DDX39A Human

DEAD Box Protein 39A Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

Chromosomal Location:

  • Human DDX39A is encoded on chromosome 19 (19p13.12) .

DDX39A participates in multiple RNA metabolic processes:

  1. mRNA Export: Facilitates nuclear export of spliced mRNAs by interacting with the transcription/export (TREX) complex .

  2. Spliceosome Assembly: Assists in pre-mRNA splicing via spliceosome formation .

  3. Innate Immune Regulation:

    • Suppresses RNA virus replication by binding conserved viral RNA structures .

    • Inhibits alphaviruses (e.g., chikungunya, Sindbis) independently of interferon pathways .

Antiviral Mechanism Against Alphaviruses

A 2023 study revealed DDX39A’s role in combating alphaviruses through:

  • Cytoplasmic Relocalization: Translocates from nucleus to cytoplasm during infection to bind viral RNA .

  • RNA Structure Recognition: Targets the 5ʹ conserved sequence element (5ʹCSE) in alphavirus genomes (Figure 1) .

  • Specificity: Active against CHIKV, SINV, VEEV, and ONNV but not coronaviruses or rhabdoviruses .

Experimental Validation:

  • siRNA knockdown of DDX39A increased CHIKV RNA levels 10-fold (p < 0.001) .

  • CRISPR-Cas9 knockout cells showed 8–12× higher viral titers .

VirusFold Increase in Replication (DDX39A Knockout)
CHIKV10×
SINV
VEEV

Tissue Expression Profile

DDX39A is ubiquitously expressed, with elevated levels in:

  • Central Nervous System: Cerebral cortex, hippocampus .

  • Immune Organs: Lymph nodes, spleen .

  • Reproductive Tissues: Testis, ovary .

TissueExpression Level
Cerebral cortexHigh
LiverModerate
Skeletal muscleLow

Clinical Significance

  • Cancer: Promotes hepatocellular carcinoma progression via Wnt/β-catenin pathway activation .

  • Therapeutic Target: Potential for broad-spectrum antiviral drug development against alphaviruses .

Research Gaps and Future Directions

  1. Structural basis of DDX39A’s specificity for 5ʹCSE.

  2. Role in non-alphavirus RNA viruses.

  3. Mechanistic links between RNA helicase activity and oncogenesis.

Product Specs

Introduction
DDX39A, a member of the DEAD box protein family, is characterized by the conserved Asp-Glu-Ala-Asp motif. This motif plays a role in various cellular processes, including RNA secondary structure alteration, nuclear and mitochondrial splicing, and ribosome and spliceosome assembly. Several DEAD box proteins are involved in embryogenesis, spermatogenesis, and cellular growth and division.
Description
Recombinant human DDX39A, expressed in E. coli, is a single, non-glycosylated polypeptide chain consisting of 274 amino acids (residues 1-249) with a molecular weight of 31 kDa. It includes a 25 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile solution without color.
Formulation
A solution of DDX39A protein (1 mg/ml) in a buffer containing 20 mM Tris-HCl (pH 8.0), 10% glycerol, and 0.4 M Urea.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For longer storage, freeze at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for extended storage. Minimize repeated freeze-thaw cycles.
Purity
Purity exceeds 80.0% as assessed by SDS-PAGE analysis.
Synonyms
DEAD (Asp-Glu-Ala-Asp) box polypeptide 39A, BAT1, BAT1L, DDX39, DDXL, URH49, ATP-dependent RNA helicase DDX39A, DEAD box protein 39, Nuclear RNA helicase URH49, DDX39A.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEFMAEQD VENDLLDYDE EEEPQAPQES TPAPPKKDIK GSYVSIHSSG FRDFLLKPEL LRAIVDCGFE HPSEVQHECI PQAILGMDVL CQAKSGMGKT AVFVLATLQQ IEPVNGQVTV LVMCHTRELA FQISKEYERF SKYMPSVKVS VFFGGLSIKK DEEVLKKNCP HVVVGTPGRI LALVRNRSFS LKNVKHFVLD ECDKMLEQLD MRRDVQEIFR LTPHEKQCMM FSATLSKDIR PVCRKFMQDP MEVF.

Q&A

What is DDX39A and what are its primary cellular functions?

DDX39A is a DExD-box RNA helicase that functions as an RNA-dependent ATPase involved in multiple aspects of RNA metabolism. As a member of the helicase superfamily 2 (SF2), DDX39A contains a structurally conserved helicase core with sequence motifs required for ATP-binding, ATPase, and helicase activities. Its primary functions include:

  • Nuclear mRNA export regulation

  • RNA splicing modulation

  • Participation in RNA-protein complex assembly

  • Control of viral RNA during infection

DDX39A predominantly localizes to the nucleus under normal conditions but can relocalize to the cytoplasm during certain viral infections, such as alphavirus infection . The protein works in conjunction with other RNA-binding proteins to regulate various steps of RNA processing, demonstrating its importance in maintaining proper RNA metabolism in human cells .

How does DDX39A differ from its paralog DDX39B?

Despite sharing approximately 90% sequence homology, DDX39A (also known as URH49) and DDX39B (also known as UAP56) display both overlapping and distinct functions:

  • Both are involved in nuclear mRNA export and certain aspects of RNA splicing

  • Both can stimulate RNA binding of the protein PHAX, suggesting shared roles in small nuclear ribonucleoprotein (snRNP) biogenesis

  • DDX39B specifically regulates splicing of certain immune transcripts such as IL7R and FOXP3, a function that cannot be rescued by DDX39A

  • DDX39A demonstrates antiviral activity against alphaviruses (CHIKV, SINV, VEEV, ONNV), while DDX39B does not affect alphavirus replication

  • Exons that specifically depend on DDX39B typically contain U-poor/C-rich polypyrimidine tracts in the upstream intron, suggesting sequence-specific regulatory differences

This functional divergence despite high sequence similarity makes the DDX39A/B system an excellent model for studying the evolution of paralogous genes and their specialized functions in higher organisms .

