Next Article in Journal
Determinants for Activation of the Ion Channel TRPV3 by Weak Acids
Previous Article in Journal
Are an Aging Gut and a Decrease in Butyrate Production the Reasons for Atherosclerosis?
Previous Article in Special Issue
Exploring Salivary Biomarkers in Pediatric Obesity: A Scoping Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MECP2 Dysfunction in Rett Syndrome: Molecular Mechanisms, Multisystem Pathology, and Emerging Therapeutic Strategies

1
Department of Stem Cell and Regenerative Biotechnology, KU Institute of Technology, Konkuk University, Seoul 05029, Republic of Korea
2
3D Tissue Culture Research Center, Konkuk University, Seoul 05029, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(17), 8277; https://doi.org/10.3390/ijms26178277
Submission received: 15 July 2025 / Revised: 14 August 2025 / Accepted: 25 August 2025 / Published: 26 August 2025
(This article belongs to the Special Issue Molecular Advances in Pediatric Diseases)

Abstract

Rett syndrome is a severe neurodevelopmental disorder that occurs primarily in females and is caused by mutations in the methyl-CpG-binding protein 2 (MECP2) gene located on the X chromosome. Though MECP2 acts as a representative transcriptional regulator and affects gene expression both directly and indirectly, a complete understanding of this disease and the treatment mechanism has not been established yet. MECP2 plays a particularly important role in synaptic development, neuronal maturation, and epigenetic regulation in the brain. In this study, we summarize the molecular structure of MECP2, mutation-specific pathogenesis, and the role of MECP2 in regulating chromatin remodeling, RNA splicing, and miRNA processing to provide a comprehensive understanding of Rett syndrome. Additionally, we describe abnormal phenotypes manifested in various brain regions and other tissues owing to MECP2 dysfunction. Finally, we discuss current and future therapeutic approaches, including AAV-based gene therapy, RNA editing, X chromosome reactivation, and pharmacological interventions. Understanding the diverse functions and pathological mechanisms of MECP2 provides an important foundation for developing targeted therapies for Rett syndrome.

1. Introduction

Rett syndrome is a genetic disease that mainly occurs in females and is caused by a mutation in the methyl-CpG-binding protein 2 (MECP2) gene located on the X chromosome (Xq28) in 95% cases [1]. In other cases, a Rett syndrome-like phenotype arises owing to mutations in the CDKL5 and FOXG1 genes [2,3]. Rett syndrome is regarded as one of the most common genetic causes of severe cognitive disability following Down syndrome, and the incidence rate is high (1 in 10,000–15,000) [4]. Representative symptoms include stereotypical hand movements without purpose, seizures, autonomic nervous system instability, and hyperventilation [5,6].
To date, 925 MECP2 variants have been identified, with 535 being pathogenic, and these can be confirmed in RettBASE [7]. Eight common pathogenic variants (R168X, R255X, R270X, R294X, R106W, R133C, T158M, and R306C) have been identified, with a C > T single-nucleotide change occurring in approximately 60–70% females with Rett syndrome [8,9,10].
Most patients with Rett syndrome have a de novo mutation in the paternally inherited X chromosome, which largely accounts for the predominance of the disorder in females [11,12,13]. Although rare, Rett syndrome can also occur in males with MECP2 mutations; however, these cases are typically associated with severe phenotypes, and the affected individuals often succumb to fatal encephalopathy before the age of two [14,15,16]. Even though MECP2 mutations can occur on both X chromosomes in females, such biallelic mutations cause embryonic lethality during fetal development in mouse models [17]. Therefore, patients with Rett syndrome typically carry a heterozygous MECP2 mutation on one of the two X chromosomes. The clinical manifestations of Rett syndrome are primarily ascribed to aberrations in synaptic structure and neuron function, which are regarded as central neuropathological mechanisms underlying the etiology of Rett syndrome.

2. X Chromosome Inactivation and MECP2 Mosaicism in Rett Syndrome Pathogenesis

Understanding the random X chromosome inactivation (XCI) that occurs during early embryonic development is crucial for understanding Rett syndrome phenotype severity. The choice of which X chromosome to inactivate occurs randomly in vivo in humans and mice [18]. Unlike in mice, where imprinted XCI initiates early during embryogenesis, canonical XCI does not occur in human preimplantation embryos. Instead, both X chromosomes remain transcriptionally active but exhibit reduced expression levels, a phenomenon referred to as X chromosome dampening [19]. Random XCI gradually occurs post-implantation, with one of the two X chromosomes undergoing inactivation [19,20]. Therefore, either the paternal or the maternal X chromosome becomes inactivated in the cells comprising the body, leading to the coexistence of both cell types [18,21]. Thus, patients with Rett syndrome include cells with a MECP2 mutation (MECP2-MUT) on the Xi (XaMECP2 XiMECP-MUT; Xa: active X chromosome, Xi: inactive X chromosome) and cells with a MECP2 mutation on the Xa (XaMECP2-MUT XiMECP2) (Figure 1A). The state in which these two types of cells are mixed is called mosaicism, and the more cells expressing mutant MECP2, the more severe the Rett syndrome phenotype appears [22] (Figure 1B).
Due to limited access to post-mortem human brain tissues and patient-derived neurons, mouse models have been widely employed to study MECP2 and Rett syndrome pathogenesis in vivo [23]. However, in mouse Rett syndrome model, females exhibit non-random XCI with preferential expression of WT Mecp2 allele. Mecp2−/y male mice are viable but display neurological impairments [17,24,25]. On the contrary, in human females with Rett syndrome, the severity of the symptom varies depending on mosaicism ratio between cells with XaMECP2 XiMECP-MUT and XaMECP2-MUT XiMECP2 through random XCI [26,27,28,29]. Moreover, human males carrying MECP2 mutation typically experience early lethality owing to severe encephalopathy (Figure 1B). Additionally, in mouse Rett syndrome models, symptom onset occurs in adulthood, whereas in humans, symptom onset occurs in childhood [17]. Although the Mecp2 mutant mouse model mimics the main characteristics of patients with Rett syndrome, it is evaluated as underrepresenting actual patients [30,31]. These differences may make it difficult to analyze the exact pathogenesis of Rett syndrome in mice. Therefore, in vitro studies using human pluripotent stem cells, derived from patients with Rett syndrome or genetically modified MECP2 mutation, offer a suitable platform for modeling Rett syndrome.

3. Molecular Mechanism of MECP2 Function

3.1. MECP2 Structure and Its Mechanism of Gene Expression Regulation

MECP2 is a major transcriptional regulator for neuronal maturation that binds to methylated DNA and acts as a global transcriptional regulator, modulating gene activation and repression, thereby influencing cellular phenotypes [5,32]. It also plays a role in regulating mRNA splicing and miRNA biogenesis [33,34,35,36]. MECP2 protein consist of (1) N-terminal domain (NTD), (2) methyl-CpG-binding domain (MBD), (3) intervening domain (ID), (4) transcriptional repression domain (TRD), (5) C-terminal domain (CTD), and (6) three AT-hooks (each located in the ID, TRD, and CTD) (Figure 2A) [37]. The MBD of MECP2 binds to DNA and regulates gene expression through various mechanisms (Figure 2B). First, MECP2 modulates chromatin compaction and DNA loop generation [38]. MECP2 binds to both methylated and non-methylated DNA and regulates genes by inducing chromatin compaction in a manner similar to histone H1 (Figure 2B) [39,40]. Furthermore, MECP2 attaches to the DLX5-DLX6 locus, an imprinted gene cluster, forming a DNA loop to induce biallelic expression (Figure 2B) [41]. Second, MECP2 regulates gene expression by interacting with co-repressors such as histone deacetylases (HDACs), switch-independent 3A (SIN3A), nuclear receptor co-repressor (NCoR), and silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) (Figure 2B) [42,43]. Located in the TRD of MECP2, the NCoR–SMRT interaction domain (NID) mediates MECP2-dependent repression of a broad set of genes [44]. Third, although MECP2 is primarily a transcriptional repressor, it can also activate transcription by activating the target gene promoter via cAMP-responsive element-binding protein 1 (CREB1) (Figure 2B) [45]. In the absence of MECP2, the number of downregulated genes exceeds that of upregulated genes, suggesting that many proteins are positively regulated by MECP2 [46]. Fourth, MECP2 can also bind to RNA, regulating alternative splicing (Figure 2B) [33,36]. The Gene ontology (GO) analysis of MECP2-interacting proteins revealed an enrichment in splicing-related functions, suggesting a role for MECP2 in the post-transcriptional gene expression regulation [33,47]. YB-1, which interacts with MECP2, is a Y-box transcription factor that regulates the transcription of various target genes and binds to mRNA to influence alternative splicing and translation (Figure 2B) [34,48]. Fifth, MECP2 directly interacts with DiGeorge syndrome critical region 8 (DGCR8) and prevents the assembly of the Drosha–DGCR8 complex, a crucial step in miRNA processing, leading to reduced miRNA production (Figure 2B) [36]. In Mecp2-null mice, miRNA production is significantly elevated in the hippocampus, suggesting a repressive role for MECP2 in miRNA biogenesis within this brain region [36,49].

3.2. MECP2 Mutations Affecting Gene Expression

To understand the pathology of Rett syndrome, the location of mutations in MECP2 affects the brain abnormality types and specific genes and proteins that are regulated should be recognized [50,51]. MECP2-mediated gene regulation is influenced by the specific location of the mutation (such as MBD, TRD, and null) [52] (Figure 2B). (1) In the case of R270X, where a mutation occurs at AT-hook 2 within the TRD and the MBD of MECP2 is intact, MECP2 cannot properly modify chromatin structure despite its preserved DNA-binding ability. The G273X mutant mouse, which has a distance of three amino acids from R270X at the end of AT-hook 2, exhibits a longer lifespan and more efficient chromatin compaction than the R270X mouse [53]. This evidence suggests that amino acids 270–272 in MECP2 are crucial for binding to the AT-rich DNA regions and that the AT-hook domain plays an important role in chromatin modification [53,54]. Furthermore, owing to premature termination, MECP2 lacks the NID, preventing it from binding to the NCoR–SMRT complex. (2) The MECP2 R306C mutation is a representative missense mutation that damages the interaction between MECP2 and NCoR–SMRT co-repressor, thus disrupting the inhibitory role [55]. In this case, the MBD remains intact and allows MECP2 to bind to chromatin; however, the mutation in the NID region impairs its interaction with co-repressor complexes, compromising transcriptional regulation. The resulting phenotype is milder than mutations that entirely abolish DNA binding [56]. (3) When a mutation occurs in the MBD, such as R106W, R133C, and T158M, MECP2 loses its DNA-binding ability and thus cannot regulate gene expression. In the case of R106W and R133C mutations, although the interactions with NID, CTD, and RNA polymerase II are normal, the mutant MECP2 fails to bind to the transcription start site (TSS) of DNA, extensively altering the expression of related genes [57]. Mutations in the MBD and NID, such as T158M and R306C, retain partial MECP2 function, as evidenced by the milder severity in patients with Rett syndrome [58,59]. Therefore, residual interactions with mSin3a, NCoR, and AT-hooks may contribute to this remaining function of MECP2 [53,60]. (4) Since MECP2 is not expressed in the null model, the regulatory mechanism is absent.

3.3. Post-Translational Modification Regulation by MECP2

3.3.1. Histone Deacetylation and Chromatin Remodeling via MECP2–HDAC Interactions

MECP2 interacts with DNA to form a complex with HDAC, regulating gene expression, and competes with histone H1 for nucleosome binding, contributing to chromatin structure and gene expression [39]. Alterations in the global chromatin state caused by MECP2 deficiency affect the expression patterns of crucial neuron-specific genes. Additionally, MECP2 deficiency hyperacetylates Histone H3, a crucial histone protein involved in the epigenetic regulation of gene expression through histone modification, in the cerebrum, cerebellum, and spleen of Mecp2308/Y mice [30,39]. H3 acetylation at lysine residues K9, K14, K18, K23, K27, and K36 can decondense the nucleosome structure, forming euchromatin and potentially activating previously repressed genes [61,62,63,64,65]. CREB–CBP and SIRT1 are representative examples of chromatin-modifying enzymes that function as histone acetyltransferase (HAT) and HDACs, respectively [66,67]. Taken together, MECP2 regulates gene expression in mature neurons through interactions with histone-modifying enzymes and alters chromatin structure through its histone H1-like role.

3.3.2. Histone Methylation

MECP2 is closely associated with histone methylation, as evidenced by its involvement in histone H3K9 methylation during IL6 regulation [68]. DNA methylation plays a more dominant role in IL6 silencing than histone deacetylation. Notably, MECP2 and H3K9 methylation are both enriched at methylated DNA regions, indicating that MECP2 binds to methylated DNA and induces H3K9 methylation to maintain a repressive chromatin state. Consistent with this, the brain cells showed higher DNA methylation levels than other tissues, which may enhance the binding affinity of MECP2 to regions marked by H3K9 and H3K27 methylation [69].
MECP2 was first shown to bind to 5-methylcytosine (5mC) through its MBD. However, subsequent studies have revealed that it also has high affinity for non-CpG methylation (mCH) and 5-hydroxymethylcytosine (5hmC) [70,71,72]. In addition, MECP2 preferentially binds to GC-rich DNA regions, and this binding becomes even stronger when histone H3K27me3 is present [73]. This suggests that histone methylation facilitates MECP2-mediated transcriptional regulation of gene expression.

3.3.3. DNA Methylation

MECP2 directly binds to Dnmt1 to form a complex that binds to hemimethylated DNA, thereby contributing to DNA methylation maintenance during replication [74]. In mouse embryonic stem cells (ESCs), despite the absence of Dnmt1, Dnmt3a, and Dnmt3b, Mecp2 continues to bind chromatin [75]. Since MECP2 can still associate with DNA without these DNA methyltransferases, whether MECP2 directly contributes to DNA methylation maintenance should be further studied.

3.3.4. MECP2 Phosphorylation and SUMOylation

Depending on the specific mutation type in MECP2, the phosphorylation pattern of MECP2 and its interaction with related proteins can vary. Phosphorylation mapping of MECP2 has identified Ser86, Ser274, Thr308, and Ser421 as critical sites [76]. Phosphorylation at these sites is induced by neuronal activity, brain-derived neurotrophic factor (BDNF), and cAMP signaling pathways, functioning as key epigenetic regulation mediators [76]. Specifically, BDNF induces Ser86 and Ser274 phosphorylation in MECP2, while Thr308 and Ser421 phosphorylation in MECP2 is mediated by calcium/calmodulin-dependent protein kinase IV (CaMKIV), which is activated by membrane depolarization [76,77,78]. Thr308 phosphorylation in MECP2 disrupts the interaction between the MECP2 NID and the NCoR complex, thereby inhibiting MECP2’s ability to repress transcription [44]. However, the MECP2 missense mutation R306C prevents Thr308 phosphorylation, making it epigenetically impossible to regulate specific genes [76]. In the T308A mutant mouse brain, the mRNA expression levels of Npas4 and Bdnf, which are essential for inhibitory synapse maturation and plasticity, were significantly decreased. This was accompanied by brain weight loss and motor coordination deficits. Interestingly, treating the T308A mouse with the GABAA receptor antagonist pentylenetetrazol (PTZ) reduced seizures [79,80].
Ser421 phosphorylation in MECP2 is closely associated with dendritic growth, spine maturation, and BDNF expression, which are crucial for development of neuronal dendrites and inhibitory synapses in the cortex. This event is also linked to activity-dependent regulation, playing an essential role in establishing connectivity within the nervous system [81,82].
Ser80 of Mecp2 is phosphorylated in resting neurons, but becomes dephosphorylated upon action potentials [83]. Active neurons exhibit Ser80 dephosphorylation but Ser421 phosphorylation. The absence of phosphorylation owing to Mecp2 Ser80 mutation increases the expression levels of Rab3d, Vamp3, and Igsf4b [84]. Rab3d plays a role in the vesicular release machinery [83,85], Vamp3 facilitates SNARE-dependent exocytosis in astrocytes [86], and Igsf4b is a nerve tissue-specific cell adhesion molecule crucial for axon–glia interaction in myelination [87], suggesting that Ser80 phosphorylation in Mecp2 plays a critical role in repressing the expression of genes involved in synaptic vesicle release, exocytosis, and axon–glia interactions, thereby regulating the neuronal activity and myelination. Ser164 phosphorylation in Mecp2 is regulated during neurodevelopment in the brain. This event significantly impairs the DNA-binding affinity of Mecp2 and influences its interaction with nucleosomes and chromatin [83,88]. These data indicate that Mecp2 phosphorylation may profoundly affect brain maturation processes.
Ser421 and Thr308 phosphorylation in MECP2 promotes its SUMOylation, which can be induced by NMDA, IGF-1, and corticotropic releasing factor (CRF) in the rat brain [89]. Mecp2 SUMOylation plays a critical role in enhancing Bdnf mRNA expression by releasing CREB from the repressor complex (MECP2–Sin3a–HDAC1–CREB) [90]. Furthermore, impairments in Mecp2 SUMOylation contribute to deficits in social interaction, long-term memory (also called long-term potentiation, LTP) and synaptic plasticity [89]. Among MECP2 mutations, SUMOylation is significantly reduced in R106W, R133C, P152A, T158M, R306C, and P3mecp76R [91]. The deficit in SUMOylation is attributed to reduced expression of IGF-2, WNT6, and WNT5b genes, aligning with findings in the etiology of patients with Rett syndrome [89,92,93].

4. Nervous Systems Affected by MECP2 Mutations

Since Rett syndrome predominantly affects the nervous system, this review explores the brain regions and associated cell types, including neurons and glial cells, that are impacted by Rett syndrome or MECP2 mutations. In addition, we discuss the role of MECP2 in neural development and its contribution to Rett syndrome pathophysiology.

