Entry - *600571 - RE1-SILENCING TRANSCRIPTION FACTOR; REST - OMIM
 
* 600571

RE1-SILENCING TRANSCRIPTION FACTOR; REST


Alternative titles; symbols

NEURON-RESTRICTIVE SILENCER FACTOR; NRSF


HGNC Approved Gene Symbol: REST

Cytogenetic location: 4q12     Genomic coordinates (GRCh38): 4:56,907,900-56,935,844 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
4q12 {Wilms tumor 6, susceptibility to} 616806 AD 3
Deafness, autosomal dominant 27 612431 AD 3
Fibromatosis, gingival, 5 617626 AD 3

TEXT

Description

REST is a transcriptional repressor that regulates gene expression throughout the body. It binds 21-bp repressor element-1 (RE1) sites, also called neuron-restrictive silencer elements (NRSEs), through its 8 C2H2 zinc fingers. REST mediates gene repression by acting as a hub for the recruitment of multiple chromatin-modifying enzymes (Ooi and Wood, 2007).


Cloning and Expression

Schoenherr and Anderson (1995) cloned a transcription factor, which they termed NRSF, that bound the NRSE present in the 5-prime regulatory region of SCG10 (600621), a neuron-specific gene. The NRSF cDNA was cloned from a HeLa cell library. The longest cDNA was predicted to encode 8 zinc fingers of the C2H2 class, with interfinger sequences that placed NRSE within the GLI (see 165220)-Kruppel family of zinc finger proteins. Northern blot analysis detected a NRSF transcript of 7 to 8 kb. Expression of NRSF mRNA was detected in most nonneuronal progenitor cells, but it was absent in differentiated neurons.

Independently, Chong et al. (1995) cloned REST from a HeLa cell cDNA library. The deduced 1,097-amino acid protein has a calculated molecular mass of 121 kD. REST has 8 N-terminal C2H2-type zinc fingers, followed by a basic region, 6 repeats of a proline-rich sequence, and a C-terminal zinc finger. It also has a nuclear localization signal. In situ hybridization of mouse embryos detected abundant Rest expression in nonneuronal tissues.

Thiel et al. (1998) determined that REST contains 2 repressor domains, one located at the N terminus and the other at the C terminus, and an N-terminal zinc finger cluster which functions as the DNA-binding domain for neuronal genes.

Palm et al. (1999) identified several REST variants that arise from alternative splicing of an exon that they designated exon N. The splice variants produce insertions that generate in-frame stop codons and encode truncated proteins with an N-terminal repressor domain and weakened DNA-binding activity. The expression levels of these variants differ in human neuroblastoma and glial cells.


Gene Function

Schoenherr and Anderson (1995) showed that NRSF bound the NRSE DNA sequence in the 5-prime region of SCG10. They noted that the SCG10 regulatory region contains both activation and repression (i.e., silencer) domains and that similar NRSE-like sequence elements have been identified in other neuron-specific genes. Schoenherr and Anderson (1995) proposed that NRSF may function as a master negative regulator of neurogenesis.

By transfecting rat myocyte and pheochromocytoma cell lines, Chong et al. (1995) showed that human REST downregulated expression of the type II voltage-dependent sodium channel (see 182390). They proposed that REST suppresses expression of the channel in nonneuronal tissues.

Thiel et al. (1998) noted that the REST binding sites of several neuron-specific genes, such as those encoding synapsin I (313440), SCG10 (600621), A1-glycine receptor (138491), the B2 subunit of the nicotinic acetylcholine receptor (118507), and the M4-subunit of the muscarinic acetylcholine receptor, are found at various positions within the sequence. By transfecting these sequences in reporter constructs together with REST, Thiel et al. (1998) found that REST blocks transcription of a gene irrespective of whether the NRSE is located upstream or downstream of the open reading frame in either orientation and in both a distance- and a gene-independent manner.

Abderrahmani et al. (2001) identified an NRSE sequence in the promoter region of MAPK8PI1 (604641), which is expressed exclusively in neuronal tissue and pancreatic B cells. They confirmed that REST binds to the NRSE element of MAPK8PI1 and found that transfection and expression of REST in a B-cell line represses MAPK8PI1 transcription. Conversely, the introduction of a mutated NRSE into the MAPK8PI1 promoter allowed MAPK8PI1 expression in non-B-cell and nonneuronal cell lines. REST-mediated repression was found to be dependent on histone deacetylase (see 601241) activity.

Lunyak et al. (2002) reported that the zinc finger gene-specific repressor element REST can mediate extraneuronal restriction by imposing either active repression via histone deacetylase recruitment or long-term gene silencing using a distinct functional complex. Silencing of neuronal-specific genes requires the recruitment of an associated corepressor, COREST (607675), that serves as a functional molecular beacon for recruitment of molecular machinery that imposes silencing across a chromosomal interval, including transcriptional units that do not themselves contain REST/NRSF response elements.

Using indexing-based differential display PCR on neuronal precursor cells to study gene expression in Down syndrome (190685), Bahn et al. (2002) found that genes regulated by the REST transcription factor were selectively repressed. One of these genes, SCG10, was almost undetectable. The REST factor itself was also downregulated by 49% compared to controls. In cell culture, the Down syndrome cells showed a reduction of neurogenesis, as well as decreased neurite length and abnormal changes in neuron morphology. The authors noted that REST-regulated genes play an important part in brain development, plasticity, and synapse formation, and they suggested a link between dysregulation of REST and some of the neurologic deficits seen in Down syndrome.

The huntingtin gene (HTT; 613004) is mutated in Huntington disease (HD; 143100). Zuccato et al. (2001) reported that wildtype but not mutant huntingtin stimulates transcription of the gene encoding brain-derived neurotrophic factor (BDNF; 113505). Zuccato et al. (2003) showed that the NRSE is the target of wildtype huntingtin activity on BDNF promoter II. Wildtype huntingtin inhibits the silencing activity of NRSE, increasing transcription of BDNF. Zuccato et al. (2003) showed that this effect occurs through cytoplasmic sequestering of REST/NRSF, the transcription factor that binds to NRSE. In contrast, aberrant accumulation of REST/NRSF in the nucleus was present in Huntington disease. They showed that wildtype huntingtin coimmunoprecipitates with REST/NRSF and that less immunoprecipitated material is found in brain tissue with Huntington disease. They also reported that wildtype huntingtin acts as a positive transcriptional regulator for other NRSE-containing genes involved in the maintenance of the neuronal phenotype. Consistently, loss of expression of NRSE-controlled neuronal genes was shown in cells, mice, and human brain with Huntington disease. Zuccato et al. (2003) concluded that wildtype huntingtin acts in the cytoplasm of neurons to regulate the availability of REST/NRSF to its nuclear NRSE-binding site and that this control is lost in the pathology of Huntington disease. These data identified a novel mechanism by which mutation of huntingtin causes loss of transcription of neuronal genes.

Kemp et al. (2003) identified NRSE-like motifs in several genes involved in pancreas development, including a highly conserved NRSE-like motif in the upstream promoter of PAX4 (167413), a gene implicated in differentiation of the insulin-producing beta-cell lineage. Using mammalian cell lines, they found that the NRSE in the upstream promoter of Pax4 formed a DNA-protein complex with Nrsf and conferred Nrsf-dependent transcriptional repression on a reporter gene promoter and the native Pax4 gene promoter.

Reactivation of the fetal cardiac gene program is a characteristic feature of hypertrophied and failing hearts. Kuwahara et al. (2003) showed that Nrsf selectively regulated expression of multiple fetal cardiac genes and played a role in reexpression of these genes in rat neonatal ventricular myocytes. Transgenic mice expressing a dominant-negative Nrsf mutant in their hearts exhibited dilated cardiomyopathy, high susceptibility to arrhythmias, and sudden death. Genes encoding 2 ion channels that carry the fetal cardiac currents I(f) and I(Ca,T), which were induced in Nrsf-transgenic mice and were potentially responsible for both the cardiac dysfunction and arrhythmogenesis, were regulated by Nrsf.

Neuronal gene transcription is repressed in nonneuronal cells by the REST/NRSF complex. To understand how this silencing is achieved, Yeo et al. (2005) examined CTDSP1 (605323), CTDSP2 (608711), and CTDSPL (608592), the small CTD phosphatases (SCP), whose expression is restricted to nonneuronal tissues. Yeo et al. (2005) showed that REST/NRSF recruits SCPs to neuronal genes that contain RE1 elements, leading to neuronal gene silencing in nonneuronal cells. Phosphatase-inactive forms of SCP interfere with REST/NRSF function and promote neuronal differentiation of P19 stem cells. Likewise, small interfering RNA directed to the single Drosophila SCP unmasks neuronal gene expression in S2 cells. Thus, Yeo et al. (2005) concluded that SCP activity is an evolutionarily conserved transcriptional regulator that acts globally to silence neuronal genes.

Cheong et al. (2005) identified a functional REST-binding sequence in the promoter region of the KCNN4 gene (602754). REST was expressed in the nuclei of human vascular smooth muscle cells (SMCs), and it downregulated KCNN4 expression in mouse and human vascular SMCs. Downregulated REST and upregulated KCNN4 were evident in SMCs of human neointimal hyperplasia grown in organ culture, and exogenous REST reduced the functional impact of KCNN4. Cheong et al. (2005) concluded that REST acts as a switch to regulate potassium channel expression and consequently the phenotype of vascular smooth muscle cells and human vascular disease.

Plaisance et al. (2005) showed that the transcriptional factor Sp1 (189906) was required for expression of most Rest target genes in mouse insulin-secreting cells and rat neuronal-like cells where Rest is absent. Inhibition of REST in HeLa cells and in mouse beta cells restored the transcriptional activity of Sp1. Coimmunoprecipitation and transfection assays indicated that the C-terminal repressor domain of REST was required for interaction with Sp1 and inhibited its activity. Silencing of Sp1 by REST required histone deacetylase activity.

Tahiliani et al. (2007) showed that JARID1C/SMCX (314690), a JmjC domain-containing protein implicated in X-linked mental retardation and epilepsy, possesses H3K4 tridemethylase activity and functions as a transcriptional repressor. An SMCX complex isolated from HeLa cells contained additional chromatin modifiers (the histone deacetylases HDAC1 (601241) and HDAC2 (605164), and the histone H3K9 methyltransferase G9a (604599)) and the transcriptional repressor REST, suggesting a direct role for SMCX in chromatin dynamics and REST-mediated repression. Chromatin immunoprecipitation revealed that SMCX and REST co-occupy the neuron-restrictive silencing elements in the promoters of a subset of REST target genes. RNA interference-mediated depletion of SMCX derepressed several of these targets and simultaneously increased H3K4 trimethylation at the sodium channel type 2A (SCN2A; 182390) and synapsin I (SYN1; 313440) promoters. Tahiliani et al. (2007) proposed that loss of SMCX activity impairs REST-mediated neuronal gene regulation, thereby contributing to SMCX-associated X-linked mental retardation.

Ding et al. (2008) found that purified HeLa cell mediator complexes that included MED12 (300188) interacted directly with the G9A and REST. Endogenous REST in HEK293 cells suppressed expression of a reporter gene bearing RE1 sites, and knockdown of either MED12 or G9A abrogated the suppression. Depletion of MED12 significantly reduced the association of G9A with RE1 elements and decreased the level of H3K9 dimethylation by G9A without influencing RE1 site occupancy by REST.

Using an unbiased screen, Guardavaccaro et al. (2008) demonstrated that REST is an interactor with the F-box protein beta-TRCP (603482). REST is degraded by means of the ubiquitin beta-TRCP during the G2 phase of the cell cycle to allow transcriptional derepression of Mad2 (601467), an essential component of the spindle assembly checkpoint. The expression in cultured cells of a stable REST mutant, which is unable to bind beta-TRCP, inhibited Mad2 expression and resulted in a phenotype analogous to that observed in Mad2 heterozygous cells. In particular, Guardavaccaro et al. (2008) observed defects that were consistent with faulty activation of the spindle checkpoint, such as shortened mitosis, premature sister-chromatid separation, chromosome bridges and missegregation in anaphase, tetraploidy, and a faster mitotic slippage in the presence of a spindle inhibitor. An indistinguishable phenotype was observed by expressing the oncogenic REST-FS mutant, which does not bind beta-TRCP. Thus, beta-TRCP-dependent degradation of REST during G2 permits the optimal activation of the spindle checkpoint, and consequently it is required for the fidelity of mitosis.

Westbrook et al. (2008) showed that REST is regulated by ubiquitin-mediated proteolysis, and used an RNA interference screen to identify a Skp1-Cul1-F-box protein complex containing the F-box protein beta-TRCP as an E3 ubiquitin ligase responsible for REST degradation. Beta-TRCP binds and ubiquitinates REST and controls its stability through a conserved phospho-degron. During neural differentiation, REST is degraded in a beta-TRCP-dependent manner. Beta-TRCP is required for proper neural differentiation only in the presence of REST, indicating that beta-TRCP facilitates this process through degradation of REST. Conversely, failure to degrade REST attenuates differentiation. Furthermore, Westbrook et al. (2008) found that beta-TRCP overexpression, which is common in human epithelial cancers, causes oncogenic transformation of human mammary epithelial cells and that this pathogenic function requires REST degradation. Thus, Westbrook et al. (2008) concluded that REST is a key target in beta-TRCP-driven transformation and that the beta-TRCP-REST axis is a new regulatory pathway controlling neurogenesis.

