ELP5 Human

Elongator Acetyltransferase Complex Subunit 5 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Table 1: Biochemical Properties of ELP5 Human

ParameterSpecification
Molecular Weight37.2 kDa
Amino Acid Sequence1-316aa (UniProt Q8TE02) with His-tag
SourceE. coli
Buffer Composition20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, 20% glycerol, 1 mM DTT
Purity>80% (SDS-PAGE)

Functional Role in Cellular Mechanisms

ELP5 is a subunit of the Elongator complex, which consists of two subcomplexes:

  • ELP123: Mediates histone acetyltransferase activity.

  • ELP456: Hexameric ATPase critical for tRNA binding and wobble U34 modification .

Key functional insights:

  • tRNA Modification: ELP5 stabilizes the Elongator complex, enabling ATPase activity required for thiolation of U34 tRNA . Loss of ELP5 disrupts tRNA modification, impairing translation of stress-responsive proteins like hnRNPQ, which regulates p53 synthesis .

  • p53 Regulation: Depletion of ELP5 reduces p53-dependent apoptosis in gallbladder cancer (GBC) cells, contributing to gemcitabine resistance .

  • Disease Links: Pathogenic ELP3 variants (linked to ELP5 stability) are associated with cancer and neurodegenerative diseases like ALS .

Table 2: Research Findings on ELP5 in Disease Contexts

Study FocusKey FindingsSource
Gemcitabine ResistanceELP5 loss reduces apoptosis in GBC via impaired p53 translation. Restoring ELP5 rescues drug sensitivity .
Cryo-EM StructuresHuman ELP123-tRNA complexes reveal conserved mechanisms in U34 modification. Pathogenic ELP3 mutations disrupt ATPase activity .
Genetic Associationsrs222857 (CLDN7/ELP5 locus) correlates with adiponectin response in diabetes, implicating ELP5 in metabolic regulation .
ToxicologyELP5 expression is modulated by environmental toxins (e.g., cadmium, bisphenol A) .

Implications for Drug Development

  • Cancer Therapy: ELP5’s role in tRNA modification and p53 activation highlights its potential as a biomarker for gemcitabine efficacy in GBC .

  • Neurodegeneration: Structural studies of ELP3-ELP5 interactions provide targets for ameliorating Elongator dysfunction in ALS .

  • Metabolic Disorders: Genetic variants in the ELP5 locus influence adiponectin levels, suggesting therapeutic avenues for obesity-related insulin resistance .

Future Research Directions

  • Mechanistic Studies: Clarify how ELP5 stabilizes the Elongator complex and modulates tRNA specificity.

  • Clinical Trials: Validate ELP5 as a predictive marker for chemotherapy response in GBC.

  • Drug Discovery: Develop small molecules targeting ELP456 ATPase activity to restore tRNA modification in Elongator-related diseases .

Product Specs

Introduction
Elongator acetyltransferase complex subunit 5 (ELP5) is a member of the ELP5 family. It functions as a subunit of the RNA polymerase II elongator complex, which is a histone acetyltransferase component of the RNA polymerase II (Pol II) holoenzyme and participates in transcriptional elongation. Elongator plays a role in chromatin remodeling and is involved in the acetylation of histones H3 and likely H4. ELP5 is involved in cell migration and is widely expressed in the heart, brain, liver, skeletal muscle, and testes.
Description
Recombinant human ELP5 protein produced in E. coli is a single, non-glycosylated polypeptide chain containing 339 amino acids (amino acids 1-316) with a molecular mass of 37.2 kDa. The ELP5 protein is fused to a 23 amino acid His-tag at the N-terminus and purified by proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
ELP5 protein solution (0.25 mg/mL) in 20 mM Tris-HCl buffer (pH 8.0), 0.15 M NaCl, 20% glycerol, and 1 mM DTT.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Greater than 80.0% purity as determined by SDS-PAGE.
Synonyms
C17orf81, DERP6, HSPC002, MST071, MSTP071, Dermal papilla-derived protein 6, S-phase 2 protein, ELP5.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMTPSEGA RAGTGRELEM LDSLLALGGL VLLRDSVEWE GRSLLKALVK KSALCGEQVH ILGCEVSEEE FREGFDSDIN NRLVYHDFFR DPLNWSKTEE AFPGGPLGAL RAMCKRTDPV PVTIALDSLS WLLLRLPCTT LCQVLHAVSH QDSCPGDSSS VGKVSVLGLL HEELHGPGPV GALSSLAQTE VTLGGTMGQA SAHILCRRPR QRPTDQTQWF SILPDFSLDL QEGPSVESQP YSDPHIPPVD PTTHLTFNLH LSKKEREARD SLILPFQFSS EKQQALLRPR PGQATSHIFY EPDAYDDLDQ EDPDDDLDI.

