EXOSC4 Human

Exosome Component 4 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Overview

AttributeDetail
Gene LocationChromosome 8 (GRCh38.p14)
Protein Length268 amino acids (1–245 aa)
Molecular Weight28.8 kDa
Subcellular LocalizationNucleus and cytoplasm
StructureNon-catalytic core subunit of the RNA exosome barrel-shaped complex

Recombinant Production

EXOSC4 Human is produced in Escherichia coli as a His-tagged polypeptide (23 aa N-terminal tag) and purified via chromatography. It is supplied in Tris-HCl buffer with urea and glycerol for stability .

Core Activities

The RNA exosome processes RNA substrates via two pathways:

  1. Nuclear Exosome:

    • Maturation of rRNA, snRNA, snoRNA.

    • Degradation of non-coding RNAs (e.g., antisense RNAs, PROMPTs) .

  2. Cytoplasmic Exosome:

    • General mRNA turnover and surveillance of aberrant transcripts .

Chromatin Interaction Mechanism

EXOSC4 binds histone H3 co-modified with H3K9me3 + acetylation (K14/K18/K23), recruiting the exosome to decondensed heterochromatin to degrade spurious non-coding RNAs . This interaction prevents genomic instability and maintains transcriptional fidelity .

Role in Cancer Progression

Cancer TypeFindingsMechanismSource
Epithelial Ovarian Cancer (EOC)Overexpression correlates with advanced FIGO stage, shorter OS/PFS.Suppresses Wnt/β-catenin pathway; inhibits EMT via reduced N-cadherin/vimentin.
Pancreatic/Liver CancerDepletion induces apoptosis; overexpression promotes proliferation.Regulates cell cycle (cyclin D1, CDK4).

Neurological Disorders

A L187P variant in EXOSC4 causes:

  • Accumulation of 7S rRNA precursors.

  • Reduced translation efficiency.

  • Impaired interaction with exosome subunits (e.g., DIS3, EXOSC10) .

Experimental Models

ModelApproachOutcomeSource
HepG2/C3A CellsdCas9-KRAB-MeCP2-mediated knockdown.↓ RNA surveillance machinery; ↑ non-coding RNAs.
Recombinant EXOSC4Pull-down assays with histone peptides.Specific binding to H3K9me3K14acK18acK23ac
Neuronal CellsKnockdown of EXOSC4-L187P variant.↓ Steady-state protein levels.

Oncological Implications

EXOSC4 amplification is observed in multiple cancers, including EOC, pancreatic, and liver cancers. Its role in promoting cell cycle progression (via cyclin D1/CDK4 upregulation) and suppressing EMT inhibitors (E-cadherin) makes it a potential therapeutic target .

Neurological Deficits

Biallelic mutations in EXOSC4 are linked to developmental brain disorders, likely due to impaired RNA processing in neurons. This aligns with similar defects in other exosome subunits (e.g., EXOSC3, DIS3) .

Key Interactors

ProteinFunctionInteraction ScoreSource
EXOSC5Exosome ring subunit.0.999
MPHOSPH6Recruits exosome to pre-rRNA.0.999
DIS3LCytoplasmic catalytic exosome component.0.999
EXOSC10Catalytic subunit; displaced by RNA-bound MTR4.0.999

Cryo-EM Data

The nuclear exosome-MTR4 complex (PDB: 6D6Q) reveals:

  • MTR4 Helicase: Unwinds RNA, funneling it into the exosome core.

  • EXOSC4 Position: Part of the non-catalytic ring, stabilizing RNA channel entry .

Biomarker Utility

High EXOSC4 expression in EOC is an independent predictor of poor prognosis, suggesting its utility as a diagnostic marker .

Targeting EXOSC4

  • Oncology: Inhibiting EXOSC4 may suppress tumor growth via Wnt pathway modulation.

  • Neurology: Restoring exosome function could mitigate RNA processing defects in neurological disorders .