Singh et al. (2008) demonstrated that REST maintains self-renewal and pluripotency in mouse ES cells through suppression of the microRNA miR21 (611020). The authors found that, as with known self-renewal markers, the level of REST expression is much higher in self-renewing mouse embryonic stem (ES) cells than in differentiating mouse ES (embryoid body, EB) cells. Heterozygous deletion of Rest and its short interfering RNA (siRNA)-mediated knockdown in mouse ES cells caused a loss of self-renewal--even when these cells were grown under self-renewal conditions--and led to the expression of markers specific for multiple lineages. Conversely, exogenously added REST maintained self-renewal in mouse EB cells. Furthermore, Rest heterozygous mouse ES cells cultured under self-renewal conditions expressed substantially reduced levels of several self-renewal regulators, including Oct4 (164177), Nanog (607937), Sox2 (184429), and c-Myc (190080), and exogenously added Rest in mouse EB cells maintained the self-renewal phenotypes and expression of these self-renewal regulators. Singh et al. (2008) also demonstrated that in mouse ES cells, Rest is bound to the gene chromatin of a set of miRNAs that potentially target self-renewal genes. Whereas mouse ES cells and mouse EB cells containing exogenously added Rest expressed lower levels of these miRNAs, EB cells, Rest heterozygous ES cells, and ES cells treated with siRNA targeting Rest expressed higher levels of these miRNAs. At least one of these REST-regulated miRNAs, miR21, specifically suppressed the self-renewal of mouse ES cells, corresponding to the decreased expression of Oct4, Nanog, Sox2, and c-Myc. Thus, Singh et al. (2008) concluded that REST is an element of the interconnected regulatory network that maintains the self-renewal and pluripotency of mouse ES cells.

Using a transchromosomic mouse model of Down syndrome, Canzonetta et al. (2008) showed that a 30 to 60% reduced expression of Nrsf/Rest, a key regulator of pluripotency and neuronal differentiation, is an alteration that persists in trisomy 21 (see 190685) from undifferentiated embryonic stem cells to adult brain and is reproducible across several Down syndrome models. Using partially trisomic ES cells, Canzonetta et al. (2008) mapped this effect to a 3-gene segment of human chromosome 21 containing DYRK1A (600855). The authors independently identified the same locus as the most significant expression quantitative trait locus (eQTL) controlling REST expression in the human genome. Canzonetta et al. (2008) found that specifically silencing the third copy of DYRK1A rescued Rest levels, and demonstrated altered Rest expression in response to inhibition of DYRK1A expression or kinase activity, and in a transgenic Dyrk1a mouse. The authors observed that undifferentiated trisomy 21 ES cells showed DYRK1A-dose-sensitive reductions in levels of some pluripotency regulators, including Nanog (607937) and Sox2 (184429), causing premature expression of transcription factors driving early endodermal and mesodermal differentiation, partially overlapping downstream effects of Rest heterozygosity. The ES cells produced embryoid bodies with elevated levels of the primitive endoderm progenitor marker Gata4 (600576) and a strongly reduced neuroectodermal progenitor compartment. Canzonetta et al. (2008) concluded that DYRK1A-mediated deregulation of REST is a very early pathologic consequence of trisomy 21 with potential to disturb the development of all embryonic lineages, warranting closer research into its contribution to Down syndrome pathology and new rationales for therapeutic approaches.

Using yeast 2-hybrid and immunoprecipitation analyses, Shimojo (2008) showed that human RILP (PRICKLE1; 608500) and huntingtin interacted directly with dynactin-1 (DCTN1; 601143) to form a triplex. REST bound to the triplex through direct interaction with RILP, forming a quaternary complex involved in nuclear translocation of REST in nonneuronal cells. In neuronal cells, the complex also contained HAP1 (600947), which affected interaction of disease-causing mutant huntingtin, but not wildtype huntingtin, with dynactin-1 and RILP. Overexpression and knockout analyses demonstrated that the presence of HAP1 in the complex prevented nuclear translocation of REST and thereby regulated REST activity.

At the point of mitotic exit within the vertebrate nervous system, when cells lose multipotency and begin to develop stable connections that will persist over life, a switch in ATP-dependent chromatin-remodeling mechanisms occurs. This switch involves the exchange of the BAF53A and BAF45A (PHF10; 613069) subunits within Swi/Snf-like neural progenitor-specific BAF (npBAF) complexes for the homologous BAF53B (ACTL6B; 612458) and BAF45B (DPF1; 601670) subunits within neuron-specific BAF (nBAF) complexes in postmitotic neurons. The subunits of the npBAF complex are essential for neural progenitor proliferation, and mice with reduced dosage for the genes encoding its subunits have defects in neural tube closure similar to those in human spina bifida. In contrast, BAF53B and the nBAF complex are essential for an evolutionarily conserved program of postmitotic neural development and dendritic morphogenesis. Yoo et al. (2009) showed that this essential transition is mediated by repression of BAF53A by miR9* (an miRNA processed from the opposite arm of the miR9 (611186) stem-loop precursor) and miR124 (609327). They found that BAF53a repression is mediated by sequences in the 3-prime untranslated region corresponding to the recognition sites for miR9* and miR124, which are selectively expressed in postmitotic neurons. Mutation of these sites led to persistent expression of BAF53A and defective activity-dependent dendritic outgrowth in neurons. In addition, overexpression of miR9* and miR124 in neural progenitors caused reduced proliferation. miR9* and miR124 are repressed by REST. Yoo et al. (2009) showed that expression of REST in postmitotic neurons led to derepression of BAF53A, indicating that REST-mediated repression of microRNAs directs the essential switch of chromatin regulatory complexes.

Loe-Mie et al. (2010) showed that an SWI/SNF-centered network including the Smarca2 gene (600014) was modified by the downregulation of REST/NRSF in a mouse neuronal cell line. REST/NRSF downregulation also modified the levels of Smarce1 (603111), Smarcd3 (601737), and SWI/SNF interactors (Hdac1, 601241; RcoR, 607675; and Mecp2, 300005). Smarca2 downregulation generated an abnormal dendritic spine morphology that was an intermediate phenotype of schizophrenia (see 181500). The authors noted that 8 genomewide-supported schizophrenia-associated genes (SMARCA2; CSF2RA, 306250; HIST1H2BJ, 615044; NOTCH4, 164951; NRGN, 602350; SHOX, 312865; TCF4, 602272; and ZNF804A, 612282) are part of an interacting network; 5 of the 8, including SMARCA2, encode transcription regulators, and 3 (TCF4, SMARCA2, and CSF2RA) were modified at the level of expression when the REST/NRSF-SWI/SNF chromatin remodeling complex was experimentally manipulated in mouse cell lines and in transgenic mouse models. REST/NRSF-SWI/SNF deregulation also resulted in the differential expression of genes that are clustered in chromosomes, suggesting the induction of genomewide epigenetic changes. Loe-Mie et al. (2010) concluded that the SWI/SNF chromatin remodeling complex is a key component of the genetic architecture of schizophrenia.

Yang et al. (2012) demonstrated that ZNF335 (610827) acts upstream of REST and regulates its expression.

Das et al. (2013) found that knockdown of REST resulted in a decline in medulloblastoma cell proliferation and accumulation of p27 (CDKN1B; 600778). In vitro analysis showed that REST and p27 expression were reciprocally correlated in human medulloblastoma samples. REST repressed expression of USP37 (620226), and USP37 expression promoted p27 deubiquitination. USP37 interacted with p27 to promote its deubiquitination and stabilization, thereby blocking cell proliferation. The authors concluded that REST regulates p27 stability and cell proliferation by controlling USP37.

Lu et al. (2014) demonstrated that induction of REST is a universal feature of normal aging in human cortical and hippocampal neurons. REST is lost, however, in mild cognitive impairment and Alzheimer disease (AD; 104300). Chromatin immunoprecipitation with deep sequencing and expression analysis showed that REST represses genes that promote cell death and AD pathology, and induces the expression of stress response genes. Moreover, REST potently protects neurons from oxidative stress and beta-amyloid (see 104760) protein toxicity, and conditional deletion of Rest in the mouse brain leads to age-related neurodegeneration. A functional ortholog of REST, C. elegans Spr4, also protects against oxidative stress and beta-amyloid protein toxicity. During normal aging, REST is induced in part by cell-nonautonomous Wnt signaling. However, in Alzheimer disease, frontotemporal dementia (FTD; 600274), and dementia with Lewy bodies (DLB; 127750), REST is lost from the nucleus and appears in autophagosomes together with pathologic misfolded proteins. Finally, REST levels during aging are closely correlated with cognitive preservation and longevity. Lu et al. (2014) therefore concluded that the activation state of REST may distinguish neuroprotection from neurodegeneration in the aging brain.

Zullo et al. (2019) showed that extended longevity in humans is associated with a distinct transcriptome signature in the cerebral cortex that is characterized by downregulation of genes related to neural excitation and synaptic function. In C. elegans, neural excitation increases with age, and inhibition of excitation globally, or in glutamatergic or cholinergic neurons, increases longevity. Furthermore, longevity is dynamically regulated by the excitatory-inhibitory balance of neural circuits. The transcription factor REST is upregulated in humans with extended longevity and represses excitation-related genes. Notably, Rest-deficient mice exhibit increased cortical activity and neuronal excitability during aging. Similarly, loss-of-function mutations in the C. elegans REST ortholog genes spr3 and spr4 elevate neural excitation and reduce the lifespan of long-lived daf2 mutants. In wildtype worms, overexpression of spr4 suppresses excitation and extends lifespan. REST, spr3, spr4, and reduced excitation activated the longevity-associated transcription factors FOXO1 (136533) and daf16 in mammals and worms, respectively. Zullo et al. (2019) concluded that their findings revealed a conserved mechanism of aging that is mediated by neural circuit activity and regulated by REST.


Gene Structure

Bayram et al. (2017) stated that the REST gene has 4 exons.


Mapping

Bayram et al. (2017) stated that the REST gene maps to chromosome 4q12.


Molecular Genetics

Wilms Tumor 6

Mahamdallie et al. (2015) identified 11 different REST mutations (see, e.g., 600571.0001-600571.0003) in 16 individuals from 4 families and 9 nonfamilial Wilms tumor (WT6; 616806) pedigrees. Ten of the 11 different mutations, including all of the nonsynonymous mutations, clustered in the DNA binding domain of REST. In 4 cases for whom parental DNA was available, 1 mutation had occurred de novo and 3 had been inherited, confirming incomplete penetrance. None was present in ICR1000 exome series of 993 or in the 61,312 individuals in the ExAC browser. All tested variants showed abrogation of REST function. Mahamdallie et al. (2015) concluded that their data established REST as a Wilms tumor predisposition gene accounting for approximately 2% of Wilms tumors, and recommended screening of REST in all familial cases.

Gingival Fibromatosis 5

In a 11 patients from 3 unrelated Turkish families with gingival fibromatosis-5 (GINGF5; 617626), Bayram et al. (2017) identified 3 different heterozygous truncating mutations in the REST gene (600571.0004-600571.0006). The mutations, which were found by exome sequencing and confirmed by Sanger sequencing, segregated with the disorder in 2 families; the mutation in the proband of the third family occurred de novo. A mildly affected father in 1 of the families was mosaic for the mutation. All mutations occurred in the final exon of the gene, which may result in escape from nonsense-mediated mRNA decay, suggesting that the alleles may act through a dominant-negative or gain-of-function effect. Bayram et al. (2017) noted that studies have suggested that the disorder results from excessive accumulation of extracellular matrix components, particularly collagen type I, which may be due to abnormal expression of TGF-beta (TGFB1; 190180) and IL6 (147620). The mutant transcripts may reduce the repressor function of REST on the collagen synthesis pathway, resulting in the accumulation of collagen in gingiva. However, functional studies of the REST variants and studies of patient cells were not performed.

Autosomal Dominant Deafness 27

In a 3-generation family with autosomal dominant hearing loss mapping to chromosome 4q12-q13.1 (DFNA27; 612431), Nakano et al. (2018) identified heterozygosity for an intronic variant in the REST gene (600571.0007) that segregated fully with deafness in the family and was not found in public variant databases. Functional analysis revealed that the DFNA27-associated REST variant causes gain or loss of function depending on the cellular context: in cells lacking the splicing factor SRRM4 (613103), it causes REST inactivation (loss of function), whereas in cells expressing SRRM4, such as neurons and mechanosensory hair cells of the ear, the variant prevents REST inactivation by alternative splicing (gain of function), thus abrogating its normal downregulation in differentiating neurons.

In a South African Xhosa mother and son with progressive prelingual sensorineural hearing loss, Manyisa et al. (2021) identified heterozygosity for a missense mutation in the REST gene (C415S; 600571.0008) that segregated with disease in the family and was not found in controls or in public variant databases. Functional analysis revealed perturbation of cellular localization and loss of function with the mutant compared to wildtype REST.


Animal Model

Chen et al. (1998) disrupted the Rest gene in mice by gene targeting in mouse embryonic stem cells. As a result, derepression of neuron-specific tubulin (602529) in a subset of nonneural tissues resulted and embryonic lethality ensued.

Lepagnol-Bestel et al. (2009) used the transgenic 152F7 mouse model of Down syndrome (190685) to show that the DYRK1A (600855) gene dosage imbalance deregulated chromosomal clusters of genes located near REST/NRSF binding sites. Dyrk1a bound the SWI/SNF complex (see 603111), which is known to interact with REST/NRSF. Mutation of a REST/NRSF binding site in the promoter of the REST/NRSF target gene L1cam (308840) modified the transcriptional effect of Dyrk1a-dosage imbalance on L1cam. Dyrk1a dosage imbalance perturbed Rest/Nrsf levels with decreased Rest/Nrsf expression in embryonic neurons and increased expression in adult neurons. In transgenic embryonic brain subregions, the authors identified a coordinated deregulation of multiple genes that responsible for dendritic growth impairment. Similarly, Dyrk1a overexpression in primary mouse cortical neurons induced severe reduction of the dendritic growth and dendritic complexity. Lepagnol-Bestel et al. (2009) proposed that both the DYRK1A overexpression-related neuronal gene deregulation (via disturbance of REST/NRSF levels) and the REST/NRSF-SWI/SNF chromatin remodeling complex significantly contribute to the neural phenotypic changes that characterize Down syndrome.