4.1. MECP2 Function in Early Neurogenesis

Early studies on MECP2 primarily investigated its role in neuronal maturation, with functional studies focusing on postnatal processes such as synapse formation and dendritic development [81,94,95,96]. In general, MECP2 does not affect neural progenitor cells (NPCs) during development and is expressed more in the nuclei of differentiated neurons than in glial cells [97,98,99,100]. However, MECP2 influences the fate of neural stem cells (NSCs)/NPCs to differentiate into neurons and glial cells [101]. The absence of MECP2 in NSCs/NPCs results in deficits of mature miR-199a. Therefore, Smad1—normally repressed by miR-199a—is upregulated, which in turn activates the downstream BMP4–Smad1 signaling pathway, triggering the shift from neuronal to astrocytic differentiation from NSCs/NPCs [101,102]. Belichenko et al. also reported decreased neurons and increased glial cells in patients with Rett syndrome [103]. Therefore, MECP2 expression loss decreases the number of mature neurons during development and, instead, increases the number of glial cells.
However, MECP2 deficiency in NSCs/NPCs can affect the differentiation and proliferation by increasing miR-199a and miR-214 expression, thereby regulating the expression of downstream target proteins [102]. Both miR-199a and miR-214 are important in regulating early neural differentiation and prenatal neurogenesis. miR-199a targets and inhibits PAK4, a serine/threonine kinase regulated by Rho family GTPases that activates the mitogen-activated protein kinase signaling pathway. PAK4 inhibition subsequently suppresses the ERK pathway [104]. In the absence of MECP2, miR-214 increases. This miRNA targets and suppresses PTEN, a negative regulator of AKT signaling, thereby enhancing AKT activation [105,106]. In general, ERK promotes neural differentiation in embryonic brain development and AKT induces NPC proliferation and survival [107,108]. In vivo studies have also demonstrated increased miR-199b-5p expression in the brains of adult Mecp2-mutant mice [49]. Additionally, the BMP/Smad signaling pathway promotes primary miRNA (pri-miRNA) processing [109]. Moreover, Rett syndrome brain organoids showed elevated BMP4 mRNA expression levels [102]. Taken together, these findings indicate that MECP2 deficiency disrupts neural development by upregulating miRNAs that modulate ERK and AKT signaling pathways. Additionally, enhanced BMP/Smad signaling may promote miRNA processing, further influencing neural differentiation and proliferation.
The expressions of specific miRNAs, such as miR-199a, vary depending on the location and type of MECP2 mutation or deletion. For example, increased miR-199a was observed in 316C > T missense mutation and 705delG in MECP2 [102], but decreased miR-199a was observed in 806delG [101]. Further research on the regulation of additional miRNAs in the context of MECP2 mutation is needed.

4.2. Neuronal Defects in Cerebral Cortex

4.2.1. Clinical Alteration of Brain Structure in Rett Syndrome

The resting-state functional MRI (rs-fMRI) analysis demonstrated that the brain size of patients with Rett syndrome was smaller than that of healthy individuals [110]. The greatest reduction was observed in the frontal lobe, and notably, the dorsal attention network (DAN), which is composed of connections between the frontal and posterior parietal cortices, showed decreased functional connectivity [111,112]. Since DAN maturation is essential for developing cognitive abilities and concentration [113], abnormal connections within the DAN may decrease concentration [114]. Accordingly, patients with Rett syndrome exhibit impaired selective focus and concentration, which may be attributed to decreased functional connectivity of the DAN. Similarly, the reduced connectivity of the corpus callosum contributes to the decreased DAN functionality in humans belonging to the autism spectrum [110].

4.2.2. Clinical Alteration of Neurons in Rett Syndrome

Clinical characteristics of neurons in patients with Rett syndrome include smaller cell bodies, reduced dendritic arborization, short and sparse dendrites, and reduced phenotypes of excitatory glutamatergic, inhibitory GABAergic, and monoaminergic neurotransmission [52,115,116,117,118]. MECP2 is crucial for synapse maturation and neurotransmitter activity regulation in neurons [119,120]. Notably, reduced MECP2 expression has been associated with decreased dendritic complexity, as evidenced by reduced spine density and short spine length, which are critical for synaptic structure formation [121,122]. Moreover, the dendritic length of pyramidal neurons in the frontal motor cortex is significantly reduced in patients with Rett syndrome [119]. In particular, the dendrites of pyramidal neurons in layers 3 and 5 of the frontal, motor, and inferior temporal regions, as well as the basal dendrites of layer 4 in the subiculum, are selectively and significantly reduced [123,124]. In a very rare case, a clinical study described a male proband with a MECP2 mutation who died from central respiratory failure at 15 months of age. The brain was small, with reduced frontal and temporal lobes, a thin corpus callosum, and a marked reduction in synaptic vesicles in the cerebellum and spinal cord. As observed in female cases, the dendritic tree of pyramidal neurons was significantly reduced in cortical layers 3 and 5 of the frontal and temporal lobes [15].

4.2.3. MECP2-Regulated Genes Involved in Neuronal Function

MECP2 is a global transcriptional regulator in neurons. It exerts both repressive and activating effects on target genes that control synaptic transmission, neuronal development, and metabolic regulation. Disruption of MECP2 function in Rett syndrome alters excitatory and inhibitory neurotransmission, impairing neuronal network activity. These alterations are believed to underlie key features of the disorder, including cognitive, behavioral, and motor deficits.
  • Excitatory Neurotransmission-Related Genes
Key excitatory neurotransmission-related genes include BDNF, serum/glucocorticoid-regulated kinase 1 (SGK1), and IGFBP3. BDNF regulates neuronal growth and is considered an important neurite outgrowth and synapse formation regulator [125,126]. Mecp2 knockdown reduces BDNF expression at glutamatergic synapses, delaying neuronal growth and maturation, but neuronal growth can be restored upon introduction of BDNF [127].
SGK1 is important for learning, memory, synaptic plasticity in neurons of the hippocampus, and is closely related to neuron morphology [128]. When SGK1 is overexpressed in the hippocampus, the total number of mature spines in dentate gyrus increases, and when SGK1 is reduced, the number of spines in CA1 increases, affecting synaptic structure and function in a region-specific manner depending on SGK1 expression [129]. Similarly, increased SGK1 expression in neurons promotes cell survival and inhibits apoptosis [130,131]. However, increased SGK1 expression in glial cells leads to exacerbation of neuroinflammation and neuronal damage [132]. MECP2 directly and indirectly affects SGK1 expression [133]. In particular, the absence of MECP2 prevents its binding to the proximal-promotor region of SGK1, thereby failing to suppress the expression of SGK1 [134].
IGFBP3 is directly regulated by MECP2, and IGFBP3 overexpression owing to MECP2 deficiency is a hallmark of Rett syndrome in both mice and humans [135]. IGFBP3 is a binding protein for IGF-1 and IGF-2 and an important factor in cell growth and neuronal dendrite and axon elongation [136,137]. Thus, increased IGFBP3 expression inhibits neurodevelopment. For example, IGFBP3-transgenic mice exhibit delayed brain growth and abnormal dendritic extension of neuronal cells [138]. As IGFBP3 binds IGF-1 and IGF-1 mimetics have shown therapeutic benefits in Rett syndrome, targeting the IGF system may be a potential strategy for Rett syndrome (see Section 5.4).
  • Inhibitory Neurotransmission-Related Genes
Key inhibitory neurotransmission-related genes, such as DLX5 and GABRB3, are also transcriptionally regulated by MECP2. DLX5 regulates the expression of glutamate decarboxylase, a key enzyme for GABA synthesis, during the development of GABAergic neurons [139]. DLX5 expression is regulated by MECP2, and thus DLX5 is overexpressed in the absence of MECP2 [140]. However, DLX5 expression also varies among Mecp2-deficient mice and is not consistently upregulated in the prefrontal cortex [141]. Further studies should determine whether this phenomenon is consistent across different brain regions and animal models. GABRB3 encodes the β3 subunit of the GABA receptor, which is a key receptor that mediates fast inhibitory neurotransmission in the brain, and is critical for maintaining excitation and inhibition (E/I balance), which is a mechanism underlying seizures [142,143]. MECP2 positively regulates GABRB3 expression, and its deficiency contributes to E/I imbalance, a key symptom of Rett syndrome [142].
  • Other Genes Regulated by MECP2
FKBP5 regulates the function of glucocorticoid receptors and plays a key role in regulating stress hormones and synaptic plasticity. FKBP5 expression is repressed by MECP2 [133,144], and is overexpressed in the Rett syndrome mouse model [133]. Thus, MECP2 deficiency causes abnormal regulation of stress and glucocorticoid signals during brain development.
All four known members of the inhibitor of DNA binding/differentiation subfamily (ID1, ID2, ID3, and ID4) are targets of MECP2 [145]. In Mecp2-dificent mice, the expression of ID protein was increased [145]. ID proteins regulate the fate of NSCs by influencing transcriptional programs important for differentiation. Specifically, they inhibit the activity of basic helix–loop–helix (bHLH) transcription factors, which play a key role in initiating neuronal differentiation by activating neuron-specific gene expression. However, the ID protein itself lacks a DNA-binding domain and thus acts as a dominant-negative regulator by forming a dimer with bHLH transcription factors to prevent DNA binding. Therefore, increased ID protein expression in the absence of MECP2 promotes NSC self-renewal and proliferation but suppresses neuronal differentiation [146,147,148]. Thus, MECP2 deficiency enhances NSC proliferation via increased expression of ID proteins, but reduces differentiation into neurons, contributing to the symptoms of Rett syndrome [102].
FXYD1, which encodes a membrane protein that regulates Na+-K+ ATPase activity important for neural function, is directly repressed by MECP2 [149]. In both patients with Rett syndrome and Mecp2-deficient mice, FXYD1 is overexpressed in the prefrontal cortex, which reduced dendritic arborization and spine formation, decreased Na+-K+ ATPase activity, disrupted potassium homeostasis, and caused abnormal neuronal activity [149]. Interestingly, partial FXYD1 expression reduction by deleting one Fxyd1 allele in Mecp2-deficient mice reverses the defects in neuronal branching and potassium regulation [150].
Early postnatal UBE3A expression is essential for proper maturation of neural circuits, including those in the striatum and hippocampus [151]. UBE3A mutations cause Angelman syndrome, where UBE3A deficiency disrupts synaptic development, neural circuit formation, and brain function during critical developmental stages [152]. UBE3A expression is also reduced in patients with Rett syndrome and Mecp2-deficient mouse models [153,154]. UBE3A expression is indirectly upregulated by MECP2, and its levels are reduced in Rett syndrome. Thus, UBE3A deficiency owing to MECP2 deficiency can help explain the pathophysiological overlap between Rett syndrome and Angelman syndrome [154].
Additionally, reduced MECP2 expression significantly impairs not only neuronal gene transcription but also mitochondrial function, including basal respiration, maximal respiration rate, and electron transport chain activity in neurons [32]. In addition, MECP2 appears to be involved in multiple metabolic processes, including lipid, nucleotide, glucose, and energy metabolism, as demonstrated in Mecp2-null mouse models [155].
Therefore, further studies are needed to elucidate the specific genes and proteins regulated by MECP2. Recent advances in artificial intelligence (AI)-based structure prediction tools, such as AlphaFold, allow us to assess whether specific MECP2 variants cause up- or down-regulation of target proteins and to predict potential binding interactions between MECP2 and specific proteins [156,157]. Leveraging these AI models will be crucial for elucidating the molecular interactome of MECP2. Future research should integrate AI-driven predictions with experimental validation to determine mutation-specific alterations in protein expression and to identify previously unrecognized molecular targets of MECP2.

4.2.4. Neuronal Imbalance

In excitatory and inhibitory (E/I) neurons, MECP2 dysfunction causes structural deficits, including smaller soma size, shorter dendritic length, and fewer excitatory synapses. Such alterations ultimately impair E/I synaptic transmission [158].
MECP2 interacts with the Drosha complex, potentially affecting the processing of primary miR-199a (Pri-miR-199a) into precursor miR-199a (Pre-miR-199a). The pre-miR-199a produced in the nucleus undergoes processing in the cytoplasm to become mature miR-199a-5p/3p, which inhibits SIRT1, HIF1a, and PDE4D. These regulators negatively modulate mTOR signaling, which is linked to neurotransmission at excitatory synapses and neuronal cell growth. Therefore, reduced MECP2 decreased Drosha complex activity and reduced miR-199a levels, thereby enhancing mTOR inhibition and resulting in abnormal phenotypes of excitatory neurons, such as reduced soma size and decreased dendritic density [35].
Hdac1 and Hdac2 interact with Mecp2 and form a complex to bind to the promoter of Sap90/Psd95-associated protein 3 (Sapap3), a key component of excitatory synapse postsynaptic proteins [159,160]. Pre- and postnatal MECP2 loss in the forebrain causes excessive grooming, a stereotypic behavior commonly observed in Rett syndrome [160]. Mecp2 deletion or both Hdac1 and Hdac2 loss downregulates Sapap3, which leads to repetitive behavior. Reintroduction of Sapap3 expression successfully rescues these behavioral abnormalities [158].
Taken together, MECP2 regulates excitatory synapse function by controlling miR-199a processing via the Drosha complex and repressing Sapap3 expression through interaction with HDAC1/2. Its deficiency disrupts mTOR signaling and synaptic protein expression, leading to neuronal and behavioral abnormalities in Rett syndrome.

4.2.5. Neuronal Types Affected by MECP2 Mutations

Cortical GABAergic neurons exhibit approximately 50% higher MECP2 expression level than non-GABAergic neurons, suggesting that MECP2 deficiency has a more significant effect on GABAergic neurons than on other neurons [161]. MECP2 loss leads to modifications in inhibitory intensity within the cortex, hippocampus, and brainstem [162,163]. MECP2 deficiency in GABAergic neurons leads to the loss of multiple functions such as social behavior, learning, memory, motor function, stereotyped behaviors, and sensorimotor gating. MECP2 deficiency also reduces presynaptic glutamic acid decarboxylase 1 (Gad1; GAD67) and glutamic acid decarboxylase 2 (Gad2; GAD65) levels, thus decreasing GABA production [161].
MECP2 deficiency reduced neurotransmitter levels in both dopaminergic and serotonergic neurons [164]. The role of the serotonergic system has been emphasized in relation to anxiety, repetitive behavior, and hyperactivity [165]. Particularly, in patients with Rett syndrome, low levels of serotonin (5-HT) have been reported, and deficiencies in serotonergic neurotransmission are associated with apnea, anxiety, and repetitive behaviors [164].
Dysfunctional dopaminergic activity is characterized by rigidity and movement abnormalities [23,166]. Reduced dopamine levels owing to MECP2 deficiency result in motor abnormalities in both mice and humans [164,167].
Both GABA and glycine are involved in neuronal development and regeneration [168,169]. Particularly, glycinergic neurons play a key role in the spinal cord and are co-localized with GABAergic neurons in cerebellar cortical interneurons. In the medulla oblongata, glycinergic neurons contribute to dual inhibitory synaptic transmissions in conjunction with GABA, playing a crucial role in regulating vital brainstem functions such as respiration. A recent study using Rett syndrome mouse models suggested that impaired GABAergic signaling appears to increase reliance on glycine-mediated inhibition [170].

4.2.6. Cerebellar Cortex Dysfunction in Rett Syndrome

Cerebellar dysfunction and progressive cerebellar circuit loss are observed in Rett syndrome [110]. Rett syndrome is associated with Purkinje cell (PC) loss in the cerebellar cortex [171,172]. PCs are GABAergic and inhibitory, highly interconnected with up to 500 synaptic connections per cell, and contribute to motor coordination [173]. MECP2 deletion in PCs increases PTP1B, a small-conductance calcium-activated potassium channel, and decreases signaling through TrkB, the receptor for BDNF, reducing intrinsic excitability. This ultimately contributes to autistic-like behaviors and impaired vestibulo-cerebellar motor learning [174]. Patients with Rett syndrome exhibit reduced cerebrum–cerebellum connectivity than the general population, with disrupted functional connectivity between the occipital lobe and cerebellum being implicated in the pathophysiology of autism spectrum disorder and schizophrenia. Additionally, decreased cerebellum–cerebrum/corpus callosum connectivity is associated with various behavioral and motor dysfunction phenotypes [110]. These reports indicate that MECP2 deficiency impairs cerebellar circuits and connectivity, contributing to the motor and behavioral symptoms of Rett syndrome.

4.2.7. Hippocampal Dysfunction in Rett Syndrome

The hippocampus plays critical roles in learning and long-term memory formation [175]. In Mecp2-deficient mice, hippocampal neurons exhibit reduced dendritic complexity and spine density, accompanied by a decreased thickness of the CA1 region of hippocampus [176,177]. Furthermore, NMDA receptor maturation in the hippocampus is delayed, and synaptic plasticity is impaired [178]. Accordingly, patients with Rett syndrome exhibit reduced expression of metabotropic glutamate receptors, which are essential for learning and memory, than healthy individuals [179].
In Rett syndrome mouse models, theta-burst stimulation fails to induce LTP in CA1 hippocampal neurons. This impairment was accompanied by E/I imbalance and altered excitability in neurogliaform (NGF) interneurons, suggesting their involvement in Rett syndrome pathophysiology [180]. Additionally, Mecp2 deficiency significantly decreases the frequency of spontaneous miniature excitatory postsynaptic currents in hippocampal neurons [181].

4.2.8. Olfactory Bulb

Olfactory receptor neurons (ORNs) in mice are continuously replaced throughout life owing to postnatal neurogenesis [182]. NSCs located in the subventricular zone may contribute to this postnatal neurogenesis by migration through the rostral migratory stream to the olfactory bulb. MECP2 function loss results in an abnormal accumulation of immature ORNs and olfactory epithelium expansion [183,184]. Immature ORNs are characterized by neuron-specific tubulin (NST)-positive markers, while mature ORNs express olfactory marker protein (OMP). In typical conditions, the ratio of immature to mature ORNs remains relatively low (approximately 1.2–2), but this ratio is markedly elevated in Rett syndrome models (2.4–13.5). Additionally, OMP-positive ORN display abnormal dendritic morphology and dysmorphic cell bodies in patients with Rett syndrome [184].

4.2.9. Autonomous Nervous System

Rett syndrome is characterized by dysfunction and dysregulation of the autonomic nervous system (ANS), which is considered a major cause of sudden death [185,186]. Rett syndrome features various characteristic respiratory patterns, including hyperventilation, apnea, apneusis, Valsalva maneuvers, breath-holding, and rapid shallow breathing, consistent with autonomic dysregulation [187,188,189]. Furthermore, in patients with Rett syndrome, even breathing that seems normal is actually more irregular than in healthy individuals, with increased average inspiratory flow rates, reduced expiratory times, and elevated respiratory frequency [190]. The activation of serotonin type 1A (5-HT1A) receptor, a key receptor involved in the termination of inspiration and located in the brainstem, promotes regular breathing and plays a role in homeostatic CO2 regulation [191].
In Rett syndrome, reduced serotonin (5-HT) levels decrease the activation of 5-HT1A receptors in the brainstem, resulting in dysregulation of ANS and contributing to the occurrence of apneas [192,193]. Heart rate regulation is also impaired in Rett syndrome, as indicated by a shortened R–R interval, corresponding to the ventricular depolarization phase of the cardiac cycle, and a weakened association between respiratory cycles and heart rate variability. In addition, unlike healthy individuals whose heart rate decreases during breath-holding, individuals with Rett syndrome show an increase in heart rate [190].