Product Specs

Introduction
EXOSC4, a non-catalytic component of the RNA exosome complex, plays a crucial role in RNA processing and degradation. This complex exhibits 3' to 5' exoribonuclease activity and is involved in various cellular processes. Within the nucleus, it contributes to the maturation of stable RNA species such as rRNA, snRNA, and snoRNA. Additionally, it participates in the removal of RNA processing byproducts, non-coding transcripts (including antisense RNA and promoter-upstream transcripts), and mRNAs with processing defects. This regulation ensures proper RNA functionality and prevents the export of defective transcripts to the cytoplasm. Notably, EXOSC4 has been linked to diseases such as hepatitis A and mouth disease.
Description
Recombinant human EXOSC4, expressed in E. coli, is a non-glycosylated polypeptide chain with a molecular weight of 28.8 kDa. It consists of 268 amino acids, encompassing residues 1-245, and includes a 23 amino acid His-tag at the N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The EXOSC4 protein solution (0.5 mg/ml) is supplied in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.4 M Urea, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is advised. Avoid repeated freeze-thaw cycles to maintain product integrity.
Purity
The purity of the EXOSC4 protein is greater than 85%, as determined by SDS-PAGE analysis.
Synonyms
Exosome Component 4, Ribosomal RNA-Processing Protein 41, Exosome Complex Exonuclease RRP41, Exosome Complex Component RRP41, RRP41A, Ski6p, SKI6, p12A.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAGLELL SDQGYRVDGR RAGELRKIQA RMGVFAQADG SAYIEQGNTK ALAVVYGPHE IRGSRARALP DRALVNCQYS SATFSTGERK RRPHGDRKSC EMGLQLRQTF EAAILTQLHP RSQIDIYVQV LQADGGTYAA CVNAATLAVL DAGIPMRDFV CACSAGFVDG TALADLSHVE EAAGGPQLAL ALLPASGQIA LLEMDARLHE DHLERVLEAA AQAARDVHTL LDRVVRQHVR EASILLGD 

Q&A

What is EXOSC4 and what is its role in the RNA exosome complex?

EXOSC4 (also known as RRP41) is a non-catalytic barrel component of the RNA exosome, a multi-subunit ribonuclease complex that is evolutionarily conserved from archaea to humans. It forms part of the essential "ring" structure (comprising EXOSC4-EXOSC9) that is fundamental for RNA processing and degradation activities . The RNA exosome complex serves as the major cellular machinery for RNA surveillance, processing, degradation, and turnover of diverse RNA species essential for cell viability . Within the three-dimensional structure of the RNA exosome, EXOSC4 participates in crucial protein-protein interactions, particularly with EXOSC9, that maintain the structural integrity of the complex .

How is the EXOSC4 gene structured and regulated in humans?

The human EXOSC4 gene is located on chromosome 8 and encodes the EXOSC4 protein that participates in the RNA exosome complex . Gene expression studies indicate that EXOSC4 regulation is context-dependent and can be influenced by cellular state, tissue type, and disease conditions. Recent research has demonstrated that EXOSC4 expression can be modulated by epigenetic mechanisms, specifically through interactions with modified histones. EXOSC4 shows specific enrichment for binding to histone H3 co-modified with K9me3 and acetylation marks (at positions K14, K18, and K23), suggesting that chromatin modifications may regulate EXOSC4 recruitment and function .

What RNA species are targeted by EXOSC4-containing complexes?

The EXOSC4-containing RNA exosome processes and degrades a wide spectrum of RNA substrates including:

RNA TypeExamplesEXOSC4 Function
Messenger RNA (mRNA)Aberrant mRNAs, transcriptional byproductsDegradation, quality control
Ribosomal RNA (rRNA)7S precursor of 5.8S rRNAProcessing, maturation
Transfer RNA (tRNA)Misfolded tRNAsSurveillance, degradation
Small nuclear RNA (snRNA)Precursor snRNAsProcessing, maturation
Non-coding RNAsAntisense RNAs, intronic sequencesRegulation, turnover

EXOSC4, as part of the RNA exosome, contributes to the recognition and processing of these diverse RNA substrates to maintain RNA homeostasis . Experimental evidence from yeast models (using the Rrp41 ortholog) and human cells confirms that disruption of EXOSC4 function leads to accumulation of RNA exosome targets, particularly the 7S precursor of 5.8S rRNA .