Nakano et al. (2018) found that heterozygous deletion of exon 4 of mouse Rest activated the apoptotic pathway and caused hair cell death in a cell-autonomous manner, resulting in balance defects and deafness. Single-cell quantitative RT-PCR revealed that alternative splicing of Rest exon 4 regulated gene expression specifically in hair cells. As a result, lack of exon 4-dependent alternative splicing in mutant mice reduced expression of many hearing-related genes, especially genes critical for development and function of hair cells, causing defects in cilia of utricles. In vitro studies demonstrated that HDAC inhibitors increased expression of many Rest target genes and prevented degeneration of outer hair cells of mutant mice in organ cultures. The HDAC inhibitor SAHA (Vorinostat) rescued hair cells and hearing of Rest mutant mice in vivo.


ALLELIC VARIANTS ( 8 Selected Examples):

.0001 WILMS TUMOR 6, SUSCEPTIBILITY TO

REST, 2-BP DEL, 831AT
  
RCV000207462...

In 2 sisters (FAM0482) and an unrelated individual (FAM00250) with Wilms tumor (WT6; 616806), Mahamdallie et al. (2015) identified a heterozygous 2-bp deletion (c.831_832delAT, ENST00000309042) in the DNA binding domain of the REST gene resulting in frameshift. The mutation was inherited from the mother by the sisters and from the father by the unrelated individual. Age at diagnosis was 3.7 and 6.0 years in the sisters, and 3.2 years in the unrelated individual. Histology of the sisters' tumors showed triphasic (blastemal, epithelial, and stromal) elements. Mahamdallie et al. (2015) noted that this mutation was not identified in the ICR1000 UK exome series or in the ExAC browser.


.0002 WILMS TUMOR 6, SUSCEPTIBILITY TO

REST, 4-BP DEL, 772GTGA
  
RCV000207468

In 2 first cousins once removed (FAM0509) with Wilms tumor (WT6; 616806), Mahamdallie et al. (2015) identified a heterozygous 4-bp deletion (c.772_775delGTGA, ENST00000309042) in the DNA binding domain of the REST gene. In one cousin the mutation was inherited from the mother, age at diagnosis was 2.6 years, and the tumor sample showed triphasic histology. In the other cousin the mutation was inherited from the father, age at diagnosis was 0.8 years, and tumor histology was predominantly blastemal. Mahamdallie et al. (2015) noted that this mutation was not identified in the ICR1000 UK exome series or in the ExAC browser.


.0003 WILMS TUMOR 6, SUSCEPTIBILITY TO

REST, HIS322ARG
  
RCV000207458

In 2 cousins (FAM0481) and in an unrelated individual (FAM1324) with Wilms tumor (WT6; 616806), Mahamdallie et al. (2015) detected a heterozygous c.965A-G transition (c.965A-G, ENST00000309042) in the zinc finger DNA-binding domain of the REST gene, resulting in a his322-to-arg (H322R) substitution. The mutation was paternally inherited in the female cousin and maternally inherited in the male cousin. Age of onset was 3 years in female patient and 0.5 years in the male patient. Inheritance was unknown in the unrelated individual; Wilms tumor developed at 0.5 years of age and was of triphasic histology. Functional studies showed that this variant was unable to repress REST target gene expression, supporting pathogenicity. Mahamdallie et al. (2015) noted that this mutation was not identified in the ICR1000 UK exome series or in the ExAC browser.


.0004 FIBROMATOSIS, GINGIVAL, 5

REST, 2-BP DEL, 2865AA (SCV000579327)
  
RCV000497680...

In 2 Turkish brothers with gingival fibromatosis-5 (GINGF5; 617626), Bayram et al. (2017) identified a heterozygous 2-bp deletion (c.2865_2866delAA, NM_005612.4) in the last exon (exon 4) of the REST gene, resulting in a frameshift and premature termination (Asn958SerfsTer9). The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the dbSNP, 1000 Genomes Project, Exome Sequencing Project, or ExAC databases, or in an in-house database of more than 6,500 exomes. The father, who had a mild form of the disorder, was mosaic for the mutation. Functional studies of the variant and studies of patient cells were not performed.


.0005 FIBROMATOSIS, GINGIVAL, 5

REST, LEU437TER (SCV000579328)
  
RCV000498225...

In 5 members of a Turkish family with gingival fibromatosis-5 (GINGF5; 617626), Bayram et al. (2017) identified a heterozygous c.1310T-A transversion (c.1310T-A, NM_005612.4) in the last exon (exon 4) of the REST gene, resulting in a leu437-to-ter (L437X) substitution. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the dbSNP, 1000 Genomes Project, Exome Sequencing Project, or ExAC databases, or in an in-house database of more than 6,500 exomes. The family was originally reported by Pehlivan et al. (2009). Functional studies of the variant and studies of patient cells were not performed.


.0006 FIBROMATOSIS, GINGIVAL, 5

REST, 1-BP DEL, 2413C (SCV000579329)
  
RCV000498949...

In a 9-year-old Turkish girl with gingival fibromatosis-5 (GINGF5; 617626), Bayram et al. (2017) identified a de novo heterozygous 1-bp deletion (c.2413delC, NM_005612.4) in the last exon (exon 4) of the REST gene, resulting in a frameshift and premature termination (Leu805PhefsTer38). The variant, which was found by exome sequencing and confirmed by Sanger sequencing, was not found in the dbSNP, 1000 Genomes Project, Exome Sequencing Project, or ExAC databases, or in an in-house database of more than 6,500 exomes. The patient was identified through the GeneMatcher database. Functional studies of the variant and studies of patient cells were not performed.


.0007 DEAFNESS, AUTOSOMAL DOMINANT 27

REST, IVS3AS, C-G, -21
  
RCV000855719

In a 3-generation family (LMG2) with autosomal dominant hearing loss (DFNA27; 612431), Nakano et al. (2018) identified heterozygosity for a C-to-G transversion (chr4.56,927,594C-G, GRCh38) at a conserved nucleotide in intron 3, 21 bp upstream of exon 4a/b of the REST gene. The mutation segregated fully with deafness in the family and was not found in 400 control DNA samples or in the gnomAD database. RT-PCR in patient and control blood cells showed that the C-G variant causes truncation of the coding region of REST by generating a novel splice acceptor site for exon 4a. The authors noted that splicing of exon 4 into REST mRNA is normally tissue-specific and requires SRRM4 (613103), which is selectively expressed in neurons and mechanosensory hair cells of the ear. Using minigenes transfected into HEK293 cells, they observed that DFNA27-associated aberrant splicing results in a novel splice form containing a 24-nucleotide variant of exon 4b, and splicing of this variant exon 4b requires both the C-G variant-dependent relocation of the splice acceptor site as well as an SRRM4-dependent shift in the splice donor site. Analysis of luciferase activity of isoforms generated in the presence or absence of SRRM4 suggested that the C-G DFNA27-associated variant has opposing effects on REST depending on whether SRRM4 is present. In cells that do not express SRRM4, the C-G variant aberrantly inactivates REST by creating a novel acceptor site for constitutive splicing upstream of exon 4a/b. Conversely, in cells that do express SRRM4, the C-G variant aberrantly results in production of active REST that cannot be inactivated by SRRM4-directed alternative splicing of exon 4, thus abrogating the normal downregulation of REST that occurs in differentiating neurons.


.0008 DEAFNESS, AUTOSOMAL DOMINANT 27

REST, CYS415SER
  
RCV002248339

In a South African Xhosa mother and son with progressive prelingual sensorineural hearing loss (DFNA27; 612431), Manyisa et al. (2021) identified heterozygosity for a c.1244G-C transversion (c.1244G-C, NM_005612.5) in exon 4 of the REST gene, resulting in a cys415-to-ser (C415S) substitution at a highly conserved residue. The mutation was not found in the proband's unaffected half-brother or unaffected maternal grandmother, in 103 black South African controls or 52 sporadic South African probands of black or mixed ancestry with nonsyndromic hearing impairment, or in the gnomAD, UK10K, Greater Middle East Variome (GME), or dbSNP databases. Experiments using GFP tagging in HEK293 cells showed that the wildtype REST protein is located exclusively within the nucleus, whereas the mutant showed localization throughout the cell, indicating loss of exclusive nuclear shuttling/localization. In addition, wildtype REST competently repressed transcription of the known REST target AF1Q (604684) in transiently transfected HEK293 cells, whereas transcriptional repression was lost in cells expressing the mutant REST protein.


REFERENCES

  1. Abderrahmani, A., Steinmann, M., Plaisance, V., Niederhauser, G., Haefliger, J.-A., Mooser, V., Bonny, C., Nicod, P., Waeber, G. The transcriptional repressor REST determines the cell-specific expression of the human MAPK8IP1 gene encoding IB1 (JIP-1). Molec. Cell. Biol. 21: 7256-7627, 2001. [PubMed: 11585908, images, related citations] [Full Text]

  2. Bahn, S., Mimmack, M., Ryan, M., Caldwell, M. A., Jauniaux, E., Starkey, M., Svendsen, C. N., Emson, P. Neuronal target genes of the neuron-restrictive silencer factor in neurospheres derived from fetuses with Down's syndrome: a gene expression study. Lancet 359: 310-315, 2002. [PubMed: 11830198, related citations] [Full Text]

  3. Bayram, Y., White, J. J., Elcioglu, N., Cho, M. T., Zadeh, N., Gedikbasi, A., Palanduz, S., Ozturk, S., Cefle, K., Kasapcopur, O., Akdemir, Z. C., Pehlivan, D., and 11 others. REST final-exon-truncating mutations cause hereditary gingival fibromatosis. Am. J. Hum. Genet. 101: 149-156, 2017. [PubMed: 28686854, images, related citations] [Full Text]

  4. Canzonetta, C., Mulligan, C., Deutsch, S., Ruf, S., O'Doherty, A., Lyle, R., Borel, C., Lin-Marq, N., Delom, F., Groet, J., Schnappauf, F., De Vita, S, and 12 others. DYRK1A-dosage imbalance perturbs NRSF/REST levels, deregulating pluripotency and embryonic stem cell fate in Down syndrome. Am. J. Hum. Genet. 83: 388-400, 2008. [PubMed: 18771760, images, related citations] [Full Text]

  5. Chen, Z.-F., Paquette, A. J., Anderson, D. J. NRSF/REST is required in vivo for repression of multiple neuronal target genes during embryogenesis. Nature Genet. 20: 136-142, 1998. [PubMed: 9771705, related citations] [Full Text]

  6. Cheong, A., Bingham, A. J., Li, J., Kumar, B., Sukumar, P., Munsch, C., Buckley, N. J., Neylon, C. B., Porter, K. E., Beech, D. J., Wood, I. C. Downregulated REST transcription factor is a switch enabling critical potassium channel expression and cell proliferation. Molec. Cell 20: 45-52, 2005. [PubMed: 16209944, related citations] [Full Text]

  7. Chong, J. A., Tapia-Ramirez, J., Kim, S., Toledo-Aral, J. J., Zheng, Y., Boutros, M. C., Altshuller, Y. M., Frohman, M. A., Kraner, S. D., Mandel, G. REST: a mammalian silencer protein that restricts sodium channel gene expression to neurons. Cell 80: 949-957, 1995. [PubMed: 7697725, related citations] [Full Text]

  8. Das, C. M., Taylor, P., Gireud, M., Singh, A., Lee, D., Fuller, G., Ji, L., Fangusaro, J., Rajaram, V., Goldman, S., Eberhart, C., Gopalakrishnan, V. The deubiquitylase USP37 links REST to the control of p27 stability and cell proliferation. Oncogene 32: 1691-1701, 2013. Note: Erratum: Oncogene 35: 6153-6154, 2016. [PubMed: 22665064, images, related citations] [Full Text]

  9. Ding, N., Zhou, H., Esteve, P.-O., Chin, H. G., Kim, S., Xu, X., Joseph, S. M., Friez, M. J., Schwartz, C. E., Pradhan, S., Boyer, T. G. Mediator links epigenetic silencing of neuronal gene expression with X-linked mental retardation. Molec. Cell 31: 347-359, 2008. [PubMed: 18691967, images, related citations] [Full Text]

  10. Guardavaccaro, D., Frescas, D., Dorrello, N. V., Peschiaroli, A., Multani, A. S., Cardozo, T., Lasorella, A., Iavarone, A., Chang, S., Hernando, E., Pagano, M. Control of chromosome stability by the beta-TrCP-REST-Mad2 axis. Nature 452: 365-369, 2008. [PubMed: 18354482, images, related citations] [Full Text]

  11. Kemp, D. M., Lin, J. C., Habener, J. F. Regulation of Pax4 paired homeodomain gene by neuron-restrictive silencer factor. J. Biol. Chem. 278: 35057-35062, 2003. [PubMed: 12829700, related citations] [Full Text]

  12. Kuwahara, K., Saito, Y., Takano, M., Arai, Y., Yasuno, S., Nakagawa, Y., Takahashi, N., Adachi, Y., Takemura, G., Horie, M., Miyamoto, Y., Morisaki, T., and 12 others. NRSF regulates the fetal cardiac gene program and maintains normal cardiac structure and function. EMBO J. 22: 6310-6321, 2003. [PubMed: 14633990, images, related citations] [Full Text]

