Natl Acad Sci
 
2000
;
97
:
7835
40
.

123.

Kasherman
 
MA
,
Premarathne
 
S
,
Burne
 
THJ
. et al.  
The ubiquitin system: a regulatory hub for intellectual disability and autism Spectrum disorder
.
Mol Neurobiol
 
2020
;
57
:
2179
93
.

124.

Furukawa
 
S
,
Nomura
 
J
,
Hanafusa
 
H
. et al.  
Germ-cell-specific transcriptome analysis illuminates the chromatin and ubiquitin pathway in autism spectrum disorders
.
Autism Res
 
2023
;
16
:
1101
10
.

125.

Hu
 
Q
,
Botuyan
 
MV
,
Mer
 
G
.
A chemical strategy to study transient states in substrate ubiquitylation by cryo-EM
.
Chem
 
2023
;
9
:
1069
71
.

126.

Ai
 
H
,
Tong
 
Z
,
Deng
 
Z
. et al.  
Synthetic E2-Ub-nucleosome conjugates for studying nucleosome ubiquitination
.
Chem
 
2023
;
9
:
1221
40
.

127.

Pettersson
 
M
,
Crews
 
CM
.
PROteolysis TArgeting chimeras (PROTACs) - past, present and future
.
Drug Discov Today Technol
 
2019
;
31
:
15
27
.

128.

Burslem
 
GM
,
Crews
 
CM
.
Proteolysis-targeting chimeras as therapeutics and tools for biological discovery
.
Cell
 
2020
;
181
:
102
14
.

129.

Salas-Lloret
 
D
,
Gonzalez-Prieto
 
R
.
Insights in Post-translational modifications: ubiquitin and SUMO
.
Int J Mol Sci
 
2022
;
23
:
3281
.

130.

Wu
 
T
,
Donohoe
 
ME
.
Sequential chromatin immunoprecipitation to detect SUMOylated MeCP2 in neurons
.
Biochem Biophys Rep
 
2016
;
5
:
374
8
.

131.

Jumper
 
J
,
Evans
 
R
,
Pritzel
 
A
. et al.  
Highly accurate protein structure prediction with AlphaFold
.
Nature
 
2021
;
596
:
583
9
.

132.

Schuster
 
MK
,
Graber
 
M
.
EMBOSS epestfind
.
2002
. https://emboss.bioinformatics.nl/cgi-bin/emboss/help/epestfind  
(accessed June 28, 2023)
.

133.

Dittmar
 
G
,
Winklhofer
 
KF
.
Linear ubiquitin chains: cellular functions and strategies for detection and quantification
.
Front Chem
 
2019
;
7
:
915
.

134.

Wilkinson
 
KA
,
Henley
 
JM
.
Mechanisms, regulation and consequences of protein SUMOylation
.
Biochem J
 
2010
;
428
:
133
45
.

135.

Adzhubei
 
AA
,
Sternberg
 
MJ
,
Makarov
 
AA
.
Polyproline-II helix in proteins: structure and function
.
J Mol Biol
 
2013
;
425
:
2100
32
.

136.

Sheikh
 
TI
,
Ausio
 
J
,
Faghfoury
 
H
. et al.  
From function to phenotype: impaired DNA binding and clustering correlates with clinical severity in males with missense mutations in MECP2
.
Sci Rep
 
2016
;
6
:
38590
.

137.

Ballestar
 
E
,
Yusufzai
 
T
,
Wolffe
 
A
.
Effects of Rett syndrome mutations of the methyl-CpG binding domain of the transcriptional repressor MeCP2 on selectivity for association with methylated DNA
.
Biochemistry
 
2000
;
39
:
7100
6
.

138.

Christodoulou
 
J
,
Grimm
 
A
,
Maher
 
T
. et al.  
RettBASE: the IRSA MECP2 variation database-a new mutation database in evolution
.
Hum Mutat
 
2003
;
21
:
466
72
.

139.