What experimental models are commonly used to study DDX39A function?

Researchers investigating DDX39A typically employ several experimental systems:

  • Cell culture models: Human cell lines such as U2OS (osteosarcoma), A549 (lung epithelial), and HeLa (cervical epithelial) cells are frequently used for DDX39A studies. These models allow for siRNA-mediated knockdown, CRISPR-Cas9 knockout, and overexpression studies .

  • Biochemical assays:

    • RNA-protein binding assays using purified recombinant proteins

    • Cross-linking immunoprecipitation (CLIP-Seq) to map RNA-protein interactions

    • Subcellular fractionation to monitor protein localization

    • GST pull-down assays to assess protein-protein interactions

  • Microscopy techniques:

    • Immunofluorescence microscopy to visualize protein localization

    • Automated high-content imaging for phenotypic screens

  • Functional assays:

    • Viral infection assays using various RNA viruses

    • RT-qPCR to quantify viral RNA and host gene expression

    • Alternative splicing analysis using RT-PCR and RNA-seq

These models collectively provide comprehensive insights into DDX39A's molecular functions, regulatory mechanisms, and physiological roles .

What role does DDX39A play in RNA splicing regulation?

DDX39A contributes to RNA splicing regulation through several mechanisms:

  • Alternative splicing modulation: DDX39A affects the splicing of specific pre-mRNAs, but with some differences compared to its paralog DDX39B. While DDX39A and DDX39B share significant redundancy in their target genes, certain transcripts specifically require one paralog or the other .

  • Sequence specificity: Unlike DDX39B, which regulates exons with U-poor/C-rich polypyrimidine tracts, DDX39A appears to have different sequence preferences. This distinction creates a molecular basis for their non-redundant roles in splicing regulation .

  • Methodological approach to study DDX39A splicing function:

    • Perform DDX39A knockdown or knockout followed by RNA-seq

    • Analyze alternative splicing events using computational tools such as rMATS or VAST-TOOLS

    • Validate changes in specific splicing events by RT-PCR

    • Compare splicing patterns between DDX39A and DDX39B depletion to identify unique and shared targets

    • Use minigene reporter assays to test direct effects on specific splicing events

Understanding DDX39A's role in splicing provides insight into how RNA helicases contribute to the complex regulation of gene expression through post-transcriptional mechanisms .

How does DDX39A contribute to U snRNP biogenesis?

DDX39A (URH49) contributes to U snRNP biogenesis through the following mechanisms:

  • PHAX loading activity: DDX39A stimulates the binding of PHAX (phosphorylated adaptor for RNA export) to RNA, a critical step in U snRNP assembly and export. This activity appears to be shared with its paralog DDX39B (UAP56) but is not observed with other RNA helicases like DBP5/DDX19 .

  • Protein-protein interactions: DDX39A directly interacts with PHAX, likely facilitating its loading onto target RNAs. This interaction appears to be specific to the DDX39 family of helicases .

  • ATP-dependent function: The loading of PHAX onto RNA likely involves ATP hydrolysis, consistent with DDX39A's function as an RNA-dependent ATPase .

  • Experimental approaches to study this process:

    • In vitro RNA-protein binding assays using labeled RNA and purified proteins

    • Co-immunoprecipitation (co-IP) experiments to detect protein-protein interactions

    • RNA co-IP assays to identify RNA-protein complexes

    • GST pull-down assays using nuclear lysates from various cell types

    • Analysis of U snRNA export using cell fractionation and RNA detection methods

This loading activity represents a novel aspect of TREX complex components in U snRNP biogenesis and highlights the specialized roles of RNA helicases in ribonucleoprotein complex assembly .

What methods are most effective for studying DDX39A's interactions with specific RNA sequences?

To effectively study DDX39A's interactions with specific RNA sequences, researchers should consider these methodological approaches:

  • CLIP-Seq (Cross-linking immunoprecipitation followed by sequencing):

    • UV cross-linking to create covalent bonds between DDX39A and its bound RNAs in vivo

    • Immunoprecipitation of DDX39A-RNA complexes using specific antibodies

    • RNA fragmentation, library preparation, and high-throughput sequencing

    • Bioinformatic analysis to identify enriched binding sites and motifs
      This approach has successfully identified DDX39A binding to the 5'CSE element in alphavirus RNA .

  • RNA electrophoretic mobility shift assays (EMSA):

    • Incubation of purified recombinant DDX39A with labeled RNA probes

    • Analysis of complex formation by native gel electrophoresis

    • Competition assays with unlabeled RNAs to determine specificity

  • RNA-protein binding assays with purified components:

    • Using purified recombinant DDX39A and synthetic RNA oligonucleotides

    • Testing ATP-dependent and ATP-independent binding

    • Analyzing the effect of RNA structure on binding efficiency

  • Structure probing of RNA-protein complexes:

    • SHAPE (Selective 2'-hydroxyl acylation analyzed by primer extension)

    • Hydroxyl radical footprinting

    • Dimethyl sulfate (DMS) probing to identify protein-protected regions of RNA

  • Fluorescence-based approaches:

    • Fluorescence anisotropy to measure binding affinities

    • FRET (Förster resonance energy transfer) to assess conformational changes during binding

These techniques, especially when used in combination, provide comprehensive insights into the sequence and structural specificity of DDX39A-RNA interactions, helping to elucidate its mechanistic roles in various cellular processes .

