ELP4 Human

Elongator Acetyltransferase Complex Subunit 4 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Function and Biological Role

ELP4 is a core subunit of the Elongator complex, a histone acetyltransferase (HAT) integral to RNA polymerase II-mediated transcriptional elongation. Key functions include:

  • Chromatin Remodeling: Acetylation of histones H3 and H4, enhancing DNA accessibility .

  • tRNA Modification: Post-transcriptional wobble uridine modification, critical for translation fidelity .

  • Neuronal Development: Regulates cortical neuron migration and axonal guidance via cytoskeletal gene expression .

The human ELP4 protein partially rescues yeast ELP4 deletion mutants, confirming conserved functional domains .

Genetic and Clinical Associations

ELP4 variants are implicated in neurodevelopmental and epileptic disorders:

Table 1: ELP4 Genetic Associations

ConditionVariant TypeKey FindingsSource
Rolandic EpilepsyIntronic SNPs (e.g., rs986527)Genome-wide linkage (11p13, HLOD 4.30); OR ≈ 2 for centrotemporal sharp waves
Autism Spectrum Disorder (ASD)Copy Number Variations (CNVs)Excess ELP4 CNVs in ASD cases vs. controls (P=2.7×10⁻³)
Language ImpairmentMicrodeletionsCo-occurrence with ASD and intellectual disability

Mechanistic Insights from Research

  • Epilepsy Pathogenesis: Non-coding ELP4 variants disrupt Elongator-mediated expression of neuronal migration genes (e.g., actin regulators), predisposing to cortical hyperexcitability .

  • Cognitive Impairments: Dysregulated ELP4 correlates with glucose metabolism abnormalities in associative cortices and synaptic maturation defects .

  • Functional Redundancy: ELP4 depletion mimics ELP1 mutations in Riley-Day syndrome, suggesting shared pathways in neurodevelopmental disorders .

Expression and Tissue Distribution

ELP4 exhibits brain-specific expression patterns, with high activity in:

  • Cerebral Cortex

  • Hippocampus

  • Cerebellum

Protein Atlas data confirms widespread but variable expression across tissues, with strongest signals in neuronal compartments .

Research Applications of Recombinant ELP4

The ELP4 Human Recombinant protein is utilized for:

  • In vitro assays studying HAT activity and tRNA modification.

  • Antibody production for immunohistochemistry and Western blotting.

  • Functional studies modeling ELP4-associated disorders in cellular systems .

Future Directions

Unresolved questions include:

  • Identification of non-coding regulatory mutations in ELP4-linked epilepsy.

  • Mechanistic links between ELP4 CNVs and ASD penetrance.

  • Therapeutic targeting of Elongator complex dysfunction.

Product Specs

Introduction
Elongator Acetyltransferase Complex Subunit 4, also known as ELP4, is a protein-coding gene. It is a subunit of the RNA polymerase II elongator complex. ELP4 complements phenotypes of yeast ELP4 deletion mutants and is linked to Rolandic epilepsy.
Description
Recombinant human ELP4 protein, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 447 amino acids (1-424 a.a.). It has a molecular mass of 49 kDa. ELP4 is fused to a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile filtered solution.
Formulation
ELP4 protein solution (1 mg/ml) in 20 mM Tris-HCl buffer (pH 8.0) containing 10% glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freezing and thawing.
Purity
Purity greater than 80.0% as determined by SDS-PAGE.
Synonyms
Elongator complex protein 4, ELP4, Elongator Acetyltransferase Complex Subunit 4, hELP4, PAX6 neighbor gene protein, C11orf19, PAXNEB, dJ68P15A.1, PAX6NEB.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAAVATC GSVAASTGSA VATASKSNVT SFQRRGPRAS VTNDSGPRLV SIAGTRPSVR NGQLLVSTGL PALDQLLGGG LAVGTVLLIE EDKYNIYSPL LFKYFLAEGI VNGHTLLVAS AKEDPANILQ ELPAPLLDDK CKKEFDEDVY NHKTPESNIK MKIAWRYQLL PKMEIGPVSS SRFGHYYDAS KRMPQELIEA SNWHGFFLPE KISSTLKVEP CSLTPGYTKL LQFIQNIIYE EGFDGSNPQK KQRNILRIGI QNLGSPLWGD DICCAENGGN SHSLTKFLYV LRGLLRTSLS ACIITMPTHL IQNKAIIARV TTLSDVVVGL ESFIGSERET NPLYKDYHGL IHIRQIPRLN NLICDESDVK DLAFKLKRKL FTIERLHLPP DLSDTVSRSS KMDLAESAKR LGPGCGMMAG GKKHLDF.

