The DHX36 antibody is a critical tool for studying the DEAH-box helicase 36 (DHX36), an ATP-dependent enzyme that resolves G4-quadruplex structures in DNA and RNA. DHX36 plays pivotal roles in genomic stability, transcriptional regulation, and immune responses . This antibody enables the detection and functional analysis of DHX36 in various cellular contexts, including cancer, neurodegeneration, and viral immunity.
DHX36 contains a conserved helicase core with six functional domains, including the RHAU-specific motif (RSM), which facilitates interaction with G-rich regions . Its helicase activity is essential for unwinding G4 structures, which are implicated in transcriptional regulation and genomic instability . DHX36 also functions as a dsRNA sensor in innate immunity, triggering antiviral pathways by activating stress granules .
Lung Cancer: DHX36 knockdown accelerates tumor growth and reduces sensitivity to chemotherapy, suggesting its role as a tumor suppressor .
Breast Cancer: High DHX36 expression correlates with improved patient survival, with knockdown enhancing tumor invasion and proliferation .
DHX36 binds to G-rich mRNA regions (e.g., 5’UTRs), regulating translation efficiency by resolving G4 structures. Its loss stabilizes target mRNAs, such as WAC and PURB, leading to altered protein synthesis .
DHX36 interacts with replication protein A (RPA) to resolve G4-DNA during replication, preventing DNA damage. Depletion increases 53BP1 foci (a marker of double-strand breaks), indicating compromised genomic stability .
The DHX36 antibody targets a multifunctional ATP-dependent helicase that unwinds G-quadruplex (G4) structures. This enzyme plays a crucial role in various biological processes, including genomic stability, gene expression regulation, and antiviral response initiation. G4 structures are helical structures characterized by guanine tetrads. DHX36 exhibits high-affinity binding to and unwinding of G4 structures formed in nucleic acids (G4-DNA and G4-RNA).
Specific roles of DHX36 include:
Further research supporting the functions of DHX36 and related helicases is detailed below:
DHX36 (also known as G4R1 or RHAU) is a multifunctional ATP-dependent helicase that specifically unwinds G-quadruplex (G4) structures in both DNA and RNA. It plays crucial roles in genomic integrity, gene expression regulation, and antiviral responses . The protein's ability to resolve G4 structures makes it particularly important for maintaining cellular function, as these structures can impede DNA replication, transcription, and translation. DHX36 is predominantly localized in the cytoplasm and actively interacts with polyadenylated RNA, suggesting its primary role in post-transcriptional regulation .
DHX36 antibodies have been validated for multiple experimental applications:
Western Blotting (WB): Reliable detection at 1:500-1:2000 dilution
Immunoprecipitation (IP): Effective at 0.5-4.0 μg for 1.0-3.0 mg total protein lysate
Immunohistochemistry (IHC): Functional at 1:50-1:500 dilution
RNA Immunoprecipitation (RIP): Validated in multiple publications
Co-immunoprecipitation (CoIP): Confirmed for protein-protein interaction studies
Immunofluorescence (IF): Validated for subcellular localization studies
For robust validation, researchers should:
Generate DHX36 knockdown (KD) or knockout (KO) cell lines using RNA interference (shRNA) or CRISPR-Cas9 technologies
Confirm knockdown efficiency by quantitative PCR and Western blot analysis
Run parallel immunoblots with wildtype and KD/KO samples under identical conditions
Include appropriate loading controls
Compare band intensity at the expected molecular weight (110-120 kDa)
Research shows that doxycycline-inducible DHX36 knockdown systems can achieve significant protein reduction after 96 hours of treatment, as demonstrated by immunoblot analysis . This approach provides a valuable control for antibody specificity verification.
When investigating G-quadruplex (G4) structures with DHX36 antibodies:
Sample preparation: Use chemical crosslinking (like formaldehyde) to preserve protein-nucleic acid interactions
G4 stabilizing conditions: Include potassium ions (K+) in buffers to stabilize G4 structures
Controls: Include parallel experiments with:
Validation methods: Combine antibody approaches with G4-specific detection methods like BG4 antibody staining or small molecule G4 ligands
Research indicates that DHX36 exhibits much higher binding and enzymatic activities for G4-DNA compared to unstructured DNA forms, making proper experimental controls critical .