How does DDX39A control alphavirus infection?

DDX39A exerts antiviral activity against alphaviruses through several interconnected mechanisms:

  • Direct binding to viral genomic RNA: DDX39A specifically recognizes and binds to the 5' conserved sequence element (5'CSE) in alphavirus genomic RNA, as demonstrated by CLIP-Seq analysis. This highly conserved RNA structure is essential for the antiviral activity of DDX39A, as deletion of this structure renders CHIKV insensitive to DDX39A-mediated restriction .

  • Cytoplasmic relocalization during infection: While DDX39A is predominantly nuclear under normal conditions, alphavirus infection triggers its accumulation in the cytoplasm where viral RNA replication occurs. This relocalization is dependent on active viral replication, as UV-inactivated viruses cannot induce this response .

  • Interferon-independent restriction: Unlike many antiviral factors, DDX39A's activity against alphaviruses is independent of the canonical interferon pathway. DDX39A is not itself an interferon-stimulated gene (ISG), and its depletion does not affect ISG induction. This represents a distinct layer of antiviral defense .

  • Broad spectrum activity against alphaviruses: DDX39A controls diverse alphaviruses including CHIKV, SINV, VEEV, and ONNV, but shows no activity against other RNA viruses such as coronaviruses (229E, OC43), picornaviruses (CVB), bunyaviruses (RVFV), or rhabdoviruses (VSV). This specificity suggests recognition of conserved features unique to the alphavirus family .

Experimental approaches to study this mechanism include viral infection assays with DDX39A-depleted cells, quantification of viral RNA by RT-qPCR, immunofluorescence microscopy to track DDX39A localization, and biochemical fractionation to monitor protein redistribution during infection .

What is the relationship between DDX39A and the interferon response?

Unlike many antiviral factors, DDX39A operates independently of the canonical interferon (IFN) pathway, as evidenced by multiple experimental observations:

  • DDX39A is not an interferon-stimulated gene (ISG):

    • DDX39A mRNA and protein levels do not increase following poly(I:C) treatment, a potent IFN inducer

    • Viral infections with SINV, CHIKV, or Sendai virus (SeV) do not alter DDX39A expression levels

    • This contrasts with canonical ISGs like IFIT1, which show strong induction

  • DDX39A does not stimulate IFN production:

    • Depletion of DDX39A does not decrease ISG production following poly(I:C) treatment

    • This indicates that DDX39A is not required for IFN pathway activation or signaling

    • Unlike some other DExD/H-box helicases (e.g., RIG-I and MDA-5), DDX39A does not appear to function as a pattern recognition receptor

  • DDX39A antiviral activity is maintained in IFN-deficient contexts:

    • The antiviral effect against alphaviruses occurs without detectable IFN signaling

    • This represents an alternative antiviral mechanism distinct from the well-characterized IFN system

  • Experimental methods to study this relationship:

    • qPCR analysis of ISG expression following DDX39A knockdown/overexpression

    • ISG protein detection by immunoblotting

    • Use of reporter assays for IFN promoter activity

    • Comparison of antiviral effects in wild-type versus IFN receptor knockout cells

This interferon-independent antiviral activity highlights DDX39A as part of a complementary immune defense system that can restrict viral replication through direct recognition of viral RNA structures rather than through the induction of interferon-stimulated genes .

How can researchers experimentally distinguish between DDX39A and DDX39B functions in antiviral immunity?

Experimentally distinguishing between DDX39A and DDX39B functions in antiviral immunity requires careful methodological approaches to overcome their high sequence similarity:

  • Gene-specific knockdown approaches:

    • Design siRNAs targeting unique regions of each transcript, typically the UTRs

    • Validate knockdown specificity by qPCR and western blot for both proteins

    • Use individual siRNAs rather than pools to minimize off-target effects

    • Perform rescue experiments with siRNA-resistant cDNA constructs to confirm specificity

  • CRISPR-Cas9 knockout strategies:

    • Generate single knockout cell lines for each gene

    • Create double knockout lines followed by complementation with individual genes

    • Design sgRNAs targeting unique exons

    • Validate knockout by genomic sequencing and protein expression analysis

  • Domain swap experiments:

    • Create chimeric proteins exchanging domains between DDX39A and DDX39B

    • Express these in knockout backgrounds to identify critical regions

    • Focus on non-conserved regions that might confer specificity

  • Infection assays with multiple virus types:

    • Test a panel of viruses from different families (e.g., alphaviruses, coronaviruses, picornaviruses)

    • Compare virus replication by various readouts (infectious titers, viral RNA, viral protein)

    • Perform microscopy to quantify infection rates in knockdown/knockout cells

    • Assess cytoplasmic relocalization of each protein during infection

  • Biochemical approaches:

    • Perform CLIP-seq for both proteins to identify unique and shared RNA targets

    • Compare binding affinities for specific viral RNA structures

    • Assess protein-protein interaction networks through IP-MS experiments

Research has demonstrated that DDX39A, but not DDX39B, exhibits antiviral activity against alphaviruses, providing a clear functional distinction despite their high sequence homology. This difference can be exploited as a model system to understand paralog-specific functions in antiviral immunity .

How does altered DDX39A expression impact cellular RNA processing in disease states?