Q&A

What is ELP5 and what are its primary cellular functions?

ELP5 functions as a critical component of the Elongator complex, which plays an essential role in wobble uridine 34 (U34) tRNA modification in human cells. The protein contributes significantly to the structural integrity and stability of the entire Elongator complex, serving as a scaffolding protein that maintains proper complex assembly. Loss of ELP5 has been demonstrated to impair the integrity and stability of the Elongator complex, directly abrogating wobble U34 tRNA modification processes that are vital for specific protein translation . Research indicates that ELP5 particularly influences the translation of specific mRNAs that rely on U34 tRNA modifications, including hnRNPQ mRNA, which functions as a P53 internal ribosomal entry site (IRES) trans-acting factor. This relationship establishes a mechanistic link between ELP5 and P53-dependent cellular processes such as apoptosis in response to chemotherapy agents like gemcitabine. Understanding this fundamental function is crucial as it explains how ELP5 contributes to cancer treatment response through epigenetic regulation of translation.

How does ELP5 relate to cancer treatment response?

ELP5 expression levels have been directly linked to gemcitabine sensitivity in gallbladder cancer (GBC), establishing it as a potential biomarker for treatment efficacy prediction. According to genome-wide CRISPR screening research, loss of ELP5 significantly reduces gemcitabine-induced apoptosis in GBC cells through a P53-dependent mechanism . This reduced sensitivity occurs because ELP5 depletion impairs the wobble U34 tRNA modification-dependent translation of hnRNPQ mRNA, which is necessary for P53 internal ribosomal entry site (IRES)-dependent translation. Clinical data supports this laboratory finding, as GBC patients with lower expression levels of ELP5, hnRNPQ, or P53 demonstrate poorer survival outcomes after gemcitabine chemotherapy . These molecular and clinical observations suggest that ELP5 expression analysis could potentially be incorporated into treatment decision algorithms for patients with gallbladder cancer. The ELP5-hnRNPQ-P53 axis represents a targetable pathway that may be leveraged to enhance chemosensitivity in resistant tumors or to develop companion diagnostics for gemcitabine therapy.

How does the Elongator complex and ELP5 influence protein translation?

The Elongator complex, of which ELP5 is a crucial component, mediates wobble U34 tRNA modification, which significantly impacts the efficiency and fidelity of protein translation for specific subsets of mRNAs. This tRNA modification process is particularly important for the translation of mRNAs containing specific codon combinations that rely on U34-modified tRNAs for optimal translation . When ELP5 is depleted, the integrity and stability of the entire Elongator complex becomes compromised, which in turn abrogates the wobble U34 tRNA modification process. This disruption specifically impedes the translation of mRNAs that depend on these modified tRNAs, such as hnRNPQ mRNA. Research has demonstrated that the translation of hnRNPQ is directly dependent on U34 tRNA modifications facilitated by the intact Elongator complex . Importantly, this translational regulation mechanism represents an additional layer of gene expression control beyond transcriptional regulation, allowing cells to rapidly adjust protein synthesis in response to various stimuli or stressors. This mechanism explains why ELP5 depletion can have profound effects on specific cellular pathways without necessarily altering global protein synthesis.

What techniques are most effective for functional analysis of ELP5?