4.3. Glial Cells Affected by MECP2 Mutations and Rett Syndrome

Astrocytes are the most abundant type of glial cells in the brain and nervous system [194]. They support neuronal functions and respond to brain and neuronal damage, thereby helping the brain to maintain normal function [195]. Astrocytes are also connected to numerous synaptic sites and play a role in regulating the number and function of synapses [196]. Therefore, owing to their extensive roles in the nervous system, astrocyte dysfunction can significantly contribute to Rett syndrome pathophysiology, either directly or by disrupting neuronal synaptic function and homeostasis.
Although MECP2 expression is relatively low in astrocytes, it exerts substantial regulatory effects on their function and fate determination. NSCs differentiated from Rett syndrome iPSCs, which were generated from the fibroblasts of patients with Rett syndrome, show reduced neuronal differentiation potential and instead tend to differentiate more into astrocytes compared to wild-type NSCs [197]. Mecp2-null mice show a significant increase in GFAP and S100β, which are major markers of astrocytes [198]. This suggests that MECP2 plays a regulatory role in the expression of astrocyte-specific genes. MECP2 binds to the methylated promoter region of GFAP to regulate its expression, and MECP2 deficiency leads to GFAP overexpression [199]. Furthermore, Mecp2 knockout or knockdown reduces astrocyte growth, and is proportional to the extent of Mecp2 expression reduction [200]. These findings indicate that Mecp2 deficiency or reduced expression can lead to astrocyte dysfunction, contributing to pathological outcomes.
Glutamate plays a crucial role in signal transmission by maintaining its proper levels [201]. When extracellular Glu concentration becomes excessively high, glutamate transporter EAAT1/GLAST and EAAT2/GLT-1 expression is suppressed [202]. Under normal conditions, EAAT1 and EAAT2 transport glutamate into astrocytes, where glutamine synthetase (GS) converts it into active neurotransmitters [202,203]. In Mecp2-deficient mice, elevated extracellular glutamate levels suppress EAAT1/EAAT2 expression and increase GS expression, resulting in dysregulation of extracellular glutamate concentration [204]. This demonstrates that Mecp2 deficiency leads to abnormal regulation of the glutamate metabolic pathway in astrocytes, contributing to Rett syndrome pathology.
Interestingly, long-term co-culture of Mecp2-null astrocytes with wild-type astrocytes reduced Mecp2 expression in the wild-type astrocytes. This non-cell-autonomous effect is mediated by Connexin-43 (Cx-43)-based gap junctions [200]. When these gap junctions are blocked by Cx-43 siRNA, this effect is diminished, suggesting that targeting gap junctions might be a potential therapeutic strategy for Rett syndrome phenotypes.
In neurons and glial cells of Rett syndrome cerebral organoids, mitochondrial length and area were reduced compared to those in the control group, and the alteration was particularly significant in glial cells [205,206,207]. Additionally, Rett syndrome astrocytes show an overall decrease in oxygen consumption rate and overall ATP production [206]. Accordingly, Rett syndrome astrocytes exhibit abnormal mitochondrial and metabolic function, along with increased amino acid metabolism to compensate for the energy deficit. Interestingly, abnormal mitochondria in Rett syndrome astrocytes can be transferred to neurons [208], which cause neuronal hyperexcitability and neuronal dysfunction [206,209].
Oligodendrocytes also express MECP2 [210] and oligodendrocyte-specific proteins, such as crystallin B and S100a13, were abundant in the post-mortem brains of patients with Rett syndrome [211]. Conditionally knocked-out mice with Mecp2 deficiency specifically in oligodendrocyte lineages demonstrate severe hindlimb clasping phenotypes, which are typically observed in Rett syndrome models [212]. MECP2 is also essential for the oligodendrocyte maturation from oligodendrocyte progenitor cells. When differentiation is induced in MECP2-mutated NSCs, oligodendrocyte differentiation rate is decreased [213]. However, glial differentiation mechanisms may vary dependent on species, brain region, and environmental factors [214]. In addition, MECP2 is associated with oligodendrocyte survival and myelination [215].
Collectively, MECP2 in glial cells, including astrocytes and oligodendrocytes, contributes to the neurological manifestations of Rett syndrome and glial cells play an active role in disease pathology beyond neuronal dysfunction directly or indirectly.

4.4. Other Organs Affected by MECP2 Mutation and Rett Syndrome

Genetic disorders usually affect multiple organ systems [216] MECP2 is abundantly expressed in not only the brain but also the heart, kidneys, lungs, and spleen, whereas its expression is minimal in the liver, stomach, and small intestine [99]. As an epigenetic regulator, MECP2 plays a critical role in cardiovascular function. Prolonged QTc intervals have been observed in some patients with Rett syndrome and Mecp2-null male mice [217]. MECP2 is also involved in cardiac development. Mecp2-deficient mouse ESCs show severely impaired development into cardiovascular progenitors, and Mecp2-null mice display dysregulated cardiac gene expression and structural abnormalities in the myocardium [218].
Urinary dysfunction is reported in some patients with Rett syndrome. Mecp2-deficient mice display frequent urination, reduced void volume, and signs of renal failure owing to urethral obstruction [219]. Interestingly, MECP2 is upregulated during ischemia–reperfusion-induced acute kidney injury, where it exerts a protective effect by suppressing the IL-6/STAT3 axis. On the contrary, MECP2 deficiency worsens renal injury and promotes cell death, inflammation, and fibrosis [220]. These findings indicate that MECP2 functions not only as a neuronal gene but also as a key kidney function regulator.
MECP2 deficiency also impacts the immune system within the spleen. MECP2 is expressed in spleen-resident macrophages in mice, and peripheral macrophage populations are reduced in Mecp2-null mice than in controls, suggesting that Mecp2 deficiency may affect the function of the peripheral immune system, such as the spleen [221]. Meanwhile, splenocytes from Mecp2-deficient mice with experimental autoimmune encephalomyelitis maintained a proinflammatory profile from acute to chronic stages upon in vitro autoantigen restimulation, indicating that Mecp2 loss may lead to excess inflammatory responses [222].
Respiratory dysfunction caused by MECP2 deficiency is primarily attributed to impaired neural regulation within the brainstem. In Mecp2-deficient mice, neurotransmitter expression related to swallowing and airway protection is reduced in the brainstem [223]. However, respiratory dysfunction in Rett syndrome is also associated with morphological abnormalities of the lungs. High-resolution CT scans of the lungs in females with MECP2 mutations revealed various abnormalities, including centrilobular nodules, bronchial wall thickening, patchy ground-glass opacities, and bronchiectasis in about half cases [224]. Additionally, pulmonary dysfunction also contributes to respiratory abnormalities. Conditional MECP2 knockout in alveolar epithelial type II cells caused abnormal lung lipid profiles and respiratory symptoms. These respiratory abnormalities were distinct from those observed when Mecp2 was deleted only in brainstem neurons [225].

5. Rett Syndrome Treatment

5.1. AAV9-Based Gene Replacement Therapy

Wild-type MECP2 gene delivery has been extensively studied as a treatment for Rett syndrome [226]. Injecting a self-complementary AAV9 vector expressing a codon-optimized Mecp2 (scAAV9-MCO) improved symptoms and survival in Mecp2-deficient mice but caused toxicity at high doses [227,228]. To address these issues, second-generation vectors with extended Mecp2 promoters and regulatory 3′-UTR elements reduced liver toxicity, enhanced brain delivery, and improved outcomes in neonatal Mecp2-deficient mice [229]. AAV-PHP.B demonstrated excellent blood–brain barrier penetration with high specificity for both neurons and glial cells in adult mice [230,231]. Intravenous injection of an AAV-PHP.eB vector expressing instability-inducing MECP2 (iMECP2) improved motor function and lifespan without liver toxicity [232]. The capsid-modified AAV.CAP-B10 achieved strong transgene expression in multiple brain regions of mice and marmosets [233]. Additionally, miniMECP2 gene therapy (TSHA-102) with a miRNA-responsive element improved respiration, weight, survival, and motor skills in Rett syndrome mice [234].
Although MECP2 gene delivery techniques for the treatment of Rett syndrome have advanced, there is still a lack of research on how long the transgenic MECP2 is expressed and when its expression may cease. To achieve clinical improvement, further studies are needed to define the optimal timing for intervention.

5.2. MECP2 Gene Editing and Xi Reactivation

The adenosine deaminases acting on RNA 2 (ADAR2) catalyzes adenosine-to-inosine conversion in double-stranded RNA, which is one of the most common forms found in brain mRNA [235,236]. Sinnamon et al. employed an AAV vector with hyperactive ADAR2 for RNA editing, restoring MECP2 expression in cultured hippocampal neurons and in the hippocampus and brainstem of Rett syndrome mouse models [237,238,239]. Meanwhile, in females with Rett syndrome, random XCI results in approximately 50% cells expressing mutant MECP2 and the remainder expressing normal MECP2 [240]. Therefore, X chromosome reactivation has been considered a potential therapeutic approach for Rett syndrome. Combined treatment with an antisense oligonucleotide targeting Xist and a DNA methylation inhibitor significantly increased Mecp2 expression from the Xi in mouse fibroblasts [241]. Subsequent studies pharmacologically inhibited X chromosome inactivation factors (XCIFs) and reactivated the Xi-linked Mecp2 in mouse cortical neurons in vivo [242]. However, its effects on Rett syndrome phenotypes remain to be determined. Recently, multiplex epigenome editing restored MECP2 expression that follows restoration of neuronal soma size and electrophysiological function in human Rett syndrome ESCs and their derivative neurons [243]. However, further validation in living animal models is needed to evaluate their clinical relevance.

5.3. Anti-Inflammation and Immunomodulation

MECP2 regulates microglial responses to inflammatory stimuli [221]. In MECP2-null microglia, increased glutamate secretion causes neurotoxicity [121]. Both Mecp2-deficient mice and patients with Rett syndrome show excess inflammation and oxidative stress [244,245]. Dendrimer-N-acetyl cysteine (D-NAC) selectively targets microglia and improves neurological and behavioral functions [246]. Inhibiting the overactive inflammatory RIPK1 kinase pathway in MECP2-deficient microglia reduces oxidative stress and inflammatory cytokine release, improving neurological function [247]. In Mecp2-null mice, the downstream gene Irak1 is overexpressed, which significantly activates the NF-κB pathway. Therefore, lowering NF-κB expression could enhance the dendritic complexity of cortical callosal projection neurons (CPNs) and substantially prolong their lifespan [122].

5.4. Pharmacological Therapy

Trofinetide (Daybue™), an oral solution at a concentration of 200 mg/mL and a synthetic analog of the N-terminal tripeptide of insulin-like growth factor 1 (IGF-1; Gly–Pro–Glu (GPE)), was the first drug approved by the United States Food and Drug Administration (FDA) for the treatment of Rett syndrome in March 2023 [248]. A phase 3 trial in 187 female patients aged 5–20 showed significant improvements in behavior, clinical assessment, communication, and symbolic behavior [248]. Trofinetide improves the structure and function of synapses, reduces neuroinflammation and oxidative stress, modulates apoptosis, and promotes neuronal homeostasis, with a longer half-life than GPE [249]. Trofinetide showed clinically meaningful improvements in Rett Syndrome Behavior Questionnaire (RSBQ) and Clinical Global Impression–Improvement (CGI-I) scores in patients with Rett syndrome. Adverse effects were mainly limited to mild vomiting and diarrhea. Overall, its favorable safety profile and demonstrated efficacy position it as an important therapeutic option for Rett syndrome [250].
In MECP2-mutant neurons, protein production is decreased, a phenomenon linked to dysregulated AKT/mTOR signaling [32]. The AKT/mTOR pathway can be activated by inhibiting PTEN, a negative regulator of PI3K, increasing neuronal soma size and enhancing neurite arborization [251,252]. Through BDNF and IGF-1 treatment, Li et al. observed an increase in protein synthesis and the recovery of AKT/mTOR signaling [32]. Activation of AKT/mTOR signaling after growth factor administration ameliorates disease phenotypes, such as reduced soma size, reduced neurite complexity, and reduced action potential rates, in MECP2-mutant human neurons.
MECP2 deficiency contributes to Rett syndrome pathology through changes in NMDA receptor (NMDAR) expression [253]. Administering the NMDAR antagonist ketamine alleviated motor and respiratory deficits and extended survival in Rett syndrome models [254]. Although MECP2 acts as a transcriptional repressor of BDNF [255], BDNF expression progressively declined in the brainstem and nodose ganglion of Mecp2-deficient mice [256]. Given the importance of these regions in cardiopulmonary homeostasis [257], reduced BDNF may underlie severe respiratory dysfunction. The ampakine drug CX546, which enhances glutamatergic AMPA receptor activation, increased BDNF levels, leading to restored normal respiratory rate and ventilation in mice [256].
Increased TrkB activity may help restore abnormal movements of PCs caused by MECP2 deficiency, as well as synapse formation and structural defects in neurons [258,259,260]. A 5-HT1A agonist (NLX-101) can reduce irregular breathing in Rett syndrome by acting on the brainstem [193,261,262]. Improved respiration is mediated through GIRK channel activation, independently of neurotransmitter release mechanisms [193,261,262].

5.5. Other Therapeutic Approaches

Cell-based therapies, neurostimulation, and environmental interventions have also been studied as potential treatments for Rett syndrome. Co-culturing Mecp2-deficient neurons with NPCs restored neuronal morphology and synaptic function [263]. In addition, NPC transplantation to the cerebellum improved memory and motor functions in mouse models [263]. Transcriptomic analysis revealed that activation of interferon γ (IFNγ) may be a key molecular mechanism underlying the improved brain function following NPC transplantation.
Forniceal deep brain stimulation (DBS), an invasive intervention, improved hippocampal LTP and neurogenesis in Rett syndrome mouse model, thereby restoring contextual fear memory and spatial learning [264]. Beyond the hippocampus, forniceal DBS also activates neural networks within limbic structures involved in emotion and memory [265].

6. Conclusions

Rett syndrome is a complex neurodevelopmental disorder caused by mutations in the MECP2 gene, leading to widespread epigenetic dysregulation and impaired neuronal and glial function. MECP2 is a global transcription factor that affects chromatin structure, alternative splicing, and miRNA processing, which in turn affect neurodevelopment, synaptic homeostasis, and cellular metabolism. These abnormalities affect various parts of the brain, including the cortex, hippocampus, cerebellum, and brainstem, as well as extracerebral areas, such as the heart, kidneys, spleen, and lungs.
We present strategies to understand Rett syndrome from various angles, including the molecular mechanisms of MECP2, MECP2-related mosaicism, and post-translational modifications. These insights have fueled the development of targeted therapeutic strategies, including AAV-based gene replacement, RNA editing, X chromosome reactivation, anti-inflammatory approaches, and pharmacological interventions such as Trofinetide, ketamine, and growth factors. Although various treatment strategies have been proposed, the MECP2 mosaicism and developing approaches that specifically target cells with abnormal MECP2, while sparing normal cells, should be considered to ensure safety and long-term efficacy. Future research should also focus on improving human-relevant disease models, developing personalized genetic and drug therapies, and exploring ways to overcome mosaicism. Additionally, MECP2 deficiency affects not only various cell types in the brain but also the function of multiple peripheral organs. Therefore, further research is needed to characterize tissue- and cell-type-specific responses to MECP2-based therapies.

Author Contributions

Conceptualization, G.C. and J.T.D.; writing—original draft preparation, G.C., S.L., and S.Y.; writing—review and editing, G.C., and J.T.D.; supervision, J.T.D.; funding acquisition, J.T.D. All authors have read and agreed to the published version of the manuscript.

Funding

This paper was supported by Konkuk University in 2025.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ADAR2Adenosine deaminases acting on RNA 2
ANSAutonomous nervous system
BDNFBrain-derived neurotrophic factor
CREB1cAMP-responsive element-binding protein 1
CRFCorticotropic releasing factor
CTDC-terminal domain
DANDorsal attention network
DBSDeep brain stimulation
ERKExtracellular signal-regulated kinase
GABAγ-aminobutyric acid
GABAAγ-aminobutyric acid a
GFAPGlial fibrillary acidic protein
GIRKG protein-gated inwardly rectifying potassium channel
GOGene Ontology
GPEGlycine-Proline-Glutamate
GSGlutamine synthetase
HATHistone acetyltransferase
HDACHistone deacetylases
IDIntervening domain
IGFInsulin-like growth factor
LTPLong-term potentiation
MBDMethyl-CpG-binding domain
MECP2Methyl-CpG-binding protein 2
NCoRNuclear receptor co-repressor
NGFNeurogliaform
NIDNCoR–SMRT interaction domain
NMDAN-methyl-D-aspartate
NMDARNMDA receptor
NPCNeural Progenitor Cell
NSCNeural stem cells
OMPOlfactory marker protein
ORNOlfactory receptor neurons
PCPurkinje cell
PTENPhosphatase and tensin homolog
PTZNeural progenitor cells
SIN3ASwitch-independent 3A
SMRTSilencing mediator of retinoic acid and thyroid hormone receptor
TrkBTropomyosin receptor kinase B
TRDTranscriptional repression domain
TSSTranscription start site
WTWild type
XCIX chromosome inactivation