Brown
 
K
,
Selfridge
 
J
,
Lagger
 
S
. et al.  
The molecular basis of variable phenotypic severity among common missense mutations causing Rett syndrome
.
Hum Mol Genet
 
2016
;
25
:
558
70
.

140.

Vermudez
 
SAD
,
Gogliotti
 
RG
,
Arthur
 
B
. et al.  
Profiling beneficial and potential adverse effects of MeCP2 overexpression in a hypomorphic Rett syndrome mouse model
.
Genes Brain Behav
 
2022
;
21
:
e12752
.

141.

Zhou
 
J
,
Hamdan
 
H
,
Yalamanchili
 
HK
. et al.  
Disruption of MeCP2-TCF20 complex underlies distinct neurodevelopmental disorders
.
Proc Natl Acad Sci
 
2022
;
119
:
e2119078119
.

142.

Wang
 
L
,
Hu
 
M
,
Zuo
 
MQ
. et al.  
Rett syndrome-causing mutations compromise MeCP2-mediated liquid-liquid phase separation of chromatin
.
Cell Res
 
2020
;
30
:
393
407
.

143.

Heckman
 
LD
,
Chahrour
 
MH
,
Zoghbi
 
HY
.
Rett-causing mutations reveal two domains critical for MeCP2 function and for toxicity in MECP2 duplication syndrome mice
.
Elife
 
2014
;
3
:
e02676
.

144.

Johnson
 
BS
,
Zhao
 
YT
,
Fasolino
 
M
. et al.  
Biotin tagging of MeCP2 in mice reveals contextual insights into the Rett syndrome transcriptome
.
Nat Med
 
2017
;
23
:
1203
14
.

145.

Ortega-Alarcon
 
D
,
Claveria-Gimeno
 
R
,
Vega
 
S
. et al.  
Molecular context-dependent effects induced by Rett syndrome-associated mutations in MeCP2
.
Biomol Ther
 
2020
;
10
:
1533
.

146.

Stefanelli
 
G
,
Gandaglia
 
A
,
Costa
 
M
. et al.  
Brain phosphorylation of MeCP2 at serine 164 is developmentally regulated and globally alters its chromatin association
.
Sci Rep
 
2016
;
6
:
28295
.

147.

Merritt
 
JK
,
Collins
 
BE
,
Erickson
 
KR
. et al.  
Pharmacological read-through of R294X Mecp2 in a novel mouse model of Rett syndrome
.
Hum Mol Genet
 
2020
;
29
:
2461
70
.

148.

Neul
 
JL
,
Fang
 
P
,
Barrish
 
J
. et al.  
Specific mutations in methyl-CpG-binding protein 2 confer different severity in Rett syndrome
.
Neurology
 
2008
;
70
:
1313
21
.

149.

Caffarelli
 
C
,
Gonnelli
 
S
,
Pitinca
 
MDT
. et al.  
Methyl-CpG-binding protein 2 (MECP2) mutation type is associated with bone disease severity in Rett syndrome
.
BMC Med Genet
 
2020
;
21
:
21
.

150.

Kruusvee
 
V
,
Lyst
 
MJ
,
Taylor
 
C
. et al.  
Structure of the MeCP2-TBLR1 complex reveals a molecular basis for Rett syndrome and related disorders
.
Proc Natl Acad Sci
 
2017
;
114
:
E3243
50
.

151.

Lyst
 
MJ
,
Ekiert
 
R
,
Ebert
 
DH
. et al.  
Rett syndrome mutations abolish the interaction of MeCP2 with the NCoR/SMRT co-repressor
.
Nat Neurosci
 
2013
;
16
:
898
902
.

152.

Boxer
 
LD
,
Renthal
 
W
,
Greben
 
AW
. et al.  
MeCP2 represses the rate of transcriptional initiation of highly methylated long genes
.
Mol Cell
 
2020
;
77
:
294
309.e9
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/pages/standard-publication-reuse-rights)

Supplementary data