  13. Lepagnol-Bestel, A.-M., Zvara, A., Maussion, G., Quignon, F., Ngimbous, B., Ramoz, N., Imbeaud, S., Loe-Mie, Y., Benihoud, K., Agier, N., Salin, P. A., Cardona, A., and 11 others. DYRK1A interacts with the REST/NRSF-SWI/SNF chromatin remodelling complex to deregulate gene clusters involved in the neuronal phenotypic traits of Down syndrome. Hum. Molec. Genet. 18: 1405-1414, 2009. Note: Erratum: Hum. Molec. Genet. 31: 2106-2107, 2022. [PubMed: 19218269, related citations] [Full Text]

  14. Loe-Mie, Y., Lepagnol-Bestel, A.-M., Maussion, G., Doron-Faigenboim, A., Imbeaud, S., Delacroix, H., Aggerbeck, L., Pupko, T., Gorwood, P., Simonneau, M., Moalic, J.-M. SMARCA2 and other genome-wide supported schizophrenia-associated genes: regulation by REST/NRSF, network organization and primate-specific evolution. Hum. Molec. Genet. 19: 2841-2857, 2010. [PubMed: 20457675, related citations] [Full Text]

  15. Lu, T., Aron, L., Zullo, J., Pan, Y., Kim, H., Chen, Y., Yang, T.-H., Kim, H.-M., Drake, D., Liu, X. S., Bennett, D. A., Colaiacovo, M. P., Yankner, B. A. REST and stress resistance in ageing and Alzheimer's disease. Nature 507: 448-454, 2014. Note: Erratum: Nature 540: 470 only, 2016. [PubMed: 24670762, related citations] [Full Text]

  16. Lunyak, V. V., Burgess, R., Prefontaine, G. G., Nelson, C., Sze, S.-H., Chenoweth, J., Schwartz, P., Pevzner, P. A., Glass, C., Mandel, G., Rosenfeld, M. G. Corepressor-dependent silencing of chromosomal regions encoding neuronal genes. Science 298: 1747-1752, 2002. Note: Erratum: Science 299: 1663 only, 2003. [PubMed: 12399542, related citations] [Full Text]

  17. Mahamdallie, S. S., Hanks, S., Karlin, K. L., Zachariou, A., Perdeaux, E. R., Ruark, E., Shaw, C. A., Renwick, A., Ramsay, E., Yost, S., Elliott, A., Birch, J., and 13 others. Mutations in the transcriptional repressor REST predispose to Wilms tumor. Nature Genet. 47: 1471-1474, 2015. Note: Erratum: Nature Genet. 48: 473 only, 2016. [PubMed: 26551668, related citations] [Full Text]

  18. Manyisa, N., Schrauwen, I., Alves de Souza Rios, L., Mowla, S., Tekendo-Ngongang, C., Popel, K., Esoh, K., Bharadwaj, T., Nouel-Saied, L. M., Acharya, A., Nasir, A., Wonkam-Tingang, E., de Kock, C., Dandara C., Leal, S. M., Wonkam, A. A monoallelic variant in REST is associated with non-syndromic autosomal dominant hearing impairment in a South African family. Genes 12: 1765, 2021. [PubMed: 34828371, images, related citations] [Full Text]

  19. Nakano, Y., Kelly, M. C., Rehman, A. U., Boger, E. T., Morell, R. J., Kelley, M. W., Friedman, T. B., Banfi, B. Defects in the alternative splicing-dependent regulation of REST cause deafness. Cell 174: 536-548, 2018. [PubMed: 29961578, images, related citations] [Full Text]

  20. Ooi, L., Wood, I. C. Chromatin crosstalk in development and disease: lessons from REST. Nature Rev. Genet. 8: 544-554, 2007. [PubMed: 17572692, related citations] [Full Text]

  21. Palm, K., Metsis, M., Timmusk, T. Neuron-specific splicing of zinc finger transcription factor REST/NRSF/XBR is frequent in neuroblastomas and conserved in human, mouse and rat. Molec. Brain Res. 72: 30-39, 1999. [PubMed: 10521596, related citations] [Full Text]

  22. Pehlivan, D., Abe, S., Ozturk, S., Kayhan, K. B., Gunduz, E., Cefle, K., Bayrak, A., Ark, N., Gunduz, M., Palanduz, S. Cytogenetic analysis and examination of SOS1 gene mutation in a Turkish family with hereditary gingival fibromatosis. J. Hard Tissue Biol. 18: 131-134, 2009.

  23. Plaisance, V., Niederhauser, G., Azzouz, F., Lenain, V., Haefliger, J.-A., Waeber, G., Abderrahmani, A. The repressor element silencing transcription factor (REST)-mediated transcriptional repression requires the inhibition of Sp1. J. Biol. Chem. 280: 401-407, 2005. [PubMed: 15528196, related citations] [Full Text]

  24. Schoenherr, C. J., Anderson, D. J. The neuron-restrictive silencer factor (NRSF): a coordinate repressor of multiple neuron-specific genes. Science 267: 1360-1363, 1995. [PubMed: 7871435, related citations] [Full Text]

  25. Shimojo, M. Huntingtin regulates RE1-silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) nuclear trafficking indirectly through a complex with REST/NRSF-interacting LIM domain protein (RILP) and dynactin p150-Glued. J. Biol. Chem. 283: 34880-34886, 2008. [PubMed: 18922795, images, related citations] [Full Text]

  26. Singh, S. K., Kagalwala, M. N., Parker-Thornburg, J., Adams, H., Majumder, S. REST maintains self-renewal and pluripotency of embryonic stem cells. Nature 453: 223-227, 2008. [PubMed: 18362916, images, related citations] [Full Text]

  27. Tahiliani, M., Mei, P., Fang, R., Leonor, T., Rutenberg, M., Shimizu, F., Li, J., Rao, A., Shi, Y. The histone H3K4 demethylase SMCX links REST target genes to X-linked mental retardation. Nature 447: 601-605, 2007. [PubMed: 17468742, related citations] [Full Text]

  28. Thiel, G., Lietz, M., Cramer, M. Biological activity and modulator structure of RE-1-silencing transcription factor (REST), a repressor of neuronal genes. J. Biol. Chem. 273: 26891-26899, 1998. [PubMed: 9756936, related citations] [Full Text]

  29. Westbrook, T. F., Hu, G., Ang, X. L., Mulligan, P., Pavlova, N. N., Liang, A., Leng, Y., Maehr, R., Shi, Y., Harper, J. W., Elledge, S. J. SCF-(beta-TRCP) controls oncogenic transformation and neural differentiation through REST degradation. Nature 452: 370-374, 2008. [PubMed: 18354483, images, related citations] [Full Text]

  30. Yang, Y. J., Baltus, A. E., Mathew, R. S., Murphy, E. A., Evrony, G. D., Gonzalez, D. M., Wang, E. P., Marshall-Walker, C. A., Barry, B. J., Murn, J., Tatarakis, A., Mahajan, M. A., Samuels, H. H., Shi, Y., Golden, J. A., Mahajnah, M., Shenhav, R., Walsh, C. A. Microcephaly gene links trithorax and REST/NRSF to control neural stem cell proliferation and differentiation. Cell 151: 1097-1112, 2012. [PubMed: 23178126, related citations] [Full Text]

  31. Yeo, M., Lee, S.-K., Lee, B., Ruiz, E. C., Pfaff, S. L., Gill, G. N. Small CTD phosphatases function in silencing neuronal gene expression. Science 307: 596-600, 2005. [PubMed: 15681389, related citations] [Full Text]

  32. Yoo, A. S., Staahl, B. T., Chen, L., Crabtree, G. R. MicroRNA-mediated switching of chromatin-remodelling complexes in neural development. Nature 460: 642-646, 2009. Note: Erratum: Nature 461: 296 only, 2009. [PubMed: 19561591, images, related citations] [Full Text]

  33. Zuccato, C., Ciammola, A., Rigamonti, D., Leavitt, B. R., Goffredo, D., Conti, L., MacDonald, M. E., Friedlander, R. M., Silani, V., Hayden, M. R., Timmusk, T., Sipione, S., Cattaneo, E. Loss of huntingtin-mediated BDNF gene transcription in Huntington's disease. Science 293: 493-498, 2001. [PubMed: 11408619, related citations] [Full Text]

  34. Zuccato, C., Tartari, M., Crotti, A., Goffredo, D., Valenza, M., Conti, L., Cataudella, T., Leavitt, B. R., Hayden, M. R., Timmusk, T., Rigamonti, D., Cattaneo, E. Huntingtin interacts with REST/NRSF to modulate the transcription of NRSE-controlled neuronal genes. Nature Genet. 35: 76-83, 2003. [PubMed: 12881722, related citations] [Full Text]

  35. Zullo, J. M., Drake, D., Aron, L., O'Hern, P., Dhamne, S. C., Davidsohn, N., Mao, C.-A., Klein, W. H., Rotenberg, A., Bennett, D. A., Church, G. M., Colaiacovo, M. P., Yankner, B. A. Regulation of lifespan by neural excitation and REST. Nature 574: 359-364, 2019. [PubMed: 31619788, images, related citations] [Full Text]


Bao Lige - updated : 01/31/2023
Marla J. F. O'Neill - updated : 04/22/2022
Ada Hamosh - updated : 04/09/2020
Bao Lige - updated : 01/09/2020
Marla J. F. O'Neill - updated : 11/12/2019
Bao Lige - updated : 09/23/2019
Cassandra L. Kniffin - updated : 08/14/2017
Ada Hamosh - updated : 02/11/2016
Ada Hamosh - updated : 4/11/2014
George E. Tiller - updated : 8/27/2013
Cassandra L. Kniffin - updated : 2/25/2013
Patricia A. Hartz - updated : 4/13/2012
George E. Tiller - updated : 11/30/2009
Ada Hamosh - updated : 8/27/2009
Patricia A. Hartz - updated : 8/20/2009
Ada Hamosh - updated : 11/5/2008
Ada Hamosh - updated : 6/12/2008
Ada Hamosh - updated : 5/22/2008
Patricia A. Hartz - updated : 9/25/2007
Ada Hamosh - updated : 6/15/2007
Patricia A. Hartz - updated : 12/12/2005
Ada Hamosh - updated : 3/1/2005
Victor A. McKusick - updated : 8/15/2003
Cassandra L. Kniffin - updated : 6/23/2003
Ada Hamosh - updated : 4/3/2003
Patricia A. Hartz - updated : 6/13/2002
Victor A. McKusick - updated : 9/28/1998
Creation Date:
Alan F. Scott : 6/1/1995
carol : 06/08/2023
mgross : 01/31/2023
alopez : 07/27/2022
carol : 04/22/2022
carol : 02/15/2022
carol : 07/17/2020
carol : 06/23/2020
alopez : 04/09/2020
mgross : 01/09/2020
carol : 11/13/2019
alopez : 11/12/2019
alopez : 11/12/2019
carol : 10/08/2019
mgross : 09/23/2019
carol : 08/24/2017
carol : 08/24/2017
carol : 08/15/2017
ckniffin : 08/14/2017
alopez : 02/11/2016
alopez : 4/11/2014
carol : 8/27/2013
tpirozzi : 8/27/2013
tpirozzi : 8/27/2013
carol : 2/25/2013
ckniffin : 2/25/2013
mgross : 5/18/2012
mgross : 5/18/2012
terry : 4/13/2012
wwang : 1/7/2010
terry : 11/30/2009
mgross : 10/5/2009
carol : 9/23/2009
carol : 9/15/2009
alopez : 9/4/2009
terry : 8/27/2009
mgross : 8/24/2009
terry : 8/20/2009
carol : 6/3/2009
alopez : 12/1/2008
terry : 11/5/2008
alopez : 6/17/2008
terry : 6/12/2008
alopez : 5/28/2008
alopez : 5/28/2008
terry : 5/22/2008
mgross : 10/2/2007
terry : 9/25/2007
alopez : 6/20/2007
terry : 6/15/2007
wwang : 12/12/2005
wwang : 3/7/2005
terry : 3/1/2005
alopez : 9/2/2003
alopez : 8/19/2003
alopez : 8/19/2003
terry : 8/15/2003
carol : 7/10/2003
carol : 7/9/2003
ckniffin : 6/23/2003
alopez : 4/3/2003
terry : 4/3/2003
carol : 6/20/2002
terry : 6/13/2002
alopez : 9/28/1998
joanna : 9/28/1998
mark : 3/6/1998
joanna : 12/6/1995
mark : 6/1/1995

* 600571

RE1-SILENCING TRANSCRIPTION FACTOR; REST


Alternative titles; symbols

NEURON-RESTRICTIVE SILENCER FACTOR; NRSF


HGNC Approved Gene Symbol: REST

Cytogenetic location: 4q12     Genomic coordinates (GRCh38): 4:56,907,900-56,935,844 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
4q12 {Wilms tumor 6, susceptibility to} 616806 Autosomal dominant 3
Deafness, autosomal dominant 27 612431 Autosomal dominant 3
Fibromatosis, gingival, 5 617626 Autosomal dominant 3

TEXT

Description

REST is a transcriptional repressor that regulates gene expression throughout the body. It binds 21-bp repressor element-1 (RE1) sites, also called neuron-restrictive silencer elements (NRSEs), through its 8 C2H2 zinc fingers. REST mediates gene repression by acting as a hub for the recruitment of multiple chromatin-modifying enzymes (Ooi and Wood, 2007).