References

  1. Zoghbi, H.Y. MeCP2 dysfunction in humans and mice. J. Child Neurol. 2005, 20, 736–740. [Google Scholar] [CrossRef]
  2. Evans, J.C.; Archer, H.L.; Colley, J.P.; Ravn, K.; Nielsen, J.B.; Kerr, A.; Williams, E.; Christodoulou, J.; Gécz, J.; Jardine, P.E. Early onset seizures and Rett-like features associated with mutations in CDKL5. Eur. J. Hum. Genet. 2005, 13, 1113–1120. [Google Scholar] [CrossRef]
  3. Philippe, C.; Amsallem, D.; Francannet, C.; Lambert, L.; Saunier, A.; Verneau, F.; Jonveaux, P. Phenotypic variability in Rett syndrome associated with FOXG1 mutations in females. J. Med. Genet. 2010, 47, 59–65. [Google Scholar] [CrossRef]
  4. Petriti, U.; Dudman, D.C.; Scosyrev, E.; Lopez-Leon, S. Global prevalence of Rett syndrome: Systematic review and meta-analysis. Syst. Rev. 2023, 12, 5. [Google Scholar] [CrossRef]
  5. Akbarian, S.; Jiang, Y.; Laforet, G. The molecular pathology of Rett syndrome: Synopsis and update. Neuromol. Med. 2006, 8, 485–494. [Google Scholar] [CrossRef]
  6. Chahrour, M.; Zoghbi, H.Y. The story of Rett syndrome: From clinic to neurobiology. Neuron 2007, 56, 422–437. [Google Scholar] [CrossRef]
  7. Krishnaraj, R.; Ho, G.; Christodoulou, J. RettBASE: Rett syndrome database update. Hum. Mutat. 2017, 38, 922–931. [Google Scholar] [CrossRef] [PubMed]
  8. Signorini, C.; Leoncini, S.; Durand, T.; Galano, J.-M.; Guy, A.; Bultel-Poncé, V.; Oger, C.; Lee, J.C.-Y.; Ciccoli, L.; Hayek, J. Circulating 4-F4t-Neuroprostane and 10-F4t-Neuroprostane Are Related to MECP2 Gene Mutation and Natural History in Rett Syndrome. Int. J. Mol. Sci. 2021, 22, 4240. [Google Scholar] [CrossRef] [PubMed]
  9. Neul, J.; Fang, P.; Barrish, J.; Lane, J.; Caeg, E.; Smith, E.; Zoghbi, H.; Percy, A.; Glaze, D. Specific mutations in methyl-CpG-binding protein 2 confer different severity in Rett syndrome. Neurology 2008, 70, 1313–1321. [Google Scholar] [CrossRef]
  10. Cuddapah, V.A.; Pillai, R.B.; Shekar, K.V.; Lane, J.B.; Motil, K.J.; Skinner, S.A.; Tarquinio, D.C.; Glaze, D.G.; McGwin, G.; Kaufmann, W.E. Methyl-CpG-binding protein 2 (MECP2) mutation type is associated with disease severity in Rett syndrome. J. Med. Genet. 2014, 51, 152–158. [Google Scholar] [CrossRef] [PubMed]
  11. Trappe, R.; Laccone, F.; Cobilanschi, J.; Meins, M.; Huppke, P.; Hanefeld, F.; Engel, W. MECP2 mutations in sporadic cases of Rett syndrome are almost exclusively of paternal origin. Am. J. Hum. Genet. 2001, 68, 1093–1101. [Google Scholar] [CrossRef]
  12. Girard, M.; Couvert, P.; Carrié, A.; Tardieu, M.; Chelly, J.; Beldjord, C.; Bienvenu, T. Parental origin of de novo MECP2 mutations in Rett syndrome. Eur. J. Hum. Genet. 2001, 9, 231–236. [Google Scholar] [CrossRef]
  13. Zhu, X.; Li, M.; Pan, H.; Bao, X.; Zhang, J.; Wu, X. Analysis of the parental origin of de novo MECP2 mutations and X chromosome inactivation in 24 sporadic patients with Rett syndrome in China. J. Child Neurol. 2010, 25, 842–848. [Google Scholar] [CrossRef]
  14. Shah, J.; Patel, H.; Jain, D.; Sheth, F.; Sheth, H. A rare case of a male child with post-zygotic de novo mosaic variant c. 538C> T in MECP2 gene: A case report of Rett syndrome. BMC Neurol. 2021, 21, 469. [Google Scholar] [CrossRef]
  15. Schüle, B.; Armstrong, D.; Vogel, H.; Oviedo, A.; Francke, U. Severe congenital encephalopathy caused by MECP2 null mutations in males: Central hypoxia and reduced neuronal dendritic structure. Clin. Genet. 2008, 74, 116–126. [Google Scholar] [CrossRef]
  16. Cheung, A.Y.; Horvath, L.M.; Grafodatskaya, D.; Pasceri, P.; Weksberg, R.; Hotta, A.; Carrel, L.; Ellis, J. Isolation of MECP2-null Rett Syndrome patient hiPS cells and isogenic controls through X-chromosome inactivation. Hum. Mol. Genet. 2011, 20, 2103–2115. [Google Scholar] [CrossRef] [PubMed]
  17. Guy, J.; Hendrich, B.; Holmes, M.; Martin, J.E.; Bird, A. A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat. Genet. 2001, 27, 322–326. [Google Scholar] [CrossRef] [PubMed]
  18. Tukiainen, T.; Villani, A.-C.; Yen, A.; Rivas, M.A.; Marshall, J.L.; Satija, R.; Aguirre, M.; Gauthier, L.; Fleharty, M.; Kirby, A.; et al. Landscape of X chromosome inactivation across human tissues. Nature 2017, 550, 244–248. [Google Scholar] [CrossRef] [PubMed]
  19. Patrat, C.; Ouimette, J.-F.; Rougeulle, C. X chromosome inactivation in human development. Development 2020, 147, dev183095. [Google Scholar] [CrossRef]
  20. van den Berg, I.M.; Laven, J.S.; Stevens, M.; Jonkers, I.; Galjaard, R.-J.; Gribnau, J.; van Doorninck, J.H. X chromosome inactivation is initiated in human preimplantation embryos. Am. J. Hum. Genet. 2009, 84, 771–779. [Google Scholar] [CrossRef]
  21. Thorpe, J.; Osei-Owusu, I.A.; Avigdor, B.E.; Tupler, R.; Pevsner, J. Mosaicism in human health and disease. Annu. Rev. Genet. 2020, 54, 487–510. [Google Scholar] [CrossRef]
  22. Renthal, W.; Boxer, L.D.; Hrvatin, S.; Li, E.; Silberfeld, A.; Nagy, M.A.; Griffith, E.C.; Vierbuchen, T.; Greenberg, M.E. Characterization of human mosaic Rett syndrome brain tissue by single-nucleus RNA sequencing. Nat. Neurosci. 2018, 21, 1670–1679. [Google Scholar] [CrossRef]
  23. Armstrong, D.D. Neuropathology of Rett syndrome. J. Child Neurol. 2005, 20, 747–753. [Google Scholar] [CrossRef]
  24. Young, J.I.; Zoghbi, H.Y. X-chromosome inactivation patterns are unbalanced and affect the phenotypic outcome in a mouse model of rett syndrome. Am. J. Hum. Genet. 2004, 74, 511–520. [Google Scholar] [CrossRef]
  25. Schaevitz, L.; Gómez, N.; Zhen, D.; Berger-Sweeney, J. MeCP2 R168X male and female mutant mice exhibit Rett-like behavioral deficits. Genes Brain Behav. 2013, 12, 732–740. [Google Scholar] [CrossRef] [PubMed]
  26. Cheung, A.Y.; Horvath, L.M.; Carrel, L.; Ellis, J. X-chromosome inactivation in rett syndrome human induced pluripotent stem cells. Front. Psychiatry 2012, 3, 24. [Google Scholar] [CrossRef]
  27. Archer, H.; Evans, J.; Leonard, H.; Colvin, L.; Ravine, D.; Christodoulou, J.; Williamson, S.; Charman, T.; Bailey, M.E.; Sampson, J. Correlation between clinical severity in patients with Rett syndrome with a p.R168X or p.T158M MECP2 mutation, and the direction and degree of skewing of X-chromosome inactivation. J. Med. Genet. 2007, 44, 148–152. [Google Scholar] [CrossRef]
  28. Pohodich, A.E.; Zoghbi, H.Y. Rett syndrome: Disruption of epigenetic control of postnatal neurological functions. Hum. Mol. Genet. 2015, 24, R10–R16. [Google Scholar] [CrossRef] [PubMed]
  29. D’Mello III, S.R. MECP2 and the biology of MECP2 duplication syndrome. J. Neurochem. 2021, 159, 29–60. [Google Scholar] [CrossRef] [PubMed]
  30. Shahbazian, M.D.; Young, J.I.; Yuva-Paylor, L.A.; Spencer, C.M.; Antalffy, B.A.; Noebels, J.L.; Armstrong, D.L.; Paylor, R.; Zoghbi, H.Y. Mice with truncated MeCP2 recapitulate many Rett syndrome features and display hyperacetylation of histone H3. Neuron 2002, 35, 243–254. [Google Scholar] [CrossRef]
  31. Chen, R.Z.; Akbarian, S.; Tudor, M.; Jaenisch, R. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nat. Genet. 2001, 27, 327–331. [Google Scholar] [CrossRef]
  32. Li, Y.; Wang, H.; Muffat, J.; Cheng, A.W.; Orlando, D.A.; Lovén, J.; Kwok, S.-m.; Feldman, D.A.; Bateup, H.S.; Gao, Q. Global transcriptional and translational repression in human-embryonic-stem-cell-derived Rett syndrome neurons. Cell Stem Cell 2013, 13, 446–458. [Google Scholar] [CrossRef] [PubMed]
  33. Cheng, T.-L.; Chen, J.; Wan, H.; Tang, B.; Tian, W.; Liao, L.; Qiu, Z. Regulation of mRNA splicing by MeCP2 via epigenetic modifications in the brain. Sci. Rep. 2017, 7, 42790. [Google Scholar] [CrossRef] [PubMed]
  34. Young, J.I.; Hong, E.P.; Castle, J.C.; Crespo-Barreto, J.; Bowman, A.B.; Rose, M.F.; Kang, D.; Richman, R.; Johnson, J.M.; Berget, S. Regulation of RNA splicing by the methylation-dependent transcriptional repressor methyl-CpG binding protein 2. Proc. Natl. Acad. Sci. USA 2005, 102, 17551–17558. [Google Scholar] [CrossRef]
  35. Tsujimura, K.; Irie, K.; Nakashima, H.; Egashira, Y.; Fukao, Y.; Fujiwara, M.; Itoh, M.; Uesaka, M.; Imamura, T.; Nakahata, Y. miR-199a links MeCP2 with mTOR signaling and its dysregulation leads to Rett syndrome phenotypes. Cell Rep. 2015, 12, 1887–1901. [Google Scholar] [CrossRef]
  36. Cheng, T.-L.; Wang, Z.; Liao, Q.; Zhu, Y.; Zhou, W.-H.; Xu, W.; Qiu, Z. MeCP2 suppresses nuclear microRNA processing and dendritic growth by regulating the DGCR8/Drosha complex. Dev. Cell 2014, 28, 547–560. [Google Scholar] [CrossRef]
  37. Pejhan, S.; Rastegar, M. Role of DNA methyl-CpG-binding protein MeCP2 in Rett syndrome pathobiology and mechanism of disease. Biomolecules 2021, 11, 75. [Google Scholar] [CrossRef]
  38. Georgel, P.T.; Horowitz-Scherer, R.A.; Adkins, N.; Woodcock, C.L.; Wade, P.A.; Hansen, J.C. Chromatin compaction by human MeCP2: Assembly of novel secondary chromatin structures in the absence of DNA methylation. J. Biol. Chem. 2003, 278, 32181–32188. [Google Scholar] [CrossRef] [PubMed]
  39. Skene, P.J.; Illingworth, R.S.; Webb, S.; Kerr, A.R.; James, K.D.; Turner, D.J.; Andrews, R.; Bird, A.P. Neuronal MeCP2 is expressed at near histone-octamer levels and globally alters the chromatin state. Mol. Cell 2010, 37, 457–468. [Google Scholar] [CrossRef]
  40. Wang, L.; Hu, M.; Zuo, M.-Q.; Zhao, J.; Wu, D.; Huang, L.; Wen, Y.; Li, Y.; Chen, P.; Bao, X. Rett syndrome-causing mutations compromise MeCP2-mediated liquid–liquid phase separation of chromatin. Cell Res. 2020, 30, 393–407. [Google Scholar] [CrossRef]
  41. Horike, S.-i.; Cai, S.; Miyano, M.; Cheng, J.-F.; Kohwi-Shigematsu, T. Loss of silent-chromatin looping and impaired imprinting of DLX5 in Rett syndrome. Nat. Genet. 2005, 37, 31–40. [Google Scholar] [CrossRef] [PubMed]
  42. Nan, X.; Ng, H.-H.; Johnson, C.A.; Laherty, C.D.; Turner, B.M.; Eisenman, R.N.; Bird, A. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 1998, 393, 386–389. [Google Scholar] [CrossRef]
  43. Jones, P.L.; Veenstra, G.J.C.; Wade, P.A.; Vermaak, D.; Kass, S.U.; Landsberger, N.; Strouboulis, J.; Wolffe, A.P. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat. Genet. 1998, 19, 187–191. [Google Scholar] [CrossRef]
  44. Lyst, M.J.; Ekiert, R.; Ebert, D.H.; Merusi, C.; Nowak, J.; Selfridge, J.; Guy, J.; Kastan, N.R.; Robinson, N.D.; de Lima Alves, F. Rett syndrome mutations abolish the interaction of MeCP2 with the NCoR/SMRT co-repressor. Nat. Neurosci. 2013, 16, 898–902. [Google Scholar] [CrossRef]
  45. Chahrour, M.; Jung, S.Y.; Shaw, C.; Zhou, X.; Wong, S.T.; Qin, J.; Zoghbi, H.Y. MeCP2, a key contributor to neurological disease, activates and represses transcription. Science 2008, 320, 1224–1229. [Google Scholar] [CrossRef]
  46. Ben-Shachar, S.; Chahrour, M.; Thaller, C.; Shaw, C.A.; Zoghbi, H.Y. Mouse models of MeCP2 disorders share gene expression changes in the cerebellum and hypothalamus. Hum. Mol. Genet. 2009, 18, 2431–2442. [Google Scholar] [CrossRef]
  47. Li, R.; Dong, Q.; Yuan, X.; Zeng, X.; Gao, Y.; Chiao, C.; Li, H.; Zhao, X.; Keles, S.; Wang, Z. Misregulation of alternative splicing in a mouse model of Rett syndrome. PLoS Genet. 2016, 12, e1006129. [Google Scholar] [CrossRef] [PubMed]
  48. Lyabin, D.N.; Eliseeva, I.A.; Ovchinnikov, L.P. YB-1 protein: Functions and regulation. Wiley Interdiscip. Rev. RNA 2014, 5, 95–110. [Google Scholar] [CrossRef]
  49. Urdinguio, R.G.; Fernandez, A.F.; Lopez-Nieva, P.; Rossi, S.; Huertas, D.; Kulis, M.; Liu, C.-G.; Croce, C.M.; Calin, G.A.; Esteller, M. Disrupted microRNA expression caused by Mecp2 loss in a mouse model of Rett syndrome. Epigenetics 2010, 5, 656–663. [Google Scholar] [CrossRef] [PubMed]
  50. Ballestar, E.; Ropero, S.; Alaminos, M.; Armstrong, J.; Setien, F.; Agrelo, R.; Fraga, M.F.; Herranz, M.; Avila, S.; Pineda, M. The impact of MECP2 mutations in the expression patterns of Rett syndrome patients. Hum. Genet. 2005, 116, 91–104. [Google Scholar] [CrossRef]
  51. Traynor, J.; Agarwal, P.; Lazzeroni, L.; Francke, U. Gene expression patterns vary in clonal cell cultures from Rett syndrome females with eight different MECP2 mutations. BMC Med. Genet. 2002, 3, 12. [Google Scholar] [CrossRef]
  52. Lyst, M.J.; Bird, A. Rett syndrome: A complex disorder with simple roots. Nat. Rev. Genet. 2015, 16, 261–275. [Google Scholar] [CrossRef] [PubMed]
  53. Baker, S.A.; Chen, L.; Wilkins, A.D.; Yu, P.; Lichtarge, O.; Zoghbi, H.Y. An AT-hook domain in MeCP2 determines the clinical course of Rett syndrome and related disorders. Cell 2013, 152, 984–996. [Google Scholar] [CrossRef]
  54. Xu, X.; Pozzo-Miller, L. A novel DNA-binding feature of MeCP2 contributes to Rett syndrome. Front. Cell. Neurosci. 2013, 7, 64. [Google Scholar] [CrossRef] [PubMed]
  55. Guy, J.; Alexander-Howden, B.; FitzPatrick, L.; DeSousa, D.; Koerner, M.V.; Selfridge, J.; Bird, A. A mutation-led search for novel functional domains in MeCP2. Hum. Mol. Genet. 2018, 27, 2531–2545. [Google Scholar] [CrossRef] [PubMed]
  56. Heckman, L.D.; Chahrour, M.H.; Zoghbi, H.Y. Rett-causing mutations reveal two domains critical for MeCP2 function and for toxicity in MECP2 duplication syndrome mice. eLife 2014, 3, e02676. [Google Scholar] [CrossRef]
  57. Liu, Y.; Flamier, A.; Bell, G.W.; Diao, A.J.; Whitfield, T.W.; Wang, H.-C.; Wu, Y.; Schulte, F.; Friesen, M.; Guo, R. MECP2 directly interacts with RNA polymerase II to modulate transcription in human neurons. Neuron 2024, 112, 1943–1958.E10. [Google Scholar] [CrossRef]
  58. Bebbington, A.; Anderson, A.; Ravine, D.; Fyfe, S.; Pineda, M.; de Klerk, N.; Ben-Zeev, B.; Yatawara, N.; Percy, A.; Kaufmann, W.E.; et al. Investigating genotype-phenotype relationships in Rett syndrome using an international data set. Neurology 2008, 70, 868–875. [Google Scholar] [CrossRef]
  59. Colvin, L.; Leonard, H.; de Klerk, N.; Davis, M.; Weaving, L.; Williamson, S.; Christodoulou, J. Refining the phenotype of common mutations in Rett syndrome. J. Med. Genet. 2004, 41, 25–30. [Google Scholar] [CrossRef]
  60. Klose, R.J.; Sarraf, S.A.; Schmiedeberg, L.; McDermott, S.M.; Stancheva, I.; Bird, A.P. DNA binding selectivity of MeCP2 due to a requirement for A/T sequences adjacent to methyl-CpG. Mol. Cell 2005, 19, 667–678. [Google Scholar] [CrossRef]
  61. Kouzarides, T. Chromatin modifications and their function. Cell 2007, 128, 693–705. [Google Scholar] [CrossRef]
  62. Wang, Z.; Zang, C.; Rosenfeld, J.A.; Schones, D.E.; Barski, A.; Cuddapah, S.; Cui, K.; Roh, T.-Y.; Peng, W.; Zhang, M.Q. Combinatorial patterns of histone acetylations and methylations in the human genome. Nat. Genet. 2008, 40, 897–903. [Google Scholar] [CrossRef] [PubMed]