Q&A

What is the ELP4 gene and what protein does it encode?

ELP4 (Elongation protein 4 homolog) is a gene that encodes a component of the six-subunit elongator complex, a histone acetyltransferase complex that associates directly with RNA polymerase II during transcriptional elongation . The human ELP4 gene can partially complement sensitivity phenotypes of yeast ELP4 deletion mutants . The gene produces multiple alternatively spliced variants encoding different protein isoforms, though the full-length nature of only one has been completely determined .

What protein complexes is ELP4 associated with and what are their primary functions?

ELP4 functions as a component of the RNA polymerase II elongator complex, a multiprotein complex involved in transcriptional elongation . This complex catalyzes the formation of carboxymethyluridine in the wobble base at position 34 in tRNAs, which is essential for proper translation . ELP4 specifically forms strong functional partnerships with other elongator complex proteins, including ELP3 and ELP6, as evidenced by high interaction scores in protein association databases . Together with ELP5 and ELP6, it forms a sub-complex that is necessary for histone acetyltransferase activity, making DNA more accessible for transcription .

How can researchers distinguish between ELP4 and other elongator complex components when investigating cellular phenotypes?

When investigating cellular phenotypes, researchers should employ a methodological approach that includes both genetic and biochemical techniques. Start with siRNA or CRISPR-based selective knockdown/knockout of ELP4 while maintaining the expression of other elongator components, then perform co-immunoprecipitation experiments to assess complex integrity. Rescue experiments using wild-type versus mutant ELP4 can determine which phenotypes are specifically attributable to ELP4 rather than general elongator complex dysfunction. Additionally, comparative transcriptomic and proteomic analyses between ELP4-deficient cells and those lacking other elongator components can reveal ELP4-specific signatures .

What experimental evidence supports ELP4's role in transcriptional elongation?

The evidence for ELP4's role in transcriptional elongation comes from multiple experimental approaches. In vivo complementation studies using yeast systems demonstrated that human ELP4 partially resumes the slow activation of genes like PHO5 and increases expression of SSA3 under heat shock conditions . This indicates its involvement in facilitating RNA polymerase II activity during transcriptional elongation. Additionally, biochemical studies have shown that ELP4, along with ELP5 and ELP6, forms a sub-complex essential for the histone acetyltransferase (HAT) activity of the elongator complex, which makes DNA more accessible for transcription . The lack of ELP4/5/6 leads to diminished HAT activity, affecting both transcription initiation and elongation through chromatin and acetyl-CoA dependent pathways .

How does ELP4 contribute to tRNA modification, and what techniques are most effective for studying this function?

ELP4, as part of the elongator complex, is crucial for the formation of carboxymethyluridine in the wobble base at position 34 in tRNAs . To effectively study this function, researchers should employ a combination of techniques:

  • Mass spectrometry to directly analyze tRNA modifications in cells with ELP4 manipulation

  • In vitro reconstitution assays using purified elongator complex components to assess enzymatic activity

  • Ribosome profiling in ELP4-deficient cells to identify translation defects resulting from impaired tRNA modification

  • Comparative analysis of codon usage and translation efficiency using reporter systems with different wobble position requirements

These approaches can elucidate how ELP4 contributes to the specificity and efficiency of the tRNA modification process, and how these modifications subsequently affect translation dynamics in different cellular contexts .

What is the relationship between ELP4's function in neurogenesis and its molecular activities?

ELP4's role in neurogenesis appears to be linked to its molecular functions in both transcriptional regulation and tRNA modification. As a component of the elongator complex, ELP4 is involved in regulating the migration and branching of projection neurons in the developing cerebral cortex . The mechanistic relationship likely involves:

  • Transcriptional regulation of genes essential for neuronal development through ELP4's role in elongator complex HAT activity

  • Translation regulation of specific neuronal proteins via ELP4-dependent tRNA modifications

  • Cell-type specific requirements for ELP4 function during different stages of neurogenesis

These molecular activities converge to influence neuronal differentiation, migration, and function, explaining why ELP4 dysfunction is particularly impactful in neurological disorders. The clinical link between ELP4 mutations and Rolandic epilepsy supports this relationship between ELP4's molecular functions and its role in brain development .