Cloning and Expression

Schoenherr and Anderson (1995) cloned a transcription factor, which they termed NRSF, that bound the NRSE present in the 5-prime regulatory region of SCG10 (600621), a neuron-specific gene. The NRSF cDNA was cloned from a HeLa cell library. The longest cDNA was predicted to encode 8 zinc fingers of the C2H2 class, with interfinger sequences that placed NRSE within the GLI (see 165220)-Kruppel family of zinc finger proteins. Northern blot analysis detected a NRSF transcript of 7 to 8 kb. Expression of NRSF mRNA was detected in most nonneuronal progenitor cells, but it was absent in differentiated neurons.

Independently, Chong et al. (1995) cloned REST from a HeLa cell cDNA library. The deduced 1,097-amino acid protein has a calculated molecular mass of 121 kD. REST has 8 N-terminal C2H2-type zinc fingers, followed by a basic region, 6 repeats of a proline-rich sequence, and a C-terminal zinc finger. It also has a nuclear localization signal. In situ hybridization of mouse embryos detected abundant Rest expression in nonneuronal tissues.

Thiel et al. (1998) determined that REST contains 2 repressor domains, one located at the N terminus and the other at the C terminus, and an N-terminal zinc finger cluster which functions as the DNA-binding domain for neuronal genes.

Palm et al. (1999) identified several REST variants that arise from alternative splicing of an exon that they designated exon N. The splice variants produce insertions that generate in-frame stop codons and encode truncated proteins with an N-terminal repressor domain and weakened DNA-binding activity. The expression levels of these variants differ in human neuroblastoma and glial cells.


Gene Function

Schoenherr and Anderson (1995) showed that NRSF bound the NRSE DNA sequence in the 5-prime region of SCG10. They noted that the SCG10 regulatory region contains both activation and repression (i.e., silencer) domains and that similar NRSE-like sequence elements have been identified in other neuron-specific genes. Schoenherr and Anderson (1995) proposed that NRSF may function as a master negative regulator of neurogenesis.

By transfecting rat myocyte and pheochromocytoma cell lines, Chong et al. (1995) showed that human REST downregulated expression of the type II voltage-dependent sodium channel (see 182390). They proposed that REST suppresses expression of the channel in nonneuronal tissues.

Thiel et al. (1998) noted that the REST binding sites of several neuron-specific genes, such as those encoding synapsin I (313440), SCG10 (600621), A1-glycine receptor (138491), the B2 subunit of the nicotinic acetylcholine receptor (118507), and the M4-subunit of the muscarinic acetylcholine receptor, are found at various positions within the sequence. By transfecting these sequences in reporter constructs together with REST, Thiel et al. (1998) found that REST blocks transcription of a gene irrespective of whether the NRSE is located upstream or downstream of the open reading frame in either orientation and in both a distance- and a gene-independent manner.

Abderrahmani et al. (2001) identified an NRSE sequence in the promoter region of MAPK8PI1 (604641), which is expressed exclusively in neuronal tissue and pancreatic B cells. They confirmed that REST binds to the NRSE element of MAPK8PI1 and found that transfection and expression of REST in a B-cell line represses MAPK8PI1 transcription. Conversely, the introduction of a mutated NRSE into the MAPK8PI1 promoter allowed MAPK8PI1 expression in non-B-cell and nonneuronal cell lines. REST-mediated repression was found to be dependent on histone deacetylase (see 601241) activity.

Lunyak et al. (2002) reported that the zinc finger gene-specific repressor element REST can mediate extraneuronal restriction by imposing either active repression via histone deacetylase recruitment or long-term gene silencing using a distinct functional complex. Silencing of neuronal-specific genes requires the recruitment of an associated corepressor, COREST (607675), that serves as a functional molecular beacon for recruitment of molecular machinery that imposes silencing across a chromosomal interval, including transcriptional units that do not themselves contain REST/NRSF response elements.

Using indexing-based differential display PCR on neuronal precursor cells to study gene expression in Down syndrome (190685), Bahn et al. (2002) found that genes regulated by the REST transcription factor were selectively repressed. One of these genes, SCG10, was almost undetectable. The REST factor itself was also downregulated by 49% compared to controls. In cell culture, the Down syndrome cells showed a reduction of neurogenesis, as well as decreased neurite length and abnormal changes in neuron morphology. The authors noted that REST-regulated genes play an important part in brain development, plasticity, and synapse formation, and they suggested a link between dysregulation of REST and some of the neurologic deficits seen in Down syndrome.

The huntingtin gene (HTT; 613004) is mutated in Huntington disease (HD; 143100). Zuccato et al. (2001) reported that wildtype but not mutant huntingtin stimulates transcription of the gene encoding brain-derived neurotrophic factor (BDNF; 113505). Zuccato et al. (2003) showed that the NRSE is the target of wildtype huntingtin activity on BDNF promoter II. Wildtype huntingtin inhibits the silencing activity of NRSE, increasing transcription of BDNF. Zuccato et al. (2003) showed that this effect occurs through cytoplasmic sequestering of REST/NRSF, the transcription factor that binds to NRSE. In contrast, aberrant accumulation of REST/NRSF in the nucleus was present in Huntington disease. They showed that wildtype huntingtin coimmunoprecipitates with REST/NRSF and that less immunoprecipitated material is found in brain tissue with Huntington disease. They also reported that wildtype huntingtin acts as a positive transcriptional regulator for other NRSE-containing genes involved in the maintenance of the neuronal phenotype. Consistently, loss of expression of NRSE-controlled neuronal genes was shown in cells, mice, and human brain with Huntington disease. Zuccato et al. (2003) concluded that wildtype huntingtin acts in the cytoplasm of neurons to regulate the availability of REST/NRSF to its nuclear NRSE-binding site and that this control is lost in the pathology of Huntington disease. These data identified a novel mechanism by which mutation of huntingtin causes loss of transcription of neuronal genes.

Kemp et al. (2003) identified NRSE-like motifs in several genes involved in pancreas development, including a highly conserved NRSE-like motif in the upstream promoter of PAX4 (167413), a gene implicated in differentiation of the insulin-producing beta-cell lineage. Using mammalian cell lines, they found that the NRSE in the upstream promoter of Pax4 formed a DNA-protein complex with Nrsf and conferred Nrsf-dependent transcriptional repression on a reporter gene promoter and the native Pax4 gene promoter.

Reactivation of the fetal cardiac gene program is a characteristic feature of hypertrophied and failing hearts. Kuwahara et al. (2003) showed that Nrsf selectively regulated expression of multiple fetal cardiac genes and played a role in reexpression of these genes in rat neonatal ventricular myocytes. Transgenic mice expressing a dominant-negative Nrsf mutant in their hearts exhibited dilated cardiomyopathy, high susceptibility to arrhythmias, and sudden death. Genes encoding 2 ion channels that carry the fetal cardiac currents I(f) and I(Ca,T), which were induced in Nrsf-transgenic mice and were potentially responsible for both the cardiac dysfunction and arrhythmogenesis, were regulated by Nrsf.

Neuronal gene transcription is repressed in nonneuronal cells by the REST/NRSF complex. To understand how this silencing is achieved, Yeo et al. (2005) examined CTDSP1 (605323), CTDSP2 (608711), and CTDSPL (608592), the small CTD phosphatases (SCP), whose expression is restricted to nonneuronal tissues. Yeo et al. (2005) showed that REST/NRSF recruits SCPs to neuronal genes that contain RE1 elements, leading to neuronal gene silencing in nonneuronal cells. Phosphatase-inactive forms of SCP interfere with REST/NRSF function and promote neuronal differentiation of P19 stem cells. Likewise, small interfering RNA directed to the single Drosophila SCP unmasks neuronal gene expression in S2 cells. Thus, Yeo et al. (2005) concluded that SCP activity is an evolutionarily conserved transcriptional regulator that acts globally to silence neuronal genes.

Cheong et al. (2005) identified a functional REST-binding sequence in the promoter region of the KCNN4 gene (602754). REST was expressed in the nuclei of human vascular smooth muscle cells (SMCs), and it downregulated KCNN4 expression in mouse and human vascular SMCs. Downregulated REST and upregulated KCNN4 were evident in SMCs of human neointimal hyperplasia grown in organ culture, and exogenous REST reduced the functional impact of KCNN4. Cheong et al. (2005) concluded that REST acts as a switch to regulate potassium channel expression and consequently the phenotype of vascular smooth muscle cells and human vascular disease.

Plaisance et al. (2005) showed that the transcriptional factor Sp1 (189906) was required for expression of most Rest target genes in mouse insulin-secreting cells and rat neuronal-like cells where Rest is absent. Inhibition of REST in HeLa cells and in mouse beta cells restored the transcriptional activity of Sp1. Coimmunoprecipitation and transfection assays indicated that the C-terminal repressor domain of REST was required for interaction with Sp1 and inhibited its activity. Silencing of Sp1 by REST required histone deacetylase activity.

Tahiliani et al. (2007) showed that JARID1C/SMCX (314690), a JmjC domain-containing protein implicated in X-linked mental retardation and epilepsy, possesses H3K4 tridemethylase activity and functions as a transcriptional repressor. An SMCX complex isolated from HeLa cells contained additional chromatin modifiers (the histone deacetylases HDAC1 (601241) and HDAC2 (605164), and the histone H3K9 methyltransferase G9a (604599)) and the transcriptional repressor REST, suggesting a direct role for SMCX in chromatin dynamics and REST-mediated repression. Chromatin immunoprecipitation revealed that SMCX and REST co-occupy the neuron-restrictive silencing elements in the promoters of a subset of REST target genes. RNA interference-mediated depletion of SMCX derepressed several of these targets and simultaneously increased H3K4 trimethylation at the sodium channel type 2A (SCN2A; 182390) and synapsin I (SYN1; 313440) promoters. Tahiliani et al. (2007) proposed that loss of SMCX activity impairs REST-mediated neuronal gene regulation, thereby contributing to SMCX-associated X-linked mental retardation.

Ding et al. (2008) found that purified HeLa cell mediator complexes that included MED12 (300188) interacted directly with the G9A and REST. Endogenous REST in HEK293 cells suppressed expression of a reporter gene bearing RE1 sites, and knockdown of either MED12 or G9A abrogated the suppression. Depletion of MED12 significantly reduced the association of G9A with RE1 elements and decreased the level of H3K9 dimethylation by G9A without influencing RE1 site occupancy by REST.

Using an unbiased screen, Guardavaccaro et al. (2008) demonstrated that REST is an interactor with the F-box protein beta-TRCP (603482). REST is degraded by means of the ubiquitin beta-TRCP during the G2 phase of the cell cycle to allow transcriptional derepression of Mad2 (601467), an essential component of the spindle assembly checkpoint. The expression in cultured cells of a stable REST mutant, which is unable to bind beta-TRCP, inhibited Mad2 expression and resulted in a phenotype analogous to that observed in Mad2 heterozygous cells. In particular, Guardavaccaro et al. (2008) observed defects that were consistent with faulty activation of the spindle checkpoint, such as shortened mitosis, premature sister-chromatid separation, chromosome bridges and missegregation in anaphase, tetraploidy, and a faster mitotic slippage in the presence of a spindle inhibitor. An indistinguishable phenotype was observed by expressing the oncogenic REST-FS mutant, which does not bind beta-TRCP. Thus, beta-TRCP-dependent degradation of REST during G2 permits the optimal activation of the spindle checkpoint, and consequently it is required for the fidelity of mitosis.

Westbrook et al. (2008) showed that REST is regulated by ubiquitin-mediated proteolysis, and used an RNA interference screen to identify a Skp1-Cul1-F-box protein complex containing the F-box protein beta-TRCP as an E3 ubiquitin ligase responsible for REST degradation. Beta-TRCP binds and ubiquitinates REST and controls its stability through a conserved phospho-degron. During neural differentiation, REST is degraded in a beta-TRCP-dependent manner. Beta-TRCP is required for proper neural differentiation only in the presence of REST, indicating that beta-TRCP facilitates this process through degradation of REST. Conversely, failure to degrade REST attenuates differentiation. Furthermore, Westbrook et al. (2008) found that beta-TRCP overexpression, which is common in human epithelial cancers, causes oncogenic transformation of human mammary epithelial cells and that this pathogenic function requires REST degradation. Thus, Westbrook et al. (2008) concluded that REST is a key target in beta-TRCP-driven transformation and that the beta-TRCP-REST axis is a new regulatory pathway controlling neurogenesis.

Singh et al. (2008) demonstrated that REST maintains self-renewal and pluripotency in mouse ES cells through suppression of the microRNA miR21 (611020). The authors found that, as with known self-renewal markers, the level of REST expression is much higher in self-renewing mouse embryonic stem (ES) cells than in differentiating mouse ES (embryoid body, EB) cells. Heterozygous deletion of Rest and its short interfering RNA (siRNA)-mediated knockdown in mouse ES cells caused a loss of self-renewal--even when these cells were grown under self-renewal conditions--and led to the expression of markers specific for multiple lineages. Conversely, exogenously added REST maintained self-renewal in mouse EB cells. Furthermore, Rest heterozygous mouse ES cells cultured under self-renewal conditions expressed substantially reduced levels of several self-renewal regulators, including Oct4 (164177), Nanog (607937), Sox2 (184429), and c-Myc (190080), and exogenously added Rest in mouse EB cells maintained the self-renewal phenotypes and expression of these self-renewal regulators. Singh et al. (2008) also demonstrated that in mouse ES cells, Rest is bound to the gene chromatin of a set of miRNAs that potentially target self-renewal genes. Whereas mouse ES cells and mouse EB cells containing exogenously added Rest expressed lower levels of these miRNAs, EB cells, Rest heterozygous ES cells, and ES cells treated with siRNA targeting Rest expressed higher levels of these miRNAs. At least one of these REST-regulated miRNAs, miR21, specifically suppressed the self-renewal of mouse ES cells, corresponding to the decreased expression of Oct4, Nanog, Sox2, and c-Myc. Thus, Singh et al. (2008) concluded that REST is an element of the interconnected regulatory network that maintains the self-renewal and pluripotency of mouse ES cells.