CRISPR-Cas9 genome editing has proven to be highly effective for ELP5 functional studies, allowing for precise gene knockout and functional validation. As demonstrated in the gemcitabine response research, genome-wide CRISPR screening successfully identified ELP5 as a determinant of chemosensitivity in gallbladder cancer cells . For researchers initiating ELP5 studies, both pooled and arrayed CRISPR screens can be employed depending on the specific research question and available resources. For targeted manipulation of ELP5 expression, RNA interference approaches using pre-designed siRNA sets specific to human ELP5 offer an alternative to genetic knockout, enabling transient knockdown for examining acute effects on cellular phenotypes . When designing functional experiments, it is crucial to include appropriate controls for off-target effects and to validate knockdown/knockout efficiency at both mRNA and protein levels. For mechanistic studies exploring ELP5's role in tRNA modification, specialized techniques such as primer extension analysis or high-performance liquid chromatography coupled with mass spectrometry (HPLC-MS) can be employed to directly measure tRNA modification status. Complementation experiments using ELP5 mutants resistant to the knockdown/knockout strategy can further confirm the specificity of observed phenotypes.

What experimental controls are critical when manipulating ELP5 expression?

When designing experiments involving ELP5 manipulation, several critical controls must be implemented to ensure result validity and interpretability. For siRNA-mediated knockdown, non-targeting control siRNAs that activate the RNAi machinery without targeting any specific transcript are essential to distinguish between specific ELP5 depletion effects and general responses to the presence of siRNA . Additionally, researchers should employ multiple independent siRNA sequences targeting different regions of ELP5 mRNA to minimize off-target effects and confirm phenotype specificity. When using CRISPR-Cas9 for genetic manipulation, both non-targeting guide RNAs and guide RNAs targeting non-essential genes should serve as controls. To definitively attribute observed phenotypes to ELP5 loss, rescue experiments using exogenous ELP5 expression resistant to the knockdown/knockout method employed should be conducted. Particularly important for ELP5 research is the inclusion of U34 modification-independent hnRNPQ mutants as controls when studying the ELP5-hnRNPQ-P53 axis, as has been demonstrated in gemcitabine sensitivity research . This approach can distinguish between direct effects of ELP5 loss and downstream consequences mediated through impaired tRNA modification and subsequent translational defects.

How does ELP5 depletion affect the tRNA modification landscape?

ELP5 depletion leads to comprehensive disruption of the Elongator complex, resulting in specific defects in wobble uridine 34 (U34) tRNA modification with cascading effects on the translation of specific mRNA subsets. When ELP5 is lost, the structural integrity of the Elongator complex becomes compromised, directly impairing its enzymatic activity responsible for catalyzing the critical U34 modification . This modification is particularly important for a subset of tRNAs that decode AA-ending codons, including tRNALysUUU, tRNAGluUUC, and tRNAGlnUUG. The loss of these modifications alters translation elongation rates in a codon-specific manner, affecting proteins with biased codon usage that rely heavily on these particular tRNAs. Research has demonstrated that following ELP5 depletion, there is a measurable reduction in specific tRNA modifications that can be detected using techniques such as liquid chromatography-mass spectrometry (LC-MS) or primer extension assays. These modification defects do not affect global protein synthesis but rather create a "translational stress" that predominantly impacts a subset of mRNAs enriched for codons dependent on U34-modified tRNAs, such as hnRNPQ mRNA . Understanding these specific translational consequences is critical for interpreting downstream phenotypes and identifying which cellular pathways may be particularly vulnerable to ELP5 dysfunction.

What is the mechanistic relationship between ELP5, hnRNPQ, and P53 in chemotherapy response?