What is the evidence linking ELP4 to Rolandic epilepsy, and how can researchers investigate this relationship?

ELP4 was identified as the first gene linked to Rolandic epilepsy (RE), the most common form of human epilepsy, in a study published in February 2009 . Children with RE have been found to carry mutations in the ELP4 gene that affect the Elongator Protein Complex function . To investigate this relationship, researchers should employ a multi-faceted approach:

  • Genetic screening studies to identify additional ELP4 variants in RE patient cohorts

  • Functional characterization of patient-derived mutations using in vitro assays for elongator complex activity

  • Development of animal models expressing human ELP4 mutations to assess epileptogenic mechanisms

  • Electrophysiological studies of neurons derived from patient iPSCs to determine how ELP4 dysfunction affects neuronal excitability

  • Comparative analysis of transcriptomic and translatomic profiles between control and ELP4-mutant neuronal populations

This comprehensive approach would provide mechanistic insights into how ELP4 mutations contribute to the epilepsy phenotype, potentially revealing new therapeutic targets .

How do ELP4 mutations affect cognitive development in patients with Rolandic epilepsy?

Cognitive impairment is a significant concern in Rolandic epilepsy (RE), which develops around age 3 and typically resolves by age 12 . The mechanisms by which ELP4 mutations affect cognitive development likely involve:

  • Disruption of neuronal migration and cortical circuit formation during critical developmental windows

  • Altered gene expression patterns in neurons due to impaired transcriptional elongation

  • Defective protein synthesis resulting from abnormal tRNA modifications

  • Potential compensatory mechanisms that may explain the age-dependent resolution of symptoms

To study these effects, researchers should conduct longitudinal assessments of cognitive function in RE patients with known ELP4 mutations, correlating specific genetic variants with cognitive trajectories. Neuroimaging studies examining structural and functional connectivity, combined with cellular models investigating synaptic plasticity in ELP4-mutant neurons, would provide additional insights into the mechanisms underlying cognitive impairment in these patients .

What methodological approaches should researchers use when investigating potential associations between ELP4 and other neurological disorders?

When investigating potential associations between ELP4 and other neurological disorders beyond Rolandic epilepsy, researchers should employ a systematic methodology:

  • Large-scale genetic association studies using next-generation sequencing to identify ELP4 variants in diverse neurological disorders

  • Case-control studies comparing ELP4 variant frequencies between specific disease cohorts and matched controls

  • Meta-analysis of existing genomic datasets to detect potential associations not apparent in individual studies

  • Functional characterization of identified variants using cellular and animal models

  • Integration of genetic findings with clinical phenotypes using machine learning approaches to identify potential ELP4-related endophenotypes

Additionally, researchers should consider the role of ELP4 in specific neurological processes, such as neuronal migration and protein synthesis, when selecting candidate disorders for investigation. Given ELP4's involvement in fundamental cellular processes, its dysfunction might contribute to a broader spectrum of neurological conditions than currently recognized .

What experimental models are most effective for studying ELP4 function in human neuronal development?

The most effective experimental models for studying ELP4 function in human neuronal development include:

Model SystemAdvantagesLimitationsApplications
Human iPSC-derived neuronsPhysiologically relevant human cellular context; ability to model patient-specific mutationsVariability between lines; time-consuming differentiationDisease modeling; drug screening; developmental studies
CRISPR-engineered cellular modelsPrecise genetic manipulation; isogenic controlsLimited to in vitro contextMechanistic studies; structure-function analyses
Yeast complementation systemsRapid assessment of functional conservation; high-throughput potentialEvolutionary distance from humansFunctional characterization of variants
Mouse modelsIn vivo development; behavioral assessmentSpecies differences; complex geneticsDevelopmental studies; systems-level analyses
Organoids3D organization; cell-type diversityVariability; incomplete maturationComplex tissue interactions; developmental timing

A multi-model approach is recommended, starting with yeast or cellular systems for initial characterization, followed by validation in more complex models like iPSC-derived neurons or mouse models for in-depth analysis of neuronal development .

What are the current technical challenges in purifying and studying the ELP4 protein biochemically?