When applying DHX36 antibodies in neurological disease research:
Sample selection: Select appropriate tissue regions based on disease pathology (e.g., medial prefrontal cortex for fear conditioning studies)
Timing considerations: DHX36 expression can vary temporally after learning or stress events (significant increase observed at 5h post-training in fear conditioning)
Cell-type specificity: Consider cell type-specific analysis as DHX36 distribution depends on neuronal activation states
Disease models: For C9orf72-related ALS/FTD research, assess both RAN translation and G4 structure formation using complementary approaches
Validation: Confirm findings in patient-derived cells with endogenous repeat expansions alongside model systems
Studies show DHX36 depletion suppresses RAN translation in a repeat length-dependent manner, offering therapeutic implications for nucleotide repeat expansion disorders .
For stress response investigations:
Stress granule detection: Co-stain with DHX36 antibody and stress granule markers (G3BP1, TIA-1) before and after stress induction
Integrated stress response: Monitor DHX36 during ISR activation using:
Subcellular fractionation: Perform cytoplasmic/nuclear fractionation followed by immunoblotting to track stress-induced relocalization
RIP-seq workflow:
Crosslink cells during stress response
Immunoprecipitate with DHX36 antibody
Sequence associated RNAs
Analyze G4-forming potential and stress-responsive mRNAs
Research indicates DHX36 knockout results in increased stress granule formation and protein kinase R (PKR/EIF2AK2) phosphorylation, suggesting its role in resolving rG4-induced cellular stress .
To investigate DHX36-RPA interactions:
Co-immunoprecipitation strategy:
Proximity ligation assay:
Fix cells during various cell cycle stages
Incubate with DHX36 antibody and anti-RPA antibodies
Perform proximity ligation following manufacturer protocols
Quantify interaction signals and correlate with cell cycle phases
Chromatin immunoprecipitation (ChIP):
Crosslink protein-DNA complexes
Perform sequential ChIP with DHX36 and RPA antibodies
Identify genomic regions where both proteins co-localize
Focus analysis on regions prone to G4 formation
Research shows DHX36 coimmunoprecipitates with RPA, suggesting functional interactions during DNA replication and transcription events when G4-DNA can form .
Common challenges and solutions include:
High background signal:
Inconsistent staining between tissues:
Validation approaches:
Include DHX36 knockdown/knockout control tissues
Perform peptide competition assays
Compare staining patterns with multiple antibodies targeting different DHX36 epitopes
When facing contradictory results:
Cell-type specific considerations:
DHX36 expression and localization can vary significantly between cell types
Verify DHX36 expression levels in each cell line by qPCR
Consider isoform expression differences that may affect antibody recognition
Technical validation steps:
Confirm antibody specificity in each cell line using knockdown controls
Test multiple antibodies targeting different DHX36 epitopes
Validate with orthogonal methods (e.g., mass spectrometry)
Experimental condition effects:
Data analysis:
Normalize to appropriate housekeeping controls for each cell line
Consider statistical approaches for handling variability
Report all experimental details to facilitate reproduction
For neurodegenerative disease research:
C9orf72 ALS/FTD models:
Use DHX36 antibodies in conjunction with dipeptide repeat protein (DPR) antibodies to assess correlation between DHX36 levels and RAN translation products
Compare DHX36 binding to C9orf72 repeat RNA using RNA immunoprecipitation followed by qPCR
Assess effects of DHX36 modulation on repeat-associated toxicity
Patient-derived samples:
Compare DHX36 levels in patient vs. control tissues
Assess co-localization with pathological aggregates
Evaluate DHX36 sequestration by disease-associated repeat expansions
Therapeutic target validation:
Monitor changes in DHX36 localization or activity following potential therapeutic interventions
Assess DHX36 helicase activity with G4-RNA substrates derived from disease-relevant sequences
Studies indicate that DHX36 depletion suppresses RAN translation from C9orf72 repeat expansions, suggesting it as a potential therapeutic target for G-rich repeat-associated neurological diseases .
