The DDX50 antibody is a specialized immunological tool designed to detect and study the DEAD-box helicase 50 (DDX50) protein, a member of the DExD/H-box RNA helicase family. This antibody is critical for investigating DDX50's role in innate immune signaling, antiviral responses, and RNA metabolism . Commercial versions, such as those from Thermo Fisher Scientific (PA5-65186) and Proteintech (10358-1-AP), are widely used in research to analyze DDX50 expression, localization, and interactions in human and murine systems .
DDX50 is a nucleolar RNA helicase involved in:
Antiviral Defense: Enhances IRF3 activation and restricts replication of RNA/DNA viruses (e.g., Zika, HSV-1, VACV) .
Innate Immune Signaling: Promotes TRIF-dependent IRF3/NF-κB pathway activation independently of RIG-I or MDA5 .
RNA Processing: Collaborates with DDX21 to unwind RNA substrates, influencing ribosomal RNA synthesis .
Viral Restriction: Loss of DDX50 increases viral replication (e.g., Zika virus yields rise by 3–5 fold in KO cells) . Antibodies enable tracking DDX50 expression changes during infection.
Mechanistic Insights: Co-immunoprecipitation studies show DDX50 interacts with TRIF, a key adaptor in antiviral signaling .
Cytokine Regulation: DDX50 knockout reduces IRF3-dependent cytokine production (e.g., CXCL10 and IL-6) during viral infection .
Western Blot: Detects DDX50 at 83 kDa in HeLa and Jurkat cells .
Immunofluorescence: Localizes DDX50 to nucleoli, consistent with its role in RNA processing .
Functional Studies: Antibodies validate DDX50’s role in IRF3 phosphorylation and ISG (e.g., Isg56, Ifnb) upregulation .
Applications: Ideal for comparative studies across species.
Validation: Confirmed in WB, IP, and IF/ICC using HeLa cells .
Storage: Stabilized in PBS with 50% glycerol for long-term use .
DDX50 antibodies are pivotal in dissecting its dual roles in RNA sensing and viral restriction. For example:
Applications : WB
Review: Western blotting analysis of lysates of cells infected with SINV.
DDX50 is a DExD-Box RNA helicase that functions as a viral restriction factor by enhancing IRF3 activation and antiviral signaling. It shares 55.6% amino acid identity with DDX21 but has non-redundant functions in innate immune responses . DDX50 plays a critical role in restricting viral replication of diverse pathogens, including DNA viruses like vaccinia virus (VACV) and herpes simplex virus (HSV-1), as well as RNA viruses such as Zika virus (ZIKV) . Additionally, recent research has identified DDX50 as having glucose-binding capabilities that alter its conformation and impact cellular differentiation processes, making it a multifunctional protein of interest across several research areas .
Based on published research, the following experimental models have proven effective for DDX50 studies:
For immunological studies, both human and mouse cell lines with CRISPR-mediated DDX50 knockout have been successfully used to investigate its role in antiviral responses .
For optimal Western blot detection of DDX50:
Use recommended dilutions of 1:500 to 1:2000 for primary antibody incubation
Be aware that DDX50 has an observed molecular weight of approximately 83 kDa
When working with tissue samples, include appropriate controls as DDX50 expression varies across cell types
For co-immunoprecipitation experiments, rabbit monoclonal anti-Flag antibodies have been successfully used with DDX50-HA cell lines
Consider using reduced-denatured protein samples as DDX50 can form dimers that may complicate band pattern interpretation
Despite sharing 55.6% amino acid identity, DDX50 and DDX21 have distinct functions that can be experimentally differentiated through several approaches:
RNA binding profiles: DDX50 binds a GC-rich SCSSSGCC RNA motif (S denotes G or C), while DDX21 preferentially binds the SCUGSDGC motif . CLIP-seq experiments can distinguish these binding patterns.
Functional differentiation:
Selective knockdown: Design siRNAs targeting non-conserved regions between the two proteins to achieve selective depletion and measure pathway-specific outcomes.
Specific mutants: Use point mutations like DDX50 K187R (ATPase deficient but glucose-binding intact) to selectively impair specific functions while maintaining others .