  63. Nishida, H.; Suzuki, T.; Kondo, S.; Miura, H.; Fujimura, Y.-i.; Hayashizaki, Y. Histone H3 acetylated at lysine 9 in promoter is associated with low nucleosome density in the vicinity of transcription start site in human cell. Chromosome Res. 2006, 14, 203–211. [Google Scholar] [CrossRef]
  64. Morris, S.A.; Rao, B.; Garcia, B.A.; Hake, S.B.; Diaz, R.L.; Shabanowitz, J.; Hunt, D.F.; Allis, C.D.; Lieb, J.D.; Strahl, B.D. Identification of histone H3 lysine 36 acetylation as a highly conserved histone modification. J. Biol. Chem. 2007, 282, 7632–7640. [Google Scholar] [CrossRef]
  65. Xu, Y.M.; Du, J.Y.; Lau, A.T. Posttranslational modifications of human histone H3: An update. Proteomics 2014, 14, 2047–2060. [Google Scholar] [CrossRef]
  66. Che, Y.-C.; Jiang, L.; Li, Q.-H. Molecular modification of a HSV-1 protein and its associated gene transcriptional regulation. Virol. Sin. 2008, 23, 394–398. [Google Scholar] [CrossRef]
  67. Ayissi, V.B.O.; Ebrahimi, A.; Schluesenner, H. Epigenetic effects of natural polyphenols: A focus on SIRT1-mediated mechanisms. Mol. Nutr. Food Res. 2014, 58, 22–32. [Google Scholar] [CrossRef] [PubMed]
  68. Dandrea, M.; Donadelli, M.; Costanzo, C.; Scarpa, A.; Palmieri, M. MeCP2/H3meK9 are involved in IL-6 gene silencing in pancreatic adenocarcinoma cell lines. Nucleic Acids Res. 2009, 37, 6681–6690. [Google Scholar] [CrossRef]
  69. Thambirajah, A.A.; Ng, M.K.; Frehlick, L.J.; Li, A.; Serpa, J.J.; Petrotchenko, E.V.; Silva-Moreno, B.; Missiaen, K.K.; Borchers, C.H.; Adam Hall, J. MeCP2 binds to nucleosome free (linker DNA) regions and to H3K9/H3K27 methylated nucleosomes in the brain. Nucleic Acids Res. 2012, 40, 2884–2897. [Google Scholar] [CrossRef]
  70. Nan, X.; Meehan, R.R.; Bird, A. Dissection of the methyl-CpG binding domain from the chromosomal protein MeCP2. Nucleic Acids Res. 1993, 21, 4886–4892. [Google Scholar] [CrossRef]
  71. Chen, L.; Chen, K.; Lavery, L.A.; Baker, S.A.; Shaw, C.A.; Li, W.; Zoghbi, H.Y. MeCP2 binds to non-CG methylated DNA as neurons mature, influencing transcription and the timing of onset for Rett syndrome. Proc. Natl. Acad. Sci. USA 2015, 112, 5509–5514. [Google Scholar] [CrossRef]
  72. Mellén, M.; Ayata, P.; Dewell, S.; Kriaucionis, S.; Heintz, N. MeCP2 binds to 5hmC enriched within active genes and accessible chromatin in the nervous system. Cell 2012, 151, 1417–1430. [Google Scholar] [CrossRef]
  73. Lee, W.; Kim, J.; Yun, J.-M.; Ohn, T.; Gong, Q. MeCP2 regulates gene expression through recognition of H3K27me3. Nat. Commun. 2020, 11, 3140. [Google Scholar] [CrossRef]
  74. Kimura, H.; Shiota, K. Methyl-CpG-binding protein, MeCP2, is a target molecule for maintenance DNA methyltransferase, Dnmt1. J. Biol. Chem. 2003, 278, 4806–4812. [Google Scholar] [CrossRef]
  75. Baubec, T.; Ivánek, R.; Lienert, F.; Schübeler, D. Methylation-dependent and-independent genomic targeting principles of the MBD protein family. Cell 2013, 153, 480–492. [Google Scholar] [CrossRef] [PubMed]
  76. Ebert, D.H.; Gabel, H.W.; Robinson, N.D.; Kastan, N.R.; Hu, L.S.; Cohen, S.; Navarro, A.J.; Lyst, M.J.; Ekiert, R.; Bird, A.P. Activity-dependent phosphorylation of MeCP2 threonine 308 regulates interaction with NCoR. Nature 2013, 499, 341–345. [Google Scholar] [CrossRef] [PubMed]
  77. Shibasaki, F.; Hallin, U.; Uchino, H. Calcineurin as a multifunctional regulator. J. Biochem. 2002, 131, 1–15. [Google Scholar] [CrossRef] [PubMed]
  78. Buchthal, B.; Lau, D.; Weiss, U.; Weislogel, J.-M.; Bading, H. Nuclear calcium signaling controls methyl-CpG-binding protein 2 (MeCP2) phosphorylation on serine 421 following synaptic activity. J. Biol. Chem. 2012, 287, 30967–30974. [Google Scholar] [CrossRef]
  79. Lin, Y.; Bloodgood, B.L.; Hauser, J.L.; Lapan, A.D.; Koon, A.C.; Kim, T.-K.; Hu, L.S.; Malik, A.N.; Greenberg, M.E. Activity-dependent regulation of inhibitory synapse development by Npas4. Nature 2008, 455, 1198–1204. [Google Scholar] [CrossRef]
  80. Banerjee, A.; Romero-Lorenzo, E.; Sur, M. MeCP2: Making sense of missense in Rett syndrome. Cell Res. 2013, 23, 1244–1246. [Google Scholar] [CrossRef]
  81. Zhou, Z.; Hong, E.J.; Cohen, S.; Zhao, W.-n.; Ho, H.-y.H.; Schmidt, L.; Chen, W.G.; Lin, Y.; Savner, E.; Griffith, E.C. Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation. Neuron 2006, 52, 255–269. [Google Scholar] [CrossRef]
  82. Cohen, S.; Gabel, H.W.; Hemberg, M.; Hutchinson, A.N.; Sadacca, L.A.; Ebert, D.H.; Harmin, D.A.; Greenberg, R.S.; Verdine, V.K.; Zhou, Z. Genome-wide activity-dependent MeCP2 phosphorylation regulates nervous system development and function. Neuron 2011, 72, 72–85. [Google Scholar] [CrossRef]
  83. Tao, J.; Hu, K.; Chang, Q.; Wu, H.; Sherman, N.E.; Martinowich, K.; Klose, R.J.; Schanen, C.; Jaenisch, R.; Wang, W. Phosphorylation of MeCP2 at Serine 80 regulates its chromatin association and neurological function. Proc. Natl. Acad. Sci. USA 2009, 106, 4882–4887. [Google Scholar] [CrossRef] [PubMed]
  84. Chao, H.-T.; Zoghbi, H.Y. The yin and yang of MeCP2 phosphorylation. Proc. Natl. Acad. Sci. USA 2009, 106, 4577–4578. [Google Scholar] [CrossRef]
  85. Schlüter, O.M.; Schmitz, F.; Jahn, R.; Rosenmund, C.; Südhof, T.C. A complete genetic analysis of neuronal Rab3 function. J. Neurosci. 2004, 24, 6629–6637. [Google Scholar] [CrossRef]
  86. Bezzi, P.; Gundersen, V.; Galbete, J.L.; Seifert, G.; Steinhäuser, C.; Pilati, E.; Volterra, A. Astrocytes contain a vesicular compartment that is competent for regulated exocytosis of glutamate. Nat. Neurosci. 2004, 7, 613–620. [Google Scholar] [CrossRef]
  87. Park, J.; Liu, B.; Chen, T.; Li, H.; Hu, X.; Gao, J.; Zhu, Y.; Zhu, Q.; Qiang, B.; Yuan, J. Disruption of Nectin-like 1 cell adhesion molecule leads to delayed axonal myelination in the CNS. J. Neurosci. 2008, 28, 12815–12819. [Google Scholar] [CrossRef]
  88. Stefanelli, G.; Gandaglia, A.; Costa, M.; Cheema, M.S.; Di Marino, D.; Barbiero, I.; Kilstrup-Nielsen, C.; Ausió, J.; Landsberger, N. Brain phosphorylation of MeCP2 at serine 164 is developmentally regulated and globally alters its chromatin association. Sci. Rep. 2016, 6, 28295. [Google Scholar] [CrossRef]
  89. Tai, D.J.; Liu, Y.C.; Hsu, W.L.; Ma, Y.L.; Cheng, S.J.; Liu, S.Y.; Lee, E.H. MeCP2 SUMOylation rescues Mecp2-mutant-induced behavioural deficits in a mouse model of Rett syndrome. Nat. Commun. 2016, 7, 10552. [Google Scholar] [CrossRef] [PubMed]
  90. Cheng, J.; Huang, M.; Zhu, Y.; Xin, Y.J.; Zhao, Y.K.; Huang, J.; Yu, J.X.; Zhou, W.H.; Qiu, Z. SUMO ylation of Me CP 2 is essential for transcriptional repression and hippocampal synapse development. J. Neurochem. 2014, 128, 798–806. [Google Scholar] [CrossRef] [PubMed]
  91. Kalani, L.; Kim, B.-H.; Vincent, J.B.; Ausió, J. MeCP2 ubiquitination and sumoylation, in search of a function. Hum. Mol. Genet. 2024, 33, 1–11. [Google Scholar] [CrossRef]
  92. Hsu, W.-L.; Ma, Y.-L.; Liu, Y.-C.; Tai, D.J.; Lee, E.H. Restoring Wnt6 signaling ameliorates behavioral deficits in MeCP2 T158A mouse model of Rett syndrome. Sci. Rep. 2020, 10, 1074. [Google Scholar] [CrossRef]
  93. Goffin, D.; Allen, M.; Zhang, L.; Amorim, M.; Wang, I.-T.J.; Reyes, A.-R.S.; Mercado-Berton, A.; Ong, C.; Cohen, S.; Hu, L. Rett syndrome mutation MeCP2 T158A disrupts DNA binding, protein stability and ERP responses. Nat. Neurosci. 2012, 15, 274–283. [Google Scholar] [CrossRef]
  94. Kishi, N.; Macklis, J.D. MECP2 is progressively expressed in post-migratory neurons and is involved in neuronal maturation rather than cell fate decisions. Mol. Cell. Neurosci. 2004, 27, 306–321. [Google Scholar] [CrossRef]
  95. Li, H.; Zhong, X.; Chau, K.F.; Williams, E.C.; Chang, Q. Loss of activity-induced phosphorylation of MeCP2 enhances synaptogenesis, LTP and spatial memory. Nat. Neurosci. 2011, 14, 1001–1008. [Google Scholar] [CrossRef] [PubMed]
  96. Jiang, M.; Ash, R.T.; Baker, S.A.; Suter, B.; Ferguson, A.; Park, J.; Rudy, J.; Torsky, S.P.; Chao, H.-T.; Zoghbi, H.Y. Dendritic arborization and spine dynamics are abnormal in the mouse model of MECP2 duplication syndrome. J. Neurosci. 2013, 33, 19518–19533. [Google Scholar] [CrossRef] [PubMed]
  97. Kim, K.-Y.; Hysolli, E.; Park, I.-H. Neuronal maturation defect in induced pluripotent stem cells from patients with Rett syndrome. Proc. Natl. Acad. Sci. USA 2011, 108, 14169–14174. [Google Scholar] [CrossRef]
  98. Jung, B.P.; Jugloff, D.G.; Zhang, G.; Logan, R.; Brown, S.; Eubanks, J.H. The expression of methyl CpG binding factor MeCP2 correlates with cellular differentiation in the developing rat brain and in cultured cells. J. Neurobiol. 2003, 55, 86–96. [Google Scholar] [CrossRef] [PubMed]
  99. Shahbazian, M.D.; Antalffy, B.; Armstrong, D.L.; Zoghbi, H.Y. Insight into Rett syndrome: MeCP2 levels display tissue-and cell-specific differences and correlate with neuronal maturation. Hum. Mol. Genet. 2002, 11, 115–124. [Google Scholar] [CrossRef]
  100. Mullaney, B.; Johnston, M.; Blue, M. Developmental expression of methyl-CpG binding protein 2 is dynamically regulated in the rodent brain. Neuroscience 2004, 123, 939–949. [Google Scholar] [CrossRef]
  101. Nakashima, H.; Tsujimura, K.; Irie, K.; Imamura, T.; Trujillo, C.A.; Ishizu, M.; Uesaka, M.; Pan, M.; Noguchi, H.; Okada, K. MeCP2 controls neural stem cell fate specification through miR-199a-mediated inhibition of BMP-Smad signaling. Cell Rep. 2021, 35, 109124. [Google Scholar] [CrossRef] [PubMed]
  102. Mellios, N.; Feldman, D.A.; Sheridan, S.D.; Ip, J.P.; Kwok, S.; Amoah, S.K.; Rosen, B.; Rodriguez, B.A.; Crawford, B.; Swaminathan, R. MeCP2-regulated miRNAs control early human neurogenesis through differential effects on ERK and AKT signaling. Mol. Psychiatry 2018, 23, 1051–1065. [Google Scholar] [CrossRef]
  103. Belichenko, P.V.; Hagberg, B.; Dahlström, A. Morphological study of neocortical areas in Rett syndrome. Acta Neuropathol. 1996, 93, 50–61. [Google Scholar] [CrossRef]
  104. Hou, J.; Lin, L.; Zhou, W.; Wang, Z.; Ding, G.; Dong, Q.; Qin, L.; Wu, X.; Zheng, Y.; Yang, Y. Identification of miRNomes in human liver and hepatocellular carcinoma reveals miR-199a/b-3p as therapeutic target for hepatocellular carcinoma. Cancer Cell 2011, 19, 232–243. [Google Scholar] [CrossRef]
  105. Penna, E.; Orso, F.; Taverna, D. miR-214 as a key hub that controls cancer networks: Small player, multiple functions. J. Investig. Dermatol. 2015, 135, 960–969. [Google Scholar] [CrossRef]
  106. Shu, P.; Fu, H.; Zhao, X.; Wu, C.; Ruan, X.; Zeng, Y.; Liu, W.; Wang, M.; Hou, L.; Chen, P. MicroRNA-214 modulates neural progenitor cell differentiation by targeting Quaking during cerebral cortex development. Sci. Rep. 2017, 7, 8014. [Google Scholar] [CrossRef]
  107. Pucilowska, J.; Puzerey, P.A.; Karlo, J.C.; Galán, R.F.; Landreth, G.E. Disrupted ERK signaling during cortical development leads to abnormal progenitor proliferation, neuronal and network excitability and behavior, modeling human neuro-cardio-facial-cutaneous and related syndromes. J. Neurosci. 2012, 32, 8663–8677. [Google Scholar] [CrossRef]
  108. Zhang, J.; Shemezis, J.R.; McQuinn, E.R.; Wang, J.; Sverdlov, M.; Chenn, A. AKT activation by N-cadherin regulates beta-catenin signaling and neuronal differentiation during cortical development. Neural Dev. 2013, 8, 7. [Google Scholar] [CrossRef] [PubMed]
  109. Davis, B.N.; Hilyard, A.C.; Lagna, G.; Hata, A. SMAD proteins control DROSHA-mediated microRNA maturation. Nature 2008, 454, 56–61. [Google Scholar] [CrossRef]
  110. Takeguchi, R.; Kuroda, M.; Tanaka, R.; Suzuki, N.; Akaba, Y.; Tsujimura, K.; Itoh, M.; Takahashi, S. Structural and functional changes in the brains of patients with Rett syndrome: A multimodal MRI study. J. Neurol. Sci. 2022, 441, 120381. [Google Scholar] [CrossRef] [PubMed]
  111. Szczepanski, S.M.; Pinsk, M.A.; Douglas, M.M.; Kastner, S.; Saalmann, Y.B. Functional and structural architecture of the human dorsal frontoparietal attention network. Proc. Natl. Acad. Sci. USA 2013, 110, 15806–15811. [Google Scholar] [CrossRef]
  112. Lanssens, A.; Pizzamiglio, G.; Mantini, D.; Gillebert, C.R. Role of the dorsal attention network in distracter suppression based on features. In The Cognitive Neuroscience of Attention; Routledge: Abingdon, UK, 2020; pp. 204–213. [Google Scholar]
  113. Rohr, C.S.; Arora, A.; Cho, I.Y.; Katlariwala, P.; Dimond, D.; Dewey, D.; Bray, S. Functional network integration and attention skills in young children. Dev. Cogn. Neurosci. 2018, 30, 200–211. [Google Scholar] [CrossRef]
  114. Rohr, C.S.; Vinette, S.A.; Parsons, K.A.; Cho, I.Y.; Dimond, D.; Benischek, A.; Lebel, C.; Dewey, D.; Bray, S. Functional connectivity of the dorsal attention network predicts selective attention in 4–7 year-old girls. Cereb. Cortex 2017, 27, 4350–4360. [Google Scholar] [CrossRef]
  115. Della Sala, G.; Pizzorusso, T. Synaptic plasticity and signaling in Rett syndrome. Dev. Neurobiol. 2014, 74, 178–196. [Google Scholar] [CrossRef] [PubMed]
  116. Xu, X.; Miller, E.C.; Pozzo-Miller, L. Dendritic spine dysgenesis in Rett syndrome. Front. Neuroanat. 2014, 8, 97. [Google Scholar] [CrossRef] [PubMed]
  117. Johnston, M.V.; Blue, M.E.; Naidu, S. Rett syndrome and neuronal development. J. Child Neurol. 2005, 20, 759–763. [Google Scholar] [CrossRef] [PubMed]
  118. Santos, M.; Summavielle, T.; Teixeira-Castro, A.; Silva-Fernandes, A.; Duarte-Silva, S.; Marques, F.; Martins, L.; Dierssen, M.; Oliveira, P.; Sousa, N. Monoamine deficits in the brain of methyl-CpG binding protein 2 null mice suggest the involvement of the cerebral cortex in early stages of Rett syndrome. Neuroscience 2010, 170, 453–467. [Google Scholar] [CrossRef]
  119. Ip, J.P.; Mellios, N.; Sur, M. Rett syndrome: Insights into genetic, molecular and circuit mechanisms. Nat. Rev. Neurosci. 2018, 19, 368–382. [Google Scholar] [CrossRef]
  120. Fukuda, T.; Itoh, M.; Ichikawa, T.; Washiyama, K.; Goto, Y.-i. Delayed maturation of neuronal architecture and synaptogenesis in cerebral cortex of Mecp2-deficient mice. J. Neuropathol. Exp. Neurol. 2005, 64, 537–544. [Google Scholar] [CrossRef]
  121. Maezawa, I.; Jin, L.-W. Rett syndrome microglia damage dendrites and synapses by the elevated release of glutamate. J. Neurosci. 2010, 30, 5346–5356. [Google Scholar] [CrossRef]
  122. Kishi, N.; MacDonald, J.L.; Ye, J.; Molyneaux, B.J.; Azim, E.; Macklis, J.D. Reduction of aberrant NF-κB signalling ameliorates Rett syndrome phenotypes in Mecp2-null mice. Nat. Commun. 2016, 7, 10520. [Google Scholar] [CrossRef] [PubMed]
  123. Armstrong, D.; Dunn, J.K.; Antalffy, B.; Trivedi, R. Selective dendritic alterations in the cortex of Rett syndrome. J. Neuropathol. Exp. Neurol. 1995, 54, 195–201. [Google Scholar] [CrossRef]