Biochemical studies of ELP4 face several technical challenges that researchers must address methodologically:

  • Protein solubility issues during expression and purification, requiring optimization of buffer conditions and potentially using solubility tags

  • Complex formation requirements, as ELP4 functions as part of a multi-subunit complex, necessitating co-expression strategies with other elongator components

  • Post-translational modifications that may affect function, requiring expression in eukaryotic systems rather than bacterial systems

  • Structural analysis difficulties due to the complex's size and potential conformational flexibility

  • Enzymatic assay development challenges for assessing tRNA modification activities

Researchers should consider employing advanced techniques such as cryo-electron microscopy for structural studies, reconstitution of minimal functional complexes, and development of cell-free expression systems to overcome these challenges. Isotope labeling strategies combined with NMR may provide insights into dynamic aspects of ELP4 function within the elongator complex .

How can high-throughput sequencing technologies be utilized to study ELP4-related transcriptional changes?

High-throughput sequencing technologies offer powerful approaches for investigating ELP4-related transcriptional changes:

  • RNA-Seq analysis comparing wild-type and ELP4-deficient cells can identify differentially expressed genes, revealing the transcriptional consequences of ELP4 dysfunction

  • NET-seq (Native Elongating Transcript Sequencing) can map RNA polymerase II positions genome-wide, potentially identifying regions where transcriptional elongation is affected by ELP4 deficiency

  • ChIP-seq targeting RNA polymerase II or histone modifications can reveal how ELP4 affects chromatin states and polymerase occupancy

  • tRNA-seq with specialized protocols for detecting modifications can directly assess the impact of ELP4 on tRNA processing

  • Ribosome profiling can identify translational consequences of altered tRNA modifications

Integration of these various sequencing approaches, combined with sophisticated bioinformatic analysis, can provide a comprehensive view of how ELP4 influences both transcriptional and translational processes. Cell-type specific analyses using single-cell sequencing technologies may be particularly valuable given ELP4's tissue-specific functions .

How might understanding the structure-function relationship of ELP4 lead to therapeutic innovations for epilepsy?

Elucidating the structure-function relationship of ELP4 could lead to several therapeutic approaches for epilepsy:

  • Development of small molecules that can restore function to mutant ELP4 proteins by stabilizing their interaction with other elongator complex components

  • Identification of specific domains that could be targeted to modulate ELP4 activity without disrupting the entire elongator complex

  • Design of peptide mimetics that can substitute for critical ELP4 functions in patients with loss-of-function mutations

  • Gene therapy approaches using CRISPR-based technologies to correct specific ELP4 mutations or modulate expression levels

Current research utilizing complementation systems has already demonstrated that human ELP4 can partially restore function in yeast deletion models , suggesting that targeted therapeutic approaches might similarly compensate for partial loss of function in patients. Structural studies combined with functional assays will be essential for identifying the specific molecular interfaces and activities that represent viable therapeutic targets .

What is the relationship between ELP4's role in tRNA modification and protein synthesis regulation in epileptogenesis?

The relationship between ELP4's function in tRNA modification and epileptogenesis represents a frontier research question with significant therapeutic implications. ELP4, as part of the elongator complex, catalyzes specific modifications at wobble positions in tRNAs , which influence translation efficiency and accuracy. In neurons, which have high demands for precise protein synthesis, disruption of these modifications could lead to:

  • Altered translation of specific subsets of mRNAs with particular codon usage patterns

  • Translational dysregulation of ion channels or synaptic proteins that control neuronal excitability

  • Proteostatic stress resulting from inefficient translation, potentially triggering cellular stress responses

  • Developmental alterations in neuronal connectivity due to impaired synthesis of proteins required for synaptogenesis

Research methodologies to investigate this relationship should include ribosome profiling in neuronal models with ELP4 deficiency, targeted analysis of translation efficiency for epilepsy-related genes, and electrophysiological characterization of neurons with disrupted tRNA modification pathways .

How do alternative splicing variants of ELP4 affect its function across different neural cell types?