To investigate DHX36 in DNA damage responses:
DNA damage induction and assessment:
Co-localization analysis:
Perform immunofluorescence co-staining with DHX36 antibody and DNA damage markers (γH2AX, 53BP1)
Analyze recruitment kinetics of DHX36 to damage sites
Quantify co-localization coefficients at different time points after damage
Cell cycle analysis:
Synchronize cells and induce damage at specific cell cycle phases
Assess DHX36 recruitment to damage sites in relation to cell cycle stage
Monitor cell cycle progression following damage in DHX36-proficient vs. deficient cells
G4-related damage assessment:
Use G4 ligands to induce G4-stabilization and analyze DHX36 recruitment
Compare damage responses at G4-forming vs. non-G4 genomic regions
Research indicates DHX36 may suppress DNA damage by promoting the clearance of G4-DNA structures, supporting cell growth and survival .
For comprehensive RIP-seq analysis:
Experimental setup:
RNA processing and sequencing:
Extract RNA from immunoprecipitates
Verify enrichment by qRT-PCR of known targets
Prepare libraries and perform deep sequencing
Analyze data for G-rich motifs and G4-forming potential
Bioinformatic analysis:
Identify enriched transcripts compared to input and IgG controls
Analyze for G-quadruplex forming sequences (G3+N1-7)n pattern
Perform Gene Ontology analysis on target transcripts
Compare with published DHX36 targets
Research has identified >4500 mRNAs as DHX36 targets, showing preferential interaction with G-rich and G4-forming sequences, and DHX36 knockout resulting in increased target mRNA abundance but decreased ribosome occupancy .
To evaluate DHX36's impact on translation:
SUnSET (Surface Sensing of Translation) assay:
Polysome profiling:
Fractionate cytoplasmic lysates on sucrose gradients
Collect fractions and analyze by western blotting for DHX36
Extract RNA from each fraction for specific target analysis
Compare polysome association of G4-containing vs. control mRNAs
Ribosome profiling with DHX36 modulation:
Generate libraries from ribosome-protected fragments in control vs. DHX36-depleted cells
Analyze translational efficiency of G4-containing mRNAs
Investigate ribosomal pausing at G4 motifs
Target-specific translation assays:
Use luciferase reporters containing G4 motifs
Compare translation efficiency in DHX36 wildtype vs. knockdown conditions
Normalize to non-G4 control reporters
Studies demonstrate that while DHX36 knockout increases target mRNA abundance, ribosome occupancy and protein output from these targets decrease, suggesting they become translationally incompetent .
For learning and memory studies:
Behavioral paradigm setup:
Brain region-specific analysis:
Process medial prefrontal cortex (mPFC) or other relevant regions
Perform DHX36 immunohistochemistry and western blotting
Compare expression levels between trained and control animals
G4-DNA immunoprecipitation:
Perform G4-DNA IP followed by sequencing
Analyze distribution patterns (5' UTR, proximal TSS, CDS, 3' UTR)
Compare G4-DNA profiles between retention control and extinction-trained animals
In vivo intervention:
Infuse DHX36 shRNA into the mPFC to knockdown expression
Assess behavioral outcomes and G4-DNA distribution
Compare with scrambled control shRNA
Research demonstrates that DHX36 mRNA expression is transiently induced by both fear conditioning and extinction learning, suggesting its role in learning-dependent genomic regulation .