  124. Armstrong, D.D. Neuropathology of Rett syndrome. Ment. Retard. Dev. Disabil. Res. Rev. 2002, 8, 72–76. [Google Scholar] [CrossRef]
  125. Wang, L.; Chang, X.; She, L.; Xu, D.; Huang, W.; Poo, M.-m. Autocrine action of BDNF on dendrite development of adult-born hippocampal neurons. J. Neurosci. 2015, 35, 8384–8393. [Google Scholar] [CrossRef]
  126. Cheng, P.-L.; Song, A.-H.; Wong, Y.-H.; Wang, S.; Zhang, X.; Poo, M.-M. Self-amplifying autocrine actions of BDNF in axon development. Proc. Natl. Acad. Sci. USA 2011, 108, 18430–18435. [Google Scholar] [CrossRef]
  127. Larimore, J.L.; Chapleau, C.A.; Kudo, S.; Theibert, A.; Percy, A.K.; Pozzo-Miller, L. Bdnf overexpression in hippocampal neurons prevents dendritic atrophy caused by Rett-associated MECP2 mutations. Neurobiol. Dis. 2009, 34, 199–211. [Google Scholar] [CrossRef]
  128. Lengel, D.; Romm, Z.L.; Bostwick, A.; Huh, J.W.; Snyder, N.W.; Smith, G.M.; Raghupathi, R. Glucocorticoid receptor overexpression in the dorsal hippocampus attenuates spatial learning and synaptic plasticity deficits after pediatric traumatic brain injury. J. Neurotrauma 2022, 39, 979–998. [Google Scholar] [CrossRef]
  129. Steffke, E.E.; Kirca, D.; Mazei-Robison, M.S.; Robison, A.J. Serum-and glucocorticoid-inducible kinase 1 activity reduces dendritic spines in dorsal hippocampus. Neurosci. Lett. 2020, 725, 134909. [Google Scholar] [CrossRef] [PubMed]
  130. Sahin, P.; McCaig, C.; Jeevahan, J.; Murray, J.T.; Hainsworth, A.H. The cell survival kinase SGK 1 and its targets FOXO 3a and NDRG 1 in aged human brain. Neuropathol. Appl. Neurobiol. 2013, 39, 623–633. [Google Scholar] [CrossRef]
  131. Martin-Batista, E.; Maglio, L.E.; Armas-Capote, N.; Hernández, G.; de la Rosa, D.A.; Giraldez, T. SGK1. 1 limits brain damage after status epilepticus through M current-dependent and independent mechanisms. Neurobiol. Dis. 2021, 153, 105317. [Google Scholar] [CrossRef] [PubMed]
  132. Kwon, O.C.; Song, J.J.; Yang, Y.; Kim, S.H.; Kim, J.Y.; Seok, M.J.; Hwang, I.; Yu, J.W.; Karmacharya, J.; Maeng, H.J. SGK1 inhibition in glia ameliorates pathologies and symptoms in Parkinson disease animal models. EMBO Mol. Med. 2021, 13, e13076. [Google Scholar] [CrossRef]
  133. Nuber, U.A.; Kriaucionis, S.; Roloff, T.C.; Guy, J.; Selfridge, J.; Steinhoff, C.; Schulz, R.; Lipkowitz, B.; Ropers, H.H.; Holmes, M.C. Up-regulation of glucocorticoid-regulated genes in a mouse model of Rett syndrome. Hum. Mol. Genet. 2005, 14, 2247–2256. [Google Scholar] [CrossRef]
  134. Géranton, S.M.; Morenilla-Palao, C.; Hunt, S.P. A role for transcriptional repressor methyl-CpG-binding protein 2 and plasticity-related gene serum-and glucocorticoid-inducible kinase 1 in the induction of inflammatory pain states. J. Neurosci. 2007, 27, 6163–6173. [Google Scholar] [CrossRef] [PubMed]
  135. Itoh, M.; Ide, S.; Takashima, S.; Kudo, S.; Nomura, Y.; Segawa, M.; Kubota, T.; Mori, H.; Tanaka, S.; Horie, H. Methyl CpG-binding protein 2 (a mutation of which causes Rett syndrome) directly regulates insulin-like growth factor binding protein 3 in mouse and human brains. J. Neuropathol. Exp. Neurol. 2007, 66, 117–123. [Google Scholar] [CrossRef] [PubMed]
  136. Payet, L.D.; Wang, X.-H.; Baxter, R.C.; Firth, S.M. Amino-and carboxyl-terminal fragments of insulin-like growth factor (IGF) binding protein-3 cooperate to bind IGFs with high affinity and inhibit IGF receptor interactions. Endocrinology 2003, 144, 2797–2806. [Google Scholar] [CrossRef]
  137. Cubbage, M.; Suwanichkul, A.; Powell, D. Insulin-like growth factor binding protein-3. Organization of the human chromosomal gene and demonstration of promoter activity. J. Biol. Chem. 1990, 265, 12642–12649. [Google Scholar] [CrossRef]
  138. Modric, T.; Silha, J.V.; Shi, Z.; Gui, Y.; Suwanichkul, A.; Durham, S.K.; Powell, D.R.; Murphy, L.J. Phenotypic manifestations of insulin-like growth factor-binding protein-3 overexpression in transgenic mice. Endocrinology 2001, 142, 1958–1967. [Google Scholar] [CrossRef]
  139. StühMer, T.; Anderson, S.A.; Ekker, M.; Rubenstein, J.L. Ectopic expression of the Dlx genes induces glutamic acid decarboxylase and Dlx expression. Development 2002, 129, 245–252. [Google Scholar] [CrossRef]
  140. Miyano, M.; Horike, S.-I.; Cai, S.; Oshimura, M.; Kohwi-Shigematsu, T. DLX5 expression is monoallelic and Dlx5 is up-regulated in the Mecp2-null frontal cortex. J. Cell. Mol. Med. 2008, 12, 1188. [Google Scholar] [CrossRef] [PubMed]
  141. Schüle, B.; Li, H.H.; Fisch-Kohl, C.; Purmann, C.; Francke, U. DLX5 and DLX6 expression is biallelic and not modulated by MeCP2 deficiency. Am. J. Hum. Genet. 2007, 81, 492–506. [Google Scholar] [CrossRef]
  142. Li, S.; Huang, H.; Wei, X.; Ye, L.; Ma, M.; Ling, M.; Wu, Y. The recycling of AMPA receptors/GABAa receptors is related to neuronal excitation/inhibition imbalance and may be regulated by KIF5A. Ann. Transl. Med. 2022, 10, 1103. [Google Scholar] [CrossRef]
  143. Davenport, E.C.; Szulc, B.R.; Drew, J.; Taylor, J.; Morgan, T.; Higgs, N.F.; López-Doménech, G.; Kittler, J.T. Autism and schizophrenia-associated CYFIP1 regulates the balance of synaptic excitation and inhibition. Cell Rep. 2019, 26, 2037–2051.e6. [Google Scholar] [CrossRef]
  144. Cox, O.H.; Song, H.Y.; Garrison-Desany, H.M.; Gadiwalla, N.; Carey, J.L.; Menzies, J.; Lee, R.S. Characterization of glucocorticoid-induced loss of DNA methylation of the stress-response gene Fkbp5 in neuronal cells. Epigenetics 2021, 16, 1377–1397. [Google Scholar] [CrossRef]
  145. Peddada, S.; Yasui, D.H.; LaSalle, J.M. Inhibitors of differentiation (ID1, ID2, ID3 and ID4) genes are neuronal targets of MeCP2 that are elevated in Rett syndrome. Hum. Mol. Genet. 2006, 15, 2003–2014. [Google Scholar] [CrossRef] [PubMed]
  146. Chu, Y.-H.; Lin, J.-d.; Nath, S.; Schachtrup, C. Id proteins: Emerging roles in CNS disease and targets for modifying neural stemcell behavior. Cell Tissue Res. 2022, 387, 433–449. [Google Scholar] [CrossRef]
  147. Jung, S.; Park, R.-H.; Kim, S.; Jeon, Y.-J.; Ham, D.-S.; Jung, M.-Y.; Kim, S.-S.; Lee, Y.-D.; Park, C.-H.; Suh-Kim, H. Id proteins facilitate self-renewal and proliferation of neural stem cells. Stem Cells Dev. 2010, 19, 831–841. [Google Scholar] [CrossRef]
  148. Langlands, K.; Yin, X.; Anand, G.; Prochownik, E.V. Differential interactions of Id proteins with basic-helix-loop-helix transcription factors. J. Biol. Chem. 1997, 272, 19785–19793. [Google Scholar] [CrossRef] [PubMed]
  149. Deng, V.; Matagne, V.; Banine, F.; Frerking, M.; Ohliger, P.; Budden, S.; Pevsner, J.; Dissen, G.A.; Sherman, L.S.; Ojeda, S.R. FXYD1 is an MeCP2 target gene overexpressed in the brains of Rett syndrome patients and Mecp2-null mice. Hum. Mol. Genet. 2007, 16, 640–650. [Google Scholar] [CrossRef] [PubMed]
  150. Matagne, V.; Wondolowski, J.; Frerking, M.; Shahidullah, M.; Delamere, N.A.; Sandau, U.S.; Budden, S.; Ojeda, S.R. Correcting deregulated Fxyd1 expression rescues deficits in neuronal arborization and potassium homeostasis in MeCP2 deficient male mice. Brain Res. 2018, 1697, 45–52. [Google Scholar] [CrossRef]
  151. Rotaru, D.C.; Wallaard, I.; de Vries, M.; van der Bie, J.; Elgersma, Y. UBE3A expression during early postnatal brain development is required for proper dorsomedial striatal maturation. JCI Insight 2023, 8, e166073. [Google Scholar] [CrossRef]
  152. Samaco, R.C.; Hogart, A.; LaSalle, J.M. Epigenetic overlap in autism-spectrum neurodevelopmental disorders: MECP2 deficiency causes reduced expression of UBE3A and GABRB3. Hum. Mol. Genet. 2005, 14, 483–492. [Google Scholar] [CrossRef]
  153. Makedonski, K.; Abuhatzira, L.; Kaufman, Y.; Razin, A.; Shemer, R. MeCP2 deficiency in Rett syndrome causes epigenetic aberrations at the PWS/AS imprinting center that affects UBE3A expression. Hum. Mol. Genet. 2005, 14, 1049–1058. [Google Scholar] [CrossRef] [PubMed]
  154. Kim, S.; Chahrour, M.; Ben-Shachar, S.; Lim, J. Ube3a/E6AP is involved in a subset of MeCP2 functions. Biochem. Biophys. Res. Commun. 2013, 437, 67–73. [Google Scholar] [CrossRef] [PubMed]
  155. Park, M.J.; Aja, S.; Li, Q.; Degano, A.L.; Penati, J.; Zhuo, J.; Roe, C.R.; Ronnett, G.V. Anaplerotic triheptanoin diet enhances mitochondrial substrate use to remodel the metabolome and improve lifespan, motor function, and sociability in MeCP2-null mice. PLoS ONE 2014, 9, e109527. [Google Scholar] [CrossRef]
  156. Senior, A.W.; Evans, R.; Jumper, J.; Kirkpatrick, J.; Sifre, L.; Green, T.; Qin, C.; Žídek, A.; Nelson, A.W.; Bridgland, A. Protein structure prediction using multiple deep neural networks in the 13th Critical Assessment of Protein Structure Prediction (CASP13). Proteins Struct. Funct. Bioinform. 2019, 87, 1141–1148. [Google Scholar] [CrossRef]
  157. Senior, A.W.; Evans, R.; Jumper, J.; Kirkpatrick, J.; Sifre, L.; Green, T.; Qin, C.; Žídek, A.; Nelson, A.W.; Bridgland, A. Improved protein structure prediction using potentials from deep learning. Nature 2020, 577, 706–710. [Google Scholar] [CrossRef]
  158. Mok, R.S.; Zhang, W.; Sheikh, T.I.; Pradeepan, K.; Fernandes, I.R.; DeJong, L.C.; Benigno, G.; Hildebrandt, M.R.; Mufteev, M.; Rodrigues, D.C. Wide spectrum of neuronal and network phenotypes in human stem cell-derived excitatory neurons with Rett syndrome-associated MECP2 mutations. Transl. Psychiatry 2022, 12, 450. [Google Scholar] [CrossRef]
  159. Lamothe, H.; Schreiweis, C.; Mondragón-González, L.S.; Rebbah, S.; Lavielle, O.; Mallet, L.; Burguière, E. The Sapap3−/− mouse reconsidered as a comorbid model expressing a spectrum of pathological repetitive behaviours. Transl. Psychiatry 2023, 13, 26. [Google Scholar] [CrossRef] [PubMed]
  160. Mahgoub, M.; Adachi, M.; Suzuki, K.; Liu, X.; Kavalali, E.T.; Chahrour, M.H.; Monteggia, L.M. MeCP2 and histone deacetylases 1 and 2 in dorsal striatum collectively suppress repetitive behaviors. Nat. Neurosci. 2016, 19, 1506–1512. [Google Scholar] [CrossRef]
  161. Chao, H.-T.; Chen, H.; Samaco, R.C.; Xue, M.; Chahrour, M.; Yoo, J.; Neul, J.L.; Gong, S.; Lu, H.-C.; Heintz, N. Dysfunction in GABA signalling mediates autism-like stereotypies and Rett syndrome phenotypes. Nature 2010, 468, 263–269. [Google Scholar] [CrossRef]
  162. Dani, V.S.; Chang, Q.; Maffei, A.; Turrigiano, G.G.; Jaenisch, R.; Nelson, S.B. Reduced cortical activity due to a shift in the balance between excitation and inhibition in a mouse model of Rett syndrome. Proc. Natl. Acad. Sci. USA 2005, 102, 12560–12565. [Google Scholar] [CrossRef]
  163. Medrihan, L.; Tantalaki, E.; Aramuni, G.; Sargsyan, V.; Dudanova, I.; Missler, M.; Zhang, W. Early defects of GABAergic synapses in the brain stem of a MeCP2 mouse model of Rett syndrome. J. Neurophysiol. 2008, 99, 112–121. [Google Scholar] [CrossRef]
  164. Samaco, R.C.; Mandel-Brehm, C.; Chao, H.-T.; Ward, C.S.; Fyffe-Maricich, S.L.; Ren, J.; Hyland, K.; Thaller, C.; Maricich, S.M.; Humphreys, P. Loss of MeCP2 in aminergic neurons causes cell-autonomous defects in neurotransmitter synthesis and specific behavioral abnormalities. Proc. Natl. Acad. Sci. USA 2009, 106, 21966–21971. [Google Scholar] [CrossRef] [PubMed]
  165. Thomas, A.; Burant, A.; Bui, N.; Graham, D.; Yuva-Paylor, L.A.; Paylor, R. Marble burying reflects a repetitive and perseverative behavior more than novelty-induced anxiety. Psychopharmacology 2009, 204, 361–373. [Google Scholar] [CrossRef]
  166. Fahn, S. The history of dopamine and levodopa in the treatment of Parkinson’s disease. Mov. Disord. 2008, 23, S497–S508. [Google Scholar] [CrossRef] [PubMed]
  167. Temudo, T.; Ramos, E.; Dias, K.; Barbot, C.; Vieira, J.P.; Moreira, A.; Calado, E.; Carrilho, I.; Oliveira, G.; Levy, A. Movement disorders in Rett syndrome: An analysis of 60 patients with detected MECP2 mutation and correlation with mutation type. Mov. Disord. 2008, 23, 1384–1390. [Google Scholar] [CrossRef]
  168. Crook, J.; Hendrickson, A.; Robinson, F. Co-localization of glycine and gaba immunoreactivity in interneurons in Macaca monkey cerebellar cortex. Neuroscience 2006, 141, 1951–1959. [Google Scholar] [CrossRef] [PubMed]
  169. Simat, M.; Parpan, F.; Fritschy, J.M. Heterogeneity of glycinergic and gabaergic interneurons in the granule cell layer of mouse cerebellum. J. Comp. Neurol. 2007, 500, 71–83. [Google Scholar] [CrossRef]
  170. Xing, H.; Cui, N.; Johnson, C.M.; Faisthalab, Z.; Jiang, C. Dual synaptic inhibitions of brainstem neurons by GABA and glycine with impact on Rett syndrome. J. Cell. Physiol. 2021, 236, 3615–3628. [Google Scholar] [CrossRef]
  171. Oldfors, A.; Sourander, P.; Armstrong, D.L.; Percy, A.K.; Witt-Engerström, I.; Hagberg, B.A. Rett syndrome: Cerebellar pathology. Pediatr. Neurol. 1990, 6, 310–314. [Google Scholar] [CrossRef]
  172. Murakami, J.W.; Courchesne, E.; Haas, R.; Press, G.; Yeung-Courchesne, R. Cerebellar and cerebral abnormalities in Rett syndrome: A quantitative MR analysis. AJR. Am. J. Roentgenol. 1992, 159, 177–183. [Google Scholar] [CrossRef]
  173. Ehrhart, F.; Coort, S.L.; Cirillo, E.; Smeets, E.; Evelo, C.T.; Curfs, L. New insights in Rett syndrome using pathway analysis for transcriptomics data. Wien. Med. Wochenschr. 2016, 166, 346. [Google Scholar] [CrossRef]
  174. Xu, F.-X.; Wang, X.-T.; Cai, X.-Y.; Liu, J.-Y.; Guo, J.-W.; Yang, F.; Chen, W.; Schonewille, M.; De Zeeuw, C.; Zhou, L. Purkinje-cell-specific MeCP2 deficiency leads to motor deficits and autistic-like behavior due to aberrations in PTP1B-TrkB-SK signaling. Cell Rep. 2023, 42, 113559. [Google Scholar] [CrossRef]
  175. Hagena, H.; Manahan-Vaughan, D. Interplay of hippocampal long-term potentiation and long-term depression in enabling memory representations. Philos. Trans. R. Soc. B Biol. Sci. 2024, 379, 20230229. [Google Scholar] [CrossRef] [PubMed]
  176. Belichenko, P.V.; Wright, E.E.; Belichenko, N.P.; Masliah, E.; Li, H.H.; Mobley, W.C.; Francke, U. Widespread changes in dendritic and axonal morphology in Mecp2-mutant mouse models of Rett syndrome: Evidence for disruption of neuronal networks. J. Comp. Neurol. 2009, 514, 240–258. [Google Scholar] [CrossRef]
  177. Nguyen, M.V.C.; Du, F.; Felice, C.A.; Shan, X.; Nigam, A.; Mandel, G.; Robinson, J.K.; Ballas, N. MeCP2 is critical for maintaining mature neuronal networks and global brain anatomy during late stages of postnatal brain development and in the mature adult brain. J. Neurosci. 2012, 32, 10021–10034. [Google Scholar] [CrossRef] [PubMed]
  178. Asaka, Y.; Jugloff, D.G.; Zhang, L.; Eubanks, J.H.; Fitzsimonds, R.M. Hippocampal synaptic plasticity is impaired in the Mecp2-null mouse model of Rett syndrome. Neurobiol. Dis. 2006, 21, 217–227. [Google Scholar] [CrossRef]