The ELP4 gene produces alternatively spliced variants encoding different protein isoforms , but their functional significance across neural cell types remains poorly understood. Advanced methodological approaches to address this question include:

  • Single-cell RNA-seq to identify cell type-specific expression patterns of ELP4 splice variants within the brain

  • Isoform-specific CRISPR knockout models to determine the functional contribution of each variant

  • Proteomics analysis to identify interaction partners unique to specific ELP4 isoforms

  • Electrophysiological assessment of neurons expressing different ELP4 isoforms to determine functional consequences

  • Developmental expression profiling to identify temporal regulation of splicing during neurogenesis

Understanding the distribution and function of these variants may explain why ELP4 mutations have specific effects on certain neuronal populations despite broader expression patterns, potentially revealing opportunities for isoform-specific therapeutic targeting that could minimize off-target effects .

What controls and validation experiments are essential when studying ELP4 using CRISPR-Cas9 genome editing?

When using CRISPR-Cas9 to study ELP4, researchers must implement rigorous controls and validation experiments:

  • Multiple guide RNA designs targeting different regions of ELP4 to confirm phenotypic consistency

  • Off-target analysis using whole-genome sequencing or targeted sequencing of predicted off-target sites

  • Rescue experiments with wild-type ELP4 expression to confirm specificity of observed phenotypes

  • Creation of isogenic control lines differing only in ELP4 status

  • Validation of editing efficiency at both DNA (sequencing) and protein (Western blot) levels

  • Functional assessment of the elongator complex integrity following ELP4 editing

Additionally, researchers should consider complementary approaches such as RNAi or small molecule inhibitors to corroborate CRISPR findings. The design of CRISPR experiments should account for potential compensatory mechanisms and cellular adaptations to ELP4 loss, which may mask acute phenotypes or create secondary effects not directly related to ELP4 function .

How should researchers approach contradictory findings between different model systems when studying ELP4?

When faced with contradictory findings across model systems, researchers should employ a systematic approach:

  • Critically evaluate methodological differences between studies, including:

    • Specific genetic backgrounds of models used

    • Technical variations in ELP4 manipulation (knockout, knockdown, point mutations)

    • Assay sensitivity and specificity

    • Developmental timing or cell-type specificity of analyses

  • Design experiments that directly address discrepancies:

    • Parallel studies in multiple systems under identical conditions

    • Cross-validation using independent methodological approaches

    • Dose-response studies to assess threshold effects

    • Time-course analyses to distinguish primary from secondary effects

  • Consider biological explanations for apparent contradictions:

    • Species-specific compensatory mechanisms

    • Functional redundancy with other proteins

    • Context-dependent requirements for ELP4 function

A methodological framework that integrates findings across diverse systems, while explicitly acknowledging their limitations, will yield the most comprehensive understanding of ELP4 biology and disease relevance .

What are the optimal approaches for translating ELP4 research findings from bench to clinical applications?

Translating ELP4 research into clinical applications requires a methodical, multidisciplinary approach:

  • Genetic screening pipeline:

    • Develop clinically validated assays for ELP4 variants in patient populations

    • Establish genotype-phenotype correlations through large cohort studies

    • Identify biomarkers that reflect elongator complex dysfunction

  • Therapeutic development strategy:

    • High-throughput screening for compounds that modulate ELP4 function or bypass requirements

    • Evaluation of gene therapy approaches for correcting specific mutations

    • Assessment of therapeutic windows and potential off-target effects

  • Clinical trial design considerations:

    • Patient stratification based on specific ELP4 variants

    • Selection of appropriate outcome measures reflecting both seizure control and cognitive function

    • Monitoring of downstream molecular effects using accessible biomarkers

  • Regulatory pathway planning:

    • Early engagement with regulatory authorities regarding novel therapeutic modalities

    • Consideration of orphan drug designation for rare ELP4-related disorders

    • Development of companion diagnostics for patient selection

This translational framework bridges fundamental research on ELP4 biology with patient-centered clinical development, maximizing the potential for successful therapeutic interventions for ELP4-related disorders .

How does ELP4 research intersect with broader studies of epigenetic regulation in neurological disorders?

ELP4 research connects to the broader field of epigenetic regulation in neurological disorders through several methodological and conceptual intersections:

  • Histone modification mechanisms: The elongator complex, including ELP4, possesses histone acetyltransferase (HAT) activity that directly influences chromatin accessibility and gene expression . This places ELP4 within the larger framework of chromatin-modifying complexes relevant to neurological disorders.