Research has elucidated a linear mechanistic axis connecting ELP5 function to chemotherapy response through hnRNPQ-mediated regulation of P53 translation. In this pathway, ELP5 maintains the integrity of the Elongator complex, which is necessary for wobble U34 tRNA modification . These modifications are specifically required for the efficient translation of hnRNPQ mRNA, which contains codons dependent on U34-modified tRNAs. When properly expressed, hnRNPQ protein functions as a critical trans-acting factor for P53 internal ribosomal entry site (IRES)-dependent translation, enabling P53 protein synthesis even under stress conditions where cap-dependent translation is compromised. During gemcitabine treatment, this IRES-dependent translation of P53 is essential for mounting an effective apoptotic response to DNA damage . When ELP5 is depleted, the subsequent reduction in U34 tRNA modification impairs hnRNPQ translation, which in turn prevents efficient P53 IRES-dependent translation, resulting in reduced P53 protein levels despite normal P53 mRNA levels. This translational defect ultimately leads to attenuated apoptotic responses to gemcitabine, explaining the chemoresistance phenotype observed in ELP5-depleted cancer cells. Importantly, rescue experiments using a U34 modification-independent hnRNPQ mutant restored P53 translation and gemcitabine sensitivity in ELP5-depleted GBC cells, confirming the causality in this mechanistic pathway .

How can contradictory findings regarding ELP5 function be reconciled in research?

When encountering contradictory findings about ELP5 function, researchers should systematically analyze several key factors that may explain discrepancies in experimental outcomes. Cell type-specific effects represent a primary consideration, as the reliance on ELP5-mediated tRNA modifications may vary considerably across different cancer types and cellular contexts. The genetic background of cell models used, particularly the P53 status, is critical since the ELP5-hnRNPQ-P53 axis depends on functional P53 for mediating gemcitabine sensitivity . Researchers should verify P53 mutation status in their experimental systems and stratify analyses accordingly. Methodological differences in ELP5 manipulation techniques may also contribute to discrepancies; CRISPR-based knockout versus siRNA-mediated knockdown approaches may result in different compensatory mechanisms or levels of depletion efficiency . The timing of analyses after ELP5 depletion is another important factor, as acute versus chronic loss may trigger different cellular adaptive responses. When reviewing contradictory literature, researchers should pay particular attention to the specific phenotypic readouts employed, as ELP5 depletion may affect different cellular processes to varying degrees depending on their reliance on U34-modified tRNA-dependent translation. Integrating findings from multiple methodological approaches and experimental systems provides the most robust understanding of ELP5 function and resolves apparent contradictions through contextualizing results within appropriate biological frameworks.

How can ELP5 expression analysis be incorporated into clinical decision-making?

ELP5 expression analysis offers promising clinical utility for predicting gemcitabine response in gallbladder cancer patients, potentially guiding treatment selection and improving outcomes. To implement ELP5 testing in clinical settings, researchers must first establish standardized assays for measuring ELP5 protein or mRNA levels in tumor specimens, with clearly defined thresholds for "low" versus "normal" expression based on correlations with clinical outcomes. Clinical validation studies with sufficient statistical power should be conducted to confirm the predictive value of ELP5 expression for gemcitabine response across diverse patient populations. Research indicates that GBC patients with lower ELP5, hnRNPQ, or P53 expression have significantly poorer survival outcomes after gemcitabine chemotherapy, suggesting that a multi-marker panel incorporating all three proteins might provide superior predictive power compared to single markers . For clinical implementation, immunohistochemistry-based assays represent the most practical approach given their compatibility with standard formalin-fixed paraffin-embedded (FFPE) specimens routinely collected during diagnostic procedures. Prospective clinical trials evaluating alternative treatment strategies for patients with low ELP5 expression would be necessary to demonstrate the clinical utility of ELP5 testing for treatment selection. These trials could explore whether patients with low ELP5 expression might benefit more from non-gemcitabine-based regimens or combination approaches designed to overcome the resistance mechanism.

What strategies could potentially overcome gemcitabine resistance mediated by ELP5 deficiency?

Several promising therapeutic strategies may counteract gemcitabine resistance resulting from ELP5 deficiency, based on understanding the underlying resistance mechanism. Since ELP5 deficiency impairs gemcitabine-induced apoptosis through reduced P53 IRES-dependent translation, approaches that bypass this defect by enhancing P53 activity represent logical interventions . Combination therapy with MDM2 inhibitors (e.g., nutlin-3a) could stabilize existing P53 protein, potentially compensating for reduced P53 translation in ELP5-deficient cells. Small molecule IRES activators that enhance P53 IRES-dependent translation through hnRNPQ-independent mechanisms could potentially restore gemcitabine sensitivity. Another approach involves developing inhibitors of enzymes that reverse U34 tRNA modifications, which might increase the pool of modified tRNAs even in ELP5-deficient states. Researchers might also explore compounds that directly enhance wobble U34 tRNA modification through Elongator-independent pathways. For tumors with ELP5 deficiency and wild-type P53, combining gemcitabine with BH3 mimetics (e.g., ABT-737) could potentially restore apoptotic potential by directly activating the intrinsic apoptotic pathway downstream of P53. Gene therapy approaches delivering modified hnRNPQ that does not require U34-modified tRNAs for efficient translation, similar to the rescue constructs used in experimental settings, represent a more speculative but mechanistically sound strategy . Each of these approaches requires rigorous preclinical validation in appropriate models of ELP5-deficient cancer before clinical translation.

How do the experimental approaches for studying ELP5 vary between basic and translational research?

The experimental approaches for studying ELP5 differ substantially between basic mechanistic research and translational applications, with each context demanding specific methodologies and considerations. Basic research typically employs genetically tractable systems such as isogenic cell lines with CRISPR-engineered ELP5 knockout or knockdown via siRNA technology . These controlled systems enable detailed molecular characterization of ELP5 function through techniques like tRNA modification analysis, ribosome profiling to assess translational impacts, and protein interaction studies to map the Elongator complex architecture. By contrast, translational research must accommodate the heterogeneity and complexity of patient-derived materials, requiring robust biomarker assays applicable to clinical specimens. While basic research might use RNA-sequencing or mass spectrometry to comprehensively analyze tRNA modifications, translational applications typically require simplified assays such as immunohistochemistry or RT-PCR that can be standardized across clinical laboratories. Basic research questions often explore mechanistic aspects like the structural basis of ELP5's role in the Elongator complex, whereas translational research focuses on questions with immediate clinical relevance, such as the correlation between ELP5 expression and treatment outcomes in patient cohorts. Both approaches are complementary, with basic discoveries informing potential translational applications, and clinical observations generating new hypotheses for basic mechanistic investigation.

What emerging technologies could advance ELP5 research?

Cutting-edge technologies are poised to accelerate ELP5 research by providing unprecedented insights into its molecular functions and clinical relevance. Single-cell RNA sequencing technologies offer opportunities to characterize ELP5 expression heterogeneity within tumors and correlate this variability with other markers of therapeutic response. This approach could reveal previously unrecognized subpopulations with distinct ELP5 expression patterns and identify cellular contexts where ELP5 function is particularly critical. CRISPR-based screens with single-cell readouts can map genetic interactions with ELP5, potentially uncovering synthetic lethal relationships that could be therapeutically exploited in ELP5-deficient tumors. Advanced proteomics approaches, particularly those analyzing post-translational modifications and protein-protein interactions, may reveal additional regulatory mechanisms controlling Elongator complex assembly and activity beyond the current understanding . Nanopore direct RNA sequencing technologies are emerging as powerful tools for detecting tRNA modifications in native conditions, potentially allowing for more comprehensive analysis of how ELP5 loss affects the entire spectrum of tRNA modifications. Structural biology techniques like cryo-electron microscopy could provide atomic-resolution insights into the Elongator complex architecture and how ELP5 contributes to its stability and function. For translational applications, spatial transcriptomics technologies could map ELP5 expression patterns within the tumor microenvironment, potentially revealing relationships between ELP5 expression, tumor heterogeneity, and therapeutic response that are not apparent in bulk analyses.

How might ELP5 research expand beyond cancer to other disease contexts?

While current research has primarily focused on ELP5's role in cancer, particularly in relation to gemcitabine response in gallbladder cancer, the fundamental function of ELP5 in tRNA modification suggests potential relevance across multiple disease contexts. Neurodevelopmental and neurodegenerative disorders represent promising areas for expanded ELP5 research, as tRNA modifications have been implicated in neuronal protein homeostasis and function. The highly regulated protein translation requirements of neurons may make them particularly sensitive to perturbations in tRNA modification pathways involving the Elongator complex. Metabolic disorders could represent another area where ELP5 function may be relevant, particularly given that specific subsets of proteins involved in metabolic regulation might depend on U34-modified tRNAs for optimal translation. Inflammatory diseases might also be influenced by ELP5 function, as stress-responsive translation regulation is crucial for appropriate immune responses. Systematic analysis of ELP5 expression patterns across human tissues and disease states using public databases could identify previously unrecognized associations between ELP5 dysregulation and specific pathologies. Genome-wide association studies (GWAS) should be examined for potential links between ELP5 genetic variants and disease susceptibility. Researchers investigating these expanded applications should develop appropriate model systems that recapitulate the tissue-specific requirements for ELP5 function, which may differ considerably from the cancer cell models currently employed in most studies .

What methodological advances would facilitate high-throughput screening of ELP5 modulators?

Development of robust high-throughput screening (HTS) assays for ELP5 modulators would accelerate both basic research and drug discovery efforts in this field. Reporter systems that link ELP5 function to easily measurable outputs represent an ideal approach for HTS applications. Researchers could develop cellular reporter systems where expression of fluorescent or luminescent proteins depends on the translation of constructs containing codon combinations that are sensitive to U34 tRNA modifications. Such reporters would produce strong signals when the Elongator complex is fully functional and diminished signals when ELP5 function is compromised . Alternatively, split reporter systems where protein complementation depends on the interaction between ELP5 and other Elongator complex components could enable screening for compounds that either stabilize or disrupt these interactions. Biochemical assays measuring the tRNA modification activity of purified Elongator complex could be adapted to microplate formats, allowing for direct assessment of how test compounds affect the enzymatic function. Cell-based phenotypic screens using ELP5-dependent processes, such as gemcitabine-induced apoptosis in appropriate cancer cell models, could identify compounds that functionally mimic or restore ELP5 activity . Advanced computational approaches such as structure-based virtual screening could complement experimental HTS once high-resolution structural information about the Elongator complex becomes available. For validating hits from primary screens, secondary assays should include direct measurements of tRNA modification status, assessment of hnRNPQ translation efficiency, and functional readouts related to P53 IRES-dependent translation.

Product Science Overview

Gene and Protein Structure

The ELP5 gene, also known as DERP6, is located on chromosome 17p13.1 and spans approximately 8 kilobases, containing 8 exons . The deduced protein consists of 316 amino acids and includes a large central HAP2 elongator superfamily domain . The protein is primarily found in the cytosol, with a smaller proportion in the nuclear fraction .

Function and Mechanism

ELP5 is a component of the elongator complex, which is required for the formation of carboxymethyluridine in the wobble base at position 34 in tRNAs . This complex is involved in several tRNA modifications, including mcm5U (5-methoxycarbonylmethyl uridine), mcm5s2U (5-methoxycarbonylmethyl-2-thiouridine), and ncm5U (5-carbamoylmethyl uridine) . These modifications are crucial for maintaining translational fidelity and proper protein synthesis.

In addition to its role in tRNA modification, ELP5 is involved in the positive regulation of cell migration . It has been shown to increase the transcriptional activity of a p53 reporter, indicating its potential role in tumorigenesis .

Expression and Localization

ELP5 is expressed in various human tissues, with the highest expression levels observed in the heart, brain, liver, skeletal muscle, and testis . Fluorescence-tagged ELP5 shows a punctate cytoplasmic distribution in transfected cells .

Clinical Significance

Mutations or dysregulation of ELP5 have been associated with several diseases, including Ciliary Dyskinesia, Primary, 30, and Multiple Pterygium Syndrome, Escobar Variant . The elongator complex, including ELP5, has also been implicated in the regulation of tumorigenicity and migration of melanoma cells .

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 2024 Thebiotek. All Rights Reserved.