  179. Gogliotti, R.G.; Senter, R.K.; Fisher, N.M.; Adams, J.; Zamorano, R.; Walker, A.G.; Blobaum, A.L.; Engers, D.W.; Hopkins, C.R.; Daniels, J.S. mGlu7 potentiation rescues cognitive, social, and respiratory phenotypes in a mouse model of Rett syndrome. Sci. Transl. Med. 2017, 9, eaai7459. [Google Scholar] [CrossRef] [PubMed]
  180. Asgarihafshejani, A.; Raveendran, V.A.; Pressey, J.C.; Woodin, M.A. LTP is Absent in the CA1 Region of the Hippocampus of Male and Female Rett Syndrome Mouse Models. Neuroscience 2024, 537, 189–204. [Google Scholar] [CrossRef]
  181. Nelson, E.D.; Kavalali, E.T.; Monteggia, L.M. MeCP2-dependent transcriptional repression regulates excitatory neurotransmission. Curr. Biol. 2006, 16, 710–716. [Google Scholar] [CrossRef]
  182. Cohen, D.R.; Matarazzo, V.; Palmer, A.M.; Tu, Y.; Jeon, O.-H.; Pevsner, J.; Ronnett, G.V. Expression of MeCP2 in olfactory receptor neurons is developmentally regulated and occurs before synaptogenesis. Mol. Cell. Neurosci. 2003, 22, 417–429. [Google Scholar] [CrossRef] [PubMed]
  183. Palmer, A.; Qayumi, J.; Ronnett, G. MeCP2 mutation causes distinguishable phases of acute and chronic defects in synaptogenesis and maintenance, respectively. Molecular. Cell. Neurosci. 2008, 37, 794–807. [Google Scholar] [CrossRef]
  184. Ronnett, G.V.; Leopold, D.; Cai, X.; Hoffbuhr, K.C.; Moses, L.; Hoffman, E.P.; Naidu, S. Olfactory biopsies demonstrate a defect in neuronal development in Rett’s syndrome. Ann. Neurol. 2003, 54, 206–218. [Google Scholar] [CrossRef] [PubMed]
  185. Guideri, F.; Acampa, M. Sudden death and cardiac arrhythmias in Rett syndrome. Pediatr. Cardiol. 2005, 26, 111. [Google Scholar] [CrossRef]
  186. Axelrod, F.B.; Chelimsky, G.G.; Weese-Mayer, D.E. Pediatric autonomic disorders. Pediatrics 2006, 118, 309–321. [Google Scholar] [CrossRef]
  187. Lugaresi, E.; Cirignotta, F.; Montagna, P. Abnormal breathing in the Rett syndrome. Brain Dev. 1985, 7, 329–333. [Google Scholar] [CrossRef] [PubMed]
  188. Schlüter, B.; Aguigah, G.; Buschatz, D.; Trowitzsch, E.; Aksu, F. Polysomnographic recordings of respiratory disturbances in Rett syndrome. J. Sleep Res. 1995, 4, 203–207. [Google Scholar] [CrossRef]
  189. Cirignotta, F.; Lugaresi, E.; Montagna, P.; Opitz, J.M.; Reynolds, J.F. Breathing impairment in Rett syndrome. Am. J. Med. Genet. 1986, 25, 167–173. [Google Scholar] [CrossRef]
  190. Weese-Mayer, D.E.; Lieske, S.P.; Boothby, C.M.; Kenny, A.S.; Bennett, H.L.; Silvestri, J.M.; Ramirez, J.-M. Autonomic nervous system dysregulation: Breathing and heart rate perturbation during wakefulness in young girls with Rett syndrome. Pediatr. Res. 2006, 60, 443–449. [Google Scholar] [CrossRef]
  191. Richter, D.W.; Manzke, T.; Wilken, B.; Ponimaskin, E. Serotonin receptors: Guardians of stable breathing. Trends Mol. Med. 2003, 9, 542–548. [Google Scholar] [CrossRef]
  192. Hodges, M.R.; Wehner, M.; Aungst, J.; Smith, J.C.; Richerson, G.B. Transgenic mice lacking serotonin neurons have severe apnea and high mortality during development. J. Neurosci. 2009, 29, 10341–10349. [Google Scholar] [CrossRef]
  193. Abdala, A.P.; Bissonnette, J.M.; Newman-Tancredi, A. Pinpointing brainstem mechanisms responsible for autonomic dysfunction in Rett syndrome: Therapeutic perspectives for 5-HT1A agonists. Front. Physiol. 2014, 5, 205. [Google Scholar] [CrossRef]
  194. He, F.; Sun, Y.E. Glial cells more than support cells? Int. J. Biochem. Cell Biol. 2007, 39, 661–665. [Google Scholar] [CrossRef]
  195. He, L.; Zhang, R.; Yang, M.; Lu, M. The role of astrocyte in neuroinflammation in traumatic brain injury. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2024, 1870, 166992. [Google Scholar] [CrossRef]
  196. Barres, B.A. The mystery and magic of glia: A perspective on their roles in health and disease. Neuron 2008, 60, 430–440. [Google Scholar] [CrossRef] [PubMed]
  197. Andoh-Noda, T.; Akamatsu, W.; Miyake, K.; Matsumoto, T.; Yamaguchi, R.; Sanosaka, T.; Okada, Y.; Kobayashi, T.; Ohyama, M.; Nakashima, K. Differentiation of multipotent neural stem cells derived from Rett syndrome patients is biased toward the astrocytic lineage. Mol. Brain 2015, 8, 31. [Google Scholar] [CrossRef] [PubMed]
  198. Forbes-Lorman, R.M.; Kurian, J.R.; Auger, A.P. MeCP2 regulates GFAP expression within the developing brain. Brain Res. 2014, 1543, 151–158. [Google Scholar] [CrossRef] [PubMed]
  199. Messing, A.; Head, M.W.; Galles, K.; Galbreath, E.J.; Goldman, J.E.; Brenner, M. Fatal encephalopathy with astrocyte inclusions in GFAP transgenic mice. Am. J. Pathol. 1998, 152, 391–398. [Google Scholar]
  200. Maezawa, I.; Swanberg, S.; Harvey, D.; LaSalle, J.M.; Jin, L.-W. Rett syndrome astrocytes are abnormal and spread MeCP2 deficiency through gap junctions. J. Neurosci. 2009, 29, 5051–5061. [Google Scholar] [CrossRef]
  201. Sheldon, A.L.; Robinson, M.B. The role of glutamate transporters in neurodegenerative diseases and potential opportunities for intervention. Neurochem. Int. 2007, 51, 333–355. [Google Scholar] [CrossRef]
  202. Lehmann, C.; Bette, S.; Engele, J. High extracellular glutamate modulates expression of glutamate transporters and glutamine synthetase in cultured astrocytes. Brain Res. 2009, 1297, 1–8. [Google Scholar] [CrossRef]
  203. Eid, T.; Williamson, A.; Lee, T.S.W.; Petroff, O.A.; De Lanerolle, N.C. Glutamate and astrocytes—Key players in human mesial temporal lobe epilepsy? Epilepsia 2008, 49, 42–52. [Google Scholar] [CrossRef]
  204. Okabe, Y.; Takahashi, T.; Mitsumasu, C.; Kosai, K.-i.; Tanaka, E.; Matsuishi, T. Alterations of gene expression and glutamate clearance in astrocytes derived from an MeCP2-null mouse model of Rett syndrome. PLoS ONE 2012, 7, e35354. [Google Scholar] [CrossRef]
  205. Müller, M.; Can, K. Aberrant redox homoeostasis and mitochondrial dysfunction in Rett syndrome. Biochem. Soc. Trans. 2014, 42, 959–964. [Google Scholar] [CrossRef]
  206. Tomasello, D.L.; Barrasa, M.I.; Mankus, D.; Alarcon, K.I.; Lytton-Jean, A.K.; Liu, X.S.; Jaenisch, R. Mitochondrial dysfunction and increased reactive oxygen species production in MECP2 mutant astrocytes and their impact on neurons. Sci. Rep. 2024, 14, 20565. [Google Scholar] [CrossRef] [PubMed]
  207. Jin, L.-W.; Horiuchi, M.; Wulff, H.; Liu, X.-B.; Cortopassi, G.A.; Erickson, J.D.; Maezawa, I. Dysregulation of glutamine transporter SNAT1 in Rett syndrome microglia: A mechanism for mitochondrial dysfunction and neurotoxicity. J. Neurosci. 2015, 35, 2516–2529. [Google Scholar] [CrossRef] [PubMed]
  208. Hayakawa, K.; Esposito, E.; Wang, X.; Terasaki, Y.; Liu, Y.; Xing, C.; Ji, X.; Lo, E.H. Transfer of mitochondria from astrocytes to neurons after stroke. Nature 2016, 535, 551–555. [Google Scholar] [CrossRef] [PubMed]
  209. Lampinen, R.; Belaya, I.; Saveleva, L.; Liddell, J.R.; Rait, D.; Huuskonen, M.T.; Giniatullina, R.; Sorvari, A.; Soppela, L.; Mikhailov, N. Neuron-astrocyte transmitophagy is altered in Alzheimer’s disease. Neurobiol. Dis. 2022, 170, 105753. [Google Scholar] [CrossRef]
  210. Sharma, K.; Singh, J.; Pillai, P.P. MeCP2 differentially regulate the myelin MBP and PLP protein expression in oligodendrocytes and C6 glioma. J. Mol. Neurosci. 2018, 65, 343–350. [Google Scholar] [CrossRef]
  211. Colantuoni, C.; Jeon, O.H.; Hyder, K.; Chenchik, A.; Khimani, A.H.; Narayanan, V.; Hoffman, E.P.; Kaufmann, W.E.; Naidu, S.; Pevsner, J. Gene expression profiling in postmortem Rett Syndrome brain: Differential gene expression and patient classification. Neurobiol. Dis. 2001, 8, 847–865. [Google Scholar] [CrossRef]
  212. Nguyen, M.V.; Felice, C.A.; Du, F.; Covey, M.V.; Robinson, J.K.; Mandel, G.; Ballas, N. Oligodendrocyte lineage cells contribute unique features to Rett syndrome neuropathology. J. Neurosci. 2013, 33, 18764–18774. [Google Scholar] [CrossRef] [PubMed]
  213. Alessio, N.; Riccitiello, F.; Squillaro, T.; Capasso, S.; Del Gaudio, S.; Di Bernardo, G.; Cipollaro, M.; Melone, M.A.B.; Peluso, G.; Galderisi, U. Neural stem cells from a mouse model of Rett syndrome are prone to senescence, show reduced capacity to cope with genotoxic stress, and are impaired in the differentiation process. Exp. Mol. Med. 2018, 50, 1. [Google Scholar] [CrossRef] [PubMed]
  214. Kahanovitch, U.; Patterson, K.C.; Hernandez, R.; Olsen, M.L. Glial Dysfunction in MeCP2 Deficiency Models: Implications for Rett Syndrome. Int. J. Mol. Sci. 2019, 20, 3813. [Google Scholar] [CrossRef] [PubMed]
  215. Lipi, B.; Jaldeep, L.; Prakash, P. Role of astrocytic MeCP2 in regulation of CNS myelination by affecting oligodendrocyte and neuronal physiology and axo–glial interactions. Exp. Brain Res. 2018, 236, 3015–3027. [Google Scholar] [CrossRef]
  216. Yoon, S.H.; Kim, G.Y.; Choi, G.T.; Do, J.T. Organ abnormalities caused by Turner syndrome. Cells 2023, 12, 1365. [Google Scholar] [CrossRef]
  217. McCauley, M.D.; Wang, T.; Mike, E.; Herrera, J.; Beavers, D.L.; Huang, T.-W.; Ward, C.S.; Skinner, S.; Percy, A.K.; Glaze, D.G. Pathogenesis of lethal cardiac arrhythmias in Mecp2 mutant mice: Implication for therapy in Rett syndrome. Sci. Transl. Med. 2011, 3, 113ra125. [Google Scholar] [CrossRef]
  218. Hara, M.; Takahashi, T.; Mitsumasu, C.; Igata, S.; Takano, M.; Minami, T.; Yasukawa, H.; Okayama, S.; Nakamura, K.; Okabe, Y. Disturbance of cardiac gene expression and cardiomyocyte structure predisposes Mecp2-null mice to arrhythmias. Sci. Rep. 2015, 5, 11204. [Google Scholar] [CrossRef]
  219. Ward, C.S.; Huang, T.-W.; Herrera, J.A.; Samaco, R.C.; Pitcher, M.R.; Herron, A.; Skinner, S.A.; Kaufmann, W.E.; Glaze, D.G.; Percy, A.K. Loss of MeCP2 causes urological dysfunction and contributes to death by kidney failure in mouse models of Rett syndrome. PLoS ONE 2016, 11, e0165550. [Google Scholar] [CrossRef]
  220. Wang, J.; Xiong, M.; Fan, Y.; Liu, C.; Wang, Q.; Yang, D.; Yuan, Y.; Huang, Y.; Wang, S.; Zhang, Y. Mecp2 protects kidney from ischemia-reperfusion injury through transcriptional repressing IL-6/STAT3 signaling. Theranostics 2022, 12, 3896–3910. [Google Scholar] [CrossRef]
  221. Cronk, J.C.; Derecki, N.C.; Ji, E.; Xu, Y.; Lampano, A.E.; Smirnov, I.; Baker, W.; Norris, G.T.; Marin, I.; Coddington, N. Methyl-CpG binding protein 2 regulates microglia and macrophage gene expression in response to inflammatory stimuli. Immunity 2015, 42, 679–691. [Google Scholar] [CrossRef]
  222. Zalosnik, M.; Fabio, M.; Bertoldi, M.; Castañares, C.; Degano, A. MeCP2 deficiency exacerbates the neuroinflammatory setting and autoreactive response during an autoimmune challenge. Sci. Rep. 2021, 11, 10997. [Google Scholar] [CrossRef]
  223. Kida, H.; Takahashi, T.; Nakamura, Y.; Kinoshita, T.; Hara, M.; Okamoto, M.; Okayama, S.; Nakamura, K.; Kosai, K.I.; Taniwaki, T.; et al. Pathogenesis of Lethal Aspiration Pneumonia in Mecp2-null Mouse Model for Rett Syndrome. Sci. Rep. 2017, 7, 12032. [Google Scholar] [CrossRef] [PubMed]
  224. De Felice, C.; Guazzi, G.; Rossi, M.; Ciccoli, L.; Signorini, C.; Leoncini, S.; Tonni, G.; Latini, G.; Valacchi, G.; Hayek, J. Unrecognized lung disease in classic Rett syndrome: A physiologic and high-resolution CT imaging study. Chest 2010, 138, 386–392. [Google Scholar] [CrossRef]
  225. Vashi, N.; Ackerley, C.; Post, M.; Justice, M.J. Aberrant lung lipids cause respiratory impairment in a Mecp2-deficient mouse model of Rett syndrome. Hum. Mol. Genet. 2021, 30, 2161–2176. [Google Scholar] [CrossRef] [PubMed]
  226. Panayotis, N.; Ehinger, Y.; Felix, M.S.; Roux, J.c. State-of-the-art therapies for Rett syndrome. Dev. Med. Child Neurol. 2023, 65, 162–170. [Google Scholar] [CrossRef] [PubMed]
  227. Matagne, V.; Ehinger, Y.; Saidi, L.; Borges-Correia, A.; Barkats, M.; Bartoli, M.; Villard, L.; Roux, J.C. A codon-optimized Mecp2 transgene corrects breathing deficits and improves survival in a mouse model of Rett syndrome. Neurobiol. Dis. 2017, 99, 1–11. [Google Scholar] [CrossRef]
  228. Matagne, V.; Borloz, E.; Ehinger, Y.; Saidi, L.; Villard, L.; Roux, J.-C. Severe offtarget effects following intravenous delivery of AAV9-MECP2 in a female mouse model of Rett syndrome. Neurobiol. Dis. 2021, 149, 105235. [Google Scholar] [CrossRef]
  229. Gadalla, K.K.E.; Vudhironarit, T.; Hector, R.D.; Sinnett, S.; Bahey, N.G.; Bailey, M.E.S.; Gray, S.J.; Cobb, S.R. Development of a Novel AAV Gene Therapy Cassette with Improved Safety Features and Efficacy in a Mouse Model of Rett Syndrome. Mol. Ther. Methods Clin. Dev. 2017, 5, 180–190. [Google Scholar] [CrossRef]
  230. Deverman, B.E.; Pravdo, P.L.; Simpson, B.P.; Kumar, S.R.; Chan, K.Y.; Banerjee, A.; Wu, W.L.; Yang, B.; Huber, N.; Pasca, S.P.; et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat. Biotechnol. 2016, 34, 204–209. [Google Scholar] [CrossRef]
  231. Morabito, G.; Giannelli, S.G.; Ordazzo, G.; Bido, S.; Castoldi, V.; Indrigo, M.; Cabassi, T.; Cattaneo, S.; Luoni, M.; Cancellieri, C.; et al. AAV-PHP.B-Mediated Global-Scale Expression in the Mouse Nervous System Enables GBA1 Gene Therapy for Wide Protection from Synucleinopathy. Mol. Ther. 2017, 25, 2727–2742. [Google Scholar] [CrossRef]
  232. Luoni, M.; Giannelli, S.; Indrigo, M.T.; Niro, A.; Massimino, L.; Iannielli, A.; Passeri, L.; Russo, F.; Morabito, G.; Calamita, P.; et al. Whole brain delivery of an instability-prone Mecp2 transgene improves behavioral and molecular pathological defects in mouse models of Rett syndrome. eLife 2020, 9, e52629. [Google Scholar] [CrossRef]
  233. Goertsen, D.; Flytzanis, N.C.; Goeden, N.; Chuapoco, M.R.; Cummins, A.; Chen, Y.; Fan, Y.; Zhang, Q.; Sharma, J.; Duan, Y.; et al. AAV capsid variants with brain-wide transgene expression and decreased liver targeting after intravenous delivery in mouse and marmoset. Nat. Neurosci. 2022, 25, 106–115. [Google Scholar] [CrossRef]
  234. Sadhu, C.; Lyons, C.; Oh, J.; Jagadeeswaran, I.; Gray, S.J.; Sinnett, S.E. The Efficacy of a Human-Ready miniMECP2 Gene Therapy in a Pre-Clinical Model of Rett Syndrome. Genes 2023, 15, 31. [Google Scholar] [CrossRef]
  235. Nishikura, K. Functions and regulation of RNA editing by ADAR deaminases. Annu. Rev. Biochem. 2010, 79, 321–349. [Google Scholar] [CrossRef]
  236. Paul, M.S.; Bass, B.L. Inosine exists in mRNA at tissue-specific levels and is most abundant in brain mRNA. EMBO J. 1998, 17, 1120–1127. [Google Scholar] [CrossRef]
  237. Sinnamon, J.R.; Kim, S.Y.; Corson, G.M.; Song, Z.; Nakai, H.; Adelman, J.P.; Mandel, G. Site-directed RNA repair of endogenous Mecp2 RNA in neurons. Proc. Natl. Acad. Sci. USA 2017, 114, E9395–E9402. [Google Scholar] [CrossRef]