  • Integrated multi-omics approaches: Comprehensive understanding requires:

    • Integration of transcriptomic, epigenomic, and proteomic data

    • Analysis of ELP4-dependent changes in chromatin states across neural cell types

    • Correlation of epigenetic profiles with neurological phenotypes

  • Developmental timing considerations:

    • Temporal analysis of ELP4 function during critical neurodevelopmental windows

    • Comparison with other epigenetic regulators implicated in neurodevelopmental disorders

    • Assessment of age-dependent epigenetic signatures in ELP4-related conditions

Through these integrative perspectives, ELP4 research contributes to and benefits from the broader understanding of how epigenetic dysregulation contributes to neurological disorders, potentially revealing common mechanisms and therapeutic approaches applicable beyond ELP4-specific conditions .

What computational approaches are most effective for predicting the functional consequences of ELP4 variants?

Effective computational approaches for predicting functional consequences of ELP4 variants combine multiple predictive methods:

  • Sequence-based prediction algorithms:

    • Conservation analysis across species to identify evolutionarily constrained regions

    • Machine learning models trained on known pathogenic variants

    • Splicing effect predictions for variants near exon-intron boundaries

  • Structure-based modeling approaches:

    • Homology modeling of ELP4 protein structure

    • Molecular dynamics simulations to assess variant effects on protein stability

    • Protein-protein interaction interface analysis for variants affecting elongator complex assembly

  • Systems biology integration:

    • Network analysis incorporating known ELP4 interaction partners

    • Pathway enrichment to identify biological processes potentially affected

    • Integration with tissue-specific expression data to predict context-dependent effects

  • Validation and refinement methodology:

    • Iterative refinement using experimental feedback

    • Cross-validation against clinical phenotype data

    • Ensemble methods combining multiple prediction algorithms

These computational approaches should be applied hierarchically, starting with rapid screening methods followed by more computationally intensive analyses for variants of particular interest. Integration of computational predictions with functional validation experiments provides the most reliable assessment of variant pathogenicity .

How should researchers design multidisciplinary studies to comprehensively understand ELP4's role in human health and disease?

Designing multidisciplinary studies to comprehensively understand ELP4 requires systematic coordination across multiple research domains:

  • Core technology platforms integration:

    • Genomics: Identification of ELP4 variants in patient populations

    • Transcriptomics: Analysis of expression changes resulting from ELP4 dysfunction

    • Proteomics: Characterization of ELP4 protein interactions and post-translational modifications

    • Structural biology: Determination of ELP4 structure within the elongator complex

    • Bioinformatics: Integration of multi-omics data and pathway analysis

  • Experimental model coordination:

    • Parallel studies in yeast, cellular, and animal models to provide complementary insights

    • Standardized phenotypic assessments across models

    • Collaborative tissue and data sharing between research groups

  • Clinical research integration:

    • Natural history studies of ELP4-related disorders

    • Biomarker development for monitoring disease progression and treatment response

    • Patient-derived sample collection coordinated with basic research efforts

  • Interdisciplinary team structure:

    • Regular communication between basic scientists, clinicians, and computational experts

    • Shared methodological standards and data formats

    • Centralized database for integrating findings across research domains

This multidisciplinary framework enables comprehensive characterization of ELP4 biology from molecular mechanisms to clinical manifestations, facilitating translation of research findings into improved patient care .

Product Science Overview

Function and Role

The elongator complex, including ELP4, is essential for multiple tRNA modifications, such as mcm5U (5-methoxycarbonylmethyl uridine), mcm5s2U (5-methoxycarbonylmethyl-2-thiouridine), and ncm5U (5-carbamoylmethyl uridine) . These modifications are critical for the proper functioning of tRNA and, consequently, for protein synthesis.

ELP4 also plays a role in the formation of carboxymethyluridine in the wobble base at position 34 in tRNAs . This modification is vital for the accurate decoding of the genetic code during translation.

Genetic and Clinical Significance

The ELP4 gene has been associated with several genetic disorders. Notably, mutations in this gene have been linked to Rolandic epilepsy, a common form of childhood epilepsy . This association underscores the importance of ELP4 in neurological development and function.

Research and Applications

Research on ELP4 has provided insights into its role in transcriptional regulation and tRNA modification. The human recombinant form of ELP4 is used in various studies to understand its function and to develop potential therapeutic interventions for related genetic disorders.

In summary, ELP4 is a vital component of the elongator complex, playing a significant role in transcriptional elongation and tRNA modification. Its association with genetic disorders like Rolandic epilepsy highlights its importance in human health and disease .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.