Using a transchromosomic mouse model of Down syndrome, Canzonetta et al. (2008) showed that a 30 to 60% reduced expression of Nrsf/Rest, a key regulator of pluripotency and neuronal differentiation, is an alteration that persists in trisomy 21 (see 190685) from undifferentiated embryonic stem cells to adult brain and is reproducible across several Down syndrome models. Using partially trisomic ES cells, Canzonetta et al. (2008) mapped this effect to a 3-gene segment of human chromosome 21 containing DYRK1A (600855). The authors independently identified the same locus as the most significant expression quantitative trait locus (eQTL) controlling REST expression in the human genome. Canzonetta et al. (2008) found that specifically silencing the third copy of DYRK1A rescued Rest levels, and demonstrated altered Rest expression in response to inhibition of DYRK1A expression or kinase activity, and in a transgenic Dyrk1a mouse. The authors observed that undifferentiated trisomy 21 ES cells showed DYRK1A-dose-sensitive reductions in levels of some pluripotency regulators, including Nanog (607937) and Sox2 (184429), causing premature expression of transcription factors driving early endodermal and mesodermal differentiation, partially overlapping downstream effects of Rest heterozygosity. The ES cells produced embryoid bodies with elevated levels of the primitive endoderm progenitor marker Gata4 (600576) and a strongly reduced neuroectodermal progenitor compartment. Canzonetta et al. (2008) concluded that DYRK1A-mediated deregulation of REST is a very early pathologic consequence of trisomy 21 with potential to disturb the development of all embryonic lineages, warranting closer research into its contribution to Down syndrome pathology and new rationales for therapeutic approaches.

Using yeast 2-hybrid and immunoprecipitation analyses, Shimojo (2008) showed that human RILP (PRICKLE1; 608500) and huntingtin interacted directly with dynactin-1 (DCTN1; 601143) to form a triplex. REST bound to the triplex through direct interaction with RILP, forming a quaternary complex involved in nuclear translocation of REST in nonneuronal cells. In neuronal cells, the complex also contained HAP1 (600947), which affected interaction of disease-causing mutant huntingtin, but not wildtype huntingtin, with dynactin-1 and RILP. Overexpression and knockout analyses demonstrated that the presence of HAP1 in the complex prevented nuclear translocation of REST and thereby regulated REST activity.

At the point of mitotic exit within the vertebrate nervous system, when cells lose multipotency and begin to develop stable connections that will persist over life, a switch in ATP-dependent chromatin-remodeling mechanisms occurs. This switch involves the exchange of the BAF53A and BAF45A (PHF10; 613069) subunits within Swi/Snf-like neural progenitor-specific BAF (npBAF) complexes for the homologous BAF53B (ACTL6B; 612458) and BAF45B (DPF1; 601670) subunits within neuron-specific BAF (nBAF) complexes in postmitotic neurons. The subunits of the npBAF complex are essential for neural progenitor proliferation, and mice with reduced dosage for the genes encoding its subunits have defects in neural tube closure similar to those in human spina bifida. In contrast, BAF53B and the nBAF complex are essential for an evolutionarily conserved program of postmitotic neural development and dendritic morphogenesis. Yoo et al. (2009) showed that this essential transition is mediated by repression of BAF53A by miR9* (an miRNA processed from the opposite arm of the miR9 (611186) stem-loop precursor) and miR124 (609327). They found that BAF53a repression is mediated by sequences in the 3-prime untranslated region corresponding to the recognition sites for miR9* and miR124, which are selectively expressed in postmitotic neurons. Mutation of these sites led to persistent expression of BAF53A and defective activity-dependent dendritic outgrowth in neurons. In addition, overexpression of miR9* and miR124 in neural progenitors caused reduced proliferation. miR9* and miR124 are repressed by REST. Yoo et al. (2009) showed that expression of REST in postmitotic neurons led to derepression of BAF53A, indicating that REST-mediated repression of microRNAs directs the essential switch of chromatin regulatory complexes.

Loe-Mie et al. (2010) showed that an SWI/SNF-centered network including the Smarca2 gene (600014) was modified by the downregulation of REST/NRSF in a mouse neuronal cell line. REST/NRSF downregulation also modified the levels of Smarce1 (603111), Smarcd3 (601737), and SWI/SNF interactors (Hdac1, 601241; RcoR, 607675; and Mecp2, 300005). Smarca2 downregulation generated an abnormal dendritic spine morphology that was an intermediate phenotype of schizophrenia (see 181500). The authors noted that 8 genomewide-supported schizophrenia-associated genes (SMARCA2; CSF2RA, 306250; HIST1H2BJ, 615044; NOTCH4, 164951; NRGN, 602350; SHOX, 312865; TCF4, 602272; and ZNF804A, 612282) are part of an interacting network; 5 of the 8, including SMARCA2, encode transcription regulators, and 3 (TCF4, SMARCA2, and CSF2RA) were modified at the level of expression when the REST/NRSF-SWI/SNF chromatin remodeling complex was experimentally manipulated in mouse cell lines and in transgenic mouse models. REST/NRSF-SWI/SNF deregulation also resulted in the differential expression of genes that are clustered in chromosomes, suggesting the induction of genomewide epigenetic changes. Loe-Mie et al. (2010) concluded that the SWI/SNF chromatin remodeling complex is a key component of the genetic architecture of schizophrenia.

Yang et al. (2012) demonstrated that ZNF335 (610827) acts upstream of REST and regulates its expression.

Das et al. (2013) found that knockdown of REST resulted in a decline in medulloblastoma cell proliferation and accumulation of p27 (CDKN1B; 600778). In vitro analysis showed that REST and p27 expression were reciprocally correlated in human medulloblastoma samples. REST repressed expression of USP37 (620226), and USP37 expression promoted p27 deubiquitination. USP37 interacted with p27 to promote its deubiquitination and stabilization, thereby blocking cell proliferation. The authors concluded that REST regulates p27 stability and cell proliferation by controlling USP37.

Lu et al. (2014) demonstrated that induction of REST is a universal feature of normal aging in human cortical and hippocampal neurons. REST is lost, however, in mild cognitive impairment and Alzheimer disease (AD; 104300). Chromatin immunoprecipitation with deep sequencing and expression analysis showed that REST represses genes that promote cell death and AD pathology, and induces the expression of stress response genes. Moreover, REST potently protects neurons from oxidative stress and beta-amyloid (see 104760) protein toxicity, and conditional deletion of Rest in the mouse brain leads to age-related neurodegeneration. A functional ortholog of REST, C. elegans Spr4, also protects against oxidative stress and beta-amyloid protein toxicity. During normal aging, REST is induced in part by cell-nonautonomous Wnt signaling. However, in Alzheimer disease, frontotemporal dementia (FTD; 600274), and dementia with Lewy bodies (DLB; 127750), REST is lost from the nucleus and appears in autophagosomes together with pathologic misfolded proteins. Finally, REST levels during aging are closely correlated with cognitive preservation and longevity. Lu et al. (2014) therefore concluded that the activation state of REST may distinguish neuroprotection from neurodegeneration in the aging brain.

Zullo et al. (2019) showed that extended longevity in humans is associated with a distinct transcriptome signature in the cerebral cortex that is characterized by downregulation of genes related to neural excitation and synaptic function. In C. elegans, neural excitation increases with age, and inhibition of excitation globally, or in glutamatergic or cholinergic neurons, increases longevity. Furthermore, longevity is dynamically regulated by the excitatory-inhibitory balance of neural circuits. The transcription factor REST is upregulated in humans with extended longevity and represses excitation-related genes. Notably, Rest-deficient mice exhibit increased cortical activity and neuronal excitability during aging. Similarly, loss-of-function mutations in the C. elegans REST ortholog genes spr3 and spr4 elevate neural excitation and reduce the lifespan of long-lived daf2 mutants. In wildtype worms, overexpression of spr4 suppresses excitation and extends lifespan. REST, spr3, spr4, and reduced excitation activated the longevity-associated transcription factors FOXO1 (136533) and daf16 in mammals and worms, respectively. Zullo et al. (2019) concluded that their findings revealed a conserved mechanism of aging that is mediated by neural circuit activity and regulated by REST.


Gene Structure

Bayram et al. (2017) stated that the REST gene has 4 exons.


Mapping

Bayram et al. (2017) stated that the REST gene maps to chromosome 4q12.


Molecular Genetics

Wilms Tumor 6

Mahamdallie et al. (2015) identified 11 different REST mutations (see, e.g., 600571.0001-600571.0003) in 16 individuals from 4 families and 9 nonfamilial Wilms tumor (WT6; 616806) pedigrees. Ten of the 11 different mutations, including all of the nonsynonymous mutations, clustered in the DNA binding domain of REST. In 4 cases for whom parental DNA was available, 1 mutation had occurred de novo and 3 had been inherited, confirming incomplete penetrance. None was present in ICR1000 exome series of 993 or in the 61,312 individuals in the ExAC browser. All tested variants showed abrogation of REST function. Mahamdallie et al. (2015) concluded that their data established REST as a Wilms tumor predisposition gene accounting for approximately 2% of Wilms tumors, and recommended screening of REST in all familial cases.

Gingival Fibromatosis 5

In a 11 patients from 3 unrelated Turkish families with gingival fibromatosis-5 (GINGF5; 617626), Bayram et al. (2017) identified 3 different heterozygous truncating mutations in the REST gene (600571.0004-600571.0006). The mutations, which were found by exome sequencing and confirmed by Sanger sequencing, segregated with the disorder in 2 families; the mutation in the proband of the third family occurred de novo. A mildly affected father in 1 of the families was mosaic for the mutation. All mutations occurred in the final exon of the gene, which may result in escape from nonsense-mediated mRNA decay, suggesting that the alleles may act through a dominant-negative or gain-of-function effect. Bayram et al. (2017) noted that studies have suggested that the disorder results from excessive accumulation of extracellular matrix components, particularly collagen type I, which may be due to abnormal expression of TGF-beta (TGFB1; 190180) and IL6 (147620). The mutant transcripts may reduce the repressor function of REST on the collagen synthesis pathway, resulting in the accumulation of collagen in gingiva. However, functional studies of the REST variants and studies of patient cells were not performed.

Autosomal Dominant Deafness 27

In a 3-generation family with autosomal dominant hearing loss mapping to chromosome 4q12-q13.1 (DFNA27; 612431), Nakano et al. (2018) identified heterozygosity for an intronic variant in the REST gene (600571.0007) that segregated fully with deafness in the family and was not found in public variant databases. Functional analysis revealed that the DFNA27-associated REST variant causes gain or loss of function depending on the cellular context: in cells lacking the splicing factor SRRM4 (613103), it causes REST inactivation (loss of function), whereas in cells expressing SRRM4, such as neurons and mechanosensory hair cells of the ear, the variant prevents REST inactivation by alternative splicing (gain of function), thus abrogating its normal downregulation in differentiating neurons.

In a South African Xhosa mother and son with progressive prelingual sensorineural hearing loss, Manyisa et al. (2021) identified heterozygosity for a missense mutation in the REST gene (C415S; 600571.0008) that segregated with disease in the family and was not found in controls or in public variant databases. Functional analysis revealed perturbation of cellular localization and loss of function with the mutant compared to wildtype REST.


Animal Model

Chen et al. (1998) disrupted the Rest gene in mice by gene targeting in mouse embryonic stem cells. As a result, derepression of neuron-specific tubulin (602529) in a subset of nonneural tissues resulted and embryonic lethality ensued.

Lepagnol-Bestel et al. (2009) used the transgenic 152F7 mouse model of Down syndrome (190685) to show that the DYRK1A (600855) gene dosage imbalance deregulated chromosomal clusters of genes located near REST/NRSF binding sites. Dyrk1a bound the SWI/SNF complex (see 603111), which is known to interact with REST/NRSF. Mutation of a REST/NRSF binding site in the promoter of the REST/NRSF target gene L1cam (308840) modified the transcriptional effect of Dyrk1a-dosage imbalance on L1cam. Dyrk1a dosage imbalance perturbed Rest/Nrsf levels with decreased Rest/Nrsf expression in embryonic neurons and increased expression in adult neurons. In transgenic embryonic brain subregions, the authors identified a coordinated deregulation of multiple genes that responsible for dendritic growth impairment. Similarly, Dyrk1a overexpression in primary mouse cortical neurons induced severe reduction of the dendritic growth and dendritic complexity. Lepagnol-Bestel et al. (2009) proposed that both the DYRK1A overexpression-related neuronal gene deregulation (via disturbance of REST/NRSF levels) and the REST/NRSF-SWI/SNF chromatin remodeling complex significantly contribute to the neural phenotypic changes that characterize Down syndrome.

Nakano et al. (2018) found that heterozygous deletion of exon 4 of mouse Rest activated the apoptotic pathway and caused hair cell death in a cell-autonomous manner, resulting in balance defects and deafness. Single-cell quantitative RT-PCR revealed that alternative splicing of Rest exon 4 regulated gene expression specifically in hair cells. As a result, lack of exon 4-dependent alternative splicing in mutant mice reduced expression of many hearing-related genes, especially genes critical for development and function of hair cells, causing defects in cilia of utricles. In vitro studies demonstrated that HDAC inhibitors increased expression of many Rest target genes and prevented degeneration of outer hair cells of mutant mice in organ cultures. The HDAC inhibitor SAHA (Vorinostat) rescued hair cells and hearing of Rest mutant mice in vivo.