Altered DDX39A expression in disease states can significantly impact cellular RNA processing through multiple mechanisms:

  • Disruption of alternative splicing regulation:

    • DDX39A regulates a specific subset of alternative splicing events distinct from those controlled by DDX39B

    • In disease states with altered DDX39A expression, these splicing events may be dysregulated

    • Experimental approach: Perform RNA-seq on cells or tissues with increased or decreased DDX39A expression and analyze alternative splicing patterns using computational tools like rMATS

    • Validation method: RT-PCR analysis of specific splicing events identified in RNA-seq data

  • Impaired nuclear export of mRNAs:

    • As DDX39A functions in the TREX (TRanscription-EXport) complex, its dysregulation can affect mRNA export

    • This may lead to nuclear retention of certain transcripts or altered cytoplasmic mRNA populations

    • Experimental approach: Subcellular fractionation followed by RT-qPCR or RNA-seq to assess nuclear/cytoplasmic distribution of mRNAs

    • Visualization method: RNA fluorescence in situ hybridization (FISH) to track specific transcripts

  • Altered U snRNP biogenesis:

    • DDX39A's role in loading PHAX onto RNA affects U snRNP assembly and trafficking

    • Disruption may lead to defects in the splicing machinery itself

    • Experimental approach: Analysis of U snRNA levels, processing, and localization

    • Biochemical method: Assessment of snRNP assembly using glycerol gradient fractionation

  • Research methodology for studying disease-related impacts:

    • Compare DDX39A expression levels across healthy and diseased tissues using public databases (TCGA, GTEx)

    • Utilize patient-derived samples to validate expression changes

    • Create cellular models with DDX39A overexpression or knockdown

    • Perform comprehensive RNA processing analysis, including splicing, export, and stability

    • Identify disease-relevant target transcripts affected by DDX39A dysregulation

Understanding these mechanisms provides insights into how DDX39A dysregulation contributes to disease pathogenesis and may reveal potential therapeutic approaches targeting RNA processing pathways .

What are the challenges in developing therapeutic approaches targeting DDX39A?

Developing therapeutic approaches targeting DDX39A presents several significant challenges that researchers must address:

  • Functional redundancy with DDX39B:

    • DDX39A shares approximately 90% sequence homology with DDX39B

    • Many cellular functions overlap between these paralogs

    • Challenge: Achieving DDX39A-specific inhibition without affecting DDX39B function

    • Experimental approach: Detailed structural analysis to identify unique binding pockets or interaction surfaces

    • Validation method: Selective inhibition assays comparing effects on DDX39A versus DDX39B activity

  • Essential nature of RNA processing:

    • DDX39A participates in fundamental cellular processes including mRNA export and splicing

    • Complete inhibition may cause substantial toxicity

    • Challenge: Identifying disease contexts where DDX39A inhibition provides a therapeutic window

    • Experimental approach: Cell viability assays comparing normal versus disease cells with DDX39A modulation

    • Analytical method: RNA-seq to identify cancer-specific DDX39A-dependent transcripts

  • Targeting protein-RNA interactions:

    • DDX39A's functions involve dynamic RNA interactions

    • These interfaces are typically large and lack deep binding pockets

    • Challenge: Developing small molecules that can disrupt protein-RNA interactions

    • Experimental approach: High-throughput screening using fluorescence polarization assays with labeled RNA

    • Alternative strategy: RNA aptamer development to interfere with DDX39A function

  • Tissue-specific effects:

    • DDX39A may have different roles in various tissues

    • Challenge: Understanding context-dependent functions to predict therapeutic outcomes

    • Experimental approach: Conditional knockout mouse models with tissue-specific DDX39A deletion

    • Analytical method: Tissue-specific RNA processing analysis

  • Delivery of RNA-targeting therapeutics:

    • Nucleic acid-based approaches (ASOs, siRNAs) face delivery challenges

    • Challenge: Achieving efficient intracellular delivery to relevant tissues

    • Experimental approach: Testing various delivery vehicles (lipid nanoparticles, conjugates)

    • Validation method: Biodistribution studies to confirm target engagement

Despite these challenges, the unique role of DDX39A in certain disease contexts, such as its potential as a cancer biomarker and its specific function in alphavirus infection , suggests that targeted therapeutic approaches may be feasible with continued research into its structure-function relationships and disease-specific activities.

How can structural biology approaches advance our understanding of DDX39A function?

Structural biology approaches can significantly advance our understanding of DDX39A function through several key methodologies:

These structural approaches, especially when integrated with functional studies, would provide molecular insights into DDX39A's roles in RNA metabolism, antiviral activity, and disease associations, potentially guiding therapeutic development .

What are the most promising techniques for identifying the complete spectrum of DDX39A RNA targets?

Identifying the complete spectrum of DDX39A RNA targets requires sophisticated methodological approaches that capture both direct binding interactions and functional impacts on RNA processing:

  • Enhanced CLIP-seq methodologies:

    • eCLIP or iCLIP provide improved signal-to-noise ratio and single-nucleotide resolution

    • frCLIP (formaldehyde RNA immunoprecipitation) can capture weaker or transient interactions

    • Experimental design: Perform in multiple cell types to identify context-dependent targets

    • Analytical approach: Integrated motif discovery and RNA structure prediction

    • Validation method: In vitro binding assays with purified components

  • RNA-map approaches:

    • Correlate DDX39A binding sites with splicing outcomes to generate RNA splicing maps

    • Experimental design: Combine CLIP-seq with RNA-seq following DDX39A modulation

    • Analytical method: Computational integration of binding and splicing data

    • Visualization: Generate positional maps relating binding position to splicing outcome

    • Comparison: Create differential RNA-maps between DDX39A and DDX39B to identify paralog-specific regulation

  • Proximity-based RNA labeling:

    • APEX-seq or RNA-APEX for spatial mapping of RNA interactions

    • Experimental design: Fusion of DDX39A with engineered peroxidase for proximity labeling

    • Advantage: Captures RNAs in native cellular context without crosslinking biases