For robust investigation of DDX50's viral restriction function:
Infection parameters:
Use both high and low MOI (multiplicity of infection) conditions, as DDX50-mediated restriction is most evident at low MOI
For VACV, use MOI 5 (high) or 0.0001-0.0003 (low) in MEF or HEK293T cells
For HSV-1, MOI 0.01 is effective for observing DDX50's restriction effects
For ZIKV, MOI 1 (high) or 0.1 (low) has demonstrated differential restriction
Readouts for viral restriction:
Control conditions:
To effectively characterize DDX50-TRIF interactions:
Co-immunoprecipitation strategies:
Stimulation conditions:
Proximity-based assays:
Proximity ligation assay (PLA) to visualize endogenous interactions
FRET or BiFC (Bimolecular Fluorescence Complementation) for live-cell interaction dynamics
Functional validation:
DDX50 has dual roles in RNA stability and antiviral response that can be experimentally distinguished:
For RNA stability assessment:
Perform actinomycin D chase experiments to measure mRNA decay rates in DDX50-depleted versus control cells
Focus on specific pro-differentiation RNAs such as JUN, OVOL1, CEBPB, PRDM1, and TINCR
Analyze DDX50-STAU1 interactions using co-immunoprecipitation followed by RNA sequencing
Measure changes in RNA structure using SHAPE (Selective 2′-hydroxyl acylation analyzed by primer extension) in the presence and absence of DDX50
For antiviral function assessment:
Measure IRF3 phosphorylation and nuclear translocation
Quantify expression of IRF3-dependent genes like Isg56 and Ifnb
Analyze cytokine production (CXCL10, IL-6) in response to viral infection
Perform viral replication assays under different glucose conditions to dissect metabolic from antiviral functions
Comparative analysis:
The literature shows contradictory roles for DDX50 in HIV-1 replication. To resolve these contradictions:
Systematically compare experimental conditions:
Cell type differences: Primary CD4+ T cells versus cell lines may show different DDX50 functions
Infection parameters: MOI, viral strains, and infection duration
Knockdown methods: siRNA versus CRISPR, acute versus stable depletion
Measure multiple aspects of viral replication: Early versus late replication events
Analyze pathway context:
Data integration matrix:
To systematically analyze this complex relationship:
Experimental design matrix:
Functional readouts across conditions:
Measure DDX50 dimerization status using native PAGE or crosslinking approaches
Assess DDX50-STAU1 complex formation using co-immunoprecipitation
Quantify binding to target RNAs using RNA immunoprecipitation
Monitor antiviral response markers (IRF3 phosphorylation, ISG expression)
Track differentiation markers in parallel experiments
Use DDX50 mutants strategically:
When troubleshooting non-specific binding:
Optimize antibody conditions:
Titrate antibody concentration (start with 1-5 μg for IP)
Try different antibody clones or host species
Consider using epitope-tagged DDX50 constructs if specificity issues persist
Modify binding and washing conditions:
Increase salt concentration (150-500 mM NaCl) to reduce non-specific interactions
Add detergents (0.1-0.5% NP-40 or Triton X-100) to reduce hydrophobic interactions
Include competitors like BSA (0.1-1%) to block non-specific binding sites
Use more stringent washing steps (increase number or duration)
Special considerations for DDX50:
Be aware that DDX50 forms dimers that can be disrupted by glucose
DDX50's interactions with RNA may influence co-IP results; consider RNase treatment controls
DDX50 shares high homology with DDX21 (55.6%); validate antibody specificity against both proteins
DDX50 shuttles between nucleus and cytoplasm; consider cell fractionation to enrich for relevant pools
For detecting DDX50 across tissues with varying expression:
Sample preparation optimization:
Use tissue-specific extraction buffers that account for varying protein content
Consider protease inhibitor cocktails optimized for each tissue type
Normalize loading based on total protein rather than housekeeping genes
For tissues with low expression, increase sample concentration or use immunoprecipitation before Western blot
Signal enhancement strategies:
Use high-sensitivity detection systems (ECL Prime or fluorescent secondaries)
Consider signal amplification methods (like biotin-streptavidin systems)
Optimize exposure times based on preliminary tissue expression data
For immunohistochemistry, use polymer detection systems with DAB enhancement
Expression reference guide:
Based on research findings, expected relative DDX50 expression levels:
Recent discovery of DDX50 as a glucose-binding protein opens new research avenues:
Experimental approaches:
Compare antiviral responses under different glucose concentrations
Use glucose-binding mutants (V227W, K187G) to dissect metabolic from non-metabolic functions
Measure DDX50 dimerization status as a readout of glucose binding in infected cells
Assess whether viral infection alters glucose binding to DDX50
Research questions to address:
Does hyperglycemia or hypoglycemia alter DDX50-mediated viral restriction?
Can glucose analogs modulate DDX50's antiviral functions?
Do viruses target DDX50-glucose binding as an immune evasion strategy?
Is DDX50 glucose binding altered in metabolic diseases, affecting antiviral immunity?
Methodological considerations:
Use cellular thermal shift assays (CETSAs) to measure DDX50-glucose binding during infection
Apply fluorescence quenching assays to quantify binding under different conditions
Employ microscale thermophoresis to measure binding affinities of DDX50 mutants
Develop biosensors to monitor DDX50 conformational changes in real-time
To explore DDX50 as a therapeutic target:
Screen for DDX50 modulators:
Develop high-throughput assays measuring DDX50-dependent IRF3 activation
Screen small molecule libraries for compounds that enhance DDX50 activity
Identify compounds that stabilize DDX50 monomers, mimicking glucose binding effects
Use fragment-based drug discovery focusing on the ATP-binding domain
Therapeutic strategy validation:
Test candidate compounds against multiple viruses (VACV, HSV-1, ZIKV) to assess broad-spectrum potential
Evaluate compounds in both prevention and treatment models
Assess effects on normal cellular differentiation to identify potential side effects
Determine if combination with established antivirals produces synergistic effects
Translational considerations:
Compare DDX50 sequence conservation across species to guide animal model selection
Develop tissue-specific delivery strategies for DDX50 modulators
Assess potential for resistance development through viral mutation
Consider repurposing glucose analogs or metabolic drugs that may interact with DDX50
To investigate structural dynamics:
Advanced structural biology techniques:
Cryo-electron microscopy to visualize DDX50 conformational states with/without glucose
Hydrogen-deuterium exchange mass spectrometry to map conformational changes
Nuclear magnetic resonance (NMR) to track dynamic changes in protein structure
X-ray crystallography of DDX50 in different binding states
Protein interaction mapping:
Use BioID or APEX proximity labeling to identify DDX50 interaction partners in different glucose conditions
Apply crosslinking mass spectrometry to capture transient interactions
Perform yeast two-hybrid screens with DDX50 mutants mimicking different conformational states
Use protein complementation assays to visualize interaction dynamics in live cells
Computational approaches:
Molecular dynamics simulations to model glucose-induced conformational changes
Protein-protein docking predictions for DDX50-STAU1 and DDX50-TRIF interactions
Machine learning analysis of binding site conservation across DDX family members
Systems biology modeling of DDX50's role in signaling networks under different metabolic conditions