ALLELIC VARIANTS 8 Selected Examples):

.0001   WILMS TUMOR 6, SUSCEPTIBILITY TO

REST, 2-BP DEL, 831AT
SNP: rs869025310, ClinVar: RCV000207462, RCV001230375

In 2 sisters (FAM0482) and an unrelated individual (FAM00250) with Wilms tumor (WT6; 616806), Mahamdallie et al. (2015) identified a heterozygous 2-bp deletion (c.831_832delAT, ENST00000309042) in the DNA binding domain of the REST gene resulting in frameshift. The mutation was inherited from the mother by the sisters and from the father by the unrelated individual. Age at diagnosis was 3.7 and 6.0 years in the sisters, and 3.2 years in the unrelated individual. Histology of the sisters' tumors showed triphasic (blastemal, epithelial, and stromal) elements. Mahamdallie et al. (2015) noted that this mutation was not identified in the ICR1000 UK exome series or in the ExAC browser.


.0002   WILMS TUMOR 6, SUSCEPTIBILITY TO

REST, 4-BP DEL, 772GTGA
SNP: rs869025311, ClinVar: RCV000207468

In 2 first cousins once removed (FAM0509) with Wilms tumor (WT6; 616806), Mahamdallie et al. (2015) identified a heterozygous 4-bp deletion (c.772_775delGTGA, ENST00000309042) in the DNA binding domain of the REST gene. In one cousin the mutation was inherited from the mother, age at diagnosis was 2.6 years, and the tumor sample showed triphasic histology. In the other cousin the mutation was inherited from the father, age at diagnosis was 0.8 years, and tumor histology was predominantly blastemal. Mahamdallie et al. (2015) noted that this mutation was not identified in the ICR1000 UK exome series or in the ExAC browser.


.0003   WILMS TUMOR 6, SUSCEPTIBILITY TO

REST, HIS322ARG
SNP: rs869025312, ClinVar: RCV000207458

In 2 cousins (FAM0481) and in an unrelated individual (FAM1324) with Wilms tumor (WT6; 616806), Mahamdallie et al. (2015) detected a heterozygous c.965A-G transition (c.965A-G, ENST00000309042) in the zinc finger DNA-binding domain of the REST gene, resulting in a his322-to-arg (H322R) substitution. The mutation was paternally inherited in the female cousin and maternally inherited in the male cousin. Age of onset was 3 years in female patient and 0.5 years in the male patient. Inheritance was unknown in the unrelated individual; Wilms tumor developed at 0.5 years of age and was of triphasic histology. Functional studies showed that this variant was unable to repress REST target gene expression, supporting pathogenicity. Mahamdallie et al. (2015) noted that this mutation was not identified in the ICR1000 UK exome series or in the ExAC browser.


.0004   FIBROMATOSIS, GINGIVAL, 5

REST, 2-BP DEL, 2865AA ({dbSNP SCV000579327})
SNP: rs1553904481, ClinVar: RCV000497680, RCV000516014, RCV001851328

In 2 Turkish brothers with gingival fibromatosis-5 (GINGF5; 617626), Bayram et al. (2017) identified a heterozygous 2-bp deletion (c.2865_2866delAA, NM_005612.4) in the last exon (exon 4) of the REST gene, resulting in a frameshift and premature termination (Asn958SerfsTer9). The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the dbSNP, 1000 Genomes Project, Exome Sequencing Project, or ExAC databases, or in an in-house database of more than 6,500 exomes. The father, who had a mild form of the disorder, was mosaic for the mutation. Functional studies of the variant and studies of patient cells were not performed.


.0005   FIBROMATOSIS, GINGIVAL, 5

REST, LEU437TER ({dbSNP SCV000579328})
SNP: rs1553904077, ClinVar: RCV000498225, RCV000516150

In 5 members of a Turkish family with gingival fibromatosis-5 (GINGF5; 617626), Bayram et al. (2017) identified a heterozygous c.1310T-A transversion (c.1310T-A, NM_005612.4) in the last exon (exon 4) of the REST gene, resulting in a leu437-to-ter (L437X) substitution. The mutation, which was found by whole-exome sequencing and confirmed by Sanger sequencing, was not found in the dbSNP, 1000 Genomes Project, Exome Sequencing Project, or ExAC databases, or in an in-house database of more than 6,500 exomes. The family was originally reported by Pehlivan et al. (2009). Functional studies of the variant and studies of patient cells were not performed.


.0006   FIBROMATOSIS, GINGIVAL, 5

REST, 1-BP DEL, 2413C ({dbSNP SCV000579329})
SNP: rs1553904346, ClinVar: RCV000498949, RCV000515908

In a 9-year-old Turkish girl with gingival fibromatosis-5 (GINGF5; 617626), Bayram et al. (2017) identified a de novo heterozygous 1-bp deletion (c.2413delC, NM_005612.4) in the last exon (exon 4) of the REST gene, resulting in a frameshift and premature termination (Leu805PhefsTer38). The variant, which was found by exome sequencing and confirmed by Sanger sequencing, was not found in the dbSNP, 1000 Genomes Project, Exome Sequencing Project, or ExAC databases, or in an in-house database of more than 6,500 exomes. The patient was identified through the GeneMatcher database. Functional studies of the variant and studies of patient cells were not performed.


.0007   DEAFNESS, AUTOSOMAL DOMINANT 27

REST, IVS3AS, C-G, -21
SNP: rs1720770872, ClinVar: RCV000855719

In a 3-generation family (LMG2) with autosomal dominant hearing loss (DFNA27; 612431), Nakano et al. (2018) identified heterozygosity for a C-to-G transversion (chr4.56,927,594C-G, GRCh38) at a conserved nucleotide in intron 3, 21 bp upstream of exon 4a/b of the REST gene. The mutation segregated fully with deafness in the family and was not found in 400 control DNA samples or in the gnomAD database. RT-PCR in patient and control blood cells showed that the C-G variant causes truncation of the coding region of REST by generating a novel splice acceptor site for exon 4a. The authors noted that splicing of exon 4 into REST mRNA is normally tissue-specific and requires SRRM4 (613103), which is selectively expressed in neurons and mechanosensory hair cells of the ear. Using minigenes transfected into HEK293 cells, they observed that DFNA27-associated aberrant splicing results in a novel splice form containing a 24-nucleotide variant of exon 4b, and splicing of this variant exon 4b requires both the C-G variant-dependent relocation of the splice acceptor site as well as an SRRM4-dependent shift in the splice donor site. Analysis of luciferase activity of isoforms generated in the presence or absence of SRRM4 suggested that the C-G DFNA27-associated variant has opposing effects on REST depending on whether SRRM4 is present. In cells that do not express SRRM4, the C-G variant aberrantly inactivates REST by creating a novel acceptor site for constitutive splicing upstream of exon 4a/b. Conversely, in cells that do express SRRM4, the C-G variant aberrantly results in production of active REST that cannot be inactivated by SRRM4-directed alternative splicing of exon 4, thus abrogating the normal downregulation of REST that occurs in differentiating neurons.


.0008   DEAFNESS, AUTOSOMAL DOMINANT 27

REST, CYS415SER
SNP: rs2109573013, ClinVar: RCV002248339

In a South African Xhosa mother and son with progressive prelingual sensorineural hearing loss (DFNA27; 612431), Manyisa et al. (2021) identified heterozygosity for a c.1244G-C transversion (c.1244G-C, NM_005612.5) in exon 4 of the REST gene, resulting in a cys415-to-ser (C415S) substitution at a highly conserved residue. The mutation was not found in the proband's unaffected half-brother or unaffected maternal grandmother, in 103 black South African controls or 52 sporadic South African probands of black or mixed ancestry with nonsyndromic hearing impairment, or in the gnomAD, UK10K, Greater Middle East Variome (GME), or dbSNP databases. Experiments using GFP tagging in HEK293 cells showed that the wildtype REST protein is located exclusively within the nucleus, whereas the mutant showed localization throughout the cell, indicating loss of exclusive nuclear shuttling/localization. In addition, wildtype REST competently repressed transcription of the known REST target AF1Q (604684) in transiently transfected HEK293 cells, whereas transcriptional repression was lost in cells expressing the mutant REST protein.


REFERENCES

  1. Abderrahmani, A., Steinmann, M., Plaisance, V., Niederhauser, G., Haefliger, J.-A., Mooser, V., Bonny, C., Nicod, P., Waeber, G. The transcriptional repressor REST determines the cell-specific expression of the human MAPK8IP1 gene encoding IB1 (JIP-1). Molec. Cell. Biol. 21: 7256-7627, 2001. [PubMed: 11585908] [Full Text: https://doi.org/10.1128/MCB.21.21.7256-7267.2001]

  2. Bahn, S., Mimmack, M., Ryan, M., Caldwell, M. A., Jauniaux, E., Starkey, M., Svendsen, C. N., Emson, P. Neuronal target genes of the neuron-restrictive silencer factor in neurospheres derived from fetuses with Down's syndrome: a gene expression study. Lancet 359: 310-315, 2002. [PubMed: 11830198] [Full Text: https://doi.org/10.1016/S0140-6736(02)07497-4]

  3. Bayram, Y., White, J. J., Elcioglu, N., Cho, M. T., Zadeh, N., Gedikbasi, A., Palanduz, S., Ozturk, S., Cefle, K., Kasapcopur, O., Akdemir, Z. C., Pehlivan, D., and 11 others. REST final-exon-truncating mutations cause hereditary gingival fibromatosis. Am. J. Hum. Genet. 101: 149-156, 2017. [PubMed: 28686854] [Full Text: https://doi.org/10.1016/j.ajhg.2017.06.006]

  4. Canzonetta, C., Mulligan, C., Deutsch, S., Ruf, S., O'Doherty, A., Lyle, R., Borel, C., Lin-Marq, N., Delom, F., Groet, J., Schnappauf, F., De Vita, S, and 12 others. DYRK1A-dosage imbalance perturbs NRSF/REST levels, deregulating pluripotency and embryonic stem cell fate in Down syndrome. Am. J. Hum. Genet. 83: 388-400, 2008. [PubMed: 18771760] [Full Text: https://doi.org/10.1016/j.ajhg.2008.08.012]

  5. Chen, Z.-F., Paquette, A. J., Anderson, D. J. NRSF/REST is required in vivo for repression of multiple neuronal target genes during embryogenesis. Nature Genet. 20: 136-142, 1998. [PubMed: 9771705] [Full Text: https://doi.org/10.1038/2431]

  6. Cheong, A., Bingham, A. J., Li, J., Kumar, B., Sukumar, P., Munsch, C., Buckley, N. J., Neylon, C. B., Porter, K. E., Beech, D. J., Wood, I. C. Downregulated REST transcription factor is a switch enabling critical potassium channel expression and cell proliferation. Molec. Cell 20: 45-52, 2005. [PubMed: 16209944] [Full Text: https://doi.org/10.1016/j.molcel.2005.08.030]

  7. Chong, J. A., Tapia-Ramirez, J., Kim, S., Toledo-Aral, J. J., Zheng, Y., Boutros, M. C., Altshuller, Y. M., Frohman, M. A., Kraner, S. D., Mandel, G. REST: a mammalian silencer protein that restricts sodium channel gene expression to neurons. Cell 80: 949-957, 1995. [PubMed: 7697725] [Full Text: https://doi.org/10.1016/0092-8674(95)90298-8]

  8. Das, C. M., Taylor, P., Gireud, M., Singh, A., Lee, D., Fuller, G., Ji, L., Fangusaro, J., Rajaram, V., Goldman, S., Eberhart, C., Gopalakrishnan, V. The deubiquitylase USP37 links REST to the control of p27 stability and cell proliferation. Oncogene 32: 1691-1701, 2013. Note: Erratum: Oncogene 35: 6153-6154, 2016. [PubMed: 22665064] [Full Text: https://doi.org/10.1038/onc.2012.182]

  9. Ding, N., Zhou, H., Esteve, P.-O., Chin, H. G., Kim, S., Xu, X., Joseph, S. M., Friez, M. J., Schwartz, C. E., Pradhan, S., Boyer, T. G. Mediator links epigenetic silencing of neuronal gene expression with X-linked mental retardation. Molec. Cell 31: 347-359, 2008. [PubMed: 18691967] [Full Text: https://doi.org/10.1016/j.molcel.2008.05.023]

  10. Guardavaccaro, D., Frescas, D., Dorrello, N. V., Peschiaroli, A., Multani, A. S., Cardozo, T., Lasorella, A., Iavarone, A., Chang, S., Hernando, E., Pagano, M. Control of chromosome stability by the beta-TrCP-REST-Mad2 axis. Nature 452: 365-369, 2008. [PubMed: 18354482] [Full Text: https://doi.org/10.1038/nature06641]

  11. Kemp, D. M., Lin, J. C., Habener, J. F. Regulation of Pax4 paired homeodomain gene by neuron-restrictive silencer factor. J. Biol. Chem. 278: 35057-35062, 2003. [PubMed: 12829700] [Full Text: https://doi.org/10.1074/jbc.M305891200]