What are the optimal methods for studying EXOSC4 protein interactions?

To study EXOSC4 protein interactions, researchers have successfully employed multiple complementary approaches:

  • Affinity Purification Mass Spectrometry (AP-MS): This technique has been effectively used to identify proteins that interact with EXOSC4. Nuclear and cytoplasmic extracts are incubated with immobilized EXOSC4, and co-precipitated proteins are detected by mass spectrometry . This approach has revealed that EXOSC4 interacts with proteins involved in different RNA processing pathways in both nuclear and cytoplasmic compartments.

  • Co-immunoprecipitation (Co-IP): This method has confirmed specific interactions between EXOSC4 and other exosome components, particularly EXOSC2 . When investigating novel interactions, optimized lysis conditions (typically using mild detergents like 0.1% NP-40) help preserve native protein complexes.

  • Structural Biology Approaches: X-ray crystallography and cryo-electron microscopy have been instrumental in defining EXOSC4's position within the RNA exosome complex, revealing its interactions with other subunits like EXOSC9. These studies have shown that specific amino acids, such as Leu187, mediate important inter-subunit contacts .

  • Protein-Peptide Interaction Assays: Pull-down experiments using modified histone peptides have demonstrated that EXOSC4 specifically interacts with histone H3 carrying K9me3 and acetylation marks, suggesting a chromatin-mediated recruitment mechanism .

What gene editing approaches are most effective for EXOSC4 functional studies?

Several gene editing approaches have proven effective for investigating EXOSC4 function:

  • CRISPR-Cas9 with dCas9-KRAB-MeCP2: This approach has been successfully used to generate stable EXOSC4 knockdown in cell lines like HepG2/C3A. The system employs a catalytically inactive Cas9 fused to transcriptional repressors to downregulate EXOSC4 expression without altering the genomic sequence . This method allows for studying the effects of EXOSC4 depletion while maintaining cell viability.

  • RNA Interference (RNAi): siRNA and shRNA approaches targeting EXOSC4 have been employed to study its role in cancer cell survival, particularly in pancreatic cancer models. These studies revealed that EXOSC4 knockdown reduces cell viability and induces apoptosis .

  • Variant Modeling in Orthologous Systems: For studying disease-associated variants like EXOSC4-L187P, modeling the equivalent mutation in yeast (Rrp41-L187P) has provided valuable insights into functional consequences. This approach allows researchers to leverage the power of yeast genetics while investigating human disease variants .

  • Rescue Experiments: To confirm the specificity of EXOSC4 depletion phenotypes, complementation with wild-type or mutant EXOSC4 constructs provides important validation. For example, studies have shown that knockdown of BIK and SESN2 could partially rescue pancreatic cells from viability reduction caused by EXOSC4 knockdown .

How can researchers best analyze RNA targets of EXOSC4?

To analyze RNA targets of EXOSC4, researchers should consider multiple complementary techniques:

  • RNA Sequencing after EXOSC4 Depletion: RNA-seq following EXOSC4 knockdown reveals transcriptome-wide changes and identifies accumulated RNA exosome targets. This approach has demonstrated that EXOSC4 depletion leads to increased expression of non-coding transcripts, including antisense RNAs .

  • RNA Stability Assays: Measuring RNA half-life (using transcription inhibitors like actinomycin D) before and after EXOSC4 depletion identifies direct targets whose stability is regulated by EXOSC4. This approach demonstrated that EXOSC4 destabilizes SESN2 mRNA by promoting its degradation .

  • Cross-linking and Immunoprecipitation (CLIP): CLIP methods can identify direct RNA binding sites of EXOSC4-containing complexes, distinguishing direct from indirect targets. Various CLIP methods (PAR-CLIP, iCLIP, eCLIP) offer different advantages in resolution and efficiency.

  • Polysome Profiling: This technique assesses the impact of EXOSC4 dysfunction on translation. Studies in yeast models (rrp41-L187P) revealed a decrease in actively translating ribosomes and incorporation of 7S pre-rRNA into polysomes , linking RNA processing defects to translational consequences.

How is EXOSC4 implicated in cancer biology?