    • Analytical approach: Compare subcellular fractions to identify compartment-specific targets

    • Validation method: RNA-FISH to confirm colocalization

  • Global RNA structure probing:

    • SHAPE-MaP or DMS-MaPseq to assess RNA structural changes induced by DDX39A

    • Experimental design: Compare structural profiles in DDX39A-depleted versus control cells

    • Analytical approach: Structure change analysis coupled with binding site data

    • Functional validation: Test whether identified structural changes affect RNA processing

    • Mathematical modeling: Predict DDX39A-dependent structural transitions

  • Functional RNA target identification:

    • RNA Antisense Purification (RAP) with DDX39A-specific antibodies

    • CRISPR-Cas13 screens targeting potential RNA substrates

    • Tethering assays using MS2-DDX39A fusions to test functional impact on reporter RNAs

    • Nascent RNA sequencing to identify co-transcriptional DDX39A targets

    • Ribosome profiling to assess impacts on translation

  • Integrative computational approaches:

    • Machine learning models trained on validated targets to predict additional substrates

    • Network analysis to identify RNA regulons controlled by DDX39A

    • Evolutionary conservation analysis of binding motifs and structures

    • Meta-analysis across multiple datasets and experimental conditions

By combining these complementary approaches, researchers can construct a comprehensive atlas of DDX39A RNA targets across different cellular contexts, providing insights into its diverse roles in RNA metabolism and disease .

How might single-cell approaches reveal new insights about DDX39A function in heterogeneous cellular populations?

Single-cell approaches offer powerful methods to uncover new insights about DDX39A function in heterogeneous cellular populations, revealing context-dependent roles that might be masked in bulk analyses:

  • Single-cell RNA sequencing (scRNA-seq) applications:

    • Compare transcriptomes of DDX39A-high versus DDX39A-low cells within natural populations

    • Experimental design: Perform scRNA-seq on tissues or heterogeneous cultures with variable DDX39A expression

    • Analytical approach: Trajectory analysis to identify cell state transitions associated with DDX39A expression

    • Splicing analysis: Use computational tools like BRIE or VAST-TOOLS for single-cell splicing analysis

    • Validation method: Single-molecule FISH to confirm cell-type-specific expression patterns

  • Single-cell CLIP techniques:

    • Emerging methods like scCLIP-seq could reveal cell-type-specific DDX39A-RNA interactions

    • Experimental approach: Combine single-cell isolation with CLIP protocols

    • Alternative strategy: Spatial transcriptomics coupled with in situ proximity ligation

    • Analytical method: Correlate binding patterns with cell state markers

    • Advantage: Reveals heterogeneity in RNA target selection across cell populations

  • Viral infection heterogeneity studies:

    • Single-cell analysis of DDX39A relocalization during alphavirus infection

    • Experimental design: Time-course imaging of DDX39A localization in infected cultures

    • Analytical approach: Quantify cell-to-cell variability in nuclear-cytoplasmic distribution

    • Correlation analysis: Relate DDX39A relocalization to viral replication efficiency

    • Functional validation: Single-cell viral RNA quantification in cells with different DDX39A patterns

  • Cellular microenvironment influences:

    • Study how tissue microenvironment affects DDX39A function

    • Experimental approach: Spatial transcriptomics or Slide-seq with DDX39A activity readouts

    • Analytical method: Identify spatial patterns of DDX39A-dependent RNA processing

    • Validation: Laser capture microdissection followed by targeted analysis

    • Application: Particularly relevant for understanding DDX39A's role in heterogeneous tumors

  • Multimodal single-cell analysis:

    • CITE-seq or REAP-seq to correlate DDX39A protein levels with transcriptome profiles

    • G&T-seq to simultaneously profile genomic DNA and RNA from the same cell

    • TEA-seq to integrate transcriptome, epitope, and chromatin accessibility data

    • Analytical approach: Multi-omics integration to place DDX39A function in broader cellular context

    • Application: Identifying cell states where DDX39A function is critical

  • Methodology for implementation:

    • Generate reporter systems to monitor DDX39A activity in single cells

    • Develop computational pipelines specific for DDX39A-dependent RNA processing events

    • Create indexed CRISPR screens to assess DDX39A function across diverse cell states

    • Implement live-cell RNA imaging to track DDX39A targets in real-time

These single-cell approaches would reveal how DDX39A function varies across cell types, states, and microenvironments, providing insights into its context-dependent roles in normal physiology and disease .

What are the common challenges in achieving effective DDX39A knockdown or knockout?

Researchers working with DDX39A face several technical challenges when attempting knockdown or knockout approaches:

  • Distinguishing between DDX39A and DDX39B:

    • The 90% sequence homology between these paralogs creates specificity challenges

    • Problem: Cross-targeting between DDX39A and DDX39B can occur with poorly designed reagents

    • Solution: Design siRNAs targeting unique regions, typically the 5' or 3' UTRs

    • Validation approach: Always check both DDX39A and DDX39B levels by qPCR and western blot

    • Control strategy: Include DDX39B-targeted conditions as comparative controls

  • Functional redundancy complications:

    • DDX39B may compensate for some DDX39A functions upon knockout

    • Problem: Subtle or absent phenotypes due to compensation

    • Solution: Consider double knockdown approaches with careful titration

    • Alternative strategy: Acute degradation systems (e.g., auxin-inducible degron) to minimize adaptation

    • Validation approach: Rescue experiments with paralog-specific constructs

  • Essential gene considerations:

    • DDX39A may be essential in certain cellular contexts

    • Problem: Cell death or strong selection against complete knockout

    • Solution: Inducible or partial knockdown systems

    • Alternative strategy: Domain-specific mutations rather than complete knockout

    • Experimental approach: Time-course analysis after knockdown induction

  • Technical challenges with CRISPR editing:

    • Problem: Low efficiency of homology-directed repair for precise editing

    • Solution: Use multiple guide RNAs and enrichment strategies

    • Alternative approach: Base editing or prime editing for specific mutations

    • Validation method: Deep sequencing to quantify editing efficiency

    • Control strategy: Generate clonal lines with sequencing validation

  • siRNA off-target effects:

    • Problem: Unintended targeting of other transcripts

    • Solution: Use multiple independent siRNAs and validate phenotypic consistency

    • Control strategy: Include rescue experiments with siRNA-resistant constructs

    • Validation approach: Transcriptome analysis to assess off-target effects

    • Alternative method: Consider CRISPRi for more specific knockdown

  • Practical protocol considerations:

    • Optimal transfection conditions vary by cell type (lipofection, electroporation, viral delivery)

    • Kinetics of knockdown should be assessed (typically 48-72h for siRNA)

    • Protein half-life may necessitate longer depletion times

    • Western blot detection requires specific antibodies that distinguish between paralogs

    • RT-qPCR primer design must ensure paralog specificity

These considerations are crucial for researchers designing experiments targeting DDX39A, ensuring specific and interpretable results when studying its cellular functions .

What controls are essential when studying DDX39A-RNA interactions?

When investigating DDX39A-RNA interactions, implementing appropriate controls is crucial for generating reliable and interpretable data:

  • Specificity controls for RNA binding assays:

    • Paralog comparison: Include DDX39B as a closely related control to identify paralog-specific interactions

    • Mutant protein controls: Use ATPase-deficient mutants (e.g., K95A) to distinguish ATP-dependent interactions

    • RNA competition assays: Include specific and non-specific competitor RNAs to assess binding specificity

    • Heterologous RNA binding protein: Include an unrelated RNA helicase (e.g., DDX6 or DDX3) as a negative control

    • Validation approach: Direct comparison of binding affinities using quantitative methods like surface plasmon resonance

  • CLIP-seq experimental controls:

    • Input controls: Always sequence input RNA for normalization

    • IgG controls: Perform mock IPs with non-specific IgG to identify background binding

    • UV crosslinking controls: Include non-crosslinked samples to identify non-specific interactions

    • RNase titration: Optimize RNase treatment to generate appropriate fragment sizes

    • Biological replicates: Perform at least three independent experiments to ensure reproducibility

    • Computational validation: Motif enrichment analysis to confirm specificity

  • Functional validation controls:

    • Rescue experiments: Reintroduce wild-type or mutant DDX39A to confirm specificity

    • Structure-specific controls: Compare binding to wild-type versus mutated RNA structures

    • Domain deletion analysis: Map the RNA-binding domains of DDX39A

    • Cellular compartment controls: Compare nuclear versus cytoplasmic interactions

    • Validation method: Follow-up individual targets with direct binding assays

  • RNA structure considerations:

    • In vitro transcribed RNAs: Ensure proper folding through thermal cycling

    • Native purification: Consider native RNA purification for structural studies

    • Competitive displacement: Test specificity of structure recognition

    • Mutational analysis: Introduce mutations that preserve or disrupt specific structural elements

    • Biophysical validation: Use techniques like SHAPE or DMS probing to confirm structural integrity

  • Technical controls for quantitative analysis:

    • Standard curves: Include RNA standards for absolute quantification

    • Spike-in controls: Use exogenous RNA species for normalization

    • Sequential immunoprecipitation: To assess completeness of target capture

    • Size-matched control RNAs: To control for length-dependent effects

    • Cross-validation: Compare results across different binding assay formats

These comprehensive controls ensure that identified DDX39A-RNA interactions are specific, functionally relevant, and distinguishable from interactions mediated by related RNA-binding proteins, thereby providing a solid foundation for mechanistic studies of DDX39A function .

How can researchers reconcile contradictory findings about DDX39A function across different experimental systems?

Reconciling contradictory findings about DDX39A function across different experimental systems requires systematic approaches to identify sources of variation and establish consensus:

  • Systematic comparison of experimental conditions:

    • Create a standardized table comparing key parameters across studies:

      ParameterStudy AStudy BStudy C
      Cell typeU2OSHeLaA549
      DDX39A depletion methodsiRNACRISPRshRNA
      Depletion efficiency85%100%70%
      DDX39B statusUnchangedCompensatory increaseNot reported
      Assay timing48h post-KDStable KO72h post-KD
      Readout methodRT-qPCRRNA-seqMicroscopy
    • Identify critical variables that correlate with different outcomes

    • Experimental approach: Systematically vary one parameter at a time to assess its impact

  • Cell type-specific effects:

    • DDX39A function may vary across cell types due to different expression levels of cofactors

    • Problem: Direct comparison across cell lines may be invalid

    • Solution: Perform parallel experiments in multiple cell types under identical conditions

    • Analytical approach: Correlate outcomes with expression profiles of known interactors

    • Validation method: Introduce factors from one cell type into another to test sufficiency

  • Paralog compensation mechanisms:

    • DDX39B may compensate for DDX39A loss to varying degrees across systems

    • Problem: Incomplete knockdown may show different results than complete knockout

    • Solution: Monitor DDX39B levels and activity following DDX39A manipulation

    • Experimental approach: Double knockdown experiments with titrated levels

    • Validation method: Rescue experiments with paralog-specific constructs

  • Temporal considerations:

    • Acute versus chronic depletion may reveal different phenotypes

    • Problem: Adaptation mechanisms may mask initial effects

    • Solution: Use inducible systems to track temporal responses

    • Analytical approach: Time-course experiments following DDX39A depletion

    • Computational method: Dynamic modeling of RNA processing changes over time

  • Resolution through meta-analysis:

    • Integrate data across multiple studies using standardized effect sizes

    • Statistical approach: Random-effects meta-analysis to account for between-study heterogeneity

    • Subgroup analysis: Stratify by experimental conditions to identify sources of variation

    • Visualization: Forest plots to display effect consistency across studies

    • Validation: Design consensus experiments addressing identified variables

  • Practical reconciliation strategy:

    • Direct collaboration between labs with discrepant findings

    • Exchange of reagents, protocols, and cell lines

    • Blinded analysis of samples to reduce bias

    • Pre-registered experimental designs with defined endpoints

    • Multi-center validation studies with standardized protocols

By systematically addressing these factors, researchers can resolve apparent contradictions and develop a more nuanced understanding of DDX39A function that accounts for biological context and experimental variables .

What are the most pressing unanswered questions about DDX39A function?

Despite significant advances in our understanding of DDX39A, several critical questions remain unanswered that represent important directions for future research:

  • Mechanistic basis for paralog-specific functions:

    • How do DDX39A and DDX39B, despite 90% sequence homology, exhibit distinct functions in splicing regulation and antiviral activity?

    • What structural or sequence elements determine their functional specificity?

    • How are their expression patterns and subcellular localizations differentially regulated?

    • This question is fundamental to understanding the evolution and specialization of these closely related helicases

  • Complete characterization of DDX39A RNA targets:

    • What is the full spectrum of RNA targets directly bound by DDX39A?

    • What sequence or structural features define DDX39A binding specificity?

    • How does target selection change under different cellular conditions or stresses?

    • Comprehensive target identification is essential for understanding DDX39A's diverse cellular functions

  • Regulation of DDX39A relocalization during viral infection:

    • What triggers the relocalization of DDX39A from the nucleus to the cytoplasm during alphavirus infection?

    • Is this relocalization mediated by post-translational modifications or protein-protein interactions?

    • Does relocalization occur in response to other cellular stresses or stimuli?

    • Understanding this regulation could reveal broader principles of nuclear-cytoplasmic shuttling of RNA-binding proteins

  • Physiological significance in development and tissue homeostasis:

    • What are the phenotypic consequences of DDX39A deficiency in different tissues in vivo?

    • How does DDX39A function change during development or cellular differentiation?

    • Are there tissue-specific requirements for DDX39A versus DDX39B?

    • These questions require animal models and developmental studies not addressed in the current literature

  • Role in human disease beyond viral infection:

    • How does altered DDX39A expression or mutation contribute to various diseases, particularly cancer?

    • What are the downstream molecular consequences of DDX39A dysregulation in disease contexts?

    • Can DDX39A serve as a prognostic biomarker or therapeutic target in specific diseases?

    • Translational studies are needed to fully understand DDX39A's clinical relevance

  • Integration with other RNA processing pathways:

    • How does DDX39A function coordinate with other RNA processing mechanisms?

    • Does DDX39A participate in regulatory feedback loops controlling gene expression?

    • How is DDX39A activity itself regulated at transcriptional, post-transcriptional, and post-translational levels?

    • Systems biology approaches are needed to place DDX39A in broader regulatory networks

Addressing these questions will require innovative experimental approaches, including advanced structural studies, in vivo models, single-cell technologies, and integrative multi-omics analyses, ultimately leading to a comprehensive understanding of DDX39A's diverse cellular functions and disease relevance .

What emerging technologies or approaches might accelerate research on DDX39A?

Several emerging technologies and innovative approaches have the potential to significantly accelerate research on DDX39A and provide new insights into its functions:

  • Advanced structural biology techniques:

    • Cryo-electron microscopy (cryo-EM) to capture DDX39A in complex with RNA and protein partners

    • AlphaFold2 and RoseTTAFold for computational structure prediction of DDX39A complexes

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map conformational dynamics

    • Integrative structural biology combining multiple data types

    • These approaches could reveal the structural basis for DDX39A's paralog-specific functions and RNA recognition

  • Long-read direct RNA sequencing:

    • Oxford Nanopore direct RNA sequencing to identify DDX39A-dependent RNA processing events

    • Detection of RNA modifications and their relationship to DDX39A binding

    • Analysis of full-length transcripts to connect multiple processing events

    • These methods provide a more complete picture of DDX39A's impact on the transcriptome

  • Spatial transcriptomics and imaging:

    • MERFISH or seqFISH+ for spatial mapping of DDX39A and its RNA targets

    • Lattice light-sheet microscopy for high-resolution live imaging of DDX39A dynamics

    • Proximity labeling techniques (TurboID, APEX) to map the DDX39A protein interaction network

    • These technologies would reveal the spatiotemporal regulation of DDX39A function

  • CRISPR-based technologies:

    • Base editing or prime editing for precise manipulation of DDX39A

    • CRISPRi/CRISPRa for titratable control of DDX39A expression

    • CRISPR RNA-targeting systems (Cas13) to disrupt DDX39A-RNA interactions

    • CRISPR screens targeting potential DDX39A cofactors

    • These tools enable more precise genetic manipulation to dissect DDX39A function

  • Single-molecule approaches:

    • sm-FRET to study conformational changes during DDX39A's catalytic cycle

    • Optical tweezers to measure forces generated during helicase activity

    • Single-molecule imaging of DDX39A-RNA interactions in cells

    • These techniques provide mechanistic insights at unprecedented resolution

  • Organoid and in vivo models:

    • Patient-derived organoids to study DDX39A in disease-relevant contexts

    • Conditional knockout mouse models for tissue-specific DDX39A deletion

    • CRISPR-engineered animal models with tagged endogenous DDX39A

    • These systems enable physiologically relevant studies of DDX39A function

  • AI and machine learning applications:

    • Deep learning for prediction of DDX39A binding sites and RNA structural preferences

    • Network analysis to identify DDX39A-regulated RNA regulons

    • Automated literature mining to integrate DDX39A knowledge across studies

    • These computational approaches accelerate hypothesis generation and data integration

  • High-throughput drug screening platforms:

    • Small molecule screens for DDX39A modulators

    • RNA-targeted drug discovery to affect DDX39A-RNA interactions

    • PROTAC technology for targeted DDX39A degradation

    • These approaches could yield therapeutic strategies and chemical probes for DDX39A function

By leveraging these emerging technologies and approaches, researchers can overcome current technical limitations and gain unprecedented insights into DDX39A's molecular mechanisms, physiological roles, and disease relevance .

How might understanding DDX39A function contribute to broader advances in RNA biology?

Understanding DDX39A function has significant potential to contribute to broader advances in RNA biology across multiple fundamental areas:

  • Paralog specialization in RNA processing:

    • DDX39A and DDX39B provide an excellent model system for studying how highly homologous paralogs evolve distinct functions

    • Insights from their functional divergence could inform our understanding of other duplicated RNA-binding protein families

    • This research may reveal general principles governing protein evolution following gene duplication

    • Methodological impact: Development of approaches to distinguish between highly similar paralogs

  • Integration of RNA processing pathways:

    • DDX39A functions at the intersection of multiple RNA processing steps (splicing, export, snRNP biogenesis)

    • Understanding how these processes are coordinated through DDX39A activity provides insights into the integrated nature of RNA metabolism

    • This research could reveal regulatory hubs connecting different RNA processing pathways

    • Theoretical advancement: Models for coupling between transcription, splicing, and export

  • Structure-function relationships in RNA recognition:

    • DDX39A's specific recognition of alphavirus RNA structures illuminates principles of RNA-protein interactions

    • Mapping the structural basis for recognition specificity advances our understanding of RNA-protein recognition

    • This research may reveal general principles governing structure-specific RNA recognition

    • Methodological impact: Improved prediction of RNA-protein interactions based on structural features

  • Nuclear-cytoplasmic dynamics of RNA-binding proteins:

    • DDX39A's relocalization during viral infection provides a model for studying regulated subcellular trafficking

    • Understanding the mechanisms controlling this relocalization may reveal broader principles of RBP localization

    • This research could uncover signaling pathways that regulate RNA processing through protein localization

    • Technical advancement: Methods for tracking protein movement between compartments

  • RNA-based antiviral immunity:

    • DDX39A's role in controlling alphavirus infection represents a non-canonical antiviral mechanism

    • Understanding its IFN-independent activity may reveal additional layers of innate immunity

    • This research could identify new strategies for targeting viral RNA

    • Theoretical advancement: Expanded model of cellular antiviral mechanisms

  • Context-dependent functions of RNA helicases:

    • DDX39A shows different activities depending on cellular context and binding partners

    • Deciphering these context-dependent functions advances our understanding of regulatory flexibility

    • This research may reveal principles governing conditional protein activities

    • Methodological impact: Context-specific protein function prediction

  • RNA processing in disease pathogenesis:

    • DDX39A dysregulation in disease contexts connects RNA metabolism to pathogenesis

    • Understanding these connections may reveal RNA processing as a therapeutic target

    • This research could identify RNA-based biomarkers and therapeutic approaches

    • Clinical significance: Novel diagnostic and treatment strategies

Product Science Overview

Introduction

DEAD Box Protein 39A (DDX39A), also known as BAT1, is a member of the DEAD box protein family. These proteins are characterized by the conserved motif Asp-Glu-Ala-Asp (DEAD) and are putative RNA helicases . They play crucial roles in various cellular processes involving the alteration of RNA secondary structure, such as translation initiation, nuclear and mitochondrial splicing, and ribosome and spliceosome assembly .

Structure and Function

DDX39A is a single, non-glycosylated polypeptide chain containing 274 amino acids and has a molecular mass of approximately 31 kDa . The protein is involved in multiple cellular processes, including:

  • Translation Initiation: DDX39A helps in the initiation of protein synthesis by unwinding RNA secondary structures that can impede the translation machinery.
  • Splicing: It plays a role in both nuclear and mitochondrial splicing, ensuring the proper removal of introns from pre-mRNA and the correct assembly of exons.
  • Ribosome and Spliceosome Assembly: DDX39A is essential for the assembly of ribosomes and spliceosomes, which are critical for protein synthesis and mRNA processing, respectively .
Biological Significance

The DEAD box protein family, including DDX39A, is implicated in several vital biological processes:

  • Embryogenesis: These proteins are believed to be involved in the early stages of embryonic development.
  • Spermatogenesis: They play a role in the development of sperm cells.
  • Cellular Growth and Division: DDX39A is crucial for cell proliferation and division, making it an important factor in tissue growth and repair .
Clinical Relevance

DDX39A has been identified as a potential biomarker for unfavorable neuroblastoma, a type of cancer that arises from nerve tissue . Its expression levels can provide insights into the prognosis of patients with this condition. Additionally, a pseudogene of DDX39A is present on chromosome 13, and alternative splicing results in multiple transcript variants, although their full-length nature is not yet fully understood .

Recombinant Production

The recombinant form of DDX39A is produced in E. coli and is used for various research applications. It is a valuable tool for studying the protein’s function and its role in different cellular processes. The recombinant protein is typically fused to a tag, such as GST, to facilitate its purification and detection in experimental settings .

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.