  238. Sinnamon, J.R.; Kim, S.Y.; Fisk, J.R.; Song, Z.; Nakai, H.; Jeng, S.; McWeeney, S.K.; Mandel, G. In Vivo Repair of a Protein Underlying a Neurological Disorder by Programmable RNA Editing. Cell Rep. 2020, 32, 107878. [Google Scholar] [CrossRef]
  239. Sinnamon, J.R.; Jacobson, M.E.; Yung, J.F.; Fisk, J.R.; Jeng, S.; McWeeney, S.K.; Parmelee, L.K.; Chan, C.N.; Yee, S.P.; Mandel, G. Targeted RNA editing in brainstem alleviates respiratory dysfunction in a mouse model of Rett syndrome. Proc. Natl. Acad. Sci. USA 2022, 119, e2206053119. [Google Scholar] [CrossRef]
  240. Gadalla, K.K.; Bailey, M.E.; Cobb, S.R. MeCP2 and Rett syndrome: Reversibility and potential avenues for therapy. Biochem. J. 2011, 439, 1–14. [Google Scholar] [CrossRef]
  241. Carrette, L.L.G.; Wang, C.Y.; Wei, C.; Press, W.; Ma, W.; Kelleher, R.J., 3rd; Lee, J.T. A mixed modality approach towards Xi reactivation for Rett syndrome and other X-linked disorders. Proc. Natl. Acad. Sci. USA 2018, 115, E668–E675. [Google Scholar] [CrossRef]
  242. Przanowski, P.; Wasko, U.; Zheng, Z.; Yu, J.; Sherman, R.; Zhu, L.J.; McConnell, M.J.; Tushir-Singh, J.; Green, M.R.; Bhatnagar, S. Pharmacological reactivation of inactive X-linked Mecp2 in cerebral cortical neurons of living mice. Proc. Natl. Acad. Sci. USA 2018, 115, 7991–7996. [Google Scholar] [CrossRef]
  243. Qian, J.; Guan, X.; Xie, B.; Xu, C.; Niu, J.; Tang, X.; Li, C.H.; Colecraft, H.M.; Jaenisch, R.; Liu, X.S. Multiplex epigenome editing of MECP2 to rescue Rett syndrome neurons. Sci. Transl. Med. 2023, 15, eadd4666. [Google Scholar] [CrossRef]
  244. De Felice, C.; Della Ragione, F.; Signorini, C.; Leoncini, S.; Pecorelli, A.; Ciccoli, L.; Scalabri, F.; Marracino, F.; Madonna, M.; Belmonte, G.; et al. Oxidative brain damage in Mecp2-mutant murine models of Rett syndrome. Neurobiol. Dis. 2014, 68, 66–77. [Google Scholar] [CrossRef]
  245. Leoncini, S.; De Felice, C.; Signorini, C.; Zollo, G.; Cortelazzo, A.; Durand, T.; Galano, J.M.; Guerranti, R.; Rossi, M.; Ciccoli, L.; et al. Cytokine Dysregulation in MECP2- and CDKL5-Related Rett Syndrome: Relationships with Aberrant Redox Homeostasis, Inflammation, and omega-3 PUFAs. Oxid. Med. Cell. Longev. 2015, 2015, 421624. [Google Scholar] [CrossRef] [PubMed]
  246. Nance, E.; Kambhampati, S.P.; Smith, E.S.; Zhang, Z.; Zhang, F.; Singh, S.; Johnston, M.V.; Kannan, R.M.; Blue, M.E.; Kannan, S. Dendrimer-mediated delivery of N-acetyl cysteine to microglia in a mouse model of Rett syndrome. J. Neuroinflamm. 2017, 14, 252. [Google Scholar] [CrossRef] [PubMed]
  247. Cao, Z.; Min, X.; Xie, X.; Huang, M.; Liu, Y.; Sun, W.; Xu, G.; He, M.; He, K.; Li, Y.; et al. RIPK1 activation in Mecp2-deficient microglia promotes inflammation and glutamate release in RTT. Proc. Natl. Acad. Sci. USA 2024, 121, e2320383121. [Google Scholar] [CrossRef]
  248. Neul, J.L.; Percy, A.K.; Benke, T.A.; Berry-Kravis, E.M.; Glaze, D.G.; Marsh, E.D.; Lin, T.; Stankovic, S.; Bishop, K.M.; Youakim, J.M. Trofinetide for the treatment of Rett syndrome: A randomized phase 3 study. Nat. Med. 2023, 29, 1468–1475. [Google Scholar] [CrossRef] [PubMed]
  249. Hudu, S.A.; Elmigdadi, F.; Qtaitat, A.A.; Almehmadi, M.; Alsaiari, A.A.; Allahyani, M.; Aljuaid, A.; Salih, M.; Alghamdi, A.; Alrofaidi, M.A. Trofinetide for Rett syndrome: Highlights on the development and related inventions of the first USFDA-approved treatment for rare pediatric unmet medical need. J. Clin. Med. 2023, 12, 5114. [Google Scholar] [CrossRef]
  250. Mohammed, H.E.; Bady, Z.; Haseeb, M.E.; Aboeldahab, H.; Sharaf-Eldin, W.E.; Zaki, M.S. Is trofinetide a future treatment for Rett syndrome? A comprehensive systematic review and meta-analysis of randomized controlled trials. BMC Med. 2024, 22, 299. [Google Scholar] [CrossRef]
  251. Sansal, I.; Sellers, W.R. The biology and clinical relevance of the PTEN tumor suppressor pathway. J. Clin. Oncol. 2004, 22, 2954–2963. [Google Scholar] [CrossRef]
  252. Ricciardi, S.; Boggio, E.M.; Grosso, S.; Lonetti, G.; Forlani, G.; Stefanelli, G.; Calcagno, E.; Morello, N.; Landsberger, N.; Biffo, S. Reduced AKT/mTOR signaling and protein synthesis dysregulation in a Rett syndrome animal model. Hum. Mol. Genet. 2011, 20, 1182–1196. [Google Scholar] [CrossRef]
  253. Blue, M.E.; Kaufmann, W.E.; Bressler, J.; Eyring, C.; O’Driscoll, C.; Naidu, S.; Johnston, M.V. Temporal and regional alterations in NMDA receptor expression in Mecp2-null mice. Anat. Rec. 2011, 294, 1624–1634. [Google Scholar] [CrossRef]
  254. Patrizi, A.; Picard, N.; Simon, A.J.; Gunner, G.; Centofante, E.; Andrews, N.A.; Fagiolini, M. Chronic Administration of the N-Methyl-D-Aspartate Receptor Antagonist Ketamine Improves Rett Syndrome Phenotype. Biol. Psychiatry 2016, 79, 755–764. [Google Scholar] [CrossRef]
  255. Chen, W.G.; Chang, Q.; Lin, Y.; Meissner, A.; West, A.E.; Griffith, E.C.; Jaenisch, R.; Greenberg, M.E. Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2. Science 2003, 302, 885–889. [Google Scholar] [CrossRef] [PubMed]
  256. Ogier, M.; Wang, H.; Hong, E.; Wang, Q.; Greenberg, M.E.; Katz, D.M. Brain-derived neurotrophic factor expression and respiratory function improve after ampakine treatment in a mouse model of Rett syndrome. J. Neurosci. 2007, 27, 10912–10917. [Google Scholar] [CrossRef] [PubMed]
  257. Mohanta, S.K.; Yin, C.; Weber, C.; Godinho-Silva, C.; Veiga-Fernandes, H.; Xu, Q.J.; Chang, R.B.; Habenicht, A.J.R. Cardiovascular Brain Circuits. Circ. Res. 2023, 132, 1546–1565. [Google Scholar] [CrossRef]
  258. Adams, I.; Yang, T.; Longo, F.M.; Katz, D.M. Restoration of motor learning in a mouse model of Rett syndrome following long-term treatment with a novel small-molecule activator of TrkB. Dis. Models Mech. 2020, 13, dmm044685. [Google Scholar] [CrossRef]
  259. Fazzari, M.; Lunghi, G.; Carsana, E.V.; Valsecchi, M.; Spiombi, E.; Breccia, M.; Casati, S.R.; Pedretti, S.; Mitro, N.; Mauri, L. GM1 Oligosaccharide Ameliorates Rett Syndrome Phenotypes In Vitro and In Vivo via Trk Receptor Activation. Int. J. Mol. Sci. 2024, 25, 11555. [Google Scholar] [CrossRef] [PubMed]
  260. Schmid, D.A.; Yang, T.; Ogier, M.; Adams, I.; Mirakhur, Y.; Wang, Q.; Massa, S.M.; Longo, F.M.; Katz, D.M. A TrkB small molecule partial agonist rescues TrkB phosphorylation deficits and improves respiratory function in a mouse model of Rett syndrome. J. Neurosci. 2012, 32, 1803–1810. [Google Scholar] [CrossRef]
  261. Levitt, E.S.; Hunnicutt, B.J.; Knopp, S.J.; Williams, J.T.; Bissonnette, J.M. A selective 5-HT1a receptor agonist improves respiration in a mouse model of Rett syndrome. J. Appl. Physiol. 2013, 115, 1626–1633. [Google Scholar] [CrossRef]
  262. Newman-Tancredi, A. Biased agonism at serotonin 5-HT1A receptors: Preferential postsynaptic activity for improved therapy of CNS disorders. Neuropsychiatry 2011, 1, 149. [Google Scholar] [CrossRef]
  263. Frasca, A.; Miramondi, F.; Butti, E.; Indrigo, M.; Balbontin Arenas, M.; Postogna, F.M.; Piffer, A.; Bedogni, F.; Pizzamiglio, L.; Cambria, C.; et al. Neural precursor cells rescue symptoms of Rett syndrome by activation of the Interferon gamma pathway. EMBO Mol. Med. 2024, 16, 3218–3246. [Google Scholar] [CrossRef] [PubMed]
  264. Hao, S.; Tang, B.; Wu, Z.; Ure, K.; Sun, Y.; Tao, H.; Gao, Y.; Patel, A.J.; Curry, D.J.; Samaco, R.C.; et al. Forniceal deep brain stimulation rescues hippocampal memory in Rett syndrome mice. Nature 2015, 526, 430–434. [Google Scholar] [CrossRef]
  265. Tang, B.; Wu, Z.; Wang, Q.; Tang, J. Neuronal Network Activation Induced by Forniceal Deep Brain Stimulation in Mice. Genes 2025, 16, 210. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Regulation of X chromosome expression during early embryonic development and the effects of methyl-CpG-binding protein 2 (MECP2) mutations in mice and humans. (A) In mice, paternal X chromosome inactivation occurred during preimplantation development during the preimplantation stages as the embryo develops into a blastocyst. X chromosome reactivation occurs only in the epiblast of the late blastocyst stage. In humans, zygotic genome activation takes place at the 8-cell stage and X chromosome dampening begins at the morula stage. In the post-implantation stage, both mouse and human embryos exhibit inactivation of either the maternal or paternal X chromosome, which leads to a mosaic pattern of X chromosome expression. (B) The effects of MECP2 mutations differ depending on sex in both mice and humans. Male mice and humans with a MECP2 mutation (MECP2−/y) show different phenotypes. In females, the severity of the phenotype increases when the proportion of cells expressing the mutant MECP2 allele becomes higher; Cells expressing mutant MECP2 (pink); cells expressing normal MECP2 (light blue).
Figure 1. Regulation of X chromosome expression during early embryonic development and the effects of methyl-CpG-binding protein 2 (MECP2) mutations in mice and humans. (A) In mice, paternal X chromosome inactivation occurred during preimplantation development during the preimplantation stages as the embryo develops into a blastocyst. X chromosome reactivation occurs only in the epiblast of the late blastocyst stage. In humans, zygotic genome activation takes place at the 8-cell stage and X chromosome dampening begins at the morula stage. In the post-implantation stage, both mouse and human embryos exhibit inactivation of either the maternal or paternal X chromosome, which leads to a mosaic pattern of X chromosome expression. (B) The effects of MECP2 mutations differ depending on sex in both mice and humans. Male mice and humans with a MECP2 mutation (MECP2−/y) show different phenotypes. In females, the severity of the phenotype increases when the proportion of cells expressing the mutant MECP2 allele becomes higher; Cells expressing mutant MECP2 (pink); cells expressing normal MECP2 (light blue).
Ijms 26 08277 g001
Figure 2. Protein structure of methyl-CpG-binding protein 2 (MECP2) and gene regulation mechanisms. (A) Schematic of MECP2 protein and common pathogenic mutations. MECP2 has functional domains including a methyl-CpG-binding domain (MBD), an intervening domain (ID), a transcriptional repression domain (TRD), an NCoR–SMRT interaction domain (NID), and three AT-hook domains. NTD, N-terminal domain; CTD, C-terminal domain. (B) Mechanisms of MECP2-mediated gene regulation under normal and mutant conditions. (1) Chromatin compaction and DNA loop formation—Normal MECP2 binds methylated and unmethylated DNA, compacting chromatin in a histone H1-like manner, and forms DNA loops at loci such as DLX5–DLX6 to induce biallelic expression. Mutations in the AT-hook 2 region (e.g., R270X, G273X) impair this chromatin compaction. G273X retains partial function relative to R270X, indicating that amino acids 270–272 are important for AT-rich DNA binding. (2) Co-repressor recruitment—MECP2 interacts with NCoR and SMRT via the NID to repress transcription. The R306C mutation disrupts NCoR–SMRT interaction despite intact DNA binding, leading to partial loss of repression. (3) Transcriptional activation—MECP2 can also activate gene expression by recruiting CREB1 to target promoters. MBD mutations (e.g., R106W, R133C abolish DNA binding, severely altering gene expression. (4) Post-transcriptional regulation—MECP2 interacts with splicing factors such as YB-1 to influence alternative splicing. MECP2 binds DGCR8, inhibiting Drosha–DGCR8 complex formation to repress miRNA processing, while loss of MECP2 elevates miRNA levels in certain brain regions. In null mutations, MECP2 expression is completely absent, resulting in loss of all regulatory mechanisms described above.
Figure 2. Protein structure of methyl-CpG-binding protein 2 (MECP2) and gene regulation mechanisms. (A) Schematic of MECP2 protein and common pathogenic mutations. MECP2 has functional domains including a methyl-CpG-binding domain (MBD), an intervening domain (ID), a transcriptional repression domain (TRD), an NCoR–SMRT interaction domain (NID), and three AT-hook domains. NTD, N-terminal domain; CTD, C-terminal domain. (B) Mechanisms of MECP2-mediated gene regulation under normal and mutant conditions. (1) Chromatin compaction and DNA loop formation—Normal MECP2 binds methylated and unmethylated DNA, compacting chromatin in a histone H1-like manner, and forms DNA loops at loci such as DLX5–DLX6 to induce biallelic expression. Mutations in the AT-hook 2 region (e.g., R270X, G273X) impair this chromatin compaction. G273X retains partial function relative to R270X, indicating that amino acids 270–272 are important for AT-rich DNA binding. (2) Co-repressor recruitment—MECP2 interacts with NCoR and SMRT via the NID to repress transcription. The R306C mutation disrupts NCoR–SMRT interaction despite intact DNA binding, leading to partial loss of repression. (3) Transcriptional activation—MECP2 can also activate gene expression by recruiting CREB1 to target promoters. MBD mutations (e.g., R106W, R133C abolish DNA binding, severely altering gene expression. (4) Post-transcriptional regulation—MECP2 interacts with splicing factors such as YB-1 to influence alternative splicing. MECP2 binds DGCR8, inhibiting Drosha–DGCR8 complex formation to repress miRNA processing, while loss of MECP2 elevates miRNA levels in certain brain regions. In null mutations, MECP2 expression is completely absent, resulting in loss of all regulatory mechanisms described above.
Ijms 26 08277 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Choi, G.; Lee, S.; Yoo, S.; Do, J.T. MECP2 Dysfunction in Rett Syndrome: Molecular Mechanisms, Multisystem Pathology, and Emerging Therapeutic Strategies. Int. J. Mol. Sci. 2025, 26, 8277. https://doi.org/10.3390/ijms26178277

AMA Style

Choi G, Lee S, Yoo S, Do JT. MECP2 Dysfunction in Rett Syndrome: Molecular Mechanisms, Multisystem Pathology, and Emerging Therapeutic Strategies. International Journal of Molecular Sciences. 2025; 26(17):8277. https://doi.org/10.3390/ijms26178277

Chicago/Turabian Style

Choi, Gyutae, Sanghyo Lee, Seungjae Yoo, and Jeung Tae Do. 2025. "MECP2 Dysfunction in Rett Syndrome: Molecular Mechanisms, Multisystem Pathology, and Emerging Therapeutic Strategies" International Journal of Molecular Sciences 26, no. 17: 8277. https://doi.org/10.3390/ijms26178277

APA Style

Choi, G., Lee, S., Yoo, S., & Do, J. T. (2025). MECP2 Dysfunction in Rett Syndrome: Molecular Mechanisms, Multisystem Pathology, and Emerging Therapeutic Strategies. International Journal of Molecular Sciences, 26(17), 8277. https://doi.org/10.3390/ijms26178277

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop