Skip to main content

Development of neurodevelopmental disorders: a regulatory mechanism involving bromodomain-containing proteins

Abstract

Neurodevelopmental disorders are classified as diseases that cause abnormal functions of the brain or central nervous system. Children with neurodevelopmental disorders show impaired language and speech abilities, learning and memory damage, and poor motor skills. However, we still know very little about the molecular etiology of these disorders. Recent evidence implicates the bromodomain-containing proteins (BCPs) in the initiation and development of neurodevelopmental disorders. BCPs have a particular domain, the bromodomain (Brd), which was originally identified as specifically binding acetyl-lysine residues at the N-terminus of histone proteins in vitro and in vivo. Other domains of BCPs are responsible for binding partner proteins to form regulatory complexes. Once these complexes are assembled, BCPs alter chromosomal states and regulate gene expression. Some BCP complexes bind nucleosomes, are involved in basal transcription regulation, and influence the transcription of many genes. However, most BCPs are involved in targeting. For example, some BCPs function as a recruitment platform or scaffold through their Brds-binding targeting sites. Others are recruited to form a complex to bind the targeting sites of their partners. The regulation mediated by these proteins is especially critical during normal and abnormal development. Mutant BCPs or dysfunctional BCP-containing complexes are implicated in the initiation and development of neurodevelopmental disorders. However, the pathogenic molecular mechanisms are not fully understood. In this review, we focus on the roles of regulatory BCPs associated with neurodevelopmental disorders, including mental retardation, Fragile X syndrome (FRX), Williams syndrome (WS), Rett syndrome and Rubinstein-Taybi syndrome (RTS). A better understanding of the molecular pathogenesis, based upon the roles of BCPs, will lead to screening of targets for the treatment of neurodevelopmental disorders.

Review

Neurodevelopmental disorders are complicated diseases. Studies are currently underway to understand their regulatory mechanisms, to explore better treatments and, ultimately, to prevent their onset.

Chromatin modification traits are associated with the pathogenic characteristics of many neurodevelopmental disorders. Bromodomain-containing proteins (BCPs), which contain a bromodomain (Brd), represent a family of proteins found in many chromatin-associated proteins and in a range of transcription factors. A hydrophobic pocket formed by the ZA loop and BC loop is responsible for acetyl-lysine recognition and specific binding, the accuracy of which could be improved by sequence and acetyl-lysine modification of target sites. Thus, several inhibitors targeting these interactions have been designed for potential clinical application.

BCPs play pivotal roles in transcription regulation. Some BCPs participate in initiation and regulation of basal transcription by binding nucleosomes. However, as targeting proteins, some BCPs can recruit their binding substrates and bind their target through the Brds as the ‘reader’ or a recruitment platform for modular protein complexes. They regulate gene transcription by both activation and repression. Conversely, some BCPs can be recruited by other transcription factors (such as p53), and function as modifying enzymes. Consequently, this affects the transcription of downstream genes of other transcription factors.

Abnormal BCPs or BCP-containing complexes in the nervous system have been shown to cause neurodevelopmental disorders, including neural tube defects (for example, GCN5 or BRD2), mental retardation (for example, BRWD3), Williams syndrome (WS; for example, BAZ1B), Rett syndrome (for example, SWI/SNF), Rett syndrome and Rubinstein-Taybi syndrome (RTS; for example, CBP/p300), and Fragile X syndrome (FRX; for example, CBP/p300).

Although the available inhibitors are not efficient enough to be used clinically, for example, by simply increasing histone acetyltransferase (HAT) activity, recent evidence has provided the starting point to investigate BCP’s role in nervous system development and neurodevelopmental disorders.

Introduction

Neurodevelopmental disorders are a group of diseases characterized by impairment of the growth and development of the brain or central nervous system. Most neurodevelopmental disorders affect behavior, resulting in economic and mental problems, not only for the patients, but also for their families and society. The most common forms of these disorders include mental retardation, autism, bipolar disorder, schizophrenia, attention-deficit/hyperactivity disorder, and learning and memory impairment [1, 2].

Understanding the regulatory mechanisms of these diseases is vital for developing better treatments and for preventing their onset. Neurodevelopmental disorders are thought to be caused by (a combination of) genetic and environmental factors. Epigenetic modification is affected by environmental factors, such as drugs, nutrition, toxicities, and mental stress, and its traits are in line with the pathogenic characteristics of neurodevelopmental disorders. Epigenetic modification of chromatin provides a dynamic platform for regulating the expression of target genes via acetylation, phosphorylation, methylation, ubiquitination, and sumoylation [3]. Among such intricate regulatory mechanisms, the equilibrium between global histone N-terminal deacetylation and acetylation changes in response to environmental stimulation through the functional interplay between HATs and histone deacetylases (HDACs). When the equilibrium is disturbed in the brain or central nervous system, patients develop neurodevelopmental disorders with complicated phenotypes.

In the present review, we focus on the roles of BCPs in the pathogenesis of neurodevelopmental disorders. BCPs represent a family of proteins found in many chromatin-associated proteins or in nearly all known nuclear HATs, and contain particular Brds. The importance of BCPs is underscored by the fact that genetic alterations of these genes and the subtle balance of aberrant acetylation are strongly linked to complicated diseases, such as cancer and neurodevelopmental disorders (Table 1). Here, we focus on the key advances in the current understanding of BCPs in the pathogenesis of neurodevelopmental disorders [4]. Special attention is paid to FRX, WS and RTS.

Table 1 BCPs implicated in neurodevelopmental disorders

Background of BCPs

The Brd, first identified in the Drosophila brahma (BRM) protein, is a highly evolutionarily conserved domain. More than 75 human BCPs have been identified in the National Center for Biotechnology Information (NCBI) protein database using Brd sequences as query sequences [20, 21]. With their particular domains and special functions, BCPs have been identified in a number of nuclear proteins, such as methyltransferases (for example, Ash1 and MLL) and HATs (for example, GCN5 and PCAF), as constituents of chromatin-remodeling complexes (for example, BAZ1A and BAZ1B) and in transcription factors (TFs) (for example, BRD1-4 and BRWD3). Here, we briefly introduce the structure and functions of BCPs, and their relevance to the pathogenesis of neurodevelopmental disorders.

The spatial structure of Brd results in recognition and binding of target regions. Other critical domains in BCPs, the amino acid sequences and posttranslational modifications of target sites combine to subtly and accurately regulate both the chromatin state and the transcription of target genes.

Originally, Brd was described as an approximately 60 amino acid domain forming two α-helices. After analyzing the Brds of several BCPs, Jeanmougin et al. identified a larger domain of about 110 residues [22]. Based on the resolved structure of Polybromo BRG1-associated factor (PCAF), a typical Brd forms an atypical left-handed, four-helix bundle topology, which includes A, B, Z, and C helixes paired in loops. A hydrophobic pocket formed by a combination of the Z-A loop with the B-C loop is responsible for acetyl-lysine recognition and specific binding [23, 24]. Brds may exist at the N- or C- terminus or in the center of proteins, with a repeat number varying from one to six (Figure 1). It is not clear whether the repeated Brds within a protein bind different targets or are a product of natural redundancy. One possible explanation is that the other Brds bind different specific substrates as integration platforms, or allocate the proteins to different nuclear compartments. Self-dimerization of some Brds plays a crucial role in their binding capacity. Using structural data, Nakamura et al. determined that Brd1 of BRD2 forms an intact homodimer, in the crystal and also in solution. Brd1 recognizes acetylated H4K12, whereas the dimer interface of Brd1 binds to hypoacetylated H4K8 [25]. Brd2 of BRD2 is monomeric in solution. The affinity between Brd2 of BRD2 and the acetylated H4K12 peptide is as low as 2.9 mM. This implies that BRD2 Brd1 and Brd2 have distinct roles in BRD2’s function [26]. Other BCPs can form dimers, including BRD4, TFIID, TAF1, and BRDT. Self-dimerization may help to avoid erasing histone codes during mitosis [27] or might improve modification accuracy [28].

Figure 1
figure 1

Bar diagram of representative BCPs and their major associated domains. ADA2, adaptor 2; BAH, bromo-adjacent homology (1 and 2); BAZ1, bromodomain adjacent to zinc finger domain; BCPs, bromodomain-containing proteins; Brd, bromodomain; C/H, Cys/His rich regions (1, 2 and 3); CTK, C-terminal kinase domain; DDT, DNA binding homeobox and different transcription factors; E1/E2, ubiquitin-activating/conjugating domain; HAT, histone acetyltransferase domain; HMG, high-mobility group domain; LH, leucine-rich helical domain; NTK, N-terminal kinase domain; WAC, WSTF/Acf1/cbp146; WD, tryptophan-aspartic acid dipeptide domain.

Brds can specifically bind target sites, and the other domains in BCPs contribute to recruit partner proteins or play other critical roles. For example, in p300/CBP, these other domains include three cysteine-histidine (CH) rich domains (CH1, CH2, and CH3), a KIX domain, and an ADA2-homology domain (Figure 1). In particular, both the PHD finger and Brd of p300 are required for interaction between p300 and nucleosomes. P300 binds nucleosomes in an acetylation-dependent manner when the PHD finger is combined with Brd. When both of these domains are isolated, the PHD finger binds to nucleosomes in an acetylation-independent manner, while the isolated Brd cannot bind nucleosomes at all [29, 30]. What is the relationship between Brd and PHD in p300? Increasing evidence shows that they form a cassette in BPTF, MLL, and TRIM24 proteins. The MLL PHD domain binds methylated H3K4 and Brd [31, 32]. PHD fingers not only recognize an acetyl-lysine residue of histone tails, as does Brd, but also recognize a wide variety of peptides, either modified by methylation, acetylation, or unmodified [32–34].

Complexes containing BCPs subtly and accurately regulate transcription through substrate recognition and by selecting and coordinating between Brds and other domains of BCPs. Brds specifically bind acetylated lysine residues, and the amino acid sequence surrounding the target site is also important for their binding ability [24, 35]. Significantly, a recent study showed that the flanking posttranslational modifications on targeting sites, such as phosphorylation and acetylation, are critical for Brds’ specific recognition and binding of substrates [33]. For Brds and other domains of BCPs, recent studies showed their combination improved the accuracy of the regulatory roles of BCPs. In p300, the Brd is important in maintaining the basal activity of HATs and in inducing transcription of the target genes [36]. Ivanov et al. uncovered an interaction between the PHD domain and Brd, where the PHD domain of the KAPI corepressor functions as an intramolecular E3 ligase for sumoylation of the adjacent Brd [37]. Based on the spatial reorganization and binding between Brds and their substrates, various inhibitors have been identified as specifically antagonizing the interaction between acetylated histones and Brds. These inhibitors include ischemin [38], novel benzodiazepines, benzotriazepine [39], and I-BET (a derivative of benzodiazepine) [40]. These inhibitory benzodiazepines have anti-inflammatory effects, and have potential benefits for treating specific carcinomas [41–43]. The results from inhibitor studies suggest the important roles of the fine regulation mechanism during normal and abnormal development. More detailed regulatory information, novel targets, and new inhibitors represent promising strategies towards delaying or halting the progression of neurological disorders.

BCPs have multiple functions that rely on particular domains that modify or recruit proteins to form regulatory complexes in response to developmental and environmental cues.

BCPs are implicated in the regulation of cellular events, including the cell cycle, growth, proliferation, differentiation, and apoptosis. Deletion models of different BCPs show a wide range of effects, such as lethality, slow growth, or no phenotypic change [44].

BCPs were initially considered as activators. The transcriptional coactivators CBP/p300, key members of the BCP family, participate in RNA polymerase II-mediated transcription. CBP/p300 possesses HAT activity and transfers an acetyl group from acetyl coenzyme A to a recipient lysine residue. Thus, CBP/p300 loosens dense chromatin, influences global chromatin acetylation and subsequently regulates transcription [45, 46]. The binding between p300 and its target gene promoters provides an accessible chromatin structure directly. Indirectly, another key property of CBP/p300 is as a protein bridge, providing a platform for other BCPs and transcriptional regulators [47]. A number of proteins bind to CBP/p300 through the CH1, CH3, and the KIX domains, which are important in mediating protein interactions and in the regulation of cellular events. The interaction between CBP/p300 and E1A is important in cell cycle regulation, involving the control of DNA synthesis and S phase progression, whereas the CBP/p300-p53 interaction is involved in cell apoptosis. The transactivation activity of p53 is improved after acetylation by CBP/p300, which is accompanied by a concomitant increase of the half-life of p53. After DNA damage, p53 might be acetylated by CBP through the Brd binding lysine 382-acetylated p53 peptide. Transcription of cyclin-dependent kinase inhibitor p21 is then activated to induce cell cycle arrest, senescence, or apoptosis. In this process, CBP acts as a HAT and regulates the transcription of p53 downstream genes through p53 binding target sites [48].

Accumulating evidence suggests that the same BCP would function as an activator or a repressor in different situations. For example, BRD7 is a component of chromatin-remodeling complexes, acting as either an activator or an inhibitor. The complex consisting of BRD7 and interferon regulatory factor 2 in the nucleus may activate chromatin transcription in situ. In the case of breast cancer with wild-type p53 and deletion of BRD7, the latter has been suggested as a cofactor in the transcriptional regulation of p53 target genes. In the complex constituted by BRD7 and p53, BRD7 affects p53 acetylation and the promoter activity of p53 target genes [49]. In the animal model, when GCN5 is deleted, mice die with increasing apoptosis. The mice survive longer if p53 is also deleted [16]. In addition, BRD7 promotes TCF4-mediated transcription through interaction with β-catenin and TCF4. A model has been proposed whereby BRD7 brings PTP-BL to the Dvl-1/axin/APC/GSK-3β/β-catenin complex, which results in enhanced Wnt signaling. This leads to GSK-3β dephosphorylation and nuclear translocation of β-catenin [50]. The BRD7-containing complex positively influences Wnt signaling; therefore, it is believed to be associated with gene activation. However, there are other types of complexes where BRD7 functions as a repressor. BRD7 can form hSWI-SNF complexes with PRMT5 and PRC2, which leads to inhibition of the expression of suppressor of tumorigenicity 7 (ST7) and retinoblastoma-like protein 2 (RBL2) [51]. In addition, reported substrate affinities range from micromolar to millimolar dissociation constant values [33]. The weak affinities suggest that BCPs possess a dual function (activation and repression). The hypothesis further suggests that BCPs form a recruitment platform or pool for holding different chromatin proteins and function to activate or repress transcription in a temporal-spatial pattern. The duality of BCPs depends on the recruitment of transcription factors and chromatin-remodelers in response to cellular or environmental signaling. Increasing data concerning the structure and function of Brds and BCPs will contribute to understanding the roles of BCPs in normal and abnormal nervous system development.

Association between BCPs and neurodevelopmental disorders

Abnormal brain development or injury during the fetal stage and during childhood leads to neurodevelopmental and psychiatric disorders. The genetic bases of certain neurodevelopmental disorders have been known for decades. Specific genes have been reported to be associated with neurodevelopmental disorders, but studies on their functional relevance are still ongoing. BCPs are involved in embryonic development and neural malformation [52]. Perhaps the most convincing evidence of the importance of BCPs and their complexes is as follows: even if the DNA sequences of all genes that directly control nervous system development and function are normal, abnormal modification by mutant or dysfunctional BCPs would result in neurodevelopmental disorders (Table 1). Thus, we pay particular attention to BCPs’ roles as TFs, HATs and chromatin-remodeling modulators in neurodevelopmental disorders.

Among 20 members of the BCP family, BRD2 and BRWD3 are related to neuronal development as TFs.

During the formation of the nervous system, a transcriptional program precisely determines the number and types of normal neurons. Precise spatiotemporal regulation controls neuron proliferation, differentiation and apoptosis. Although we know that a regulatory network comprising many TFs is required for neuronal development, we have only scratched the surface in our understanding of this process. Neurodevelopmental disorders show high genetic heterogeneity; therefore, it is difficult to screen and obtain all related genes that directly influence neural and mental development. Increasing evidence implies that BCPs are involved in neural development. More than 20 members of the BCP family have been identified as TFs. Here, we use the roles of BRD2 and BRWD3 in neurodevelopmental disorders as examples (Table 1).

BRD2 (also called RING3 and Fsrg1) is an important transcriptional modulator that is expressed in brain vesicles, the neural tube, the spinal cord and dorsal root ganglia. At the cellular level, BRD2 is localized to the nucleus in proliferating cells and to the cytoplasm in differentiating neuronal precursors [53]. BRD2−/− mice deviate from normal developmental programs at embryonic day 9.0 (E9.0), and die at E11.5, with a smaller embryonic size and neural tube defects (NTDs) [54, 55] (Table 1). The NTDs appear as exencephaly of the hindbrain. The causal mechanism between BRD2 and NTDs remains poorly understood. BRD2 is implicated in the regulation of cellular events, including growth, proliferation, differentiation, and apoptosis. Several associated proteins were identified as forming a BRD2 complex using Brd2 rabbit polyclonal immune-affinity chromatography. Among them, BRD2 combines with E2F to regulate cell cycle by binding to the cyclin A promoter [56]. In addition, BRD2 is the constituent of TFIID and SWI/SNF complexes. The available evidence suggests that BRD2 plays an essential role as an integrator of transcription and chromatin structure during mammalian embryogenesis and neurogenesis.

BRWD3 contains two Brds and nine WD repeats. The WD repeat domains are responsible for interaction with other proteins to regulate cellular events, such as cell cycle, signaling transduction, and apoptosis. Truncation mutants of BRWD3 were identified in the etiology of X-linked mental retardation during systematic screening of the X-chromosome coding sequences in 250 families (Online Mendelian Inheritance in Man (OMIM) ID: 300659) (Table 1). The affected males showed symptoms of prominent forehead, long face, large cupped ears and mild-to-moderate intellectual disability [11]. Recently, we demonstrated that the SNP loci rs7049509 and rs12689192 near the second Brd domain were linked with mental retardation [9] and two new mutations in Brds and WD40 domains, which might be the causes of mental retardation related to BRWD3 (unpublished data). Research into the causative mechanism is underway. BRWD3 is assumed to be involved in modulation of the JAK/STAT signaling pathway [57]. Notably, the mutant phenotype is related with a frame-shift, and the concomitant loss of a key Brd. Thus, BRWD3 might be associated with learning and memory.

HATs play important roles in regulating gene expression during brain development and memory formation. Aberrant BCPs, as HATs, contribute to neurodevelopmental disorders.

GCN5 is a transcriptional co-activator with HAT activity [58]. Mutant GCN5 is associated with NTDs. The structures of GCN5 and its histone substrates are conserved throughout eukaryotes. GCN5 is indispensable for amino acid metabolism, as a component of the ADA (Adapter) and SPT-ADA-GCN5 acetyltransferase (SAGA) transcription complexes [59]. These complexes regulate transcription processes rather than initiation [16]. The structure of the complex revealed that the single Brd of GCN5 would preferentially bind acetylated H4K16. In yeast, GCN5 acetylates other nearby lysine sites [60]. GCN5 participates in neural and mental development. After being treated with a fear-conditioning stimulus, murine GCN5/KAT2A and acetylation levels were upregulated within one hour. Deletion of GCN5 leads to early embryonic lethality with cranial NTDs. Conditional knockout of GCN5 showed that the normal expression level of GCN5 is critical for neural tube closure in mice, suggesting that mutations of Gcn5 may be associated with increasing risk of NTDs in humans [61] (Table 1). GCN5 acetylates p53 in vitro and in vivo, and double mutants of Gcn5 and p53 mouse embryos survive longer than Gcn5 null mice. Deletion of p53 cannot rescue the embryonic lethality caused by the mutant of Gcn5. When the HAT activity of GCN5 is abnormal, cranial neural tube closure in mice is defective [16]. These results reveal that GCN5 is required for survival, and the HAT activity of GCN5 is critical for proper neural tube closure. The exact mechanism remains unclear.

Although NTDs are the second commonest disorders among birth defects in humans, their molecular regulation mechanisms remain poorly understood. Available evidence suggests that acetylation is involved in the likely mechanism of the etiology of NTDs. The process of neural tube closure, including shaping, elevating, bending, and sealing, is finely regulated. In addition to the role of GCN5, neural tube closure is related to acetylation-associated proteins such as CBP, p300, and Hat1 [62]. Knockouts of p300 and CBP show similar phenotypes to GCN5, such as embryonic lethality and NTDs. Consistent with this phenotype, mutations in CBP (that is, chromosomal translocations, microdeletions, and point mutations) are associated with the congenital developmental disorder, RTS.

As mentioned previously, CBP/p300 is another important HAT in the BCP family. It is becoming clear that mutations in CBP/p300 cause 55% of RTS (OMIM ID: 180849), which is characterized by mild to severe mental retardation, craniofacial defects, short stature, skeletal abnormalities, broad big toes, and broad thumbs [14, 15, 63] (Table 1). This syndrome is relatively common, accounting for 1 in 300 patients with mental retardation. Haploinsufficiency of CBP probably contributes to RTS in humans. Mutations of both the PHD finger and Brds have been identified in RTS patients. In mice, heterozygous deletion or truncation of CBP produced a phenotype resembling RTS [15]. Mutations of CBP (Y1175C, E1278K and R1379P) are associated with RTS, again suggesting the importance of HAT activity [64]. In addition, abnormal CBP/p300 can be found in progressive neurodegenerative diseases, such as FRX, myotonic dystrophy, and X-linked spinal and bulbar muscular atrophy [65]. FRX (OMIM ID: 300624) patients usually suffer from learning disabilities, macroorchidism, seizures, anxiety, mood disturbance, and attention problems. In some cases, individuals display autistic symptoms, including poor eye contact, shyness, self-talk, hand flapping, and hand biting. In line with their similar physical symptoms to mental retardation, the affected males present with more typical physical features, such as prominent ears, macrocephaly, a long face, and a high arched palate [66].

CBP and p300 are also implicated in memory formation. P300 of mice is crucial for long-term memory and learning in a histone acetylation-dependent manner. Long-term memory is impaired in CBP knockdown mice, whereas short-term memory is not affected [67]. If the HAT activity of CBP is abolished, long-term memory is impaired, but short-term memory is not affected. The learning ability of mice could be improved after treatment with HDAC inhibitors of the CREB1/CBP complex. Interestingly, long-term memory can also be rescued with HDACs inhibitors [68, 69]. Such studies have illustrated the critical roles of HAT activity of BCPs for long-term memory and synaptic plasticity. Therefore, members of the BCP family with HAT activity regulate transcription by acting as a recruiting platform and through their HAT function. Thus, their mutations may influence transcriptional regulation in two ways: by disrupting the recruiting process and by preventing chromatin remodeling. Together with the properties discussed earlier, at the cellular level, the events leading to the processes of normal or abnormal neurodevelopment, such as cell proliferation, differentiation, survival and polarity may be influenced by acetylation/deacetylation homeostasis events mediated by these special BCPs. At a mechanistic level, intensive research on members of complexes and downstream genes will be helpful in providing clues to the underlying causes of neurodevelopmental disorders.

BCPs also function beyond their main role as transcriptional modulators and enzymes. Some BCPs are associated with neurodevelopmental disorders as chromatin-remodeling modulators.

As chromatin-remodeling modulators, some BCPs affect aspects of chromatin remodeling, in addition to transcriptional regulation, such as increasing the efficiency of chromatin assembly and modulation, DNA replication, DNA repair/recombination, and chromosomal alteration. The function of chromosome remodeling appears to be different from that of transcription regulation [70]. The SWI/SNF complex was first identified as a chromatin-remodeling modulator that remodels and increases access of transcription factors to nucleosomes [71, 72]. Abnormal compositions of the SWI/SNF complex are involved in α-thalassemia mental retardation, with patients displaying common characteristics, such as severe cognitive delay, α-thalassemia, facial dysmorphism, microcephaly, skeletal and genital abnormalities, and severe neonatal hypotonia [73, 74]. The null and dominant-negative gene mutations exhibit defects of the peripheral nervous system in adults, homeotic transformations, and decreased viability [75].

WSTF including the nucleosome assembly complex (WINAC) is another SWI/SNF-type complex that has ATP-dependent chromatin-remodeling activity. It interacts with the vitamin D receptor (VDR) through the Williams syndrome transcription factor (WSTF, also named bromodomain adjacent zinc finger, BAZ1B). WINAC is a key complex for repressing and activating transcription [76], and is required to enable DNA replication through highly condensed regions of chromatin [77]. The BAZ1B protein contains a Brd, a PHD-type zinc finger motif, a WAKZ motif, and a leucine-rich helical domain (Figure 1). BAZ1B is ubiquitously expressed in both adult and fetal tissues, such as limb buds, tail and brain from around E11.5 in the mouse embryo [7]. It is also expressed strongly in the cranial neural crest-derived mesenchyme, which drives facial morphogenesis. Phosphorylation of BAZ1B in a MAPK-dependent manner is important to maintain the WINAC complex assembly [76]. BAZ1B has been previously identified as contributing to WS [78] (Table 1). Total deletion was detected in 50 of 50 WS individuals using fluorescence in situ hybridization analysis. WS (OMIM ID: 194050) is a microdeletion or contiguous gene deletion syndrome characterized by hemizygous deletion of 1.5 to 1.8 Mb on chromosome 7q11.23 [78]. The frequency of WS is estimated to be 1 in 10,000. Subjects with WS show typical craniofacial dysmorphology (a small upturned nose with a flat nasal bridge, mandibular hypoplasia, malocclusion, bi-temporal narrowing and prominent forehead), supravalvular aortic stenosis, multiple peripheral pulmonary arterial stenosis, statural deficiency, infantile hypocalcemia and a distinct cognitive profile with mild mental retardation [79, 80]. The patients suffer from specific cognitive deficits, including poor visual-motor integration and attention deficit. In terms of molecular pathogenesis, mutations of BAZ1B and dysfunction of WINAC contribute to WS [5]. The phosphorylation of Ser-158 in the WSTF/Acf1/cbpq46 (WAC) domain is essential for maintaining the association between BAZ1B and core BAF complex components, thereby maintaining the ATPase activity of WINAC. As a sensor, BAZ1B can turn on its chromatin-remodeling activity in response to intracellular signaling [81]. DNA sequence mutations or abnormal modification would disrupt the regulatory complex. These studies might support the view that a chromatin remodeler, such as WINAC, plays a key role in the development of nervous systems and the pathophysiology of WS (Table 1).

Deregulation of methylation and BCPs may alter complex networks of gene expression and brain function, contributing to neurodevelopmental disorders.

Brain development is a complicated process, involving neuron proliferation, differentiation, migration, communication, and apoptosis. Genetic deficits and negative environmental exposures may lead to abnormal neurological development. The characteristics of neurodevelopmental disorders strongly coincide with the traits of dysfunctional epigenetic modification at the chromatin level, alone or in combination. Here, we use Rett syndrome (OMIM ID: 312750) as an example to elucidate the contribution of aberrant methylation and dysfunction of BCPs to neurodevelopmental disorders.

Rett syndrome, a progressive childhood neurodevelopmental disorder, is characterized by stereotypies and mental retardation, and autistic behavior in females. The affected individuals show neurodevelopmental defects, such as common hand movements, aberrant gait, and seizures [82]. Rett syndrome occurs in 1 in 10,000 to 15,000 births and is caused by mutations in the methyl CpG binding protein 2 (MeCP2) gene [83]. Most patients have heterozygous mutations in MeCP2. If MeCP2 is heterozygous in mice, the female also exhibits behavioral symptoms. If totally deleted, the mice show severe neurological symptoms [84]. Although MeCP2 was discovered about 20 years ago, we still have limited information about its molecular function. On the one hand, MeCP2 selectively binds CpG dinucleotides in the mammalian genome, mediating transcriptional repression through interaction with a chromatin-remodeling complex. The SWI/SNF complex can be recruited by MeCP2 to heterochromatic foci in living mouse cells in a DNA methylation-dependent manner. The interaction between ATRX and MeCP2 may control expression of MeCP2-binding genes. When the MeCP2-ATRX interaction is disrupted, pathological changes could be identified in a number of X-linked mental retardations, including Rett syndrome [85]. In contrast, if MeCP2 recruits CREB, it acts as a transcriptional activator. The exact mechanism of MeCP2 action remains unclear as it acts as a repressor or an activator, depending on its interacting protein partners. MeCP2 regulates the brain-derived neurotrophic factor gene (Bdnf) in resting neurons. Overexpression of Bdnf can rescue a subset of RTS-like phenotypes [86]. In summary, increasing data suggest that MeCP2 acts as a transcriptional modulator, repressing genes by binding to methylated CpG DNA or activating genes through chromatin reorganization. Further research is required to understand the molecular pathogenesis of neurodevelopmental disorders based upon the network created by genetic determination and epigenetic modification.

Conclusions

The growing number of identified BCPs and the links between some of these members and neurodevelopmental disorders underscore the importance of this class of proteins. BCPs, which are ubiquitous and evolutionarily conserved, play pivotal roles in chromatin modifications, remodeling, and transcriptional regulation. Remarkably, all HATs contain Brds, but not all BCPs are HATs. Some of them are now considered as modulators of chromatin remodeling (such as BAZ1A and CECR2), while some are transcriptional regulators (such as BRD2 and BRWD3) (Table 1). Significantly, some BCP complexes are implicated in basal transcriptional regulation through their Brds or the domains of their partners, influencing the expressions of a range of genes. However, many BCPs regulate transcription through Brds or the particular domains of other TFs that bind to the promoters of target genes (Figure 1). BCPs are critical for cellular events; therefore, complicated phenotypes inevitably result from abnormal genotypes of BCPs or from their dysfunctional complexes. Neurodevelopmental disorders associated with BCPs are not limited to those discussed in this review, for example, bipolar affective disorder and schizophrenia [87]. Further studies addressing the continued functional analyses of each BCP family member are required to better assess the physiological roles of these proteins.

In general, BCPs bind to target sites through their Brds, and they regulate transcription together with their binding partners. BCPs not only ‘read’ the histone code [88], but also dynamically alter chromatin status [89]. HATs (containing some BCPs) write markers on nucleosomes, and HDACs erase them from nucleosomes, and the Brds of some BCPs read the markers [90]. Alternatively, some BCPs regulate gene transcription through the binding of their partners to target sites. In the process of hormonal and nutrient regulation, acetyltransferase complexes containing GCN5 repress glucose metabolism via GCN5 acetylation of PGC-1α [91]. The evidence presented in this review does not represent special cases; there are many other members of the BCP family that may adopt this model as modifying enzymes. Many relevant mechanisms remain to be investigated.

Furthermore, histone acetylation can improve the memory and learning ability through direct control of epigenetic suppression of gene expression [92]. This expectation presents an interesting challenge for future research concerning the wide variety of BCPs. Although they are unlikely to restore patients’ intelligence, the HDAC inhibitors SAHA and trichostatin A exhibit potential rescue deficits in long-term emotional memory and recognition memory in animal models of RTS. Thus, it may be possible to enhance quality of life by improving social behavior and to ameliorate the cognitive and motor deficits in neurodegenerative disorders (that is, Huntington’s, Parkinson’s, and Alzheimer’s diseases). Although the current inhibitors are not efficient enough for clinical use because they simply increase HAT activity, emerging advances highlight the potential of applying BCPs and chromosome acetylation in treating neurodevelopmental disorders.

Authors’ information

JL focuses on developmental biology and pathogenesis of complicated diseases. Recently, his laboratory has identified SNP loci that are positively associated with mental retardation in the Qinba mountain region, northwest China. Both loci rs7049509 and rs12689192 are near the second Brd domain in the BRWD3 gene. After investigating the functional domains and phosphorylation sites of BRWD3, the group is currently studying the molecular mechanism of neurodevelopment based upon BRWD3. GZ is a full professor, the director of Key Laboratory of Resource Biology and Biotechnology in western China. XG is a full professor, and the vice-director of the Institute of Population and Health. Since its inception in 1995, the Institute of Population and Health has been devoted to the study of neurodevelopmental disorders, such as mental retardation and Fragile X syndrome. The authors’ research teams have carried out investigations on the pathogenesis of neurodevelopmental disorders in the Qinba mountain area, where the incidence is about 3%. In addition to environmental factors, genetic deficits are critical. Susceptible loci and several genes have been identified using gene scanning methods.

Abbreviations

Ash1:

Absent, small, or homeotic disc1

ATAD:

ATPase family AAA domain-containing protein

BAZ1A:

Bromodomain adjacent zinc finger, 1A

BAZ1B:

Bromodomain adjacent zinc finger, 1B

BCPs:

Bromodomain-containing proteins

Brd1:

The first bromodomain protein gene

Brd2:

The second Brd protein gene

BRM:

Brahma

BET:

Bromodomain and the extra terminal domain

BPTF:

BRD and PHD finger-containing transcription factor

Brd:

Bromodomain

CBP:

CREB-binding protein

DRG:

Dorsal root ganglia

FRX:

Fragile X syndrome

HAT domain:

Histone acetyltransferase domain

HDACs:

Histone deacetylases

LAP finger:

Leukemia-associated-protein finger

MeCP2:

Methyl CpG binding protein 2

MLL:

Mixed-lineage leukemia

NTDs:

Neural tube defects

OMIM:

Online Mendelian Inheritance in Man

PBAF:

Polybromo BRG1-associated factor

PCAF:

P300/CREB-binding protein-associated factor

PDE-4:

Phospodiesterase-4

P-TEFb:

Positive transcription elongation factor b

RTS:

Rubinstein-Taybi syndrome

SAGA:

Spt-Ada-Gcn5 acetyltransferase

SAHA:

Suberoylanilide hydroxamic acid

SNP:

Single nucleotide polymorphism

TAF1:

TATA-binding protein-associated factor-1

TAZ:

Transcription adaptor putative zinc finger

TFs:

Transcription factors

Tg:

Transgenic

TPA:

Tetradecanoyl phorbol acetate

VDR:

Vitamin D receptor

VPA:

Valproic acid

WD:

Tryptophan-aspartate

WS:

Williams syndrome

TFIID:

transcription initiation factor.

References

  1. Silva AJ, Ehninger D: Adult reversal of cognitive phenotypes in neurodevelopmental disorders. J Neurodev Disord. 2009, 1: 150-157. 10.1007/s11689-009-9018-7.

    Article  PubMed Central  PubMed  Google Scholar 

  2. van Loo KM, Martens GJ: Genetic and environmental factors in complex neurodevelopmental disorders. Curr Genomics. 2007, 8: 429-444. 10.2174/138920207783591717.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  3. Weigel AV, Simon B, Tamkun MM, Krapf D: Ergodic and nonergodic processes coexist in the plasma membrane as observed by single-molecule tracking. Proc Natl Acad Sci U S A. 2011, 108: 6438-6443. 10.1073/pnas.1016325108.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Kaufman L, Ayub M, Vincent JB: The genetic basis of non-syndromic intellectual disability: a review. J Neurodev Disord. 2010, 2: 182-209. 10.1007/s11689-010-9055-2.

    Article  PubMed Central  PubMed  Google Scholar 

  5. Bochar DA, Savard J, Wang W, Lafleur DW, Moore P, Cote J, Shiekhattar R: A family of chromatin remodeling factors related to Williams syndrome transcription factor. Proc Natl Acad Sci U S A. 2000, 97: 1038-1043. 10.1073/pnas.97.3.1038.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Jones MH, Hamana N, Nezu J, Shimane M: A novel family of bromodomain genes. Genomics. 2000, 63: 40-45. 10.1006/geno.1999.6071.

    Article  CAS  PubMed  Google Scholar 

  7. Kitagawa H, Fujiki R, Yoshimura K, Mezaki Y, Uematsu Y, Matsui D, Ogawa S, Unno K, Okubo M, Tokita A, Nakagawa T, Ito T, Ishimi Y, Nagasawa H, Matsumoto T, Yanagisawa J, Kato S: The chromatin-remodeling complex WINAC targets a nuclear receptor to promoters and is impaired in Williams syndrome. Cell. 2003, 113: 905-917. 10.1016/S0092-8674(03)00436-7.

    Article  CAS  PubMed  Google Scholar 

  8. Cavalleri GL, Walley NM, Soranzo N, Mulley J, Doherty CP, Kapoor A, Depondt C, Lynch JM, Scheffer IE, Heils A, Gehrmann A, Kinirons P, Gandhi S, Satishchandra P, Wood NW, Anand A, Sander T, Berkovic SF, Delanty N, Goldstein DB, Sisodiya SM: A multicenter study of BRD2 as a risk factor for juvenile myoclonic epilepsy. Epilepsia. 2007, 48: 706-712. 10.1111/j.1528-1167.2007.00977.x.

    Article  CAS  PubMed  Google Scholar 

  9. Li J, Li L, Xing L, Zhang M, Yang X, Zheng Z, Li J, Zhang F, Gao X: Association of BRWD3 gene with mental retardation in Qin-ba mountains region. Journal of Northwest University(Natural Science Edition). 2012, 42: 961-965.

    CAS  Google Scholar 

  10. Kuwano Y, Kamio Y, Kawai T, Katsuura S, Inada N, Takaki A, Rokutan K: Autism-associated gene expression in peripheral leucocytes commonly observed between subjects with autism and healthy women having autistic children. PLoS One. 2011, 6: e24723. 10.1371/journal.pone.0024723.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  11. Field M, Tarpey PS, Smith R, Edkins S, O’Meara S, Stevens C, Tofts C, Teague J, Butler A, Dicks E, Barthorpe S, Buck G, Cole J, Gray K, Halliday K, Hills K, Jenkinson A, Jones D, Menzies A, Mironenko T, Perry J, Raine K, Richardson D, Shepherd R, Small A, Varian J, West S, Widaa S, Mallya U, Wooster R: Mutations in the BRWD3 gene cause X-linked mental retardation associated with macrocephaly. Am J Hum Genet. 2007, 81: 367-374. 10.1086/520677.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  12. Tarpey PS, Smith R, Pleasance E, Whibley A, Edkins S, Hardy C, O’Meara S, Latimer C, Dicks E, Menzies A, Stephens P, Blow M, Greenman C, Xue Y, Tyler-Smith C, Thompson D, Gray K, Andrews J, Barthorpe S, Buck G, Cole J, Dunmore R, Jones D, Maddison M, Mironenko T, Turner R, Turrell K, Varian J, West S, Widaa S: A systematic, large-scale resequencing screen of X-chromosome coding exons in mental retardation. Nat Genet. 2009, 41: 535-543. 10.1038/ng.367.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  13. Banting GS, Barak O, Ames TM, Burnham AC, Kardel MD, Cooch NS, Davidson CE, Godbout R, McDermid HE, Shiekhattar R: CECR2, a protein involved in neurulation, forms a novel chromatin remodeling complex with SNF2L. Hum Mol Genet. 2005, 14: 513-524.

    Article  CAS  PubMed  Google Scholar 

  14. Petrij F, Giles RH, Dauwerse HG, Saris JJ, Hennekam RC, Masuno M, Tommerup N, van Ommen GJ, Goodman RH, Peters DJ: Rubinstein-Taybi syndrome caused by mutations in the transcriptional co-activator CBP. Nature. 1995, 376: 348-351. 10.1038/376348a0.

    Article  CAS  PubMed  Google Scholar 

  15. Roelfsema JH, White SJ, Ariyurek Y, Bartholdi D, Niedrist D, Papadia F, Bacino CA, den Dunnen JT, van Ommen GJ, Breuning MH, Hennekam RC, Peters DJ: Genetic heterogeneity in Rubinstein-Taybi syndrome: mutations in both the CBP and EP300 genes cause disease. Am J Hum Genet. 2005, 76: 572-580. 10.1086/429130.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  16. Bu P, Evrard YA, Lozano G, Dent SY: Loss of Gcn5 acetyltransferase activity leads to neural tube closure defects and exencephaly in mouse embryos. Mol Cell Biol. 2007, 27: 3405-3416. 10.1128/MCB.00066-07.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  17. Van Houdt JK, Nowakowska BA, Sousa SB, van Schaik BD, Seuntjens E, Avonce N, Sifrim A, Abdul-Rahman OA, van den Boogaard MJ, Bottani A: Heterozygous missense mutations in SMARCA2 cause Nicolaides-Baraitser syndrome. Nat Genet. 2012, 44: 445-449. 10.1038/ng.1105. S1

    Article  CAS  PubMed  Google Scholar 

  18. Tsurusaki Y, Okamoto N, Ohashi H, Kosho T, Imai Y, Hibi-Ko Y, Kaname T, Naritomi K, Kawame H, Wakui K, Fukushima Y, Homma T, Kato M, Hiraki Y, Yamagata T, Yano S, Mizuno S, Sakazume S, Ishii T, Nagai T, Shiina M, Ogata K, Ohta T, Niikawa N, Miyatake S, Okada I, Mizuguchi T, Doi H, Saitsu H, Miyake N: Mutations affecting components of the SWI/SNF complex cause Coffin-Siris syndrome. Nat Genet. 2012, 44: 376-378. 10.1038/ng.2219.

    Article  CAS  PubMed  Google Scholar 

  19. Makino S, Kaji R, Ando S, Tomizawa M, Yasuno K, Goto S, Matsumoto S, Tabuena MD, Maranon E, Dantes M, Lee LV, Ogasawara K, Tooyama I, Akatsu H, Nishimura M, Tamiya G: Reduced neuron-specific expression of the TAF1 gene is associated with X-linked dystonia-parkinsonism. Am J Hum Genet. 2007, 80: 393-406. 10.1086/512129.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  20. Peserico A, Simone C: Physical and functional HAT/HDAC interplay regulates protein acetylation balance. J Biomed Biotechnol. 2011, 2011: 1-10.

    Article  Google Scholar 

  21. Jenuwein T, Allis CD: Translating the histone code. Science. 2001, 293: 1074-1080. 10.1126/science.1063127.

    Article  CAS  PubMed  Google Scholar 

  22. Jeanmougin F, Wurtz JM, Le Douarin B, Chambon P, Losson R: The bromodomain revisited. Trends Biochem Sci. 1997, 22: 151-153. 10.1016/S0968-0004(97)01042-6.

    Article  CAS  PubMed  Google Scholar 

  23. Dhalluin C, Carlson JE, Zeng L, He C, Aggarwal AK, Zhou MM: Structure and ligand of a histone acetyltransferase bromodomain. Nature. 1999, 399: 491-496. 10.1038/20974.

    Article  CAS  PubMed  Google Scholar 

  24. Zeng L, Zhou MM: Bromodomain: an acetyl-lysine binding domain. FEBS Lett. 2002, 513: 124-128. 10.1016/S0014-5793(01)03309-9.

    Article  CAS  PubMed  Google Scholar 

  25. Nakamura Y, Umehara T, Nakano K, Jang MK, Shirouzu M, Morita S, Uda-Tochio H, Hamana H, Terada T, Adachi N, Matsumoto T, Tanaka A, Horikoshi M, Ozato K, Padmanabhan B, Yokoyama S: Crystal structure of the human BRD2 bromodomain: insights into dimerization and recognition of acetylated histone H4. J Biol Chem. 2007, 282: 4193-4201.

    Article  CAS  PubMed  Google Scholar 

  26. Huang H, Zhang J, Shen W, Wang X, Wu J, Shi Y: Solution structure of the second bromodomain of Brd2 and its specific interaction with acetylated histone tails. BMC Struct Biol. 2007, 7: 57. 10.1186/1472-6807-7-57.

    Article  PubMed Central  PubMed  Google Scholar 

  27. Umehara T, Nakamura Y, Jang MK, Nakano K, Tanaka A, Ozato K, Padmanabhan B, Yokoyama S: Structural basis for acetylated histone H4 recognition by the human BRD2 bromodomain. J Biol Chem. 2010, 285: 7610-7618. 10.1074/jbc.M109.062422.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  28. Umehara T, Nakamura Y, Wakamori M, Ozato K, Yokoyama S, Padmanabhan B: Structural implications for K5/K12-di-acetylated histone H4 recognition by the second bromodomain of BRD2. FEBS Lett. 2010, 584: 3901-3908. 10.1016/j.febslet.2010.08.013.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  29. Kalkhoven E: CBP and p300: HATs for different occasions. Biochem Pharmacol. 2004, 68: 1145-1155. 10.1016/j.bcp.2004.03.045.

    Article  CAS  PubMed  Google Scholar 

  30. Ragvin A, Valvatne H, Erdal S, Arskog V, Tufteland KR, Breen K, OYan AM, Eberharter A, Gibson TJ, Becker PB, Aasland R: Nucleosome binding by the bromodomain and PHD finger of the transcriptional cofactor p300. J Mol Biol. 2004, 337: 773-788. 10.1016/j.jmb.2004.01.051.

    Article  CAS  PubMed  Google Scholar 

  31. Bycroft M: Recognition of non-methyl histone marks. Curr Opin Struct Biol. 2011, 21: 761-766. 10.1016/j.sbi.2011.09.006.

    Article  CAS  PubMed  Google Scholar 

  32. Zeng L, Zhang Q, Li S, Plotnikov AN, Walsh MJ, Zhou MM: Mechanism and regulation of acetylated histone binding by the tandem PHD finger of DPF3b. Nature. 2010, 466: 258-262. 10.1038/nature09139.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  33. Filippakopoulos P, Picaud S, Mangos M, Keates T, Lambert JP, Barsyte-Lovejoy D, Felletar I, Volkmer R, Muller S, Pawson T, Gingras AC, Arrowsmith CH, Knapp S: Histone recognition and large-scale structural analysis of the human bromodomain family. Cell. 2012, 149: 214-231. 10.1016/j.cell.2012.02.013.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  34. Ruthenburg AJ, Li H, Milne TA, Dewell S, McGinty RK, Yuen M, Ueberheide B, Dou Y, Muir TW, Patel DJ, Allis CD: Recognition of a mononucleosomal histone modification pattern by BPTF via multivalent interactions. Cell. 2011, 145: 692-706. 10.1016/j.cell.2011.03.053.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  35. Mujtaba S, He Y, Zeng L, Farooq A, Carlson JE, Ott M, Verdin E, Zhou MM: Structural basis of lysine-acetylated HIV-1 Tat recognition by PCAF bromodomain. Mol Cell. 2002, 9: 575-586. 10.1016/S1097-2765(02)00483-5.

    Article  CAS  PubMed  Google Scholar 

  36. Chen J, Ghazawi FM, Li Q: Interplay of bromodomain and histone acetylation in the regulation of p300-dependent genes. Epigenetics. 2010, 5: 509-515. 10.4161/epi.5.6.12224.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  37. Ivanov AV, Peng H, Yurchenko V, Yap KL, Negorev DG, Schultz DC, Psulkowski E, Fredericks WJ, White DE, Maul GG, Sadofsky MJ, Zhou MM, Rauscher FJ: PHD domain-mediated E3 ligase activity directs intramolecular sumoylation of an adjacent bromodomain required for gene silencing. Mol Cell. 2007, 28: 823-837. 10.1016/j.molcel.2007.11.012.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  38. Borah JC, Mujtaba S, Karakikes I, Zeng L, Muller M, Patel J, Moshkina N, Morohashi K, Zhang W, Gerona-Navarro G, Hajjar RJ, Zhou MM: A small molecule binding to the coactivator CREB-binding protein blocks apoptosis in cardiomyocytes. Chem Biol. 2011, 18: 531-541. 10.1016/j.chembiol.2010.12.021.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  39. Filippakopoulos P, Picaud S, Fedorov O, Keller M, Wrobel M, Morgenstern O, Bracher F, Knapp S: Benzodiazepines and benzotriazepines as protein interaction inhibitors targeting bromodomains of the BET family. Bioorg Med Chem. 2012, 20: 1878-1886. 10.1016/j.bmc.2011.10.080.

    Article  CAS  PubMed  Google Scholar 

  40. Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, Chung CW, Chandwani R, Marazzi I, Wilson P, Coste H, White J, Kirilovsky J, Rice CM, Lora JM, Prinjha RK, Lee K, Tarakhovsky A: Suppression of inflammation by a synthetic histone mimic. Nature. 2010, 468: 1119-1123. 10.1038/nature09589.

    Article  CAS  PubMed  Google Scholar 

  41. Chung CW, Coste H, White JH, Mirguet O, Wilde J, Gosmini RL, Delves C, Magny SM, Woodward R, Hughes SA, Boursier EV, Flynn H, Bouillot AM, Bamborough P, Brusq JM, Gellibert FJ, Jones EJ, Riou AM, Homes P, Martin SL, Uings IJ, Toum J, Clement CA, Boullay AB, Grimley RL, Blandel FM, Prinjha RK, Lee K, Kirilovsky J, Nicodeme E: Discovery and characterization of small molecule inhibitors of the BET family bromodomains. J Med Chem. 2011, 54: 3827-3838. 10.1021/jm200108t.

    Article  CAS  PubMed  Google Scholar 

  42. Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J, Chesi M, Schinzel AC, McKeown MR, Heffernan TP, Vakoc CR, Bergsagel PL, Ghobrial IM, Richardson PG, Young RA, Hahn WC, Anderson KC, Kung AL, Bradner JE, Mitsiades CS: BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011, 146: 904-917. 10.1016/j.cell.2011.08.017.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  43. Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, Morse EM, Keates T, Hickman TT, Felletar I, Philpott M, Munro S, McKeown MR, Wang Y, Christie AL, West N, Cameron MJ, Schwartz B, Heightman TD, La Thangue N, French CA, Wiest O, Kung AL, Knapp S, Bradner JE: Selective inhibition of BET bromodomains. Nature. 2010, 468: 1067-1073. 10.1038/nature09504.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  44. Denis GV: Duality in bromodomain-containing protein complexes. Front Biosci. 2001, 6: D849-D852. 10.2741/Denis.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  45. Bannister AJ, Kouzarides T: The CBP co-activator is a histone acetyltransferase. Nature. 1996, 384: 641-643. 10.1038/384641a0.

    Article  CAS  PubMed  Google Scholar 

  46. Ogryzko VV, Schiltz RL, Russanova V, Howard BH, Nakatani Y: The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell. 1996, 87: 953-959. 10.1016/S0092-8674(00)82001-2.

    Article  CAS  PubMed  Google Scholar 

  47. Pugh BF: HATs off to PIC assembly. Mol Cell. 2006, 23: 776-777. 10.1016/j.molcel.2006.08.022.

    Article  CAS  PubMed  Google Scholar 

  48. Mujtaba S, He Y, Zeng L, Yan S, Plotnikova O, Sachchidanand , Sanchez R, Zeleznik-Le NJ, Ronai Z, Zhou MM: Structural mechanism of the bromodomain of the coactivator CBP in p53 transcriptional activation. Mol Cell. 2004, 13: 251-263. 10.1016/S1097-2765(03)00528-8.

    Article  CAS  PubMed  Google Scholar 

  49. Mantovani F, Drost J, Voorhoeve PM, Del Sal G, Agami R: Gene regulation and tumor suppression by the bromodomain-containing protein BRD7. Cell Cycle. 2010, 9: 2777-2781. 10.4161/cc.9.14.12309.

    Article  CAS  PubMed  Google Scholar 

  50. Kim S, Lee J, Park J, Chung J: BP75, bromodomain-containing M(r) 75,000 protein, binds dishevelled-1 and enhances Wnt signaling by inactivating glycogen synthase kinase-3 beta. Cancer Res. 2003, 63: 4792-4795.

    CAS  PubMed  Google Scholar 

  51. Tae S, Karkhanis V, Velasco K, Yaneva M, Erdjument-Bromage H, Tempst P, Sif S: Bromodomain protein 7 interacts with PRMT5 and PRC2, and is involved in transcriptional repression of their target genes. Nucleic Acids Res. 2011, 39: 5424-5438. 10.1093/nar/gkr170.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  52. Denis GV: Bromodomain coactivators in cancer, obesity, type 2 diabetes, and inflammation. Discov Med. 2010, 10: 489-499. 10.1038/nrc2875.

    PubMed Central  PubMed  Google Scholar 

  53. Crowley T, Brunori M, Rhee K, Wang X, Wolgemuth DJ: Change in nuclear-cytoplasmic localization of a double-bromodomain protein during proliferation and differentiation of mouse spinal cord and dorsal root ganglia. Brain Res Dev Brain Res. 2004, 149: 93-101. 10.1016/j.devbrainres.2003.12.011.

    Article  CAS  PubMed  Google Scholar 

  54. Shang E, Wang X, Wen D, Greenberg DA, Wolgemuth DJ: Double bromodomain-containing gene Brd2 is essential for embryonic development in mouse. Dev Dyn. 2009, 238: 908-917. 10.1002/dvdy.21911.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  55. Gyuris A, Donovan DJ, Seymour KA, Lovasco LA, Smilowitz NR, Halperin AL, Klysik JE, Freiman RN: The chromatin-targeting protein Brd2 is required for neural tube closure and embryogenesis. Biochim Biophys Acta. 2009, 1789: 413-421. 10.1016/j.bbagrm.2009.03.005.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  56. Denis GV, McComb ME, Faller DV, Sinha A, Romesser PB, Costello CE: Identification of transcription complexes that contain the double bromodomain protein Brd2 and chromatin remodeling machines. J Proteome Res. 2006, 5: 502-511. 10.1021/pr050430u.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  57. Muller P, Kuttenkeuler D, Gesellchen V, Zeidler MP, Boutros M: Identification of JAK/STAT signalling components by genome-wide RNA interference. Nature. 2005, 436: 871-875. 10.1038/nature03869.

    Article  PubMed  Google Scholar 

  58. Brownell JE, Zhou J, Ranalli T, Kobayashi R, Edmondson DG, Roth SY, Allis CD: Tetrahymena histone acetyltransferase A: a homolog to yeast Gcn5p linking histone acetylation to gene activation. Cell. 1996, 84: 843-851. 10.1016/S0092-8674(00)81063-6.

    Article  CAS  PubMed  Google Scholar 

  59. Wang L, Liu L, Berger SL: Critical residues for histone acetylation by Gcn5, functioning in Ada and SAGA complexes, are also required for transcriptional function in vivo. Genes Dev. 1998, 12: 640-653. 10.1101/gad.12.5.640.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  60. Owen DJ, Ornaghi P, Yang JC, Lowe N, Evans PR, Ballario P, Neuhaus D, Filetici P, Travers AA: The structural basis for the recognition of acetylated histone H4 by the bromodomain of histone acetyltransferase gcn5p. EMBO J. 2000, 19: 6141-6149. 10.1093/emboj/19.22.6141.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  61. Lin W, Zhang Z, Chen CH, Behringer RR, Dent SY: Proper Gcn5 histone acetyltransferase expression is required for normal anteroposterior patterning of the mouse skeleton. Dev Growth Differ. 2008, 50: 321-330. 10.1111/j.1440-169X.2008.01041.x.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  62. Dutnall RN, Tafrov ST, Sternglanz R, Ramakrishnan V: Structure of the histone acetyltransferase Hat1: a paradigm for the GCN5-related N-acetyltransferase superfamily. Cell. 1998, 94: 427-438. 10.1016/S0092-8674(00)81584-6.

    Article  CAS  PubMed  Google Scholar 

  63. Timmermann S, Lehrmann H, Polesskaya A, Harel-Bellan A: Histone acetylation and disease. Cell Mol Life Sci. 2001, 58: 728-736. 10.1007/PL00000896.

    Article  CAS  PubMed  Google Scholar 

  64. Murata T, Kurokawa R, Krones A, Tatsumi K, Ishii M, Taki T, Masuno M, Ohashi H, Yanagisawa M, Rosenfeld MG, Glass CK, Hayashi Y: Defect of histone acetyltransferase activity of the nuclear transcriptional coactivator CBP in Rubinstein-Taybi syndrome. Hum Mol Genet. 2001, 10: 1071-1076. 10.1093/hmg/10.10.1071.

    Article  CAS  PubMed  Google Scholar 

  65. Paulson HL, Fischbeck KH: Trinucleotide repeats in neurogenetic disorders. Annu Rev Neurosci. 1996, 19: 79-107. 10.1146/annurev.ne.19.030196.000455.

    Article  CAS  PubMed  Google Scholar 

  66. Berry-Kravis E, Knox A, Hervey C: Targeted treatments for fragile X syndrome. J Neurodev Disord. 2011, 3: 193-210. 10.1007/s11689-011-9074-7.

    Article  PubMed Central  PubMed  Google Scholar 

  67. Alarcon JM, Malleret G, Touzani K, Vronskaya S, Ishii S, Kandel ER, Barco A: Chromatin acetylation, memory, and LTP are impaired in CBP+/− mice: a model for the cognitive deficit in Rubinstein-Taybi syndrome and its amelioration. Neuron. 2004, 42: 947-959. 10.1016/j.neuron.2004.05.021.

    Article  CAS  PubMed  Google Scholar 

  68. Urdinguio RG, Sanchez-Mut JV, Esteller M: Epigenetic mechanisms in neurological diseases: genes, syndromes, and therapies. Lancet Neurol. 2009, 8: 1056-1072. 10.1016/S1474-4422(09)70262-5.

    Article  CAS  PubMed  Google Scholar 

  69. Kramer JM, van Bokhoven H: Genetic and epigenetic defects in mental retardation. Int J Biochem Cell Biol. 2009, 41: 96-107. 10.1016/j.biocel.2008.08.009.

    Article  CAS  PubMed  Google Scholar 

  70. Fyodorov DV, Kadonaga JT: The many faces of chromatin remodeling: SWItching beyond transcription. Cell. 2001, 106: 523-525. 10.1016/S0092-8674(01)00478-0.

    Article  CAS  PubMed  Google Scholar 

  71. Khavari PA, Peterson CL, Tamkun JW, Mendel DB, Crabtree GR: BRG1 contains a conserved domain of the SWI2/SNF2 family necessary for normal mitotic growth and transcription. Nature. 1993, 366: 170-174. 10.1038/366170a0.

    Article  CAS  PubMed  Google Scholar 

  72. Laurent BC, Treich I, Carlson M: Role of yeast SNF and SWI proteins in transcriptional activation. Cold Spring Harb Symp Quant Biol. 1993, 58: 257-263. 10.1101/SQB.1993.058.01.030.

    Article  CAS  PubMed  Google Scholar 

  73. Gibbons RJ, Picketts DJ, Villard L, Higgs DR: Mutations in a putative global transcriptional regulator cause X-linked mental retardation with alpha-thalassemia (ATR-X syndrome). Cell. 1995, 80: 837-845. 10.1016/0092-8674(95)90287-2.

    Article  CAS  PubMed  Google Scholar 

  74. Gibbons RJ, Higgs DR: Molecular-clinical spectrum of the ATR-X syndrome. Am J Med Genet. 2000, 97: 204-212. 10.1002/1096-8628(200023)97:3<204::AID-AJMG1038>3.0.CO;2-X.

    Article  CAS  PubMed  Google Scholar 

  75. Elfring LK, Daniel C, Papoulas O, Deuring R, Sarte M, Moseley S, Beek SJ, Waldrip WR, Daubresse G, DePace A, Kennison JA, Tamkun JW: Genetic analysis of brahma: the Drosophila homolog of the yeast chromatin remodeling factor SWI2/SNF2. Genetics. 1998, 148: 251-265.

    PubMed Central  CAS  PubMed  Google Scholar 

  76. Oya H, Yokoyama A, Yamaoka I, Fujiki R, Yonezawa M, Youn MY, Takada I, Kato S, Kitagawa H: Phosphorylation of Williams syndrome transcription factor by MAPK induces a switching between two distinct chromatin remodeling complexes. J Biol Chem. 2009, 284: 32472-32482. 10.1074/jbc.M109.009738.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  77. Collins N, Poot RA, Kukimoto I, Garcia-Jimenez C, Dellaire G, Varga-Weisz PD: An ACF1-ISWI chromatin-remodeling complex is required for DNA replication through heterochromatin. Nat Genet. 2002, 32: 627-632. 10.1038/ng1046.

    Article  CAS  PubMed  Google Scholar 

  78. Lu X, Meng X, Morris CA, Keating MT: A novel human gene, WSTF, is deleted in Williams syndrome. Genomics. 1998, 54: 241-249. 10.1006/geno.1998.5578.

    Article  CAS  PubMed  Google Scholar 

  79. Perez Jurado AL: Williams-Beuren syndrome: a model of recurrent genomic mutation. Horm Res. 2003, 59 (Suppl 1): 106-113.

    Article  CAS  PubMed  Google Scholar 

  80. Pober BR: Williams-Beuren syndrome. N Engl J Med. 2010, 362: 239-252. 10.1056/NEJMra0903074.

    Article  CAS  PubMed  Google Scholar 

  81. Kitagawa H, Fujiki R, Yoshimura K, Oya H, Kato S: Williams syndrome is an epigenome-regulator disease. Endocr J. 2011, 58: 77-85. 10.1507/endocrj.K10E-393.

    Article  CAS  PubMed  Google Scholar 

  82. Zoghbi HY: Postnatal neurodevelopmental disorders: meeting at the synapse?. Science. 2003, 302: 826-830. 10.1126/science.1089071.

    Article  CAS  PubMed  Google Scholar 

  83. Kavalali ET, Nelson ED, Monteggia LM: Role of MeCP2, DNA methylation, and HDACs in regulating synapse function. J Neurodev Disord. 2011, 3: 250-256. 10.1007/s11689-011-9078-3.

    Article  PubMed Central  PubMed  Google Scholar 

  84. Guy J, Hendrich B, Holmes M, Martin JE, Bird A: A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat Genet. 2001, 27: 322-326. 10.1038/85899.

    Article  CAS  PubMed  Google Scholar 

  85. Nan X, Hou J, Maclean A, Nasir J, Lafuente MJ, Shu X, Kriaucionis S, Bird A: Interaction between chromatin proteins MECP2 and ATRX is disrupted by mutations that cause inherited mental retardation. Proc Natl Acad Sci U S A. 2007, 104: 2709-2714. 10.1073/pnas.0608056104.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  86. Zachariah RM, Rastegar M: Linking epigenetics to human disease and Rett syndrome: the emerging novel and challenging concepts in MeCP2 research. Neural Plast. 2012, 2012: 415825-

    PubMed Central  PubMed  Google Scholar 

  87. Severinsen JE, Bjarkam CR, Kiaer-Larsen S, Olsen IM, Nielsen MM, Blechingberg J, Nielsen AL, Holm IE, Foldager L, Young BD, Muir WJ, Blackwood DH, Corydon TJ, Mors O, Børglum AD: Evidence implicating BRD1 with brain development and susceptibility to both schizophrenia and bipolar affective disorder. Mol Psychiatry. 2006, 11: 1126-1138. 10.1038/sj.mp.4001885.

    Article  CAS  PubMed  Google Scholar 

  88. Glatt S, Alfieri C, Muller CW: Recognizing and remodeling the nucleosome. Curr Opin Struct Biol. 2011, 21: 335-341. 10.1016/j.sbi.2011.02.003.

    Article  CAS  PubMed  Google Scholar 

  89. Matangkasombut O, Buratowski RM, Swilling NW, Buratowski S: Bromodomain factor 1 corresponds to a missing piece of yeast TFIID. Genes Dev. 2000, 14: 951-962.

    PubMed Central  CAS  PubMed  Google Scholar 

  90. Muller S, Filippakopoulos P, Knapp S: Bromodomains as therapeutic targets. Expert Rev Mol Med. 2011, 13: e29-

    Article  PubMed Central  PubMed  Google Scholar 

  91. Lerin C, Rodgers JT, Kalume DE, Kim SH, Pandey A, Puigserver P: GCN5 acetyltransferase complex controls glucose metabolism through transcriptional repression of PGC-1alpha. Cell Metab. 2006, 3: 429-438. 10.1016/j.cmet.2006.04.013.

    Article  CAS  PubMed  Google Scholar 

  92. Schaefer A, Tarakhovsky A, Greengard P: Epigenetic mechanisms of mental retardation. Prog Drug Res. 2011, 67: 125-146.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Preparation of this review was financially supported by grants from the National Natural Science Foundation of China (31071103 to XC Gao), and the Western Resource Biology and Modern Biological Technology Lab Open Foundation (ZS11005 and 11JS094 to JL Li).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Xiaocai Gao.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

JL, GZ and XG made substantial contributions to design of this study, and JL drafted and manuscript, and GZ and XG revised the manuscript. All authors contributed to writing the manuscript. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Li, J., Zhao, G. & Gao, X. Development of neurodevelopmental disorders: a regulatory mechanism involving bromodomain-containing proteins. J Neurodevelop Disord 5, 4 (2013). https://doi.org/10.1186/1866-1955-5-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1866-1955-5-4

Keywords