  12. Kuwahara, K., Saito, Y., Takano, M., Arai, Y., Yasuno, S., Nakagawa, Y., Takahashi, N., Adachi, Y., Takemura, G., Horie, M., Miyamoto, Y., Morisaki, T., and 12 others. NRSF regulates the fetal cardiac gene program and maintains normal cardiac structure and function. EMBO J. 22: 6310-6321, 2003. [PubMed: 14633990] [Full Text: https://doi.org/10.1093/emboj/cdg601]

  13. Lepagnol-Bestel, A.-M., Zvara, A., Maussion, G., Quignon, F., Ngimbous, B., Ramoz, N., Imbeaud, S., Loe-Mie, Y., Benihoud, K., Agier, N., Salin, P. A., Cardona, A., and 11 others. DYRK1A interacts with the REST/NRSF-SWI/SNF chromatin remodelling complex to deregulate gene clusters involved in the neuronal phenotypic traits of Down syndrome. Hum. Molec. Genet. 18: 1405-1414, 2009. Note: Erratum: Hum. Molec. Genet. 31: 2106-2107, 2022. [PubMed: 19218269] [Full Text: https://doi.org/10.1093/hmg/ddp047]

  14. Loe-Mie, Y., Lepagnol-Bestel, A.-M., Maussion, G., Doron-Faigenboim, A., Imbeaud, S., Delacroix, H., Aggerbeck, L., Pupko, T., Gorwood, P., Simonneau, M., Moalic, J.-M. SMARCA2 and other genome-wide supported schizophrenia-associated genes: regulation by REST/NRSF, network organization and primate-specific evolution. Hum. Molec. Genet. 19: 2841-2857, 2010. [PubMed: 20457675] [Full Text: https://doi.org/10.1093/hmg/ddq184]

  15. Lu, T., Aron, L., Zullo, J., Pan, Y., Kim, H., Chen, Y., Yang, T.-H., Kim, H.-M., Drake, D., Liu, X. S., Bennett, D. A., Colaiacovo, M. P., Yankner, B. A. REST and stress resistance in ageing and Alzheimer's disease. Nature 507: 448-454, 2014. Note: Erratum: Nature 540: 470 only, 2016. [PubMed: 24670762] [Full Text: https://doi.org/10.1038/nature13163]

  16. Lunyak, V. V., Burgess, R., Prefontaine, G. G., Nelson, C., Sze, S.-H., Chenoweth, J., Schwartz, P., Pevzner, P. A., Glass, C., Mandel, G., Rosenfeld, M. G. Corepressor-dependent silencing of chromosomal regions encoding neuronal genes. Science 298: 1747-1752, 2002. Note: Erratum: Science 299: 1663 only, 2003. [PubMed: 12399542] [Full Text: https://doi.org/10.1126/science.1076469]

  17. Mahamdallie, S. S., Hanks, S., Karlin, K. L., Zachariou, A., Perdeaux, E. R., Ruark, E., Shaw, C. A., Renwick, A., Ramsay, E., Yost, S., Elliott, A., Birch, J., and 13 others. Mutations in the transcriptional repressor REST predispose to Wilms tumor. Nature Genet. 47: 1471-1474, 2015. Note: Erratum: Nature Genet. 48: 473 only, 2016. [PubMed: 26551668] [Full Text: https://doi.org/10.1038/ng.3440]

  18. Manyisa, N., Schrauwen, I., Alves de Souza Rios, L., Mowla, S., Tekendo-Ngongang, C., Popel, K., Esoh, K., Bharadwaj, T., Nouel-Saied, L. M., Acharya, A., Nasir, A., Wonkam-Tingang, E., de Kock, C., Dandara C., Leal, S. M., Wonkam, A. A monoallelic variant in REST is associated with non-syndromic autosomal dominant hearing impairment in a South African family. Genes 12: 1765, 2021. [PubMed: 34828371] [Full Text: https://doi.org/10.3390/genes12111765]

  19. Nakano, Y., Kelly, M. C., Rehman, A. U., Boger, E. T., Morell, R. J., Kelley, M. W., Friedman, T. B., Banfi, B. Defects in the alternative splicing-dependent regulation of REST cause deafness. Cell 174: 536-548, 2018. [PubMed: 29961578] [Full Text: https://doi.org/10.1016/j.cell.2018.06.004]

  20. Ooi, L., Wood, I. C. Chromatin crosstalk in development and disease: lessons from REST. Nature Rev. Genet. 8: 544-554, 2007. [PubMed: 17572692] [Full Text: https://doi.org/10.1038/nrg2100]

  21. Palm, K., Metsis, M., Timmusk, T. Neuron-specific splicing of zinc finger transcription factor REST/NRSF/XBR is frequent in neuroblastomas and conserved in human, mouse and rat. Molec. Brain Res. 72: 30-39, 1999. [PubMed: 10521596] [Full Text: https://doi.org/10.1016/s0169-328x(99)00196-5]

  22. Pehlivan, D., Abe, S., Ozturk, S., Kayhan, K. B., Gunduz, E., Cefle, K., Bayrak, A., Ark, N., Gunduz, M., Palanduz, S. Cytogenetic analysis and examination of SOS1 gene mutation in a Turkish family with hereditary gingival fibromatosis. J. Hard Tissue Biol. 18: 131-134, 2009.

  23. Plaisance, V., Niederhauser, G., Azzouz, F., Lenain, V., Haefliger, J.-A., Waeber, G., Abderrahmani, A. The repressor element silencing transcription factor (REST)-mediated transcriptional repression requires the inhibition of Sp1. J. Biol. Chem. 280: 401-407, 2005. [PubMed: 15528196] [Full Text: https://doi.org/10.1074/jbc.M411825200]

  24. Schoenherr, C. J., Anderson, D. J. The neuron-restrictive silencer factor (NRSF): a coordinate repressor of multiple neuron-specific genes. Science 267: 1360-1363, 1995. [PubMed: 7871435] [Full Text: https://doi.org/10.1126/science.7871435]

  25. Shimojo, M. Huntingtin regulates RE1-silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) nuclear trafficking indirectly through a complex with REST/NRSF-interacting LIM domain protein (RILP) and dynactin p150-Glued. J. Biol. Chem. 283: 34880-34886, 2008. [PubMed: 18922795] [Full Text: https://doi.org/10.1074/jbc.M804183200]

  26. Singh, S. K., Kagalwala, M. N., Parker-Thornburg, J., Adams, H., Majumder, S. REST maintains self-renewal and pluripotency of embryonic stem cells. Nature 453: 223-227, 2008. [PubMed: 18362916] [Full Text: https://doi.org/10.1038/nature06863]

  27. Tahiliani, M., Mei, P., Fang, R., Leonor, T., Rutenberg, M., Shimizu, F., Li, J., Rao, A., Shi, Y. The histone H3K4 demethylase SMCX links REST target genes to X-linked mental retardation. Nature 447: 601-605, 2007. [PubMed: 17468742] [Full Text: https://doi.org/10.1038/nature05823]

  28. Thiel, G., Lietz, M., Cramer, M. Biological activity and modulator structure of RE-1-silencing transcription factor (REST), a repressor of neuronal genes. J. Biol. Chem. 273: 26891-26899, 1998. [PubMed: 9756936] [Full Text: https://doi.org/10.1074/jbc.273.41.26891]

  29. Westbrook, T. F., Hu, G., Ang, X. L., Mulligan, P., Pavlova, N. N., Liang, A., Leng, Y., Maehr, R., Shi, Y., Harper, J. W., Elledge, S. J. SCF-(beta-TRCP) controls oncogenic transformation and neural differentiation through REST degradation. Nature 452: 370-374, 2008. [PubMed: 18354483] [Full Text: https://doi.org/10.1038/nature06780]

  30. Yang, Y. J., Baltus, A. E., Mathew, R. S., Murphy, E. A., Evrony, G. D., Gonzalez, D. M., Wang, E. P., Marshall-Walker, C. A., Barry, B. J., Murn, J., Tatarakis, A., Mahajan, M. A., Samuels, H. H., Shi, Y., Golden, J. A., Mahajnah, M., Shenhav, R., Walsh, C. A. Microcephaly gene links trithorax and REST/NRSF to control neural stem cell proliferation and differentiation. Cell 151: 1097-1112, 2012. [PubMed: 23178126] [Full Text: https://doi.org/10.1016/j.cell.2012.10.043]

  31. Yeo, M., Lee, S.-K., Lee, B., Ruiz, E. C., Pfaff, S. L., Gill, G. N. Small CTD phosphatases function in silencing neuronal gene expression. Science 307: 596-600, 2005. [PubMed: 15681389] [Full Text: https://doi.org/10.1126/science.1100801]

  32. Yoo, A. S., Staahl, B. T., Chen, L., Crabtree, G. R. MicroRNA-mediated switching of chromatin-remodelling complexes in neural development. Nature 460: 642-646, 2009. Note: Erratum: Nature 461: 296 only, 2009. [PubMed: 19561591] [Full Text: https://doi.org/10.1038/nature08139]

  33. Zuccato, C., Ciammola, A., Rigamonti, D., Leavitt, B. R., Goffredo, D., Conti, L., MacDonald, M. E., Friedlander, R. M., Silani, V., Hayden, M. R., Timmusk, T., Sipione, S., Cattaneo, E. Loss of huntingtin-mediated BDNF gene transcription in Huntington's disease. Science 293: 493-498, 2001. [PubMed: 11408619] [Full Text: https://doi.org/10.1126/science.1059581]

  34. Zuccato, C., Tartari, M., Crotti, A., Goffredo, D., Valenza, M., Conti, L., Cataudella, T., Leavitt, B. R., Hayden, M. R., Timmusk, T., Rigamonti, D., Cattaneo, E. Huntingtin interacts with REST/NRSF to modulate the transcription of NRSE-controlled neuronal genes. Nature Genet. 35: 76-83, 2003. [PubMed: 12881722] [Full Text: https://doi.org/10.1038/ng1219]

  35. Zullo, J. M., Drake, D., Aron, L., O'Hern, P., Dhamne, S. C., Davidsohn, N., Mao, C.-A., Klein, W. H., Rotenberg, A., Bennett, D. A., Church, G. M., Colaiacovo, M. P., Yankner, B. A. Regulation of lifespan by neural excitation and REST. Nature 574: 359-364, 2019. [PubMed: 31619788] [Full Text: https://doi.org/10.1038/s41586-019-1647-8]


Contributors:
Bao Lige - updated : 01/31/2023
Marla J. F. O'Neill - updated : 04/22/2022
Ada Hamosh - updated : 04/09/2020
Bao Lige - updated : 01/09/2020
Marla J. F. O'Neill - updated : 11/12/2019
Bao Lige - updated : 09/23/2019
Cassandra L. Kniffin - updated : 08/14/2017
Ada Hamosh - updated : 02/11/2016
Ada Hamosh - updated : 4/11/2014
George E. Tiller - updated : 8/27/2013
Cassandra L. Kniffin - updated : 2/25/2013
Patricia A. Hartz - updated : 4/13/2012
George E. Tiller - updated : 11/30/2009
Ada Hamosh - updated : 8/27/2009
Patricia A. Hartz - updated : 8/20/2009
Ada Hamosh - updated : 11/5/2008
Ada Hamosh - updated : 6/12/2008
Ada Hamosh - updated : 5/22/2008
Patricia A. Hartz - updated : 9/25/2007
Ada Hamosh - updated : 6/15/2007
Patricia A. Hartz - updated : 12/12/2005
Ada Hamosh - updated : 3/1/2005
Victor A. McKusick - updated : 8/15/2003
Cassandra L. Kniffin - updated : 6/23/2003
Ada Hamosh - updated : 4/3/2003
Patricia A. Hartz - updated : 6/13/2002
Victor A. McKusick - updated : 9/28/1998

Creation Date:
Alan F. Scott : 6/1/1995

Edit History:
carol : 06/08/2023
mgross : 01/31/2023
alopez : 07/27/2022
carol : 04/22/2022
carol : 02/15/2022
carol : 07/17/2020
carol : 06/23/2020
alopez : 04/09/2020
mgross : 01/09/2020
carol : 11/13/2019
alopez : 11/12/2019
alopez : 11/12/2019
carol : 10/08/2019
mgross : 09/23/2019
carol : 08/24/2017
carol : 08/24/2017
carol : 08/15/2017
ckniffin : 08/14/2017
alopez : 02/11/2016
alopez : 4/11/2014
carol : 8/27/2013
tpirozzi : 8/27/2013
tpirozzi : 8/27/2013
carol : 2/25/2013
ckniffin : 2/25/2013
mgross : 5/18/2012
mgross : 5/18/2012
terry : 4/13/2012
wwang : 1/7/2010
terry : 11/30/2009
mgross : 10/5/2009
carol : 9/23/2009
carol : 9/15/2009
alopez : 9/4/2009
terry : 8/27/2009
mgross : 8/24/2009
terry : 8/20/2009
carol : 6/3/2009
alopez : 12/1/2008
terry : 11/5/2008
alopez : 6/17/2008
terry : 6/12/2008
alopez : 5/28/2008
alopez : 5/28/2008
terry : 5/22/2008
mgross : 10/2/2007
terry : 9/25/2007
alopez : 6/20/2007
terry : 6/15/2007
wwang : 12/12/2005
wwang : 3/7/2005
terry : 3/1/2005
alopez : 9/2/2003
alopez : 8/19/2003
alopez : 8/19/2003
terry : 8/15/2003
carol : 7/10/2003
carol : 7/9/2003
ckniffin : 6/23/2003
alopez : 4/3/2003
terry : 4/3/2003
carol : 6/20/2002
terry : 6/13/2002
alopez : 9/28/1998
joanna : 9/28/1998
mark : 3/6/1998
joanna : 12/6/1995
mark : 6/1/1995