For studying DHX36 in antiviral responses:
Viral infection models:
Select appropriate viral systems (RNA virus preferred)
Monitor DHX36 localization before and after infection
Assess co-localization with viral RNA using immunofluorescence and in situ hybridization
Multi-helicase-TICAM1 complex analysis:
Immunoprecipitate DHX36 from infected cells
Analyze co-precipitation of TICAM1 and other complex components
Compare complex formation in response to different viral stimuli
DHX36 functions as a component of a multi-helicase-TICAM1 complex that acts as a cytoplasmic sensor of viral double-stranded RNA
Signaling pathway investigation:
Monitor pro-inflammatory cytokine induction in control vs. DHX36-depleted cells
Assess phosphorylation of downstream signaling components
Correlate with viral replication efficiency
G4-viral RNA interaction:
Analyze viral sequences for G4-forming potential
Perform RNA immunoprecipitation to detect DHX36-viral RNA interactions
Use G4-stabilizing ligands to assess effects on DHX36-mediated sensing
For robust computational analysis:
Binding site identification:
Use peak calling algorithms optimized for G-rich regions
Apply appropriate background models
Consider G-content normalization to avoid bias
Motif analysis:
Search for G-quadruplex consensus sequences (G3+N1-7)n
Analyze for other structural elements beyond primary sequence
Compare with known G4 databases
Cross-validation approaches:
Correlate DHX36 binding sites with G4 structures identified by:
G4-seq
BG4 ChIP-seq
G4-selective chemical probing
Validate a subset of targets using orthogonal techniques
Functional analysis of targets:
When analyzing seemingly contradictory effects:
Integrated analysis approach:
Mechanistic interpretation framework:
DHX36 binding to G4 structures can have dual effects:
Resolution of G4s in 5' UTRs may enhance translation initiation
Resolution of G4s may also decrease RNA stability
Consider subcellular localization of affected transcripts
Analyze stress granule association of targets
G4 structure prediction:
Correlate effects with predicted G4 stability
Consider G4 location within the transcript
Analyze for alternative secondary structures
Data visualization:
Create scatterplots of mRNA abundance vs. translational efficiency
Identify outlier transcripts for further mechanistic studies
Group targets by shared biological pathways
Research demonstrates that loss of DHX36 results in target mRNAs with G-rich structures becoming translationally inactive despite increased abundance, potentially due to stress granule sequestration .
For therapeutic development research:
Target validation in disease models:
Create cell and animal models expressing pathogenic repeat expansions
Modulate DHX36 levels through knockdown, knockout, or overexpression
Measure repeat-associated non-AUG (RAN) translation using reporter assays
Research shows DHX36 depletion suppresses RAN translation in a repeat length-dependent manner
Small molecule screening:
Design assays to identify compounds that modulate DHX36-G4 interactions
Measure effects on:
RAN translation efficiency
G4 resolution activity
Target RNA stability and translation
Therapeutic delivery approaches:
Test antisense oligonucleotides targeting DHX36
Evaluate viral vector-mediated DHX36 modulation
Assess cell-type specific effects in relevant disease tissues
Combinatorial therapeutic strategies:
Test DHX36 modulation with G4-stabilizing compounds
Combine with integrated stress response modulators
Evaluate effects on disease-relevant phenotypes
Studies suggest that modulation of G4-resolving helicases like DHX36 represents a candidate therapeutic strategy for G-rich repeat-associated neurological diseases .
To study protein-protein interactions in G4 biology:
Sequential ChIP (ChIP-reChIP):
Perform first immunoprecipitation with DHX36 antibody
Elute complexes and perform second IP with antibodies against other G4 binding proteins
Identify genomic regions with co-occupancy
Correlate with G4 formation potential
Proximity-based protein identification:
Express DHX36 fused to a proximity labeling enzyme (BioID or APEX)
Identify biotinylated proteins in G4-rich environments
Validate interactions with co-immunoprecipitation
Analyze temporal dynamics of interactions
Competition assays:
Assess binding affinity changes in the presence of other G4 binding proteins
Determine if relationships are competitive, cooperative, or sequential
Measure effects on G4 resolution efficiency
Functional validation:
Create combined knockdowns of DHX36 and other G4 binding proteins
Assess synergistic or antagonistic effects on:
Target mRNA abundance
Translation efficiency
G4-associated genomic stability