EXOSC4 plays complex roles in cancer biology with both oncogenic and tumor-suppressive activities depending on context:

  • Amplification in Multiple Cancers: Integrated genomic analyses using The Cancer Genome Atlas (TCGA) PanCancer Atlas Studies have revealed that the EXOSC4 gene is amplified across multiple cancer types , suggesting potential oncogenic functions.

  • Prognostic Significance: EXOSC4 alteration is associated with poor prognosis in pancreatic cancer patients, indicating its potential value as a prognostic biomarker . The table below summarizes survival data from TCGA analysis:

Cancer TypeEXOSC4 StatusMedian Survivalp-value
Pancreatic CancerAmplified/AlteredSignificantly reduced<0.05
Other GI CancersAmplified/AlteredVariable effects-
  • Cell Survival Mechanisms: EXOSC4 is required for the survival of pancreatic cancer cells. Mechanistically, EXOSC4 represses BIK expression (a pro-apoptotic protein) and destabilizes SESN2 mRNA through promoting its degradation . Knockdown of BIK and SESN2 can partially rescue the reduction in cell viability caused by EXOSC4 knockdown.

  • Dual Effects on Cell Growth: Paradoxically, both dysregulated EXOSC4 expression and EXOSC4 overexpression have been linked to cancer cell biology. While some studies show EXOSC4 depletion reduces pancreatic and liver cancer cell growth and triggers apoptosis , others indicate that EXOSC4 overexpression increases proliferation , suggesting context-dependent functions.

What is known about EXOSC4 variants in human diseases?

Recent research has identified disease-associated variants in EXOSC4:

  • EXOSC4-L187P Variant: A biallelic missense variant (NM_019037.3:exon3:c.560T>C) changing leucine to proline at position 187 (p.Leu187Pro) has been linked to a novel neurodevelopmental disorder . This homozygous variant was identified in two affected siblings who presented with:

    • Prenatal growth restriction

    • Failure to thrive

    • Global developmental delay

    • Intracerebral and basal ganglia calcifications

    • Kidney failure

  • Functional Consequences: Molecular characterization of the L187P variant revealed:

    • Decreased steady-state levels of the mutant protein

    • Reduced interaction with other RNA exosome subunits

    • Accumulation of RNA exosome target transcripts, including the 7S precursor of 5.8S rRNA

    • Decreased actively translating ribosomes

    • Incorporation of immature 7S pre-rRNA into polysomes

  • Structure-Function Relationships: Structural analyses indicate that Leu187 of EXOSC4 lies in an α-helix region that interacts with Leu199 in a neighboring β-strand, which contacts Ile234 of EXOSC9. The L187P substitution likely disrupts these interactions, destabilizing EXOSC4 and affecting its integration into the RNA exosome complex .

  • Conservation and Ortholog Studies: The Leu187 position is highly conserved among 100 vertebrate genomes. Studies modeling the equivalent mutation in the yeast ortholog (Rrp41-L187P) show similar molecular defects, indicating evolutionary conservation of this functional domain .

How do EXOSC4 interactions with epigenetic modifications impact gene expression?

Recent studies have revealed a novel connection between EXOSC4 and chromatin regulation:

  • Histone Modification Recognition: EXOSC4 shows specific enrichment for binding to histone H3 co-modified with K9me3 and acetylation marks at positions K14, K18, and K23. This interaction is highly specific, as no binding was observed with other histone H3 peptides .

  • Chromatin-RNA Processing Connection: This interaction suggests a mechanistic link between chromatin state and RNA processing machinery, where EXOSC4 may be recruited to specific chromatin regions through modified histones to regulate local RNA processing and surveillance.

  • Functional Consequences: EXOSC4 depletion leads to:

    • Down-regulation of the RNA surveillance machinery

    • Increased expression of non-coding transcripts, including antisense RNAs

    • Disruption of RNA quality control pathways

  • Co-depletion Effect: Knockdown of EXOSC4 leads to co-depletion of other exosome components, demonstrating its importance for the stability of the entire RNA exosome complex .

How does EXOSC4 coordinate with other exosome components in substrate specificity?

The RNA exosome complex exhibits remarkable substrate specificity despite handling diverse RNA targets. EXOSC4's role in this specificity involves:

  • Structural Contributions: EXOSC4 forms part of the "ring" complex (EXOSC4-EXOSC9) and contributes to the central channel through which RNA substrates pass. The architecture of this channel influences which RNAs can be processed by the complex .

  • Cofactor Interactions: EXOSC4 likely associates with different cofactors in nuclear versus cytoplasmic compartments. Mass spectrometry analysis of EXOSC4 interactors revealed distinct protein networks in these compartments:

    • Nuclear interactors are enriched for proteins involved in chromatin remodeling and protein-DNA complex assembly

    • Cytoplasmic interactors are associated with ncRNA processing and regulation of mRNA stability

  • Substrate Recognition Mechanisms: While EXOSC4 itself doesn't have catalytic activity, it may influence substrate recognition through:

    • Structural stabilization of the complex

    • Facilitating interactions with cofactors that recognize specific RNA features

    • Potential direct interactions with RNA substrates or RNA-binding proteins

  • Comparative Studies: Interestingly, while proteomics detected all RNA exosome complex subunits, only EXOSC4 showed significant enrichment in binding to the hybrid histone mark (H3K9me3 with acetylation), suggesting a unique role for EXOSC4 in chromatin-associated RNA processing .

What mechanisms explain the tissue-specific effects of EXOSC4 dysfunction?

The tissue-specific manifestations of EXOSC4 dysfunction, particularly in neurodevelopmental disorders and cancer, likely arise from several factors:

  • Differential Expression Patterns: While RNA exosome components are expressed in all tissues, their relative abundance varies. Certain tissues, particularly those with high transcriptional activity or specialized RNA processing requirements (brain, pancreas), may be more sensitive to EXOSC4 dysfunction.

  • Tissue-Specific RNA Targets: Different tissues may have unique RNA species that particularly depend on EXOSC4-mediated processing or degradation. For example, the neurological manifestations of EXOSC4-L187P may reflect critical RNA processing requirements in neural development.

  • Developmental Timing: The prenatal growth restriction and developmental delay observed in patients with EXOSC4-L187P suggest that EXOSC4 function may be particularly critical during early development , when precise RNA processing coordinates complex developmental programs.

  • Compensatory Mechanisms: Some tissues may have redundant RNA surveillance pathways that can compensate for EXOSC4 dysfunction, while others lack these backup systems. This differential resilience could explain why certain organs (brain, kidneys) are more affected by EXOSC4 mutations.

  • Cell-Type Specific Stresses: Cancer cells often experience unique stresses (replication stress, metabolic alterations) that may create specific dependencies on RNA quality control mechanisms involving EXOSC4, explaining its role in cancer cell survival .

How do evolutionary changes in EXOSC4 relate to its functional specialization?

The RNA exosome is evolutionarily ancient, with core components conserved from archaea to humans. Analysis of EXOSC4 evolution provides insights into its functional specialization:

  • Sequence Conservation: The high conservation of key residues like Leu187 across 100 vertebrate genomes underscores their functional importance . Comparative sequence analysis shows that the regions involved in inter-subunit interactions (like the interface with EXOSC9) are particularly conserved.

  • Functional Conservation: Modeling the human EXOSC4-L187P variant in the yeast ortholog Rrp41 produced similar molecular defects, including:

    • Growth defects in cells expressing Rrp41-L187P

    • Decreased steady-state levels of the mutant protein

    • Accumulation of RNA exosome target transcripts

    • Decreased actively translating ribosomes

This functional conservation across evolutionarily distant species highlights the fundamental importance of these mechanisms.

  • Specialization in Higher Organisms: Despite core conservation, EXOSC4 in humans may have acquired additional functions, such as:

    • Interactions with epigenetic marks (H3K9me3 with acetylation)

    • Specialized roles in developmental processes

    • Functions in tissue-specific RNA surveillance mechanisms

  • Comparative Interactome Studies: Comparing EXOSC4 protein-protein interactions across species could reveal evolutionarily conserved core interactions versus species-specific associations that reflect functional specialization.

What are the most reliable antibodies and reagents for studying EXOSC4?

Researchers investigating EXOSC4 should consider the following validated reagents:

  • Antibodies for Western Blotting and Immunoprecipitation:

    • Commercial antibodies against human EXOSC4 have been successfully used for western blot detection of both endogenous and overexpressed protein

    • For co-immunoprecipitation studies examining EXOSC4 interactions with other exosome components, antibodies with epitopes outside interaction interfaces are preferable

    • Validation using EXOSC4-depleted cells as negative controls is essential to confirm specificity

  • Expression Constructs:

    • Tagged EXOSC4 constructs (e.g., with FLAG, HA, or GFP tags) have been successfully used for overexpression and interaction studies

    • When introducing disease-associated variants like L187P, both N- and C-terminal tags should be tested to ensure the tag doesn't interfere with the functional consequences of the mutation

  • CRISPR Reagents:

    • The dCas9-KRAB-MeCP2 system has been validated for effective EXOSC4 knockdown

    • Guide RNA design should target promoter regions or early exons for maximum repression efficiency

    • Complete knockout may not be achievable due to the essential nature of EXOSC4, making knockdown approaches preferable

  • Recombinant Proteins:

    • For in vitro biochemical studies, recombinant EXOSC4 protein can be produced, although co-expression with other exosome components may be necessary to maintain stability and proper folding

What are the challenges in interpreting EXOSC4 depletion experiments?

Interpreting EXOSC4 depletion experiments presents several challenges that researchers should consider:

How can researchers distinguish direct from indirect targets of EXOSC4?

Distinguishing direct from indirect targets of EXOSC4 requires multifaceted approaches:

  • Integrative Analysis: Combining multiple datasets helps identify high-confidence direct targets:

    • RNA-seq after EXOSC4 depletion identifies accumulated transcripts

    • RNA half-life measurements identify stabilized RNAs

    • CLIP-seq data reveals direct binding sites

    • Targets identified across multiple approaches are likely direct EXOSC4 substrates

  • Kinetic Studies: Early timepoints after EXOSC4 depletion are more likely to reveal direct targets before secondary effects accumulate. Time-resolved experiments can distinguish immediate from delayed responses.

  • In Vitro Degradation Assays: Reconstituted systems using purified RNA exosome components (including EXOSC4) and candidate RNA substrates can directly test processing activity. Comparing wild-type and mutant (e.g., L187P) EXOSC4 in these assays can provide insights into disease mechanisms.

  • Substrate Features Analysis: Computational analysis of EXOSC4-dependent transcripts may reveal common sequence or structural features that define direct targets. Machine learning approaches can help identify these substrate recognition signatures.

  • Genetic Interaction Studies: Combining EXOSC4 depletion with manipulation of putative cofactors or other RNA degradation pathways can help delineate the direct EXOSC4 targetome through genetic epistasis relationships.

What is the potential role of EXOSC4 in regulating non-coding RNA biology?

EXOSC4's involvement in non-coding RNA regulation represents an exciting frontier:

  • Antisense RNA Regulation: EXOSC4 depletion leads to increased expression of antisense RNAs , suggesting it normally suppresses these regulatory transcripts. This finding has implications for gene expression regulation, as antisense RNAs can modulate the expression of their sense counterparts.

  • lncRNA Processing: EXOSC4, as part of the RNA exosome, likely participates in the processing and turnover of long non-coding RNAs. The specific subset of lncRNAs regulated by EXOSC4 and the consequences for cellular functions remain to be fully characterized.

  • Regulatory RNA Quality Control: EXOSC4 may help distinguish functional non-coding RNAs from transcriptional noise by facilitating the degradation of spurious transcripts. This quality control function could be particularly important in transcriptionally complex regions of the genome.

  • Bidirectional Transcription Regulation: Recent studies suggest RNA exosome components regulate bidirectional transcription from promoters. EXOSC4's role in processing these transcripts and its impact on gene expression directionality warrant further investigation.

How does EXOSC4 contribute to cellular stress responses?

Emerging evidence suggests EXOSC4 plays important roles in cellular stress responses:

  • Oxidative Stress Connection: EXOSC4 regulates SESN2 mRNA stability . Since SESN2 is an important stress-responsive gene involved in antioxidant defense and mTOR regulation, EXOSC4 may modulate cellular responses to oxidative stress.

  • Translation Stress: Studies in yeast show that cells expressing the Rrp41-L187P variant (modeling human EXOSC4-L187P) have decreased actively translating ribosomes and incorporation of immature rRNA into polysomes . This suggests EXOSC4 dysfunction creates translational stress that could activate cellular stress responses.

  • RNA Damage Response: During cellular stress, damaged RNAs can accumulate and require efficient degradation. EXOSC4-containing complexes likely participate in eliminating these damaged transcripts to maintain cellular homeostasis.

  • Cancer Stress Adaptation: EXOSC4 amplification in cancer may represent an adaptation to cancer-specific stresses, including increased transcription rates and replication stress. Cancer cells might become dependent on enhanced RNA surveillance to manage these stresses.

What are the therapeutic implications of targeting EXOSC4 function?

The emerging understanding of EXOSC4 biology suggests several therapeutic directions:

  • Cancer Therapy Potential: Given that EXOSC4 is required for pancreatic cancer cell survival and is amplified in multiple cancer types, it represents a potential therapeutic target. Several approaches could be considered:

    • Small molecule inhibitors disrupting EXOSC4 interactions with other exosome components

    • Peptide-based inhibitors targeting specific EXOSC4 interactions

    • RNA-based therapeutics to downregulate EXOSC4 expression

  • Synthetic Lethality Approaches: EXOSC4 inhibition might create vulnerabilities that could be exploited in combination therapies. Identifying genes that show synthetic lethality with EXOSC4 depletion could reveal promising drug combinations.

  • Biomarker Development: EXOSC4 amplification or expression levels could serve as biomarkers to guide treatment decisions, particularly in pancreatic cancer where EXOSC4 alteration correlates with poor prognosis .

  • Developmental Disorder Treatment: Understanding the molecular consequences of EXOSC4 mutations like L187P could guide development of therapies for associated developmental disorders. Approaches might include:

    • Small molecules that stabilize mutant EXOSC4 protein

    • Targeting downstream pathways affected by EXOSC4 dysfunction

    • RNA-based therapies to modulate levels of specific EXOSC4 targets

  • Delivery Challenges: A major consideration for therapeutic development will be achieving tissue-specific targeting, particularly for neurological manifestations of EXOSC4 dysfunction that would require blood-brain barrier penetration.

Product Science Overview

Background and Structure

EXOSC4 is a part of the exosome complex, a multi-protein complex responsible for degrading RNA molecules in eukaryotic cells. The exosome complex plays a vital role in maintaining RNA homeostasis by processing and degrading different types of RNA, including messenger RNA (mRNA), ribosomal RNA (rRNA), and small nuclear RNA (snRNA).

The human recombinant form of EXOSC4 is typically produced in Escherichia coli (E. coli) and is often tagged with a His-tag at the N-terminus to facilitate purification. The recombinant protein corresponds to the amino acids 1-245 of the human EXOSC4 protein .

Function and Mechanism

EXOSC4, as part of the exosome complex, is involved in the 3’ to 5’ degradation of RNA molecules. This degradation process is essential for the regulation of gene expression, RNA quality control, and the removal of defective or unnecessary RNA molecules. The exosome complex, including EXOSC4, ensures that RNA molecules are processed correctly and degraded when they are no longer needed .

Biological Significance

EXOSC4 and the exosome complex are critical for various cellular processes, including:

  • RNA Processing: The exosome complex processes precursor rRNA, snRNA, and small nucleolar RNA (snoRNA) into their mature forms.
  • RNA Degradation: It degrades aberrant or excess RNA molecules, preventing the accumulation of defective RNA that could disrupt cellular functions.
  • Gene Expression Regulation: By degrading specific RNA molecules, the exosome complex helps regulate the levels of different RNAs, thereby influencing gene expression .
Applications in Research

Recombinant EXOSC4 is widely used in research to study the structure and function of the exosome complex. It is particularly useful in biochemical assays, structural biology studies, and functional analyses. The recombinant form allows researchers to investigate the protein’s properties in a controlled environment, facilitating a deeper understanding of its role in RNA metabolism .

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.